Extracellular vesicles in cancers: mechanisms, biomarkers, and therapeutic strategies

Yuxi Ma , Xiaohui Zhang , Cuiwei Liu , Yanxia Zhao

MedComm ›› 2024, Vol. 5 ›› Issue (12) : e70009

PDF
MedComm ›› 2024, Vol. 5 ›› Issue (12) : e70009 DOI: 10.1002/mco2.70009
REVIEW

Extracellular vesicles in cancers: mechanisms, biomarkers, and therapeutic strategies

Author information +
History +
PDF

Abstract

Extracellular vesicles (EVs) composed of various biologically active constituents, such as proteins, nucleic acids, lipids, and metabolites, have emerged as a noteworthy mode of intercellular communication. There are several categories of EVs, including exosomes, microvesicles, and apoptotic bodies, which largely differ in their mechanisms of formation and secretion. The amount of evidence indicated that changes in the EV quantity and composition play a role in multiple aspects of cancer development, such as the transfer of oncogenic signals, angiogenesis, metabolism remodeling, and immunosuppressive effects. As EV isolation technology and characteristics recognition improve, EVs are becoming more commonly used in the early diagnosis and evaluation of treatment effectiveness for cancers. Actually, EVs have sparked clinical interest in their potential use as delivery vehicles or vaccines for innovative antitumor techniques. This review will focus on the function of biological molecules contained in EVs linked to cancer progression and their participation in the intricate interrelationship within the tumor microenvironment. Furthermore, the potential efficacy of an EV-based liquid biopsy and delivery cargo for treatment will be explored. Finally, we explicitly delineate the limitations of EV-based anticancer therapies and provide an overview of the clinical trials aimed at improving EV development.

Keywords

biomarkers / cancer / cargo / challenges / delivery vehicles / exosomes / extracellular vesicles / therapy resistance / tumor microenvironment

Cite this article

Download citation ▾
Yuxi Ma, Xiaohui Zhang, Cuiwei Liu, Yanxia Zhao. Extracellular vesicles in cancers: mechanisms, biomarkers, and therapeutic strategies. MedComm, 2024, 5(12): e70009 DOI:10.1002/mco2.70009

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Anand S, Samuel M, Kumar S, Mathivanan S. Ticket to a bubble ride: cargo sorting into exosomes and extracellular vesicles. BBA-Proteins Proteom. 2019; 1867(12): 140203.

[2]

Sundararajan V, Sarkar FH, Ramasamy TS. The versatile role of exosomes in cancer progression: diagnostic and therapeutic implications. Cell Oncol. 2018; 41: 223-252.

[3]

Kok VC, Yu CC. Cancer-derived exosomes: their role in cancer biology and biomarker development. Int J Nanomedicine. 2020; 15: 8019-8036.

[4]

Bahrami A, Binabaj MM, Ferns GA. Exosomes: emerging modulators of signal transduction in colorectal cancer from molecular understanding to clinical application. Biomed Pharmacother. 2021; 141: 111882.

[5]

Hsu M-T, Wang Y-K, Tseng YJ. Exosomal proteins and lipids as potential biomarkers for lung cancer diagnosis, prognosis, and treatment. Cancers. 2022; 14(3): 732.

[6]

Al-Nedawi K, Meehan B, Kerbel RS, Allison AC, Rak J. Endothelial expression of autocrine VEGF upon the uptake of tumor-derived microvesicles containing oncogenic EGFR. Proc Natl Acad Sci USA. 2009; 106(10): 3794-3799.

[7]

Liu Y, Cao X. Immunosuppressive cells in tumor immune escape and metastasis. J Mol Med. 2016; 94: 509-522.

[8]

Taghikhani A, Farzaneh F, Sharifzad F, Mardpour S, Ebrahimi M, Hassan ZM. Engineered tumor-derived extracellular vesicles: potentials in cancer immunotherapy. Front Immunol. 2020; 11: 221.

[9]

Ahmadi M, Abbasi R, Rezaie J. Tumor immune escape: extracellular vesicles roles and therapeutics application. Cell Commun Signal. 2024; 22(1): 9.

[10]

Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011; 144(5): 646-674.

[11]

Fu X, Song J, Yan W, Downs BM, Wang W, Li J. The biological function of tumor-derived extracellular vesicles on metabolism. Cell Commun Signal. 2023; 21(1): 150.

[12]

Mashouri L, Yousefi H, Aref AR, Ahadi AM, Molaei F, Alahari SK. Exosomes: composition, biogenesis, and mechanisms in cancer metastasis and drug resistance. Mol Cancer. 2019; 18(1): 75.

[13]

Catalano M, O’Driscoll L. Inhibiting extracellular vesicles formation and release: a review of EV inhibitors. J Extracell Vesicles. 2020; 9(1): 1703244.

[14]

Fujita Y, Yoshioka Y, Ochiya T. Extracellular vesicle transfer of cancer pathogenic components. Cancer Sci. 2016; 107(4): 385-390.

[15]

Xu R, Rai A, Chen M, Suwakulsiri W, Greening DW, Simpson RJ. Extracellular vesicles in cancer—implications for future improvements in cancer care. Nat Rev Clin Oncol. 2018; 15(10): 617-638.

[16]

Chulpanova DS, Kitaeva KV, James V, Rizvanov AA, Solovyeva VV. Therapeutic prospects of extracellular vesicles in cancer treatment. Front Immunol. 2018; 9: 1534.

[17]

Pan B-T, Teng K, Wu C, Adam M, Johnstone RM. Electron microscopic evidence for externalization of the transferrin receptor in vesicular form in sheep reticulocytes. J Cell Biol. 1985; 101(3): 942-948.

[18]

Budnik V, Ruiz-Cañada C, Wendler F. Extracellular vesicles round off communication in the nervous system. Nat Rev Neurosci. 2016; 17(3): 160-172.

[19]

Ludwig A-K, De Miroschedji K, Doeppner TR, et al. Precipitation with polyethylene glycol followed by washing and pelleting by ultracentrifugation enriches extracellular vesicles from tissue culture supernatants in small and large scales. J Extracell Vesicles. 2018; 7(1): 1528109.

[20]

Tkach M, Théry C. Communication by extracellular vesicles: where we are and where we need to go. Cell. 2016; 164(6): 1226-1232.

[21]

Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science. 2020; 367(6478): eaau6977.

[22]

McAndrews KM, Kalluri R. Mechanisms associated with biogenesis of exosomes in cancer. Mol Cancer. 2019; 18(1): 1-11.

[23]

Kahlert C, Kalluri R. Exosomes in tumor microenvironment influence cancer progression and metastasis. J Mol Med. 2013; 91(4): 431-437.

[24]

Henne WM, Buchkovich NJ, Emr SD. The ESCRT pathway. Dev Cell. 2011; 21(1): 77-91.

[25]

Keller S, Sanderson MP, Stoeck A, Altevogt P. Exosomes: from biogenesis and secretion to biological function. Immunol Lett. 2006; 107(2): 102-108.

[26]

Bicalho B, Holovati JL, Acker JP. Phospholipidomics reveals differences in glycerophosphoserine profiles of hypothermically stored red blood cells and microvesicles. BBA-Biomembranes. 2013; 1828(2): 317-326.

[27]

Teng F, Fussenegger M. Shedding light on extracellular vesicle biogenesis and bioengineering. Adv Sci. 2021; 8(1): 2003505.

[28]

Peinado H, Alečković M, Lavotshkin S, et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med. 2012; 18(6): 883-891.

[29]

Ho J, Chaiswing L, St Clair DK. Extracellular vesicles and cancer therapy: insights into the role of oxidative stress. Antioxidants. 2022; 11(6): 1194.

[30]

Morimoto K, Ishitobi J, Noguchi K, et al. Extracellular microvesicles modified with arginine-rich peptides for active macropinocytosis induction and delivery of therapeutic molecules. ACS Appl Mater Interfaces. 2024; 16(14): 17069–17079.

[31]

Atkin-Smith GK, Tixeira R, Paone S, et al. A novel mechanism of generating extracellular vesicles during apoptosis via a beads-on-a-string membrane structure. Nat Commun. 2015; 6(1): 1-10.

[32]

Charras GT, Yarrow JC, Horton MA, Mahadevan L, Mitchison T. Non-equilibration of hydrostatic pressure in blebbing cells. Nature. 2005; 435(7040): 365-369.

[33]

Barros L, Kanaseki T, Sabirov R, et al. Apoptotic and necrotic blebs in epithelial cells display similar neck diameters but different kinase dependency. Cell Death Differ. 2003; 10(6): 687-697.

[34]

Croft DR, Coleman ML, Li S, et al. Actin-myosin–based contraction is responsible for apoptotic nuclear disintegration. J Cell Biol. 2005; 168(2): 245-255.

[35]

Xu X, Lai Y, Hua Z-C. Apoptosis and apoptotic body: disease message and therapeutic target potentials. Bioscience Rep. 2019; 39(1): BSR20180992.

[36]

Kranich J, Krautler NJ, Falsig J, et al. Engulfment of cerebral apoptotic bodies controls the course of prion disease in a mouse strain–dependent manner. J Exp Med. 2010; 207(10): 2271-2281.

[37]

Wiley RD, Gummuluru S. Immature dendritic cell-derived exosomes can mediate HIV-1 trans infection. Proc Natl Acad Sci USA. 2006; 103(3): 738-743.

[38]

Record M, Subra C, Silvente-Poirot S, Poirot M. Exosomes as intercellular signalosomes and pharmacological effectors. Biochem Pharmacol. 2011; 81(10): 1171-1182.

[39]

Ludwig A-K, Giebel B. Exosomes: small vesicles participating in intercellular communication. Int J Biochem Cell B. 2012; 44(1): 11-15.

[40]

Nazarenko I, Rana S, Baumann A, et al. Cell surface tetraspanin Tspan8 contributes to molecular pathways of exosome-induced endothelial cell activation. Cancer Res. 2010; 70(4): 1668-1678.

[41]

Mathieu M, Martin-Jaular L, Lavieu G, Théry C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat cell Biol 2019; 21(1): 9-17.

[42]

Mimeault M, Batra SK. Molecular biomarkers of cancer stem/progenitor cells associated with progression, metastases, and treatment resistance of aggressive cancers. Cancer Epidem Biomar. 2014; 23(2): 234-254.

[43]

Dai J, Su Y, Zhong S, et al. Exosomes: key players in cancer and potential therapeutic strategy. Signal Transduct Tar 2020; 5(1): 145.

[44]

Andreu Z, Yáñez-Mó M. Tetraspanins in extracellular vesicle formation and function. Front Immunol. 2014; 5: 442.

[45]

Phanie Rieu S, GeÂminard C, Rabesandratana H, Sainte-Marie J, Vidal M. Exosomes released during reticulocyte maturation bind to fibronectin via integrin a4b1. Eur J Biochem. 2000; 267: 583–590

[46]

Segura E, Nicco C, Lombard B, et al. ICAM-1 on exosomes from mature dendritic cells is critical for efficient naive T-cell priming. Blood. 2005; 106(1): 216-223.

[47]

Lee YJ, Shin KJ, Chae YC. Regulation of cargo selection in exosome biogenesis and its biomedical applications in cancer. Exp Mol Med. 2024; 56(4): 877-889.

[48]

Mazurov D, Barbashova L, Filatov A. Tetraspanin protein CD 9 interacts with metalloprotease CD 10 and enhances its release via exosomes. FEBS J. 2013; 280(5): 1200-1213.

[49]

Taha EA, Ono K, Eguchi T. Roles of extracellular HSPs as biomarkers in immune surveillance and immune evasion. Int J Mol Sci. 2019; 20(18): 4588.

[50]

Arnold P, Lückstädt W, Li W, et al. Joint reconstituted signaling of the IL-6 receptor via extracellular vesicles. Cells. 2020; 9(5): 1307.

[51]

Chen G, Huang AC, Zhang W, et al. Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature. 2018; 560(7718): 382-386.

[52]

Blanchard N, Lankar D, Faure F, et al. TCR activation of human T cells induces the production of exosomes bearing the TCR/CD3/ζ complex. J Immunol. 2002; 168(7): 3235-3241.

[53]

Phuyal S, Hessvik NP, Skotland T, Sandvig K, Llorente A. Regulation of exosome release by glycosphingolipids and flotillins. FEBS J. 2014; 281(9): 2214-2227.

[54]

Diamandis EP, Plebani M. Glypican-1 as a highly sensitive and specific pancreatic cancer biomarker. Clin Chem Lab Med. 2016; 54(1): e1-e2.

[55]

Juan T, Fürthauer M. Biogenesis and function of ESCRT-dependent extracellular vesicles. Semin Cell Dev Biol. 2018: 66-77.

[56]

Hoshino A, Kim HS, Bojmar L, et al. Extracellular vesicle and particle biomarkers define multiple human cancers. Cell. 2020; 182(4): 1044-1061. e18.

[57]

Kim KM, Abdelmohsen K, Mustapic M, Kapogiannis D, Gorospe M. RNA in extracellular vesicles. WIRES RNA. 2017; 8(4): e1413.

[58]

Villarroya-Beltri C, Gutiérrez-Vázquez C, Sánchez-Cabo F, et al. Sumoylated hnRNPA2B1 controls the sorting of miRNAs into exosomes through binding to specific motifs. Nat Commun. 2013; 4(1): 2980.

[59]

Chen C, Luo Y, He W, et al. Exosomal long noncoding RNA LNMAT2 promotes lymphatic metastasis in bladder cancer. J Clin Invest. 2020; 130(1): 404-421.

[60]

Yang F, Ning Z, Ma L, et al. Exosomal miRNAs and miRNA dysregulation in cancer-associated fibroblasts. Mol Cancer. 2017; 16: 1-10.

[61]

Fortunato O, Gasparini P, Boeri M, Sozzi G. Exo-miRNAs as a new tool for liquid biopsy in lung cancer. Cancers. 2019; 11(6): 888.

