DOI QR코드

DOI QR Code

The therapeutic potential of immune cell-derived exosomes as an alternative to adoptive cell transfer

  • Hong, Yeonsun (Biomedical Research Institute, Korea Institute of Science and Technology (KIST)) ;
  • Kim, In-San (Biomedical Research Institute, Korea Institute of Science and Technology (KIST))
  • Received : 2021.05.24
  • Accepted : 2021.08.05
  • Published : 2022.01.31

Abstract

Adoptive cell transfer (ACT), a form of cell-based immunotherapy that eliminates cancer by restoring and strengthening the body's immune system, has revolutionized cancer treatment. ACT entails intravenous transfer of either tumor-resident or peripheral blood-modified immune cells into cancer patients to mediate anti-tumor response. Although these immune cells control and eradicate cancer via enhanced cytotoxicity against specific tumor antigens, several side effects have been frequently reported in clinical trials. Recently, exosomes, potential cell-free therapeutics, have emerged as an alternative to cell-based immunotherapies, due to their higher stability under same storage condition, lower risk of GvHD and CRS, and higher resistance to immunosuppressive tumor microenvironment. Exosomes, which are nano-sized lipid vesicles, are secreted by living cells, including immune cells. Exosomes contain proteins, lipids, and nucleic acids, and the functional role of each exosome is determined by the specific cargo derived from parental cells. Exosomes derived from cytotoxic effectors including T cells and NK cells exert anti-tumor effects via proteins such as granzyme B and FasL. In this mini-review, we describe the current understanding of the ACT and immune cell-derived exosomes and discuss the limitations of ACT and the opportunities for immune cell-derived exosomes as immune therapies.

Keywords

Acknowledgement

This work was supported by grants from the National Research Foundation of Korea (NRF) funded by the Korean government (2017R1A3B1023418), KU- KIST Graduate School of Convertging Science and Technology Program, and KIST Institutional Program.

References

  1. Hinshaw DC and Shevde LA (2019) the tumor microenvironment innately modulates cancer progression. Cancer Res 79, 4557-4566 https://doi.org/10.1158/0008-5472.can-18-3962
  2. Roma-Rodrigues C, Mendes R, Baptista PV and Fernandes AR (2019) Targeting tumor microenvironment for cancer therapy. Int J Mol Sci 20, 840 https://doi.org/10.3390/ijms20040840
  3. Baghban R, Roshangar L, Jahanban-Esfahlan R et al (2020) Tumor microenvironment complexity and therapeutic implications at a glance. Cell Commun Signal 18, 59 https://doi.org/10.1186/s12964-020-0530-4
  4. Yu Y and Cui J (2018) Present and future of cancer immunotherapy: a tumor microenvironmental perspective. Oncol Lett 16, 4105-4113
  5. Galluzzi L, Chan TA, Kroemer G, Wolchok JD and Lopez-Soto A (2018) The hallmarks of successful anticancer immunotherapy. Sci Transl Med 10, eaat7807 https://doi.org/10.1126/scitranslmed.aat7807
  6. Chuah S and Chew V (2020) High-dimensional immuneprofiling in cancer: implications for immunotherapy. J Immunother Cancer 8, e000363 https://doi.org/10.1136/jitc-2019-000363
  7. Disis ML (2014) Mechanism of action of immunotherapy. Semin Oncol 41 Suppl 5, S3-13 https://doi.org/10.1053/j.seminoncol.2014.09.004
  8. Houot R, Schultz LM, Marabelle A and Kohrt H (2015) T-cell-based immunotherapy: adoptive cell transfer and checkpoint inhibition. Cancer Immunol Res 3, 1115-1122 https://doi.org/10.1158/2326-6066.CIR-15-0190
  9. Rosenberg SA and Restifo NP (2015) Adoptive cell transfer as personalized immunotherapy for human cancer. Science 348, 62-68 https://doi.org/10.1126/science.aaa4967
  10. Laskowski T and Rezvani K (2020) Adoptive cell therapy: Living drugs against cancer. J Exp Med 217, e20200377 https://doi.org/10.1084/jem.20200377
