Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Vesiclemia: counting on extracellular vesicles for glioblastoma patients

Abstract

Although rare, glioblastoma is a devastating tumor of the central nervous system characterized by a poor survival and an extremely dark prognosis, making its diagnosis, treatment, and monitoring highly challenging. Numerous studies have highlighted extracellular vesicles (EVs) as key players of tumor growth, invasiveness, and resistance, as they carry oncogenic material. Moreover, EVs have been shown to communicate locally in a paracrine way but also at remote throughout the organism. Indeed, recent reports demonstrated the presence of brain tumor-derived EVs into body fluids such as plasma and cerebrospinal fluid. Fluid-associated EVs have indeed been suspected to reflect quantitative and qualitative information about the status and fate of the tumor and can potentially act as a resource for noninvasive biomarkers that might assist in diagnosis, treatment, and follow-up of glioblastoma patients. Here, we coined the name vesiclemia to define the concentration of plasmatic EVs, an intuitive term to be directly transposed in the clinical jargon.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Biogenesis, nomenclature, and specific markers of extracellular vesicles.
Fig. 2: Separation and characterization of extracellular vesicles.
Fig. 3: Possible functions of extracellular vesicles within the glioblastoma microenvironment.
Fig. 4: Use of plasmatic extracellular vesicles in clinics.

Similar content being viewed by others

References

  1. Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJB, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352:987–96.

    CAS  PubMed  Google Scholar 

  2. Vijapura C, Saad Aldin E, Capizzano AA, Policeni B, Sato Y, Moritani T. Genetic syndromes associated with central nervous system tumors. Radiographics. 2017;37:258–80.

    PubMed  Google Scholar 

  3. Bondy ML, Scheurer ME, Malmer B, Barnholtz-Sloan JS, Davis FG, Il’yasova D, et al. Brain tumor epidemiology: consensus from the brain tumor epidemiology consortium. Cancer. 2008;113:1953–68.

    PubMed  Google Scholar 

  4. Linos E, Raine T, Alonso A, Michaud D. Atopy and risk of brain tumors: a meta-analysis. J Natl Cancer Inst. 2007;99:1544–50.

    PubMed  Google Scholar 

  5. Lu-Emerson C, Duda DG, Emblem KE, Taylor JW, Gerstner ER, Loeffler JS, et al. Lessons from anti-vascular endothelial growth factor and anti-vascular endothelial growth factor receptor trials in patients with glioblastoma. J Clin Oncol. 2015;33:1197–213.

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Chinot OL, Wick W, Mason W, Henriksson R, Saran F, Nishikawa R, et al. Bevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastoma. N Engl J Med. 2014;370:709–22.

    CAS  PubMed  Google Scholar 

  7. Friedman HS, Prados MD, Wen PY, Mikkelsen T, Schiff D, Abrey LE, et al. Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J Clin Oncol. 2009;27:4733–40.

    CAS  PubMed  Google Scholar 

  8. Ghiaseddin A, Peters KB. Use of bevacizumab in recurrent glioblastoma. CNS Oncol. 2015;4:157–69.

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Jain KK. A critical overview of targeted therapies for glioblastoma. Front Oncol. 2018;8:419.

    PubMed  PubMed Central  Google Scholar 

  10. Lathia JD, Mack SC, Mulkearns-Hubert EE, Valentim CLL, Rich JN. Cancer stem cells in glioblastoma. Genes Dev. 2015;29:1203–17.

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Gimple RC, Bhargava S, Dixit D, Rich JN. Glioblastoma stem cells: lessons from the tumor hierarchy in a lethal cancer. Genes Dev. 2019;33:591–609.

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Pan BT, 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;101:942–8.

    CAS  PubMed  Google Scholar 

  13. Akers JC, Gonda D, Kim R, Carter BS, Chen CC. Biogenesis of extracellular vesicles (EV): exosomes, microvesicles, retrovirus-like vesicles, and apoptotic bodies. J Neurooncol. 2013;113:1–11.