[62]

Huang X, Yuan T, Liang M, et al. Exosomal miR-1290 and miR-375 as prognostic markers in castration-resistant prostate cancer. Eur Urol. 2015; 67(1): 33-41.

[63]

Baroni S, Romero-Cordoba S, Plantamura I, et al. Exosome-mediated delivery of miR-9 induces cancer-associated fibroblast-like properties in human breast fibroblasts. Cell Death Dis. 2016; 7(7): e2312-e2312.

[64]

Endzeliņš E, Berger A, Melne V, et al. Detection of circulating miRNAs: comparative analysis of extracellular vesicle-incorporated miRNAs and cell-free miRNAs in whole plasma of prostate cancer patients. BMC cancer. 2017; 17: 1-13.

[65]

Shi Y, Wang Z, Zhu X, et al. Exosomal miR-1246 in serum as a potential biomarker for early diagnosis of gastric cancer. Int J Clin Oncol. 2020; 25: 89-99.

[66]

Zhao W, Shan B, He D, et al. Recent progress in characterizing long noncoding RNAs in cancer drug resistance. J Cancer. 2019; 10(26): 6693.

[67]

Liu C, Lu C, Yixi L, et al. Exosomal Linc00969 induces trastuzumab resistance in breast cancer by increasing HER-2 protein expression and mRNA stability by binding to HUR. Breast Cancer Res. 2023; 25(1): 124.

[68]

Li J, Li Z, Zheng W, et al. Lnc RNA-ATB: an indispensable cancer-related long noncoding RNA. Cell Proliferat. 2017; 50(6): e12381.

[69]

Kristensen LS, Andersen MS, Stagsted LV, Ebbesen KK, Hansen TB, Kjems J. The biogenesis, biology and characterization of circular RNAs. Nat Rev Genet. 2019; 20(11): 675-691.

[70]

Li J, Zhang G, Liu C-G, et al. The potential role of exosomal circRNAs in the tumor microenvironment: insights into cancer diagnosis and therapy. Theranostics. 2022; 12(1): 87.

[71]

Wang J, Zhao X, Wang Y, et al. circRNA-002178 act as a ceRNA to promote PDL1/PD1 expression in lung adenocarcinoma. Cell Death Dis. 2020; 11(1): 32.

[72]

Lu J, Wang Y-h, Yoon C, et al. Circular RNA circ-RanGAP1 regulates VEGFA expression by targeting miR-877–3p to facilitate gastric cancer invasion and metastasis. Cancer Lett. 2020; 471: 38-48.

[73]

Wang X, Zhang H, Yang H, et al. Exosome-delivered circRNA promotes glycolysis to induce chemoresistance through the miR-122-PKM2 axis in colorectal cancer. Mol Oncol. 2020; 14(3): 539-555.

[74]

Sork H, Corso G, Krjutskov K, et al. Heterogeneity and interplay of the extracellular vesicle small RNA transcriptome and proteome. Sci Rep. 2018; 8(1): 10813.

[75]

Fabbiano F, Corsi J, Gurrieri E, Trevisan C, Notarangelo M, D’Agostino VG. RNA packaging into extracellular vesicles: an orchestra of RNA-binding proteins? J Extracell Vesicles. 2020; 10(2): e12043.

[76]

Stoorvogel W. Functional transfer of microRNA by exosomes. Blood, J Am Soc Hematol. 2012; 119(3): 646-648.

[77]

Cai J, Wu G, Jose PA, Zeng C. Functional transferred DNA within extracellular vesicles. Exp Cell Res. 2016; 349(1): 179-183.

[78]

Vagner T, Spinelli C, Minciacchi VR, et al. Large extracellular vesicles carry most of the tumour DNA circulating in prostate cancer patient plasma. J Extracell Vesicles. 2018; 7(1): 1505403.

[79]

Tatischeff I. Extracellular vesicle-DNA: the next liquid biopsy biomarker for early cancer diagnosis? Cancers. 2023; 15(5): 1456.

[80]

Kalluri R, LeBleu VS. Discovery of double-stranded genomic DNA in circulating exosomes. Cold Spring Harbor Laboratory Press. 2016; 81: 275-280.

[81]

Hagey DW, Kordes M, Görgens A, et al. Extracellular vesicles are the primary source of blood-borne tumour-derived mutant KRAS DNA early in pancreatic cancer. J Extracell Vesicles. 2021; 10(12): e12142.

[82]

Yokoi A, Villar-Prados A, Oliphint PA, et al. Mechanisms of nuclear content loading to exosomes. Sci Adv. 2019; 5(11): eaax8849.

[83]

Llorente A, Skotland T, Sylvänne T, et al. Molecular lipidomics of exosomes released by PC-3 prostate cancer cells. BBA-Mol Cell Biol L. 2013; 1831(7): 1302-1309.

[84]

Haraszti RA, Didiot M-C, Sapp E, et al. High-resolution proteomic and lipidomic analysis of exosomes and microvesicles from different cell sources. J Extracell Vesicles. 2016; 5(1): 32570.

[85]

Skotland T, Ekroos K, Kauhanen D, et al. Molecular lipid species in urinary exosomes as potential prostate cancer biomarkers. Eur J Cancer. 2017; 70: 122-132.

[86]

Skotland T, Sagini K, Sandvig K, Llorente A. An emerging focus on lipids in extracellular vesicles. Adv Drug deliver Rev. 2020; 159: 308-321.

[87]

Zebrowska A, Skowronek A, Wojakowska A, Widlak P, Pietrowska M. Metabolome of exosomes: focus on vesicles released by cancer cells and present in human body fluids. Int J Mol Sci. 2019; 20(14): 3461.

[88]

Wojakowska A, Zebrowska A, Skowronek A, et al. Metabolic profiles of whole serum and serum-derived exosomes are different in head and neck cancer patients treated by radiotherapy. J Pers Med. 2020; 10(4): 229.

[89]

Lazar I, Clement E, Attane C, Muller C, Nieto L. A new role for extracellular vesicles: how small vesicles can feed tumors’ big appetite. J Lipid Res. 2018; 59(10): 1793-1804.

[90]

Fridman ES, Ginini L, Gil Z. The role of extracellular vesicles in metabolic reprogramming of the tumor microenvironment. Cells. 2022; 11(9): 1433.

[91]

Zhu Q, Huang Y, Yang Q, Liu F. Recent technical advances to study metabolomics of extracellular vesicles. Microchem J. 2021; 171: 106816.

[92]

Talukdar S, Emdad L, Das SK, Fisher PB. EGFR: an essential receptor tyrosine kinase-regulator of cancer stem cells. Adv Cancer Res. 2020; 147: 161-188.

[93]

Al-Nedawi K, Meehan B, Micallef J, et al. Intercellular transfer of the oncogenic receptor EGFRvIII by microvesicles derived from tumour cells. Nat Cell biol. 2008; 10(5): 619-624.

[94]

Skog J, Würdinger T, Van Rijn S, et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol. 2008; 10(12): 1470-1476.

[95]

Zhang H, Deng T, Liu R, et al. Exosome-delivered EGFR regulates liver microenvironment to promote gastric cancer liver metastasis. Nat Commun. 2017; 8(1): 15016.

[96]

Wu S, Luo M, To KK, et al. Intercellular transfer of exosomal wild type EGFR triggers osimertinib resistance in non-small cell lung cancer. Mol Cancer. 2021; 20: 1-17.

[97]

Jin H, Liu P, Wu Y, et al. Exosomal zinc transporter ZIP4 promotes cancer growth and is a novel diagnostic biomarker for pancreatic cancer. Cancer Sci. 2018; 109(9): 2946-2956.

[98]

Demory Beckler M, Higginbotham JN, Franklin JL, et al. Proteomic analysis of exosomes from mutant KRAS colon cancer cells identifies intercellular transfer of mutant KRAS. Mol Cell Proteomics. 2013; 12(2): 343-55.

[99]

Qiao Z, Zhang Y, Ge M, et al. Cancer cell derived small extracellular vesicles contribute to recipient cell metastasis through promoting hgf/c-met pathway*[s]. Mol Cell Proteomics. 2019; 18(8): 1619-1629.

[100]

Webber J, Steadman R, Mason MD, Tabi Z, Clayton A. Cancer exosomes trigger fibroblast to myofibroblast differentiation. Cancer Res. 2010; 70(23): 9621-30.

[101]

Ringuette Goulet C, Bernard G, Tremblay S, Chabaud S, Bolduc S, Pouliot F. Exosomes induce fibroblast differentiation into cancer-associated fibroblasts through TGFβ signaling. Mol Cancer Res. 2018; 16(7): 1196-1204.

[102]

Fang T, Lv H, Lv G, et al. Tumor-derived exosomal miR-1247-3p induces cancer-associated fibroblast activation to foster lung metastasis of liver cancer. Nat Commun. 2018; 9(1): 191.

[103]

Vu LT, Peng B, Zhang DX, et al. Tumor-secreted extracellular vesicles promote the activation of cancer-associated fibroblasts via the transfer of microRNA-125b. J Extracell Vesicles. 2019; 8(1): 1599680.

[104]

Sahai E, Astsaturov I, Cukierman E, et al. A framework for advancing our understanding of cancer-associated fibroblasts. Nat Rev Cancer. 2020; 20(3): 174-186.

[105]

Olumi AF, Grossfeld GD, Hayward SW, Carroll PR, Tlsty TD, Cunha GR. Carcinoma-associated fibroblasts direct tumor progression of initiated human prostatic epithelium. Cancer Res. 1999; 59: 5002–5011.

[106]

Dourado MR, Korvala J, Åström P, et al. Extracellular vesicles derived from cancer-associated fibroblasts induce the migration and invasion of oral squamous cell carcinoma. J Extracell Vesicles. 2019; 8(1): 1578525.

[107]

Luga V, Zhang L, Viloria-Petit AM, et al. Exosomes mediate stromal mobilization of autocrine Wnt-PCP signaling in breast cancer cell migration. Cell. 2012; 151(7): 1542-1556.

[108]

Zhang H, Deng T, Liu R, et al. CAF secreted miR-522 suppresses ferroptosis and promotes acquired chemo-resistance in gastric cancer. Mol Cancer. 2020; 19: 1-17.

[109]

Zhao H, Yang L, Baddour J, et al. Tumor microenvironment derived exosomes pleiotropically modulate cancer cell metabolism. Elife. 2016; 5: e10250.

[110]

Deng H, Sun C, Sun Y, et al. Lipid, protein, and microRNA composition within mesenchymal stem cell-derived exosomes. Cell Reprogram. 2018; 20(3): 178-186.

[111]

Seo M, Kim SM, Woo EY, et al. Stemness-attenuating miR-503-3p as a paracrine factor to regulate growth of cancer stem cells. Stem Cells Int. 2018; 2018(1): 4851949.

[112]

Sharma A. Role of stem cell derived exosomes in tumor biology. Int J Cancer. 2018; 142(6): 1086-1092.

[113]

Figueroa J, Phillips LM, Shahar T, et al. Exosomes from glioma-associated mesenchymal stem cells increase the tumorigenicity of glioma stem-like cells via transfer of miR-1587. Cancer Res. 2017; 77(21): 5808-5819.

[114]

Pakravan K, Babashah S, Sadeghizadeh M, et al. MicroRNA-100 shuttled by mesenchymal stem cell-derived exosomes suppresses in vitro angiogenesis through modulating the mTOR/HIF-1α/VEGF signaling axis in breast cancer cells. Cell Oncol. 2017; 40: 457-470.

[115]

Sun Z, Wang L, Dong L, Wang X. Emerging role of exosome signalling in maintaining cancer stem cell dynamic equilibrium. J Cell Mol Med. 2018; 22(8): 3719-3728.

[116]

Xu J, Liao K, Zhou W. Exosomes regulate the transformation of cancer cells in cancer stem cell homeostasis. Stem cells Int. 2018; 2018(1): 4837370.

[117]

Li W, Zhang L, Guo B, et al. Exosomal FMR1-AS1 facilitates maintaining cancer stem-like cell dynamic equilibrium via TLR7/NFκB/c-Myc signaling in female esophageal carcinoma. Mol Cancer. 2019; 18: 1-15.

[118]

Wang J, Zheng Y, Zhao M. Exosome-based cancer therapy: implication for targeting cancer stem cells. Front Pharmacol. 2017; 7: 533.

[119]

Torreggiani E, Roncuzzi L, Perut F, Zini N, Baldini N. Multimodal transfer of MDR by exosomes in human osteosarcoma. Int J Oncol. 2016; 49(1): 189-196.

[120]

Lv M-M, Zhu X-Y, Chen W-X, et al. Exosomes mediate drug resistance transfer in MCF-7 breast cancer cells and a probable mechanism is delivery of P-glycoprotein. Tumor Biol. 2014; 35: 10773-10779.

[121]

Zhang F-F, Zhu Y-F, Zhao Q-N, et al. Microvesicles mediate transfer of P-glycoprotein to paclitaxel-sensitive A2780 human ovarian cancer cells, conferring paclitaxel-resistance. Eur J Pharmacol. 2014; 738: 83-90.

[122]

Corcoran C, Rani S, O’Brien K, et al. Docetaxel-resistance in prostate cancer: evaluating associated phenotypic changes and potential for resistance transfer via exosomes. PLoS One. 2012; 7(12): e50999.

[123]

Boelens MC, Wu TJ, Nabet BY, et al. Exosome transfer from stromal to breast cancer cells regulates therapy resistance pathways. Cell. 2014; 159(3): 499-513.

[124]

Dong G, Mao Q, Xia W, et al. PKM2 and cancer: the function of PKM2 beyond glycolysis. Oncol Lett. 2016; 11(3): 1980-1986.

[125]

Iqbal MA, Gupta V, Gopinath P, Mazurek S, Bamezai RN. Pyruvate kinase M2 and cancer: an updated assessment. FEBS Lett. 2014; 588(16): 2685-2692.

