DOI QR코드

DOI QR Code

Immune cell-derived small extracellular vesicles in cancer treatment

  • Choi, Sung-Jin (Department of New Biology, DGIST) ;
  • Cho, Hanchae (Department of Molecular Medicine, CMRI, Exosome Convergence Research Center (ECRC), School of Medicine, Kyungpook National University) ;
  • Yea, Kyungmoo (Department of New Biology, DGIST) ;
  • Baek, Moon-Chang (Department of Molecular Medicine, CMRI, Exosome Convergence Research Center (ECRC), School of Medicine, Kyungpook National University)
  • Received : 2021.08.26
  • Accepted : 2021.10.26
  • Published : 2022.01.31

Abstract

Small extracellular vesicles (sEVs) secreted by most cells carry bioactive macromolecules including proteins, lipids, and nucleic acids for intercellular communication. Given that some immune cell-derived sEVs exhibit anti-cancer properties, these sEVs have received scientific attention for the development of novel anti-cancer immunotherapeutic agents. In this paper, we reviewed the latest advances concerning the biological roles of immune cell-derived sEVs for cancer therapy. sEVs derived from immune cells including dendritic cells (DCs), T cells, natural-killer (NK) cells, and macrophages are good candidates for sEV-based cancer therapy. Besides their role of cancer vaccines, DC-shed sEVs activated cytotoxic lymphocytes and killed tumor cells. sEVs isolated from NK cells and chimeric antigen receptor (CAR) T cells exhibited cytotoxicity against cancer cells. sEVs derived from CD8+ T and CD4+ T cells inhibited cancer-associated cells in tumor microenvironment (TME) and activated B cells, respectively. M1-macrophage-derived sEVs induced M2 to M1 repolarization and also created a pro-inflammatory environment. Hence, these sEVs, via mono or combination therapy, could be considered in the treatment of cancer patients in the future. In addition, sEVs derived from cytokine-stimulated immune cells or sEV engineering could improve their anti-tumor potency.

Keywords

Acknowledgement

This research was supported by the Bio & Medical Technology Development Program of the National Research Foundation (NRF) of Korea funded by the Ministry of Science & ICT (2017M3A9G8083382 and 2020M3A9I4039539), the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) (No.2021R1A5A2021614), and the DGIST Start-up Fund Program of the Ministry of Science and ICT (2018010109).

