303
Views
0
CrossRef citations to date
0
Altmetric
ORIGINAL RESEARCH

A Novel Neutrophil Extracellular Traps Signature for Overall Survival Prediction and Tumor Microenvironment Identification in Gastric Cancer

, , , , , ORCID Icon, , , , , & show all
Pages 3419-3436 | Received 28 Apr 2023, Accepted 04 Jul 2023, Published online: 14 Aug 2023

References

  • Smyth EC, Nilsson M, Grabsch HI, van Grieken NC, Lordick F. Gastric cancer. Lancet. 2020;396(10251):635–648.
  • Bang YJ, Van Cutsem E, Feyereislova A, et al. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a Phase 3, open-label, randomised controlled trial. Lancet. 2010;376(9742):687–697. doi:10.1016/S0140-6736(10)61121-X
  • Janjigian YY, Shitara K, Moehler M, et al. First-line nivolumab plus chemotherapy versus chemotherapy alone for advanced gastric, gastro-oesophageal junction, and oesophageal adenocarcinoma (CheckMate 649): a randomised, open-label, phase 3 trial. Lancet. 2021;398(10294):27–40. doi:10.1016/S0140-6736(21)00797-2
  • Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–249. doi:10.3322/caac.21660
  • Binnewies M, Roberts EW, Kersten K, et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med. 2018;24(5):541–550. doi:10.1038/s41591-018-0014-x
  • Joyce JA, Fearon DT. T cell exclusion, immune privilege, and the tumor microenvironment. Science. 2015;348(6230):74–80. doi:10.1126/science.aaa6204
  • Gentles AJ, Newman AM, Liu CL, et al. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat Med. 2015;21(8):938–945. doi:10.1038/nm.3909
  • Qu Z, Wang Q, Wang H, et al. The effect of inflammatory markers on the survival of advanced gastric cancer patients who underwent anti-programmed death 1 therapy. Front Oncol. 2022;12:783197. doi:10.3389/fonc.2022.783197
  • Brinkmann V, Reichard U, Goosmann C, et al. Neutrophil extracellular traps kill bacteria. Science. 2004;303(5663):1532–1535. doi:10.1126/science.1092385
  • Papayannopoulos V. Neutrophil extracellular traps in immunity and disease. Nat Rev Immunol. 2018;18(2):134–147.
  • Park J, Wysocki RW, Amoozgar Z, et al. Cancer cells induce metastasis-supporting neutrophil extracellular DNA traps. Sci Transl Med. 2016;8(361):361ra138.
  • Berger-Achituv S, Brinkmann V, Abed UA, et al. A proposed role for neutrophil extracellular traps in cancer immunoediting. Front Immunol. 2013;4:48. doi:10.3389/fimmu.2013.00048
  • 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. doi:10.7150/jca.44215
  • 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.
  • Lee W, Ko SY, Mohamed MS, Kenny HA, Lengyel E, Naora H. Neutrophils facilitate ovarian cancer premetastatic niche formation in the omentum. J Exp Med. 2019;216(1):176–194. doi:10.1084/jem.20181170
  • Kessenbrock K, Krumbholz M, Schönermarck U, et al. Netting neutrophils in autoimmune small-vessel vasculitis. Nat Med. 2009;15(6):623–625. doi:10.1038/nm.1959
  • Grilz E, Mauracher LM, Posch F, et al. Citrullinated histone H3, a biomarker for neutrophil extracellular trap formation, predicts the risk of mortality in patients with cancer. Br J Haematol. 2019;186(2):311–320. doi:10.1111/bjh.15906
  • Rosell A, Aguilera K, Hisada Y, et al. Prognostic value of circulating markers of neutrophil activation, neutrophil extracellular traps, coagulation and fibrinolysis in patients with terminal cancer. Sci Rep. 2021;11(1):5074. doi:10.1038/s41598-021-84476-3
  • Li Q, Chen W, Li Q, Mao J, Chen X. A novel neutrophil extracellular trap signature to predict prognosis and immunotherapy response in head and neck squamous cell carcinoma. Front Immunol. 2022;13:1019967. doi:10.3389/fimmu.2022.1019967
  • 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. doi:10.3389/fonc.2020.01036
  • 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). doi:10.1136/jitc-2021-004210
