Targeting neutrophils: Mechanism and advances in cancer therapy

Shuaixi Yang , Jiachi Jia , Fuqi Wang , Yuhang Wang , Yingshuai Fang , Yabing Yang , Quanbo Zhou , Weitang Yuan , Zhilei Bian

Clinical and Translational Medicine ›› 2024, Vol. 14 ›› Issue (3) : e1599

PDF
Clinical and Translational Medicine ›› 2024, Vol. 14 ›› Issue (3) : e1599 DOI: 10.1002/ctm2.1599
REVIEW

Targeting neutrophils: Mechanism and advances in cancer therapy

Author information +
History +
PDF

Abstract

Background: Cancer is a thorny problem which cannot be conquered by mankind at present and recent researchers have put their focus on tumor microenviroment. Neutrophils, the prominent leukocytes in peripheral blood that accumulate in tumours, serves as frontline cells in response to tumour progression owing to the rapid development of micro biotechnology. Hence, targeted therapy with these neutrophils has made targeting treatment a promising field in cancer therapy.

Main body: We broadly summarise some studies on the phenotypes and functions of tumour-associated neutrophils as well as the unique web-like products of neutrophils that play a role in cancer progression—neutrophil extracellular traps—and the interactions between neutrophils and the tumour microenvironment. Moreover, several targeted neutrophils therapeutic studies have made some progress and provided potential strategies for the treatment of cancer.

Conclusion: This review aims to offer a holistic perspective on therapeutic interventions targeting neutrophils to further inspire more researches on cancer therapies.

Keywords

NETs / neutrophils / targeting therapy / tumour microenvironment

Cite this article

Download citation ▾
Shuaixi Yang, Jiachi Jia, Fuqi Wang, Yuhang Wang, Yingshuai Fang, Yabing Yang, Quanbo Zhou, Weitang Yuan, Zhilei Bian. Targeting neutrophils: Mechanism and advances in cancer therapy. Clinical and Translational Medicine, 2024, 14(3): e1599 DOI:10.1002/ctm2.1599

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Coffelt SB, Wellenstein MD, De Visser KE. Neutrophils in cancer: neutral no more. Nat Rev Cancer. 2016;16:431-446.

[2]

Chiang C-C, Cheng W-J, Korinek M, Lin C-Y, Hwang T-L. Neutrophils in psoriasis. Front Immunol. 2019;10:2376.

[3]

Hedrick CC, Malanchi I. Neutrophils in cancer: heterogeneous and multifaceted. Nat Rev Immunol. 2022;22:173-187.

[4]

Cerezo-Wallis D, Ballesteros I. Neutrophils in cancer, a love-hate affair. FEBS J. 2022;289:3692-3703.

[5]

Fridlender ZG, Sun J, Kim S, et al. Polarization of tumor-associated neutrophil phenotype by TGF-beta: “N1” versus “N2” TAN. Cancer Cell. 2009;16:183-194.

[6]

Mollinedo F. Neutrophil degranulation, plasticity, and cancer metastasis. Trends Immunol. 2019;40:228-242.

[7]

Xie X, Shi Q, Wu P, et al. Single-cell transcriptome profiling reveals neutrophil heterogeneity in homeostasis and infection. Nat Immunol. 2020;21:1119-1133.

[8]

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

[9]

Bianchi A, De Castro Silva I, Deshpande NU, et al. Cell-autonomous Cxcl1 sustains tolerogenic circuitries and stromal inflammation via neutrophil-derived TNF in pancreatic cancer. Cancer Discov. 2023;13:1428-1453.

[10]

Yvan-Charvet L, Ng LG. Granulopoiesis and neutrophil homeostasis: a metabolic, daily balancing act. Trends Immunol. 2019;40:598-612.

[11]

Liew PX, Kubes P. The neutrophil's role during health and disease. Physiol Rev. 2019;99:1223-1248.

[12]

Mistry P, Nakabo S, O'Neil L, et al. Transcriptomic, epigenetic, and functional analyses implicate neutrophil diversity in the pathogenesis of systemic lupus erythematosus. Proc Natl Acad Sci U S A. 2019;116:25222-25228.

[13]

Dahl R, Walsh JC, Lancki D, et al. Regulation of macrophage and neutrophil cell fates by the PU.1:c/EBPalpha ratio and granulocyte colony-stimulating factor. Nat Immunol. 2003;4:1029-1036.

[14]

Rosmarin AG, Yang Z, Resendes KK. Transcriptional regulation in myelopoiesis: hematopoietic fate choice, myeloid differentiation, and leukemogenesis. Exp Hematol. 2005;33:131-143.

