Exosomes and the Future of Immunotherapy in Pancreatic Cancer
Abstract
:1. Introduction
2. Biogenesis of Exosomes
3. Cancer Exosomes and Immune Response in PDAC
4. Advantages of the Use of Exosomes in Immunotherapy
5. Exosomes-Based Immunotherapy Approaches for the Treatment of PDAC
6. Final Remarks
Funding
Conflicts of Interest
References
- Ferlay, J.; Ervik, M.; Lam, F.; Colombet, M.; Mery, L.; Piñeros, M.; Znaor, A.; Soerjomataram, I.; Bray, F. Global Cancer Observatory. Lyon, France: International Agency for Research on Cancer. 2018. Available online: https://gco.Iarc.Fr (accessed on 8 October 2018).
- Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: Globocan estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [PubMed]
- Hidalgo, M.; Cascinu, S.; Kleeff, J.; Labianca, R.; Löhr, J.M.; Neoptolemos, J.; Real, F.X.; Van Laethem, J.L.; Heinemann, V. Addressing the challenges of pancreatic cancer: Future directions for improving outcomes. Pancreatology 2015, 15, 8–18. [Google Scholar] [CrossRef] [Green Version]
- Huang, L.; Jansen, L.; Balavarca, Y.; Babaei, M.; van der Geest, L.; Lemmens, V.; Van Eycken, L.; De Schutter, H.; Johannesen, T.B.; Primic-Žakelj, M.; et al. Stratified survival of resected and overall pancreatic cancer patients in europe and the USA in the early twenty-first century: A large, international population-based study. BMC Med. 2018, 16, 125. [Google Scholar] [CrossRef]
- Ilic, M.; Ilic, I. Epidemiology of pancreatic cancer. World J. Gastroenterol. 2016, 22, 9694–9705. [Google Scholar] [CrossRef] [PubMed]
- Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2018. CA Cancer J. Clin. 2018, 68, 7–30. [Google Scholar] [CrossRef] [PubMed]
- Garrido-Laguna, I.; Hidalgo, M. Pancreatic cancer: From state-of-the-art treatments to promising novel therapies. Nat. Rev. Clin. Oncol. 2015, 12, 319–334. [Google Scholar] [CrossRef]
- Werner, J.; Combs, S.E.; Springfeld, C.; Hartwig, W.; Hackert, T.; Büchler, M.W. Advanced-stage pancreatic cancer: Therapy options. Nat. Rev. Clin. Oncol. 2013, 10, 323–333. [Google Scholar] [CrossRef] [PubMed]
- Groot, V.P.; Rezaee, N.; Wu, W.; Cameron, J.L.; Fishman, E.K.; Hruban, R.H.; Weiss, M.J.; Zheng, L.; Wolfgang, C.L.; He, J. Patterns, timing, and predictors of recurrence following pancreatectomy for pancreatic ductal adenocarcinoma. Ann. Surg. 2018, 267, 936–945. [Google Scholar] [CrossRef]
- Karachristos, A.; Esnaola, N.F. Surgical management of pancreatic neoplasms: What’s new? Curr. Gastroenterol. Rep. 2014, 16, 397. [Google Scholar] [CrossRef] [PubMed]
- Artinyan, A.; Anaya, D.A.; McKenzie, S.; Ellenhorn, J.D.; Kim, J. Neoadjuvant therapy is associated with improved survival in resectable pancreatic adenocarcinoma. Cancer 2011, 117, 2044–2049. [Google Scholar] [CrossRef]
- Inman, K.S.; Francis, A.A.; Murray, N.R. Complex role for the immune system in initiation and progression of pancreatic cancer. World J. Gastroenterol. 2014, 20, 11160–11181. [Google Scholar] [CrossRef]
- Clark, C.E.; Hingorani, S.R.; Mick, R.; Combs, C.; Tuveson, D.A.; Vonderheide, R.H. Dynamics of the immune reaction to pancreatic cancer from inception to invasion. Cancer Res. 2007, 67, 9518–9527. [Google Scholar] [CrossRef] [PubMed]
- Sahin, I.H.; Askan, G.; Hu, Z.I.; O’Reilly, E.M. Immunotherapy in pancreatic ductal adenocarcinoma: An emerging entity? Ann. Oncol. 2017, 28, 2950–2961. [Google Scholar] [CrossRef]
- Winograd, R.; Byrne, K.T.; Evans, R.A.; Odorizzi, P.M.; Meyer, A.R.; Bajor, D.L.; Clendenin, C.; Stanger, B.Z.; Furth, E.E.; Wherry, E.J.; et al. Induction of t-cell immunity overcomes complete resistance to pd-1 and ctla-4 blockade and improves survival in pancreatic carcinoma. Cancer Immunol. Res. 2015, 3, 399–411. [Google Scholar] [CrossRef] [PubMed]
- Royal, R.E.; Levy, C.; Turner, K.; Mathur, A.; Hughes, M.; Kammula, U.S.; Sherry, R.M.; Topalian, S.L.; Yang, J.C.; Lowy, I.; et al. Phase 2 trial of single agent ipilimumab (anti-ctla-4) for locally advanced or metastatic pancreatic adenocarcinoma. J. Immunother. 2010, 33, 828–833. [Google Scholar] [CrossRef] [PubMed]
- Aglietta, M.; Barone, C.; Sawyer, M.B.; Moore, M.J.; Miller, W.H.; Bagalà, C.; Colombi, F.; Cagnazzo, C.; Gioeni, L.; Wang, E.; et al. A phase i dose escalation trial of tremelimumab (cp-675,206) in combination with gemcitabine in chemotherapy-naive patients with metastatic pancreatic cancer. Ann. Oncol. 2014, 25, 1750–1755. [Google Scholar] [CrossRef]
- Brahmer, J.R.; Tykodi, S.S.; Chow, L.Q.; Hwu, W.J.; Topalian, S.L.; Hwu, P.; Drake, C.G.; Camacho, L.H.; Kauh, J.; Odunsi, K.; et al. Safety and activity of anti-pd-l1 antibody in patients with advanced cancer. N. Engl. J. Med. 2012, 366, 2455–2465. [Google Scholar] [CrossRef]
