Extracellular vesicles in cancer drug resistance: Mechanistic insights and therapeutic implications

Jing Zuo , Han Yan , Siyuan Qin , Yonghao Yan , Shiqi Wang , Li Yang , Jinlin Yang , Jun Yang

MEDCOMM - Oncology ›› 2024, Vol. 3 ›› Issue (4) : e94

PDF
MEDCOMM - Oncology ›› 2024, Vol. 3 ›› Issue (4) : e94 DOI: 10.1002/mog2.94
REVIEW ARTICLE

Extracellular vesicles in cancer drug resistance: Mechanistic insights and therapeutic implications

Author information +
History +
PDF

Abstract

Despite significant advancements in the treatment of cancer, therapeutic resistance remains a major hurdle for the achievement of full cures. Besides being typically driven by intratumoral heterogeneity, such acquired resistance also relies heavily on cell-cell communication whereby extracellular vesicles (EVs) carrying resistance-related components can be transferred from drug-resistant cells or nontumorigenic members within tumor microenvironment (TME) to their sensitive neighbors. The cargo and membrane surface proteins of EVs derived from drug-resistant cells and TME cells carry abundant biological information, transferring therapy-resistant capacity to sensitive cancer cells. Hence, a deep understanding of the roles of EVs in cancer therapy resistance will facilitate identifying biomarkers and developing new approaches to restore therapy sensitivity. In this review, we summarize our current understanding regarding the causes and effects of EV-mediated cell-cell communication in drug resistance, with a particular notice on how various kinds of cargoes derived from different cells within TME are linked to the resistant phenotype. We also discuss how this knowledge can contribute to improvements in clinical practice, that is, the opportunities and challenges of EVs in functioning as potential biomarkers in predicting therapeutic resistance and, by extension, EV-based therapy in achieving deeper and longer-lasting clinical responses.

Keywords

cancer therapy resistance / cargo delivery / cell–cell communication / extracellular vesicles

Cite this article

Download citation ▾
Jing Zuo, Han Yan, Siyuan Qin, Yonghao Yan, Shiqi Wang, Li Yang, Jinlin Yang, Jun Yang. Extracellular vesicles in cancer drug resistance: Mechanistic insights and therapeutic implications. MEDCOMM - Oncology, 2024, 3(4): e94 DOI:10.1002/mog2.94

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

WardRA, FawellS, Floc’hN, FlemingtonV, McKerrecher D, SmithPD. Challenges and opportunities in cancer drug resistance. Chem Rev. 2021;121(6):3297-3351.

[2]

NussinovR, TsaiCJ, JangH. Anticancer drug resistance: an update and perspective. Drug Resist Updates. 2021;59:100796.

[3]

HerzogBH, Devarakonda S, GovindanR. Overcoming chemotherapy resistance in SCLC. J Thorac Oncol. 2021;16(12):2002-2015.

[4]

DiasMP, MoserSC, GanesanS, Jonkers J. Understanding and overcoming resistance to PARP inhibitors in cancer therapy. Nat Rev Clin Oncol. 2021;18(12):773-791.

[5]

VeselyMD, ZhangT, ChenL. Resistance mechanisms to anti-PD cancer immunotherapy. Annu Rev Immunol. 2022;40:45-74.

[6]

CottuP, Bièche I, AssayagF, et al. Acquired resistance to endocrine treatments is associated with tumor-specific molecular changes in patient-derived luminal breast cancer xenografts. Clin Cancer Res. 2014;20(16):4314-4325.

[7]

PassaroA, Brahmer J, AntoniaS, MokT, PetersS. Managing resistance to immune checkpoint inhibitors in lung cancer: treatment and novel strategies. J Clin Oncol. 2022;40(6):598-610.

[8]

ShiK, WangG, PeiJ, et al. Emerging strategies to overcome resistance to third-generation EGFR inhibitors. J Hematol Oncol. 2022;15(1):94.

[9]

QinS, JiangJ, LuY, et al. Emerging role of tumor cell plasticity in modifying therapeutic response. Signal Transduct Target Ther. 2020;5(1):228.

[10]

DhanyamrajuPK, SchellTD, AminS, Robertson GP. Drug-tolerant persister cells in cancer therapy resistance. Cancer Res. 2022;82(14):2503-2514.

[11]

CooperAJ, Sequist LV, LinJJ. Third-generation EGFR and ALK inhibitors: mechanisms of resistance and management. Nat Rev Clin Oncol. 2022;19(8):499-514.

[12]

FletcherJI, Williams RT, HendersonMJ, NorrisMD, HaberM. ABC transporters as mediators of drug resistance and contributors to cancer cell biology. Drug Resist Updates. 2016;26:1-9.

[13]

MarinJJG, BrizO, Rodríguez-MaciasG, Díez-MartínJL, MaciasRIR. Role of drug transport and metabolism in the chemoresistance of acute myeloid leukemia. Blood Rev. 2016;30(1):55-64.

[14]

LiuW, DuY, WenR, YangM, XuJ. Drug resistance to targeted therapeutic strategies in non-small cell lung cancer. Pharmacol Ther. 2020;206:107438.

[15]

QinS, LiB, MingH, Nice EC, ZouB, HuangC. Harnessing redox signaling to overcome therapeutic-resistant cancer dormancy. Biochim Biophys Acta Rev Cancer. 2022;1877(4):188749.

[16]

HeB, HuangZ, QinS, et al. Enhanced SLC35B2/SAV1 sulfation axis promotes tumor growth by inhibiting Hippo signaling in HCC. Hepatology. 2024.

[17]

LiB, JiangJ, AssarafYG, Xiao H, ChenZS, HuangC. Surmounting cancer drug resistance: new insights from the perspective of N6-methyladenosine RNA modification. Drug Resist Updates. 2020;53:100720.

[18]

ZuoJ, ZhangZ, LuoM, et al. Redox signaling at the crossroads of human health and disease. MedComm. 2022;3(2):e127.

[19]

BhattacharyaS, Mohanty A, AchuthanS, et al. Group behavior and emergence of cancer drug resistance. Trends Cancer. 2021;7(4):323-334.

[20]

ZhangQ, LiuRX, ChanKW, et al. Exosomal transfer of p-STAT3 promotes acquired 5-FU resistance in colorectal cancer cells. J Exp Clin Cancer Res. 2019;38(1):320.