[15]

Goldschmeding R, Van Dalen CM, Faber N, et al. Further characterization of the NB 1 antigen as a variably expressed 56–62 kD GPI-linked glycoprotein of plasma membranes and specific granules of neutrophils. Br J Haematol. 1992;81:336-345.

[16]

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

[17]

Geh D, Leslie J, Rumney R, Reeves HL, Bird TG, Mann DA. Neutrophils as potential therapeutic targets in hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol. 2022;19:257-273.

[18]

Gershkovitz M, Caspi Y, Fainsod-Levi T, et al. TRPM2 mediates neutrophil killing of disseminated tumor cells. Cancer Res. 2018;78:2680-2690.

[19]

Granot Z, Henke E, Comen EA, King TA, Norton L, Benezra R. Tumor entrained neutrophils inhibit seeding in the premetastatic lung. Cancer Cell. 2011;20:300-314.

[20]

Kruk J, Aboul-Enein HY. Reactive oxygen and nitrogen species in carcinogenesis: implications of oxidative stress on the progression and development of several cancer types. Mini Rev Med Chem. 2017;17:904-919.

[21]

Das A, Monteiro M, Barai A, Kumar S, Sen S. MMP proteolytic activity regulates cancer invasiveness by modulating integrins. Sci Rep. 2017;7:14219.

[22]

Petri B, Sanz M-J. Neutrophil chemotaxis. Cell Tissue Res. 2018;371:425-436.

[23]

De Oliveira S, Rosowski EE, Huttenlocher A. Neutrophil migration in infection and wound repair: going forward in reverse. Nat Rev Immunol. 2016;16:378-391.

[24]

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;72:958-971.

[25]

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

[26]

Thanabalasuriar A, Kubes P. Rise and shine: open your eyes to produce anti-inflammatory NETs. J Leukoc Biol. 2019;105:1083-1084.

[27]

Gungabeesoon J, Gort-Freitas NA, Kiss M, et al. A neutrophil response linked to tumor control in immunotherapy. Cell. 2023;186:1448-1464.e1420.

[28]

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;204:1362-1372.

[29]

Papayannopoulos V. Neutrophil extracellular traps in immunity and disease. Nat Rev Immunol. 2018;18:134-147.

[30]

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

[31]

Fuchs TA, Abed U, Goosmann C, et al. Novel cell death program leads to neutrophil extracellular traps. J Cell Biol. 2007;176:231-241.

[32]

Chen T, Li Y, Sun R, et al. Receptor-mediated NETosis on neutrophils. Front Immunol. 2021;12:775267.

[33]

Alemán OR, Mora N, Cortes-Vieyra R, Uribe-Querol E, Rosales C. Differential use of human neutrophil Fcγ receptors for inducing neutrophil extracellular trap formation. J Immunol Res. 2016;2016:2908034.

[34]

Alemán OR, Mora N, Cortes-Vieyra R, Uribe-Querol E, Rosales C. Transforming growth factor-β-activated kinase 1 is required for human FcγRIIIb-induced neutrophil extracellular trap formation. Front Immunol. 2016;7:277.

[35]

Khan U, Chowdhury S, Billah MM, Islam KMD, Thorlacius H, Rahman M. Neutrophil extracellular traps in colorectal cancer progression and metastasis. Int J Mol Sci. 2021;22:7260.

[36]

Sørensen OE, Borregaard N. Neutrophil extracellular traps—the dark side of neutrophils. J Clin Invest. 2016;126:1612-1620.

[37]

Castanheira FVS, Kubes P. Neutrophils and NETs in modulating acute and chronic inflammation. Blood. 2019;133:2178-2185.

[38]

Meegan JE, Yang X, Coleman DC, Jannaway M, Yuan SY. Neutrophil-mediated vascular barrier injury: role of neutrophil extracellular traps. Microcirculation. 2017;24.

[39]

Wang Z, Kim SY, Tu W, et al. Extracellular vesicles in fatty liver promote a metastatic tumor microenvironment. Cell Metab. 2023;35:1209-1226.e1213.

[40]

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

[41]

Gautam SK, Batra SK, Jain M. Molecular and metabolic regulation of immunosuppression in metastatic pancreatic ductal adenocarcinoma. Mol Cancer. 2023;22:118.

[42]

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

[43]

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. 2018;216:176-194.

[44]

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;13:1017.