  • Shen XT, Xie SZ, Xu J, Yang LY, Qin LX. Pan-cancer analysis reveals a distinct neutrophil extracellular trap-associated regulatory pattern. Front Immunol. 2022;13:798022. doi:10.3389/fimmu.2022.798022
  • Subramanian A, Tamayo P, Mootha VK, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005;102(43):15545–15550. doi:10.1073/pnas.0506580102
  • Cristinziano L, Modestino L, Antonelli A, et al. Neutrophil extracellular traps in cancer. Semin Cancer Biol. 2022;79:91–104. doi:10.1016/j.semcancer.2021.07.011
  • Yang D, Liu J. Neutrophil extracellular traps: a new player in cancer metastasis and therapeutic target. J Exp Clin Cancer Res. 2021;40(1):233. doi:10.1186/s13046-021-02013-6
  • Wang H, Zhang Y, Wang Q, Wei X, Wang H, Gu K. The regulatory mechanism of neutrophil extracellular traps in cancer biological behavior. Cell Biosci. 2021;11(1):193. doi:10.1186/s13578-021-00708-z
  • Fang C, Liu F, Wang Y, et al. A innovative prognostic symbol based on neutrophil extracellular traps (NETs)-related lncRNA signature in non-small-cell lung cancer. Aging. 2021;13(13):17864–17879. doi:10.18632/aging.203289
  • Chen N, He D, Cui J. A neutrophil extracellular traps signature predicts the clinical outcomes and immunotherapy response in head and neck squamous cell carcinoma. Front Mol Biosci. 2022;9:833771. doi:10.3389/fmolb.2022.833771
  • Albrengues J, Shields MA, Ng D, et al. Neutrophil extracellular traps produced during inflammation awaken dormant cancer cells in mice. Science. 2018;361(6409). doi:10.1126/science.aao4227
  • 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. doi:10.1038/s41467-021-20982-2
  • Mittal V. Epithelial mesenchymal transition in tumor metastasis. Annu Rev Pathol. 2018;13:395–412. doi:10.1146/annurev-pathol-020117-043854
  • Zhu T, Zou X, Yang C, et al. Neutrophil extracellular traps promote gastric cancer metastasis by inducing epithelial‑mesenchymal transition. Int J Mol Med. 2021;48(1). doi:10.3892/ijmm.2021.4960
  • Cools-Lartigue J, Spicer J, McDonald B, et al. Neutrophil extracellular traps sequester circulating tumor cells and promote metastasis. J Clin Invest. 2013;123(8):3446–3458. doi:10.1172/JCI67484
  • Cassetta L, Pollard JW. Targeting macrophages: therapeutic approaches in cancer. Nat Rev Drug Discov. 2018;17(12):887–904. doi:10.1038/nrd.2018.169
  • 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. doi:10.1016/j.immuni.2020.03.001
  • Kaltenmeier C, Yazdani HO, Morder K, Geller DA, Simmons RL, Tohme S. Neutrophil extracellular traps promote T cell exhaustion in the tumor microenvironment. Front Immunol. 2021;12:785222. doi:10.3389/fimmu.2021.785222
  • 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. doi:10.1038/nm.2084
  • Papayannopoulos V, Metzler KD, Hakkim A, Zychlinsky A. Neutrophil elastase and myeloperoxidase regulate the formation of neutrophil extracellular traps. J Cell Biol. 2010;191(3):677–691. doi:10.1083/jcb.201006052
  • Yazdani HO, Roy E, Comerci AJ, et al. Neutrophil extracellular traps drive mitochondrial homeostasis in tumors to augment growth. Cancer Res. 2019;79(21):5626–5639. doi:10.1158/0008-5472.CAN-19-0800
  • Yoo DG, Floyd M, Winn M, Moskowitz SM, Rada B. NET formation induced by Pseudomonas aeruginosa cystic fibrosis isolates measured as release of myeloperoxidase-DNA and neutrophil elastase-DNA complexes. Immunol Lett. 2014;160(2):186–194. doi:10.1016/j.imlet.2014.03.003
  • Kaltenmeier CT, Yazdani H, van der Windt D, et al. Neutrophil extracellular traps as a novel biomarker to predict recurrence-free and overall survival in patients with primary hepatic malignancies. HPB (Oxford). 2021;23(2):309–320. doi:10.1016/j.hpb.2020.06.012
  • Moik F, Zöchbauer-Müller S, Posch F, Pabinger I, Ay C. Systemic inflammation and activation of haemostasis predict poor prognosis and response to chemotherapy in patients with advanced lung cancer. Cancers. 2020;12(6):1619. doi:10.3390/cancers12061619