  11. Rohaan MW, Wilgenhof S and Haanen J (2019) Adoptive cellular therapies: the current landscape. Virchows Arch 474, 449-461 https://doi.org/10.1007/s00428-018-2484-0
  12. Kalluri R and LeBleu VS (2020) The biology, function, and biomedical applications of exosomes. Science 367, eaau6977 https://doi.org/10.1126/science.aau6977
  13. Zhang Y, Liu Y, Liu H and Tang WH (2019) Exosomes: biogenesis, biologic function and clinical potential. Cell Biosci 9, 19 https://doi.org/10.1186/s13578-019-0282-2
  14. Mathieu M, Martin-Jaular L, Lavieu G and Thery C (2019) Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat Cell Biol 21, 9-17 https://doi.org/10.1038/s41556-018-0250-9
  15. Fais S (2013) NK cell-released exosomes: natural nanobullets against tumors. Oncoimmunology 2, e22337 https://doi.org/10.4161/onci.22337
  16. Pitt JM, Andre F, Amigorena S et al (2016) Dendritic cell-derived exosomes for cancer therapy. J Clin Invest 126, 1224-1232 https://doi.org/10.1172/jci81137
  17. Hong Y, Kim YK, Kim GB et al (2019) Degradation of tumour stromal hyaluronan by small extracellular vesicle-PH20 stimulates CD103(+) dendritic cells and in combination with PD-L1 blockade boosts anti-tumour immunity. J Extracell Vesicles 8, 1670893 https://doi.org/10.1080/20013078.2019.1670893
  18. Kim GB, Nam GH, Hong Y et al (2020) Xenogenization of tumor cells by fusogenic exosomes in tumor microenvironment ignites and propagates antitumor immunity. Sci Adv 6, eaaz2083 https://doi.org/10.1126/sciadv.aaz2083
  19. Koh E, Lee EJ, Nam GH et al (2017) Exosome-SIRPalpha, a CD47 blockade increases cancer cell phagocytosis. Biomaterials 121, 121-129 https://doi.org/10.1016/j.biomaterials.2017.01.004
  20. Tran TH, Mattheolabakis G, Aldawsari H and Amiji M (2015) Exosomes as nanocarriers for immunotherapy of cancer and inflammatory diseases. Clin Immunol 160, 46-58 https://doi.org/10.1016/j.clim.2015.03.021
  21. Yang Y, Hong Y, Cho E, Kim GB and Kim IS (2018) Extracellular vesicles as a platform for membrane-associated therapeutic protein delivery. J Extracell Vesicles 7, 1440131 https://doi.org/10.1080/20013078.2018.1440131
  22. Hong Y, Nam GH, Koh E et al (2018) Exosome as a vehicle for delivery of membrane protein therapeutics, PH20, for enhanced tumor penetration and antitumor efficacy. Adv Funct Mater 28, 1703074 https://doi.org/10.1002/adfm.201703074
  23. Yang JC and Rosenberg SA (2016) Adoptive T-cell therapy for cancer. Adv Immunol 130, 279-294 https://doi.org/10.1016/bs.ai.2015.12.006
  24. Dudley ME and Rosenberg SA (2003) Adoptive-cell-transfer therapy for the treatment of patients with cancer. Nat Rev Cancer 3, 666-675 https://doi.org/10.1038/nrc1167
  25. Rosenberg SA, Packard BS, Aebersold PM et al (1988) Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma. A preliminary report. N Engl J Med 319, 1676-1680 https://doi.org/10.1056/NEJM198812223192527
  26. Ott PA, Dotti G, Yee C and Goff SL (2019) An update on adoptive T-cell therapy and neoantigen vaccines. Am Soc Clin Oncol Educ Book 39, e70-e78
  27. Essand M and Loskog AS (2013) Genetically engineered T cells for the treatment of cancer. J Intern Med 273, 166-181 https://doi.org/10.1111/joim.12020
  28. Morgan RA, Dudley ME, Wunderlich JR et al (2006) Cancer regression in patients after transfer of genetically engineered lymphocytes. Science 314, 126-129 https://doi.org/10.1126/science.1129003
  29. Johnson LA, Morgan RA, Dudley ME et al (2009) Gene therapy with human and mouse T-cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen. Blood 114, 535-546
  30. Robbins PF, Morgan RA, Feldman SA et al (2011) Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1. J Clin Oncol 29, 917-924 https://doi.org/10.1200/JCO.2010.32.2537