[45]

Ronchetti L, Boubaker NS, Barba M, Vici P, Gurtner A, Piaggio G. Neutrophil extracellular traps in cancer: not only catching microbes. J Exp Clin Cancer Res. 2021;40:231.

[46]

Shahzad MH, Feng L, Su X, et al. Neutrophil extracellular traps in cancer therapy resistance. Cancers (Basel). 2022;14:1359.

[47]

Najmeh S, Cools-Lartigue J, Rayes RF, et al. Neutrophil extracellular traps sequester circulating tumor cells via β1-integrin mediated interactions. Int J Cancer. 2017;140:2321-2330.

[48]

Rayes RF, Mouhanna JG, Nicolau I, et al. Primary tumors induce neutrophil extracellular traps with targetable metastasis promoting effects. JCI Insight. 2019;5:128008.

[49]

Lavoie SS, Dumas E, Vulesevic B, Neagoe P-E, White M, Sirois MG. Synthesis of human neutrophil extracellular traps contributes to angiopoietin-mediated in vitro proinflammatory and proangiogenic activities. J Immunol. 2018;200:3801-3813.

[50]

Hidalgo A, Libby P, Soehnlein O, Aramburu IV, Papayannopoulos V, Silvestre-Roig C. Neutrophil extracellular traps: from physiology to pathology. Cardiovasc Res. 2022;118:2737-2753.

[51]

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

[52]

Lin C, Herlihy SE, Li M, et al. Abstract 2103: NETs promote tumor resistance to anthracyclines. Cancer Res. 2019;79:2103-2103.

[53]

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;52:856-871.e858.

[54]

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

[55]

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

[56]

Neutrophil extracellular traps confer chemoresistance through TGFβ. Cancer Discov. 2023;13:OF2.

[57]

Zhang Yu, 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:e20190354.

[58]

Tesi RJ. MDSC; the most important cell you have never heard of. Trends Pharmacol Sci. 2019;40:4-7.

[59]

Gabrilovich DI. Myeloid-derived suppressor cells. Cancer Immunol Res. 2017;5:3-8.

[60]

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.

[61]

Ramachandran IR, Condamine T, Lin C, et al. Bone marrow PMN-MDSCs and neutrophils are functionally similar in protection of multiple myeloma from chemotherapy. Cancer Lett. 2016;371:117-124.

[62]

Ugel S, De Sanctis F, Mandruzzato S, Bronte V. Tumor-induced myeloid deviation: when myeloid-derived suppressor cells meet tumor-associated macrophages. J Clin Invest. 2015;125:3365-3376.

[63]

Condamine T, Dominguez GA, Youn JI, et al. Lectin-type oxidized LDL receptor-1 distinguishes population of human polymorphonuclear myeloid-derived suppressor cells in cancer patients. Sci Immunol. 2016;1:aaf8943.

[64]

Zhao X, Lu Ye, Cui Li. Neutrophil-sourced TNF in cancer: deciphering an intricate orchestrator of immunosuppressive communication in the tumor microenvironment. Signal Transduct Target Ther. 2023;8:272.

[65]

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;314:250-279.

[66]

Tcyganov E, Mastio J, Chen E, Gabrilovich DI. Plasticity of myeloid-derived suppressor cells in cancer. Curr Opin Immunol. 2018;51:76-82.

[67]

Rahmy S, Lu X. Chapter 6—Targeting tumor-associated neutrophils in immunotherapy. In: Amiji MM, Milane LS, eds. Systemic Drug Delivery Strategies. Academic Press; 2022:147-161.

[68]

Kim J, Bae J-S. Tumor-associated macrophages and neutrophils in tumor microenvironment. Mediat Inflamm. 2016;2016:6058147.

[69]

Soehnlein O, Lindbom L, Weber C. Mechanisms underlying neutrophil-mediated monocyte recruitment. Blood. 2009;114:4613-4623.

[70]

Kumar V, Sharma A. Neutrophils: cinderella of innate immune system. Int Immunopharmacol. 2010;10:1325-1334.

[71]

Zhou Z, Wang P, Sun R, et al. Tumor-associated neutrophils and macrophages interaction contributes to intrahepatic cholangiocarcinoma progression by activating STAT3. J Immunother Cancer. 2021;9:e001946.

[72]

Tam TH, Chan KL, Boroumand P, et al. Nucleotides released from palmitate-activated murine macrophages attract neutrophils. J Biol Chem. 2020;295:4902-4911.

[73]

Hinshaw DC, Shevde LA. The tumor microenvironment innately modulates cancer progression. Cancer Res. 2019;79:4557-4566.