[126]

Yang W, Lu Z. Pyruvate kinase M2 at a glance. J Cell Sci. 2015; 128(9): 1655-1660.

[127]

Li Y, Zhao Z, Liu W, Li X. SNHG3 functions as miRNA sponge to promote breast cancer cells growth through the metabolic reprogramming. Appl Biochem Biotechnol. 2020; 191(3): 1084-1099.

[128]

Yi H, Zheng X, Song J, Shen R, Su Y, Lin D. Exosomes mediated pentose phosphate pathway in ovarian cancer metastasis: a proteomics analysis. Int J Clin Exp Patho. 2015; 8(12): 15719.

[129]

Pavlova NN, Thompson CB. The emerging hallmarks of cancer metabolism. Cell Metab. 2016; 23(1): 27-47.

[130]

Reinfeld BI, Madden MZ, Wolf MM, et al. Cell-programmed nutrient partitioning in the tumour microenvironment. Nature. 2021; 593(7858): 282-288.

[131]

Wang M. Preferential glutamine uptake in cancer cells. Nat Rev Nephrol. 2021; 17(6): 368-368.

[132]

Yang C, Wu S, Mou Z, et al. Exosome-derived circTRPS1 promotes malignant phenotype and CD8+ T cell exhaustion in bladder cancer microenvironments. Mol Ther. 2022; 30(3): 1054-1070.

[133]

Wang W, Zhu N, Yan T, et al. The crosstalk: exosomes and lipid metabolism. Cell Commun Signal. 2020; 18(1): 119.

[134]

Wang X, Li H, Lu X, et al. Melittin-induced long non-coding RNA NONHSAT105177 inhibits proliferation and migration of pancreatic ductal adenocarcinoma. Cell Death Dis. 2018; 9(10): 940.

[135]

Lazar I, Clement E, Dauvillier S, et al. Adipocyte exosomes promote melanoma aggressiveness through fatty acid oxidation: a novel mechanism linking obesity and cancer. Cancer Res. 2016; 76(14): 4051-7.

[136]

Faict S, Oudaert I, D’Auria L, et al. The transfer of sphingomyelinase contributes to drug resistance in multiple myeloma. Cancers (Basel). 2019; 11(12): 1823.

[137]

Sansone P, Savini C, Kurelac I, et al. Packaging and transfer of mitochondrial DNA via exosomes regulate escape from dormancy in hormonal therapy-resistant breast cancer. Proc Natl Acad Sci USA. 2017; 114(43): E9066-E9075.

[138]

Tao L, Xu C, Shen W, et al. HIPK3 inhibition by exosomal hsa-miR-101-3p is related to metabolic reprogramming in colorectal cancer. Front Oncol. 2022; 11: 5513.

[139]

Yan W, Wu X, Zhou W, et al. Cancer-cell-secreted exosomal miR-105 promotes tumour growth through the MYC-dependent metabolic reprogramming of stromal cells. Nat Cell Biol. 2018; 20(5): 597-609.

[140]

Rai A, Greening DW, Chen M, Xu R, Ji H, Simpson RJ. Exosomes derived from human primary and metastatic colorectal cancer cells contribute to functional heterogeneity of activated fibroblasts by reprogramming their proteome. Proteomics. 2019; 19(8): 1800148.

[141]

Short NJ, Rytting ME, Cortes JE. Acute myeloid leukaemia. Lancet. 2018; 392(10147): 593-606.

[142]

Passaro D, Di Tullio A, Abarrategi A, et al. Increased vascular permeability in the bone marrow microenvironment contributes to disease progression and drug response in acute myeloid leukemia. Cancer Cell. 2017; 32(3): 324-341. e6.

[143]

Wang B, Wang X, Hou D, et al. Exosomes derived from acute myeloid leukemia cells promote chemoresistance by enhancing glycolysis-mediated vascular remodeling. J Cell Physiol. 2019; 234(7): 10602-10614.

[144]

Wang S, Li X, Xu M, Wang J, Zhao RC. Reduced adipogenesis after lung tumor exosomes priming in human mesenchymal stem cells via TGFbeta signaling pathway. Mol Cell Biochem. 2017; 435(1-2): 59-66.

[145]

Dirat B, Bochet L, Dabek M, et al. Cancer-associated adipocytes exhibit an activated phenotype and contribute to breast cancer invasion. Cancer Res. 2011; 71(7): 2455-2465.

[146]

Lapeire L, Hendrix A, Lambein K, et al. Cancer-associated adipose tissue promotes breast cancer progression by paracrine oncostatin m and jak/stat3 signalingparacrine oncostatin M promotes breast cancer progression. Cancer Res. 2014; 74(23): 6806-6819.

[147]

Sagar G, Sah RP, Javeed N, et al. Pathogenesis of pancreatic cancer exosome-induced lipolysis in adipose tissue. Gut. 2016; 65(7): 1165-74.

[148]

Hu W, Ru Z, Xiao W, et al. Adipose tissue browning in cancer-associated cachexia can be attenuated by inhibition of exosome generation. Biochem Biophys Res Commun. 2018; 506(1): 122-129.

[149]

Sosnowska A, Czystowska-Kuzmicz M, Golab J. Extracellular vesicles released by ovarian carcinoma contain arginase 1 that mitigates antitumor immune response. Oncoimmunology. 2019; 8(11): e1655370.

[150]

Bland CL, Byrne-Hoffman CN, Fernandez A, Rellick SL, Deng W, Klinke DJ. Exosomes derived from B16F0 melanoma cells alter the transcriptome of cytotoxic T cells that impacts mitochondrial respiration. FEBS J. 2018; 285(6): 1033-1050.

[151]

Xu F, Wang X, Huang Y, et al. Prostate cancer cell-derived exosomal IL-8 fosters immune evasion by disturbing glucolipid metabolism of CD8+ T cell. Cell Rep. 2023; 42(11): 113424.

[152]

Xiang X, Poliakov A, Liu C, et al. Induction of myeloid-derived suppressor cells by tumor exosomes. Int J Cancer. 2009; 124(11): 2621-33.

[153]

Morrissey SM, Zhang F, Ding C, et al. Tumor-derived exosomes drive immunosuppressive macrophages in a pre-metastatic niche through glycolytic dominant metabolic reprogramming. Cell Metab. 2021; 33(10): 2040-2058. e10.

[154]

Kim J, Hong SW, Kim S, et al. Cyclooxygenase-2 expression is induced by celecoxib treatment in lung cancer cells and is transferred to neighbor cells via exosomes. Int J Oncol. 2018; 52(2): 613-620.

[155]

Wong SK, Mohamad N-V, Giaze TR, Chin K-Y, Mohamed N, Ima-Nirwana S. Prostate cancer and bone metastases: the underlying mechanisms. Int J Mol Sci. 2019; 20(10): 2587.

[156]

Dai J, Escara-Wilke J, Keller JM, et al. Primary prostate cancer educates bone stroma through exosomal pyruvate kinase M2 to promote bone metastasis. J Exp Med. 2019; 216(12): 2883-2899.

[157]

Liu Z-L, Chen H-H, Zheng L-L, Sun L-P, Shi L. Angiogenic signaling pathways and anti-angiogenic therapy for cancer. Signal Transduct Tar. 2023; 8(1): 198.

[158]

Treps L, Perret R, Edmond S, Ricard D, Gavard J. Glioblastoma stem-like cells secrete the pro-angiogenic VEGF-A factor in extracellular vesicles. J Extracell Vesicles. 2017; 6(1): 1359479.

[159]

Ko SY, Lee W, Kenny HA, et al. Cancer-derived small extracellular vesicles promote angiogenesis by heparin-bound, bevacizumab-insensitive VEGF, independent of vesicle uptake. Commun Biol. 2019; 2(1): 386.

[160]

Lang H-L, Hu G-W, Zhang B, et al. Glioma cells enhance angiogenesis and inhibit endothelial cell apoptosis through the release of exosomes that contain long non-coding RNA CCAT2. Oncol Rep. 2017; 38(2): 785-798.

[161]

Zeng Z, Li Y, Pan Y, et al. Cancer-derived exosomal miR-25-3p promotes pre-metastatic niche formation by inducing vascular permeability and angiogenesis. Nat Commun. 2018; 9(1): 5395.

[162]

Zhuang G, Wu X, Jiang Z, et al. Tumour-secreted miR-9 promotes endothelial cell migration and angiogenesis by activating the JAK-STAT pathway. EMBO J. 2012; 31(17): 3513-3523.

[163]

Xu Y, Leng K, Yao Y, et al. A circular RNA, cholangiocarcinoma-associated circular RNA 1, contributes to cholangiocarcinoma progression, induces angiogenesis, and disrupts vascular endothelial barriers. Hepatology. 2021; 73(4): 1419-1435.

[164]

Ludwig N, Yerneni SS, Razzo BM, Whiteside TL. Exosomes from HNSCC promote angiogenesis through reprogramming of endothelial cells. Mol Cancer Res. 2018; 16(11): 1798-1808.

[165]

Ludwig N, Yerneni SS, Azambuja JH, et al. TGFβ+ small extracellular vesicles from head and neck squamous cell carcinoma cells reprogram macrophages towards a pro-angiogenic phenotype. J Extracell Vesicles. 2022; 11(12): 12294.

[166]

Zhu H, Gu X, Xia L, et al. A novel TGFβ trap blocks chemotherapeutics-induced TGFβ1 signaling and enhances their anticancer activity in gynecologic cancers. Clin Cancer Res. 2018; 24(12): 2780-2793.

[167]

Zhang X, Wang Y, Wang X, et al. Extracellular vesicles-encapsulated microRNA-10a-5p shed from cancer-associated fibroblast facilitates cervical squamous cell carcinoma cell angiogenesis and tumorigenicity via Hedgehog signaling pathway. Cancer gene Ther. 2021; 28(5): 529-542.

[168]

Miaomiao S, Xiaoqian W, Yuwei S, et al. Cancer-associated fibroblast-derived exosome microRNA-21 promotes angiogenesis in multiple myeloma. Sci Rep. 2023; 13(1): 9671.

[169]

Giusti I, Delle Monache S, Di Francesco M, et al. From glioblastoma to endothelial cells through extracellular vesicles: messages for angiogenesis. Tumor Biol 2016; 37: 12743-12753.

[170]

Johan S, Tom W, Sjoerd R, et al. Glioblastoma microvesicles transport RNA and proteins that promote tumor growth and provide diagnostic biomarkers. Neurol Surg. 2009; 37(8): 823-823.

[171]

You Y, Shan Y, Chen J, et al. Matrix metalloproteinase 13-containing exosomes promote nasopharyngeal carcinoma metastasis. Cancer Sci. 2015; 106(12): 1669-1677.

[172]

Chan YK, Zhang H, Liu P, et al. Proteomic analysis of exosomes from nasopharyngeal carcinoma cell identifies intercellular transfer of angiogenic proteins. Int J Cancer. 2015; 137(8): 1830-1841.

[173]

Maji S, Chaudhary P, Akopova I, et al. Exosomal annexin II promotes angiogenesis and breast cancer metastasis. Mol Cancer Res. 2017; 15(1): 93-105.

[174]

Beckham CJ, Olsen J, Yin P-N, et al. Bladder cancer exosomes contain EDIL-3/Del1 and facilitate cancer progression. J Urol. 2014; 192(2): 583-592.

[175]

Wang J, De Veirman K, Faict S, et al. Multiple myeloma exosomes establish a favourable bone marrow microenvironment with enhanced angiogenesis and immunosuppression. J Pathol. 2016; 239(2): 162-173.

[176]

Wilson CM, Naves T, Vincent F, et al. Sortilin mediates the release and transfer of exosomes in concert with two tyrosine kinase receptors. J Cell Sci. 2014; 127(18): 3983-3997.

[177]

Zimna A, Kurpisz M. Hypoxia-inducible factor-1 in physiological and pathophysiological angiogenesis: applications and therapies. BioMed Res Int. 2015; 2015(1): 549412.

[178]

Ravi R, Mookerjee B, Bhujwalla ZM, et al. Regulation of tumor angiogenesis by p53-induced degradation of hypoxia-inducible factor 1α. Gene Dev. 2000; 14(1): 34-44.

[179]

Aga M, Bentz GL, Raffa S, et al. Exosomal HIF1α supports invasive potential of nasopharyngeal carcinoma-associated LMP1-positive exosomes. Oncogene. 2014; 33(37): 4613-4622.

[180]

de Jong OG, van Balkom BW, Gremmels H, Verhaar MC. Exosomes from hypoxic endothelial cells have increased collagen crosslinking activity through up-regulation of lysyl oxidase-like 2. J Cell Mol Med. 2016; 20(2): 342-350.

[181]

Kucharzewska P, Christianson HC, Welch JE, et al. Exosomes reflect the hypoxic status of glioma cells and mediate hypoxia-dependent activation of vascular cells during tumor development. Proc Natl Acad Sci USA. 2013; 110(18): 7312-7317.

[182]

Kore RA, Edmondson JL, Jenkins SV, et al. Hypoxia-derived exosomes induce putative altered pathways in biosynthesis and ion regulatory channels in glioblastoma cells. Biochem Biophys Rep. 2018; 14: 104-113.

[183]

Ramteke A, Ting H, Agarwal C, et al. Exosomes secreted under hypoxia enhance invasiveness and stemness of prostate cancer cells by targeting adherens junction molecules. Mol Carcinogen. 2015; 54(7): 554-565.

[184]

Dorayappan KDP, Wanner R, Wallbillich JJ, et al. Hypoxia-induced exosomes contribute to a more aggressive and chemoresistant ovarian cancer phenotype: a novel mechanism linking STAT3/Rab proteins. Oncogene. 2018; 37(28): 3806-3821.

[185]

Gao L, Wang L, Dai T, et al. Tumor-derived exosomes antagonize innate antiviral immunity. Nat Immunol. 2018; 19(3): 233-245.

[186]

Théry C, Ostrowski M, Segura E. Membrane vesicles as conveyors of immune responses. Nat Rev Immunol. 2009; 9(8): 581-593.

