Extracellular Vesicles and Their Roles in the Tumor Immune Microenvironment
Abstract
1. Introduction
1.1. Extracellular Vesicles
1.2. The Tumor Microenvironment
1.3. EVs and the Tumor Immune Microenvironment
2. EVs and Tumor Immune Suppression
2.1. EVs and Tumor Immune Suppression: Role of T and B Cells
2.2. EVs and Tumor Immune Suppression: Role of Tregs
2.3. EVs and Tumor Immune Suppression: Role of Natural Killer Cells
2.4. EVs and Tumor Immune Suppression: Role of MDSCs
2.5. EVs and Tumor Immune Suppression: Role of Antigen Presenting Cells
2.6. EVs and Tumor Immune Suppression: Role of Monocytes and Macrophages
3. Immune Cells-Derived EVs
4. EV-Mediated Immune Activation
5. Translational Applications: Targeting EV-Mediated Tumor Immune Suppression
6. Limitations and Future Perspectives
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
Abbreviations
A | |
AML | acute myeloid leukaemia |
APCs | antigen presenting cells |
ARG1 | arginase-1 |
B | |
BAG6 | large proline-rich protein BAG6 |
C | |
CCL2 | C-C motif chemokine ligand 2 |
CCRs | chemokine receptors |
CLL | chronic lymphocytic leukemia |
CRC | colorectal cancer |
D | |
DAMPs | damage-associated molecular patterns |
DC | dendritic cells |
E | |
eTreg | effector Treg |
EGFR | epidermal growth factor receptor |
EMT | epithelial mesenchymal transition |
EOC | epithelial ovarian cancer |
ECM | extracellular matrix |
EVs | Extracellular Vesicles |
F | |
FAS-L | tumor necrosis factor ligand superfamily, member 6 |
FDA | Food and Drug Administration |
G | |
LGALS9 | galectin 9 |
GBM | glioblastoma multiforme |
gp100 | glycoprotein 100 |
H | |
HNSCC | head and neck squamous |
HDGF | hepatoma-derived growth factor |
HSP70 | heat shock protein 70 |
HA | hyaluronan |
HA | hyaluronic acid |
I | |
iDC | Immature DCs |
ICP | immune checkpoint proteins |
ICD | immunogenic cell death |
IFN-β | interferon-β |
L | |
LGALS9 | galectin 9 |
LPS | lipopolysaccharide |
lnc | long non-coding |
LNs | lymph nodes |
CD107a/LAMP1 | lysosomal-associated membrane protein 1 |
M | |
MEKK2 | MEK kinase 2 |
MART1 | melanoma-associated antigen 3-MAGE-A3 |
MIF | macrophage migration inhibitory factor |
MUC1 | mucin-1 |
MM | multiple myeloma |
MDSCs | Myeloid-derived suppressor cells |
N | |
NK | natural killer |
NKG2DL | NKG2D ligand |
NPM1 | nucleophosmin |
P | |
PDAC | pancreatic ductal adenocarcinoma |
PDCD4 | programmed cell death factor 4 |
PD1 | programmed death 1 |
PD-L1 | programmed death ligand 1 |
PGE2 | prostaglandin E2 |
PLP2 | proteolipid protein 2 |
R | |
RFXAP | regulatory factor X-associated protein |
Tregs | regulatory T cells |
S | |
SIRPalpha | signal regulatory protein alpha |
SCLC | small cell lung cancer |
sNMase | neutral sphingomyelinase |
nSMase | sphingomyelinase |
SPHK1 | sphingosine kinase-1 |
SPHK1 | sphingosine kinase 1 |
SCC | squamous cell carcinoma |
T | |
TCR | T-cell receptor |
TERF1 | telomeric repeat binding factor 1 |
TβRII | TGF-β type II receptor |
TLR4 | Toll-like receptor 4 |
TGF-β | transforming growth factor β |
SMAD3 | transforming growth factor-beta signaling protein 3 |
TAAs | tumor associated antigens |
TME | Tumor Microenvironment |
TNF | tumor necrosis factor |
TRAIL | tumor necrosis factor (TNF)-related apoptosis-inducing ligand |
TAMs | tumor-associated macrophages |
TEVs | tumor-derived EVs |
References
- Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; 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. [Google Scholar] [CrossRef]
- Kalluri, R.; LeBleu, V.S. The biology, function, and biomedical applications of exosomes. Science 2020, 367, eaau6977. [Google Scholar] [CrossRef]
- Wang, S.E. Extracellular vesicles in cancer therapy. Semin. Cancer Biol. 2022, 86, 296–309. [Google Scholar] [CrossRef]
- Reale, A.; Khong, T.; Xu, R.; Savvidou, I.; Lim, S.; Carmichael, I.; Greening, D.W.; Spencer, A. Understanding the role of extracellular vesicles in multiple myeloma. In Hematologic Malignancies; Copland, B., Ed.; Nova Science Publishers, Inc.: Hauppauge, NY, USA, 2020; pp. 217–261. [Google Scholar]
- Pietrowska, M.; Wlosowicz, A.; Gawin, M.; Widlak, P. MS-Based Proteomic Analysis of Serum and Plasma: Problem of High Abundant Components and Lights and Shadows of Albumin Removal. Adv. Exp. Med. Biol. 2019, 1073, 57–76. [Google Scholar] [CrossRef]
