The Multifaceted Role of Neutrophils in NSCLC in the Era of Immune Checkpoint Inhibitors
Abstract
Simple Summary
Abstract
1. Introduction
2. TAN Heterogeneity in Tumors and Correlation with ICI Response
Anti-Tumor Neutrophils (N1) | Pro-Tumor Neutrophils (N2) | PMN-MDSCs/g-MDSCs | |
---|---|---|---|
Surface markers in mice | CD11b+, Ly6G+, CD170low, CD177+, CD54+, CD16+, CD32+, CD89+ [14,20,23,28] | CD11b+, Ly6G+, Ly6Clow, CD170high, PDL1+, S100A8/A9+, CXCR2+, CXCR4−, Ym1+ [14,21,22,23,39,40] | CD11b+/low, Ly6G+, GR1+, Ly6Clow, S100A8/A9+, CXCR4+, CD84+, CD115+, CD244+ [28,29,34,35,36] |
Surface markers in human | CD66b+, CD11b+, CD170low, CD66b+, CD86+, CD54+, CD15high [14,30] | CD66b+, CD11b+, PDL1+, CD170high, CD14−, CD15+, CXCR2+ [14,27,30] | CD11b+, CD14−, CD15+, CD66b+, LOX+, CD33+, CD84+, CD124+ [27,29,30,35,36,41] |
Polarization factors | LPS, IFNγ [20,24,42,43] | TGFβ, IFNβ, IL-4 [23,43,44,45] | GCN2 [46] |
Factors released | ROS, NO, MPO, H2O2 [14,17,20,47] | ROS, NO, ARG1, PGE2, MMP9, EGF, HGF, PDGF, NE, S100A8/A9 [14,20,45,48] | ROS, NO, ARG1, PGE2 [35,48] |
Morphology | Mature, poly-nuclear, hyper-segmented, high density [23,27] | Mature, poly-nuclear, segmented nuclear, low density [23,27] | Mature or immature, ring-shaped nuclear, low density [41] |
Related cancer type | Non-Hodgkin lymphoma, hepatocellular carcinoma, Lewis lung carcinoma, colon adenocarcinoma, renal cell carcinoma, breast cancer | NSCLC, hepatocellular carcinoma, melanoma, colon rectal cancer | Pancreatic ductal adenocarcinoma, NSCLC, breast cancer, renal cell carcinoma, head and neck squamous cell carcinoma, glioblastoma, melanoma |
3. TAN Interaction with Malignant Cells
3.1. ROS
3.2. Growth Factors
3.3. EMT Proteins
3.4. Neutrophil Extracellular Trap (NET)
3.5. Chemokine Receptor
3.6. Metabolites
3.7. Non-Coding RNAs
4. TAN Interactions with T-Cells
4.1. Neutrophil–Lymphocyte Ratio
4.2. Direct Interaction with T-Cells
4.3. Indirect Interaction with T-Cells
5. TAN Interaction with Other Immune Cells
6. Targeting Pro-Tumor Neutrophils and Clinical Trials That Combined with ICIs
Drug | Target | Disease | Status | NCT | Combination |
---|---|---|---|---|---|
Galunisertib | TGFβ | NSCLC | Phase II | NCT02423343 | Nivolumab [120] |
Pegfilgrastim | G-CSF | NSCLC | Phase I | NCT01840579 | Pembrolizumab and chemo |
Cabiralizumab | CSF1R | NSCLC | Phase I | NCT03502330 | Nivolumab |
Sidenafil | PDE5 | NSCLC | Phase III | NCT00752115 | Carboplatin, paclitaxel |
Tadalafil | PDE5 | NSCLC | Terminated at phase II | NCT04069936 | Nivolumab |
Celecoxib | COX2 | NSCLC | Phase II | NCT00030407 | Docetaxel |
BMS-813160 | CXCR2/5 | NSCLC | Phase II | NCT04123379 | Nivolumab |
Navarixin | CXCR2 | NSCLC | Phase II | NCT03473925 | Pembrolizumab |
Reparixin | CXCR2 | Metastatic breast cancer | Phase I | NCT02370238 | paclitaxel |
Telaglenastat | glutaminase | NSCLC | Terminated at phase II | NCT04265534 | Pembrolizumab and chemo |
Tocilizumab | IL6 | NSCLC | Phase II | NCT04691817 | Atezolizumab |
DV281 | TLR9 | NSCLC | Phase I | NCT03326752 | Nivolumab |
Anlotinib | VEGFR, PDGFR | NSCLC | Phase II | NCT05001971 | Penpulimab [121] |
Marimastat | MMP | NSCLC | Phase III | NCT00002911 | NA |
Numidargistat | ARG1 | Metastatic/solid tumor | Phase II | NCT02903914 | Pembrolizumab |
GSK484 | PAD4, NETosis | Lung carcinoma | Preclinical [122] | NA | Anti-PD1, anti-CTLA4 |
Tasquinimod | S100A9 | Prostate cancer | Approved [119] | NCT01234311 | NA |
1A8 | Ly6G | Glioma | Preclinical [95] | NA | Anti-PD1 |
Bindarit | CCL2 | Diabetic nephropathy | Phase II | NCT01109212 | NA |
Sivelestat | NE | Acute lung injury in esophageal cancer | Approved in Japan and Korea [123] | NCT01170845 | NA |
Bardoxolone methyl | ROS | Advanced solid tumor | Phase I | NCT00529438 | NA |
7. Conclusions
Author Contributions
Funding
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Alexander, M.; Kim, S.Y.; Cheng, H. Update 2020: Management of Non-Small Cell Lung Cancer. Lung 2020, 198, 897–907. [Google Scholar] [CrossRef] [PubMed]
- Jain, A.S.; Prasad, A.; Pradeep, S.; Dharmashekar, C.; Achar, R.R.; Ekaterina, S.; Victor, S.; Amachawadi, R.G.; Prasad, S.K.; Pruthvish, R.; et al. Everything Old Is New Again: Drug Repurposing Approach for Non-Small Cell Lung Cancer Targeting MAPK Signaling Pathway. Front. Oncol. 2021, 11, 741326. [Google Scholar] [CrossRef] [PubMed]
- Herbst, R.S.; Morgensztern, D.; Boshoff, C. The biology and management of non-small cell lung cancer. Nature 2018, 553, 446–454. [Google Scholar] [CrossRef]