[21]

XavierCPR, Belisario DC, RebeloR, et al. The role of extracellular vesicles in the transfer of drug resistance competences to cancer cells. Drug Resist Updates. 2022;62:100833.

[22]

XuJ, JiL, LiangY, et al. CircRNA-SORE mediates sorafenib resistance in hepatocellular carcinoma by stabilizing YBX1. Signal Transduct Target Ther. 2020;5(1):298.

[23]

SousaD, LimaRT, VasconcelosMH. Intercellular transfer of cancer drug resistance traits by extracellular vesicles. Trends Mol Med. 2015;21(10):595-608.

[24]

FattoreL, Sacconi A, ManciniR, CilibertoG. MicroRNA-driven deregulation of cytokine expression helps development of drug resistance in metastatic melanoma. Cytokine Growth Factor Rev. 2017;36:39-48.

[25]

WangSE. Extracellular vesicles in cancer therapy. Sem Cancer Biol. 2022;86:296-309.

[26]

CaoY, WangZ, YanY, et al. Enterotoxigenic bacteroidesfragilis promotes intestinal inflammation and malignancy by inhibiting exosome-packaged miR-149-3p. Gastroenterology. 2021;161(5):1552-1566.e12.

[27]

ZhaoS, MiY, ZhengB, et al. Highly-metastatic colorectal cancer cell released miR-181a-5p-rich extracellular vesicles promote liver metastasis by activating hepatic stellate cells and remodelling the tumour microenvironment. J Extracell Vesicles. 2022;11(1):e12186.

[28]

IzquierdoE, Vorholt D, BlakemoreS, et al. Extracellular vesicles and PD-L1 suppress macrophages, inducing therapy resistance in TP53-deficient B-cell malignancies. Blood. 2022;139(25):3617-3629.

[29]

SamuelM, Fonseka P, SanwlaniR, et al. Oral administration of bovine milk-derived extracellular vesicles induces senescence in the primary tumor but accelerates cancer metastasis. Nat Commun. 2021;12(1):3950.

[30]

WangX, HuangH, SzeKMF, et al. S100A10 promotes HCC development and progression via transfer in extracellular vesicles and regulating their protein cargos. Gut. 2023;72(7):1370-1384.

[31]

SandifordOA, Donnelly RJ, El-FarMH, et al. Mesenchymal stem cell-secreted extracellular vesicles instruct stepwise dedifferentiation of breast cancer cells into dormancy at the bone marrow perivascular region. Cancer Res. 2021;81(6):1567-1582.

[32]

SansoneP, SaviniC, KurelacI, 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.

[33]

ChengC, WangP, YangY, et al. Smoking-induced M2-TAMs, via circEML4 in EVs, promote the progression of NSCLC through ALKBH5-Regulated m6A modification of SOCS2 in NSCLC cells. Adv Sci (Weinh). 2023;10(22):e2300953.

[34]

PoçasJ, Marques C, GomesC, et al. Syndecan-4 is a maestro of gastric cancer cell invasion and communication that underscores poor survival. Proc Natl Acad Sci USA. 2023;120(20):e2214853120.

[35]

YamanaK, InoueJ, YoshidaR, et al. Extracellular vesicles derived from radioresistant oral squamous cell carcinoma cells contribute to the acquisition of radioresistance via the miR-503-3p-BAK axis. J Extracell Vesicles. 2021;10(14):e12169.

[36]

VeermanRE, Akpinar GG, OffensA, et al. Antigen-loaded extracellular vesicles induce responsiveness to anti-PD-1 and anti-PD-L1 treatment in a checkpoint refractory melanoma model. Cancer Immunol Res. 2023;11(2):217-227.

[37]

RubioK, Romero-Olmedo AJ, SarvariP, et al. Non-canonical integrin signaling activates EGFR and RAS-MAPK-ERK signaling in small cell lung cancer. Theranostics. 2023;13(8):2384-2407.

[38]

ChenQY, GaoB, TongD, Huang C. Crosstalk between extracellular vesicles and tumor-associated macrophage in the tumor microenvironment. Cancer Lett. 2023;552:215979.

[39]

RebeloR, XavierCPR, GiovannettiE, VasconcelosMH. Fibroblasts in pancreatic cancer: molecular and clinical perspectives. Trends Mol Med. 2023;29(6):439-453.

[40]

KalluriR, McAndrews KM. The role of extracellular vesicles in cancer. Cell. 2023;186(8):1610-1626.

[41]

ClancyJW, D’souza-Schorey C. Tumor-derived extracellular vesicles: multifunctional entities in the tumor microenvironment. Annu Rev Pathol: Mech Dis. 2023;18:205-229.

[42]

RaiA, FangH, ClaridgeB, Simpson RJ, GreeningDW. Proteomic dissection of large extracellular vesicle surfaceome unravels interactive surface platform. J Extracell Vesicles. 2021;10(13):e12164.

[43]

van NielG, CarterDRF, ClaytonA, Lambert DW, RaposoG, VaderP. Challenges and directions in studying cell-cell communication by extracellular vesicles. Nat Rev Mol Cell Biol. 2022;23(5):369-382.

[44]

ZhangDX, VuLT, IsmailNN, Le MTN, GrimsonA. Landscape of extracellular vesicles in the tumour microenvironment: interactions with stromal cells and with non-cell components, and impacts on metabolic reprogramming, horizontal transfer of neoplastic traits, and the emergence of therapeutic resistance. Sem Cancer Biol. 2021;74:24-44.

[45]

TricaricoC, ClancyJ, D’souza-SchoreyC. Biology and biogenesis of shed microvesicles. Small GTPases. 2017;8(4):220-232.

[46]

SegawaK, Yanagihashi Y, YamadaK, SuzukiC, Uchiyama Y, NagataS. Phospholipid flippases enable precursor B cells to flee engulfment by macrophages. Proc Natl Acad Sci USA. 2018;115(48):12212-12217.

[47]

BeerKB, Rivas-Castillo J, KuhnK, et al. Extracellular vesicle budding is inhibited by redundant regulators of TAT-5 flippase localization and phospholipid asymmetry. Proc Natl Acad Sci USA. 2018;115(6):E1127-E1136.

[48]

HanTW, YeW, BethelNP, et al. Chemically induced vesiculation as a platform for studying TMEM16F activity. Proc Natl Acad Sci USA. 2019;116(4):1309-1318.