- Reale, A.; Khong, T.; Xu, R.; Chen, M.; Mithraprabhu, S.; Bingham, N.; Spencer, A.; Greening, D.W. Human Plasma Extracellular Vesicle Isolation and Proteomic Characterization for the Optimization of Liquid Biopsy in Multiple Myeloma. Methods Mol. Biol. 2021, 2261, 151–191. [Google Scholar] [CrossRef] [PubMed]
- Hoshino, A.; Kim, H.S.; Bojmar, L.; Gyan, K.E.; Cioffi, M.; Hernandez, J.; Zambirinis, C.P.; Rodrigues, G.; Molina, H.; Heissel, S.; et al. Extracellular Vesicle and Particle Biomarkers Define Multiple Human Cancers. Cell 2020, 182, 1044–1061.e18. [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] [PubMed]
- Peinado, H.; Alečković, M.; Lavotshkin, S.; Matei, I.; Costa-Silva, B.; Moreno-Bueno, G.; Hergueta-Redondo, M.; Williams, C.; García-Santos, G.; Ghajar, C.; et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat. Med. 2012, 18, 883–891. [Google Scholar] [CrossRef]
- Wang, J.; Hendrix, A.; Hernot, S.; Lemaire, M.; De Bruyne, E.; Van Valckenborgh, E.; Lahoutte, T.; De Wever, O.; Vanderkerken, K.; Menu, E. Bone marrow stromal cell-derived exosomes as communicators in drug resistance in multiple myeloma cells. Blood 2014, 124, 555–566. [Google Scholar] [CrossRef] [PubMed]
- Roccaro, A.M.; Sacco, A.; Maiso, P.; Azab, A.K.; Tai, Y.T.; Reagan, M.; Azab, F.; Flores, L.M.; Campigotto, F.; Weller, E.; et al. BM mesenchymal stromal cell-derived exosomes facilitate multiple myeloma progression. J. Clin. Investig. 2013, 123, 1542–1555. [Google Scholar] [CrossRef]
- Buzas, E.I. The roles of extracellular vesicles in the immune system. Nat. Rev. Immunol. 2022. [Google Scholar] [CrossRef] [PubMed]
- Hao, Q.; Wu, Y.; Wu, Y.; Wang, P.; Vadgama, J.V. Tumor-Derived Exosomes in Tumor-Induced Immune Suppression. Int. J. Mol. Sci. 2022, 23, 1461. [Google Scholar] [CrossRef] [PubMed]
- Xu, R.; Rai, A.; Chen, M.; Suwakulsiri, W.; Greening, D.W.; Simpson, R.J. Extracellular vesicles in cancer—Implications for future improvements in cancer care. Nat. Rev. Clin. Oncol. 2018, 15, 617–638. [Google Scholar] [CrossRef] [PubMed]
- Fontana, F.; Carollo, E.; Melling, G.E.; Carter, D.R.F. Extracellular Vesicles: Emerging Modulators of Cancer Drug Resistance. Cancers 2021, 13, 749. [Google Scholar] [CrossRef] [PubMed]
- Uchihara, T.; Miyake, K.; Yonemura, A.; Komohara, Y.; Itoyama, R.; Koiwa, M.; Yasuda, T.; Arima, K.; Harada, K.; Eto, K.; et al. Extracellular Vesicles from Cancer-Associated Fibroblasts Containing Annexin A6 Induces FAK-YAP Activation by Stabilizing beta1 Integrin, Enhancing Drug Resistance. Cancer Res. 2020, 80, 3222–3235. [Google Scholar] [CrossRef] [PubMed]
- Tutrone, R.; Donovan, M.J.; Torkler, P.; Tadigotla, V.; McLain, T.; Noerholm, M.; Skog, J.; McKiernan, J. Clinical utility of the exosome based ExoDx Prostate(IntelliScore) EPI test in men presenting for initial Biopsy with a PSA 2–10 ng/mL. Prostate Cancer Prostatic Dis. 2020, 23, 607–614. [Google Scholar] [CrossRef] [PubMed]
- Reale, A.; Carmichael, I.; Xu, R.; Mithraprabhu, S.; Khong, T.; Chen, M.; Fang, H.; Savvidou, I.; Ramachandran, M.; Bingham, N.; et al. Human myeloma cell- and plasma-derived extracellular vesicles contribute to functional regulation of stromal cells. Proteomics 2021, 21, e2000119. [Google Scholar] [CrossRef] [PubMed]
- Desantis, V.; Savino, F.D.; Scaringella, A.; Potenza, M.A.; Nacci, C.; Frassanito, M.A.; Vacca, A.; Montagnani, M. The Leading Role of the Immune Microenvironment in Multiple Myeloma: A New Target with a Great Prognostic and Clinical Value. J. Clin. Med. 2022, 11, 2513. [Google Scholar] [CrossRef]
- Weiss, F.; Lauffenburger, D.; Friedl, P. Towards targeting of shared mechanisms of cancer metastasis and therapy resistance. Nat. Rev. Cancer 2022, 22, 157–173. [Google Scholar] [CrossRef] [PubMed]
- Wu, F.; Yang, J.; Liu, J.; Wang, Y.; Mu, J.; Zeng, Q.; Deng, S.; Zhou, H. Signaling pathways in cancer-associated fibroblasts and targeted therapy for cancer. Signal Transduct. Target. Ther. 2021, 6, 218. [Google Scholar] [CrossRef]
- Morana, O.; Wood, W.; Gregory, C.D. The Apoptosis Paradox in Cancer. Int. J. Mol. Sci. 2022, 23, 1328. [Google Scholar] [CrossRef] [PubMed]
- Singleton, D.C.; Macann, A.; Wilson, W.R. Therapeutic targeting of the hypoxic tumour microenvironment. Nat. Rev. Clin. Oncol. 2021, 18, 751–772. [Google Scholar] [CrossRef] [PubMed]
- Valkenburg, K.C.; de Groot, A.E.; Pienta, K.J. Targeting the tumour stroma to improve cancer therapy. Nat. Rev. Clin. Oncol. 2018, 15, 366–381. [Google Scholar] [CrossRef]