[187]

Greening DW, Gopal SK, Xu R, Simpson RJ, Chen W. Exosomes and their roles in immune regulation and cancer. Semin Cell Dec Biol. 2015; 40: 72-81.

[188]

Chen L, Han X. Anti–PD-1/PD-L1 therapy of human cancer: past, present, and future. J Clin Invest. 2015; 125(9): 3384-3391.

[189]

Yang Y, Li C-W, Chan L-C, et al. Exosomal PD-L1 harbors active defense function to suppress T cell killing of breast cancer cells and promote tumor growth. Cell Res. 2018; 28(8): 862-864.

[190]

Poggio M, Hu T, Pai C-C, et al. Suppression of exosomal PD-L1 induces systemic anti-tumor immunity and memory. Cell. 2019; 177(2): 414-427. e13.

[191]

Jing H, Meng M, Ye M, et al. Integrin α2 promotes immune escape in non-small-cell lung cancer by enhancing PD-L1 expression in exosomes to inhibit CD8+ T-cell activity. J Invest Med. 2024; 72(1): 57-66.

[192]

Chen J, Song Y, Miao F, et al. PDL1-positive exosomes suppress antitumor immunity by inducing tumor-specific CD8+ T cell exhaustion during metastasis. Cancer Sci. 2021; 112(9): 3437-3454.

[193]

Li C, Qiu S, Jin K, et al. Tumor-derived microparticles promote the progression of triple-negative breast cancer via PD-L1-associated immune suppression. Cancer Lett. 2021; 523: 43-56.

[194]

Zhang W, Zhong W, Wang B, et al. ICAM-1-mediated adhesion is a prerequisite for exosome-induced T cell suppression. Dev Cell. 2022; 57(3): 329-343. e7.

[195]

Zhong W, Lu Y, Han X, et al. Upregulation of exosome secretion from tumor-associated macrophages plays a key role in the suppression of anti-tumor immunity. Cell Rep. 2023; 42(10): 113224.

[196]

Shen D-D, Pang J-R, Bi Y-P, et al. LSD1 deletion decreases exosomal PD-L1 and restores T-cell response in gastric cancer. Mol Cancer. 2022; 21(1): 75.

[197]

Guan L, Wu B, Li T, et al. HRS phosphorylation drives immunosuppressive exosome secretion and restricts CD8+ T-cell infiltration into tumors. Nat Commun. 2022; 13(1): 4078.

[198]

Wang J, Deng R, Chen S, et al. Helicobacter pylori CagA promotes immune evasion of gastric cancer by upregulating PD-L1 level in exosomes. Iscience. 2023; 26(12): 108414.

[199]

Gong W, Donnelly C, Heath B, et al. Cancer-specific type-I interferon receptor signaling promotes cancer stemness and effector CD8+ T-cell exhaustion. Oncoimmunology. 2021; 10: 1997385.

[200]

Chatterjee S, Chatterjee A, Jana S, et al. Transforming growth factor beta orchestrates PD-L1 enrichment in tumor-derived exosomes and mediates CD8 T-cell dysfunction regulating early phosphorylation of TCR signalome in breast cancer. Carcinogenesis. 2021; 42(1): 38-47.

[201]

Chen S-W, Zhu S-Q, Pei X, et al. Cancer cell-derived exosomal circUSP7 induces CD8+ T cell dysfunction and anti-PD1 resistance by regulating the miR-934/SHP2 axis in NSCLC. Mol Cancer. 2021; 20(1): 1-18.

[202]

Ye S-b, Li Z-L, Luo D-h, et al. Tumor-derived exosomes promote tumor progression and T-cell dysfunction through the regulation of enriched exosomal microRNAs in human nasopharyngeal carcinoma. Oncotarget. 2014; 5(14): 5439.

[203]

Vignard V, Labbé M, Marec N, et al. MicroRNAs in tumor exosomes drive immune escape in melanoma. Cancer Immunol Res. 2020; 8(2): 255-267.

[204]

Li G, Chen W, Jiang K, et al. Exosome-mediated delivery of miR-519e-5p promotes malignant tumor phenotype and CD8+ T-cell exhaustion in metastatic PTC. J Clin Endocrinol Metab. 2023:dgad725.

[205]

Zhang Z, Yang L, Lei X, et al. Mechanism of non-small cell lung cancer cell-derived exosome miR-196b-5p promoting pyroptosis of tumor T cells and tumor cell proliferation by downregulating ING5. J Biochem Mol Toxic. 2024; 38(1): e23629.

[206]

Li W, Han G, Li F, et al. Cancer cell-derived exosomal miR-20a-5p inhibits CD8+ T-cell function and confers anti-programmed cell death 1 therapy resistance in triple-negative breast cancer. Cancer Sci. 2024; 115: 347–356

[207]

Otmani K, Rouas R, Lagneaux L, et al. Acute myeloid leukemia-derived exosomes deliver miR-24-3p to hinder the T-cell immune response through DENN/MADD targeting in the NF-κB signaling pathways. Cell Commun Signal. 2023; 21(1): 253.

[208]

Hong W, Xue M, Jiang J, Zhang Y, Gao X. Circular RNA circ-CPA4/let-7 miRNA/PD-L1 axis regulates cell growth, stemness, drug resistance and immune evasion in non-small cell lung cancer (NSCLC). J Exp Clin Canc Res. 2020; 39: 1-19.

[209]

Yuan Y, Wang L, Ge D, et al. Exosomal O-GlcNAc transferase from esophageal carcinoma stem cell promotes cancer immunosuppression through up-regulation of PD-1 in CD8+ T cells. Cancer Lett. 2021; 500: 98-106.

[210]

Zhang H-Y, Liang H-X, Wu S-H, Jiang H-Q, Wang Q, Yu Z-J. Overexpressed tumor suppressor exosomal miR-15a-5p in cancer cells inhibits PD1 expression in CD8+ T cells and suppresses the hepatocellular carcinoma progression. Front Oncol. 2021; 11: 622263.

[211]

Mondal SK, Haas D, Han J, Whiteside TL. Small EV in plasma of triple negative breast cancer patients induce intrinsic apoptosis in activated T cells. Commun Biol. 2023; 6(1): 815.

[212]

Shen T, Huang Z, Shi C, et al. Pancreatic cancer-derived exosomes induce apoptosis of T lymphocytes through the p38 MAPK-mediated endoplasmic reticulum stress. FASEB J. 2020; 34(6): 8442-8458.

[213]

Abusamra AJ, Zhong Z, Zheng X, et al. Tumor exosomes expressing Fas ligand mediate CD8+ T-cell apoptosis. Blood Cell Mol Dis 2005; 35(2): 169-173.

[214]

Leary N, Walser S, He Y, et al. Melanoma-derived extracellular vesicles mediate lymphatic remodelling and impair tumour immunity in draining lymph nodes. J Extracell Vesicles. 2022; 11(2): e12197.

[215]

Kondelková K, Vokurková D, Krejsek J, Borská L, Fiala Z, Ctirad A. Regulatory T cells (TREG) and their roles in immune system with respect to immunopathological disorders. Acta Medica (Hradec Kralove). 2010; 53(2): 73-77.

[216]

Gholipour E, Kahroba H, Soltani N, et al. Paediatric pre-B acute lymphoblastic leukaemia-derived exosomes regulate immune function in human T cells. J Cell Mol Med. 2022; 26(16): 4566-4576.

[217]

Graham R, Gazinska P, Zhang B, et al. Serum-derived extracellular vesicles from breast cancer patients contribute to differential regulation of T-cell-mediated immune-escape mechanisms in breast cancer subtypes. Front Immunol. 2023; 14: 1204224.

[218]

Ni H, Zhang H, Li L, et al. T cell-intrinsic STING signaling promotes regulatory T cell induction and immunosuppression by upregulating FOXP3 transcription in cervical cancer. J Immunother Cancer. 2022; 10(9): e005151.

[219]

Wei F, Fang R, Lyu K, et al. Exosomal PD-L1 derived from head and neck squamous cell carcinoma promotes immune evasion by activating the positive feedback loop of activated regulatory T cell-M2 macrophage. Oral Oncol. 2023; 145: 106532.

[220]

Chen Y, Li Z, Liang J, et al. CircRNA has_circ_0069313 induced OSCC immunity escape by miR-325-3p-Foxp3 axes in both OSCC cells and Treg cells. Aging (Albany NY). 2022; 14(10): 4376.

[221]

Pallett LJ, Swadling L, Diniz M, et al. Tissue CD14+ CD8+ T cells reprogrammed by myeloid cells and modulated by LPS. Nature. 2023; 614(7947): 334-342.

[222]

Ding X-Q, Wang Z-Y, Xia D, Wang R-X, Pan X-R, Tong J-H. Proteomic profiling of serum exosomes from patients with metastatic gastric cancer. Front Oncol. 2020; 10: 1113.

[223]

Rashid MH, Borin TF, Ara R, et al. Critical immunosuppressive effect of MDSC-derived exosomes in the tumor microenvironment. Oncol Rep. 2021; 45(3): 1171-1181.

[224]

Wang Y, Ding Y, Guo N, Wang S. MDSCs: key criminals of tumor pre-metastatic niche formation. Front Immunol. 2019; 10: 172.

[225]

Liu J, Liu J, Qin G, et al. MDSCs-derived GPR84 induces CD8+ T-cell senescence via p53 activation to suppress the antitumor response. J Immunother Cancer. 2023; 11(11): e007802.

[226]

Jia X, Xi J, Tian B, et al. The tautomerase activity of tumor exosomal MIF promotes pancreatic cancer progression by modulating MDSC differentiation. Cancer Immunol Res. 2024; 12(1): 72-90.

[227]

Du Z, Feng Y, Zhang H, Liu J, Wang J. Melanoma-derived small extracellular vesicles remodel the systemic onco-immunity via disrupting hematopoietic stem cell proliferation and differentiation. Cancer Lett. 2022; 545: 215841.

[228]

Zhang X, Li F, Tang Y, et al. miR-21a in exosomes from Lewis lung carcinoma cells accelerates tumor growth through targeting PDCD4 to enhance expansion of myeloid-derived suppressor cells. Oncogene. 2020; 39(40): 6354-6369.

[229]

Guo X, Qiu W, Liu Q, et al. Immunosuppressive effects of hypoxia-induced glioma exosomes through myeloid-derived suppressor cells via the miR-10a/Rora and miR-21/Pten Pathways. Oncogene. 2018; 37(31): 4239-4259.

[230]

Gao J, Ao Y-Q, Zhang L-X, et al. Exosomal circZNF451 restrains anti-PD1 treatment in lung adenocarcinoma via polarizing macrophages by complexing with TRIM56 and FXR1. J Exp Clin Cancer Res. 2022; 41(1): 295.

[231]

Shinohara H, Kuranaga Y, Kumazaki M, et al. Regulated polarization of tumor-associated macrophages by mir-145 via colorectal cancer–derived extracellular vesicles. J Immunol. 2017; 199(4): 1505-1515.

[232]

Linton SS, Abraham T, Liao J, et al. Tumor-promoting effects of pancreatic cancer cell exosomes on THP-1-derived macrophages. PLoS One. 2018; 13(11): e0206759.

[233]

Wang S, Gao Y. Pancreatic cancer cell-derived microRNA-155-5p-containing extracellular vesicles promote immune evasion by triggering EHF-dependent activation of Akt/NF-κB signaling pathway. Int Immunopharmacol. 2021; 100: 107990.

[234]

Chang Y-T, Peng H-Y, Hu C-M, Huang S-C, Tien S-C, Jeng Y-M. Pancreatic cancer-derived small extracellular vesical Ezrin regulates macrophage polarization and promotes metastasis. Am J Cancer Res. 2020; 10(1): 12.

[235]

Lu J-C, Zhang P-F, Huang X-Y, et al. Amplification of spatially isolated adenosine pathway by tumor–macrophage interaction induces anti-PD1 resistance in hepatocellular carcinoma. J Hematol Oncol. 2021; 14: 1-20.

[236]

Chen J, Lin Z, Liu L, et al. GOLM1 exacerbates CD8+ T cell suppression in hepatocellular carcinoma by promoting exosomal PD-L1 transport into tumor-associated macrophages. Signal Transduct Tar. 2021; 6(1): 397.

[237]

Tan HY, Wang N, Zhang C, Chan YT, Yuen MF, Feng Y. Lysyl oxidase-like 4 fosters an immunosuppressive microenvironment during hepatocarcinogenesis. Hepatology. 2021; 73(6): 2326-2341.

[238]

Yao X, Tu Y, Xu Y, Guo Y, Yao F, Zhang X. Endoplasmic reticulum stress-induced exosomal miR-27a-3p promotes immune escape in breast cancer via regulating PD-L1 expression in macrophages. J Cell Mol Med. 2020; 24(17): 9560-9573.

[239]

Li X, Wang S, Mu W, et al. Reactive oxygen species reprogram macrophages to suppress antitumor immune response through the exosomal miR-155-5p/PD-L1 pathway. J Exp Clin Cancer Res. 2022; 41(1): 1-19.

[240]

Burassakarn A, Srisathaporn S, Pientong C, et al. Exosomes-carrying Epstein-Barr virus-encoded small RNA-1 induces indoleamine 2, 3-dioxygenase expression in tumor-infiltrating macrophages of oral squamous-cell carcinomas and suppresses T-cell activity by activating RIG-I/IL-6/TNF-α pathway. Oral Oncol. 2021; 117: 105279.

[241]

Zhang P-F, Gao C, Huang X-Y, et al. Cancer cell-derived exosomal circUHRF1 induces natural killer cell exhaustion and may cause resistance to anti-PD1 therapy in hepatocellular carcinoma. Mol Cancer. 2020; 19: 1-15.

[242]

Qi M, Xia Y, Wu Y, et al. Lin28B-high breast cancer cells promote immune suppression in the lung pre-metastatic niche via exosomes and support cancer progression. Nat Commun. 2022; 13(1): 897.