References

  1. Thery C, Witwer KW, Aikawa E et al (2018) Minimal information for studies of extracellular vesicles 2018 (MISEV 2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles 7, 1535750 https://doi.org/10.1080/20013078.2018.1535750
  2. Kalluri R and LeBleu VS (2020) The biology, function, and biomedical applications of exosomes. Science 367, eaau6977 https://doi.org/10.1126/science.aau6977
  3. 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
  4. Kahlert C and Kalluri R (2013) Exosomes in tumor microenvironment influence cancer progression and metastasis. J Mol Med (Berl) 91, 431-437 https://doi.org/10.1007/s00109-013-1020-6
  5. 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
  6. Bebelman MP, Smit MJ, Pegtel DM and Baglio SR (2018) Biogenesis and function of extracellular vesicles in cancer. Pharmacol Ther 188, 1-11 https://doi.org/10.1016/j.pharmthera.2018.02.013
  7. Ciardiello C, Cavallini L, Spinelli C et al (2016) Focus on extracellular vesicles: new frontiers of cell-to-cell communication in cancer. Int J Mol Sci 17, 175 https://doi.org/10.3390/ijms17020175
  8. van Niel G, D'Angelo G and Raposo G (2018) Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol 19, 213-228 https://doi.org/10.1038/nrm.2017.125
  9. Urbanelli L, Magini A, Buratta S et al (2013) Signaling pathways in exosomes biogenesis, secretion and fate. Genes (Basel) 4, 152-170 https://doi.org/10.3390/genes4020152
  10. McLellan AD (2009) Exosome release by primary B cells. Crit Rev Immunol 29, 203-217 https://doi.org/10.1615/CritRevImmunol.v29.i3.20
  11. Segura E, Amigorena S and Thery C (2005) Mature dendritic cells secrete exosomes with strong ability to induce antigen-specific effector immune responses. Blood Cells Mol Dis 35, 89-93 https://doi.org/10.1016/j.bcmd.2005.05.003
  12. Paolillo M and Schinelli S (2017) Integrins and exosomes, a dangerous liaison in cancer progression. Cancers (Basel) 9
  13. Segura E, Guerin C, Hogg N, Amigorena S and Thery C (2007) CD8+ dendritic cells use LFA-1 to capture MHC-peptide complexes from exosomes in vivo. J Immunol 179, 1489-1496 https://doi.org/10.4049/jimmunol.179.3.1489
  14. Kalluri R (2016) The biology and function of exosomes in cancer. J Clin Invest 126, 1208-1215 https://doi.org/10.1172/jci81135
  15. Gao D and Jiang L (2018) Exosomes in cancer therapy: a novel experimental strategy. Am J Cancer Res 8, 2165-2175
  16. Batrakova EV and Kim MS (2015) Using exosomes, naturally-equipped nanocarriers, for drug delivery. J Control Release 219, 396-405 https://doi.org/10.1016/j.jconrel.2015.07.030
  17. Kibria G, Ramos EK, Wan Y, Gius DR and Liu H (2018) Exosomes as a drug delivery system in cancer therapy: potential and challenges. Mol Pharm 15, 3625-3633 https://doi.org/10.1021/acs.molpharmaceut.8b00277
  18. Yong T, Zhang X, Bie N et al (2019) Tumor exosome-based nanoparticles are efficient drug carriers for chemotherapy. Nat Commun 10, 3838 https://doi.org/10.1038/s41467-019-11718-4
  19. Li Y, Wan YY and Zhu B (2017) Immune cell metabolism in tumor microenvironment. Adv Exp Med Biol 1011, 163-196 https://doi.org/10.1007/978-94-024-1170-6_5
  20. Guo S and Deng CX (2018) Effect of stromal cells in tumor microenvironment on metastasis initiation. Int J Biol Sci 14, 2083-2093 https://doi.org/10.7150/ijbs.25720
  21. Steinman RM (2012) Decisions about dendritic cells: past, present, and future. Annu Rev Immunol 30, 1-22 https://doi.org/10.1146/annurev-immunol-100311-102839
  22. Pitt JM, Charrier M, Viaud S et al (2014) Dendritic cell-derived exosomes as immunotherapies in the fight against cancer. J Immunol 193, 1006-1011 https://doi.org/10.4049/jimmunol.1400703
  23. Chen DS and Mellman I (2013) Oncology meets immunology: the cancer-immunity cycle. Immunity 39, 1-10 https://doi.org/10.1016/j.immuni.2013.07.012
  24. Saxena M, van der Burg SH, Melief CJM and Bhardwaj N (2021) Therapeutic cancer vaccines. Nat Rev Cancer 21, 360-378 https://doi.org/10.1038/s41568-021-00346-0
  25. Santos P and Almeida F (2021) Exosome-based vaccines: history, current state, and clinical trials. Front Immunol 12, 711565 https://doi.org/10.3389/fimmu.2021.711565