- Pittet, M.J.; Michielin, O.; Migliorini, D. Clinical relevance of tumour-associated macrophages. Nat. Rev. Clin. Oncol. 2022, 19, 402–421. [Google Scholar] [CrossRef] [PubMed]
- Bruni, D.; Angell, H.K.; Galon, J. The immune contexture and Immunoscore in cancer prognosis and therapeutic efficacy. Nat. Rev. Cancer 2020, 20, 662–680. [Google Scholar] [CrossRef]
- Kalaora, S.; Nagler, A.; Wargo, J.A.; Samuels, Y. Mechanisms of immune activation and regulation: Lessons from melanoma. Nat. Rev. Cancer 2022, 22, 195–207. [Google Scholar] [CrossRef]
- Labani-Motlagh, A.; Ashja-Mahdavi, M.; Loskog, A. The Tumor Microenvironment: A Milieu Hindering and Obstructing Antitumor Immune Responses. Front. Immunol. 2020, 11, 940. [Google Scholar] [CrossRef]
- Kraehenbuehl, L.; Weng, C.H.; Eghbali, S.; Wolchok, J.D.; Merghoub, T. Enhancing immunotherapy in cancer by targeting emerging immunomodulatory pathways. Nat. Rev. Clin. Oncol. 2022, 19, 37–50. [Google Scholar] [CrossRef]
- Couzin-Frankel, J. Breakthrough of the year 2013. Cancer immunotherapy. Science 2013, 342, 1432–1433. [Google Scholar] [CrossRef]
- Wolchok, J. Putting the Immunologic Brakes on Cancer. Cell 2018, 175, 1452–1454. [Google Scholar] [CrossRef]
- Koury, J.; Lucero, M.; Cato, C.; Chang, L.; Geiger, J.; Henry, D.; Hernandez, J.; Hung, F.; Kaur, P.; Teskey, G.; et al. Immunotherapies: Exploiting the Immune System for Cancer Treatment. J. Immunol. Res. 2018, 2018, 9585614. [Google Scholar] [CrossRef] [PubMed]
- Murciano-Goroff, Y.R.; Warner, A.B.; Wolchok, J.D. The future of cancer immunotherapy: Microenvironment-targeting combinations. Cell Res. 2020, 30, 507–519. [Google Scholar] [CrossRef] [PubMed]
- Zhao, S.; Mi, Y.; Zheng, B.; Wei, P.; Gu, Y.; Zhang, Z.; Xu, Y.; Cai, S.; Li, X.; Li, D. Highly-metastatic colorectal cancer cell released miR-181a-5p-rich extracellular vesicles promote liver metastasis by activating hepatic stellate cells and remodelling the tumour microenvironment. J. Extracell. Vesicles 2022, 11, e12186. [Google Scholar] [CrossRef] [PubMed]
- Sirkisoon, S.R.; Wong, G.L.; Aguayo, N.R.; Doheny, D.L.; Zhu, D.; Regua, A.T.; Arrigo, A.; Manore, S.G.; Wagner, C.; Thomas, A.; et al. Breast cancer extracellular vesicles-derived miR-1290 activates astrocytes in the brain metastatic microenvironment via the FOXA2-->CNTF axis to promote progression of brain metastases. Cancer Lett. 2022, 540, 215726. [Google Scholar] [CrossRef]
- Giusti, I.; Di Francesco, M.; D’Ascenzo, S.; Palmerini, M.G.; Macchiarelli, G.; Carta, G.; Dolo, V. Ovarian cancer-derived extracellular vesicles affect normal human fibroblast behavior. Cancer Biol. Ther. 2018, 19, 722–734. [Google Scholar] [CrossRef]
- Fang, T.; Lv, H.; Lv, G.; Li, T.; Wang, C.; Han, Q.; Yu, L.; Su, B.; Guo, L.; Huang, S.; et al. Tumor-derived exosomal miR-1247-3p induces cancer-associated fibroblast activation to foster lung metastasis of liver cancer. Nat. Commun. 2018, 9, 191. [Google Scholar] [CrossRef]
- Forder, A.; Hsing, C.Y.; Trejo Vazquez, J.; Garnis, C. Emerging Role of Extracellular Vesicles and Cellular Communication in Metastasis. Cells 2021, 10, 3429. [Google Scholar] [CrossRef]
- Faict, S.; Muller, J.; De Veirman, K.; De Bruyne, E.; Maes, K.; Vrancken, L.; Heusschen, R.; De Raeve, H.; Schots, R.; Vanderkerken, K.; et al. Exosomes play a role in multiple myeloma bone disease and tumor development by targeting osteoclasts and osteoblasts. Blood Cancer J. 2018, 8, 105. [Google Scholar] [CrossRef]
- Menu, E.; Vanderkerken, K. Exosomes in Multiple Myeloma: From bench to bedside. Blood 2022. [Google Scholar] [CrossRef]
- Wang, J.; De Veirman, K.; Faict, S.; Frassanito, M.A.; Ribatti, D.; Vacca, A.; Menu, E. Multiple myeloma exosomes establish a favourable bone marrow microenvironment with enhanced angiogenesis and immunosuppression. J. Pathol. 2016, 239, 162–173. [Google Scholar] [CrossRef]
- Yamaguchi, T.; Kawamoto, E.; Gaowa, A.; Park, E.J.; Shimaoka, M. Remodeling of Bone Marrow Niches and Roles of Exosomes in Leukemia. Int. J. Mol. Sci. 2021, 22, 1881. [Google Scholar] [CrossRef]
- Vergani, E.; Daveri, E.; Vallacchi, V.; Bergamaschi, L.; Lalli, L.; Castelli, C.; Rodolfo, M.; Rivoltini, L.; Huber, V. Extracellular vesicles in anti-tumor immunity. Semin. Cancer Biol. 2022, 86, 64–79. [Google Scholar] [CrossRef] [PubMed]
- Marar, C.; Starich, B.; Wirtz, D. Extracellular vesicles in immunomodulation and tumor progression. Nat. Immunol. 2021, 22, 560–570. [Google Scholar] [CrossRef]