[243]

Yu W, Hurley J, Roberts D, et al. Exosome-based liquid biopsies in cancer: opportunities and challenges. Ann Oncol. 2021; 32(4): 466-477.

[244]

Bang YH, Shim JH, Ryu KJ, et al. Clinical relevance of serum-derived exosomal messenger RNA sequencing in patients with non-Hodgkin lymphoma. J Cancer. 2022; 13(5): 1388.

[245]

Marrugo-Ramírez J, Mir M, Samitier J. Blood-based cancer biomarkers in liquid biopsy: a promising non-invasive alternative to tissue biopsy. Int J Mol Sci. 2018; 19(10): 2877.

[246]

Wang K, Wang X, Pan Q, Zhao B. Liquid biopsy techniques and pancreatic cancer: diagnosis, monitoring, and evaluation. Mol Cancer. 2023; 22(1): 167.

[247]

Zhou H, Zhu L, Song J, et al. Liquid biopsy at the frontier of detection, prognosis and progression monitoring in colorectal cancer. Mol Cancer. 2022; 21(1): 86.

[248]

Moon P-G, Lee J-E, Cho Y-E, et al. Identification of developmental endothelial locus-1 on circulating extracellular vesicles as a novel biomarker for early breast cancer detection. Clin Cancer Res. 2016; 22(7): 1757-1766.

[249]

Alegre E, Zubiri L, Perez-Gracia JL, et al. Circulating melanoma exosomes as diagnostic and prognosis biomarkers. Clin Chim Acta. 2016; 454: 28-32.

[250]

Liang K, Liu F, Fan J, et al. Nanoplasmonic quantification of tumor-derived extracellular vesicles in plasma microsamples for diagnosis and treatment monitoring. Nat Biomed Eng. 2017; 1(4): 0021.

[251]

Madhankumar A, Mrowczynski OD, Patel SR, et al. Interleukin-13 conjugated quantum dots for identification of glioma initiating cells and their extracellular vesicles. Acta Biomater. 2017; 58: 205-213.

[252]

Chen C-L, Lai Y-F, Tang P, et al. Comparative and targeted proteomic analyses of urinary microparticles from bladder cancer and hernia patients. J Phys Chem Lett. 2012; 11(12): 5611-5629.

[253]

Raimondo F, Morosi L, Corbetta S, et al. Differential protein profiling of renal cell carcinoma urinary exosomes. Mol BioSyst. 2013; 9(6): 1220-1233.

[254]

Im H, Shao H, Park YI, et al. Label-free detection and molecular profiling of exosomes with a nano-plasmonic sensor. Nat Biotechnol. 2014; 32(5): 490-495.

[255]

Dabral P, Bhasin N, Ranjan M, Makhlouf MM, Abd Elmageed ZY. Tumor-derived extracellular vesicles as liquid biopsy for diagnosis and prognosis of solid tumors: their clinical utility and reliability as tumor biomarkers. Cancers. 2024; 16(13): 2462.

[256]

Graner MW, Alzate O, Dechkovskaia AM, et al. Proteomic and immunologic analyses of brain tumor exosomes. FASEB J. 2009; 23(5): 1541.

[257]

Zhang C, Chong X, Jiang F, et al. Plasma extracellular vesicle derived protein profile predicting and monitoring immunotherapeutic outcomes of gastric cancer. J Extracell Vesicles. 2022; 11(4): e12209.

[258]

Khan S, Jutzy JM, Valenzuela MMA, et al. Plasma-derived exosomal survivin, a plausible biomarker for early detection of prostate cancer. PLoS One. 2012; 7(10): e46737.

[259]

Kawakami K, Fujita Y, Matsuda Y, et al. Gamma-glutamyltransferase activity in exosomes as a potential marker for prostate cancer. BMC cancer. 2017; 17: 1-12.

[260]

Logozzi M, Angelini DF, Iessi E, et al. Increased PSA expression on prostate cancer exosomes in in vitro condition and in cancer patients. Cancer Lett. 2017; 403: 318-329.

[261]

Gaballa R, Ali HE, Mahmoud MO, et al. Exosomes-mediated transfer of Itga2 promotes migration and invasion of prostate cancer cells by inducing epithelial-mesenchymal transition. Cancers. 2020; 12(8): 2300.

[262]

Melo SA, Luecke LB, Kahlert C, et al. Glypican-1 identifies cancer exosomes and detects early pancreatic cancer. Nature. 2015; 523(7559): 177-182.

[263]

Logozzi M, De Milito A, Lugini L, et al. High levels of exosomes expressing CD63 and caveolin-1 in plasma of melanoma patients. PLoS One. 2009; 4(4): e5219.

[264]

Ueda K, Ishikawa N, Tatsuguchi A, Saichi N, Fujii R, Nakagawa H. Antibody-coupled monolithic silica microtips for highthroughput molecular profiling of circulating exosomes. Sci Rep. 2014; 4(1): 6232.

[265]

Sandfeld-Paulsen B, Aggerholm-Pedersen N, Bæk R, et al. Exosomal proteins as prognostic biomarkers in non-small cell lung cancer. Mol Oncol. 2016; 10(10): 1595-1602.

[266]

Arbelaiz A, Azkargorta M, Krawczyk M, et al. Serum extracellular vesicles contain protein biomarkers for primary sclerosing cholangitis and cholangiocarcinoma. Hepatology. 2017; 66(4): 1125-1143.

[267]

Li J, Chen Y, Guo X, et al. GPC 1 exosome and its regulatory mi RNA s are specific markers for the detection and target therapy of colorectal cancer. J Cell Mol Med. 2017; 21(5): 838-847.

[268]

Yoshioka Y, Kosaka N, Konishi Y, et al. Ultra-sensitive liquid biopsy of circulating extracellular vesicles using ExoScreen. Nat Commun. 2014; 5(1): 3591.

[269]

Sun B, Li Y, Zhou Y, et al. Circulating exosomal CPNE3 as a diagnostic and prognostic biomarker for colorectal cancer. J Cell Physiol. 2019; 234(2): 1416-1425.

[270]

Lee C-H, Im E-J, Moon P-G, Baek M-C. Discovery of a diagnostic biomarker for colon cancer through proteomic profiling of small extracellular vesicles. BMC Cancer. 2018; 18: 1-11.

[271]

Hannafon BN, Trigoso YD, Calloway CL, et al. Plasma exosome microRNAs are indicative of breast cancer. Breast Cancer Res. 2016; 18: 1-14.

[272]

Zhai L-Y, Li M-X, Pan W-L, et al. In situ detection of plasma exosomal microRNA-1246 for breast cancer diagnostics by a Au nanoflare probe. ACS Appl Mater Interfaces. 2018; 10(46): 39478-39486.

[273]

Rodríguez-Martínez A, de Miguel-Pérez D, Ortega FG, et al. Exosomal miRNA profile as complementary tool in the diagnostic and prediction of treatment response in localized breast cancer under neoadjuvant chemotherapy. Breast Cancer Res. 2019; 21: 1-9.

[274]

Stevic I, Müller V, Weber K, et al. Specific microRNA signatures in exosomes of triple-negative and HER2-positive breast cancer patients undergoing neoadjuvant therapy within the GeparSixto trial. BMC Med. 2018; 16: 1-16.

[275]

Wang J, Yan F, Zhao Q, et al. Circulating exosomal miR-125a-3p as a novel biomarker for early-stage colon cancer. Sci Rep. 2017; 7(1): 4150.

[276]

Matsumura T, Sugimachi K, Iinuma H, et al. Exosomal microRNA in serum is a novel biomarker of recurrence in human colorectal cancer. Brit J Cancer. 2015; 113(2): 275-281.

[277]

Ogata-Kawata H, Izumiya M, Kurioka D, et al. Circulating exosomal microRNAs as biomarkers of colon cancer. PLoS One. 2014; 9(4): e92921.

[278]

Manterola L, Guruceaga E, Pérez-Larraya JG, et al. A small noncoding RNA signature found in exosomes of GBM patient serum as a diagnostic tool. Neuro-oncology. 2014; 16(4): 520-527.

[279]

Sohn W, Kim J, Kang SH, et al. Serum exosomal microRNAs as novel biomarkers for hepatocellular carcinoma. Exp Mol Med. 2015; 47(9): e184-e184.

[280]

Bryant R, Pawlowski T, Catto J, et al. Changes in circulating microRNA levels associated with prostate cancer. Brit J Cancer. 2012; 106(4): 768-774.

[281]

Ali HE, Gaballah MS, Gaballa R, et al. Small extracellular vesicle-derived microRNAs stratify prostate cancer patients according to gleason score, race and associate with survival of African American and Caucasian men. Cancers. 2021; 13(20): 5236.

[282]

Madhavan B, Yue S, Galli U, et al. Combined evaluation of a panel of protein and miRNA serum-exosome biomarkers for pancreatic cancer diagnosis increases sensitivity and specificity. Int J Cancer. 2015; 136(11): 2616-2627.

[283]

Que R, Ding G, Chen J, Cao L. Analysis of serum exosomal microRNAs and clinicopathologic features of patients with pancreatic adenocarcinoma. World J Surg Oncol. 2013; 11: 1-9.

[284]

Joshi GK, Deitz-McElyea S, Liyanage T, et al. Label-free nanoplasmonic-based short noncoding RNA sensing at attomolar concentrations allows for quantitative and highly specific assay of microRNA-10b in biological fluids and circulating exosomes. ACS Nano. 2015; 9(11): 11075-11089.

[285]

Lai X, Wang M, McElyea SD, Sherman S, House M, Korc M. A microRNA signature in circulating exosomes is superior to exosomal glypican-1 levels for diagnosing pancreatic cancer. Cancer Lett. 2017; 393: 86-93.

[286]

Jin X, Chen Y, Chen H, et al. Evaluation of tumor-derived exosomal miRNA as potential diagnostic biomarkers for early-stage non–small cell lung cancer using next-generation sequencing. Clin Cancer Res. 2017; 23(17): 5311-5319.

[287]

Cazzoli R, Buttitta F, Di Nicola M, et al. microRNAs derived from circulating exosomes as noninvasive biomarkers for screening and diagnosing lung cancer. J Thorac Oncol. 2013; 8(9): 1156-1162.

[288]

Wen Q, Wang Y, Li X, Jin X, Wang G. Decreased serum exosomal miR-29a expression and its clinical significance in papillary thyroid carcinoma. J Clin Lab Anal. 2021; 35(1): e23560.

[289]

Zou X, Gao F, Wang Z-Y, et al. A three-microRNA panel in serum as novel biomarker for papillary thyroid carcinoma diagnosis. Chinese Med J. 2020; 133(21): 2543-2551.

[290]

Pan Q, Zhao J, Li M, et al. Exosomal miRNAs are potential diagnostic biomarkers between malignant and benign thyroid nodules based on next-generation sequencing. Carcinogenesis. 2020; 41(1): 18-24.

[291]

Liang M, Yu S, Tang S, et al. A panel of plasma exosomal miRNAs as potential biomarkers for differential diagnosis of thyroid nodules. Front Genet. 2020; 11: 449.

[292]

Dai D, Tan Y, Guo L, Tang A, Zhao Y. Identification of exosomal miRNA biomarkers for diagnosis of papillary thyroid cancer by small RNA sequencing. Euro J Endocrinol. 2020; 182(1): 111-121.

[293]

Akers JC, Ramakrishnan V, Kim R, et al. MiR-21 in the extracellular vesicles (EVs) of cerebrospinal fluid (CSF): a platform for glioblastoma biomarker development. PLoS One. 2013; 8(10): e78115.

[294]

Cappellesso R, Tinazzi A, Giurici T, et al. Programmed cell death 4 and micro RNA 21 inverse expression is maintained in cells and exosomes from ovarian serous carcinoma effusions. Cancer Cytopathol. 2014; 122(9): 685-693.

[295]

Zhou J, Gong G, Tan H, et al. Urinary microRNA-30a-5p is a potential biomarker for ovarian serous adenocarcinoma. Oncol Rep. 2015; 33(6): 2915-2923.

[296]

Goldvaser H, Gutkin A, Beery E, et al. Characterisation of blood-derived exosomal hTERT mRNA secretion in cancer patients: a potential pan-cancer marker. Brit J Cancer. 2017; 117(3): 353-357.

[297]

Yokoi A, Yoshioka Y, Yamamoto Y, et al. Malignant extracellular vesicles carrying MMP1 mRNA facilitate peritoneal dissemination in ovarian cancer. Nat Commun. 2017; 8(1): 14470.

[298]

Woo H-K, Park J, Ku JY, et al. Urine-based liquid biopsy: non-invasive and sensitive AR-V7 detection in urinary EVs from patients with prostate cancer. Lab on a Chip. 2019; 19(1): 87-97.

[299]

Kahlert C, Melo SA, Protopopov A, et al. Identification of double-stranded genomic DNA spanning all chromosomes with mutated KRAS and p53 DNA in the serum exosomes of patients with pancreatic cancer. J Biol Chem. 2014; 289(7): 3869-3875.

[300]

Allenson K, Castillo J, San Lucas F, et al. High prevalence of mutantKRAS in circulating exosome-derived DNA from early-stage pancreatic cancer patients. Ann Oncol. 2017; 28(4): 741-747.

[301]

San Lucas F, Allenson K, Bernard V, et al. Minimally invasive genomic and transcriptomic profiling of visceral cancers by next-generation sequencing of circulating exosomes. Ann Oncol. 2016; 27(4): 635-641.

[302]

Lázaro-Ibáñez E, Sanz-Garcia A, Visakorpi T, et al. Different gDNA content in the subpopulations of prostate cancer extracellular vesicles: apoptotic bodies, microvesicles, and exosomes. Prostate. 2014; 74(14): 1379-1390.

[303]

Yu D, Li Y, Wang M, et al. Exosomes as a new frontier of cancer liquid biopsy. Mol Cancer. 2022; 21(1): 56.