    PubMed  PubMed Central  Google Scholar 

  14. Gandham S, Su X, Wood J, Nocera AL, Alli SC, Milane L, et al. Technologies and standardization in research on extracellular vesicles. Trends Biotechnol. 2020. https://doi.org/10.1016/j.tibtech.2020.05.012.

  15. Théry C, Witwer KW, Aikawa E, Alcaraz MJ, Anderson JD, Andriantsitohaina R, et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles. 2018;7:1535750.

    PubMed  PubMed Central  Google Scholar 

  16. Escola J-M, Kleijmeer MJ, Stoorvogel W, Griffith JM, Yoshie O, Geuze HJ. Selective enrichment of tetraspan proteins on the internal vesicles of multivesicular endosomes and on exosomes secreted by human B-lymphocytes. J Biol Chem. 1998;273:20121–7.

    CAS  PubMed  Google Scholar 

  17. Wubbolts R, Leckie RS, Veenhuizen PTM, Schwarzmann G, Möbius W, Hoernschemeyer J, et al. Proteomic and biochemical analyses of human B cell-derived exosomes: potential implications for their function and multivesicular body formatION. J Biol Chem. 2003;278:10963–72.

    CAS  PubMed  Google Scholar 

  18. van Niel G, D’Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol. 2018;19:213–28.

    PubMed  Google Scholar 

  19. Nolte-’t Hoen ENM, Buermans HPJ, Waasdorp M, Stoorvogel W, Wauben MHM, ’t Hoen PAC. Deep sequencing of RNA from immune cell-derived vesicles uncovers the selective incorporation of small non-coding RNA biotypes with potential regulatory functions. Nucleic Acids Res. 2012;40:9272–85.

    PubMed  PubMed Central  Google Scholar 

  20. Skog J, Würdinger T, van Rijn S, Meijer DH, Gainche L, Curry WT, et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol. 2008;10:1470–6.

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Weaver AM, Patton JG. Argonautes in extracellular vesicles: artifact or selected cargo? Cancer Res. 2020;80:379–81.

    CAS  PubMed  Google Scholar 

  22. Van Deun J, Mestdagh P, Sormunen R, Cocquyt V, Vermaelen K, Vandesompele J, et al. The impact of disparate isolation methods for extracellular vesicles on downstream RNA profiling. J Extracell Vesicles. 2014;3:24858.

    Google Scholar 

  23. Jeppesen DK, Fenix AM, Franklin JL, Higginbotham JN, Zhang Q, Zimmerman LJ, et al. Reassessment of exosome composition. Cell. 2019;177:428–45.e18.

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Bellingham SA, Coleman BM, Hill AF. Small RNA deep sequencing reveals a distinct miRNA signature released in exosomes from prion-infected neuronal cells. Nucleic Acids Res. 2012;40:10937–49.

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Laulagnier K, Motta C, Hamdi S, Roy S, Fauvelle F, Pageaux J-F, et al. Mast cell- and dendritic cell-derived exosomes display a specific lipid composition and an unusual membrane organization. Biochem J. 2004;380:161–71.

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Subra C, Laulagnier K, Perret B, Record M. Exosome lipidomics unravels lipid sorting at the level of multivesicular bodies. Biochimie. 2007;89:205–12.

    CAS  PubMed  Google Scholar 

  27. Skotland T, Sagini K, Sandvig K, Llorente A. An emerging focus on lipids in extracellular vesicles. Adv Drug Deliv Rev. 2020;S0169-409X:30014–4.

    Google Scholar 

  28. Williams C, Royo F, Aizpurua-Olaizola O, Pazos R, Boons G-J, Reichardt N-C, et al. Glycosylation of extracellular vesicles: current knowledge, tools and clinical perspectives. J Extracell Vesicles. 2018;7:1442985.