- Kugeratski, F.G.; Kalluri, R. Exosomes as mediators of immune regulation and immunotherapy in cancer. FEBS J. 2021, 288, 10–35. [Google Scholar] [CrossRef]
- Ludwig, S.; Floros, T.; Theodoraki, M.N.; Hong, C.S.; Jackson, E.K.; Lang, S.; Whiteside, T.L. Suppression of Lymphocyte Functions by Plasma Exosomes Correlates with Disease Activity in Patients with Head and Neck Cancer. Clin. Cancer Res. 2017, 23, 4843–4854. [Google Scholar] [CrossRef]
- Ricklefs, F.L.; Alayo, Q.; Krenzlin, H.; Mahmoud, A.B.; Speranza, M.C.; Nakashima, H.; Hayes, J.L.; Lee, K.; Balaj, L.; Passaro, C.; et al. Immune evasion mediated by PD-L1 on glioblastoma-derived extracellular vesicles. Sci. Adv. 2018, 4, eaar2766. [Google Scholar] [CrossRef]
- Czystowska-Kuzmicz, M.; Sosnowska, A.; Nowis, D.; Ramji, K.; Szajnik, M.; Chlebowska-Tuz, J.; Wolinska, E.; Gaj, P.; Grazul, M.; Pilch, Z.; et al. Small extracellular vesicles containing arginase-1 suppress T-cell responses and promote tumor growth in ovarian carcinoma. Nat. Commun. 2019, 10, 3000. [Google Scholar] [CrossRef]
- Haderk, F.; Schulz, R.; Iskar, M.; Cid, L.L.; Worst, T.; Willmund, K.V.; Schulz, A.; Warnken, U.; Seiler, J.; Benner, A.; et al. Tumor-derived exosomes modulate PD-L1 expression in monocytes. Sci. Immunol. 2017, 2, eaah5509. [Google Scholar] [CrossRef]
- Twomey, J.D.; Zhang, B. Cancer Immunotherapy Update: FDA-Approved Checkpoint Inhibitors and Companion Diagnostics. AAPS J. 2021, 23, 39. [Google Scholar] [CrossRef]
- Daassi, D.; Mahoney, K.M.; Freeman, G.J. The importance of exosomal PDL1 in tumour immune evasion. Nat. Rev. Immunol. 2020, 20, 209–215. [Google Scholar] [CrossRef]
- 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]
- Dou, X.; Hua, Y.; Chen, Z.; Chao, F.; Li, M. Extracellular vesicles containing PD-L1 contribute to CD8+ T-cell immune suppression and predict poor outcomes in small cell lung cancer. Clin. Exp. Immunol. 2022, 207, 307–317. [Google Scholar] [CrossRef] [PubMed]
- Gupta, P.; Kadamberi, I.P.; Mittal, S.; Tsaih, S.W.; George, J.; Kumar, S.; Vijayan, D.K.; Geethadevi, A.; Parashar, D.; Topchyan, P.; et al. Tumor Derived Extracellular Vesicles Drive T Cell Exhaustion in Tumor Microenvironment through Sphingosine Mediated Signaling and Impacting Immunotherapy Outcomes in Ovarian Cancer. Adv. Sci. 2022, 9, e2104452. [Google Scholar] [CrossRef] [PubMed]
- Xie, F.; Zhou, X.; Su, P.; Li, H.; Tu, Y.; Du, J.; Pan, C.; Wei, X.; Zheng, M.; Jin, K.; et al. Breast cancer cell-derived extracellular vesicles promote CD8(+) T cell exhaustion via TGF-beta type II receptor signaling. Nat. Commun. 2022, 13, 4461. [Google Scholar] [CrossRef]
- Vignard, V.; Labbe, M.; Marec, N.; Andre-Gregoire, G.; Jouand, N.; Fonteneau, J.F.; Labarriere, N.; Fradin, D. MicroRNAs in Tumor Exosomes Drive Immune Escape in Melanoma. Cancer Immunol. Res. 2020, 8, 255–267. [Google Scholar] [CrossRef] [PubMed]
- Peng, M.; Ren, J.; Jing, Y.; Jiang, X.; Xiao, Q.; Huang, J.; Tao, Y.; Lei, L.; Wang, X.; Yang, Z.; et al. Tumour-derived small extracellular vesicles suppress CD8+ T cell immune function by inhibiting SLC6A8-mediated creatine import in NPM1-mutated acute myeloid leukaemia. J. Extracell. Vesicles 2021, 10, e12168. [Google Scholar] [CrossRef]
- Wieckowski, E.U.; Visus, C.; Szajnik, M.; Szczepanski, M.J.; Storkus, W.J.; Whiteside, T.L. Tumor-derived microvesicles promote regulatory T cell expansion and induce apoptosis in tumor-reactive activated CD8+ T lymphocytes. J. Immunol. 2009, 183, 3720–3730. [Google Scholar] [CrossRef]
- Theodoraki, M.N.; Hoffmann, T.K.; Jackson, E.K.; Whiteside, T.L. Exosomes in HNSCC plasma as surrogate markers of tumour progression and immune competence. Clin. Exp. Immunol. 2018, 194, 67–78. [Google Scholar] [CrossRef]
- Yang, Y.; Li, C.W.; Chan, L.C.; Wei, Y.; Hsu, J.M.; Xia, W.; Cha, J.H.; Hou, J.; Hsu, J.L.; Sun, L.; et al. Exosomal PD-L1 harbors active defense function to suppress T cell killing of breast cancer cells and promote tumor growth. Cell Res. 2018, 28, 862–864. [Google Scholar] [CrossRef]
- Poggio, M.; Hu, T.; Pai, C.C.; Chu, B.; Belair, C.D.; Chang, A.; Montabana, E.; Lang, U.E.; Fu, Q.; Fong, L.; et al. Suppression of Exosomal PD-L1 Induces Systemic Anti-tumor Immunity and Memory. Cell 2019, 177, 414–427.e13. [Google Scholar] [CrossRef]
- Kim, D.H.; Kim, H.; Choi, Y.J.; Kim, S.Y.; Lee, J.E.; Sung, K.J.; Sung, Y.H.; Pack, C.G.; Jung, M.K.; Han, B.; et al. Exosomal PD-L1 promotes tumor growth through immune escape in non-small cell lung cancer. Exp. Mol. Med. 2019, 51, 1–13. [Google Scholar] [CrossRef] [PubMed]