[304]

Hosseini K, Ranjbar M, Pirpour Tazehkand A, et al. Evaluation of exosomal non-coding RNAs in cancer using high-throughput sequencing. J Transl Med. 2022; 20(1): 30.

[305]

Hur JY, Lee KY. Characteristics and clinical application of extracellular vesicle-derived DNA. Cancers. 2021; 13(15): 3827.

[306]

Taylor C, Chacko S, Davey M, et al. Peptide-affinity precipitation of extracellular vesicles and cell-free DNA improves sequencing performance for the detection of pathogenic mutations in lung cancer patient plasma. Int J Mol Sci. 2020; 21(23): 9083.

[307]

Puhka M, Takatalo M, Nordberg ME, et al. Metabolomic profiling of extracellular vesicles and alternative normalization methods reveal enriched metabolites and strategies to study prostate cancer-related changes. Theranostics. 2017; 7(16): 3824-3841.

[308]

Clos-Garcia M, Loizaga-Iriarte A, Zuñiga-Garcia P, et al. Metabolic alterations in urine extracellular vesicles are associated to prostate cancer pathogenesis and progression. J Extracell Vesicles. 2018; 7(1): 1470442.

[309]

Cheng L, Zhang K, Qing Y, et al. Proteomic and lipidomic analysis of exosomes derived from ovarian cancer cells and ovarian surface epithelial cells. J Ovarian Res. 2020; 13: 1-13.

[310]

Elmallah MI, Ortega-Deballon P, Hermite L, Pais-De-Barros JP, Gobbo J, Garrido C. Lipidomic profiling of exosomes from colorectal cancer cells and patients reveals potential biomarkers. Mol Oncol. 2022; 16(14): 2710-2718.

[311]

Sanchez JI, Jiao J, Kwan S-Y, et al. Lipidomic profiles of plasma exosomes identify candidate biomarkers for early detection of hepatocellular carcinoma in patients with cirrhosis. Cancer Prev Res. 2021; 14(10): 955-962.

[312]

Tiziani S, Lopes V, Günther UL. Early stage diagnosis of oral cancer using 1H NMR–based metabolomics. Neoplasia. 2009; 11(3): 269-276.

[313]

Shin H, Choi BH, Shim O, et al. Single test-based diagnosis of multiple cancer types using Exosome-SERS-AI for early stage cancers. Nat Commun. 2023; 14(1): 1644.

[314]

Li B, Kugeratski FG, Kalluri R. A novel machine learning algorithm selects proteome signature to specifically identify cancer exosomes. Elife. 2024; 12: RP90390.

[315]

Achreja A, Zhao H, Yang L, Yun TH, Marini J, Nagrath D. Exo-MFA - A 13C metabolic flux analysis framework to dissect tumor microenvironment-secreted exosome contributions towards cancer cell metabolism. Metab Eng. 2017; 43(Pt B): 156-172.

[316]

Ko J, Bhagwat N, Yee SS, et al. Combining machine learning and nanofluidic technology to diagnose pancreatic cancer using exosomes. ACS Nano. 2017; 11(11): 11182-11193.

[317]

Kim S, Choi BH, Shin H, et al. Plasma exosome analysis for protein mutation identification using a combination of raman spectroscopy and deep learning. ACS sens. 2023; 8(6): 2391-2400.

[318]

Gao J, Qiu X, Li X, et al. Expression profiles and clinical value of plasma exosomal Tim-3 and Galectin-9 in non-small cell lung cancer. Biochem Bioph Res Co. 2018; 498(3): 409-415.

[319]

Del Re M, Marconcini R, Pasquini G, et al. PD-L1 mRNA expression in plasma-derived exosomes is associated with response to anti-PD-1 antibodies in melanoma and NSCLC. Brit J Cancer. 2018; 118(6): 820-824.

[320]

Del Re M, Cucchiara F, Rofi E, et al. A multiparametric approach to improve the prediction of response to immunotherapy in patients with metastatic NSCLC. Cancer Immunol Immun. 2021; 70: 1667-1678.

[321]

de Miguel-Perez D, Russo A, Arrieta O, et al. Extracellular vesicle PD-L1 dynamics predict durable response to immune-checkpoint inhibitors and survival in patients with non-small cell lung cancer. J Exp Clin Cancer Res. 2022; 41(1): 186.

[322]

Lee C-H, Bae J-H, Choe E-J, et al. Macitentan improves antitumor immune responses by inhibiting the secretion of tumor-derived extracellular vesicle PD-L1. Theranostics. 2022; 12(5): 1971.

[323]

Zhao R, Zhang Y, Zhang X, et al. Exosomal long noncoding RNA HOTTIP as potential novel diagnostic and prognostic biomarker test for gastric cancer. Mol Cancer. 2018; 17: 1-5.

[324]

Zhan Y, Du L, Wang L, et al. Expression signatures of exosomal long non-coding RNAs in urine serve as novel non-invasive biomarkers for diagnosis and recurrence prediction of bladder cancer. Mol Cancer. 2018; 17: 1-5.

[325]

Lee YR, Kim G, Tak WY, et al. Circulating exosomal noncoding RNAs as prognostic biomarkers in human hepatocellular carcinoma. Int J Cancer. 2019; 144(6): 1444-1452.

[326]

Chen X, Chen R-X, Wei W-S, et al. PRMT5 circular RNA promotes metastasis of urothelial carcinoma of the bladder through sponging miR-30c to induce epithelial–mesenchymal transition. Clin Cancer Res. 2018; 24(24): 6319-6330.

[327]

Abramowicz A, Story MD. The long and short of it: the emerging roles of non-coding RNA in small extracellular vesicles. Cancers. 2020; 12(6): 1445.

[328]

Tao L, Zhou J, Yuan C, et al. Metabolomics identifies serum and exosomes metabolite markers of pancreatic cancer. Metabolomics. 2019; 15(6): 1-11.

[329]

Zhu Q, Huang L, Yang Q, et al. Metabolomic analysis of exosomal-markers in esophageal squamous cell carcinoma. Nanoscale. 2021; 13(39): 16457-16464.

[330]

Tatischeff I. Assets of circulating extracellular vesicles for early diagnosis and prognosis of gastric cancer by liquid biopsy. Jpn J Gstro Hepato. 2022; 8: 1-9.

[331]

Chen I-H, Xue L, Hsu C-C, et al. Phosphoproteins in extracellular vesicles as candidate markers for breast cancer. Proc Natl Acad Sci USA. 2017; 114(12): 3175-3180.

[332]

Su YY, Sun L, Guo ZR, et al. Upregulated expression of serum exosomal miR-375 and miR-1307 enhance the diagnostic power of CA125 for ovarian cancer. J Ovarian Res. 2019; 12: 1-9.

[333]

He Y, Deng F, Yang S, et al. Exosomal microRNA: a novel biomarker for breast cancer. Biomark Med. 2018; 12(2): 177-188.

[334]

Wang W, Jo H, Park S, et al. Integrated analysis of ascites and plasma extracellular vesicles identifies a miRNA-based diagnostic signature in ovarian cancer. Cancer Lett. 2022; 542: 215735.

[335]

Zhou L, Wang W, Wang F, et al. Plasma-derived exosomal miR-15a-5p as a promising diagnostic biomarker for early detection of endometrial carcinoma. Mol Cancer. 2021; 20: 1-6.

[336]

Nakamura K, Zhu Z, Roy S, et al. An exosome-based transcriptomic signature for noninvasive, early detection of patients with pancreatic ductal adenocarcinoma: a multicenter cohort study. Gastroenterology. 2022; 163(5): 1252-1266. e2.

[337]

Roseblade A, Luk F, Ung A, Bebawy M. Targeting microparticle biogenesis: a novel approach to the circumvention of cancer multidrug resistance. Curr Cancer Drug Tar. 2015; 15(3): 205-214.

[338]

Kosgodage US, Trindade RP, Thompson PR, Inal JM, Lange S. Chloramidine/bisindolylmaleimide-I-mediated inhibition of exosome and microvesicle release and enhanced efficacy of cancer chemotherapy. Int J Mol Sci. 2017; 18(5): 1007.

[339]

Hu Y, Yan C, Mu L, et al. Fibroblast-derived exosomes contribute to chemoresistance through priming cancer stem cells in colorectal cancer. PLoS One. 2015; 10(5): e0125625.

[340]

Xie L, Li J, Wang G, et al. Phototheranostic metal-phenolic networks with antiexosomal PD-L1 enhanced ferroptosis for synergistic immunotherapy. J Am Chem Soc. 2022; 144(2): 787-797.

[341]

Richards KE, Zeleniak AE, Fishel ML, Wu J, Littlepage LE, Hill R. Cancer-associated fibroblast exosomes regulate survival and proliferation of pancreatic cancer cells. Oncogene. 2017; 36(13): 1770-1778.

[342]

Menck K, Sönmezer C, Worst TS, et al. Neutral sphingomyelinases control extracellular vesicles budding from the plasma membrane. J Extracell Vesicles. 2017; 6(1): 1378056.

[343]

Li J, Liu K, Liu Y, et al. Exosomes mediate the cell-to-cell transmission of IFN-α-induced antiviral activity. Nat Immunol. 2013; 14(8): 793-803.

[344]

Rojas C, Barnaeva E, Thomas AG, et al. DPTIP, a newly identified potent brain penetrant neutral sphingomyelinase 2 inhibitor, regulates astrocyte-peripheral immune communication following brain inflammation. Sci Rep. 2018; 8(1): 17715.

[345]

Henriksson C, Hellum M, Haug K, et al. Anticoagulant effects of an antidiabetic drug on monocytes in vitro. Thromb Res. 2011; 128(5): e100-e106.

[346]

Koch R, Aung T, Vogel D, et al. Nuclear trapping through inhibition of exosomal export by indomethacin increases cytostatic efficacy of doxorubicin and pixantrone. Clin Cancer Res. 2016; 22(2): 395-404.

[347]

Jorfi S, Ansa-Addo EA, Kholia S, et al. Inhibition of microvesiculation sensitizes prostate cancer cells to chemotherapy and reduces docetaxel dose required to limit tumor growth in vivo. Sci Rep. 2015; 5(1): 13006.

[348]

Li B, Antonyak MA, Zhang J, Cerione RA. RhoA triggers a specific signaling pathway that generates transforming microvesicles in cancer cells. Oncogene. 2012; 31(45): 4740-4749.

[349]

Khan S, Jutzy JM, Aspe JR, McGregor DW, Neidigh JW, Wall NR. Survivin is released from cancer cells via exosomes. Apoptosis. 2011; 16: 1-12.

[350]

Li M, Yu D, Williams KJ, Liu M-L. Tobacco smoke induces the generation of procoagulant microvesicles from human monocytes/macrophages. Arterioscl Throm Vas. 2010; 30(9): 1818-1824.

[351]

Wang Y, Luo L, Mörgelin M, Thorlacius H. Rac1 regulates sepsis-induced formation of platelet-derived microparticles and thrombin generation. Biochem Bioph Res Co. 2017; 487(4): 887-891.

[352]

Datta A, Kim H, Lal M, et al. Manumycin A suppresses exosome biogenesis and secretion via targeted inhibition of Ras/Raf/ERK1/2 signaling and hnRNP H1 in castration-resistant prostate cancer cells. Cancer Lett. 2017; 408: 73-81.

[353]

Martin L-A, Head JE, Pancholi S, et al. The farnesyltransferase inhibitor R115777 (tipifarnib) in combination with tamoxifen acts synergistically to inhibit MCF-7 breast cancer cell proliferation and cell cycle progression in vitro and in vivo. Mol Cancer Ther. 2007; 6(9): 2458-2467.

[354]

Kholia S, Jorfi S, Thompson PR, et al. A novel role for peptidylarginine deiminases in microvesicle release reveals therapeutic potential of PAD inhibition in sensitizing prostate cancer cells to chemotherapy. J Extracell Vesicles. 2015; 4(1): 26192.

[355]

Stratton D, Moore C, Zheng L, Lange S, Inal J. Prostate cancer cells stimulated by calcium-mediated activation of protein kinase C undergo a refractory period before re-releasing calcium-bearing microvesicles. Biochem Bioph Res Co. 2015; 460(3): 511-517.

[356]

Im E-J, Lee C-H, Moon P-G, et al. Sulfisoxazole inhibits the secretion of small extracellular vesicles by targeting the endothelin receptor A. Nat Commun. 2019; 10(1): 1387.

[357]

Leblanc R, Kashyap R, Barral K, et al. Pharmacological inhibition of syntenin PDZ2 domain impairs breast cancer cell activities and exosome loading with syndecan and EpCAM cargo. J Extracell Vesicles. 2020; 10(2): e12039.

[358]

Khan FM, Saleh E, Alawadhi H, Harati R, Zimmermann W-H, El-Awady R. Inhibition of exosome release by ketotifen enhances sensitivity of cancer cells to doxorubicin. Cancer Biol Ther. 2018; 19(1): 25-33.

[359]

Chalmin F, Ladoire S, Mignot G, et al. Membrane-associated Hsp72 from tumor-derived exosomes mediates STAT3-dependent immunosuppressive function of mouse and human myeloid-derived suppressor cells. J Clin Invest. 2010; 120(2): 457-471.

[360]

Christianson HC, Svensson KJ, Van Kuppevelt TH, Li J-P, Belting M. Cancer cell exosomes depend on cell-surface heparan sulfate proteoglycans for their internalization and functional activity. Proc Natl Acad Sci USA. 2013; 110(43): 17380-17385.

[361]

Kawamoto T, Ohga N, Akiyama K, et al. Tumor-derived microvesicles induce proangiogenic phenotype in endothelial cells via endocytosis. PLoS One. 2012; 7(3): e34045.

[362]

Escrevente C, Keller S, Altevogt P, Costa J. Interaction and uptake of exosomes by ovarian cancer cells. BMC cancer. 2011; 11: 1-10.