- Brahmer, J.; Reckamp, K.L.; Baas, P.; Crino, L.; Eberhardt, W.E.; Poddubskaya, E.; Antonia, S.; Pluzanski, A.; Vokes, E.E.; Holgado, E.; et al. Nivolumab versus Docetaxel in Advanced Squamous-Cell Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2015, 373, 123–135. [Google Scholar] [CrossRef] [PubMed]
- Schoenfeld, A.J.; Hellmann, M.D. Acquired Resistance to Immune Checkpoint Inhibitors. Cancer Cell 2020, 37, 443–455. [Google Scholar] [CrossRef]
- Darvin, P.; Toor, S.M.; Sasidharan Nair, V.; Elkord, E. Immune checkpoint inhibitors: Recent progress and potential biomarkers. Exp. Mol. Med. 2018, 50, 1–11. [Google Scholar] [CrossRef] [PubMed]
- de Visser, K.E.; Eichten, A.; Coussens, L.M. Paradoxical roles of the immune system during cancer development. Nat. Rev. Cancer 2006, 6, 24–37. [Google Scholar] [CrossRef] [PubMed]
- Graves, E.E.; Maity, A.; Le, Q.T. The tumor microenvironment in non-small-cell lung cancer. Semin. Radiat. Oncol. 2010, 20, 156–163. [Google Scholar] [CrossRef] [PubMed]
- Yamauchi, M.; Gibbons, D.L.; Zong, C.; Fradette, J.J.; Bota-Rabassedas, N.; Kurie, J.M. Fibroblast heterogeneity and its impact on extracellular matrix and immune landscape remodeling in cancer. Matrix Biol. 2020, 91–92, 8–18. [Google Scholar] [CrossRef]
- Peng, D.H.; Rodriguez, B.L.; Diao, L.; Chen, L.; Wang, J.; Byers, L.A.; Wei, Y.; Chapman, H.A.; Yamauchi, M.; Behrens, C.; et al. Collagen promotes anti-PD-1/PD-L1 resistance in cancer through LAIR1-dependent CD8+ T cell exhaustion. Nat. Commun. 2020, 11, 4520. [Google Scholar] [CrossRef]
- Kargl, J.; Busch, S.E.; Yang, G.H.; Kim, K.H.; Hanke, M.L.; Metz, H.E.; Hubbard, J.J.; Lee, S.M.; Madtes, D.K.; McIntosh, M.W.; et al. Neutrophils dominate the immune cell composition in non-small cell lung cancer. Nat. Commun. 2017, 8, 14381. [Google Scholar] [CrossRef]
- Stankovic, B.; Bjorhovde, H.A.K.; Skarshaug, R.; Aamodt, H.; Frafjord, A.; Muller, E.; Hammarstrom, C.; Beraki, K.; Baekkevold, E.S.; Woldbaek, P.R.; et al. Immune Cell Composition in Human Non-small Cell Lung Cancer. Front. Immunol. 2018, 9, 3101. [Google Scholar] [CrossRef]
- Peng, L.; Wang, Y.; Liu, F.; Qiu, X.; Zhang, X.; Fang, C.; Qian, X.; Li, Y. Peripheral blood markers predictive of outcome and immune-related adverse events in advanced non-small cell lung cancer treated with PD-1 inhibitors. Cancer Immunol. Immunother. 2020, 69, 1813–1822. [Google Scholar] [CrossRef]
- Jaillon, S.; Ponzetta, A.; Di Mitri, D.; Santoni, A.; Bonecchi, R.; Mantovani, A. Neutrophil diversity and plasticity in tumour progression and therapy. Nat. Rev. Cancer 2020, 20, 485–503. [Google Scholar] [CrossRef]
- Diem, S.; Schmid, S.; Krapf, M.; Flatz, L.; Born, D.; Jochum, W.; Templeton, A.J.; Fruh, M. Neutrophil-to-Lymphocyte ratio (NLR) and Platelet-to-Lymphocyte ratio (PLR) as prognostic markers in patients with non-small cell lung cancer (NSCLC) treated with nivolumab. Lung Cancer 2017, 111, 176–181. [Google Scholar] [CrossRef] [PubMed]
- Mestas, J.; Hughes, C.C. Of mice and not men: Differences between mouse and human immunology. J. Immunol. 2004, 172, 2731–2738. [Google Scholar] [CrossRef] [PubMed]
- Hedrick, C.C.; Malanchi, I. Neutrophils in cancer: Heterogeneous and multifaceted. Nat. Rev. Immunol. 2022, 22, 173–187. [Google Scholar] [CrossRef] [PubMed]
- Aloe, C.; Wang, H.; Vlahos, R.; Irving, L.; Steinfort, D.; Bozinovski, S. Emerging and multifaceted role of neutrophils in lung cancer. Transl. Lung Cancer Res. 2021, 10, 2806–2818. [Google Scholar] [CrossRef]
- Shaul, M.E.; Fridlender, Z.G. Tumour-associated neutrophils in patients with cancer. Nat. Rev. Clin. Oncol. 2019, 16, 601–620. [Google Scholar] [CrossRef]
- Masucci, M.T.; Minopoli, M.; Carriero, M.V. Tumor Associated Neutrophils. Their Role in Tumorigenesis, Metastasis, Prognosis and Therapy. Front. Oncol. 2019, 9, 1146. [Google Scholar] [CrossRef]
- Quail, D.F.; Amulic, B.; Aziz, M.; Barnes, B.J.; Eruslanov, E.; Fridlender, Z.G.; Goodridge, H.S.; Granot, Z.; Hidalgo, A.; Huttenlocher, A.; et al. Neutrophil phenotypes and functions in cancer: A consensus statement. J. Exp. Med. 2022, 219, e20220011. [Google Scholar] [CrossRef] [PubMed]
- Faget, J.; Peters, S.; Quantin, X.; Meylan, E.; Bonnefoy, N. Neutrophils in the era of immune checkpoint blockade. J. Immunother. Cancer 2021, 9, e002242. [Google Scholar] [CrossRef] [PubMed]
- Fridlender, Z.G.; Sun, J.; Kim, S.; Kapoor, V.; Cheng, G.; Ling, L.; Worthen, G.S.; Albelda, S.M. Polarization of tumor-associated neutrophil phenotype by TGF-beta: “N1” versus “N2” TAN. Cancer Cell 2009, 16, 183–194. [Google Scholar] [CrossRef] [PubMed]