- Cordonnier, M.; Nardin, C.; Chanteloup, G.; Derangere, V.; Algros, M.P.; Arnould, L.; Garrido, C.; Aubin, F.; Gobbo, J. Tracking the evolution of circulating exosomal-PD-L1 to monitor melanoma patients. J. Extracell. Vesicles 2020, 9, 1710899. [Google Scholar] [CrossRef]
- Zhang, M.; Fan, Y.; Che, X.; Hou, K.; Zhang, C.; Li, C.; Wen, T.; Wang, S.; Cheng, Y.; Liu, Y.; et al. 5-FU-Induced Upregulation of Exosomal PD-L1 Causes Immunosuppression in Advanced Gastric Cancer Patients. Front. Oncol. 2020, 10, 492. [Google Scholar] [CrossRef]
- Wang, X.; Shen, H.; Zhangyuan, G.; Huang, R.; Zhang, W.; He, Q.; Jin, K.; Zhuo, H.; Zhang, Z.; Wang, J.; et al. 14-3-3zeta delivered by hepatocellular carcinoma-derived exosomes impaired anti-tumor function of tumor-infiltrating T lymphocytes. Cell Death Dis. 2018, 9, 159. [Google Scholar] [CrossRef]
- Shenoy, G.N.; Loyall, J.; Berenson, C.S.; Kelleher, R.J., Jr.; Iyer, V.; Balu-Iyer, S.V.; Odunsi, K.; Bankert, R.B. Sialic Acid-Dependent Inhibition of T Cells by Exosomal Ganglioside GD3 in Ovarian Tumor Microenvironments. J. Immunol. 2018, 201, 3750–3758. [Google Scholar] [CrossRef] [PubMed]
- Shenoy, G.N.; Loyall, J.; Maguire, O.; Iyer, V.; Kelleher, R.J., Jr.; Minderman, H.; Wallace, P.K.; Odunsi, K.; Balu-Iyer, S.V.; Bankert, R.B. Exosomes Associated with Human Ovarian Tumors Harbor a Reversible Checkpoint of T-cell Responses. Cancer Immunol. Res. 2018, 6, 236–247. [Google Scholar] [CrossRef]
- Sharma, P.; Diergaarde, B.; Ferrone, S.; Kirkwood, J.M.; Whiteside, T.L. Melanoma cell-derived exosomes in plasma of melanoma patients suppress functions of immune effector cells. Sci. Rep. 2020, 10, 92. [Google Scholar] [CrossRef] [PubMed]
- Zhou, J.; Yang, Y.; Wang, W.; Zhang, Y.; Chen, Z.; Hao, C.; Zhang, J. Melanoma-released exosomes directly activate the mitochondrial apoptotic pathway of CD4(+) T cells through their microRNA cargo. Exp. Cell Res. 2018, 371, 364–371. [Google Scholar] [CrossRef]
- Kalvala, A.; Wallet, P.; Yang, L.; Wang, C.; Li, H.; Nam, A.; Nathan, A.; Mambetsariev, I.; Poroyko, V.; Gao, H.; et al. Phenotypic Switching of Naive T Cells to Immune-Suppressive Treg-Like Cells by Mutant KRAS. J. Clin. Med. 2019, 8, 1726. [Google Scholar] [CrossRef]
- Ni, C.; Fang, Q.Q.; Chen, W.Z.; Jiang, J.X.; Jiang, Z.; Ye, J.; Zhang, T.; Yang, L.; Meng, F.B.; Xia, W.J.; et al. Breast cancer-derived exosomes transmit lncRNA SNHG16 to induce CD73+gammadelta1 Treg cells. Signal Transduct. Target. Ther. 2020, 5, 41. [Google Scholar] [CrossRef] [PubMed]
- Clayton, A.; Al-Taei, S.; Webber, J.; Mason, M.D.; Tabi, Z. Cancer exosomes express CD39 and CD73, which suppress T cells through adenosine production. J. Immunol. 2011, 187, 676–683. [Google Scholar] [CrossRef] [PubMed]
- Ye, L.; Zhang, Q.; Cheng, Y.; Chen, X.; Wang, G.; Shi, M.; Zhang, T.; Cao, Y.; Pan, H.; Zhang, L.; et al. Tumor-derived exosomal HMGB1 fosters hepatocellular carcinoma immune evasion by promoting TIM-1(+) regulatory B cell expansion. J. Immunother. Cancer 2018, 6, 145. [Google Scholar] [CrossRef] [PubMed]
- Capello, M.; Vykoukal, J.V.; Katayama, H.; Bantis, L.E.; Wang, H.; Kundnani, D.L.; Aguilar-Bonavides, C.; Aguilar, M.; Tripathi, S.C.; Dhillon, D.S.; et al. Exosomes harbor B cell targets in pancreatic adenocarcinoma and exert decoy function against complement-mediated cytotoxicity. Nat. Commun. 2019, 10, 254. [Google Scholar] [CrossRef] [PubMed]
- Yin, Y.; Cai, X.; Chen, X.; Liang, H.; Zhang, Y.; Li, J.; Wang, Z.; Chen, X.; Zhang, W.; Yokoyama, S.; et al. Tumor-secreted miR-214 induces regulatory T cells: A major link between immune evasion and tumor growth. Cell Res. 2014, 24, 1164–1180. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Gao, Y.; Xu, H.; Li, N.; Wang, H.; Ma, L.; Chen, S.; Liu, J.; Zheng, Y.; Zhang, Y. Renal cancer-derived exosomes induce tumor immune tolerance by MDSCs-mediated antigen-specific immunosuppression. Cell Commun. Signal 2020, 18, 106. [Google Scholar] [CrossRef]
- Jiang, M.; Zhang, W.; Zhang, R.; Liu, P.; Ye, Y.; Yu, W.; Guo, X.; Yu, J. Cancer exosome-derived miR-9 and miR-181a promote the development of early-stage MDSCs via interfering with SOCS3 and PIAS3 respectively in breast cancer. Oncogene 2020, 39, 4681–4694. [Google Scholar] [CrossRef]