  31. Robbins PF, Kassim SH, Tran TL et al (2015) A pilot trial using lymphocytes genetically engineered with an NY-ESO-1-reactive T-cell receptor: long-term follow-up and correlates with response. Clin Cancer Res 21, 1019-1027 https://doi.org/10.1158/1078-0432.CCR-14-2708
  32. Miliotou AN and Papadopoulou LC (2018) CAR T-cell therapy: a new era in cancer immunotherapy. Curr Pharm Biotechnol 19, 5-18 https://doi.org/10.2174/1389201019666180418095526
  33. Feins S, Kong W, Williams EF, Milone MC and Fraietta JA (2019) An introduction to chimeric antigen receptor (CAR) T-cell immunotherapy for human cancer. Am J Hematol 94, S3-S9 https://doi.org/10.1002/ajh.25418
  34. Maude SL, Frey N, Shaw PA et al (2014) Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med 371, 1507-1517 https://doi.org/10.1056/NEJMoa1407222
  35. Schuster SJ, Svoboda J, Chong EA et al (2017) Chimeric antigen receptor T cells in refractory B-cell lymphomas. N Engl J Med 377, 2545-2554 https://doi.org/10.1056/NEJMoa1708566
  36. Alexander M, Culos K, Roddy J et al (2021) Chimeric antigen receptor T-cell therapy: a comprehensive review of clinical efficacy, toxicity, and best practices for outpatient administration.Transplant Cell Ther 27, 558-570 https://doi.org/10.1016/j.jtct.2021.01.014
  37. Hu W, Wang G, Huang D, Sui M and Xu Y (2019) cancer immunotherapy based on natural killer cells: current progress and new opportunities. Front Immunol 10, 1205 https://doi.org/10.3389/fimmu.2019.01205
  38. Shimasaki N, Jain A and Campana D (2020) NK cells for cancer immunotherapy. Nat Rev Drug Discov 19, 200-218 https://doi.org/10.1038/s41573-019-0052-1
  39. Liu S, Galat V, Galat Y, Lee YKA, Wainwright D and Wu J (2021) NK cell-based cancer immunotherapy: from basic biology to clinical development. J Hematol Oncol 14, 7 https://doi.org/10.1186/s13045-020-01014-w
  40. Klingemann H, Boissel L and Toneguzzo F (2016) Natural killer cells for immunotherapy - advantages of the NK-92 cell line over blood NK Cells. Front Immunol 7, 91 https://doi.org/10.3389/fimmu.2016.00091
  41. Kundu S, Gurney M and O'Dwyer M (2021) Generating natural killer cells for adoptive transfer: expanding horizons. Cytotherapy 23, 559-566 https://doi.org/10.1016/j.jcyt.2020.12.002
  42. Bollino D and Webb TJ (2017) Chimeric antigen receptor-engineered natural killer and natural killer T cells for cancer immunotherapy. Transl Res 187, 32-43 https://doi.org/10.1016/j.trsl.2017.06.003
  43. Tang X, Yang L, Li Z et al (2018) First-in-man clinical trial of CAR NK-92 cells: safety test of CD33-CAR NK-92 cells in patients with relapsed and refractory acute myeloid leukemia. Am J Cancer Res 8, 1083-1089
  44. Rapoport AP, Stadtmauer EA, Aqui N et al (2009) Rapid immune recovery and graft-versus-host disease-like engraftment syndrome following adoptive transfer of costimulated autologous T cells. Clin Cancer Res 15, 4499-4507 https://doi.org/10.1158/1078-0432.ccr-09-0418
  45. Magalhaes I, Carvalho-Queiroz C, Hartana CA et al (2019) Facing the future: challenges and opportunities in adoptive T cell therapy in cancer. Expert Opin Biol Ther 19, 811-827 https://doi.org/10.1080/14712598.2019.1608179
  46. Parkhurst MR, Yang JC, Langan RC et al (2011) T cells targeting carcinoembryonic antigen can mediate regression of metastatic colorectal cancer but induce severe transient colitis. Mol Ther 19, 620-626 https://doi.org/10.1038/mt.2010.272
  47. Kochenderfer JN, Dudley ME, Feldman SA et al (2012) B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-transduced T cells. Blood 119, 2709-2720 https://doi.org/10.1182/blood-2011-10-384388
  48. Morgan RA, Yang JC, Kitano M, Dudley ME, Laurencot CM and Rosenberg SA (2010) Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol Ther 18, 843-851 https://doi.org/10.1038/mt.2010.24