    PubMed  PubMed Central  Google Scholar 

  29. Gardiner C, Vizio DD, Sahoo S, Théry C, Witwer KW, Wauben M, et al. Techniques used for the isolation and characterization of extracellular vesicles: results of a worldwide survey. J Extracell Vesicles. 2016;5:32945.

    PubMed  Google Scholar 

  30. EV-TRACK Consortium, Van Deun J, Mestdagh P, Agostinis P, Akay Ö, Anand S, et al. EV-TRACK: transparent reporting and centralizing knowledge in extracellular vesicle research. Nat Methods. 2017;14:228–32.

    Google Scholar 

  31. André-Grégoire G, Gavard J. Spitting out the demons: extracellular vesicles in glioblastoma. Cell Adhes Migr. 2017;11:164–72.

    Google Scholar 

  32. Al-Nedawi K, Meehan B, Micallef J, Lhotak V, May L, Guha A, et al. Intercellular transfer of the oncogenic receptor EGFRvIII by microvesicles derived from tumour cells. Nat Cell Biol. 2008;10:619–24.

    CAS  PubMed  Google Scholar 

  33. Pace KR, Dutt R, Galileo DS. Exosomal L1CAM stimulates glioblastoma cell motility, proliferation, and invasiveness. Int J Mol Sci. 2019;20:3982.

    CAS  PubMed Central  Google Scholar 

  34. An JH, Lee SY, Jeon JY, Cho KG, Kim SU, Lee MA. Identification of gliotropic factors that induce human stem cell migration to malignant tumor. J Proteome Res. 2009;8:2873–81.

    CAS  PubMed  Google Scholar 

  35. Bronisz A, Wang Y, Nowicki MO, Peruzzi P, Ansari KI, Ogawa D, et al. Extracellular vesicles modulate the glioblastoma microenvironment via a tumor suppression signaling network directed by miR-1. Cancer Res. 2014;74:738–50.

    CAS  PubMed  Google Scholar 

  36. Son J, Lyssiotis CA, Ying H, Wang X, Hua S, Ligorio M, et al. Glutamine supports pancreatic cancer growth through a KRAS-regulated metabolic pathway. Nature. 2013;496:101–5.

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Yekula A, Minciacchi VR, Morello M, Shao H, Park Y, Zhang X, et al. Large and small extracellular vesicles released by glioma cells in vitro and in vivo. J Extracell Vesicles. 2020;9:1689784.

    CAS  PubMed  Google Scholar 

  38. Pavlyukov MS, Yu H, Bastola S, Minata M, Shender VO, Lee Y, et al. Apoptotic cell-derived extracellular vesicles promote malignancy of glioblastoma via intercellular transfer of splicing factors. Cancer Cell. 2018;34:119–35.e10.

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Figueroa J, Phillips LM, Shahar T, Hossain A, Gumin J, Kim H, 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;77:5808–19.

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Li D, Tian Y, Hu Y, Qi Y, Tian N, Li S, et al. Glioma-associated human endothelial cell-derived extracellular vesicles specifically promote the tumourigenicity of glioma stem cells via CD9. Oncogene. 2019;38:6898–912.

    CAS  PubMed  Google Scholar 

  41. Berenguer J, Lagerweij T, Zhao XW, Dusoswa S, van der Stoop P, Westerman B, et al. Glycosylated extracellular vesicles released by glioblastoma cells are decorated by CCL18 allowing for cellular uptake via chemokine receptor CCR8. J Extracell Vesicles. 2018;7:1446660.

    PubMed  PubMed Central  Google Scholar 

  42. Dusoswa SA, Horrevorts SK, Ambrosini M, Kalay H, Paauw NJ, Nieuwland R, et al. Glycan modification of glioblastoma-derived extracellular vesicles enhances receptor-mediated targeting of dendritic cells. J Extracell Vesicles. 2019;8:1648995.