- Guo, X.; Qiu, W.; Liu, Q.; Qian, M.; Wang, S.; Zhang, Z.; Gao, X.; Chen, Z.; Xue, H.; Li, G. Immunosuppressive effects of hypoxia-induced glioma exosomes through myeloid-derived suppressor cells via the miR-10a/Rora and miR-21/Pten Pathways. Oncogene 2018, 37, 4239–4259. [Google Scholar] [CrossRef]
- Wang, X.; Luo, G.; Zhang, K.; Cao, J.; Huang, C.; Jiang, T.; Liu, B.; Su, L.; Qiu, Z. Hypoxic Tumor-Derived Exosomal miR-301a Mediates M2 Macrophage Polarization via PTEN/PI3Kgamma to Promote Pancreatic Cancer Metastasis. Cancer Res. 2018, 78, 4586–4598. [Google Scholar] [CrossRef]
- Zhao, J.; Schlosser, H.A.; Wang, Z.; Qin, J.; Li, J.; Popp, F.; Popp, M.C.; Alakus, H.; Chon, S.H.; Hansen, H.P.; et al. Tumor-Derived Extracellular Vesicles Inhibit Natural Killer Cell Function in Pancreatic Cancer. Cancers 2019, 11, 874. [Google Scholar] [CrossRef]
- Chang, W.A.; Tsai, M.J.; Hung, J.Y.; Wu, K.L.; Tsai, Y.M.; Huang, Y.C.; Chang, C.Y.; Tsai, P.H.; Hsu, Y.L. miR-150-5p-Containing Extracellular Vesicles Are a New Immunoregulator That Favor the Progression of Lung Cancer in Hypoxic Microenvironments by Altering the Phenotype of NK Cells. Cancers 2021, 13, 6252. [Google Scholar] [CrossRef] [PubMed]
- Ding, G.; Zhou, L.; Qian, Y.; Fu, M.; Chen, J.; 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] [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]
- Wang, M.; Cai, Y.; Peng, Y.; Xu, B.; Hui, W.; Jiang, Y. Exosomal LGALS9 in the cerebrospinal fluid of glioblastoma patients suppressed dendritic cell antigen presentation and cytotoxic T-cell immunity. Cell Death Dis. 2020, 11, 896. [Google Scholar] [CrossRef]
- De Paula Silva, E.; Marti, L.C.; Andreghetto, F.M.; de Sales, R.O.; Hoberman, M.; Dos Santos Dias, B.; Diniz, L.F.A.; Dos Santos, A.M.; Moyses, R.A.; Curioni, O.A.; et al. Extracellular vesicles cargo from head and neck cancer cell lines disrupt dendritic cells function and match plasma microRNAs. Sci. Rep. 2021, 11, 18534. [Google Scholar] [CrossRef] [PubMed]
- Salimu, J.; Webber, J.; Gurney, M.; Al-Taei, S.; Clayton, A.; Tabi, Z. Dominant immunosuppression of dendritic cell function by prostate-cancer-derived exosomes. J. Extracell. Vesicles 2017, 6, 1368823. [Google Scholar] [CrossRef]
- Ning, Y.; Shen, K.; Wu, Q.; Sun, X.; Bai, Y.; Xie, Y.; Pan, J.; Qi, C. Tumor exosomes block dendritic cells maturation to decrease the T cell immune response. Immunol. Lett. 2018, 199, 36–43. [Google Scholar] [CrossRef]
- Huber, V.; Vallacchi, V.; Fleming, V.; Hu, X.; Cova, A.; Dugo, M.; Shahaj, E.; Sulsenti, R.; Vergani, E.; Filipazzi, P.; et al. Tumor-derived microRNAs induce myeloid suppressor cells and predict immunotherapy resistance in melanoma. J. Clin. Investig. 2018, 128, 5505–5516. [Google Scholar] [CrossRef]
- Fleming, V.; Hu, X.; Weller, C.; Weber, R.; Groth, C.; Riester, Z.; Huser, L.; Sun, Q.; Nagibin, V.; Kirschning, C.; et al. Melanoma Extracellular Vesicles Generate Immunosuppressive Myeloid Cells by Upregulating PD-L1 via TLR4 Signaling. Cancer Res. 2019, 79, 4715–4728. [Google Scholar] [CrossRef]
- Xiang, X.; Poliakov, A.; Liu, C.; Liu, Y.; Deng, Z.B.; Wang, J.; Cheng, Z.; Shah, S.V.; Wang, G.J.; Zhang, L.; et al. Induction of myeloid-derived suppressor cells by tumor exosomes. Int. J. Cancer 2009, 124, 2621–2633. [Google Scholar] [CrossRef]
- Li, L.; Cao, B.; Liang, X.; Lu, S.; Luo, H.; Wang, Z.; Wang, S.; Jiang, J.; Lang, J.; Zhu, G. Microenvironmental oxygen pressure orchestrates an anti- and pro-tumoral gammadelta T cell equilibrium via tumor-derived exosomes. Oncogene 2019, 38, 2830–2843. [Google Scholar] [CrossRef] [PubMed]
- Hsu, Y.L.; Hung, J.Y.; Chang, W.A.; Jian, S.F.; Lin, Y.S.; Pan, Y.C.; Wu, C.Y.; Kuo, P.L. Hypoxic Lung-Cancer-Derived Extracellular Vesicle MicroRNA-103a Increases the Oncogenic Effects of Macrophages by Targeting PTEN. Mol. Ther. 2018, 26, 568–581. [Google Scholar] [CrossRef] [PubMed]
- Gao, L.; Wang, L.; Dai, T.; Jin, K.; Zhang, Z.; Wang, S.; Xie, F.; Fang, P.; Yang, B.; Huang, H.; et al. Tumor-derived exosomes antagonize innate antiviral immunity. Nat. Immunol. 2018, 19, 233–245. [Google Scholar] [CrossRef] [PubMed]
- Park, J.E.; Dutta, B.; Tse, S.W.; Gupta, N.; Tan, C.F.; Low, J.K.; Yeoh, K.W.; Kon, O.L.; Tam, J.P.; Sze, S.K. Hypoxia-induced tumor exosomes promote M2-like macrophage polarization of infiltrating myeloid cells and microRNA-mediated metabolic shift. Oncogene 2019, 38, 5158–5173. [Google Scholar] [CrossRef]