- Topalian, S.L.; Drake, C.G.; Pardoll, D.M. Immune checkpoint blockade: A common denominator approach to cancer therapy. Cancer Cell 2015, 27, 450–461. [Google Scholar] [CrossRef]
- Guo, S.; Contratto, M.; Miller, G.; Leichman, L.; Wu, J. Immunotherapy in pancreatic cancer: Unleash its potential through novel combinations. World J. Clin. Oncol. 2017, 8, 230–240. [Google Scholar] [CrossRef]
- Théry, C.; Zitvogel, L.; Amigorena, S. Exosomes: Composition, biogenesis and function. Nat. Rev. Immunol. 2002, 2, 569–579. [Google Scholar] [CrossRef]
- Tkach, M.; Théry, C. Communication by extracellular vesicles: Where we are and where we need to go. Cell 2016, 164, 1226–1232. [Google Scholar] [CrossRef]
- Kalluri, R. The biology and function of exosomes in cancer. J. Clin. Investig. 2016, 126, 1208–1215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kalluri, R.; LeBleu, V.S. Discovery of double-stranded genomic DNA in circulating exosomes. Cold Spring Harb. Symp. Quant. Biol. 2016, 81, 275–280. [Google Scholar] [CrossRef]
- Kamerkar, S.; LeBleu, V.S.; Sugimoto, H.; Yang, S.; Ruivo, C.F.; Melo, S.A.; Lee, J.J.; Kalluri, R. Exosomes facilitate therapeutic targeting of oncogenic kras in pancreatic cancer. Nature 2017, 546, 498–503. [Google Scholar] [CrossRef]
- Théry, C.; Ostrowski, M.; Segura, E. Membrane vesicles as conveyors of immune responses. Nat. Rev. Immunol. 2009, 9, 581–593. [Google Scholar] [CrossRef] [PubMed]
- Pitt, J.M.; Charrier, M.; Viaud, S.; André, F.; Besse, B.; Chaput, N.; Zitvogel, L. Dendritic cell-derived exosomes as immunotherapies in the fight against cancer. J. Immunol. 2014, 193, 1006–1011. [Google Scholar] [CrossRef] [PubMed]
- Pan, B.T.; Johnstone, R.M. Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: Selective externalization of the receptor. Cell 1983, 33, 967–978. [Google Scholar] [CrossRef]
- Rajagopal, C.; Harikumar, K.B. The origin and functions of exosomes in cancer. Front. Oncol. 2018, 8, 66. [Google Scholar] [CrossRef]
- EL Andaloussi, S.; Mäger, I.; Breakefield, X.O.; Wood, M.J. Extracellular vesicles: Biology and emerging therapeutic opportunities. Nat. Rev. Drug Discov. 2013, 12, 347–357. [Google Scholar] [CrossRef]
- Thakur, B.K.; Zhang, H.; Becker, A.; Matei, I.; Huang, Y.; Costa-Silva, B.; Zheng, Y.; Hoshino, A.; Brazier, H.; Xiang, J.; et al. Double-stranded DNA in exosomes: A novel biomarker in cancer detection. Cell. Res. 2014, 24, 766–769. [Google Scholar] [CrossRef]
- Yang, S.; Che, S.P.; Kurywchak, P.; Tavormina, J.L.; Gansmo, L.B.; Correa de Sampaio, P.; Tachezy, M.; Bockhorn, M.; Gebauer, F.; Haltom, A.R.; et al. Detection of mutant kras and tp53 DNA in circulating exosomes from healthy individuals and patients with pancreatic cancer. Cancer Biol. Ther. 2017, 18, 158–165. [Google Scholar] [CrossRef]
- Zhang, X.; Yuan, X.; Shi, H.; Wu, L.; Qian, H.; Xu, W. Exosomes in cancer: Small particle, big player. J. Hematol. Oncol. 2015, 8, 83. [Google Scholar] [CrossRef] [PubMed]
- Taylor, D.D.; Gercel-Taylor, C. Microrna signatures of tumor-derived exosomes as diagnostic biomarkers of ovarian cancer. Gynecol. Oncol. 2008, 110, 13–21. [Google Scholar] [CrossRef] [PubMed]
- Morelli, A.E.; Larregina, A.T.; Shufesky, W.J.; Sullivan, M.L.; Stolz, D.B.; Papworth, G.D.; Zahorchak, A.F.; Logar, A.J.; Wang, Z.; Watkins, S.C.; et al. Endocytosis, intracellular sorting, and processing of exosomes by dendritic cells. Blood 2004, 104, 3257–3266. [Google Scholar] [CrossRef] [Green Version]
- Segura, E.; Guérin, C.; Hogg, N.; Amigorena, S.; Théry, C. Cd8+ dendritic cells use lfa-1 to capture mhc-peptide complexes from exosomes in vivo. J. Immunol. 2007, 179, 1489–1496. [Google Scholar] [CrossRef] [PubMed]
- Nolte-’t Hoen, E.N.; Buschow, S.I.; Anderton, S.M.; Stoorvogel, W.; Wauben, M.H. Activated t cells recruit exosomes secreted by dendritic cells via lfa-1. Blood 2009, 113, 1977–1981. [Google Scholar] [CrossRef]
- Rana, S.; Yue, S.; Stadel, D.; Zöller, M. Toward tailored exosomes: The exosomal tetraspanin web contributes to target cell selection. Int. J. Biochem. Cell Biol. 2012, 44, 1574–1584. [Google Scholar] [CrossRef]
- Minciacchi, V.R.; Freeman, M.R.; Di Vizio, D. Extracellular vesicles in cancer: Exosomes, microvesicles and the emerging role of large oncosomes. Semin. Cell Dev. Biol. 2015, 40, 41–51. [Google Scholar] [CrossRef]
- Cordonnier, M.; Chanteloup, G.; Isambert, N.; Seigneuric, R.; Fumoleau, P.; Garrido, C.; Gobbo, J. Exosomes in cancer theranostic: Diamonds in the rough. Cell. Adh. Migr. 2017, 11, 151–163. [Google Scholar] [CrossRef]