  49. Shimabukuro-Vornhagen A, Godel P, Subklewe M et al (2018) Cytokine release syndrome. J Immunother Cancer 6, 56 https://doi.org/10.1186/s40425-018-0343-9
  50. Kochenderfer JN, Somerville RPT, Lu T et al (2017) Lymphoma remissions caused by anti-CD19 chimeric antigen receptor T cells are associated with high serum interleukin-15 levels. J Clin Oncol 35, 1803-1813 https://doi.org/10.1200/JCO.2016.71.3024
  51. Neelapu SS, Tummala S, Kebriaei P et al (2018) Chimeric antigen receptor T-cell therapy - assessment and management of toxicities. Nat Rev Clin Oncol 15, 47-62 https://doi.org/10.1038/nrclinonc.2017.148
  52. Bonifant CL, Jackson HJ, Brentjens RJ and Curran KJ (2016) Toxicity and management in CAR T-cell therapy. Mol Ther Oncolytics 3, 16011 https://doi.org/10.1038/mto.2016.11
  53. Khadka RH, Sakemura R, Kenderian SS and Johnson AJ (2019) Management of cytokine release syndrome: an update on emerging antigen-specific T cell engaging immunotherapies. Immunotherapy 11, 851-857 https://doi.org/10.2217/imt-2019-0074
  54. Murthy H, Iqbal M, Chavez JC and Kharfan-Dabaja MA (2019) Cytokine release syndrome: current perspectives. Immunotargets Ther 8, 43-52 https://doi.org/10.2147/ITT.S202015
  55. Tang H, Qiao J and Fu YX (2016) Immunotherapy and tumor microenvironment. Cancer Lett 370, 85-90 https://doi.org/10.1016/j.canlet.2015.10.009
  56. Beavis PA, Slaney CY, Kershaw MH, Gyorki D, Neeson PJ and Darcy PK (2016) Reprogramming the tumor microenvironment to enhance adoptive cellular therapy. Semin Immunol 28, 64-72 https://doi.org/10.1016/j.smim.2015.11.003
  57. Thommen DS and Schumacher TN (2018) T Cell dysfunction in cancer. Cancer Cell 33, 547-562 https://doi.org/10.1016/j.ccell.2018.03.012
  58. Slattery K and Gardiner CM (2019) NK cell metabolism and TGFbeta - implications for immunotherapy. Front Immunol 10, 2915 https://doi.org/10.3389/fimmu.2019.02915
  59. Riggan L, Shah S and O'Sullivan TE (2021) Arrested development: suppression of NK cell function in the tumor microenvironment. Clin Transl Immunology 10, e1238
  60. Melaiu O, Lucarini V, Cifaldi L and Fruci D (2019) Influence of the tumor microenvironment on NK cell function in solid tumors. Front Immunol 10, 3038 https://doi.org/10.3389/fimmu.2019.03038
  61. Comoli P, Chabannon C, Koehl U et al (2019) Development of adaptive immune effector therapies in solid tumors. Ann Oncol 30, 1740-1750 https://doi.org/10.1093/annonc/mdz285
  62. Thery C, Zitvogel L and Amigorena S (2002) Exosomes: composition, biogenesis and function. Nat Rev Immunol 2, 569-579 https://doi.org/10.1038/nri855
  63. Colombo M, Raposo G and Thery C (2014) Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu Rev Cell Dev Biol 30, 255-289 https://doi.org/10.1146/annurev-cellbio-101512-122326
  64. Hessvik NP and Llorente A (2018) Current knowledge on exosome biogenesis and release. Cell Mol Life Sci 75, 193-208 https://doi.org/10.1007/s00018-017-2595-9
  65. Jeppesen DK, Fenix AM, Franklin JL et al (2019) Reassessment of exosome composition. Cell 177, 428-445 e418 https://doi.org/10.1016/j.cell.2019.02.029
  66. Thery C, Ostrowski M and Segura E (2009) Membrane vesicles as conveyors of immune responses. Nat Rev Immunol 9, 581-593 https://doi.org/10.1038/nri2567
  67. Skotland T, Sandvig K and Llorente A (2017) Lipids in exosomes: current knowledge and the way forward. Prog Lipid Res 66, 30-41 https://doi.org/10.1016/j.plipres.2017.03.001
  68. Bobrie A, Colombo M, Raposo G and Thery C (2011) Exosome secretion: molecular mechanisms and roles in immune responses. Traffic 12, 1659-1668 https://doi.org/10.1111/j.1600-0854.2011.01225.x
  69. Nam GH, Choi Y, Kim GB, Kim S, Kim SA and Kim IS (2020) Emerging prospects of exosomes for cancer treatment: from conventional therapy to immunotherapy. Adv Mater 32, e2002440