- Cooks, T.; Pateras, I.S.; Jenkins, L.M.; Patel, K.M.; Robles, A.I.; Morris, J.; Forshew, T.; Appella, E.; Gorgoulis, V.G.; Harris, C.C. Mutant p53 cancers reprogram macrophages to tumor supporting macrophages via exosomal miR-1246. Nat. Commun. 2018, 9, 771. [Google Scholar] [CrossRef]
- Gerloff, D.; Lutzkendorf, J.; Moritz, R.K.C.; Wersig, T.; Mader, K.; Muller, L.P.; Sunderkotter, C. Melanoma-Derived Exosomal miR-125b-5p Educates Tumor Associated Macrophages (TAMs) by Targeting Lysosomal Acid Lipase A (LIPA). Cancers 2020, 12, 464. [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]
- Dai, E.; Han, L.; Liu, J.; Xie, Y.; Kroemer, G.; Klionsky, D.J.; Zeh, H.J.; Kang, R.; Wang, J.; Tang, D. Autophagy-dependent ferroptosis drives tumor-associated macrophage polarization via release and uptake of oncogenic KRAS protein. Autophagy 2020, 16, 2069–2083. [Google Scholar] [CrossRef]
- Wang, S.; Gao, Y. Pancreatic cancer cell-derived microRNA-155-5p-containing extracellular vesicles promote immune evasion by triggering EHF-dependent activation of Akt/NF-kappaB signaling pathway. Int. Immunopharmacol. 2021, 100, 107990. [Google Scholar] [CrossRef]
- Wolf-Dennen, K.; Gordon, N.; Kleinerman, E.S. Exosomal communication by metastatic osteosarcoma cells modulates alveolar macrophages to an M2 tumor-promoting phenotype and inhibits tumoricidal functions. Oncoimmunology 2020, 9, 1747677. [Google Scholar] [CrossRef]
- Liu, Y.; Lu, M.; Chen, J.; Li, S.; Deng, Y.; Yang, S.; Ou, Q.; Li, J.; Gao, P.; Luo, Z.; et al. Extracellular vesicles derived from lung cancer cells exposed to intermittent hypoxia upregulate programmed death ligand 1 expression in macrophages. Sleep Breath. 2022, 26, 893–906. [Google Scholar] [CrossRef] [PubMed]
- Wang, D.; Wang, X.; Si, M.; Yang, J.; Sun, S.; Wu, H.; Cui, S.; Qu, X.; Yu, X. Exosome-encapsulated miRNAs contribute to CXCL12/CXCR4-induced liver metastasis of colorectal cancer by enhancing M2 polarization of macrophages. Cancer Lett. 2020, 474, 36–52. [Google Scholar] [CrossRef] [PubMed]
- Popena, I.; Abols, A.; Saulite, L.; Pleiko, K.; Zandberga, E.; Jekabsons, K.; Endzelins, E.; Llorente, A.; Line, A.; Riekstina, U. Effect of colorectal cancer-derived extracellular vesicles on the immunophenotype and cytokine secretion profile of monocytes and macrophages. Cell Commun. Signal 2018, 16, 17. [Google Scholar] [CrossRef]
- Ham, S.; Lima, L.G.; Chai, E.P.Z.; Muller, A.; Lobb, R.J.; Krumeich, S.; Wen, S.W.; Wiegmans, A.P.; Moller, A. Breast Cancer-Derived Exosomes Alter Macrophage Polarization via gp130/STAT3 Signaling. Front. Immunol. 2018, 9, 871. [Google Scholar] [CrossRef] [PubMed]
- He, C.; Hua, W.; Liu, J.; Fan, L.; Wang, H.; Sun, G. Exosomes derived from endoplasmic reticulum-stressed liver cancer cells enhance the expression of cytokines in macrophages via the STAT3 signaling pathway. Oncol. Lett. 2020, 20, 589–600. [Google Scholar] [CrossRef] [PubMed]
- Yao, X.; Tu, Y.; Xu, Y.; Guo, Y.; Yao, F.; Zhang, X. Endoplasmic reticulum stress-induced exosomal miR-27a-3p promotes immune escape in breast cancer via regulating PD-L1 expression in macrophages. J. Cell Mol. Med. 2020, 24, 9560–9573. [Google Scholar] [CrossRef] [PubMed]
- Chow, A.; Zhou, W.; Liu, L.; Fong, M.Y.; Champer, J.; Van Haute, D.; Chin, A.R.; Ren, X.; Gugiu, B.G.; Meng, Z.; et al. Macrophage immunomodulation by breast cancer-derived exosomes requires Toll-like receptor 2-mediated activation of NF-kappaB. Sci. Rep. 2014, 4, 5750. [Google Scholar] [CrossRef] [PubMed]
- Wu, L.; Zhang, X.; Zhang, B.; Shi, H.; Yuan, X.; Sun, Y.; Pan, Z.; Qian, H.; Xu, W. Exosomes derived from gastric cancer cells activate NF-kappaB pathway in macrophages to promote cancer progression. Tumour. Biol. 2016, 37, 12169–12180. [Google Scholar] [CrossRef]
- Fabbri, M.; Paone, A.; Calore, F.; Galli, R.; Gaudio, E.; Santhanam, R.; Lovat, F.; Fadda, P.; Mao, C.; Nuovo, G.J.; et al. MicroRNAs bind to Toll-like receptors to induce prometastatic inflammatory response. Proc. Natl. Acad. Sci. USA 2012, 109, E2110–E2116. [Google Scholar] [CrossRef]
- Chen, X.; Zhou, J.; Li, X.; Wang, X.; Lin, Y.; Wang, X. Exosomes derived from hypoxic epithelial ovarian cancer cells deliver microRNAs to macrophages and elicit a tumor-promoted phenotype. Cancer Lett. 2018, 435, 80–91. [Google Scholar] [CrossRef]