- Hurley, J.H. Escrt complexes and the biogenesis of multivesicular bodies. Curr. Opin. Cell Biol. 2008, 20, 4–11. [Google Scholar] [CrossRef]
- Williams, R.L.; Urbé, S. The emerging shape of the escrt machinery. Nat. Rev. Mol. Cell Biol. 2007, 8, 355–368. [Google Scholar] [CrossRef] [PubMed]
- Colombo, M.; Raposo, G.; Théry, C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu. Rev. Cell Dev. Biol. 2014, 30, 255–289. [Google Scholar] [CrossRef] [PubMed]
- Tamai, K.; Tanaka, N.; Nakano, T.; Kakazu, E.; Kondo, Y.; Inoue, J.; Shiina, M.; Fukushima, K.; Hoshino, T.; Sano, K.; et al. Exosome secretion of dendritic cells is regulated by hrs, an escrt-0 protein. Biochem. Biophys. Res. Commun. 2010, 399, 384–390. [Google Scholar] [CrossRef]
- Gross, J.C.; Chaudhary, V.; Bartscherer, K.; Boutros, M. Active wnt proteins are secreted on exosomes. Nat. Cell Biol. 2012, 14, 1036–1045. [Google Scholar] [CrossRef]
- Colombo, M.; Moita, C.; van Niel, G.; Kowal, J.; Vigneron, J.; Benaroch, P.; Manel, N.; Moita, L.F.; Théry, C.; Raposo, G. Analysis of escrt functions in exosome biogenesis, composition and secretion highlights the heterogeneity of extracellular vesicles. J. Cell Sci. 2013, 126, 5553–5565. [Google Scholar] [CrossRef]
- Stuffers, S.; Sem Wegner, C.; Stenmark, H.; Brech, A. Multivesicular endosome biogenesis in the absence of escrts. Traffic 2009, 10, 925–937. [Google Scholar] [CrossRef] [PubMed]
- Trajkovic, K.; Hsu, C.; Chiantia, S.; Rajendran, L.; Wenzel, D.; Wieland, F.; Schwille, P.; Brügger, B.; Simons, M. Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science 2008, 319, 1244–1247. [Google Scholar] [CrossRef]
- Van Niel, G.; Charrin, S.; Simoes, S.; Romao, M.; Rochin, L.; Saftig, P.; Marks, M.S.; Rubinstein, E.; Raposo, G. The tetraspanin cd63 regulates escrt-independent and -dependent endosomal sorting during melanogenesis. Dev. Cell 2011, 21, 708–721. [Google Scholar] [CrossRef]
- Perez-Hernandez, D.; Gutiérrez-Vázquez, C.; Jorge, I.; López-Martín, S.; Ursa, A.; Sánchez-Madrid, F.; Vázquez, J.; Yáñez-Mó, M. The intracellular interactome of tetraspanin-enriched microdomains reveals their function as sorting machineries toward exosomes. J. Biol. Chem. 2013, 288, 11649–11661. [Google Scholar] [CrossRef]
- Nazarenko, I.; Rana, S.; Baumann, A.; McAlear, J.; Hellwig, A.; Trendelenburg, M.; Lochnit, G.; Preissner, K.T.; Zöller, M. Cell surface tetraspanin tspan8 contributes to molecular pathways of exosome-induced endothelial cell activation. Cancer Res. 2010, 70, 1668–1678. [Google Scholar] [CrossRef]
- Géminard, C.; De Gassart, A.; Blanc, L.; Vidal, M. Degradation of ap2 during reticulocyte maturation enhances binding of hsc70 and alix to a common site on tfr for sorting into exosomes. Traffic 2004, 5, 181–193. [Google Scholar] [CrossRef] [PubMed]
- Bastos, N.; Ruivo, C.F.; da Silva, S.; Melo, S.A. Exosomes in cancer: Use them or target them? Semin. Cell. Dev. Biol. 2018, 78, 13–21. [Google Scholar] [CrossRef] [PubMed]
- Ruivo, C.F.; Adem, B.; Silva, M.; Melo, S.A. The biology of cancer exosomes: Insights and new perspectives. Cancer Res. 2017, 77, 6480–6488. [Google Scholar] [CrossRef] [PubMed]
- Gastpar, R.; Gehrmann, M.; Bausero, M.A.; Asea, A.; Gross, C.; Schroeder, J.A.; Multhoff, G. Heat shock protein 70 surface-positive tumor exosomes stimulate migratory and cytolytic activity of natural killer cells. Cancer Res. 2005, 65, 5238–5247. [Google Scholar] [CrossRef]
- Bloomston, M.; Frankel, W.L.; Petrocca, F.; Volinia, S.; Alder, H.; Hagan, J.P.; Liu, C.G.; Bhatt, D.; Taccioli, C.; Croce, C.M. Microrna expression patterns to differentiate pancreatic adenocarcinoma from normal pancreas and chronic pancreatitis. JAMA 2007, 297, 1901–1908. [Google Scholar] [CrossRef] [PubMed]
- Zhou, M.; Chen, J.; Zhou, L.; Chen, W.; Ding, G.; Cao, L. Pancreatic cancer derived exosomes regulate the expression of tlr4 in dendritic cells via mir-203. Cell. Immunol. 2014, 292, 65–69. [Google Scholar] [CrossRef]
- Apetoh, L.; Ghiringhelli, F.; Tesniere, A.; Obeid, M.; Ortiz, C.; Criollo, A.; Mignot, G.; Maiuri, M.C.; Ullrich, E.; Saulnier, P.; et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat. Med. 2007, 13, 1050–1059. [Google Scholar] [CrossRef] [PubMed]
- Ding, G.; Zhou, L.; Qian, Y.; Fu, M.; Chen, J.; Xiang, J.; Wu, Z.; Jiang, G.; Cao, L. Pancreatic cancer-derived exosomes transfer mirnas to dendritic cells and inhibit rfxap expression via mir-212-3p. Oncotarget 2015, 6, 29877–29888. [Google Scholar] [CrossRef]
- Basso, D.; Gnatta, E.; Padoan, A.; Fogar, P.; Furlanello, S.; Aita, A.; Bozzato, D.; Zambon, C.F.; Arrigoni, G.; Frasson, C.; et al. Pdac-derived exosomes enrich the microenvironment in mdscs in a smad4-dependent manner through a new calcium related axis. Oncotarget 2017, 8, 84928–84944. [Google Scholar] [CrossRef]