- Andzinski, L.; Kasnitz, N.; Stahnke, S.; Wu, C.F.; Gereke, M.; von Kockritz-Blickwede, M.; Schilling, B.; Brandau, S.; Weiss, S.; Jablonska, J. Type I IFNs induce anti-tumor polarization of tumor associated neutrophils in mice and human. Int. J. Cancer 2016, 138, 1982–1993. [Google Scholar] [CrossRef] [PubMed]
- Shaul, M.E.; Levy, L.; Sun, J.; Mishalian, I.; Singhal, S.; Kapoor, V.; Horng, W.; Fridlender, G.; Albelda, S.M.; Fridlender, Z.G. Tumor-associated neutrophils display a distinct N1 profile following TGFbeta modulation: A transcriptomics analysis of pro- vs. antitumor TANs. Oncoimmunology 2016, 5, e1232221. [Google Scholar] [CrossRef]
- Yang, Z.; Guo, J.; Weng, L.; Tang, W.; Jin, S.; Ma, W. Myeloid-derived suppressor cells-new and exciting players in lung cancer. J. Hematol. Oncol. 2020, 13, 10. [Google Scholar] [CrossRef] [PubMed]
- Pillay, J.; Tak, T.; Kamp, V.M.; Koenderman, L. Immune suppression by neutrophils and granulocytic myeloid-derived suppressor cells: Similarities and differences. Cell Mol. Life Sci. 2013, 70, 3813–3827. [Google Scholar] [CrossRef]
- Zilio, S.; Serafini, P. Neutrophils and Granulocytic MDSC: The Janus God of Cancer Immunotherapy. Vaccines 2016, 4, 31. [Google Scholar] [CrossRef]
- Hao, Z.; Li, R.; Wang, Y.; Li, S.; Hong, Z.; Han, Z. Landscape of Myeloid-derived Suppressor Cell in Tumor Immunotherapy. Biomark. Res. 2021, 9, 77. [Google Scholar] [CrossRef]
- Veglia, F.; Sanseviero, E.; Gabrilovich, D.I. Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity. Nat. Rev. Immunol. 2021, 21, 485–498. [Google Scholar] [CrossRef]
- Bergenfelz, C.; Leandersson, K. The Generation and Identity of Human Myeloid-Derived Suppressor Cells. Front. Oncol. 2020, 10, 109. [Google Scholar] [CrossRef] [PubMed]
- Alshetaiwi, H.; Pervolarakis, N.; McIntyre, L.L.; Ma, D.; Nguyen, Q.; Rath, J.A.; Nee, K.; Hernandez, G.; Evans, K.; Torosian, L.; et al. Defining the emergence of myeloid-derived suppressor cells in breast cancer using single-cell transcriptomics. Sci. Immunol. 2020, 5, eaay6017. [Google Scholar] [CrossRef]
- Veglia, F.; Hashimoto, A.; Dweep, H.; Sanseviero, E.; De Leo, A.; Tcyganov, E.; Kossenkov, A.; Mulligan, C.; Nam, B.; Masters, G.; et al. Analysis of classical neutrophils and polymorphonuclear myeloid-derived suppressor cells in cancer patients and tumor-bearing mice. J. Exp. Med. 2021, 218, e20201803. [Google Scholar] [CrossRef]
- Engblom, C.; Pfirschke, C.; Pittet, M.J. The role of myeloid cells in cancer therapies. Nat. Rev. Cancer 2016, 16, 447–462. [Google Scholar] [CrossRef] [PubMed]
- Dumitru, C.A.; Moses, K.; Trellakis, S.; Lang, S.; Brandau, S. Neutrophils and granulocytic myeloid-derived suppressor cells: Immunophenotyping, cell biology and clinical relevance in human oncology. Cancer Immunol. Immunother. 2012, 61, 1155–1167. [Google Scholar] [CrossRef] [PubMed]
- Youn, J.I.; Collazo, M.; Shalova, I.N.; Biswas, S.K.; Gabrilovich, D.I. Characterization of the nature of granulocytic myeloid-derived suppressor cells in tumor-bearing mice. J. Leukoc. Biol. 2012, 91, 167–181. [Google Scholar] [CrossRef] [PubMed]
- Zhou, J.; Nefedova, Y.; Lei, A.; Gabrilovich, D. Neutrophils and PMN-MDSC: Their biological role and interaction with stromal cells. Semin. Immunol. 2018, 35, 19–28. [Google Scholar] [CrossRef] [PubMed]
- Benguigui, M.; Cooper, T.J.; Kalkar, P.; Schif-Zuck, S.; Halaban, R.; Bacchiocchi, A.; Kamer, I.; Deo, A.; Manobla, B.; Menachem, R.; et al. Interferon-stimulated neutrophils as a predictor of immunotherapy response. Cancer Cell 2024, 42, 253–265.e212. [Google Scholar] [CrossRef] [PubMed]
- Engblom, C.; Pfirschke, C.; Zilionis, R.; Da Silva Martins, J.; Bos, S.A.; Courties, G.; Rickelt, S.; Severe, N.; Baryawno, N.; Faget, J.; et al. Osteoblasts remotely supply lung tumors with cancer-promoting SiglecF(high) neutrophils. Science 2017, 358, eaal5081. [Google Scholar] [CrossRef]
- Garcia-Culebras, A.; Duran-Laforet, V.; Pena-Martinez, C.; Moraga, A.; Ballesteros, I.; Cuartero, M.I.; de la Parra, J.; Palma-Tortosa, S.; Hidalgo, A.; Corbi, A.L.; et al. Role of TLR4 (Toll-Like Receptor 4) in N1/N2 Neutrophil Programming after Stroke. Stroke 2019, 50, 2922–2932. [Google Scholar] [CrossRef]
- Peranzoni, E.; Zilio, S.; Marigo, I.; Dolcetti, L.; Zanovello, P.; Mandruzzato, S.; Bronte, V. Myeloid-derived suppressor cell heterogeneity and subset definition. Curr. Opin. Immunol. 2010, 22, 238–244. [Google Scholar] [CrossRef]