[49]

DaiH, ZhangS, DuX, et al. RhoA inhibitor suppresses the production of microvesicles and rescues high ventilation induced lung injury. Int Immunopharmacol. 2019;72:74-81.

[50]

DixsonAC, DawsonTR, Di VizioD, Weaver AM. Context-specific regulation of extracellular vesicle biogenesis and cargo selection. Nat Rev Mol Cell Biol. 2023;24(7):454-476.

[51]

KalluriR, LeBleuVS. The biology, function, and biomedical applications of exosomes. Science. 2020;367(6478):eaau6977.

[52]

ColomboM, RaposoG, ThéryC. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu Rev Cell Dev Biol. 2014;30:255-289.

[53]

JenaBC, MandalM. The emerging roles of exosomes in anti-cancer drug resistance and tumor progression: an insight towards tumor-microenvironment interaction. Biochim Biophys Acta Rev Cancer. 2021;1875(1):188488.

[54]

HemlerME. Tetraspanin proteins mediate cellular penetration, invasion, and fusion events and define a novel type of membrane microdomain. Annu Rev Cell Dev Biol. 2003;19:397-422.

[55]

ZhouZ, YangZ, ZhouL, Yang M, HeS. The versatile roles of testrapanins in cancer from intracellular signaling to cell-cell communication: cell membrane proteins without ligands. Cell Biosci. 2023;13(1):59.

[56]

LariosJ, Mercier V, RouxA, GruenbergJ. ALIX-and ESCRT-III-dependent sorting of tetraspanins to exosomes. J Cell Biol. 2020;219(3):e201904113.

[57]

HenneWM, Buchkovich NJ, EmrSD. The ESCRT pathway. Dev Cell. 2011;21(1):77-91.

[58]

LiuJ, RenL, LiS, et al. The biology, function, and applications of exosomes in cancer. Acta Pharm Sin B. 2021;11(9):2783-2797.

[59]

AddiC, PresleA, FrémontS, et al. The Flemmingsome reveals an ESCRT-to-membrane coupling via ALIX/syntenin/syndecan-4 required for completion of cytokinesis. Nat Commun. 2020;11(1):1941.

[60]

BaiettiMF, ZhangZ, MortierE, et al. Syndecan-syntenin-ALIX regulates the biogenesis of exosomes. Nat Cell Biol. 2012;14(7):677-685.

[61]

van NielG, Charrin S, SimoesS, et al. The tetraspanin CD63 regulates ESCRT-independent and -dependent endosomal sorting during melanogenesis. Dev Cell. 2011;21(4):708-721.

[62]

ToribioV, Yáñez-Mó M. Tetraspanins interweave EV secretion, endosomal network dynamics and cellular metabolism. EJCB. 2022;101(3):151229.

[63]

GhossoubR, Chéry M, AudebertS, et al. Tetraspanin-6 negatively regulates exosome production. Proc Natl Acad Sci USA. 2020;117(11):5913-5922.

[64]

GuixFX, Sannerud R, BerditchevskiF, et al. Tetraspanin 6: a pivotal protein of the multiple vesicular body determining exosome release and lysosomal degradation of amyloid precursor protein fragments. Mol Neurodegener. 2017;12(1):25.

[65]

ThomSR, Bhopale VM, YuK, HuangW, KaneMA, MargolisDJ. Neutrophil microparticle production and inflammasome activation by hyperglycemia due to cytoskeletal instability. J Biol Chem. 2017;292(44):18312-18324.

[66]

GenschmerKR, Russell DW, LalC, et al. Activated PMN exosomes: pathogenic entities causing matrix destruction and disease in the lung. Cell. 2019;176(1-2):113-126.e15.

[67]

AnandS, FootN, AngCS, et al. Arrestin-domain containing protein 1 (Arrdc1) regulates the protein cargo and release of extracellular vesicles. Proteomics. 2018;18(17):e1800266.

[68]

SinhaS, Hoshino D, HongNH, et al. Cortactin promotes exosome secretion by controlling branched actin dynamics. J Cell Biol. 2016;214(2):197-213.

[69]

BécotA, Volgers C, van NielG. Transmissible endosomal intoxication: a balance between exosomes and lysosomes at the basis of intercellular amyloid propagation. Biomedicines. 2020;8(8):272.

[70]

WeiD, ZhanW, GaoY, et al. RAB31 marks and controls an ESCRT-independent exosome pathway. Cell Res. 2021;31(2):157-177.

[71]

BaiS, HouW, YaoY, et al. Exocyst controls exosome biogenesis via Rab11a. Mol Ther Nucleic Acids. 2022;27:535-546.

[72]

MessengerSW, WooSS, SunZ, MartinTFJ. A Ca2+-stimulated exosome release pathway in cancer cells is regulated by Munc13-4. J Cell Biol. 2018;217(8):2877-2890.

[73]

VillatoroAJ, Alcoholado C, Martín-AstorgaMC, FernándezV, CifuentesM, Becerra J. Comparative analysis and characterization of soluble factors and exosomes from cultured adipose tissue and bone marrow mesenchymal stem cells in canine species. Vet Immunol Immunopathol. 2019;208:6-15.

[74]

LiemM, AngCS, MathivananS. Inside front cover: insulin mediated activation of PI3K/Akt signalling pathway modifies the proteomic cargo of extracellular vesicles. Proteomics. 2017;17(23-24):17700182.

[75]

HoribeS, Tanahashi T, KawauchiS, MurakamiY, Rikitake Y. Mechanism of recipient cell-dependent differences in exosome uptake. BMC Cancer. 2018;18(1):47.

[76]

ChivetM, Javalet C, LaulagnierK, BlotB, Hemming FJ, SadoulR. Exosomes secreted by cortical neurons upon glutamatergic synapse activation specifically interact with neurons. J Extracell Vesicles. 2014;3:24722.

[77]

HeY, Rodrigues RM, WangX, et al. Neutrophil-to-hepatocyte communication via LDLR-dependent miR-223-enriched extracellular vesicle transfer ameliorates nonalcoholic steatohepatitis. J Clin Invest. 2021;131(3):e141513.

[78]

YinY, ChenH, WangY, Zhang L, WangX. Roles of extracellular vesicles in the aging microenvironment and age-related diseases. J Extracell Vesicles. 2021;10(12):e12154.