[74]

Bhattacharya P, Ismail N, Saxena A, et al. Neutrophil-dendritic cell interaction plays an important role in live attenuated Leishmania vaccine induced immunity. PLoS Negl Trop Dis. 2022;16:e0010224.

[75]

Sabado RL, Balan S, Bhardwaj N. Dendritic cell-based immunotherapy. Cell Res. 2017;27:74-95.

[76]

Pelletier M, Maggi L, Micheletti A, et al. Evidence for a cross-talk between human neutrophils and Th17 cells. Blood. 2010;115:335-343.

[77]

Minns D, Smith KJ, Alessandrini V, et al. The neutrophil antimicrobial peptide cathelicidin promotes Th17 differentiation. Nat Commun. 2021;12:1285.

[78]

Song M, He J, Pan Q-Z, et al. Cancer-associated fibroblast-mediated cellular crosstalk supports hepatocellular carcinoma progression. Hepatology. 2021;73:1717-1735.

[79]

Weiskopf D, Weinberger B, Grubeck-Loebenstein B. The aging of the immune system. Transpl Int. 2009;22:1041-1050.

[80]

Bonaventura A, Montecucco F, Dallegri F, et al. Novel findings in neutrophil biology and their impact on cardiovascular disease. Cardiovasc Res. 2019;115:1266-1285.

[81]

Wenisch C, Patruta S, Daxböck F, Krause R, Hörl W. Effect of age on human neutrophil function. J Leukoc Biol. 2000;67:40-45.

[82]

Alonso-Fernández P, Puerto M, Maté I, Ribera JM, De La Fuente M. Neutrophils of centenarians show function levels similar to those of young adults. J Am Geriatr Soc. 2008;56:2244-2251.

[83]

Fulop T, Le Page A, Fortin C, Witkowski JM, Dupuis G, Larbi A. Cellular signaling in the aging immune system. Curr Opin Immunol. 2014;29:105-111.

[84]

Bodac A, Meylan E. Neutrophil metabolism in the cancer context. Semin Immunol. 2021;57:101583.

[85]

Gibellini L, Borella R, Santacroce E, et al. Circulating and tumor-associated neutrophils in the era of immune checkpoint inhibitors: dynamics, phenotypes, metabolism, and functions. Cancers (Basel). 2023;15:3327.

[86]

Curi R, Levada-Pires AC, Silva EBD, et al. The critical role of cell metabolism for essential neutrophil functions. Cell Physiol Biochem. 2020;54:629-647.

[87]

Jun HS, Lee YM, Song KD, Mansfield BC, Chou JY. G-CSF improves murine G6PC3-deficient neutrophil function by modulating apoptosis and energy homeostasis. Blood. 2011;117:3881-3892.

[88]

Semerad CL, Liu F, Gregory AD, Stumpf K, Link DC. G-CSF is an essential regulator of neutrophil trafficking from the bone marrow to the blood. Immunity. 2002;17:413-423.

[89]

Hannah S, Mecklenburgh K, Rahman I, et al. Hypoxia prolongs neutrophil survival in vitro. FEBS Lett. 1995;372:233-237.

[90]

Lodge KM, Cowburn AS, Li W, Condliffe AM. The impact of hypoxia on neutrophil degranulation and consequences for the host. Int J Mol Sci. 2020;21:1183.

[91]

Németh T, Sperandio M, Mócsai A. Neutrophils as emerging therapeutic targets. Nat Rev Drug Discov. 2020;19:253-275.

[92]

Hirschhorn D, Budhu S, Kraehenbuehl L, et al. T cell immunotherapies engage neutrophils to eliminate tumor antigen escape variants. Cell. 2023;186:1432-1447.e1417.

[93]

Váraljai R, Zimmer L, Al-Matary Y, et al. Interleukin 17 signaling supports clinical benefit of dual CTLA-4 and PD-1 checkpoint inhibition in melanoma. Nat Cancer. 2023;4:1292-1308.

[94]

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;22:4.

[95]

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:zcaa012.

[96]

Chan L-P, Wang L-F, Chiang F-Y, Lee K-W, Kuo P-L, Liang C-H. IL-8 promotes HNSCC progression on CXCR1/2-meidated NOD1/RIP2 signaling pathway. Oncotarget. 2016;7:61820-61831.

[97]

Buck AK, Serfling SE, Lindner T, et al. CXCR4-targeted theranostics in oncology. Eur J Nucl Med Mol Imaging. 2022;49:4133-4144.