  70. Raposo G, Nijman HW, Stoorvogel W et al (1996) B lymphocytes secrete antigen-presenting vesicles. J Exp Med 183, 1161-1172 https://doi.org/10.1084/jem.183.3.1161
  71. Li Q, Wang H, Peng H, Huyan T and Cacalano NA (2019) Exosomes: versatile nano mediators of immune regulation. Cancers (Basel) 11, 1557 https://doi.org/10.3390/cancers11101557
  72. Jankovicova J, Secova P, Michalkova K and Antalikova J (2020) Tetraspanins, more than markers of extracellular vesicles in reproduction. Int J Mol Sci 21, 7568 https://doi.org/10.3390/ijms21207568
  73. Nikfarjam S, Rezaie J, Kashanchi F and Jafari R (2020) Dexosomes as a cell-free vaccine for cancer immunotherapy. J Exp Clin Cancer Res 39, 258 https://doi.org/10.1186/s13046-020-01781-x
  74. Ventimiglia LN and Alonso MA (2016) Biogenesis and function of T cell-derived exosomes. Front Cell Dev Biol 4, 84 https://doi.org/10.3389/fcell.2016.00084
  75. Ventimiglia LN, Fernandez-Martin L, Martinez-Alonso E et al (2015) Cutting edge: regulation of exosome secretion by the integral MAL protein in T cells. J Immunol 195, 810-814 https://doi.org/10.4049/jimmunol.1500891
  76. Wahlgren J, Karlson Tde L, Glader P, Telemo E and Valadi H (2012) Activated human T cells secrete exosomes that participate in IL-2 mediated immune response signaling. PLoS One 7, e49723 https://doi.org/10.1371/journal.pone.0049723
  77. Li L, Jay SM, Wang Y, Wu SW and Xiao Z (2017) IL-12 stimulates CTLs to secrete exosomes capable of activating bystander CD8(+) T cells. Sci Rep 7, 13365 https://doi.org/10.1038/s41598-017-14000-z
  78. Mittelbrunn M, Gutierrez-Vazquez C, Villarroya-Beltri C et al (2011) Unidirectional transfer of microRNA-loaded exosomes from T cells to antigen-presenting cells. Nat Commun 2, 282 https://doi.org/10.1038/ncomms1285
  79. Fu W, Lei C, Liu S et al (2019) CAR exosomes derived from effector CAR-T cells have potent antitumour effects and low toxicity. Nat Commun 10, 4355 https://doi.org/10.1038/s41467-019-12321-3
  80. Yang P, Cao X, Cai H et al (2021) The exosomes derived from CAR-T cell efficiently target mesothelin and reduce triple-negative breast cancer growth. Cell Immunol 360, 104262 https://doi.org/10.1016/j.cellimm.2020.104262
  81. Lugini L, Cecchetti S, Huber V et al (2012) Immune surveillance properties of human NK cell-derived exosomes. J Immunol 189, 2833-2842 https://doi.org/10.4049/jimmunol.1101988
  82. Jong AY, Wu CH, Li J et al (2017) Large-scale isolation and cytotoxicity of extracellular vesicles derived from activated human natural killer cells. J Extracell Vesicles 6, 1294368 https://doi.org/10.1080/20013078.2017.1294368
  83. Zhu L, Kalimuthu S, Gangadaran P et al (2017) Exosomes derived from natural killer cells exert therapeutic effect in melanoma. Theranostics 7, 2732-2745 https://doi.org/10.7150/thno.18752
  84. Shoae-Hassani A, Hamidieh AA, Behfar M, Mohseni R, Mortazavi-Tabatabaei SA and Asgharzadeh S (2017) NK cell-derived exosomes from NK cells previously exposed to neuroblastoma cells augment the antitumor activity of cytokine-activated NK cells. J Immunother 40, 265-276 https://doi.org/10.1097/CJI.0000000000000179
  85. Di Pace AL, Tumino N, Besi F et al (2020) Characterization of human NK cell-derived exosomes: Role of DNAM1 receptor in exosome-mediated cytotoxicity against tumor. Cancers (Basel) 12, 661 https://doi.org/10.3390/cancers12030661
  86. Neviani P, Wise PM, Murtadha M et al (2019) Natural killer-derived exosomal miR-186 inhibits neuroblastoma growth and immune escape mechanisms. Cancer Res 79, 1151-1164 https://doi.org/10.1158/0008-5472.can-18-0779
  87. Wculek SK, Cueto FJ, Mujal AM, Melero I, Krummel MF and Sancho D (2020) Dendritic cells in cancer immunology and immunotherapy. Nat Rev Immunol 20, 7-24 https://doi.org/10.1038/s41577-019-0210-z