[79]

KimJW, JeongMH, YuHT, ParkYJ, KimHS, Chung KH. Fibrinogen on extracellular vesicles derived from polyhexamethylene guanidine phosphate-exposed mice induces inflammatory effects via integrin β. Ecotoxicol Environ Safety. 2023;252:114600.

[80]

NigriJ, LecaJ, TubianaSS, et al. CD9 mediates the uptake of extracellular vesicles from cancer-associated fibroblasts that promote pancreatic cancer cell aggressiveness. Sci Signal. 2022;15(745):eabg8191.

[81]

HoshinoA, KimHS, BojmarL, et al. Extracellular vesicle and particle biomarkers define multiple human cancers. Cell. 2020;182(4):1044-1061.e18.

[82]

DusoswaSA, Horrevorts SK, AmbrosiniM, et al. Glycan modification of glioblastoma-derived extracellular vesicles enhances receptor-mediated targeting of dendritic cells. J Extracell Vesicles. 2019;8(1):1648995.

[83]

VietriM, Radulovic M, StenmarkH. The many functions of ESCRTs. Nat Rev Mol Cell Biol. 2020;21(1):25-42.

[84]

JoshiBS, de Beer MA, GiepmansBNG, ZuhornIS. Endocytosis of extracellular vesicles and release of their cargo from endosomes. ACS Nano. 2020;14(4):4444-4455.

[85]

RaudenskaM, BalvanJ, MasarikM. Crosstalk between autophagy inhibitors and endosome-related secretory pathways: a challenge for autophagy-based treatment of solid cancers. Mol Cancer. 2021;20(1):140.

[86]

HeJ, HeJ, MinL, et al. Extracellular vesicles transmitted miR-31-5p promotes sorafenib resistance by targeting MLH1 in renal cell carcinoma. Int J Cancer. 2020;146(4):1052-1063.

[87]

KosakaN, Yoshioka Y, FujitaY, OchiyaT. Versatile roles of extracellular vesicles in cancer. J Clin Invest. 2016;126(4):1163-1172.

[88]

TeseiA, Arienti C, BossiG, et al. TP53 drives abscopal effect by secretion of senescence-associated molecular signals in non-small cell lung cancer. J Exp Clin Cancer Res. 2021;40(1):89.

[89]

RaghavanKS, Francescone R, Franco-BarrazaJ, et al. NetrinG1+ cancer-associated fibroblasts generate unique extracellular vesicles that support the survival of pancreatic cancer cells under nutritional stress. Cancer Res Commun. 2022;2(9):1017-1036.

[90]

MaS, Mangala LS, HuW, et al. CD63-mediated cloaking of VEGF in small extracellular vesicles contributes to anti-VEGF therapy resistance. Cell Rep. 2021;36(7):109549.

[91]

KobayashiM, Fujiwara K, TakahashiK, et al. Transforming growth factor-β-induced secretion of extracellular vesicles from oral cancer cells evokes endothelial barrier instability via endothelial-mesenchymal transition. Inflamm Regen. 2022;42(1):38.

[92]

ZhangS, LiaoX, ChenS, et al. Large oncosome-loaded VAPA promotes bone-tropic metastasis of hepatocellular carcinoma via formation of osteoclastic pre-metastatic niche. Adv Sci. 2022;9(31):e2201974.

[93]

WuB, ShiX, JiangM, Liu H. Cross-talk between cancer stem cells and immune cells: potential therapeutic targets in the tumor immune microenvironment. Mol Cancer. 2023;22(1):38.

[94]

NjockMS, O’Grady T, NivellesO, et al. Endothelial extracellular vesicles promote tumour growth by tumour-associated macrophage reprogramming. J Extracell Vesicles. 2022;11(6):e12228.

[95]

MusiA, Bongiovanni L. Extracellular vesicles in cancer drug resistance: implications on melanoma therapy. Cancers. 2023;15(4):1074.

[96]

Lopes-RodriguesV, Di Luca A, SousaD, et al. Multidrug resistant tumour cells shed more microvesicle-like EVs and less exosomes than their drug-sensitive counterpart cells. Biochim Biophys Acta. 2016;1860(3):618-627.

[97]

CuiX, ChenY, ZhaoL, Ding X. Extracellular vesicles derived from paclitaxel-sensitive nasopharyngeal carcinoma cells deliver miR-183-5p and impart paclitaxel sensitivity through a mechanism involving P-gp. Cell Biol Toxicol. 2023;39(6):2953-2970.

[98]

SerratìS, GuidaM, Di FonteR, et al. Circulating extracellular vesicles expressing PD1 and PD-L1 predict response and mediate resistance to checkpoint inhibitors immunotherapy in metastatic melanoma. Mol Cancer. 2022;21(1):20.

[99]

ChoiDS, ChoiDY, HongBS, et al. Quantitative proteomics of extracellular vesicles derived from human primary and metastatic colorectal cancer cells. J Extracell Vesicles. 2012;11:1.

[100]

Goler-BaronV, Assaraf YG. Overcoming multidrug resistance via photodestruction of ABCG2-rich extracellular vesicles sequestering photosensitive chemotherapeutics. PLoS One. 2012;7(4):e35487.

[101]

CarneiroBA, El-Deiry WS. Targeting apoptosis in cancer therapy. Nat Rev Clin Oncol. 2020;17(7):395-417.

[102]

ZhangS, LiuS, RenJ, et al. Tumor-derived extracellular vesicles confer 5-fluorouracil resistance in esophageal cancer via long noncoding RNA AC116025.2 delivery. Mol Carcinog. 2022;61(12):1177-1190.

[103]

ChaoF, ZhangY, LvL, et al. Extracellular vesicles derived circSH3PXD2A inhibits chemoresistance of small cell lung cancer by miR-375-3p/YAP1. Int J Nanomedicine. 2023;18:2989-3006.

[104]

YangE, WangL, JinW, et al. PTRF/Cavin-1 enhances chemo-resistance and promotes temozolomide efflux through extracellular vesicles in glioblastoma. Theranostics. 2022;12(9):4330-4347.

[105]

AbadE, Lyakhovich A. Movement of mitochondria with mutant DNA through extracellular vesicles helps cancer cells acquire chemoresistance. ChemMedChem. 2022;17(4):e202100642.

[106]

WuY, SongY, WangR, Wang T. Molecular mechanisms of tumor resistance to radiotherapy. Mol Cancer. 2023;22(1):96.

