Neutrophil extracellular traps in cancer: From mechanisms to treatments

Yifan Wang , Kangjie Yang , Juan Li , Chuanxin Wang , Peilong Li , Lutao Du

Clinical and Translational Medicine ›› 2025, Vol. 15 ›› Issue (6) : e70368

PDF
Clinical and Translational Medicine ›› 2025, Vol. 15 ›› Issue (6) : e70368 DOI: 10.1002/ctm2.70368
REVIEW

Neutrophil extracellular traps in cancer: From mechanisms to treatments

Author information +
History +
PDF

Abstract

Neutrophil extracellular traps (NETs) are reticular ultrastructures released by activated neutrophils. As the reaction products of neutrophils, NETs have been identified as crucial effectors in pathogen defence and autoimmune diseases. Recently, increasing evidence suggest that this process also occurs in cancer. The formation and clearance of NETs are dynamically influenced by the tumour microenvironment, while NETs reciprocally play a dual role in either promoting or inhibiting tumour progression through their DNA scaffold, proteases and other granule-derived proteins. Given the interplay between NETs and tumours, active exploration is currently underway to harness their potential as tumour biomarkers and therapeutic targets. Here, we delve into the biochemical and immunological mechanisms underlying NETs formation within the tumour microenvironment, along with recent advances elucidating their multifaceted roles in tumourigenesis, metastasis and tumour-associated co-morbidities. Furthermore, we present emerging strategies for NETs-based tumour diagnostic approaches and therapeutics, with a special focus on the challenging questions that need to be answered within this field.

Keywords

biomarkers / cancer / neutrophil extracellular traps (NETs) / therapeutic target

Cite this article

Download citation ▾
Yifan Wang, Kangjie Yang, Juan Li, Chuanxin Wang, Peilong Li, Lutao Du. Neutrophil extracellular traps in cancer: From mechanisms to treatments. Clinical and Translational Medicine, 2025, 15(6): e70368 DOI:10.1002/ctm2.70368

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Brinkmann V, Reichard U, Goosmann C, et al. Neutrophil extracellular traps kill bacteria. Science. 2004; 303(5663): 1532-1535.

[2]

Brinkmann V. Neutrophil extracellular traps in the second decade. J Innate Immun. 2018; 10(5-6): 414-421.

[3]

Silva CMS, Wanderley CWS, Veras FP, et al. Gasdermin D inhibition prevents multiple organ dysfunction during sepsis by blocking NET formation. Blood. 2021; 138(25): 2702-2713.

[4]

Chen F, Chu C, Wang X, et al. Hesperetin attenuates sepsis-induced intestinal barrier injury by regulating neutrophil extracellular trap formation via the ROS/autophagy signaling pathway. Food Funct. 2023; 14(9): 4213-4227.

[5]

Tokuyama M, Gunn BM, Venkataraman A, et al. Antibodies against human endogenous retrovirus K102 envelope activate neutrophils in systemic lupus erythematosus. J Exp Med. 2021; 218(7): e20191766.

[6]

Dou H, Kotini A, Liu W, et al. Oxidized phospholipids promote NETosis and arterial thrombosis in LNK(SH2B3) deficiency. Circulation. 2021; 144(24): 1940-1954.

[7]

Fang H, Shao S, Xue K, et al. Neutrophil extracellular traps contribute to immune dysregulation in bullous pemphigoid via inducing B-cell differentiation and antibody production. FASEB J. 2021; 35(7): e21746.

[8]

van der Velden S, van Osch TLJ, Seghier A, et al. Complement activation drives antibody-mediated transfusion-related acute lung injury via macrophage trafficking and formation of NETs. Blood. 2024; 143(1): 79-91.

[9]

Zhu YP, Speir M, Tan Z, et al. NET formation is a default epigenetic program controlled by PAD4 in apoptotic neutrophils. Sci Adv. 2023; 9(51): eadj1397.

[10]

Li S, Ma Y, Ye S, et al. ERK/p38/ROS burst responses to environmentally relevant concentrations of diphenyl phosphate-evoked neutrophil extracellular traps formation: assessing the role of autophagy. J Hazard Mater. 2022; 421: 126758.

[11]

Yu J, Fu Y, Gao J, et al. Cathepsin C from extracellular histone-induced M1 alveolar macrophages promotes NETosis during lung ischemia-reperfusion injury. Redox Biol. 2024; 74: 103231.

[12]

Powell LC, Cullen JK, Boyle GM, et al. Topical, immunomodulatory epoxy-tiglianes induce biofilm disruption and healing in acute and chronic skin wounds. Sci Transl Med. 2022; 14(662): eabn3758.

[13]

Hintermann E, Tondello C, Fuchs S, et al. Blockade of neutrophil extracellular trap components ameliorates cholestatic liver disease in Mdr2 (Abcb4) knockout mice. J Autoimmun. 2024; 146: 103229.

[14]

Reis LR, Souza Junior DR, Tomasin R, et al. Citrullination of actin-ligand and nuclear structural proteins, cytoskeleton reorganization and protein redistribution across cellular fractions are early events in ionomycin-induced NETosis. Redox Biol. 2023; 64: 102784.

[15]

Cristinziano L, Modestino L, Antonelli A, et al. Neutrophil extracellular traps in cancer. Semin Cancer Biol. 2022; 79: 91-104.

[16]

Sollberger G, Choidas A, Burn GL, et al. Gasdermin D plays a vital role in the generation of neutrophil extracellular traps. Sci Immunol. 2018; 3(26): eaar6689.

[17]

Pilsczek FH, Salina D, Poon KK, et al. A novel mechanism of rapid nuclear neutrophil extracellular trap formation in response to Staphylococcus aureus. J Immunol. 2010; 185(12): 7413-7425.

[18]

Kusakabe T, Lin WY, Cheong JG, et al. Fungal microbiota sustains lasting immune activation of neutrophils and their progenitors in severe COVID-19. Nat Immunol. 2023; 24(11): 1879-1889.

[19]

Firouzjaie F, Taghipour N, Akhavan AA, et al. Neutrophil extracellular traps formation: effect of Leishmania major promastigotes and salivary gland homogenates of Phlebotomus papatasi in human neutrophil culture. BMC Microbiol. 2024; 24(1): 117.

[20]

Yipp BG, Petri B, Salina D, et al. Infection-induced NETosis is a dynamic process involving neutrophil multitasking in vivo. Nat Med. 2012; 18(9): 1386-1393.

[21]

Muñoz-Caro T, Gibson AJ, Conejeros I, et al. The role of TLR2 and TLR4 in recognition and uptake of the apicomplexan parasite Eimeria bovis and their effects on NET formation. Pathogens. 2021; 10(2): 118.

[22]

Yousefi S, Mihalache C, Kozlowski E, et al. Viable neutrophils release mitochondrial DNA to form neutrophil extracellular traps. Cell Death Differ. 2009; 16(11): 1438-1444.

[23]

Dunham-Snary KJ, Surewaard BG, Mewburn JD, et al. Mitochondria in human neutrophils mediate killing of Staphylococcus aureus. Redox Biol. 2022; 49: 102225.

[24]

Frost JN, Wideman SK, Preston AE, et al. Plasma iron controls neutrophil production and function. Sci Adv. 2022; 8(40): eabq5384.

[25]

Yang C, Wang Z, Li L, et al. Aged neutrophils form mitochondria-dependent vital NETs to promote breast cancer lung metastasis. J Immunother Cancer. 2021; 9(10): 1.