  26. Zitvogel L, Regnault A, Lozier A et al (1998) Eradication of established murine tumors using a novel cell-free vaccine: dendritic cell-derived exosomes. Nat Med 4, 594-600 https://doi.org/10.1038/nm0598-594
  27. Andre F, Chaput N, Schartz NE et al (2004) Exosomes as potent cell-free peptide-based vaccine. I. Dendritic cell-derived exosomes transfer functional MHC class I/peptide complexes to dendritic cells. J Immunol 172, 2126-2136 https://doi.org/10.4049/jimmunol.172.4.2126
  28. 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
  29. Matsumoto K, Morisaki T, Kuroki H et al (2004) Exosomes secreted from monocyte-derived dendritic cells support in vitro naive CD4+ T cell survival through NF-(kappa)B activation. Cell Immunol 231, 20-29 https://doi.org/10.1016/j.cellimm.2004.11.002
  30. Qazi KR, Gehrmann U, Domange Jordo E, Karlsson MC and Gabrielsson S (2009) Antigen-loaded exosomes alone induce Th1-type memory through a B-cell-dependent mechanism. Blood 113, 2673-2683 https://doi.org/10.1182/blood-2008-04-153536
  31. Segura E, Nicco C, Lombard B et al (2005) ICAM-1 on exosomes from mature dendritic cells is critical for efficient naive T-cell priming. Blood 106, 216-223 https://doi.org/10.1182/blood-2005-01-0220
  32. Naslund TI, Gehrmann U, Qazi KR, Karlsson MC and Gabrielsson S (2013) Dendritic cell-derived exosomes need to activate both T and B cells to induce antitumor immunity. J Immunol 190, 2712-2719 https://doi.org/10.4049/jimmunol.1203082
  33. Wei G, Jie Y, Haibo L et al (2017) Dendritic cells derived exosomes migration to spleen and induction of inflammation are regulated by CCR7. Sci Rep 7, 42996 https://doi.org/10.1038/srep42996
  34. Nolte-'t Hoen EN, Buschow SI, Anderton SM, Stoorvogel W and Wauben MH (2009) Activated T cells recruit exosomes secreted by dendritic cells via LFA-1. Blood 113, 1977-1981 https://doi.org/10.1182/blood-2008-08-174094
  35. Thery C, Duban L, Segura E, Veron P, Lantz O and Amigorena S (2002) Indirect activation of naive CD4+ T cells by dendritic cell-derived exosomes. Nat Immunol 3, 1156-1162 https://doi.org/10.1038/ni854
  36. Romagnoli GG, Zelante BB, Toniolo PA, Migliori IK and Barbuto JA (2014) Dendritic cell-derived exosomes may be a tool for cancer immunotherapy by converting tumor cells into immunogenic targets. Front Immunol 5, 692 https://doi.org/10.3389/fimmu.2014.00692
  37. Guan S, Li Q, Liu P, Xuan X and Du Y (2014) Umbilical cord blood-derived dendritic cells loaded with BGC823 tumor antigens and DC-derived exosomes stimulate efficient cytotoxic T-lymphocyte responses and antitumor immunity in vitro and in vivo. Cent Eur J Immunol 39, 142-151
  38. Lu Z, Zuo B, Jing R et al (2017) Dendritic cell-derived exosomes elicit tumor regression in autochthonous hepatocellular carcinoma mouse models. J Hepatol 67, 739-748 https://doi.org/10.1016/j.jhep.2017.05.019
  39. Viaud S, Terme M, Flament C et al (2009) Dendritic cellderived 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
  40. 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
  41. 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
  42. Sobo-Vujanovic A, Munich S and Vujanovic NL (2014) Dendritic-cell exosomes cross-present Toll-like receptor-ligands and activate bystander dendritic cells. Cell Immunol 289, 119-127 https://doi.org/10.1016/j.cellimm.2014.03.016
  43. Simhadri VR, Reiners KS, Hansen HP et al (2008) Dendritic cells release HLA-B-associated transcript-3 positive exosomes to regulate natural killer function. PLoS One 3, e3377 https://doi.org/10.1371/journal.pone.0003377
  44. Wang J, Wang Z, Mo Y, Zeng Z, Wei P and Li T (2015) Effect of hyperthermic CO2-treated dendritic cell-derived exosomes on the human gastric cancer AGS cell line. Oncol Lett 10, 71-76 https://doi.org/10.3892/ol.2015.3155
  45. Guo F, Chang CK, Fan HH et al (2008) Anti-tumour effects of exosomes in combination with cyclophosphamide and polyinosinic-polycytidylic acid. J Int Med Res 36, 1342-1353 https://doi.org/10.1177/147323000803600623
  46. 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