[107]

TortoliciF, Vumbaca S, IncocciatiB, et al. Ionizing radiation-induced extracellular vesicle release promotes AKT-associated survival response in SH-SY5Y neuroblastoma cells. Cells. 2021;10(1):107.

[108]

ZhangJ, HuangD, SawPE, Song E. Turning cold tumors hot: from molecular mechanisms to clinical applications. Trends Immunol. 2022;43(7):523-545.

[109]

XieN, ShenG, GaoW, HuangZ, HuangC, Fu L. Neoantigens: promising targets for cancer therapy. Signal Transduct Target Ther. 2023;8(1):9.

[110]

VerganiE, DaveriE, VallacchiV, et al. Extracellular vesicles in anti-tumor immunity. Sem Cancer Biol. 2022;86:64-79.

[111]

ZhangL, ZhouS, ZhouT, Li X, TangJ. Potential of the tumor derived extracellular vesicles carrying the miR 125b 5p target TNFAIP3 in reducing the sensitivity of diffuse large B cell lymphoma to rituximab. Int J Oncol. 2021;58(6):31.

[112]

Bougras-CartronG, Nadaradjane A, JoallandMP, Lalier-BretaudeauL, Raimbourg J, CartronPF. Adenosine methylation level of miR-125a-5p promotes anti-PD-1 therapy escape through the regulation of IGSF11/VSIG3 expression. Cancers. 2023;15(12):3188.

[113]

ZhouJ, QuG, ZhangG, et al. Glycerol kinase 5 confers gefitinib resistance through SREBP1/SCD1 signaling pathway. J Exp Clin Cancer Res. 2019;38(1):96.

[114]

LunavatTR, ChengL, EinarsdottirBO, et al. BRAFV600inhibition alters the microRNA cargo in the vesicular secretome of malignant melanoma cells. Proc Natl Acad Sci USA. 2017;114(29):E5930-E5939.

[115]

RodríguezY, Unno K, TruicaMI, et al. A genome-wide CRISPR activation screen identifies PRRX2 as a regulator of enzalutamide resistance in prostate cancer. Cancer Res. 2022;82(11):2110-2123.

[116]

AntonarakisES, LuC, WangH, et al. AR-V7 and resistance to enzalutamide and abiraterone in prostate cancer. N Engl J Med. 2014;371(11):1028-1038.

[117]

LeeHC, OuCH, HuangYC, et al. YAP1 overexpression contributes to the development of enzalutamide resistance by induction of cancer stemness and lipid metabolism in prostate cancer. Oncogene. 2021;40(13):2407-2421.

[118]

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

[119]

ChenY, McAndrews KM, KalluriR. Clinical and therapeutic relevance of cancer-associated fibroblasts. Nat Rev Clin Oncol. 2021;18(12):792-804.

[120]

KangSH, OhSY, LeeKY, et al. Differential effect of cancer-associated fibroblast-derived extracellular vesicles on cisplatin resistance in oral squamous cell carcinoma via miR-876-3p. Theranostics. 2024;14(2):460-479.

[121]

TanD, LiG, ZhangP, Peng C, HeB. LncRNA SNHG12 in extracellular vesicles derived from carcinoma-associated fibroblasts promotes cisplatin resistance in non-small cell lung cancer cells. Bioengineered. 2022;13(1):1838-1857.

[122]

ZhangY, PanQ, ShaoZ. Extracellular vesicles derived from cancer-associated fibroblasts carry tumor-promotive microRNA-1228-3p to enhance the resistance of hepatocellular carcinoma cells to sorafenib. Hum Cell. 2023;36(1):296-311.

[123]

MantovaniA, Allavena P, MarchesiF, GarlandaC. Macrophages as tools and targets in cancer therapy. Nat Rev Drug Discov. 2022;21(11):799-820.

[124]

ChenF, ChenJ, YangL, et al. Extracellular vesicle-packaged HIF-1α-stabilizing lncRNA from tumour-associated macrophages regulates aerobic glycolysis of breast cancer cells. Nat Cell Biol. 2019;21(4):498-510.

[125]

ChenC, ZhangL, RuanZ. GATA3 encapsulated by tumor-associated macrophage-derived extracellular vesicles promotes immune escape and chemotherapy resistance of ovarian cancer cells by upregulating the CD24/Siglec-10 axis. Mol Pharmaceutics. 2023;20(2):971-986.

[126]

LanT, LuoM, WeiX. Mesenchymal stem/stromal cells in cancer therapy. J Hematol Oncol. 2021;14(1):195.

[127]

WengZ, ZhangB, WuC, et al. Therapeutic roles of mesenchymal stem cell-derived extracellular vesicles in cancer. J Hematol Oncol. 2021;14(1):136.

[128]

ZhuT, HuZ, WangZ, et al. microRNA-301b-3p from mesenchymal stem cells-derived extracellular vesicles inhibits TXNIP to promote multidrug resistance of gastric cancer cells. Cell Biol Toxicol. 2022;39(5):1923-1937.

[129]

WuR, SuZ, ZhaoL, et al. Extracellular vesicle-loaded oncogenic lncRNA NEAT1 from adipose-derived mesenchymal stem cells confers gemcitabine resistance in pancreatic cancer via miR-491-5p/Snail/SOCS3 axis. Stem Cells Int. 2023;2023:1-21.

[130]

WangX, JiangL, LiuQ. miR-18a-5p derived from mesenchymal stem cells-extracellular vesicles inhibits ovarian cancer cell proliferation, migration, invasion, and chemotherapy resistance. J Transl Med. 2022;20(1):258.

[131]

RuanZ, LuL, ZhangL, Dong M. Bone marrow stromal cells-derived microRNA-181-containing extracellular vesicles inhibit ovarian cancer cell chemoresistance by downregulating MEST via the Wnt/β-catenin signaling pathway. Cancer Gene Ther. 2021;28(7-8):785-798.

[132]

RohlenovaK, VeysK, Miranda-SantosI, De BockK, Carmeliet P. Endothelial cell metabolism in health and disease. Trends Cell Biol. 2018;28(3):224-236.

[133]

ZengY, YaoX, LiuX, et al. Anti-angiogenesis triggers exosomes release from endothelial cells to promote tumor vasculogenesis. J Extracell Vesicles. 2019;8(1):1629865.