[26]

Cristinziano L, Modestino L, Loffredo S, et al. Anaplastic thyroid cancer cells induce the release of mitochondrial extracellular DNA traps by viable neutrophils. J Immunol. 2020; 204(5): 1362-1372.

[27]

Munir H, Jones JO, Janowitz T, et al. Stromal-driven and amyloid β-dependent induction of neutrophil extracellular traps modulates tumor growth. Nat Commun. 2021; 12(1): 683.

[28]

Bejarano L, Jordāo MJC, Joyce JA. Therapeutic targeting of the tumor microenvironment. Cancer Discov. 2021; 11(4): 933-959.

[29]

Zhang Y, Chandra V, Riquelme Sanchez E, et al. Interleukin-17-induced neutrophil extracellular traps mediate resistance to checkpoint blockade in pancreatic cancer. J Exp Med. 2020; 217(12): e20190354.

[30]

Cambier S, Gouwy M, Proost P. The chemokines CXCL8 and CXCL12: molecular and functional properties, role in disease and efforts towards pharmacological intervention. Cell Mol Immunol. 2023; 20(3): 217-251.

[31]

Linde IL, Prestwood TR, Qiu J, et al. Neutrophil-activating therapy for the treatment of cancer. Cancer Cell. 2023; 41(2): 356-372.e310.

[32]

Modestino L, Cristinziano L, Trocchia M, et al. Melanoma-derived soluble mediators modulate neutrophil biological properties and the release of neutrophil extracellular traps. Cancer Immunol Immunother. 2023; 72(10): 3363-3376.

[33]

Adel RM, Helal H, Ahmed Fouad M, et al. Regulation of miRNA-155-5p ameliorates NETosis in pulmonary fibrosis rat model via inhibiting its target cytokines IL-1β, TNF-α and TGF-β1. Int Immunopharmacol. 2024; 127: 111456.

[34]

Taifour T, Attalla SS, Zuo D, et al. The tumor-derived cytokine Chi3l1 induces neutrophil extracellular traps that promote T cell exclusion in triple-negative breast cancer. Immunity. 2023; 56(12): 2755-2772.e2758.

[35]

Nie M, Yang L, Bi X, et al. Neutrophil extracellular traps induced by IL8 promote diffuse large B-cell lymphoma progression via the TLR9 signaling. Clin Cancer Res. 2019; 25(6): 1867-1879.

[36]

Muqaku B, Pils D, Mader JC, et al. Neutrophil extracellular trap formation correlates with favorable overall survival in high grade ovarian cancer. Cancers (Basel). 2020; 12(2): 505.

[37]

Ortiz-Espinosa S, Morales X, Senent Y, et al. Complement C5a induces the formation of neutrophil extracellular traps by myeloid-derived suppressor cells to promote metastasis. Cancer Lett. 2022; 529: 70-84.

[38]

Mallavia B, Liu F, Lefrançais E, et al. Mitochondrial DNA stimulates TLR9-dependent neutrophil extracellular trap formation in primary graft dysfunction. Am J Respir Cell Mol Biol. 2020; 62(3): 364-372.

[39]

Xiao Y, Cong M, Li J, et al. Cathepsin C promotes breast cancer lung metastasis by modulating neutrophil infiltration and neutrophil extracellular trap formation. Cancer Cell. 2021; 39(3): 423-437.e427.

[40]

Guimarães-Bastos D, Frony AC, Barja-Fidalgo C, et al. Melanoma-derived extracellular vesicles skew neutrophils into a pro-tumor phenotype. J Leukoc Biol. 2022; 111(3): 585-596.

[41]

Shang A, Gu C, Zhou C, et al. Exosomal KRAS mutation promotes the formation of tumor-associated neutrophil extracellular traps and causes deterioration of colorectal cancer by inducing IL-8 expression. Cell Commun Signal. 2020; 18(1): 52.

[42]

Leal AC, Mizurini DM, Gomes T, et al. Tumor-derived exosomes induce the formation of neutrophil extracellular traps: implications for the establishment of cancer-associated thrombosis. Sci Rep. 2017; 7(1): 6438.

[43]

de Visser KE, Joyce JA. The evolving tumor microenvironment: from cancer initiation to metastatic outgrowth. Cancer Cell. 2023; 41(3): 374-403.

[44]

Li Y, Yang Y, Gan T, et al. Extracellular RNAs from lung cancer cells activate epithelial cells and induce neutrophil extracellular traps. Int J Oncol. 2019; 55(1): 69-80.

[45]

Zheng XB, Wang X, Gao SQ, et al. NINJ1-mediated plasma membrane rupture of pyroptotic endothelial cells exacerbates blood-brain barrier destruction caused by neutrophil extracellular traps in traumatic brain injury. Cell Death Discov. 2025; 11(1): 69.

[46]

Pan J, Zhang L, Wang X, et al. Chronic stress induces pulmonary epithelial cells to produce acetylcholine that remodels lung pre-metastatic niche of breast cancer by enhancing NETosis. J Exp Clin Cancer Res. 2023; 42(1): 255.

[47]

Chen L, Liu Y, Wang Z, et al. Mesenchymal stem cell-derived extracellular vesicles protect against abdominal aortic aneurysm formation by inhibiting NET-induced ferroptosis. Exp Mol Med. 2023; 55(5): 939-951.

[48]

Ou Q, Tan L, Shao Y, et al. Electrostatic charge-mediated apoptotic vesicle biodistribution attenuates sepsis by switching neutrophil NETosis to apoptosis. Small. 2022; 18(20): e2200306.

[49]

Zheng Z, Li YN, Jia S, et al. Lung mesenchymal stromal cells influenced by Th2 cytokines mobilize neutrophils and facilitate metastasis by producing complement C3. Nat Commun. 2021; 12(1): 6202.

[50]

Sutherland TE, Dyer DP, Allen JE. The extracellular matrix and the immune system: a mutually dependent relationship. Science. 2023; 379(6633): eabp8964.

[51]

Sangaletti S, Tripodo C, Vitali C, et al. Defective stromal remodeling and neutrophil extracellular traps in lymphoid tissues favor the transition from autoimmunity to lymphoma. Cancer Discov. 2014; 4(1): 110-129.

[52]

Deng J, Kang Y, Cheng CC, et al. DDR1-induced neutrophil extracellular traps drive pancreatic cancer metastasis. JCI Insight. 2021; 6(17): e146133.

[53]

Shen XT, Xie SZ, Zheng X, et al. Cirrhotic-extracellular matrix attenuates aPD-1 treatment response by initiating immunosuppressive neutrophil extracellular traps formation in hepatocellular carcinoma. Exp Hematol Oncol. 2024; 13(1): 20.

[54]

Chrysanthopoulou A, Gkaliagkousi E, Lazaridis A, et al. Angiotensin II triggers release of neutrophil extracellular traps, linking thromboinflammation with essential hypertension. JCI Insight. 2021; 6(18): e148668.

[55]

Abdol Razak N, Elaskalani O, Metharom P. Pancreatic cancer-induced neutrophil extracellular traps: a potential contributor to cancer-associated thrombosis. Int J Mol Sci. 2017; 18(3): 487.

[56]

Inoue M, Nakashima R, Enomoto M, et al. Plasma redox imbalance caused by albumin oxidation promotes lung-predominant NETosis and pulmonary cancer metastasis. Nat Commun. 2018; 9(1): 5116.

[57]

Kong X, Zhang Y, Xiang L, et al. Fusobacterium nucleatum-triggered neutrophil extracellular traps facilitate colorectal carcinoma progression. J Exp Clin Cancer Res. 2023; 42(1): 236.