- Sagiv, J.Y.; Michaeli, J.; Assi, S.; Mishalian, I.; Kisos, H.; Levy, L.; Damti, P.; Lumbroso, D.; Polyansky, L.; Sionov, R.V.; et al. Phenotypic diversity and plasticity in circulating neutrophil subpopulations in cancer. Cell Rep. 2015, 10, 562–573. [Google Scholar] [CrossRef] [PubMed]
- Mihaila, A.C.; Ciortan, L.; Macarie, R.D.; Vadana, M.; Cecoltan, S.; Preda, M.B.; Hudita, A.; Gan, A.M.; Jakobsson, G.; Tucureanu, M.M.; et al. Transcriptional Profiling and Functional Analysis of N1/N2 Neutrophils Reveal an Immunomodulatory Effect of S100A9-Blockade on the Pro-Inflammatory N1 Subpopulation. Front. Immunol. 2021, 12, 708770. [Google Scholar] [CrossRef]
- Egholm, C.; Heeb, L.E.M.; Impellizzieri, D.; Boyman, O. The Regulatory Effects of Interleukin-4 Receptor Signaling on Neutrophils in Type 2 Immune Responses. Front. Immunol. 2019, 10, 2507. [Google Scholar] [CrossRef] [PubMed]
- Germann, M.; Zangger, N.; Sauvain, M.O.; Sempoux, C.; Bowler, A.D.; Wirapati, P.; Kandalaft, L.E.; Delorenzi, M.; Tejpar, S.; Coukos, G.; et al. Neutrophils suppress tumor-infiltrating T cells in colon cancer via matrix metalloproteinase-mediated activation of TGFbeta. EMBO Mol. Med. 2020, 12, e10681. [Google Scholar] [CrossRef] [PubMed]
- Halaby, M.J.; Hezaveh, K.; Lamorte, S.; Ciudad, M.T.; Kloetgen, A.; MacLeod, B.L.; Guo, M.; Chakravarthy, A.; Medina, T.D.S.; Ugel, S.; et al. GCN2 drives macrophage and MDSC function and immunosuppression in the tumor microenvironment. Sci. Immunol. 2019, 4, eaax8189. [Google Scholar] [CrossRef]
- Morgan, A.; Giese, L.E.H.; Huttenlocher, A. Neutrophil plasticity in the tumor microenvironment. Blood 2019, 133, 2159–2167. [Google Scholar]
- Leliefeld, P.H.; Koenderman, L.; Pillay, J. How Neutrophils Shape Adaptive Immune Responses. Front. Immunol. 2015, 6, 471. [Google Scholar] [CrossRef]
- McFarlane, A.J.; Fercoq, F.; Coffelt, S.B.; Carlin, L.M. Neutrophil dynamics in the tumor microenvironment. J. Clin. Investig. 2021, 131, e143759. [Google Scholar] [CrossRef]
- Mitchell, K.G.; Diao, L.; Karpinets, T.; Negrao, M.V.; Tran, H.T.; Parra, E.R.; Corsini, E.M.; Reuben, A.; Federico, L.; Bernatchez, C.; et al. Neutrophil expansion defines an immunoinhibitory peripheral and intratumoral inflammatory milieu in resected non-small cell lung cancer: A descriptive analysis of a prospectively immunoprofiled cohort. J. Immunother. Cancer 2020, 8, e000405. [Google Scholar] [CrossRef]
- Knaapen, A.M.; Gungor, N.; Schins, R.P.; Borm, P.J.; Van Schooten, F.J. Neutrophils and respiratory tract DNA damage and mutagenesis: A review. Mutagenesis 2006, 21, 225–236. [Google Scholar] [CrossRef] [PubMed]
- Wculek, S.K.; Bridgeman, V.L.; Peakman, F.; Malanchi, I. Early Neutrophil Responses to Chemical Carcinogenesis Shape Long-Term Lung Cancer Susceptibility. iScience 2020, 23, 101277. [Google Scholar] [CrossRef] [PubMed]
- Tecchio, C.; Scapini, P.; Pizzolo, G.; Cassatella, M.A. On the cytokines produced by human neutrophils in tumors. Semin. Cancer Biol. 2013, 23, 159–170. [Google Scholar] [CrossRef] [PubMed]
- Wislez, M.; Rabbe, N.; Marchal, J.; Milleron, B.; Crestani, B.; Mayaud, C.; Antoine, M.; Soler, P.; Cadranel, J. Hepatocyte growth factor production by neutrophils infiltrating bronchioloalveolar subtype pulmonary adenocarcinoma: Role in tumor progression and death. Cancer Res. 2003, 63, 1405–1412. [Google Scholar] [PubMed]
- Hattar, K.; Franz, K.; Ludwig, M.; Sibelius, U.; Wilhelm, J.; Lohmeyer, J.; Savai, R.; Subtil, F.S.; 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. 2014, 63, 1297–1306. [Google Scholar] [CrossRef]
- Wolff, H.; Saukkonen, K.; Anttila, S.; Karjalainen, A.; Vainio, H.; Ristimaki, A. Expression of cyclooxygenase-2 in human lung carcinoma. Cancer Res. 1998, 58, 4997–5001. [Google Scholar]
- Faget, J.; Groeneveld, S.; Boivin, G.; Sankar, M.; Zangger, N.; Garcia, M.; Guex, N.; Zlobec, I.; Steiner, L.; Piersigilli, A.; et al. Neutrophils and Snail Orchestrate the Establishment of a Pro-tumor Microenvironment in Lung Cancer. Cell Rep. 2017, 21, 3190–3204. [Google Scholar] [CrossRef]
- Cools-Lartigue, J.; Spicer, J.; McDonald, B.; Gowing, S.; Chow, S.; Giannias, B.; Bourdeau, F.; Kubes, P.; Ferri, L. Neutrophil extracellular traps sequester circulating tumor cells and promote metastasis. J. Clin. Investig. 2013, 123, 3446–3458. [Google Scholar] [CrossRef]
- Brinkmann, V.R.U.; Goosmann, C.; Fauler, B.; Uhlemann, Y.; Weiss, D.S.; Weinrauch, Y.; Zychlinsky, A. Neutrophil Extracellular Traps Kill Bacteria. Science 2004, 303, 1532–1535. [Google Scholar] [CrossRef]