[134]

KoSY, LeeW, KennyHA, et al. Cancer-derived small extracellular vesicles promote angiogenesis by heparin-bound, bevacizumab-insensitive VEGF, independent of vesicle uptake. Commun Biol. 2019;2:386.

[135]

KoSY, NaoraH. Extracellular vesicle membrane-associated proteins: emerging roles in tumor angiogenesis and anti-angiogenesis therapy resistance. Int J Mol Sci. 2020;21(15):5418.

[136]

SatoS, Vasaikar S, EskarosA, et al. EPHB2 carried on small extracellular vesicles induces tumor angiogenesis via activation of ephrin reverse signaling. JCI Insight. 2019;4(23):e132447.

[137]

ZhangY, ZhangZ. The history and advances in cancer immunotherapy: understanding the characteristics of tumor-infiltrating immune cells and their therapeutic implications. Cell Mol Immunol. 2020;17(8):807-821.

[138]

XuZ, ChenY, MaL, et al. Role of exosomal non-coding RNAs from tumor cells and tumor-associated macrophages in the tumor microenvironment. Mol Ther. 2022;30(10):3133-3154.

[139]

WangJ, LongR, HanY. The role of exosomes in the tumour microenvironment on macrophage polarisation. Biochim Biophys Acta Rev Cancer. 2022;1877(6):188811.

[140]

BernareggiD, XieQ, PragerBC, et al. CHMP2A regulates tumor sensitivity to natural killer cell-mediated cytotoxicity. Nat Commun. 2022;13(1):1899.

[141]

LeeCH, BaeJH, ChoeEJ, et al. Macitentan improves antitumor immune responses by inhibiting the secretion of tumor-derived extracellular vesicle PD-L1. Theranostics. 2022;12(5):1971-1987.

[142]

ZhaoX, YuanC, WangmoD, Subramanian S. Tumor-secreted extracellular vesicles regulate T-cell costimulation and can be manipulated to induce tumor-specific T-cell responses. Gastroenterology. 2021;161(2):560-574.e11.

[143]

ChallagundlaKB, WisePM, NevianiP, et al. Exosome-mediated transfer of microRNAs within the tumor microenvironment and neuroblastoma resistance to chemotherapy. J Natl Cancer Inst. 2015;107(7):djv135.

[144]

NingT, LiJ, HeY, et al. Exosomal miR-208b related with oxaliplatin resistance promotes Treg expansion in colorectal cancer. Mol Ther. 2021;29(9):2723-2736.

[145]

LiJ, ShuX, XuJ, et al. S100A9-CXCL12 activation in BRCA1-mutant breast cancer promotes an immunosuppressive microenvironment associated with resistance to immunotherapy. Nat Commun. 2022;13(1):1481.

[146]

HuberV, Vallacchi V, FlemingV, et al. Tumor-derived microRNAs induce myeloid suppressor cells and predict immunotherapy resistance in melanoma. J Clin Invest. 2018;128(12):5505-5516.

[147]

DengZ, RongY, TengY, et al. Exosomes miR-126a released from MDSC induced by DOX treatment promotes lung metastasis. Oncogene. 2017;36(5):639-651.

[148]

YuT, WangX, ZhiT, et al. Delivery of MGMT mRNA to glioma cells by reactive astrocyte-derived exosomes confers a temozolomide resistance phenotype. Cancer Lett. 2018;433:210-220.

[149]

WuX, ZhouZ, XuS, et al. Extracellular vesicle packaged LMP1-activated fibroblasts promote tumor progression via autophagy and stroma-tumor metabolism coupling. Cancer Lett. 2020;478:93-106.

[150]

HanC, Godfrey V, LiuZ, et al. The AIM2 and NLRP3 inflammasomes trigger IL-1-mediated antitumor effects during radiation. Sci Immunol. 2021;6(59):eabc6998.

[151]

XiaoH, DuX, TaoZ, et al. Taurine inhibits ferroptosis mediated by the crosstalk between tumor cells and tumor-associated macrophages in prostate cancer. Adv Sci. 2024;11(3):e2303894.

[152]

HeideggerS, Stritzke F, DahlS, et al. Targeting nucleic acid sensors in tumor cells to reprogram biogenesis and RNA cargo of extracellular vesicles for T cell-mediated cancer immunotherapy. Cell Rep Med. 2023;4(9):101171.

[153]

SimonT, Jackson E, GiamasG. Breaking through the glioblastoma micro-environment via extracellular vesicles. Oncogene. 2020;39(23):4477-4490.

[154]

LeeY, GrahamP, LiY. Extracellular vesicles as a novel approach for breast cancer therapeutics. Cancer Lett. 2023;555:216036.

[155]

ChenX, DengY, NiuR, et al. Cancer-derived small extracellular vesicles PICKER. Anal Chem. 2022;94(38):13019-13027.

[156]

YuHA, PerezL, ChangQ, et al. A phase 1/2 trial of ruxolitinib and erlotinib in patients with EGFR-mutant lung adenocarcinomas with acquired resistance to erlotinib. J Thorac Oncol. 2017;12(1):102-109.

[157]

MartinezVG, O’Neill S, SalimuJ, et al. Resistance to HER2-targeted anti-cancer drugs is associated with immune evasion in cancer cells and their derived extracellular vesicles. Oncoimmunology. 2017;6(12):e1362530.

[158]

de Miguel-PerezD, Russo A, GunasekaranM, et al. Baseline extracellular vesicle TGF-βis a predictive biomarker for response to immune checkpoint inhibitors and survival in non-small cell lung cancer. Cancer. 2023;129(4):521-530.

[159]

CiravoloV, HuberV, GhediniGC, et al. Potential role of HER2-overexpressing exosomes in countering trastuzumab-based therapy. J Cell Physiol. 2012;227(2):658-667.

[160]

WangT, NingK, LuT, et al. Increasing circulating exosomes-carrying TRPC5 predicts chemoresistance in metastatic breast cancer patients. Cancer Sci. 2017;108(3):448-454.

[161]

ChenG, HuangAC, ZhangW, et al. Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature. 2018;560(7718):382-386.

[162]

BrennanK, Iversen KF, Blanco-FernándezA, LundT, Plesner T, Mc GeeMM. Extracellular vesicles isolated from plasma of multiple myeloma patients treated with daratumumab express CD38, PD-L1, and the complement inhibitory proteins CD55 and CD59. Cells. 2022;11(21):3365.