[58]

Li G, Liu L, Lu T, et al. Gut microbiota aggravates neutrophil extracellular traps-induced pancreatic injury in hypertriglyceridemic pancreatitis. Nat Commun. 2023; 14(1): 6179.

[59]

Yang L, Li A, Wang Y, et al. Intratumoral microbiota: roles in cancer initiation, development and therapeutic efficacy. Signal Transduct Target Ther. 2023; 8(1): 35.

[60]

Xue R, Zhang Q, Cao Q, et al. Liver tumour immune microenvironment subtypes and neutrophil heterogeneity. Nature. 2022; 612(7938): 141-147.

[61]

Masucci MT, Minopoli M, Del Vecchio S, et al. The emerging role of neutrophil extracellular traps (NETs) in tumor progression and metastasis. Front Immunol. 2020; 11: 1749.

[62]

Chung JY, Tang PC, Chan MK, et al. Smad3 is essential for polarization of tumor-associated neutrophils in non-small cell lung carcinoma. Nat Commun. 2023; 14(1): 1794.

[63]

Antuamwine BB, Bosnjakovic R, Hofmann-Vega F, et al. N1 versus N2 and PMN-MDSC: a critical appraisal of current concepts on tumor-associated neutrophils and new directions for human oncology. Immunol Rev. 2023; 314(1): 250-279.

[64]

Chan YT, Tan HY, Lu Y, et al. Pancreatic melatonin enhances anti-tumor immunity in pancreatic adenocarcinoma through regulating tumor-associated neutrophils infiltration and NETosis. Acta Pharm Sin B. 2023; 13(4): 1554-1567.

[65]

Xie M, Hao Y, Feng L, et al. Neutrophil heterogeneity and its roles in the inflammatory network after ischemic stroke. Curr Neuropharmacol. 2023; 21(3): 621-650.

[66]

Salcher S, Sturm G, Horvath L, et al. High-resolution single-cell atlas reveals diversity and plasticity of tissue-resident neutrophils in non-small cell lung cancer. Cancer Cell. 2022; 40(12): 1503-1520.e1508.

[67]

Ng MSF, Kwok I, Tan L, et al. Deterministic reprogramming of neutrophils within tumors. Science. 2024; 383(6679): eadf6493.

[68]

Li X, Zhou J, Zhao C, et al. Research progress of tumor-associated neutrophils in the occurrence and development of lung cancer. Zhongguo Fei Ai Za Zhi. 2025; 28(1): 55-62.

[69]

You Y, Du Z, Tian Z, et al. Tumor-associated macrophages drive heterogenetic CD10(High) cancer stem cells to implement tumor-associated neutrophils reprogramming in oral squamous cell carcinoma. Int J Biol Sci. 2025; 21(3): 1110-1126.

[70]

Wang L, Liu Y, Dai Y, et al. Single-cell RNA-seq analysis reveals BHLHE40-driven pro-tumour neutrophils with hyperactivated glycolysis in pancreatic tumour microenvironment. Gut. 2023; 72(5): 958-971.

[71]

Masucci MT, Minopoli M, Carriero MV. Tumor associated neutrophils. Their role in tumorigenesis, metastasis, prognosis and therapy. Front Oncol. 2019; 9: 1146.

[72]

Li J, Xia Y, Sun B, et al. Neutrophil extracellular traps induced by the hypoxic microenvironment in gastric cancer augment tumour growth. Cell Commun Signal. 2023; 21(1): 86.

[73]

Voutouri C, Stylianopoulos T. Evolution of osmotic pressure in solid tumors. J Biomech. 2014; 47(14): 3441-3447.

[74]

Tibrewal S, Ivanir Y, Sarkar J, et al. Hyperosmolar stress induces neutrophil extracellular trap formation: implications for dry eye disease. Invest Ophthalmol Vis Sci. 2014; 55(12): 7961-7969.

[75]

Demkow U. Molecular mechanisms of neutrophil extracellular trap (NETs) degradation. Int J Mol Sci. 2023; 24(5): 4896.

[76]

Santocki M, Kolaczkowska E. On neutrophil extracellular trap (NET) removal: what we know thus far and why so little. Cells. 2020; 9(9): 2079.

[77]

Haider P, Kral-Pointner JB, Mayer J, et al. Neutrophil extracellular trap degradation by differently polarized macrophage subsets. Arterioscler Thromb Vasc Biol. 2020; 40(9): 2265-2278.

[78]

Liu Y, Ma J, Ma Y, et al. Neutrophil extracellular traps impede cancer metastatic seeding via protease-activated receptor 2-mediated downregulation of phagocytic checkpoint CD24. J Immunother Cancer. 2025; 13(2): e010813.

[79]

Zhao M, Chen Y, Bao X, et al. HuoXueTongFu formula induces M2c macrophages via the MerTK/PI3K/AKT pathway to eliminate NETs in intraperitoneal adhesion in mice. J Ethnopharmacol. 2024; 331: 118290.

[80]

Demers M, Krause DS, Schatzberg D, et al. Cancers predispose neutrophils to release extracellular DNA traps that contribute to cancer-associated thrombosis. Proc Natl Acad Sci U S A. 2012; 109(32): 13076-13081.

[81]

Houghton AM, Rzymkiewicz DM, Ji H, et al. Neutrophil elastase-mediated degradation of IRS-1 accelerates lung tumor growth. Nat Med. 2010; 16(2): 219-223.

[82]

Xu L, Kong Y, Li K, et al. Neutrophil extracellular traps promote growth of lung adenocarcinoma by mediating the stability of m6A-mediated SLC2A3 mRNA-induced ferroptosis resistance and CD8(+) T cell inhibition. Clin Transl Med. 2025; 15(2): e70192.

[83]

Wang Y, Liu F, Chen L, et al. Neutrophil extracellular traps (NETs) promote non-small cell lung cancer metastasis by suppressing lncRNA MIR503HG to activate the NF-κB/NLRP3 inflammasome pathway. Front Immunol. 2022; 13: 867516.

[84]

Albrengues J, Shields MA, Ng D, et al. Neutrophil extracellular traps produced during inflammation awaken dormant cancer cells in mice. Science. 2018; 361(6409): eaao4227.

[85]

Rawat K, Syeda S, Shrivastava A. Neutrophil-derived granule cargoes: paving the way for tumor growth and progression. Cancer Metastasis Rev. 2021; 40(1): 221-244.

[86]

Martins-Cardoso K, Almeida VH, Bagri KM, et al. Neutrophil extracellular traps (NETs) promote pro-metastatic phenotype in human breast cancer cells through epithelial-mesenchymal transition. Cancers (Basel). 2020; 12(6): 1542.

[87]

Wang X, Qu Y, Xu Q, et al. NQO1 triggers neutrophil recruitment and NET formation to drive lung metastasis of invasive breast cancer. Cancer Res. 2024; 84(21): 3538-3555.

[88]

Park J, Wysocki RW, Amoozgar Z, et al. Cancer cells induce metastasis-supporting neutrophil extracellular DNA traps. Sci Transl Med. 2016; 8(361): 361ra138.

[89]

Zhu B, Zhang X, Sun S, et al. NF-κB and neutrophil extracellular traps cooperate to promote breast cancer progression and metastasis. Exp Cell Res. 2021; 405(2): 112707.

[90]

Yang L, Liu Q, Zhang X, et al. DNA of neutrophil extracellular traps promotes cancer metastasis via CCDC25. Nature. 2020; 583(7814): 133-138.