- Ino, Y.; Yamazaki-Itoh, R.; Shimada, K.; Iwasaki, M.; Kosuge, T.; Kanai, Y.; Hiraoka, N. Immune cell infiltration as an indicator of the immune microenvironment of pancreatic cancer. Br. J. Cancer 2013, 108, 914–923. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, G.; Huang, A.C.; Zhang, W.; Zhang, G.; Wu, M.; Xu, W.; Yu, Z.; Yang, J.; Wang, B.; Sun, H.; et al. Exosomal pd-l1 contributes to immunosuppression and is associated with anti-pd-1 response. Nature 2018, 560, 382–386. [Google Scholar] [CrossRef]
- Maybruck, B.T.; Pfannenstiel, L.W.; Diaz-Montero, M.; Gastman, B.R. Tumor-derived exosomes induce cd8+ t cell suppressors. J. Immunother. Cancer 2017, 5, 65. [Google Scholar] [CrossRef] [PubMed]
- Martínez-Bosch, N.; Fernández-Barrena, M.G.; Moreno, M.; Ortiz-Zapater, E.; Munné-Collado, J.; Iglesias, M.; André, S.; Gabius, H.J.; Hwang, R.F.; Poirier, F.; et al. Galectin-1 drives pancreatic carcinogenesis through stroma remodeling and hedgehog signaling activation. Cancer Res. 2014, 74, 3512–3524. [Google Scholar] [CrossRef] [PubMed]
- Kurahara, H.; Shinchi, H.; Mataki, Y.; Maemura, K.; Noma, H.; Kubo, F.; Sakoda, M.; Ueno, S.; Natsugoe, S.; Takao, S. Significance of m2-polarized tumor-associated macrophage in pancreatic cancer. J. Surg. Res. 2011, 167, e211–e219. [Google Scholar] [CrossRef] [PubMed]
- Hu, H.; Hang, J.J.; Han, T.; Zhuo, M.; Jiao, F.; Wang, L.W. The m2 phenotype of tumor-associated macrophages in the stroma confers a poor prognosis in pancreatic cancer. Tumour. Biol. 2016, 37, 8657–8664. [Google Scholar] [CrossRef]
- Linton, S.S.; Abraham, T.; Liao, J.; Clawson, G.A.; Butler, P.J.; Fox, T.; Kester, M.; Matters, G.L. Tumor-promoting effects of pancreatic cancer cell exosomes on thp-1-derived macrophages. PLoS ONE 2018, 13, e0206759. [Google Scholar] [CrossRef]
- Ahmadi, M.; Emery, D.C.; Morgan, D.J. Prevention of both direct and cross-priming of antitumor cd8+ t-cell responses following overproduction of prostaglandin e2 by tumor cells in vivo. Cancer Res. 2008, 68, 7520–7529. [Google Scholar] [CrossRef]
- Baratelli, F.; Lin, Y.; Zhu, L.; Yang, S.C.; Heuzé-Vourc’h, N.; Zeng, G.; Reckamp, K.; Dohadwala, M.; Sharma, S.; Dubinett, S.M. Prostaglandin e2 induces foxp3 gene expression and t regulatory cell function in human cd4+ t cells. J. Immunol. 2005, 175, 1483–1490. [Google Scholar] [CrossRef]
- Wang, Z.; von Au, A.; Schnölzer, M.; Hackert, T.; Zöller, M. Cd44v6-competent tumor exosomes promote motility, invasion and cancer-initiating cell marker expression in pancreatic and colorectal cancer cells. Oncotarget 2016, 7, 55409–55436. [Google Scholar] [CrossRef]
- Hoshino, A.; Costa-Silva, B.; Shen, T.L.; Rodrigues, G.; Hashimoto, A.; Tesic Mark, M.; Molina, H.; Kohsaka, S.; Di Giannatale, A.; Ceder, S.; et al. Tumour exosome integrins determine organotropic metastasis. Nature 2015, 527, 329–335. [Google Scholar] [CrossRef] [Green Version]
- Costa-Silva, B.; Aiello, N.M.; Ocean, A.J.; Singh, S.; Zhang, H.; Thakur, B.K.; Becker, A.; Hoshino, A.; Mark, M.T.; Molina, H.; et al. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat. Cell Biol. 2015, 17, 816–826. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yu, Z.; Zhao, S.; Ren, L.; Wang, L.; Chen, Z.; Hoffman, R.M.; Zhou, J. Pancreatic cancer-derived exosomes promote tumor metastasis and liver pre-metastatic niche formation. Oncotarget 2017, 8, 63461–63483. [Google Scholar] [CrossRef] [Green Version]
- Chalmin, F.; Ladoire, S.; Mignot, G.; Vincent, J.; Bruchard, M.; Remy-Martin, J.P.; Boireau, W.; Rouleau, A.; Simon, B.; Lanneau, D.; et al. Membrane-associated hsp72 from tumor-derived exosomes mediates stat3-dependent immunosuppressive function of mouse and human myeloid-derived suppressor cells. J. Clin. Investig. 2010, 120, 457–471. [Google Scholar] [CrossRef]
- Melo, S.A.; Sugimoto, H.; O’Connell, J.T.; Kato, N.; Villanueva, A.; Vidal, A.; Qiu, L.; Vitkin, E.; Perelman, L.T.; Melo, C.A.; et al. Cancer exosomes perform cell-independent microrna biogenesis and promote tumorigenesis. Cancer Cell. 2014, 26, 707–721. [Google Scholar] [CrossRef] [PubMed]
- Abd Elmageed, Z.Y.; Yang, Y.; Thomas, R.; Ranjan, M.; Mondal, D.; Moroz, K.; Fang, Z.; Rezk, B.M.; Moparty, K.; Sikka, S.C.; et al. Neoplastic reprogramming of patient-derived adipose stem cells by prostate cancer cell-associated exosomes. Stem Cells 2014, 32, 983–997. [Google Scholar] [CrossRef]
- Takikawa, T.; Masamune, A.; Yoshida, N.; Hamada, S.; Kogure, T.; Shimosegawa, T. Exosomes derived from pancreatic stellate cells: Microrna signature and effects on pancreatic cancer cells. Pancreas 2017, 46, 19–27. [Google Scholar] [CrossRef]