[163]

PorcelliL, GuidaM, De SummaS, et al. uPAR+extracellular vesicles: a robust biomarker of resistance to checkpoint inhibitor immunotherapy in metastatic melanoma patients. J Immunother Cancer. 2021;9(5):e002372.

[164]

Del ReM, Bertolini I, CrucittaS, et al. Overexpression of TK1 and CDK9 in plasma-derived exosomes is associated with clinical resistance to CDK4/6 inhibitors in metastatic breast cancer patients. Breast Cancer Res Treat. 2019;178(1):57-62.

[165]

YangS, WangD, LiJ, et al. Predictive role of GSTP1-containing exosomes in chemotherapy-resistant breast cancer. Gene. 2017;623:5-14.

[166]

Ampudia-MesiasE, El-Hadad S, CameronCS, et al. SRPX emerges as a potential tumor marker in the extracellular vesicles of glioblastoma. Cancers. 2022;14(8):1984.

[167]

ChangWH, NguyenTTT, HsuCH, et al. KRAS-dependent cancer cells promote survival by producing exosomes enriched in Survivin. Cancer Lett. 2021;517:66-77.

[168]

BarzegarM, Allahbakhshian Farsan M, AmiriV, et al. AML-derived extracellular vesicles confer de novo chemoresistance to leukemic myeloblast cells by promoting drug export genes expression and ROS inhibition. Iran J Pharm Res. 2021;20(1):384-397.

[169]

ChenY, WangL, ZhuY, et al. Breast cancer resistance protein (BCRP)-containing circulating microvesicles contribute to chemoresistance in breast cancer. Oncol Lett. 2015;10(6):3742-3748.

[170]

ČerneK, KelharN, ResnikN, et al. Characteristics of extracellular vesicles from a high-grade serous ovarian cancer cell line derived from a platinum-resistant patient as a potential tool for aiding the prediction of responses to chemotherapy. Pharmaceuticals. 2023;16(6):907.

[171]

Asare-WereheneM, HunterRA, GerberE, et al. The application of an extracellular vesicle-based biosensor in early diagnosis and prediction of chemoresponsiveness in ovarian cancer. Cancers. 2023;15(9):2566.

[172]

MillerCE, XuF, ZhaoY, et al. Hydrogen peroxide promotes the production of radiation-derived EVs containing mitochondrial proteins. Antioxidants. 2022;11(11):2119.

[173]

JennrichS, PelzerM, TertelT, et al. CD9-and CD81-positive extracellular vesicles provide a marker to monitor glioblastoma cell response to photon-based and proton-based radiotherapy. Front Oncol. 2022;12:947439.

[174]

KatoT, Mizutani K, KawakamiK, FujitaY, EharaH, ItoM. CD44v8-10 mRNA contained in serum exosomes as a diagnostic marker for docetaxel resistance in prostate cancer patients. Heliyon. 2020;6(7):e04138.

[175]

ShaoH, ChungJ, LeeK, et al. Chip-based analysis of exosomal mRNA mediating drug resistance in glioblastoma. Nat Commun. 2015;6:6999.

[176]

Del ReM, BiascoE, CrucittaS, et al. The detection of androgen receptor splice variant 7 in plasma-derived exosomal RNA strongly predicts resistance to hormonal therapy in metastatic prostate cancer patients. Eur Urol. 2017;71(4):680-687.

[177]

AlharbiM, SharmaS, GuanzonD, et al. miRNa signature in small extracellular vesicles and their association with platinum resistance and cancer recurrence in ovarian cancer. Nanomedicine. 2020;28:102207.

[178]

ZhangZ, ZhangL, YuG, et al. Exosomal miR-1246 and miR-155 as predictive and prognostic biomarkers for trastuzumab-based therapy resistance in HER2-positive breast cancer. Cancer Chemother Pharmacol. 2020;86(6):761-772.

[179]

CannistraciA, Federici G, AddarioA, et al. C-Met/miR-130b axis as novel mechanism and biomarker for castration resistance state acquisition. Oncogene. 2017;36(26):3718-3728.

[180]

CorcoranC, RaniS, O’DriscollL. miR-34a is an intracellular and exosomal predictive biomarker for response to docetaxel with clinical relevance to prostate cancer progression. Prostate. 2014;74(13):1320-1334.

[181]

YuwenD, MaY, WangD, et al. Prognostic role of circulating exosomal miR-425-3p for the response of NSCLC to platinum-based chemotherapy. Cancer Epidemiol Biomarkers Prev. 2019;28(1):163-173.

[182]

ShiQ, JiT, MaZ, TanQ, LiangJ. 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 Radiopharm. 2023;38(8):558-571.

[183]

YangY, ZhangR, DuJ, et al. Predictive role of UCA1-containing exosomes in cetuximab-resistant colorectal cancer. Cancer Cell Int. 2018;18:164.

[184]

ChengAN, ChengLC, KuoCL, et al. Mitochondrial lon-induced mtDNA leakage contributes to PD-L1-mediated immunoescape via STING-IFN signaling and extracellular vesicles. J Immunother Cancer. 2020;8(2):e001372.

[185]

OldriniB, Vaquero-Siguero N, MuQ, et al. MGMT genomic rearrangements contribute to chemotherapy resistance in gliomas. Nat Commun. 2020;11(1):3883.

[186]

GalbiatiS, DaminF, BrambillaD, et al. Small EVs-associated DNA as complementary biomarker to circulating tumor DNA in plasma of metastatic colorectal cancer patients. Pharmaceuticals. 2021;14(2):128.

[187]

CrescitelliR, FilgesS, KarimiN, et al. Extracellular vesicle DNA from human melanoma tissues contains cancer-specific mutations. Front Cell Dev Biol. 2022;10:1028854.

[188]

HurJY, KimHJ, LeeJS, et al. Extracellular vesicle-derived DNA for performing EGFR genotyping of NSCLC patients. Mol Cancer. 2018;17(1):15.

[189]

TrajkovicK, HsuC, ChiantiaS, et al. Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science. 2008;319(5867):1244-1247.

[190]

CaoYL, ZhuangT, XingBH, Li N, LiQ. Exosomal DNMT1 mediates cisplatin resistance in ovarian cancer. Cell Biochem Funct. 2017;35(6):296-303.

[191]

PoggioM, HuT, PaiCC, et al. Suppression of exosomal PD-L1 induces systemic anti-tumor immunity and memory. Cell. 2019;177(2):414-427.e13.