[91]

Mousset A, Lecorgne E, Bourget I, et al. Neutrophil extracellular traps formed during chemotherapy confer treatment resistance via TGF-β activation. Cancer Cell. 2023; 41(4): 757-775.e710.

[92]

Global age-sex-specific mortality, life expectancy, and population estimates in 204 countries and territories and 811 subnational locations, 1950-2021, and the impact of the COVID-19 pandemic: a comprehensive demographic analysis for the Global Burden of Disease Study 2021. Lancet. 2024; 403(10440): 1989-2056.

[93]

Demkow U. Neutrophil extracellular traps (NETs) in cancer invasion, evasion and metastasis. Cancers (Basel). 2021; 13(17): 4495.

[94]

Yang S, Sun B, Li J, et al. Neutrophil extracellular traps promote angiogenesis in gastric cancer. Cell Commun Signal. 2023; 21(1): 176.

[95]

Zhu T, Zou X, Yang C, et al. Neutrophil extracellular traps promote gastric cancer metastasis by inducing epithelialmesenchymal transition. Int J Mol Med. 2021; 48(1): 127.

[96]

Kanamaru R, Ohzawa H, Miyato H, et al. Low density neutrophils (LDN) in postoperative abdominal cavity assist the peritoneal recurrence through the production of neutrophil extracellular traps (NETs). Sci Rep. 2018; 8(1): 632.

[97]

Zhang A, Zou X, Yang S, et al. Effect of NETs/COX-2 pathway on immune microenvironment and metastasis in gastric cancer. Front Immunol. 2023; 14: 1177604.

[98]

Liu D, Yang X, Wang X. Neutrophil extracellular traps promote gastric cancer cell metastasis via the NAT10-mediated N4-acetylcytidine modification of SMYD2. Cell Signal. 2024; 116: 111014.

[99]

Yang S, Zou X, Li J, et al. Immunoregulation and clinical significance of neutrophils/NETs-ANGPT2 in tumor microenvironment of gastric cancer. Front Immunol. 2022; 13: 1010434.

[100]

Li JC, Zou XM, Yang SF, et al. Neutrophil extracellular traps participate in the development of cancer-associated thrombosis in patients with gastric cancer. World J Gastroenterol. 2022; 28(26): 3132-3149.

[101]

Stehr AM, Wang G, Demmler R, et al. Neutrophil extracellular traps drive epithelial-mesenchymal transition of human colon cancer. J Pathol. 2022; 256(4): 455-467.

[102]

Pastor B, Abraham JD, Pisareva E, et al. Association of neutrophil extracellular traps with the production of circulating DNA in patients with colorectal cancer. iScience. 2022; 25(2): 103826.

[103]

Yang L, Liu L, Zhang R, et al. IL-8 mediates a positive loop connecting increased neutrophil extracellular traps (NETs) and colorectal cancer liver metastasis. J Cancer. 2020; 11(15): 4384-4396.

[104]

Tohme S, Yazdani HO, Al-Khafaji AB, et al. Neutrophil extracellular traps promote the development and progression of liver metastases after surgical stress. Cancer Res. 2016; 76(6): 1367-1380.

[105]

Cheng P, Wu J, Zong G, et al. Capsaicin shapes gut microbiota and pre-metastatic niche to facilitate cancer metastasis to liver. Pharmacol Res. 2023; 188: 106643.

[106]

Ascher S, Wilms E, Pontarollo G, et al. Gut microbiota restricts NETosis in acute mesenteric ischemia-reperfusion injury. Arterioscler Thromb Vasc Biol. 2020; 40(9): 2279-2292.

[107]

Balboa-Barreiro V, Pértega-Díaz S, García-Rodríguez T, et al. Colorectal cancer recurrence and its impact on survival after curative surgery: an analysis based on multistate models. Dig Liver Dis. 2024; 56(7): 1229-1236.

[108]

Gao J, Liu J, Lu J, et al. SKAP1 expression in cancer cells enhances colon tumor growth and impairs cytotoxic immunity by promoting neutrophil extracellular trap formation via the NFATc1/CXCL8 axis. Adv Sci (Weinh). 2024; 11(41): e2403430.

[109]

Vogel A, Meyer T, Sapisochin G, et al. Hepatocellular carcinoma. Lancet. 2022; 400(10360): 1345-1362.

[110]

Zhan X, Wu R, Kong XH, et al. Elevated neutrophil extracellular traps by HBV-mediated S100A9-TLR4/RAGE-ROS cascade facilitate the growth and metastasis of hepatocellular carcinoma. Cancer Commun (Lond). 2023; 43(2): 225-245.

[111]

Wang H, Zhang H, Wang Y, et al. Regulatory T-cell and neutrophil extracellular trap interaction contributes to carcinogenesis in non-alcoholic steatohepatitis. J Hepatol. 2021; 75(6): 1271-1283.

[112]

Xin H, Lai Q, Zhou Y, et al. Noninvasive evaluation of neutrophil extracellular traps signature predicts clinical outcomes and immunotherapy response in hepatocellular carcinoma. Front Immunol. 2023; 14: 1134521.

[113]

Yang LY, Luo Q, Lu L, et al. Increased neutrophil extracellular traps promote metastasis potential of hepatocellular carcinoma via provoking tumorous inflammatory response. J Hematol Oncol. 2020; 13(1): 3.

[114]

Velliou RI, Mitroulis I, Chatzigeorgiou A. Neutrophil extracellular traps contribute to the development of hepatocellular carcinoma in NASH by promoting Treg differentiation. Hepatobiliary Surg Nutr. 2022; 11(3): 415-418.

[115]

Chen J, Huang ZB, Liao CJ, et al. LncRNA TP73-AS1/miR-539/MMP-8 axis modulates M2 macrophage polarization in hepatocellular carcinoma via TGF-β1 signaling. Cell Signal. 2020; 75: 109738.

[116]

Li N, Zheng X, Chen M, et al. Deficient DNASE1L3 facilitates neutrophil extracellular traps-induced invasion via cyclic GMP-AMP synthase and the non-canonical NF-κB pathway in diabetic hepatocellular carcinoma. Clin Transl Immunol. 2022; 11(4): e1386.

[117]

Santocki M, Such A, Drab D, et al. NETs persisting in vasculature undergo self-renewal with consequences for subsequent infection: a mouse model study. Blood. 2025; 145(18): 2070-2085.

[118]

Li X, Wang Z, Jiao C, et al. Hepatocyte SGK1 activated by hepatic ischemia-reperfusion promotes the recurrence of liver metastasis via IL-6/STAT3. J Transl Med. 2023; 21(1): 121.

[119]

Ren J, He J, Zhang H, et al. Platelet TLR4-ERK5 axis facilitates NET-mediated capturing of circulating tumor cells and distant metastasis after surgical stress. Cancer Res. 2021; 81(9): 2373-2385.

[120]

Kajioka H, Kagawa S, Ito A, et al. Targeting neutrophil extracellular traps with thrombomodulin prevents pancreatic cancer metastasis. Cancer Lett. 2021; 497: 1-13.

[121]

Takesue S, Ohuchida K, Shinkawa T, et al. Neutrophil extracellular traps promote liver micrometastasis in pancreatic ductal adenocarcinoma via the activation of cancerassociated fibroblasts. Int J Oncol. 2020; 56(2): 596-605.

[122]

Jin W, Yin H, Li H, et al. Neutrophil extracellular DNA traps promote pancreatic cancer cells migration and invasion by activating EGFR/ERK pathway. J Cell Mol Med. 2021; 25(12): 5443-5456.