[363]

Hazan-Halevy I, Rosenblum D, Weinstein S, Bairey O, Raanani P, Peer D. Cell-specific uptake of mantle cell lymphoma-derived exosomes by malignant and non-malignant B-lymphocytes. Cancer Lett. 2015; 364(1): 59-69.

[364]

Skotland T, Sandvig K, Llorente A. Lipids in exosomes: current knowledge and the way forward. Prog Lipid Res. 2017; 66: 30-41.

[365]

Record M, Poirot M, Silvente-Poirot S. Emerging concepts on the role of exosomes in lipid metabolic diseases. Biochimie. 2014; 96: 67-74.

[366]

Ranganathan S, Jackson RL, Harmony JA. Effect of pantethine on the biosynthesis of cholesterol in human skin fibroblasts. Atherosclerosis. 1982; 44(3): 261-273.

[367]

Siklos M, BenAissa M, Thatcher GR. Cysteine proteases as therapeutic targets: does selectivity matter? A systematic review of calpain and cathepsin inhibitors. Acta Pharm Sin B. 2015; 5(6): 506-519.

[368]

Watanabe K, Ueno M, Kamiya D, et al. A ROCK inhibitor permits survival of dissociated human embryonic stem cells. Nat Biotechnol. 2007; 25(6): 681-686.

[369]

Savina A, Furlán M, Vidal M, Colombo MI. Exosome release is regulated by a calcium-dependent mechanism in K562 cells. J Biol Chem. 2003; 278(22): 20083-20090.

[370]

Isola AL, Chen S. Exosomes: the link between GPCR activation and metastatic potential? Front Genet. 2016; 7: 56.

[371]

Jadli AS, Ballasy N, Edalat P, Patel VB. Inside (sight) of tiny communicator: exosome biogenesis, secretion, and uptake. Mol Cell Biochem. 2020; 467: 77-94.

[372]

Yong T, Wang D, Li X, et al. Extracellular vesicles for tumor targeting delivery based on five features principle. J Control Release. 2020; 322: 555-565.

[373]

Kim MS, Haney MJ, Zhao Y, et al. Development of exosome-encapsulated paclitaxel to overcome MDR in cancer cells. Nanomed-Nanotechnol. 2016; 12(3): 655-664.

[374]

Wei H, Chen J, Wang S, et al. A nanodrug consisting of doxorubicin and exosome derived from mesenchymal stem cells for osteosarcoma treatment in vitro. Int J Nanomed. 2019; 14: 8603-8610.

[375]

Wei H, Chen F, Chen J, et al. Mesenchymal stem cell derived exosomes as nanodrug carrier of doxorubicin for targeted osteosarcoma therapy via SDF1-CXCR4 axis. Int J Nanomed. 2022; 17: 3483.

[376]

Cho H, Jung I, Ju H, Baek M-C, Yea K. Engineered CD8+ T cell-derived extracellular vesicles induce enhanced anti-cancer efficacy and targeting to lung cancer cells. Cytokine. 2023; 169: 156249.

[377]

Dianat-Moghadam H, Heidarifard M, Mahari A, et al. TRAIL in oncology: from recombinant TRAIL to nano-and self-targeted TRAIL-based therapies. Pharmacol Res. 2020; 155: 104716.

[378]

Jiang L, Gu Y, Du Y, Tang X, Wu X, Liu J. Engineering exosomes endowed with targeted delivery of triptolide for malignant melanoma therapy. ACS Appl Mater Interfaces. 2021; 13(36): 42411-42428.

[379]

Qiu Y, Sun J, Qiu J, et al. Antitumor activity of cabazitaxel and MSC-TRAIL derived extracellular vesicles in drug-resistant oral squamous cell carcinoma. Cancer Manag Res. 2020; 12: 10809-10820.

[380]

Yuan Q, Su K, Li S, et al. Pulmonary delivery of extracellular vesicle-encapsulated dinaciclib as an effective lung cancer therapy. Cancers. 2022; 14(14): 3550.

[381]

Gu Y, Du Y, Jiang L, et al. αvβ3 integrin-specific exosomes engineered with cyclopeptide for targeted delivery of triptolide against malignant melanoma. J Nanobiotechnol. 2022; 20(1): 384.

[382]

Aspe JR, Diaz Osterman CJ, Jutzy JM, Deshields S, Whang S, Wall NR. Enhancement of Gemcitabine sensitivity in pancreatic adenocarcinoma by novel exosome-mediated delivery of the Survivin-T34A mutant. J Extracell Vesicles. 2014; 3(1): 23244.

[383]

Yim N, Ryu S-W, Choi K, et al. Exosome engineering for efficient intracellular delivery of soluble proteins using optically reversible protein–protein interaction module. Nat Commun. 2016; 7(1): 1-9.

[384]

Nordmeier S, Ke W, Afonin KA, Portnoy V. Exosome mediated delivery of functional nucleic acid nanoparticles (NANPs). Therap RNA Nanotechnol. 2021: 539-564.

[385]

Wilbie D, Walther J, Mastrobattista E. Delivery aspects of CRISPR/Cas for in vivo genome editing. Acc Chem Res. 2019; 52(6): 1555-1564.

[386]

McAndrews KM, Xiao F, Chronopoulos A, LeBleu VS, Kugeratski FG, Kalluri R. Exosome-mediated delivery of CRISPR/Cas9 for targeting of oncogenic KrasG12D in pancreatic cancer. Life Sci Alliance. 2021; 4(9): e202000875.

[387]

Du J, Wan Z, Wang C, et al. Designer exosomes for targeted and efficient ferroptosis induction in cancer via chemo-photodynamic therapy. Theranostics. 2021; 11(17): 8185.

[388]

Dumontel B, Susa F, Limongi T, et al. Nanotechnological engineering of extracellular vesicles for the development of actively targeted hybrid nanodevices. Cell Biosci. 2022; 12(1): 61.

[389]

Wang J, Liu Y, Zhang Y, Li X, Fang M, Qian D. Targeting exosomes enveloped EBV-miR-BART1-5p-antagomiRs for NPC therapy through both anti-vasculogenic mimicry and anti-angiogenesis. Cancer Med. 2023; 12(11): 12608-12621.

[390]

Najaflou M, Bani F, Khosroushahi AY. Immunotherapeutic effect of photothermal-mediated exosomes secreted from breast cancer cells. Nanomedicine. 2023; 18(22): 1535-1552.

[391]

Liang Y, Duan L, Lu J, Xia J. Engineering exosomes for targeted drug delivery. Theranostics. 2021; 11(7): 3183-3195.

[392]

Fu S, Wang Y, Xia X, Zheng JC. Exosome engineering: current progress in cargo loading and targeted delivery. NanoImpact. 2020; 20: 100261.

[393]

Li T, Li X, Han G, et al. The therapeutic potential and clinical significance of exosomes as carriers of drug delivery system. Pharmaceutics. 2023; 15(21): 2022.

[394]

Tian J, Han Z, Song D, et al. Engineered exosome for drug delivery: recent development and clinical applications. Int J Nanomed. 2023; 18: 7923-7940.

[395]

Barrueto L, Caminero F, Cash L, Makris C, Lamichhane P, Deshmukh RR. Resistance to checkpoint inhibition in cancer immunotherapy. Transl Oncol. 2020; 13(3): 100738.

[396]

Kalbasi A, Ribas A. Tumour-intrinsic resistance to immune checkpoint blockade. Nat Rev Immunol. 2020; 20(1): 25-39.

[397]

Lin C, He H, Liu H, et al. Tumour-associated macrophages-derived CXCL8 determines immune evasion through autonomous PD-L1 expression in gastric cancer. Gut. 2019; 68(10): 1764-1773.

[398]

Diskin B, Adam S, Cassini MF, et al. PD-L1 engagement on T cells promotes self-tolerance and suppression of neighboring macrophages and effector T cells in cancer. Nat Immunol. 2020; 21(4): 442-454.

[399]

Zheng N, Wang T, Luo Q, et al. M2 macrophage-derived exosomes suppress tumor intrinsic immunogenicity to confer immunotherapy resistance. OncoImmunology. 2023; 12(1): 2210959.

[400]

Li J, Wang K, Yang C, et al. Tumor-associated macrophage-derived exosomal LINC01232 induces the immune escape in glioma by decreasing surface MHC-I expression. Adv Sci. 2023; 10(17): 2207067.

[401]

Liu Z, Hu C, Zheng L, et al. BMI1 promotes cholangiocarcinoma progression and correlates with antitumor immunity in an exosome-dependent manner. Cell Mol Life Sci. 2022; 79(9): 469.

[402]

Tian T, Han J, Huang J, Li S, Pang H. Hypoxia-induced intracellular and extracellular heat shock protein gp96 increases paclitaxel-resistance and facilitates immune evasion in breast cancer. Front Oncol. 2021; 11: 784777.

[403]

Zhu X, Shen H, Yin X, et al. Macrophages derived exosomes deliver miR-223 to epithelial ovarian cancer cells to elicit a chemoresistant phenotype. J Exp Clin Cancer Res. 2019; 38: 1-14.

[404]

Cheng H-Y, Hsieh C-H, Lin P-H, et al. Snail-regulated exosomal microRNA-21 suppresses NLRP3 inflammasome activity to enhance cisplatin resistance. J Immunother Cancer. 2022; 10(8): e004832.

[405]

Qin J, Luo M, Qian H, Chen W. Upregulated miR-182 increases drug resistance in cisplatin-treated HCC cell by regulating TP53INP1. Gene. 2014; 538(2): 342-347.

[406]

Qin X, Yu S, Zhou L, et al. Cisplatin-resistant lung cancer cell–derived exosomes increase cisplatin resistance of recipient cells in exosomal miR-100–5p-dependent manner. Int J Nanomed. 2017; 12: 3721-3733.

[407]

Wang B, Zhang Y, Ye M, Wu J, Ma L, Chen H. Cisplatin-resistant MDA-MB-231 cell-derived exosomes increase the resistance of recipient cells in an exosomal miR-423-5p-dependent manner. Curr Drug Metab. 2019; 20(10): 804-814.

[408]

Shi Q, Ji T, Ma Z, Tan Q, Liang J. Serum exosomes-based biomarker circ_0008928 regulates cisplatin sensitivity, tumor progression, and glycolysis metabolism by miR-488/HK2 axis in cisplatin-resistant nonsmall cell lung carcinoma. Cancer Biother Radio. 2021.

[409]

Wang D, Zhao C, Xu F, et al. Cisplatin-resistant NSCLC cells induced by hypoxia transmit resistance to sensitive cells through exosomal PKM2. Theranostics. 2021; 11(6): 2860.

[410]

Dai Y, Liu Y, Li J, Jin M, Yang H, Huang G. Shikonin inhibited glycolysis and sensitized cisplatin treatment in non-small cell lung cancer cells via the exosomal pyruvate kinase M2 pathway. Bioengineered. 2022; 13(5): 13906-13918.

[411]

Asare-Werehene M, Nakka K, Reunov A, et al. The exosome-mediated autocrine and paracrine actions of plasma gelsolin in ovarian cancer chemoresistance. Oncogene. 2020; 39(7): 1600-1616.

[412]

Xavier CP, Castro I, Caires HR, et al. Chitinase 3-like-1 and fibronectin in the cargo of extracellular vesicles shed by human macrophages influence pancreatic cancer cellular response to gemcitabine. Cancer Lett. 2021; 501: 210-223.

[413]

Chi Y, Xin H, Liu Z. Exosomal lncRNA UCA1 derived from pancreatic stellate cells promotes gemcitabine resistance in pancreatic cancer via the SOCS3/EZH2 axis. Front Oncol. 2021; 11: 671082.

[414]

Zeng Z, Zhao Y, Chen Q, et al. Hypoxic exosomal HIF-1α-stabilizing circZNF91 promotes chemoresistance of normoxic pancreatic cancer cells via enhancing glycolysis. Oncogene. 2021; 40(36): 5505-5517.

[415]

Alharbi M, Lai A, Sharma S, et al. Extracellular vesicle transmission of chemoresistance to ovarian cancer cells is associated with hypoxia-induced expression of glycolytic pathway proteins, and prediction of epithelial ovarian cancer disease recurrence. Cancers. 2021; 13(14): 3388.

[416]

Li C, Li X. Exosome-derived Circ_0094343 promotes chemosensitivity of colorectal cancer cells by regulating glycolysis via the miR-766-5p/TRIM67 Axis. Contrast Media Mol I. 2022; 2022: 2878557.

[417]

Ding C, Yi X, Chen X, et al. Warburg effect-promoted exosomal circ_0072083 releasing up-regulates NANGO expression through multiple pathways and enhances temozolomide resistance in glioma. J Exp Clin Cancer Res. 2021; 40(1): 164.

[418]

Wu P, Guo J, Yang H, Yuan D, Wang C, Wang Z. Exosomes derived from hypoxic glioma cells reduce the sensitivity of glioma cells to temozolomide through carrying miR-106a-5p. Drug Des Dev Ther. 2022; 16: 3589-3598.

[419]

Yin J, Ge X, Shi Z, et al. Extracellular vesicles derived from hypoxic glioma stem-like cells confer temozolomide resistance on glioblastoma by delivering miR-30b-3p. Theranostics. 2021; 11(4): 1763.

[420]

Han R, Guo H, Shi J, et al. Tumour microenvironment changes after osimertinib treatment resistance in non-small cell lung cancer. Eur J Cancer. 2023; 189: 112919.

[421]

Ochiai R, Hayashi K, Yamamoto H, et al. Plasma exosomal DOK3 reflects immunological states in lung tumor and predicts prognosis of gefitinib treatment. Cancer Sci. 2022; 113(11): 3960.

[422]

Wang C, Xu J, Yuan D, et al. Exosomes carrying ALDOA and ALDH3A1 from irradiated lung cancer cells enhance migration and invasion of recipients by accelerating glycolysis. Mol Cell Biochem. 2020; 469(1-2): 77-87.