  47. Min H, Sun X, Yang X et al (2018) Exosomes derived from irradiated esophageal carcinoma-infiltrating T cells promote metastasis by inducing the epithelial-mesenchymal transition in esophageal cancer cells. Pathol Oncol Res 24, 11-18 https://doi.org/10.1007/s12253-016-0185-z
  48. 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
  49. Zhang H, Xie Y, Li W, Chibbar R, Xiong S and Xiang J (2011) CD4(+) T cell-released exosomes inhibit CD8(+) cytotoxic T-lymphocyte responses and antitumor immunity. Cell Mol Immunol 8, 23-30 https://doi.org/10.1038/cmi.2010.59
  50. Lu J, Wu J, Xie F et al (2019) CD4(+) T cell-released extracellular vesicles potentiate the efficacy of the HBsAg vaccine by enhancing B cell responses. Adv Sci (Weinh) 6, 1802219 https://doi.org/10.1002/advs.201802219
  51. Peters PJ, Borst J, Oorschot V et al (1991) Cytotoxic T lymphocyte granules are secretory lysosomes, containing both perforin and granzymes. J Exp Med 173, 1099-1109 https://doi.org/10.1084/jem.173.5.1099
  52. 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
  53. Seo N, Shirakura Y, Tahara Y et al (2018) Activated CD8(+) T cell extracellular vesicles prevent tumour progression by targeting of lesional mesenchymal cells. Nat Commun 9, 435 https://doi.org/10.1038/s41467-018-02865-1
  54. Cai Z, Yang F, Yu L et al (2012) Activated T cell exosomes promote tumor invasion via Fas signaling pathway. J Immunol 188, 5954-5961 https://doi.org/10.4049/jimmunol.1103466
  55. Xie Y, Zhang H, Li W et al (2010) Dendritic cells recruit T cell exosomes via exosomal LFA-1 leading to inhibition of CD8+ CTL responses through downregulation of peptide/MHC class I and Fas ligand-mediated cytotoxicity. J Immunol 185, 5268-5278 https://doi.org/10.4049/jimmunol.1000386
  56. 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
  57. 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
  58. 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
  59. 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
  60. Zhu L, Kalimuthu S, Oh JM et al (2019) Enhancement of antitumor potency of extracellular vesicles derived from natural killer cells by IL-15 priming. Biomaterials 190-191, 38-50 https://doi.org/10.1016/j.biomaterials.2018.10.034
  61. Ngambenjawong C, Gustafson HH and Pun SH (2017) Progress in tumor-associated macrophage (TAM)-targeted therapeutics. Adv Drug Deliv Rev 114, 206-221 https://doi.org/10.1016/j.addr.2017.04.010
  62. Bernsmeier C, van der Merwe S and Perianin A (2020) Innate immune cells in cirrhosis. J Hepatol 73, 186-201 https://doi.org/10.1016/j.jhep.2020.03.027
  63. 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
  64. 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
  65. Li Z, Suo B, Long G et al (2020) Exosomal miRNA-16-5p derived from M1 macrophages enhances T cell-dependent immune response by regulating PD-L1 in gastric cancer. Front Cell Dev Biol 8, 572689 https://doi.org/10.3389/fcell.2020.572689
  66. Lin Y, Xu J and Lan H (2019) Tumor-associated macrophages in tumor metastasis: biological roles and clinical therapeutic applications. J Hematol Oncol 12, 76 https://doi.org/10.1186/s13045-019-0760-3
  67. Wu XQ, Dai Y, Yang Y et al (2016) Emerging role of microRNAs in regulating macrophage activation and polarization in immune response and inflammation. Immunology 148, 237-248 https://doi.org/10.1111/imm.12608
  68. Choo YW, Kang M, Kim HY et al (2018) M1 macrophage-derived nanovesicles potentiate the anticancer efficacy of immune checkpoint inhibitors. ACS Nano 12, 8977-8993 https://doi.org/10.1021/acsnano.8b02446
  69. Derakhshani A, Vahidian F, Alihasanzadeh M, Mokhtarzadeh A, Lotfi Nezhad P and Baradaran B (2019) Mast cells: a double-edged sword in cancer. Immunol Lett 209, 28-35 https://doi.org/10.1016/j.imlet.2019.03.011
  70. Farcas M and Inngjerdingen M (2020) Natural killer cellderived extracellular vesicles in cancer therapy. Scand J Immunol 92, e12938 https://doi.org/10.1111/sji.12938
  71. 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
  72. 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
  73. 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