[192]

KochR, AungT, VogelD, 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.

[193]

TuC, DuZ, ZhangH, et al. Endocytic pathway inhibition attenuates extracellular vesicle-induced reduction of chemosensitivity to bortezomib in multiple myeloma cells. Theranostics. 2021;11(5):2364-2380.

[194]

Rodrigues-JuniorDM, Tan SS, LimSK, et al. Circulating extracellular vesicle-associated TGFβ3 modulates response to cytotoxic therapy in head and neck squamous cell carcinoma. Carcinogenesis. 2019;40(12):1452-1461.

[195]

LentzMR. Continuous whole blood UltraPheresis procedure in patients with metastatic cancer. J Biol Response Mod. 1989;8(5):511-527.

[196]

MarleauAM, ChenCS, JoyceJA, Tullis RH. Exosome removal as a therapeutic adjuvant in cancer. J Transl Med. 2012;10:134.

[197]

ZhangL, YuD. Exosomes in cancer development, metastasis, and immunity. Biochim Biophys Acta Rev Cancer. 2019;1871(2):455-468.

[198]

BinenbaumY, Fridman E, YaariZ, et al. Transfer of miRNA in macrophage-derived exosomes induces drug resistance in pancreatic adenocarcinoma. Cancer Res. 2018;78(18):5287-5299.

[199]

PengD, WangH, LiL, et al. miR-34c-5p promotes eradication of acute myeloid leukemia stem cells by inducing senescence through selective RAB27B targeting to inhibit exosome shedding. Leukemia. 2018;32(5):1180-1188.

[200]

YuanZ, Kolluri KK, GowersKHC, JanesSM. TRAIL delivery by MSC-derived extracellular vesicles is an effective anticancer therapy. J Extracell Vesicles. 2017;6(1):1265291.

[201]

SonSH, Gangadaran P, AhnBC. A novel strategy of transferring NIS protein to cells using extracellular vesicles leads to increase in iodine uptake and cytotoxicity. Int J Nanomedicine. 2019;14:1779-1787.

[202]

HeC, Jaffar Ali D, QiY, et al. Engineered extracellular vesicles mediated CRISPR-induced deficiency of IQGAP1/FOXM1 reverses sorafenib resistance in HCC by suppressing cancer stem cells. J Nanobiotechnology. 2023;21(1):154.

[203]

MunozJL, BlissSA, GrecoSJ, Ramkissoon SH, LigonKL, RameshwarP. Delivery of functional anti-miR-9 by mesenchymal stem cell-derived exosomes to glioblastoma multiforme cells conferred chemosensitivity. Mol Ther Nucleic Acids. 2013;2:e126.

[204]

LiangG, ZhuY, AliDJ, et al. Engineered exosomes for targeted co-delivery of miR-21 inhibitor and chemotherapeutics to reverse drug resistance in colon cancer. J Nanobiotechnology. 2020;18(1):10.

[205]

LinCC, WuCY, TsengJTC, et al. Extracellular vesicle miR-200c enhances gefitinib sensitivity in heterogeneous EGFR-Mutant NSCLC. Biomedicines. 2021;9(3):243.

[206]

XiaoZ, LiuY, LiQ, et al. EVs delivery of miR-1915-3p improves the chemotherapeutic efficacy of oxaliplatin in colorectal cancer. Cancer Chemother Pharmacol. 2021;88(6):1021-1031.

[207]

Rodrigues-JuniorDM, Pelarin MFA, NaderHB, VettoreAL, PinhalMAS. MicroRNA-1252-5p associated with extracellular vesicles enhances bortezomib sensitivity in multiple myeloma cells by targeting heparanase. Onco Targets Ther. 2021;14:455-467.

[208]

DiaoY, WangG, ZhuB, et al. Loading of “cocktail siRNAs”into extracellular vesicles via TAT-DRBD peptide for the treatment of castration-resistant prostate cancer. Cancer Biol Ther. 2022;23(1):163-172.

[209]

MatsudaA, Ishiguro K, YanIK, PatelT. Extracellular vesicle-based therapeutic targeting of β-Catenin to modulate anticancer immune responses in hepatocellular cancer. Hepatol Commun. 2019;3(4):525-541.

[210]

ZhangQ, ZhangH, NingT, et al. Exosome-delivered c-met siRNA could reverse chemoresistance to cisplatin in gastric cancer. Int J Nanomedicine. 2020;15:2323-2335.

[211]

DengJ, KeH. Overcoming the resistance of hepatocellular carcinoma to PD-1/PD-L1 inhibitor and the resultant immunosuppression by CD38 siRNA-loaded extracellular vesicles. Oncoimmunology. 2023;12(1):2152635.

[212]

LiuJ, YeZ, XiangM, et al. Functional extracellular vesicles engineered with lipid-grafted hyaluronic acid effectively reverse cancer drug resistance. Biomaterials. 2019;223:119475.

[213]

XiaoQ, ZhaoW, WuC, et al. Lemon-derived extracellular vesicles nanodrugs enable to efficiently overcome cancer multidrug resistance by endocytosis-triggered energy dissipation and energy production reduction. Adv Sci. 2022;9(20):e2105274.

[214]

RajendranRL, PaudelS, GangadaranP, et al. Extracellular vesicles act as nano-transporters of tyrosine kinase inhibitors to revert iodine avidity in thyroid cancer. Pharmaceutics. 2021;13(2):248.

[215]

ZuoJ, ZhangZ, LiM, et al. The crosstalk between reactive oxygen species and noncoding RNAs: from cancer code to drug role. Mol Cancer. 2022;21(1):30.

[216]

DuanJ, HuangZ, QinS, et al. Oxidative stress induces extracellular vesicle release by upregulation of HEXB to facilitate tumour growth in experimental hepatocellular carcinoma. J Extracell Vesicles. 2024;13(7):e12468.

[217]

KaurS, Nathani A, SinghM. Exosomal delivery of cannabinoids against cancer. Cancer Lett. 2023;566:216243.

[218]

MaJ, ZhangY, TangK, et al. Reversing drug resistance of soft tumor-repopulating cells by tumor cell-derived chemotherapeutic microparticles. Cell Res. 2016;26(6):713-727.

RIGHTS & PERMISSIONS

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

AI Summary AI Mindmap
PDF

349

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/