- Beloribi-Djefaflia, S.; Siret, C.; Lombardo, D. Exosomal lipids induce human pancreatic tumoral miapaca-2 cells resistance through the cxcr4-sdf-1α signaling axis. Oncoscience 2015, 2, 15–30. [Google Scholar] [PubMed]
- Kucharzewska, P.; Christianson, H.C.; Welch, J.E.; Svensson, K.J.; Fredlund, E.; Ringnér, M.; Mörgelin, M.; Bourseau-Guilmain, E.; Bengzon, J.; Belting, M. 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, 7312–7317. [Google Scholar] [CrossRef] [Green Version]
- Richards, K.E.; Zeleniak, A.E.; Fishel, M.L.; Wu, J.; Littlepage, L.E.; Hill, R. Cancer-associated fibroblast exosomes regulate survival and proliferation of pancreatic cancer cells. Oncogene 2017, 36, 1770–1778. [Google Scholar] [CrossRef]
- Binenbaum, Y.; Fridman, E.; Yaari, Z.; Milman, N.; Schroeder, A.; Ben David, G.; Shlomi, T.; Gil, Z. Transfer of mirna in macrophage-derived exosomes induces drug resistance in pancreatic adenocarcinoma. Cancer Res. 2018, 78, 5287–5299. [Google Scholar] [CrossRef]
- Hendrix, A.; Maynard, D.; Pauwels, P.; Braems, G.; Denys, H.; Van den Broecke, R.; Lambert, J.; Van Belle, S.; Cocquyt, V.; Gespach, C.; et al. Effect of the secretory small gtpase rab27b on breast cancer growth, invasion, and metastasis. J. Natl. Cancer Inst. 2010, 102, 866–880. [Google Scholar] [CrossRef] [PubMed]
- Sidhu, S.S.; Mengistab, A.T.; Tauscher, A.N.; LaVail, J.; Basbaum, C. The microvesicle as a vehicle for emmprin in tumor-stromal interactions. Oncogene 2004, 23, 956–963. [Google Scholar] [CrossRef]
- Li, Z.; Jiang, P.; Li, J.; Peng, M.; Zhao, X.; Zhang, X.; Chen, K.; Zhang, Y.; Liu, H.; Gan, L.; et al. Tumor-derived exosomal lnc-sox2ot promotes emt and stemness by acting as a cerna in pancreatic ductal adenocarcinoma. Oncogene 2018, 37, 3822–3838. [Google Scholar] [CrossRef] [PubMed]
- Blackwell, R.H.; Foreman, K.E.; Gupta, G.N. The role of cancer-derived exosomes in tumorigenicity & epithelial-to-mesenchymal transition. Cancers (Basel) 2017, 9, 105. [Google Scholar] [CrossRef]
- Wang, L.; Zhang, B.; Zheng, W.; Kang, M.; Chen, Q.; Qin, W.; Li, C.; Zhang, Y.; Shao, Y.; Wu, Y. Exosomes derived from pancreatic cancer cells induce insulin resistance in c2c12 myotube cells through the PI3K/akt/foxo1 pathway. Sci. Rep. 2017, 7, 5384. [Google Scholar] [CrossRef] [PubMed]
- Van den Boorn, J.G.; Schlee, M.; Coch, C.; Hartmann, G. Sirna delivery with exosome nanoparticles. Nat. Biotechnol. 2011, 29, 325–326. [Google Scholar] [CrossRef] [PubMed]
- Alvarez-Erviti, L.; Seow, Y.; Yin, H.; Betts, C.; Lakhal, S.; Wood, M.J. Delivery of sirna to the mouse brain by systemic injection of targeted exosomes. Nat. Biotechnol. 2011, 29, 341–345. [Google Scholar] [CrossRef]
- Tran, T.H.; Mattheolabakis, G.; Aldawsari, H.; Amiji, M. Exosomes as nanocarriers for immunotherapy of cancer and inflammatory diseases. Clin. Immunol. 2015, 160, 46–58. [Google Scholar] [CrossRef]
- Sercombe, L.; Veerati, T.; Moheimani, F.; Wu, S.Y.; Sood, A.K.; Hua, S. Advances and challenges of liposome assisted drug delivery. Front. Pharmacol. 2015, 6, 286. [Google Scholar] [CrossRef]
- Szebeni, J.; Moghimi, S.M. Liposome triggering of innate immune responses: A perspective on benefits and adverse reactions. J. Liposome Res. 2009, 19, 85–90. [Google Scholar] [CrossRef]
- Raposo, G.; Stoorvogel, W. Extracellular vesicles: Exosomes, microvesicles, and friends. J. Cell Biol. 2013, 200, 373–383. [Google Scholar] [CrossRef] [Green Version]
- Clayton, A.; Harris, C.L.; Court, J.; Mason, M.D.; Morgan, B.P. Antigen-presenting cell exosomes are protected from complement-mediated lysis by expression of cd55 and cd59. Eur. J. Immunol. 2003, 33, 522–531. [Google Scholar] [CrossRef] [PubMed]
- Sun, D.; Zhuang, X.; Xiang, X.; Liu, Y.; Zhang, S.; Liu, C.; Barnes, S.; Grizzle, W.; Miller, D.; Zhang, H.G. A novel nanoparticle drug delivery system: The anti-inflammatory activity of curcumin is enhanced when encapsulated in exosomes. Mol. Ther. 2010, 18, 1606–1614. [Google Scholar] [CrossRef]
- Munagala, R.; Aqil, F.; Jeyabalan, J.; Gupta, R.C. Bovine milk-derived exosomes for drug delivery. Cancer Lett. 2016, 371, 48–61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cheng, L.; Sharples, R.A.; Scicluna, B.J.; Hill, A.F. Exosomes provide a protective and enriched source of mirna for biomarker profiling compared to intracellular and cell-free blood. J. Extracell. Vesicles 2014, 3. [Google Scholar] [CrossRef]
- Chen, C.C.; Liu, L.; Ma, F.; Wong, C.W.; Guo, X.E.; Chacko, J.V.; Farhoodi, H.P.; Zhang, S.X.; Zimak, J.; Ségaliny, A.; et al. Elucidation of exosome migration across the blood-brain barrier model in vitro. Cell. Mol. Bioeng. 2016, 9, 509–529. [Google Scholar] [CrossRef] [PubMed]