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Treps L, Edmond S, Harford-Wright E, Galan-Moya EM, Schmitt A, Azzi S, et al. Extracellular vesicle-transported Semaphorin3A promotes vascular permeability in glioblastoma. Oncogene. 2016;35:2615–23.

    CAS  PubMed  Google Scholar 

  44. 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;6:1359479.

    PubMed  PubMed Central  Google Scholar 

  45. Lucero R, Zappulli V, Sammarco A, Murillo OD, Cheah PS, Srinivasan S, et al. Glioma-derived miRNA-containing extracellular vesicles induce angiogenesis by reprogramming brain endothelial cells. Cell Rep. 2020;30:2065–74.e4.

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Kucharzewska P, Christianson HC, Welch JE, Svensson KJ, Fredlund E, Ringner M, 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;110:7312–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Hallal S, Mallawaaratchy DM, Wei H, Ebrahimkhani S, Stringer BW, Day BW, et al. Extracellular vesicles released by glioblastoma cells stimulate normal astrocytes to acquire a tumor-supportive phenotype via p53 and MYC signaling pathways. Mol Neurobiol. 2019;56:4566–81.

    CAS  PubMed  Google Scholar 

  48. Oushy S, Hellwinkel JE, Wang M, Nguyen GJ, Gunaydin D, Harland TA, et al. Glioblastoma multiforme-derived extracellular vesicles drive normal astrocytes towards a tumour-enhancing phenotype. Philos Trans R Soc B. 2018;373:20160477.

    Google Scholar 

  49. Colangelo NW, Azzam EI. Extracellular vesicles originating from glioblastoma cells increase metalloproteinase release by astrocytes: the role of CD147 (EMMPRIN) and ionizing radiation. Cell Commun Signal. 2020;18:21.

    CAS  PubMed  PubMed Central  Google Scholar 

  50. de Vrij J, Maas SLN, Kwappenberg KMC, Schnoor R, Kleijn A, Dekker L, et al. Glioblastoma-derived extracellular vesicles modify the phenotype of monocytic cells: glioblastoma vesicles affect monocytic cells. Int J Cancer. 2015;137:1630–42.

    PubMed  Google Scholar 

  51. van der Vos KE, Abels ER, Zhang X, Lai C, Carrizosa E, Oakley D, et al. Directly visualized glioblastoma-derived extracellular vesicles transfer RNA to microglia/macrophages in the brain. Neuro Oncol. 2016;18:58–69.

    PubMed  Google Scholar 

  52. Himes BT, Peterson TE, de Mooij T, Garcia LMC, Jung M-Y, Uhm S, et al. The role of extracellular vesicles and PD-L1 in glioblastoma-mediated immunosuppressive monocyte induction. Neuro Oncol. 2020;22:967–78.

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Domenis R, Cesselli D, Toffoletto B, Bourkoula E, Caponnetto F, Manini I, et al. Systemic T cells immunosuppression of glioma stem cell-derived exosomes is mediated by monocytic myeloid-derived suppressor cells. PLoS ONE. 2017;12:e0169932.

    PubMed  PubMed Central  Google Scholar 

  54. Guo X, Qiu W, Wang J, Liu Q, Qian M, Wang S, et al. Glioma exosomes mediate the expansion and function of myeloid‐derived suppressor cells through microRNA‐29a/Hbp1 and microRNA‐92a/Prkar1a pathways. Int J Cancer. 2019;144:3111–26.

    CAS  PubMed  Google Scholar 

  55. Shao H, Chung J, Lee K, Balaj L, Min C, Carter BS, et al. Chip-based analysis of exosomal mRNA mediating drug resistance in glioblastoma. Nat Commun. 2015;6:6999.

    CAS  PubMed  Google Scholar 

  56. Yu T, Wang X, Zhi T, Zhang J, Wang Y, Nie E, et al. Delivery of MGMT mRNA to glioma cells by reactive astrocyte-derived exosomes confers a temozolomide resistance phenotype. Cancer Lett. 2018;433:210–20.