- Zhang, L.; Yi, H.; Chen, J.; Li, H.; Luo, Y.; Cheng, T.; Yang, H.; Jiang, Z.; Pan, C. Neutrophil Extracellular Traps Facilitate A549 Cell Invasion and Migration in a Macrophage-Maintained Inflammatory Microenvironment. Biomed. Res. Int. 2022, 2022, 8316525. [Google Scholar] [CrossRef]
- Mauracher, L.M.; Hell, L.; Moik, F.; Krall, M.; Englisch, C.; Roiss, J.; Grilz, E.; Hofbauer, T.M.; Brostjan, C.; Knapp, S.; et al. Neutrophils in lung cancer patients: Activation potential and neutrophil extracellular trap formation. Res. Pract. Thromb. Haemost. 2023, 7, 100126. [Google Scholar] [CrossRef]
- Albrengues, J.; Shields, M.A.; Ng, D.; Park, C.G.; Ambrico, A.; Poindexter, M.E.; Upadhyay, P.; Uyeminami, D.L.; Pommier, A.; Kuttner, V.; et al. Neutrophil extracellular traps produced during inflammation awaken dormant cancer cells in mice. Science 2018, 36, eaao4227. [Google Scholar] [CrossRef] [PubMed]
- Gabriele Bergers, R.B.; McMahon, G.; Thiennu, H.; Vu, T.I.; Tamaki, K.; Tanzawa, K.; Thorpe, P.; Itohara, S.; Werb, Z.; Hanahan, D. Matrix metalloproteinase-9 triggers the angiogenic switch during carcinogenesis. Nat. Cell Biol. 2000, 2, 737–744. [Google Scholar] [CrossRef]
- Jablonska, J.; Leschner, S.; Westphal, K.; Lienenklaus, S.; Weiss, S. Neutrophils responsive to endogenous IFN-beta regulate tumor angiogenesis and growth in a mouse tumor model. J. Clin. Investig. 2010, 120, 1151–1164. [Google Scholar] [CrossRef] [PubMed]
- Merchant, N.; Nagaraju, G.P.; Rajitha, B.; Lammata, S.; Jella, K.K.; Buchwald, Z.S.; Lakka, S.S.; Ali, A.N. Matrix metalloproteinases: Their functional role in lung cancer. Carcinogenesis 2017, 38, 766–780. [Google Scholar] [CrossRef]
- Curtis, V.F.; Wang, H.; Yang, P.; McLendon, R.E.; Li, X.; Zhou, Q.Y.; Wang, X.F. A PK2/Bv8/PROK2 antagonist suppresses tumorigenic processes by inhibiting angiogenesis in glioma and blocking myeloid cell infiltration in pancreatic cancer. PLoS ONE 2013, 8, e54916. [Google Scholar] [CrossRef]
- Wang, Y.; Liu, F.; Chen, L.; Fang, C.; Li, S.; Yuan, S.; Qian, X.; Yin, Y.; Yu, B.; Fu, B.; et al. Neutrophil Extracellular Traps (NETs) Promote Non-Small Cell Lung Cancer Metastasis by Suppressing lncRNA MIR503HG to Activate the NF-kappaB/NLRP3 Inflammasome Pathway. Front. Immunol. 2022, 13, 867516. [Google Scholar] [CrossRef]
- Craig, L.S.; Liu, F.; Gregory, D.A.; Gregory, K.S.; Link, D.C. G-CSF Is an Essential Regulator of Neutrophil Trafficking from the Bone Marrow to the Blood. Immunity 2002, 17, 413–423. [Google Scholar] [CrossRef]
- Capucetti, A.; Albano, F.; Bonecchi, R. Multiple Roles for Chemokines in Neutrophil Biology. Front. Immunol. 2020, 11, 1259. [Google Scholar] [CrossRef]
- Martin, C.; Burdon, P.C.; Bridger, G.; Gutierrez-Ramos, J.C.; Williams, T.J.; Rankin, S.M. Chemokines Acting via CXCR2 and CXCR4 Control the Release of Neutrophils from the Bone Marrow and Their Return following Senescence. Immunity 2003, 19, 583–593. [Google Scholar] [CrossRef]
- Kohler, A.; De Filippo, K.; Hasenberg, M.; van den Brandt, C.; Nye, E.; Hosking, M.P.; Lane, T.E.; Mann, L.; Ransohoff, R.M.; Hauser, A.E.; et al. G-CSF-mediated thrombopoietin release triggers neutrophil motility and mobilization from bone marrow via induction of Cxcr2 ligands. Blood 2011, 117, 4349–4357. [Google Scholar] [CrossRef] [PubMed]
- Coffelt, S.B.; Wellenstein, M.D.; de Visser, K.E. Neutrophils in cancer: Neutral no more. Nat. Rev. Cancer 2016, 16, 431–446. [Google Scholar] [CrossRef] [PubMed]
- Mollica Poeta, V.; Massara, M.; Capucetti, A.; Bonecchi, R. Chemokines and Chemokine Receptors: New Targets for Cancer Immunotherapy. Front. Immunol. 2019, 10, 379. [Google Scholar] [CrossRef] [PubMed]
- Jamieson, T.; Clarke, M.; Steele, C.W.; Samuel, M.S.; Neumann, J.; Jung, A.; Huels, D.; Olson, M.F.; Das, S.; Nibbs, R.J.; et al. Inhibition of CXCR2 profoundly suppresses inflammation-driven and spontaneous tumorigenesis. J. Clin. Investig. 2012, 122, 3127–3144. [Google Scholar] [CrossRef] [PubMed]
- Cheng, Y.; Ma, X.L.; Wei, Y.Q.; Wei, X.W. Potential roles and targeted therapy of the CXCLs/CXCR2 axis in cancer and inflammatory diseases. Biochim. Biophys. Acta Rev. Cancer 2019, 1871, 289–312. [Google Scholar] [CrossRef] [PubMed]
- Wislez, M.; Fujimoto, N.; Izzo, J.G.; Hanna, A.E.; Cody, D.D.; Langley, R.R.; Tang, H.; Burdick, M.D.; Sato, M.; Minna, J.D.; et al. High expression of ligands for chemokine receptor CXCR2 in alveolar epithelial neoplasia induced by oncogenic kras. Cancer Res. 2006, 66, 4198–4207. [Google Scholar] [CrossRef] [PubMed]