- Yang, T.; Martin, P.; Fogarty, B.; Brown, A.; Schurman, K.; Phipps, R.; Yin, V.P.; Lockman, P.; Bai, S. Exosome delivered anticancer drugs across the blood-brain barrier for brain cancer therapy in danio rerio. Pharm. Res. 2015, 32, 2003–2014. [Google Scholar] [CrossRef] [PubMed]
- Vader, P.; Mol, E.A.; Pasterkamp, G.; Schiffelers, R.M. Extracellular vesicles for drug delivery. Adv. Drug Deliv. Rev. 2016, 106, 148–156. [Google Scholar] [CrossRef] [PubMed]
- Bellavia, D.; Raimondo, S.; Calabrese, G.; Forte, S.; Cristaldi, M.; Patinella, A.; Memeo, L.; Manno, M.; Raccosta, S.; Diana, P.; et al. Interleukin 3- receptor targeted exosomes inhibit in vitro and in vivo chronic myelogenous leukemia cell growth. Theranostics 2017, 7, 1333–1345. [Google Scholar] [CrossRef]
- Qi, H.; Liu, C.; Long, L.; Ren, Y.; Zhang, S.; Chang, X.; Qian, X.; Jia, H.; Zhao, J.; Sun, J.; et al. Blood exosomes endowed with magnetic and targeting properties for cancer therapy. ACS Nano 2016, 10, 3323–3333. [Google Scholar] [CrossRef]
- Valadi, H.; Ekström, K.; Bossios, A.; Sjöstrand, M.; Lee, J.J.; Lötvall, J.O. Exosome-mediated transfer of mrnas and micrornas is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 2007, 9, 654–659. [Google Scholar] [CrossRef] [PubMed]
- Boelens, M.C.; Wu, T.J.; Nabet, B.Y.; Xu, B.; Qiu, Y.; Yoon, T.; Azzam, D.J.; Twyman-Saint Victor, C.; Wiemann, B.Z.; Ishwaran, H.; et al. Exosome transfer from stromal to breast cancer cells regulates therapy resistance pathways. Cell 2014, 159, 499–513. [Google Scholar] [CrossRef] [PubMed]
- Li, W.; Zhang, X.; Wang, J.; Li, M.; Cao, C.; Tan, J.; Ma, D.; Gao, Q. Tgfβ1 in fibroblasts-derived exosomes promotes epithelial-mesenchymal transition of ovarian cancer cells. Oncotarget 2017, 8, 96035–96047. [Google Scholar] [PubMed]
- Ringuette Goulet, C.; Bernard, G.; Tremblay, S.; Chabaud, S.; Bolduc, S.; Pouliot, F. Exosomes induce fibroblast differentiation into cancer-associated fibroblasts through tgfβ signaling. Mol. Cancer Res. 2018, 16, 1196–1204. [Google Scholar] [CrossRef] [PubMed]
- Chowdhury, R.; Webber, J.P.; Gurney, M.; Mason, M.D.; Tabi, Z.; Clayton, A. Cancer exosomes trigger mesenchymal stem cell differentiation into pro-angiogenic and pro-invasive myofibroblasts. Oncotarget 2015, 6, 715–731. [Google Scholar] [CrossRef] [Green Version]
- Aspe, J.R.; Diaz Osterman, C.J.; Jutzy, J.M.; Deshields, S.; Whang, S.; Wall, N.R. Enhancement of gemcitabine sensitivity in pancreatic adenocarcinoma by novel exosome-mediated delivery of the survivin-t34a mutant. J. Extracell. Vesicles 2014, 3. [Google Scholar] [CrossRef] [PubMed]
- Mahmoodzadeh Hosseini, H.; Ali Imani Fooladi, A.; Soleimanirad, J.; Reza Nourani, M.; Mahdavi, M. Exosome/staphylococcal enterotoxin b, an anti tumor compound against pancreatic cancer. J. BUON 2014, 19, 440–448. [Google Scholar] [PubMed]
- Wahlgren, J.; De L Karlson, T.; Brisslert, M.; Vaziri Sani, F.; Telemo, E.; Sunnerhagen, P.; Valadi, H. Plasma exosomes can deliver exogenous short interfering rna to monocytes and lymphocytes. Nucleic Acids Res. 2012, 40, e130. [Google Scholar] [CrossRef] [PubMed]
- Zitvogel, L.; Regnault, A.; Lozier, A.; Wolfers, J.; Flament, C.; Tenza, D.; Ricciardi-Castagnoli, P.; Raposo, G.; Amigorena, S. Eradication of established murine tumors using a novel cell-free vaccine: Dendritic cell-derived exosomes. Nat. Med. 1998, 4, 594–600. [Google Scholar] [CrossRef] [PubMed]
- Viaud, S.; Terme, M.; Flament, C.; Taieb, J.; André, F.; Novault, S.; Escudier, B.; Robert, C.; Caillat-Zucman, S.; Tursz, T.; et al. Dendritic cell-derived exosomes promote natural killer cell activation and proliferation: A role for nkg2d ligands and il-15ralpha. PLoS ONE 2009, 4, e4942. [Google Scholar] [CrossRef] [PubMed]
- Raposo, G.; Nijman, H.W.; Stoorvogel, W.; Liejendekker, R.; Harding, C.V.; Melief, C.J.; Geuze, H.J. B lymphocytes secrete antigen-presenting vesicles. J. Exp. Med. 1996, 183, 1161–1172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Besse, B.; Charrier, M.; Lapierre, V.; Dansin, E.; Lantz, O.; Planchard, D.; Le Chevalier, T.; Livartoski, A.; Barlesi, F.; Laplanche, A.; et al. Dendritic cell-derived exosomes as maintenance immunotherapy after first line chemotherapy in nsclc. Oncoimmunology 2016, 5, e1071008. [Google Scholar] [CrossRef]
- Dai, S.; Wei, D.; Wu, Z.; Zhou, X.; Wei, X.; Huang, H.; Li, G. Phase i clinical trial of autologous ascites-derived exosomes combined with gm-csf for colorectal cancer. Mol. Ther. 2008, 16, 782–790. [Google Scholar] [CrossRef] [PubMed]