    CAS  PubMed  Google Scholar 

  57. Bebawy M, Combes V, Lee E, Jaiswal R, Gong J, Bonhoure A, et al. Membrane microparticles mediate transfer of P-glycoprotein to drug sensitive cancer cells. Leukemia. 2009;23:1643–9.

    CAS  PubMed  Google Scholar 

  58. Yin J, Zeng A, Zhang Z, Shi Z, Yan W, You Y. Exosomal transfer of miR-1238 contributes to temozolomide-resistance in glioblastoma. EBioMedicine. 2019;42:238–51.

    PubMed  PubMed Central  Google Scholar 

  59. Cosset EC, Godet J, Entz-Werlé N, Guérin E, Guenot D, Froelich S, et al. Involvement of the TGFβ pathway in the regulation of α5β1 integrins by caveolin-1 in human glioblastoma. Int J Cancer. 2012;131:601–11.

    CAS  PubMed  Google Scholar 

  60. Yang J-K, Yang J-P, Tong J, Jing S-Y, Fan B, Wang F, et al. Exosomal miR-221 targets DNM3 to induce tumor progression and temozolomide resistance in glioma. J Neurooncol. 2017;131:255–65.

    CAS  PubMed  Google Scholar 

  61. Zhang C-Z, Zhang J-X, Zhang A-L, Shi Z-D, Han L, Jia Z-F, et al. MiR-221 and miR-222 target PUMA to induce cell survival in glioblastoma. Mol Cancer. 2010;9:229.

    PubMed  PubMed Central  Google Scholar 

  62. Zhang Z, Yin J, Lu C, Wei Y, Zeng A, You Y. Exosomal transfer of long non-coding RNA SBF2-AS1 enhances chemoresistance to temozolomide in glioblastoma. J Exp Clin Cancer Res. 2019;38:166.

    PubMed  PubMed Central  Google Scholar 

  63. Simon T, Pinioti S, Schellenberger P, Rajeeve V, Wendler F, Cutillas PR, et al. Shedding of bevacizumab in tumour cells-derived extracellular vesicles as a new therapeutic escape mechanism in glioblastoma. Mol Cancer. 2018;17:132.

    PubMed  PubMed Central  Google Scholar 

  64. 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;27:1939–49.

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Andre-Gregoire G, Bidere N, Gavard J. Temozolomide affects extracellular vesicles released by glioblastoma cells. Biochimie. 2018;155:11–5.

    CAS  PubMed  Google Scholar 

  66. Garnier D, Meehan B, Kislinger T, Daniel P, Sinha A, Abdulkarim B, et al. Divergent evolution of temozolomide resistance in glioblastoma stem cells is reflected in extracellular vesicles and coupled with radiosensitization. Neuro Oncol. 2018;20:236–48.

    CAS  PubMed  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  68. Osti D, Del Bene M, Rappa G, Santos M, Matafora V, Richichi C, et al. Clinical significance of extracellular vesicles in plasma from glioblastoma patients. Clin Cancer Res. 2019;25:266–76.

    CAS  PubMed  Google Scholar 

  69. Zhang H, Lyden D. Asymmetric-flow field-flow fractionation technology for exomere and small extracellular vesicle separation and characterization. Nat Protoc. 2019;14:1027–53.

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Zhang H, Freitas D, Kim HS, Fabijanic K, Li Z, Chen H, et al. Identification of distinct nanoparticles and subsets of extracellular vesicles by asymmetric flow field-flow fractionation. Nat Cell Biol. 2018;20:332–43.