- Cheng, Y.; Mo, F.; Li, Q.; Han, X.; Shi, H.; Chen, S.; Wei, Y.; Wei, X. Targeting CXCR2 inhibits the progression of lung cancer and promotes therapeutic effect of cisplatin. Mol. Cancer 2021, 20, 62. [Google Scholar] [CrossRef] [PubMed]
- Li, P.; Lu, M.; Shi, J.; Gong, Z.; Hua, L.; Li, Q.; Lim, B.; Zhang, X.H.; Chen, X.; Li, S.; et al. Lung mesenchymal cells elicit lipid storage in neutrophils that fuel breast cancer lung metastasis. Nat. Immunol. 2020, 21, 1444–1455. [Google Scholar] [CrossRef] [PubMed]
- Xing, Y.; Ruan, G.; Ni, H.; Qin, H.; Chen, S.; Gu, X.; Shang, J.; Zhou, Y.; Tao, X.; Zheng, L. Tumor Immune Microenvironment and Its Related miRNAs in Tumor Progression. Front. Immunol. 2021, 12, 624725. [Google Scholar] [CrossRef]
- Jeffries, J.; Zhou, W.; Hsu, A.Y.; Deng, Q. miRNA-223 at the crossroads of inflammation and cancer. Cancer Lett. 2019, 451, 136–141. [Google Scholar] [CrossRef]
- Zangari, J.; Ilie, M.; Rouaud, F.; Signetti, L.; Ohanna, M.; Didier, R.; Romeo, B.; Goldoni, D.; Nottet, N.; Staedel, C.; et al. Rapid decay of engulfed extracellular miRNA by XRN1 exonuclease promotes transient epithelial-mesenchymal transition. Nucleic Acids Res. 2017, 45, 4131–4141. [Google Scholar] [CrossRef]
- Guo, Y.; Xie, Y.; Luo, Y. The Role of Long Non-Coding RNAs in the Tumor Immune Microenvironment. Front. Immunol. 2022, 13, 851004. [Google Scholar] [CrossRef] [PubMed]
- Wang, T.; Cao, L.; Dong, X.; Wu, F.; De, W.; Huang, L.; Wan, Q. LINC01116 promotes tumor proliferation and neutrophil recruitment via DDX5-mediated regulation of IL-1beta in glioma cell. Cell Death Dis. 2020, 11, 302. [Google Scholar] [CrossRef]
- Kotzin, J.J.; Spencer, S.P.; McCright, S.J.; Kumar, D.B.U.; Collet, M.A.; Mowel, W.K.; Elliott, E.N.; Uyar, A.; Makiya, M.A.; Dunagin, M.C.; et al. The long non-coding RNA Morrbid regulates Bim and short-lived myeloid cell lifespan. Nature 2016, 537, 239–243. [Google Scholar] [CrossRef]
- Shang, A.; Wang, W.; Gu, C.; Chen, C.; Zeng, B.; Yang, Y.; Ji, P.; Sun, J.; Wu, J.; Lu, W.; et al. Long non-coding RNA HOTTIP enhances IL-6 expression to potentiate immune escape of ovarian cancer cells by upregulating the expression of PD-L1 in neutrophils. J. Exp. Clin. Cancer Res. 2019, 38, 411. [Google Scholar] [CrossRef]
- Kumar, D.; Gurrapu, S.; Wang, Y.; Bae, S.Y.; Pandey, P.R.; Chen, H.; Mondal, J.; Han, H.; Wu, C.J.; Karaiskos, S.; et al. LncRNA Malat1 suppresses pyroptosis and T cell-mediated killing of incipient metastatic cells. Nat. Cancer 2024, 5, 262–282. [Google Scholar] [CrossRef]
- Tang, Q.; Chen, Y.; Li, X.; Long, S.; Shi, Y.; Yu, Y.; Wu, W.; Han, L.; Wang, S. The role of PD-1/PD-L1 and application of immune-checkpoint inhibitors in human cancers. Front. Immunol. 2022, 13, 964442. [Google Scholar] [CrossRef]
- Buchbinder, E.I.; Desai, A. CTLA-4 and PD-1 Pathways: Similarities, Differences, and Implications of Their Inhibition. Am. J. Clin. Oncol. 2016, 39, 98–106. [Google Scholar] [CrossRef] [PubMed]
- Yu, Y.; Qian, L.; Cui, J. Value of neutrophil-to-lymphocyte ratio for predicting lung cancer prognosis: A meta-analysis of 7,219 patients. Mol. Clin. Oncol. 2017, 7, 498–506. [Google Scholar] [CrossRef] [PubMed]
- Kargl, J.; Zhu, X.; Zhang, H.; Yang, G.H.Y.; Friesen, T.J.; Shipley, M.; Maeda, D.Y.; Zebala, J.A.; McKay-Fleisch, J.; Meredith, G.; et al. Neutrophil content predicts lymphocyte depletion and anti-PD1 treatment failure in NSCLC. JCI Insight 2019, 4, e130850. [Google Scholar] [CrossRef]
- Alessi, J.V.; Ricciuti, B.; Alden, S.L.; Bertram, A.A.; Lin, J.J.; Sakhi, M.; Nishino, M.; Vaz, V.R.; Lindsay, J.; Turner, M.M.; et al. Low peripheral blood derived neutrophil-to-lymphocyte ratio (dNLR) is associated with increased tumor T-cell infiltration and favorable outcomes to first-line pembrolizumab in non-small cell lung cancer. J. Immunother. Cancer 2021, 9, e003536. [Google Scholar] [CrossRef] [PubMed]
- Zhou, J.G.; Wong, A.H.; Wang, H.; Tan, F.; Chen, X.; Jin, S.H.; He, S.S.; Shen, G.; Wang, Y.J.; Frey, B.; et al. Elucidation of the Application of Blood Test Biomarkers to Predict Immune-Related Adverse Events in Atezolizumab-Treated NSCLC Patients Using Machine Learning Methods. Front. Immunol. 2022, 13, 862752. [Google Scholar] [CrossRef] [PubMed]
- 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. 2015, 34, 141. [Google Scholar] [CrossRef] [PubMed]
- Yamauchi, Y.; Safi, S.; Blattner, C.; Rathinasamy, A.; Umansky, L.; Juenger, S.; Warth, A.; Eichhorn, M.; Muley, T.; Herth, F.J.F.; et al. Circulating and Tumor Myeloid-derived Suppressor Cells in Resectable Non-Small Cell Lung Cancer. Am. J. Respir. Crit. Care Med. 2018, 198, 777–787. [Google Scholar] [CrossRef] [PubMed]