[123]

Luo H, Chen G. Neutrophil extracellular traps promote the proliferation, invasion and migration of prostate cancer cells by upregulating IL-8 expression in DU145 human prostate cancer cells. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi. 2023; 39(3): 261-267.

[124]

Ozel AB, Dagsuyu E, Aydın PK, et al. Brain boron level, DNA content, and myeloperoxidase activity of metformin-treated rats in diabetes and prostate cancer model. Biol Trace Elem Res. 2022; 200(3): 1164-1170.

[125]

Castaño M, Tomás-Pérez S, González-Cantó E, et al. Neutrophil extracellular traps and cancer: trapping our attention with their involvement in ovarian cancer. Int J Mol Sci. 2023; 24(6): 5995.

[126]

De Meo ML, Spicer JD. The role of neutrophil extracellular traps in cancer progression and metastasis. Semin Immunol. 2021; 57: 101595.

[127]

Rahat MA, Galdiero MR. Editorial: extracellular traps in cancer immunity and immunotherapy. Front Immunol. 2023; 14: 1292819.

[128]

Lee W, Ko SY, Akasaka H, et al. Neutrophil extracellular traps promote pre-metastatic niche formation in the omentum by expanding innate-like B cells that express IL-10. Cancer Cell. 2025; 43(1): 69-85.e11.

[129]

Ricciuti J, Liu Q, Khan A, et al. Prognostic significance of serum complement activation, neutrophil extracellular traps and extracellular DNA in newly diagnosed epithelial ovarian cancer. Gynecol Oncol. 2025; 193: 49-57.

[130]

Tomás-Pérez S, Oto J, Aghababyan C, et al. Increased levels of NETosis biomarkers in high-grade serous ovarian cancer patients' biofluids: potential role in disease diagnosis and management. Front Immunol. 2023; 14: 1111344.

[131]

Modestino L, Cristinziano L, Poto R, et al. Neutrophil extracellular traps and neutrophil-related mediators in human thyroid cancer. Front Immunol. 2023; 14: 1167404.

[132]

Guo H, Wang Z, Yin K, et al. Sciellin promotes the development and progression of thyroid cancer through the JAK2/STAT3 signaling pathway. Mol Carcinog. 2024; 63(4): 701-713.

[133]

Monti M, De Rosa V, Iommelli F, et al. Neutrophil extracellular traps as an adhesion substrate for different tumor cells expressing RGD-binding integrins. Int J Mol Sci. 2018; 19(8): 2350.

[134]

Zha C, Meng X, Li L, 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(1): 154-168.

[135]

Liu X, Wang Y, Bauer AT, et al. Neutrophils activated by membrane attack complexes increase the permeability of melanoma blood vessels. Proc Natl Acad Sci U S A. 2022; 119(33): e2122716119.

[136]

Weide LM, Schedel F, Weishaupt C. Neutrophil extracellular traps correlate with tumor necrosis and size in human malignant melanoma metastases. Biology (Basel). 2023; 12(6): 822.

[137]

Schedel F, Mayer-Hain S, Pappelbaum KI, et al. Evidence and impact of neutrophil extracellular traps in malignant melanoma. Pigment Cell Melanoma Res. 2020; 33(1): 63-73.

[138]

Demers M, Wong SL, Martinod K, et al. Priming of neutrophils toward NETosis promotes tumor growth. Oncoimmunology. 2016; 5(5): e1134073.

[139]

Cao W, Zhu MY, Lee SH, et al. Modulation of cellular NAD(+) attenuates cancer-associated hypercoagulability and thrombosis via the inhibition of tissue factor and formation of neutrophil extracellular traps. Int J Mol Sci. 2021; 22(21): 12085.

[140]

Abrams ST, Su D, Sahraoui Y, et al. Assembly of alternative prothrombinase by extracellular histones initiates and disseminates intravascular coagulation. Blood. 2021; 137(1): 103-114.

[141]

Guan X, Lu Y, Zhu H, et al. The crosstalk between cancer cells and neutrophils enhances hepatocellular carcinoma metastasis via neutrophil extracellular traps-associated cathepsin G component: a potential therapeutic target. J Hepatocell Carcinoma. 2021; 8: 451-465.

[142]

Miller-Ocuin JL, Liang X, Boone BA, et al. DNA released from neutrophil extracellular traps (NETs) activates pancreatic stellate cells and enhances pancreatic tumor growth. Oncoimmunology. 2019; 8(9): e1605822.

[143]

Boone BA, Murthy P, Miller-Ocuin J, et al. Chloroquine reduces hypercoagulability in pancreatic cancer through inhibition of neutrophil extracellular traps. BMC Cancer. 2018; 18(1): 678.

[144]

Zhang H, Xu X, Xu R, et al. Drug repurposing of ivermectin abrogates neutrophil extracellular traps and prevents melanoma metastasis. Front Oncol. 2022; 12: 989167.

[145]

Liu K, Sun E, Lei M, et al. BCG-induced formation of neutrophil extracellular traps play an important role in bladder cancer treatment. Clin Immunol. 2019; 201: 4-14.

[146]

de Buhr N, von Köckritz-Blickwede M. Detection, visualization, and quantification of neutrophil extracellular traps (NETs) and NET markers. Methods Mol Biol. 2020; 2087: 425-442.

[147]

Lv D, Xu Y, Cheng H, et al. A novel cell-based assay for dynamically detecting neutrophil extracellular traps-induced lung epithelial injuries. Exp Cell Res. 2020; 394(2): 112101.

[148]

Barbu EA, Dominical VM, Mendelsohn L, et al. Detection and quantification of histone H4 citrullination in early NETosis with image flow cytometry version 4. Front Immunol. 2020; 11: 1335.

[149]

Barbu EA, Dominical VM, Mendelsohn L, et al. An imaging flow cytometry method to measure citrullination of H4 histone as a read-out for neutrophil extracellular traps formation. Bio Protoc. 2021; 11(4): e3927.

[150]

Gupta S, Chan DW, Zaal KJ, et al. A high-throughput real-time imaging technique to quantify NETosis and distinguish mechanisms of cell death in human neutrophils. J Immunol. 2018; 200(2): 869-879.

[151]

Zhao W, Fogg DK, Kaplan MJ. A novel image-based quantitative method for the characterization of NETosis. J Immunol Methods. 2015; 423: 104-110.

[152]

Radermecker C, Hego A, Delvenne P, et al. Identification and quantitation of neutrophil extracellular traps in human tissue sections. Bio Protoc. 2021; 11(18): e4159.

[153]

Manda-Handzlik A, Fiok K, Cieloch A, et al. Convolutional neural networks-based image analysis for the detection and quantification of neutrophil extracellular traps. Cells. 2020; 9(2): 508.

[154]

Brinkmann V, Goosmann C, Kühn LI, et al. Automatic quantification of in vitro NET formation. Front Immunol. 2012; 3: 413.

[155]

Singhal A, Yadav S, Chandra T, et al. An imaging and computational algorithm for efficient identification and quantification of neutrophil extracellular traps. Cells. 2022; 11(2): 191.

[156]

de Andrea CE, Ochoa MC, Villalba-Esparza M, et al. Heterogenous presence of neutrophil extracellular traps in human solid tumours is partially dependent on IL-8. J Pathol. 2021; 255(2): 190-201.

[157]

Cheng P, He S, Zhang C, et al. A tandem-locked fluorescent NETosis reporter for the prognosis assessment of cancer immunotherapy. Angew Chem Int Ed Engl. 2023; 62(26): e202301625.