[423]

Zhang Y, Liu X, Zeng L, et al. Exosomal protein angiopoietin-like 4 mediated radioresistance of lung cancer by inhibiting ferroptosis under hypoxic microenvironment. Brit J Cancer. 2022; 127(10): 1760-1772.

[424]

Yue X, Lan F, Xia T. Hypoxic glioma cell-secreted exosomal miR-301a activates Wnt/β-catenin signaling and promotes radiation resistance by targeting TCEAL7. Mol Ther. 2019; 27(11): 1939-1949.

[425]

Min H-Y, Lee H-Y. Molecular targeted therapy for anticancer treatment. Exp Mol Med. 2022; 54(10): 1670-1694.

[426]

Piper-Vallillo AJ, Sequist LV, Piotrowska Z. Emerging treatment paradigms for EGFR-mutant lung cancers progressing on osimertinib: a review. J Clin Oncol. 2020; 38(25): 2926-2936.

[427]

Ortiz A, Gui J, Zahedi F, et al. An interferon-driven oxysterol-based defense against tumor-derived extracellular vesicles. Cancer Cell. 2019; 35(1): 33-45. e6.

[428]

Ramakrishnan V, Xu B, Akers J, et al. Radiation-induced extracellular vesicle (EV) release of miR-603 promotes IGF1-mediated stem cell state in glioblastomas. EBioMedicine. 2020; 55: 102736.

[429]

Tang Y, Cui Y, Li Z, et al. Radiation-induced miR-208a increases the proliferation and radioresistance by targeting p21 in human lung cancer cells. J Exp Clin Canc Res. 2016; 35: 1-14.

[430]

Yan W, Jiang S. Immune cell-derived exosomes in the cancer-immunity cycle. Trends Cancer. 2020; 6(6): 506-517.

[431]

Kwak S, Lee J-Y, Kim MJ, et al. Combination of PD-1 checkpoint blockade and botulinum toxin type A1 improves antitumor responses in mouse tumor models of melanoma and colon carcinoma. Immunol Invest. 2023; 52(6): 749-766.

[432]

Shin JM, Lee CH, Son S, et al. Sulfisoxazole elicits robust antitumour immune response along with immune checkpoint therapy by inhibiting exosomal PD-L1. Adv Sci. 2022; 9(5): 2103245.

[433]

Bhatta M, Shenoy GN, Loyall JL, et al. Novel phosphatidylserine-binding molecule enhances antitumor T-cell responses by targeting immunosuppressive exosomes in human tumor microenvironments. J ImmunoTher Cancer. 2021; 9(10): e003148.

[434]

Yi B, Cheng H, Wyczechowska D, et al. Sulindac modulates the response of proficient MMR colorectal cancer to anti–PD-L1 immunotherapy. Mol Cancer Ther. 2021; 20(7): 1295-1304.

[435]

Jackson SE, Chester JD. Personalised cancer medicine. Int J Cancer. 2015; 137(2): 262-266.

[436]

Capdeville R, Buchdunger E, Zimmermann J, Matter A. Glivec (STI571, imatinib), a rationally developed, targeted anticancer drug. Nat Rev Drug Discov. 2002; 1(7): 493-502.

[437]

Rowley JD. A new consistent chromosomal abnormality in chronic myelogenous leukaemia identified by quinacrine fluorescence and Giemsa staining. Nature. 1973; 243(5405): 290-293.

[438]

Herbst RS, Soria J-C, Kowanetz M, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 2014; 515(7528): 563-567.

[439]

Muro K, Chung HC, Shankaran V, et al. Pembrolizumab for patients with PD-L1-positive advanced gastric cancer (KEYNOTE-012): a multicentre, open-label, phase 1b trial. Lancet Oncol. 2016; 17(6): 717-726.

[440]

Braig ZV. Personalized medicine: from diagnostic to adaptive. Biomed J. 2022; 45(1): 132-142.

[441]

van Dommelen SM, van der Meel R, van Solinge WW, Coimbra M, Vader P, Schiffelers RM. Cetuximab treatment alters the content of extracellular vesicles released from tumor cells. Nanomedicine. 2016; 11(8): 881-890.

[442]

Montermini L, Meehan B, Garnier D, et al. Inhibition of oncogenic epidermal growth factor receptor kinase triggers release of exosome-like extracellular vesicles and impacts their phosphoprotein and DNA content. J Biol Chem. 2015; 290(40): 24534-24546.

[443]

Zhao G, Jiang Y, Ma P, Wang S, Nie G, Li N. The clinical landscape of therapeutic cancer vaccines: the next breakthrough in cancer immunotherapy? Eur J Cancer. 2023; 181: 38-41.

[444]

Pitt JM, André F, Amigorena S, et al. Dendritic cell–derived exosomes for cancer therapy. J Clin Invest. 2016; 126(4): 1224-1232.

[445]

Chang C, Pei Y, Zhang C, Zhang W, Qin Y, Shi S. Combination therapy with dendritic cell loaded-exosomes supplemented with PD-1 inhibition at different time points have superior antitumor effect in hepatocellular carcinoma. Cancer Immunol Immun. 2023; 72(11): 3727-3738.

[446]

Zhu H, Wang K, Wang Z, et al. An efficient and safe MUC1-dendritic cell-derived exosome conjugate vaccine elicits potent cellular and humoral immunity and tumor inhibition in vivo. Acta Biomater. 2022; 138: 491-504.

[447]

Gassmann H, Schneider K, Evdokimova V, et al. Ewing sarcoma-derived extracellular vesicles impair dendritic cell maturation and function. Cells. 2021; 10(8): 2081.

[448]

Xia J, Miao Y, Wang X, Huang X, Dai J. Recent progress of dendritic cell-derived exosomes (Dex) as an anti-cancer nanovaccine. Biomed Pharmacother. 2022; 152: 113250.

[449]

Barnwal A, Gaur V, Sengupta A, Tyagi W, Das S, Bhattacharyya J. Tumor antigen-primed dendritic cell-derived exosome synergizes with colony stimulating factor-1 receptor inhibitor by modulating the tumor microenvironment and systemic immunity. ACS Biomater. Sci. Eng. 2023; 9(11): 6409-6424.

[450]

Morse MA, Garst J, Osada T, et al. A phase I study of dexosome immunotherapy in patients with advanced non-small cell lung cancer. J Transl Med. 2005; 3: 1-8.

[451]

Escudier B, Dorval T, Chaput N, et al. Vaccination of metastatic melanoma patients with autologous dendritic cell (DC) derived-exosomes: results of thefirst phase I clinical trial. J Transl Med. 2005; 3: 1-13.

[452]

Besse B, Charrier M, Lapierre V, et al. Dendritic cell-derived exosomes as maintenance immunotherapy after first line chemotherapy in NSCLC. Oncoimmunology. 2016; 5(4): e1071008.

[453]

Narita M, Kanda T, Abe T, et al. Immune responses in patients with esophageal cancer treated with SART1 peptide-pulsed dendritic cell vaccine. Int J Oncol. 2015; 46(4): 1699-1709.

[454]

Dai S, Wei D, Wu Z, et al. Phase I clinical trial of autologous ascites-derived exosomes combined with GM-CSF for colorectal cancer. Mol Ther. 2008; 16(4): 782-790.

[455]

Sun T, Li Y, Wu J, et al. Downregulation of exosomal MHC-I promotes glioma cells escaping from systemic immunosurveillance. Nanomed-Nanotechnol. 2022; 46: 102605.

[456]

Shin S, Jung I, Jung D, et al. Novel antitumor therapeutic strategy using CD4+ T cell-derived extracellular vesicles. Biomaterials. 2022; 289: 121765.

[457]

Zhou W-J, Zhang J, Xie F, et al. CD45RO-CD8+ T cell-derived exosomes restrict estrogen-driven endometrial cancer development via the ERβ/miR-765/PLP2/Notch axis. Theranostics. 2021; 11(11): 5330.

[458]

Wang X, Xiang Z, Liu Y, et al. Exosomes derived from Vδ2-T cells control Epstein-Barr virus-associated tumors and induce T cell antitumor immunity. Sci Transl Med. 2020; 12(563): eaaz3426.

[459]

Fu W, Lei C, Liu S, et al. CAR exosomes derived from effector CAR-T cells have potent antitumour effects and low toxicity. Nat Commun. 2019; 10(1): 4355.

[460]

Cheng L, Wang Y, Huang L. Exosomes from M1-polarized macrophages potentiate the cancer vaccine by creating a pro-inflammatory microenvironment in the lymph node. Mol Ther. 2017; 25(7): 1665-1675.

[461]

Kang YT, Niu Z, Hadlock T, et al. On-chip biogenesis of circulating NK cell-derived exosomes in non-small cell lung cancer exhibits antitumoral activity. Adv Sci. 2021; 8(6): 2003747.

[462]

Chen H, Zhao L, Meng Y, et al. Sulfonylurea receptor 1-expressing cancer cells induce cancer-associated fibroblasts to promote non-small cell lung cancer progression. Cancer Lett. 2022; 536: 215611.

[463]

Rui T, Wang K, Xiang A, et al. Serum exosome-derived piRNAs could be promising biomarkers for HCC diagnosis. Int J Nanomed. 2023; 18: 1989-2001.

[464]

Xu K, Jin Y, Li Y, Huang Y, Zhao R. Recent progress of exosome isolation and peptide recognition-guided strategies for exosome research. Front Chem. 2022; 10: 844124.

[465]

Lai JJ, Chau ZL, Chen SY, et al. Exosome processing and characterization approaches for research and technology development. Adv Sci. 2022; 9(15): 2103222.

[466]

Hu P, Wang X, Wei L, et al. Selective recognition of CdTe QDs and strand displacement signal amplification-assisted label-free and homogeneous fluorescence assay of nucleic acid and protein. J Mater Chem B. 2019; 7(31): 4778-4783.

[467]

Martins TS, Vaz M, Henriques AG. A review on comparative studies addressing exosome isolation methods from body fluids. Anal Bioanal Chem. 2023; 415(7): 1239-1263.

[468]

Liangsupree T, Multia E, Riekkola M-L. Modern isolation and separation techniques for extracellular vesicles. J Chromatogr A. 2021; 1636: 461773.

[469]

Weng Y, Sui Z, Shan Y, et al. Effective isolation of exosomes with polyethylene glycol from cell culture supernatant for in-depth proteome profiling. Analyst. 2016; 141(15): 4640-4646.

[470]

Chen J, Li P, Zhang T, et al. Review on strategies and technologies for exosome isolation and purification. Front Bioeng Biotech. 2022; 9: 811971.

[471]

Sharma P, Ludwig S, Muller L, et al. Immunoaffinity-based isolation of melanoma cell-derived exosomes from plasma of patients with melanoma. J Extracell Vesicles. 2018; 7(1): 1435138.

[472]

Hong CS, Muller L, Boyiadzis M, Whiteside TL. Isolation and characterization of CD34+ blast-derived exosomes in acute myeloid leukemia. PLoS One. 2014; 9(8): e103310.

[473]

Kim JY, Rhim W-K, Yoo Y-I, et al. Defined MSC exosome with high yield and purity to improve regenerative activity. J Tissue Eng. 2021; 12: 20417314211008626.

[474]

Lin S, Yu Z, Chen D, et al. Progress in microfluidics-based exosome separation and detection technologies for diagnostic applications. Small. 2020; 16(9): 1903916.

[475]

Yu Y, Li Y-T, Jin D, et al. Electrical and label-free quantification of exosomes with a reduced graphene oxide field effect transistor biosensor. Anal Chem. 2019; 91(16): 10679-10686.

[476]

Cosenza S, Ruiz M, Toupet K, Jorgensen C, Noël D. Mesenchymal stem cells derived exosomes and microparticles protect cartilage and bone from degradation in osteoarthritis. Sci Rep. 2017; 7(1): 16214.

[477]

Zhang Q, Wang H, Liu Q, et al. Exosomes as powerful biomarkers in cancer: recent advances in isolation and detection techniques. Int J Nanomed. 2024; 19: 1923-1949.

[478]

Zheng L, Li J, Li Y, et al. Empowering exosomes with aptamers for precision theranostics. Small Methods. 2024:2400551.

[479]

Richards T, Patel H, Patel K, Schanne F. Endogenous lipid carriers—bench-to-bedside roadblocks in production and drug loading of exosomes. Pharmaceuticals. 2023; 16(3): 421.

[480]

Kong L, Yang C, Zhang Z. Organism-generated biological vesicles in situ: an emerging drug delivery strategy. Adv Sci. 2023; 10(2): 2204178.

[481]

Hussen BM, Faraj GSH, Rasul MF, et al. Strategies to overcome the main challenges of the use of exosomes as drug carrier for cancer therapy. Cancer Cell Int. 2022; 22(1): 323.

[482]

Yang Q, Li S, Ou H, et al. Exosome-based delivery strategies for tumor therapy: an update on modification, loading, and clinical application. J Nanobiotechnol. 2024; 22(1): 41.

[483]

Chang W-H, Cerione RA, Antonyak MA. Extracellular vesicles and their roles in cancer progression. Methods Mol Biol. 2021; 2174: 143-170.

[484]

Li Q, Feng Q, Zhou H, et al. Mechanisms and therapeutic strategies of extracellular vesicles in cardiovascular diseases. MedComm. 2023; 4(6): e454.

[485]

Li X, Zhu Y, Wang Y, Xia X, Zheng JC. Neural stem/progenitor cell-derived extracellular vesicles: a novel therapy for neurological diseases and beyond. MedComm. 2023; 4(1): e214.

[486]

Caobi A, Nair M, Raymond AD. Extracellular vesicles in the pathogenesis of viral infections in humans. Viruses. 2020; 12(10): 1200.

RIGHTS & PERMISSIONS

2024 The Author(s). MedComm published by Sichuan International Medical Exchange & Promotion Association (SCIMEA) and John Wiley & Sons Australia, Ltd.

AI Summary AI Mindmap
PDF

224

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/