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Hallal S, Azimi A, Wei H, Ho N, Lee MYT, Sim H-W. A comprehensive proteomic SWATH-MS workflow for profiling blood extracellular vesicles: a new avenue for glioma tumour surveillance. Int J Mol Sci. 2020;21:4754

    CAS  PubMed Central  Google Scholar 

  72. Indira Chandran V, Welinder C, Månsson A-S, Offer S, Freyhult E, Pernemalm M, et al. Ultrasensitive immunoprofiling of plasma extracellular vesicles identifies Syndecan-1 as a potential tool for minimally invasive diagnosis of glioma. Clin Cancer Res. 2019;25:3115–27.

    PubMed  Google Scholar 

  73. Godlewski J, Ferrer-Luna R, Rooj AK, Mineo M, Ricklefs F, Takeda YS, et al. MicroRNA signatures and molecular subtypes of glioblastoma: the role of extracellular transfer. Stem Cell Rep. 2017;8:1497–505.

    CAS  Google Scholar 

  74. Ebrahimkhani S, Vafaee F, Hallal S, Wei H, Lee MYT, Young PE, et al. Deep sequencing of circulating exosomal microRNA allows non-invasive glioblastoma diagnosis. npj Precis Oncol. 2018;2:28.

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Tan SK, Pastori C, Penas C, Komotar RJ, Ivan ME, Wahlestedt C, et al. Serum long noncoding RNA HOTAIR as a novel diagnostic and prognostic biomarker in glioblastoma multiforme. Mol Cancer. 2018;17:74.

    PubMed  PubMed Central  Google Scholar 

  76. Figueroa JM, Skog J, Akers J, Li H, Komotar R, Jensen R, et al. Detection of wild-type EGFR amplification and EGFRvIII mutation in CSF-derived extracellular vesicles of glioblastoma patients. Neuro Oncol. 2017;19:1494–502.

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Saugstad JA, Lusardi TA, Van Keuren-Jensen KR, Phillips JI, Lind B, Harrington CA, et al. Analysis of extracellular RNA in cerebrospinal fluid. J Extracell Vesicles. 2017;6:1317577.

    PubMed  PubMed Central  Google Scholar 

  78. Manda SV, Kataria Y, Tatireddy BR, Ramakrishnan B, Ratnam BG, Lath R, et al. Exosomes as a biomarker platform for detecting epidermal growth factor receptor–positive high-grade gliomas. J Neurosurg. 2018;128:1091–101.

    CAS  PubMed  Google Scholar 

  79. Evans SM, Putt M, Yang X-Y, Lustig RA, Martinez-Lage M, Williams D, et al. Initial evidence that blood-borne microvesicles are biomarkers for recurrence and survival in newly diagnosed glioblastoma patients. J Neurooncol. 2016;127:391–400.

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Lan F, Qing Q, Pan Q, Hu M, Yu H, Yue X. Serum exosomal miR-301a as a potential diagnostic and prognostic biomarker for human glioma. Cell Oncol. 2018;41:25–33.

    CAS  Google Scholar 

Download references

Acknowledgements

We thank SOAP team members (Nantes, France). The research from the team was funded by Fondation pour la Recherche Medicale (Equipe labellisée DEQ20180339184), Fondation ARC contre le Cancer (PJA20171206146, PJA20191209477, PGA1 RF20190208474), INCa PLBIO (2019-151, 2019-291), Ligue nationale contre le cancer comités de Loire-Atlantique, Maine et Loire, Vendée, Ille-et-Vilaine, SIRIC ILIAD (INCa-DGOS-Inserm_12558) and Région Pays de la Loire et Nantes Métropole under Connect Talent Grant. QS received Master internship fellowship from ITMO Cancer (Plan Cancer 2014−2019).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Julie Gavard.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sabbagh, Q., Andre-Gregoire, G., Guevel, L. et al. Vesiclemia: counting on extracellular vesicles for glioblastoma patients. Oncogene 39, 6043–6052 (2020). https://doi.org/10.1038/s41388-020-01420-x

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41388-020-01420-x

This article is cited by

Search

Quick links