[158]

Gavillet M, Martinod K, Renella R, et al. Flow cytometric assay for direct quantification of neutrophil extracellular traps in blood samples. Am J Hematol. 2015; 90(12): 1155-1158.

[159]

Coelho LP, Pato C, Friães A, et al. Automatic determination of NET (neutrophil extracellular traps) coverage in fluorescent microscopy images. Bioinformatics. 2015; 31(14): 2364-2370.

[160]

Cho Y, Bukong TN, Tornai D, et al. Neutrophil extracellular traps contribute to liver damage and increase defective low-density neutrophils in alcohol-associated hepatitis. J Hepatol. 2023; 78(1): 28-44.

[161]

Zhang Y, Hu Y, Ma C, et al. Diagnostic, therapeutic predictive, and prognostic value of neutrophil extracellular traps in patients with gastric adenocarcinoma. Front Oncol. 2020; 10: 1036.

[162]

Zhang G, Zhang K. Screening and identification of neutrophil extracellular trap-related diagnostic biomarkers for pediatric sepsis by machine learning. Inflammation. 2025; 48(1): 212-222.

[163]

Konieva A, Deineka V, Diedkova K, et al. MXene-polydopamine-antiCEACAM1 antibody complex as a strategy for targeted ablation of melanoma. ACS Appl Mater Interfaces. 2024; 16(33): 43302-43316.

[164]

Rayes RF, Vourtzoumis P, Bou Rjeily M, et al. Neutrophil extracellular trap-associated CEACAM1 as a putative therapeutic target to prevent metastatic progression of colon carcinoma. J Immunol. 2020; 204(8): 2285-2294.

[165]

Igami K, Uchiumi T, Shiota M, et al. Extracellular vesicles expressing CEACAM proteins in the urine of bladder cancer patients. Cancer Sci. 2022; 113(9): 3120-3133.

[166]

Zhang B, Liu R, Huang H, et al. Identifying CEACAM1 as a potential prognostic biomarker for basal-like breast cancer by bioinformatics analysis and in vitro experiments. J Cancer. 2024; 15(19): 6468-6478.

[167]

Ronchetti L, Terrenato I, Ferretti M, et al. Circulating cell free DNA and citrullinated histone H3 as useful biomarkers of NETosis in endometrial cancer. J Exp Clin Cancer Res. 2022; 41(1): 151.

[168]

Heitzer E, Auinger L, Speicher MR. Cell-free DNA and apoptosis: how dead cells inform about the living. Trends Mol Med. 2020; 26(5): 519-528.

[169]

Pollak U, Zemmour H, Shaked E, et al. Novel cfDNA methylation biomarkers reveal delayed cardiac cell death after open-heart surgery. J Cardiovasc Transl Res. 2023; 16(1): 199-208.

[170]

Yokokawa T, Misaka T, Kimishima Y, et al. Clinical significance of circulating cardiomyocyte-specific cell-free DNA in patients with heart failure: a proof-of-concept study. Can J Cardiol. 2020; 36(6): 931-935.

[171]

Zhang Y, Guo L, Dai Q, et al. A signature for pan-cancer prognosis based on neutrophil extracellular traps. J Immunother Cancer. 2022; 10(6): e004210.

[172]

Schoeps B, Eckfeld C, Prokopchuk O, et al. TIMP1 triggers neutrophil extracellular trap formation in pancreatic cancer. Cancer Res. 2021; 81(13): 3568-3579.

[173]

Xu SS, Li H, Li TJ, et al. Neutrophil extracellular traps and macrophage extracellular traps predict postoperative recurrence in resectable nonfunctional pancreatic neuroendocrine tumors. Front Immunol. 2021; 12: 577517.

[174]

Zhang H, Lv H, Weng M, et al. Preoperative leukocytosis is associated with increased tumor-infiltrating neutrophil extracellular traps and worse outcomes in esophageal cancer. Ann Transl Med. 2020; 8(7): 441.

[175]

Shinde-Jadhav S, Mansure JJ, Rayes RF, et al. Role of neutrophil extracellular traps in radiation resistance of invasive bladder cancer. Nat Commun. 2021; 12(1): 2776.

[176]

Millrud CR, Kågedal Å, Kumlien Georén S, et al. NET-producing CD16(high) CD62L(dim) neutrophils migrate to tumor sites and predict improved survival in patients with HNSCC. Int J Cancer. 2017; 140(11): 2557-2567.

[177]

Rosell A, Gautam G, Wannberg F, et al. Neutrophil extracellular trap formation is an independent risk factor for occult cancer in patients presenting with venous thromboembolism. J Thromb Haemost. 2023; 21(11): 3166-3174.

[178]

Oto J, Plana E, Solmoirago MJ, et al. microRNAs and markers of neutrophil activation as predictors of early incidental post-surgical pulmonary embolism in patients with intracranial tumors. Cancers (Basel). 2020; 12(6): 1536.

[179]

Oto J, Navarro S, Larsen AC, et al. MicroRNAs and neutrophil activation markers predict venous thrombosis in pancreatic ductal adenocarcinoma and distal extrahepatic cholangiocarcinoma. Int J Mol Sci. 2020; 21(3): 840.

[180]

Armstrong AJ, Geva R, Chung HC, et al. CXCR2 antagonist navarixin in combination with pembrolizumab in select advanced solid tumors: a phase 2 randomized trial. Invest New Drugs. 2024; 42(1): 145-159.

[181]

Teijeira Á, Garasa S, Gato M, et al. CXCR1 and CXCR2 chemokine receptor agonists produced by tumors induce neutrophil extracellular traps that interfere with immune cytotoxicity. Immunity. 2020; 52(5): 856-871.e858.

[182]

Goldstein LJ, Perez RP, Yardley D, et al. A window-of-opportunity trial of the CXCR1/2 inhibitor reparixin in operable HER-2-negative breast cancer. Breast Cancer Res. 2020; 22(1): 4.

[183]

Guo C, Sharp A, Gurel B, et al. Targeting myeloid chemotaxis to reverse prostate cancer therapy resistance. Nature. 2023; 623(7989): 1053-1061.

[184]

Armstrong CWD, Coulter JA, Ong CW, et al. Clinical and functional characterization of CXCR1/CXCR2 biology in the relapse and radiotherapy resistance of primary PTEN-deficient prostate carcinoma. NAR Cancer. 2020; 2(3): zcaa012.

[185]

Yang J, Bergdorf K, Yan C, et al. CXCR2 expression during melanoma tumorigenesis controls transcriptional programs that facilitate tumor growth. Mol Cancer. 2023; 22(1): 92.

[186]

Alsabani M, Abrams ST, Cheng Z, et al. Reduction of NETosis by targeting CXCR1/2 reduces thrombosis, lung injury, and mortality in experimental human and murine sepsis. Br J Anaesth. 2022; 128(2): 283-293.

[187]

Steele CW, Karim SA, Leach JDG, et al. CXCR2 inhibition profoundly suppresses metastases and augments immunotherapy in pancreatic ductal adenocarcinoma. Cancer Cell. 2016; 29(6): 832-845.

[188]

Chen H, Wei L, Luo M, et al. PAD4 inhibitor promotes DNA damage and radiosensitivity of nasopharyngeal carcinoma cells. Environ Toxicol. 2021; 36(11): 2291-2301.

[189]

Li M, Lin C, Deng H, et al. A novel peptidylarginine deiminase 4 (PAD4) inhibitor BMS-P5 blocks formation of neutrophil extracellular traps and delays progression of multiple myeloma. Mol Cancer Ther. 2020; 19(7): 1530-1538.