- Wang, P.-F.; Zhang, Y.-X.; Su, J.; Yao, K.; Li, S.-W.; Huang, G.-R.; Yan, C.-X. Neutrophil depletion enhances the therapeutic effect of PD-1 antibody on glioma. Aging 2020, 12, 15290. [Google Scholar] [CrossRef] [PubMed]
- Eruslanov, E.B.; Bhojnagarwala, P.S.; Quatromoni, J.G.; Stephen, T.L.; Ranganathan, A.; Deshpande, C.; Akimova, T.; Vachani, A.; Litzky, L.; Hancock, W.W.; et al. Tumor-associated neutrophils stimulate T cell responses in early-stage human lung cancer. J. Clin. Investig. 2014, 124, 5466–5480. [Google Scholar] [CrossRef]
- Liu, C.Y.; Wang, Y.M.; Wang, C.L.; Feng, P.H.; Ko, H.W.; Liu, Y.H.; Wu, Y.C.; Chu, Y.; Chung, F.T.; Kuo, C.H.; et al. Population alterations of L-arginase- and inducible nitric oxide synthase-expressed CD11b+/CD14-/CD15+/CD33+ myeloid-derived suppressor cells and CD8+ T lymphocytes in patients with advanced-stage non-small cell lung cancer. J. Cancer Res. Clin. Oncol. 2010, 136, 35–45. [Google Scholar] [CrossRef] [PubMed]
- Miret, J.J.; Kirschmeier, P.; Koyama, S.; Zhu, M.; Li, Y.Y.; Naito, Y.; Wu, M.; Malladi, V.S.; Huang, W.; Walker, W.; et al. Suppression of Myeloid Cell Arginase Activity leads to Therapeutic Response in a NSCLC Mouse Model by Activating Anti-Tumor Immunity. J. Immunother. Cancer 2019, 7, 32. [Google Scholar] [CrossRef] [PubMed]
- Yang, J.; Jia, Y.; Wang, B.; Yang, S.; Du, K.; Luo, Y.; Li, Y.; Zhu, B. Circular RNA CHST15 Sponges miR-155-5p and miR-194-5p to Promote the Immune Escape of Lung Cancer Cells Mediated by PD-L1. Front. Oncol. 2021, 11, 595609. [Google Scholar] [CrossRef]
- Zhou, S.L.; Zhou, Z.J.; Hu, Z.Q.; Huang, X.W.; Wang, Z.; Chen, E.B.; Fan, J.; Cao, Y.; Dai, Z.; Zhou, J. Tumor-Associated Neutrophils Recruit Macrophages and T-Regulatory Cells to Promote Progression of Hepatocellular Carcinoma and Resistance to Sorafenib. Gastroenterology 2016, 150, 1646–1658.e1617. [Google Scholar] [CrossRef]
- Ganesan, A.P.; Johansson, M.; Ruffell, B.; Yagui-Beltran, A.; Lau, J.; Jablons, D.M.; Coussens, L.M. Tumor-infiltrating regulatory T cells inhibit endogenous cytotoxic T cell responses to lung adenocarcinoma. J. Immunol. 2013, 191, 2009–2017. [Google Scholar] [CrossRef]
- Mishalian, I.; Bayuh, R.; Eruslanov, E.; Michaeli, J.; Levy, L.; Zolotarov, L.; Singhal, S.; Albelda, S.M.; Granot, Z.; Fridlender, Z.G. Neutrophils recruit regulatory T-cells into tumors via secretion of CCL17--a new mechanism of impaired antitumor immunity. Int. J. Cancer 2014, 135, 1178–1186. [Google Scholar] [CrossRef] [PubMed]
- Shabani, F.; Farasat, A.; Mahdavi, M.; Gheibi, N. Calprotectin (S100A8/S100A9): A key protein between inflammation and cancer. Inflamm. Res. 2018, 67, 801–812. [Google Scholar] [CrossRef]
- Chao, Y.; Jiang, W.; Wang, X.; Wang, X.; Song, J.; Chen, C.; Zhou, J.; Huang, Q.; Hu, J.; Song, Y. Discovery of efficacy biomarkers for non-small cell lung cancer with first-line anti-PD-1 immunotherapy by data-independent acquisition mass spectrometry. Clin. Exp. Immunol. 2022, 208, 60–71. [Google Scholar] [CrossRef] [PubMed]
- Lindau, D.; Gielen, P.; Kroesen, M.; Wesseling, P.; Adema, G.J. The immunosuppressive tumour network: Myeloid-derived suppressor cells, regulatory T cells and natural killer T cells. Immunology 2013, 138, 105–115. [Google Scholar] [CrossRef]
- Ortiz, M.L.; Lu, L.; Ramachandran, I.; Gabrilovich, D.I. Myeloid-derived suppressor cells in the development of lung cancer. Cancer Immunol. Res. 2014, 2, 50–58. [Google Scholar] [CrossRef] [PubMed]
- Akbay, E.A.; Koyama, S.; Liu, Y.; Dries, R.; Bufe, L.E.; Silkes, M.; Alam, M.M.; Magee, D.M.; Jones, R.; Jinushi, M.; et al. Interleukin-17A Promotes Lung Tumor Progression through Neutrophil Attraction to Tumor Sites and Mediating Resistance to PD-1 Blockade. J. Thorac. Oncol. 2017, 12, 1268–1279. [Google Scholar] [CrossRef]
- Kaltenmeier, C.; Yazdani, H.O.; Morder, K.; Geller, D.A.; Simmons, R.L.; Tohme, S. Neutrophil Extracellular Traps Promote T Cell Exhaustion in the Tumor Microenvironment. Front. Immunol. 2021, 12, 785222. [Google Scholar] [CrossRef]
- Li, Z.; Wang, J.; Zhang, X.; Liu, P.; Zhang, X.; Wang, J.; Zheng, X.; Wei, L.; Peng, Q.; Liu, C.; et al. Proinflammatory S100A8 Induces PD-L1 Expression in Macrophages, Mediating Tumor Immune Escape. J. Immunol. 2020, 204, 2589–2599. [Google Scholar] [CrossRef]