- Muller, L.; Mitsuhashi, M.; Simms, P.; Gooding, W.E.; Whiteside, T.L. Tumor-derived exosomes regulate expression of immune function-related genes in human t cell subsets. Sci. Rep. 2016, 6, 20254. [Google Scholar] [CrossRef] [PubMed]
- Szajnik, M.; Czystowska, M.; Szczepanski, M.J.; Mandapathil, M.; Whiteside, T.L. Tumor-derived microvesicles induce, expand and up-regulate biological activities of human regulatory t cells (treg). PLoS ONE 2010, 5, e11469. [Google Scholar] [CrossRef]
- Xiao, L.; Erb, U.; Zhao, K.; Hackert, T.; Zöller, M. Efficacy of vaccination with tumor-exosome loaded dendritic cells combined with cytotoxic drug treatment in pancreatic cancer. Oncoimmunology 2017, 6, e1319044. [Google Scholar] [CrossRef] [Green Version]
- Su, M.J.; Aldawsari, H.; Amiji, M. Pancreatic cancer cell exosome-mediated macrophage reprogramming and the role of micrornas 155 and 125b2 transfection using nanoparticle delivery systems. Sci. Rep. 2016, 6, 30110. [Google Scholar] [CrossRef]
- Bobrie, A.; Krumeich, S.; Reyal, F.; Recchi, C.; Moita, L.F.; Seabra, M.C.; Ostrowski, M.; Théry, C. Rab27a supports exosome-dependent and -independent mechanisms that modify the tumor microenvironment and can promote tumor progression. Cancer Res. 2012, 72, 4920–4930. [Google Scholar] [CrossRef]
- Ostenfeld, M.S.; Jeppesen, D.K.; Laurberg, J.R.; Boysen, A.T.; Bramsen, J.B.; Primdal-Bengtson, B.; Hendrix, A.; Lamy, P.; Dagnaes-Hansen, F.; Rasmussen, M.H.; et al. Cellular disposal of mir23b by rab27-dependent exosome release is linked to acquisition of metastatic properties. Cancer Res. 2014, 74, 5758–5771. [Google Scholar] [CrossRef]
- Datta, A.; Kim, H.; McGee, L.; Johnson, A.E.; Talwar, S.; Marugan, J.; Southall, N.; Hu, X.; Lal, M.; Mondal, D.; et al. High-throughput screening identified selective inhibitors of exosome biogenesis and secretion: A drug repurposing strategy for advanced cancer. Sci. Rep. 2018, 8, 8161. [Google Scholar] [CrossRef]
- Ha, D.; Yang, N.; Nadithe, V. Exosomes as therapeutic drug carriers and delivery vehicles across biological membranes: Current perspectives and future challenges. Acta Pharm. Sin. B 2016, 6, 287–296. [Google Scholar] [CrossRef] [PubMed]
- Munson, P.; Shukla, A. Exosomes: Potential in cancer diagnosis and therapy. Medicines 2015, 2, 310–327. [Google Scholar] [CrossRef] [PubMed]
- Kim, M.S.; Haney, M.J.; Zhao, Y.; Mahajan, V.; Deygen, I.; Klyachko, N.L.; Inskoe, E.; Piroyan, A.; Sokolsky, M.; Okolie, O.; et al. Development of exosome-encapsulated paclitaxel to overcome mdr in cancer cells. Nanomedicine 2016, 12, 655–664. [Google Scholar] [CrossRef] [PubMed]
- André, F.; Chaput, N.; Schartz, N.E.; Flament, C.; Aubert, N.; Bernard, J.; Lemonnier, F.; Raposo, G.; Escudier, B.; Hsu, D.H.; et al. 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. 2004, 172, 2126–2136. [Google Scholar] [CrossRef] [PubMed]
Function in Cancer | Specific Role | References |
---|---|---|
Help Tumor Cells to Evade the Immune System | Activate NK cells | [55] |
Prevent DC antigen presentation | [57,59] | |
Favor an immunosuppressive phenotype in macrophages | [67] | |
Induce differentiation of myeloid cells towards MDSCs | [70] | |
Induce proliferation of TReg cells | [60] | |
Prevent proliferation and activation of effector T cells | [62,64] | |
Drive Tumor Development and Progression | Induce tumor growth and transformation and inhibit cancer cell death | [75,76,77,78] |
Stimulate angiogenesis | [51,79] | |
Contribute to drug resistance | [80,81] | |
Extracellular matrix remodeling | [82,83] | |
Promote EMT | [84,85] | |
Induce metabolic reprograming | [86] | |
Promote metastasis | Educate the cells and trigger inflammatory responses at the metastatic site | [71,72] |
Promote the recruitment of suppressive immune cells to promote the formation of a pre-metastatic niche | [71,72,73,74] |
Treatment Options | Cell of Origin | Type of Modification/Effect | Research | Main Findings | Ref | |
---|---|---|---|---|---|---|
Delivery of immuno-stimulatory molecules | Small molecules | Reticulocytes | Incubation of exosomal solution with doxorubicin | Delivery of doxorubicin to tumor cells via exosomes anchored to superparamagnetic nanoparticles | Doxorubicin was more effectively delivered to tumor cells via exosomes which were anchored to superparamagnetic nanoparticles; mice inoculated subcutaneously with H22 hepatocarcinoma cells presented decreased tumor growth when treated with these exosomes. | [101] |