[190]

Lee W, Ko SY, Mohamed MS, et al. Neutrophils facilitate ovarian cancer premetastatic niche formation in the omentum. J Exp Med. 2019; 216(1): 176-194.

[191]

Jiang ZZ, Peng ZP, Liu XC, et al. Neutrophil extracellular traps induce tumor metastasis through dual effects on cancer and endothelial cells. Oncoimmunology. 2022; 11(1): 2052418.

[192]

Ahmed D, Puthussery H, Basnett P, et al. Controlled delivery of Pan-PAD-inhibitor Cl-amidine using poly(3-hydroxybutyrate) microspheres. Int J Mol Sci. 2021; 22(23): 12852.

[193]

Uysal-Onganer P, D'Alessio S, Mortoglou M, et al. Peptidylarginine deiminase inhibitor application, using Cl-amidine, PAD2, PAD3 and PAD4 isozyme-specific inhibitors in pancreatic cancer cells, reveals roles for PAD2 and PAD3 in cancer invasion and modulation of extracellular vesicle signatures. Int J Mol Sci. 2021; 22(3): 1396.

[194]

Zhu D, Lu Y, Gui L, et al. Self-assembling, pH-responsive nanoflowers for inhibiting PAD4 and neutrophil extracellular trap formation and improving the tumor immune microenvironment. Acta Pharm Sin B. 2022; 12(5): 2592-2608.

[195]

Mou Z, Chen Y, Hu J, et al. Icaritin inhibits the progression of urothelial cancer by suppressing PADI2-mediated neutrophil infiltration and neutrophil extracellular trap formation. Acta Pharm Sin B. 2024; 14(9): 3916-3930.

[196]

Ivey AD, Matthew Fagan B, Murthy P, et al. Chloroquine reduces neutrophil extracellular trap (NET) formation through inhibition of peptidyl arginine deiminase 4 (PAD4). Clin Exp Immunol. 2023; 211(3): 239-247.

[197]

Zeng J, Xu H, Fan PZ, et al. Kaempferol blocks neutrophil extracellular traps formation and reduces tumour metastasis by inhibiting ROS-PAD4 pathway. J Cell Mol Med. 2020; 24(13): 7590-7599.

[198]

Zhu W, Yang S, Meng D, et al. Targeting NADPH oxidase and integrin α5β1 to inhibit neutrophil extracellular traps-mediated metastasis in colorectal cancer. Int J Mol Sci. 2023; 24(21): 16001.

[199]

Ali RA, Gandhi AA, Dai L, et al. Antineutrophil properties of natural gingerols in models of lupus. JCI Insight. 2021; 6(3): e138385.

[200]

Ishikawa M, Yamashita H, Oka N, et al. Antithrombin III improved neutrophil extracellular traps in lung after the onset of endotoxemia. J Surg Res. 2017; 208: 140-150.

[201]

Xia X, Zhang Z, Zhu C, et al. Neutrophil extracellular traps promote metastasis in gastric cancer patients with postoperative abdominal infectious complications. Nat Commun. 2022; 13(1): 1017.

[202]

Peng H, Shen J, Long X, et al. Local release of TGF-β inhibitor modulates tumor-associated neutrophils and enhances pancreatic cancer response to combined irreversible electroporation and immunotherapy. Adv Sci (Weinh). 2022; 9(10): e2105240.

[203]

Subhan MA, Torchilin VP. Neutrophils as an emerging therapeutic target and tool for cancer therapy. Life Sci. 2021; 285: 119952.

[204]

Akbay EA, Koyama S, Liu Y, 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(8): 1268-1279.

[205]

Efrimescu CI, Buggy PM, Buggy DJ. Neutrophil extracellular trapping role in cancer, metastases, and cancer-related thrombosis: a narrative review of the current evidence base. Curr Oncol Rep. 2021; 23(10): 118.

[206]

Mutua V, Gershwin LJ. A review of neutrophil extracellular traps (NETs) in disease: potential anti-NETs therapeutics. Clin Rev Allergy Immunol. 2021; 61(2): 194-211.

[207]

Sun Y, He J, Chen W, et al. Inhalable DNase I@Au hybrid nanoparticles for radiation sensitization and metastasis inhibition by elimination of neutrophil extracellular traps. Biomaterials. 2025; 317: 123095.

[208]

Yin H, Lu H, Xiong Y, et al. Tumor-associated neutrophil extracellular traps regulating nanocarrier-enhanced inhibition of malignant tumor growth and distant metastasis. ACS Appl Mater Interfaces. 2021; 13(50): 59683-59694.

[209]

Chen J, Hou S, Liang Q, et al. Localized degradation of neutrophil extracellular traps by photoregulated enzyme delivery for cancer immunotherapy and metastasis suppression. ACS Nano. 2022; 16(2): 2585-2597.

[210]

Khan U, Chowdhury S, Billah MM, et al. Neutrophil extracellular traps in colorectal cancer progression and metastasis. Int J Mol Sci. 2021; 22(14): 7260.

[211]

Aikawa N, Ishizaka A, Hirasawa H, et al. Reevaluation of the efficacy and safety of the neutrophil elastase inhibitor, Sivelestat, for the treatment of acute lung injury associated with systemic inflammatory response syndrome; a phase IV study. Pulm Pharmacol Ther. 2011; 24(5): 549-554.

[212]

Okamoto M, Mizuno R, Kawada K, 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(2): 1118.

[213]

Caruso JA, Akli S, Pageon L, et al. The serine protease inhibitor elafin maintains normal growth control by opposing the mitogenic effects of neutrophil elastase. Oncogene. 2015; 34(27): 3556-3567.

[214]

Zhang H, Wang Y, Onuma A, et al. Neutrophils extracellular traps inhibition improves PD-1 blockade immunotherapy in colorectal cancer. Cancers (Basel). 2021; 13(21): 5333.

[215]

Xie Y, Zhou T, Li X, et al. Targeting ESE3/EHF with nifurtimox inhibits CXCR2(+) neutrophil infiltration and overcomes pancreatic cancer resistance to chemotherapy and immunotherapy. Gastroenterology. 2024; 167(2): 281-297.

[216]

Chen H, Luo M, Wang X, et al. Inhibition of PAD4 enhances radiosensitivity and inhibits aggressive phenotypes of nasopharyngeal carcinoma cells. Cell Mol Biol Lett. 2021; 26(1): 9.

[217]

Hawez A, Taha D, Algaber A, et al. MiR-155 regulates neutrophil extracellular trap formation and lung injury in abdominal sepsis. J Leukoc Biol. 2022; 111(2): 391-400.

[218]

Lewis HD, Liddle J, Coote JE, et al. Inhibition of PAD4 activity is sufficient to disrupt mouse and human NET formation. Nat Chem Biol. 2015; 11(3): 189-191.

[219]

Manfredi AA, Rovere-Querini P, D'Angelo A, et al. Low molecular weight heparins prevent the induction of autophagy of activated neutrophils and the formation of neutrophil extracellular traps. Pharmacol Res. 2017; 123: 146-156.

[220]

Jung HS, Gu J, Kim JE, et al. Cancer cell-induced neutrophil extracellular traps promote both hypercoagulability and cancer progression. PLoS One. 2019; 14(4): e0216055.

[221]

Zhu Y, Wang D, Luo J, et al. Zingerone inhibits the neutrophil extracellular trap formation and protects against sepsis via Nrf2-mediated ROS inhibition. Oxid Med Cell Longev. 2022; 2022: 3990607.