- Spiegel, A.; Brooks, M.W.; Houshyar, S.; Reinhardt, F.; Ardolino, M.; Fessler, E.; Chen, M.B.; Krall, J.A.; DeCock, J.; Zervantonakis, I.K.; et al. Neutrophils Suppress Intraluminal NK Cell-Mediated Tumor Cell Clearance and Enhance Extravasation of Disseminated Carcinoma Cells. Cancer Discov. 2016, 6, 630–649. [Google Scholar] [CrossRef]
- Sarvaria, A.; Madrigal, J.A.; Saudemont, A. B cell regulation in cancer and anti-tumor immunity. Cell Mol. Immunol. 2017, 14, 662–674. [Google Scholar] [CrossRef] [PubMed]
- Leong, T.L.; Bryant, V.L. B cells in lung cancer-not just a bystander cell: A literature review. Transl. Lung Cancer Res. 2021, 10, 2830–2841. [Google Scholar] [CrossRef] [PubMed]
- Yang, C.; Lee, H.; Pal, S.; Jove, V.; Deng, J.; Zhang, W.; Hoon, D.S.; Wakabayashi, M.; Forman, S.; Yu, H. B cells promote tumor progression via STAT3 regulated-angiogenesis. PLoS ONE 2013, 8, e64159. [Google Scholar] [CrossRef] [PubMed]
- Shaul, M.E.; Zlotnik, A.; Tidhar, E.; Schwartz, A.; Arpinati, L.; Kaisar-Iluz, N.; Mahroum, S.; Mishalian, I.; Fridlender, Z.G. Tumor-Associated Neutrophils Drive B-cell Recruitment and Their Differentiation to Plasma Cells. Cancer Immunol. Res. 2021, 9, 811–824. [Google Scholar] [CrossRef] [PubMed]
- Costa, S.; Bevilacqua, D.; Cassatella, M.A.; Scapini, P. Recent advances on the crosstalk between neutrophils and B or T lymphocytes. Immunology 2019, 156, 23–32. [Google Scholar] [CrossRef]
- Lelis, F.J.N.; Jaufmann, J.; Singh, A.; Fromm, K.; Teschner, A.C.; Poschel, S.; Schafer, I.; Beer-Hammer, S.; Rieber, N.; Hartl, D. Myeloid-derived suppressor cells modulate B-cell responses. Immunol. Lett. 2017, 188, 108–115. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Guoqiang, L.; Sun, M.; Lu, X. Targeting and exploitation of tumor-associated neutrophils to enhance immunotherapy and drug delivery for cancer treatment. Cancer Biol. Med. 2020, 17, 32–43. [Google Scholar] [CrossRef]
- De Sanctis, F.; Adamo, A.; Cane, S.; Ugel, S. Targeting tumour-reprogrammed myeloid cells: The new battleground in cancer immunotherapy. Semin. Immunopathol. 2022, 45, 163–186. [Google Scholar] [CrossRef]
- Gong, P.; Liu, H.; Liu, X.; Zhou, G.; Liu, M.; Yang, X.; Xiong, W.; Wang, Q.; Ma, J.; Ren, Z.; et al. Efficacy of tasquinimod in men with metastatic castration-resistant prostate cancer: A meta-analysis of randomized controlled trials. Medicine 2018, 97, e13204. [Google Scholar] [CrossRef]
- Holmgaard, R.B.; Schaer, D.A.; Li, Y.; Castaneda, S.P.; Murphy, M.Y.; Xu, X.; Inigo, I.; Dobkin, J.; Manro, J.R.; Iversen, P.W.; et al. Targeting the TGFbeta pathway with galunisertib, a TGFbetaRI small molecule inhibitor, promotes anti-tumor immunity leading to durable, complete responses, as monotherapy and in combination with checkpoint blockade. J. Immunother. Cancer 2018, 6, 47. [Google Scholar] [CrossRef]
- Yuan, M.; Zhai, Y.; Men, Y.; Zhao, M.; Sun, X.; Ma, Z.; Bao, Y.; Yang, X.; Sun, S.; Liu, Y.; et al. Anlotinib Enhances the Antitumor Activity of High-Dose Irradiation Combined with Anti-PD-L1 by Potentiating the Tumor Immune Microenvironment in Murine Lung Cancer. Oxid. Med. Cell Longev. 2022, 2022, 5479491. [Google Scholar] [CrossRef] [PubMed]
- Teijeira, A.; Garasa, S.; Gato, M.; Alfaro, C.; Migueliz, I.; Cirella, A.; de Andrea, C.; Ochoa, M.C.; Otano, I.; Etxeberria, I.; et al. CXCR1 and CXCR2 Chemokine Receptor Agonists Produced by Tumors Induce Neutrophil Extracellular Traps that Interfere with Immune Cytotoxicity. Immunity 2020, 52, 856–871.e858. [Google Scholar] [CrossRef] [PubMed]
- Crocetti, L.; Giovannoni, M.P.; Cantini, N.; Guerrini, G.; Vergelli, C.; Schepetkin, I.A.; Khlebnikov, A.I.; Quinn, M.T. Novel Sulfonamide Analogs of Sivelestat as Potent Human Neutrophil Elastase Inhibitors. Front. Chem. 2020, 8, 795. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Miao, S.; Rodriguez, B.L.; Gibbons, D.L. The Multifaceted Role of Neutrophils in NSCLC in the Era of Immune Checkpoint Inhibitors. Cancers 2024, 16, 2507. https://doi.org/10.3390/cancers16142507
Miao S, Rodriguez BL, Gibbons DL. The Multifaceted Role of Neutrophils in NSCLC in the Era of Immune Checkpoint Inhibitors. Cancers. 2024; 16(14):2507. https://doi.org/10.3390/cancers16142507
Chicago/Turabian StyleMiao, Shucheng, Bertha Leticia Rodriguez, and Don L. Gibbons. 2024. "The Multifaceted Role of Neutrophils in NSCLC in the Era of Immune Checkpoint Inhibitors" Cancers 16, no. 14: 2507. https://doi.org/10.3390/cancers16142507
APA StyleMiao, S., Rodriguez, B. L., & Gibbons, D. L. (2024). The Multifaceted Role of Neutrophils in NSCLC in the Era of Immune Checkpoint Inhibitors. Cancers, 16(14), 2507. https://doi.org/10.3390/cancers16142507