[98]

Kashyap MK, Kumar D, Jones H, et al. Ulocuplumab (BMS-936564 /MDX1338): a fully human anti-CXCR4 antibody induces cell death in chronic lymphocytic leukemia mediated through a reactive oxygen species-dependent pathway. Oncotarget. 2016;7:2809-2822.

[99]

Bockorny B, Semenisty V, Macarulla T, et al. BL-8040, a CXCR4 antagonist, in combination with pembrolizumab and chemotherapy for pancreatic cancer: the COMBAT trial. Nat Med. 2020;26:878-885.

[100]

Fearon DT, Janowitz T. AMD3100/Plerixafor overcomes immune inhibition by the CXCL12-KRT19 coating on pancreatic and colorectal cancer cells. Br J Cancer. 2021;125:149-151.

[101]

Liu J, Wang L, Zhao F, et al. Pre-clinical development of a humanized anti-CD47 antibody with anti-cancer therapeutic potential. PLoS One. 2015;10:e0137345.

[102]

Le Saux O, Ray-Coquard I, Labidi-Galy SI. Challenges for immunotherapy for the treatment of platinum resistant ovarian cancer. Semin Cancer Biol. 2021;77:127-143.

[103]

Petrova PS, Viller NN, Wong M, et al. TTI-621 (SIRPαFc): a CD47-blocking innate immune checkpoint inhibitor with broad antitumor activity and minimal erythrocyte binding. Clin Cancer Res. 2017;23:1068-1079.

[104]

Sakamoto C, Kohara H, Inoue H, et al. Therapeutic vaccination based on side population cells transduced by the granulocyte-macrophage colony-stimulating factor gene elicits potent antitumor immunity. Cancer Gene Ther. 2017;24:165-174.

[105]

Califano JA, Khan Z, Noonan KA, et al. Tadalafil augments tumor specific immunity in patients with head and neck squamous cell carcinoma. Clin Cancer Res. 2015;21:30-38.

[106]

Burvenich IJG, Lee F-T, Guo N, et al. In vitro and in vivo evaluation of (89)Zr-DS-8273a as a theranostic for anti-death receptor 5 therapy. Theranostics. 2016;6:2225-2234.

[107]

Oh M-H, Sun I-H, Zhao L, et al. Targeting glutamine metabolism enhances tumor-specific immunity by modulating suppressive myeloid cells. J Clin Invest. 2020;130:3865-3884.

[108]

Demers M, Wagner DD. Neutrophil extracellular traps: a new link to cancer-associated thrombosis and potential implications for tumor progression. Oncoimmunology. 2013;2:e22946.

[109]

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;81:2373-2385.

[110]

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.

[111]

Huang H, Zhang H, Onuma AE, Tsung A. Neutrophil elastase and neutrophil extracellular traps in the tumor microenvironment. Tumor Microenvironment. Springer International Publishing; 2020:13-23.

[112]

Cui C, Chakraborty K, Tang XuA, et al. Neutrophil elastase selectively kills cancer cells and attenuates tumorigenesis. Cell. 2021;184:3163-3177.e3121.

[113]

Dudani JS, Warren AD, Bhatia SN. Harnessing protease activity to improve cancer care. Ann Rev Cancer Biol. 2018;2:353-376.

[114]

Chu D, Dong X, Shi X, Zhang C, Wang Z. Neutrophil-based drug delivery systems. Adv Mater. 2018;30:e1706245.

[115]

Qiao Qi, Liu X, Yang T, et al. Nanomedicine for acute respiratory distress syndrome: the latest application, targeting strategy, and rational design. Acta Pharm Sin B. 2021;11:3060-3091.

[116]

Chang Y, Cai X, Syahirah R, et al. CAR-neutrophil mediated delivery of tumor-microenvironment responsive nanodrugs for glioblastoma chemo-immunotherapy. Nat Commun. 2023;14:2266.

[117]

Elsharkasy OM, Nordin JZ, Hagey DW, et al. Extracellular vesicles as drug delivery systems: why and how? Adv Drug Deliv Rev. 2020;159:332-343.

[118]

Simpson RJ. Disruption of cultured cells by nitrogen cavitation. Cold Spring Harb Protoc. 2010;2010:pdb.prot5513.

[119]

Coffelt SB, Kersten K, Doornebal CW, et al. IL-17-producing γδ T cells and neutrophils conspire to promote breast cancer metastasis. Nature. 2015;522:345-348.

RIGHTS & PERMISSIONS

2024 The Authors. Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.

AI Summary AI Mindmap
PDF

160

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/