Murine macrophages (i.e., RAW 264.7 cell line) | Sonication to load paclitaxel into exosomes | Study the efficacy of paclitaxel for the treatment of multiple drug resistant cancers when delivered via exosomes | Exosomes-delivered paclitaxel successfully reached tumor cells and inhibited the growth of pulmonary metastases in vivo (in mice injected via intra-tail vein with 3LL-M27 cells). | [124] | ||
Antibodies and antigens | Monocyte-derived, IFNγ-maturated DCs | Passive intracellular sorting of MHC molecules | Presentation of cancer antigens to cytotoxic immune cells via MHC-I and MHC-II expressed on the surface of DC-derived exosomes | Exosomes derived from IFNγ-maturated DCs stimulate the activation of NK cells, but not T cells; such effect is sufficient to improve the rate of progression-free survival in unresectable non-small cell lung cancer patients. However, these DC-derived exosomes were only effective as maintenance immunotherapy in less than 50% of the patient cohort. | [113] | |
Monocyte-derived, immature DCs | Pulsing of exosomal MHC-I proteins with Mart1 via acid elution | Presentation of cancer antigens to cytotoxic immune cells via MHC-I expressed on the surface of DC-derived exosomes | Exosomes containing MHC-I/Mart1 complexes are able to activate CD8+ T cells. | [125] | ||
Mouse bone marrow-derived DCs | Treatment of DCs with exosomes isolated from UNKC6141 murine pancreatic cancer cells | Study the activation of immune responses and survival in pancreatic cancer orthotopic mice models treated with tumor exosomes-loaded DCs | Tumor exosomes-loaded DCs were able to activate and recruit effector T cells to pancreatic cancer and improve prognosis when administered together with ATRA, sunitinib, or gemcitabine. | [117] | ||
HLA-DR15-positive human B cells | Incubation of cells and exosomes with Hsp65 antigen or antigenic peptide. | Study whether B cell-derived exosomes are able to activate T cells via MHC-mediated presentation of Hsp65 antigen | Exosomes incubated with Hsp65 or derived from the medium of B cells pre-incubated with Hsp65 were able to activate T cells. | [112] | ||
siRNA and RNAi | Human plasma cells | Electroporation of siRNA | Delivery of siRNA against MAPK1 to monocytes and T cells | Electroporated exosomes are able to enter human monocytes and lymphocytes (in vitro), deliver siRNA and successfully downregulate MAPK-1 transcription. | [109] | |
Human foreskin fibroblast cell line (i.e., BJ cell line) | Electroporation of RNAi molecules | Delivery of RNAi molecules against the mutant KRASG12D to pancreatic cancer cells | BJ-derived exosomes expressing RNAi against KRASG12D are able to specifically target mouse pancreatic cancer cells in vivo, diminishing the expression of KRASG12D and considerably diminishing cancer proliferation and metastasis, which resulted in an increase in mice overall survival. | [25] | ||
miRNA | Human pancreatic ductal adenocarcinoma cell line Panc-1 | Transfection of DNA plasmids for miR-155 and miR-125b2 into Panc-1 cells | Study phenotype changes on J774A.1 murine macrophages when treated with exosomes that overexpress miR-155 and miR-125b2 | M2 macrophages treated with exosomes derived from the transfected Panc-1 cells were reprogrammed back to an M1, anti-tumor phenotype. | [118] | |
Blocking exosomes biogenesis and secretion | GW4869 | All cells | Inhibition of exosomes biogenesis | Study the combination of gemcitabine with an inhibitor of exosomes biogenesis as a treatment option | GW4869 prevented chemoresistance caused by the increased amount of exosomes released by CAFs that were exposed to gemcitabine. | [80] |
Inhibition of Rab27a | 4T1 murine breast cancer cells | Infection with shRNA-expressing lentiviruses | Study of the role of Rab27a in exosomes secretion as well as in breast cancer growth and metastasis. | Blockade of Rab27a and, thus, of exosomes secretion decreased tumor growth and lung metastasis in mice inoculated with metastatic 4T1 cells. | [119] |
© 2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Batista, I.A.; Melo, S.A. Exosomes and the Future of Immunotherapy in Pancreatic Cancer. Int. J. Mol. Sci. 2019, 20, 567. https://doi.org/10.3390/ijms20030567
Batista IA, Melo SA. Exosomes and the Future of Immunotherapy in Pancreatic Cancer. International Journal of Molecular Sciences. 2019; 20(3):567. https://doi.org/10.3390/ijms20030567
Chicago/Turabian StyleBatista, Ines A., and Sonia A. Melo. 2019. "Exosomes and the Future of Immunotherapy in Pancreatic Cancer" International Journal of Molecular Sciences 20, no. 3: 567. https://doi.org/10.3390/ijms20030567
APA StyleBatista, I. A., & Melo, S. A. (2019). Exosomes and the Future of Immunotherapy in Pancreatic Cancer. International Journal of Molecular Sciences, 20(3), 567. https://doi.org/10.3390/ijms20030567