  88. Viaud S, Terme M, Flament C et al (2009) Dendritic cell-derived exosomes promote natural killer cell activation and proliferation: a role for NKG2D ligands and IL-15Ralpha. PLoS One 4, e4942 https://doi.org/10.1371/journal.pone.0004942
  89. Munich S, Sobo-Vujanovic A, Buchser WJ, Beer-Stolz D and Vujanovic NL (2012) Dendritic cell exosomes directly kill tumor cells and activate natural killer cells via TNF superfamily ligands. Oncoimmunology 1, 1074-1083 https://doi.org/10.4161/onci.20897
  90. Morse MA, Garst J, Osada T et al (2005) A phase I study of dexosome immunotherapy in patients with advanced non-small cell lung cancer. J Transl Med 3, 9 https://doi.org/10.1186/1479-5876-3-9
  91. Besse B, Charrier M, Lapierre V et al (2016) Dendritic cell-derived exosomes as maintenance immunotherapy after first line chemotherapy in NSCLC. Oncoimmunology 5, e1071008 https://doi.org/10.1080/2162402X.2015.1071008
  92. Escudier B, Dorval T, Chaput N et al (2005) Vaccination of metastatic melanoma patients with autologous dendritic cell (DC) derived-exosomes: results of thefirst phase I clinical trial. J Transl Med 3, 10 https://doi.org/10.1186/1479-5876-3-10
  93. Dai S, Wei D, Wu Z et al (2008) Phase I clinical trial of autologous ascites-derived exosomes combined with GM-CSF for colorectal cancer. Mol Ther 16, 782-790 https://doi.org/10.1038/mt.2008.1
  94. Cheng L, Wang Y and Huang L (2017) Exosomes from M1-polarized macrophages potentiate the cancer vaccine by creating a pro-inflammatory microenvironment in the lymph node. Mol Ther 25, 1665-1675 https://doi.org/10.1016/j.ymthe.2017.02.007
  95. Wang P, Wang H, Huang Q et al (2019) Exosomes from M1-polarized macrophages enhance paclitaxel antitumor activity by activating macrophages-mediated inflammation. Theranostics 9, 1714-1727 https://doi.org/10.7150/thno.30716
  96. Li J, Li N and Wang J (2020) M1 macrophage-derived exosome-encapsulated cisplatin can enhance its anti-lung cancer effect. Minerva Med [Online ahead of print]
  97. Zhang X, Liu L, Tang M, Li H, Guo X and Yang X (2020) The effects of umbilical cord-derived macrophage exosomes loaded with cisplatin on the growth and drug resistance of ovarian cancer cells. Drug Dev Ind Pharm 46, 1150-1162 https://doi.org/10.1080/03639045.2020.1776320
  98. Kim MS, Haney MJ, Zhao Y et al (2016) Development of exosome-encapsulated paclitaxel to overcome MDR in cancer cells. Nanomedicine 12, 655-664 https://doi.org/10.1016/j.nano.2015.10.012
  99. Zhang Y, Bi J, Huang J, Tang Y, Du S and Li P (2020) Exosome: a review of its classification, isolation techniques, storage, diagnostic and targeted therapy applications. Int J Nanomedicine 15, 6917-6934 https://doi.org/10.2147/IJN.S264498
  100. Tian Y, Li S, Song J et al (2014) A doxorubicin delivery platform using engineered natural membrane vesicle exosomes for targeted tumor therapy. Biomaterials 35, 2383-2390 https://doi.org/10.1016/j.biomaterials.2013.11.083
  101. Yang D, Zhang W, Zhang H et al (2020) Progress, opportunity, and perspective on exosome isolation - efforts for efficient exosome-based theranostics. Theranostics 10, 3684-3707 https://doi.org/10.7150/thno.41580
  102. Busatto S, Vilanilam G, Ticer T et al (2018) Tangential flow filtration for highly efficient concentration of extracellular vesicles from large volumes of fluid. Cells 7, 273 https://doi.org/10.3390/cells7120273
  103. Chen YS, Lin EY, Chiou TW and Harn HJ (2020) Exosomes in clinical trial and their production in compliance with good manufacturing practice. Ci Ji Yi Xue Za Zhi 32, 113-120
  104. Whitford W and Guterstam P (2019) Exosome manufacturing status. Future Med Chem 11, 1225-1236 https://doi.org/10.4155/fmc-2018-0417