[222]

Haute GV, Luft C, Pedrazza L, et al. Octyl gallate decrease lymphocyte activation and regulates neutrophil extracellular traps release. Mol Biol Rep. 2022; 49(2): 1593-1599.

[223]

Liu Y, Qin X, Lei Z, et al. Tetramethylpyrazine inhibits neutrophil extracellular traps formation and alleviates hepatic ischemia/reperfusion injury in rat liver transplantation. Exp Cell Res. 2021; 406(1): 112719.

[224]

Chen KW, Demarco B, Broz P. Beyond inflammasomes: emerging function of gasdermins during apoptosis and NETosis. Embo J. 2020; 39(2): e103397.

[225]

Chen KW, Monteleone M, Boucher D, et al. Noncanonical inflammasome signaling elicits gasdermin D-dependent neutrophil extracellular traps. Sci Immunol. 2018; 3(26): eaar6676.

[226]

Nie D, Chen C, Li Y, et al. Disulfiram, an aldehyde dehydrogenase inhibitor, works as a potent drug against sepsis and cancer via NETosis, pyroptosis, apoptosis, ferroptosis, and cuproptosis. Blood Sci. 2022; 4(3): 152-154.

[227]

Cai W, Wu Z, Lai J, et al. LDC7559 inhibits microglial activation and GSDMD-dependent pyroptosis after subarachnoid hemorrhage. Front Immunol. 2023; 14: 1117310.

[228]

Lapponi MJ, Carestia A, Landoni VI, et al. Regulation of neutrophil extracellular trap formation by anti-inflammatory drugs. J Pharmacol Exp Ther. 2013; 345(3): 430-437.

[229]

Liu J, Zheng F, Yang M, et al. Effect of aspirin use on survival benefits of breast cancer patients: a meta-analysis. Medicine (Baltimore). 2021; 100(33): e26870.

[230]

Burkard P, Schonhart C, Vögtle T, et al. A key role for platelet GPVI in neutrophil recruitment, migration, and NETosis in the early stages of acute lung injury. Blood. 2023; 142(17): 1463-1477.

[231]

Xu C, Ye Z, Jiang W, et al. Cyclosporine A alleviates colitis by inhibiting the formation of neutrophil extracellular traps via the regulating pentose phosphate pathway. Mol Med. 2023; 29(1): 169.

[232]

Li S, Wang Y, Yu D, et al. Triclocarban evoked neutrophil extracellular trap formation in common carp (Cyprinus carpio L.) by modulating SIRT3-mediated ROS crosstalk with ERK1/2/p38 signaling. Fish Shellfish Immunol. 2022; 129: 85-95.

[233]

Papayannopoulos V, Metzler KD, Hakkim A, et al. Neutrophil elastase and myeloperoxidase regulate the formation of neutrophil extracellular traps. J Cell Biol. 2010; 191(3): 677-691.

[234]

Lulla AR, Akli S, Karakas C, et al. Neutrophil elastase remodels mammary tumors to facilitate lung metastasis. Mol Cancer Ther. 2024; 23(4): 492-506.

[235]

Du Y, Chen Y, Li F, et al. Genetically engineered cellular nanovesicle as targeted DNase I delivery system for the clearance of neutrophil extracellular traps in acute lung injury. Adv Sci (Weinh). 2023; 10(32): e2303053.

[236]

Wang Z, Chen C, Shi C, et al. Cell membrane derived liposomes loaded with DNase I target neutrophil extracellular traps which inhibits colorectal cancer liver metastases. J Control Release. 2023; 357: 620-629.

[237]

Hisada Y, Grover SP, Maqsood A, et al. Neutrophils and neutrophil extracellular traps enhance venous thrombosis in mice bearing human pancreatic tumors. Haematologica. 2020; 105(1): 218-225.

[238]

Chen Q, Zhang L, Li X, et al. Neutrophil extracellular traps in tumor metastasis: pathological functions and clinical applications. Cancers (Basel). 2021; 13(11): 2832.

[239]

Herranz R, Oto J, Hueso M, et al. Bladder cancer patients have increased NETosis and impaired DNaseI-mediated NET degradation that can be therapeutically restored in vitro. Front Immunol. 2023; 14: 1171065.

[240]

Hosseinnejad A, Ludwig N, Wienkamp AK, et al. DNase I functional microgels for neutrophil extracellular trap disruption. Biomater Sci. 2021; 10(1): 85-99.

[241]

Xia Y, He J, Zhang H, et al. AAV-mediated gene transfer of DNase I in the liver of mice with colorectal cancer reduces liver metastasis and restores local innate and adaptive immune response. Mol Oncol. 2020; 14(11): 2920-2935.

[242]

Lu Y, Peng Z, Zhu D, et al. RGD peptide and PAD4 inhibitor-loaded gold nanorods for chemo-photothermal combined therapy to inhibit tumor growth, prevent lung metastasis and improve biosafety. Int J Nanomedicine. 2021; 16: 5565-5580.

[243]

Antonelou M, Michaëlsson E, Evans RDR, et al. Therapeutic myeloperoxidase inhibition attenuates neutrophil activation, ANCA-mediated endothelial damage, and crescentic GN. J Am Soc Nephrol. 2020; 31(2): 350-364.

[244]

Liu TW, Gammon ST, Yang P, et al. Inhibition of myeloperoxidase enhances immune checkpoint therapy for melanoma. J Immunother Cancer. 2023; 11(2): e005837.

[245]

Regard JB, Harrison TJ, Axford J, et al. Discovery of a novel, highly potent and orally bioavailable pyrrolidinone indole series of irreversible myeloperoxidase (MPO) inhibitors. Biochem Pharmacol. 2023; 209: 115418.

[246]

Williams CJM, Peddle AM, Kasi PM, et al. Neoadjuvant immunotherapy for dMMR and pMMR colorectal cancers: therapeutic strategies and putative biomarkers of response. Nat Rev Clin Oncol. 2024; 21(12): 839-851.

[247]

van Bijsterveldt L, Durley SC, Maughan TS, et al. The challenge of combining chemo- and radiotherapy with checkpoint kinase inhibitors. Clin Cancer Res. 2021; 27(4): 937-962.

[248]

Kroeze SGC, Pavic M, Stellamans K, et al. Metastases-directed stereotactic body radiotherapy in combination with targeted therapy or immunotherapy: systematic review and consensus recommendations by the EORTC-ESTRO OligoCare consortium. Lancet Oncol. 2023; 24(3): e121-e132.

[249]

Jiang Y, Wang C, Zhou S. Artificial intelligence-based risk stratification, accurate diagnosis and treatment prediction in gynecologic oncology. Semin Cancer Biol. 2023; 96: 82-99.

[250]

Esfahani K, Elkrief A, Calabrese C, et al. Moving towards personalized treatments of immune-related adverse events. Nat Rev Clin Oncol. 2020; 17(8): 504-515.

[251]

Corbeau A, Kuipers SC, de Boer SM, et al. Correlations between bone marrow radiation dose and hematologic toxicity in locally advanced cervical cancer patients receiving chemoradiation with cisplatin: a systematic review. Radiother Oncol. 2021; 164: 128-137.

[252]

Shen XT, Xie SZ, Xu J, et al. Pan-cancer analysis reveals a distinct neutrophil extracellular trap-associated regulatory pattern. Front Immunol. 2022; 13: 798022.

RIGHTS & PERMISSIONS

2025 The Author(s). Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.

AI Summary AI Mindmap
PDF

8

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/