- Javeed, N.; Gustafson, M.P.; Dutta, S.K.; Lin, Y.; Bamlet, W.R.; Oberg, A.L.; Petersen, G.M.; Chari, S.T.; Dietz, A.B.; Mukhopadhyay, D. Immunosuppressive CD14(+)HLA-DR(lo/neg) monocytes are elevated in pancreatic cancer and "primed" by tumor-derived exosomes. Oncoimmunology 2017, 6, e1252013. [Google Scholar] [CrossRef] [PubMed]
- Hsieh, C.H.; Tai, S.K.; Yang, M.H. Snail-overexpressing Cancer Cells Promote M2-Like Polarization of Tumor-Associated Macrophages by Delivering MiR-21-Abundant Exosomes. Neoplasia 2018, 20, 775–788. [Google Scholar] [CrossRef] [PubMed]
- Gabrusiewicz, K.; Li, X.; Wei, J.; Hashimoto, Y.; Marisetty, A.L.; Ott, M.; Wang, F.; Hawke, D.; Yu, J.; Healy, L.M.; et al. Glioblastoma stem cell-derived exosomes induce M2 macrophages and PD-L1 expression on human monocytes. Oncoimmunology 2018, 7, e1412909. [Google Scholar] [CrossRef] [PubMed]
- Wang, F.; Li, B.; Wei, Y.; Zhao, Y.; Wang, L.; Zhang, P.; Yang, J.; He, W.; Chen, H.; Jiao, Z.; et al. Tumor-derived exosomes induce PD1(+) macrophage population in human gastric cancer that promotes disease progression. Oncogenesis 2018, 7, 41. [Google Scholar] [CrossRef] [PubMed]
- Hoelzinger, D.B.; Quinton, S.J.; Walters, D.K.; Vardam-Kaur, T.; Tschumper, R.C.; Borges da Silva, H.; Jelinek, D.F. Extracellular vesicle proteomic analysis leads to the discovery of HDGF as a new factor in multiple myeloma biology. Blood Adv. 2022, 6, 3458–3471. [Google Scholar] [CrossRef] [PubMed]
- Challagundla, K.B.; Wise, P.M.; Neviani, P.; Chava, H.; Murtadha, M.; Xu, T.; Kennedy, R.; Ivan, C.; Zhang, X.; Vannini, I.; et al. Exosome-mediated transfer of microRNAs within the tumor microenvironment and neuroblastoma resistance to chemotherapy. J. Natl. Cancer Inst. 2015, 107, djv135. [Google Scholar] [CrossRef]
- Mpakali, A.; Stratikos, E. The Role of Antigen Processing and Presentation in Cancer and the Efficacy of Immune Checkpoint Inhibitor Immunotherapy. Cancers 2021, 13, 134. [Google Scholar] [CrossRef]
- Imbert, P.R.C.; Saric, A.; Pedram, K.; Bertozzi, C.R.; Grinstein, S.; Freeman, S.A. An Acquired and Endogenous Glycocalyx Forms a Bidirectional “Don’t Eat” and “Don’t Eat Me” Barrier to Phagocytosis. Curr. Biol. 2021, 31, 77–89.e5. [Google Scholar] [CrossRef]
- Wang, J.; Zeng, H.; Zhang, H.; Han, Y. The role of exosomal PD-L1 in tumor immunotherapy. Transl. Oncol. 2021, 14, 101047. [Google Scholar] [CrossRef]
- Theodoraki, M.N.; Yerneni, S.; Gooding, W.E.; Ohr, J.; Clump, D.A.; Bauman, J.E.; Ferris, R.L.; Whiteside, T.L. Circulating exosomes measure responses to therapy in head and neck cancer patients treated with cetuximab, ipilimumab, and IMRT. Oncoimmunology 2019, 8, 1593805. [Google Scholar] [CrossRef]
- Kato, T.; Fahrmann, J.F.; Hanash, S.M.; Vykoukal, J. Extracellular Vesicles Mediate B Cell Immune Response and Are a Potential Target for Cancer Therapy. Cells 2020, 9, 1518. [Google Scholar] [CrossRef] [PubMed]
- Aung, T.; Chapuy, B.; Vogel, D.; Wenzel, D.; Oppermann, M.; Lahmann, M.; Weinhage, T.; Menck, K.; Hupfeld, T.; Koch, R.; et al. Exosomal evasion of humoral immunotherapy in aggressive B-cell lymphoma modulated by ATP-binding cassette transporter A3. Proc. Natl. Acad. Sci. USA 2011, 108, 15336–15341. [Google Scholar] [CrossRef] [PubMed]
- Ciravolo, V.; Huber, V.; Ghedini, G.C.; Venturelli, E.; Bianchi, F.; Campiglio, M.; Morelli, D.; Villa, A.; Della Mina, P.; Menard, S.; et al. Potential role of HER2-overexpressing exosomes in countering trastuzumab-based therapy. J. Cell Physiol. 2012, 227, 658–667. [Google Scholar] [CrossRef] [PubMed]
- Togashi, Y.; Shitara, K.; Nishikawa, H. Regulatory T cells in cancer immunosuppression—Implications for anticancer therapy. Nat. Rev. Clin. Oncol. 2019, 16, 356–371. [Google Scholar] [CrossRef]
- De Simone, M.; Arrigoni, A.; Rossetti, G.; Gruarin, P.; Ranzani, V.; Politano, C.; Bonnal, R.J.P.; Provasi, E.; Sarnicola, M.L.; Panzeri, I.; et al. Transcriptional Landscape of Human Tissue Lymphocytes Unveils Uniqueness of Tumor-Infiltrating T Regulatory Cells. Immunity 2016, 45, 1135–1147. [Google Scholar] [CrossRef]
- Saito, T.; Nishikawa, H.; Wada, H.; Nagano, Y.; Sugiyama, D.; Atarashi, K.; Maeda, Y.; Hamaguchi, M.; Ohkura, N.; Sato, E.; et al. Two FOXP3(+)CD4(+) T cell subpopulations distinctly control the prognosis of colorectal cancers. Nat. Med. 2016, 22, 679–684. [Google Scholar] [CrossRef]
- Kidani, Y.; Nogami, W.; Yasumizu, Y.; Kawashima, A.; Tanaka, A.; Sonoda, Y.; Tona, Y.; Nashiki, K.; Matsumoto, R.; Hagiwara, M.; et al. CCR8-targeted specific depletion of clonally expanded Treg cells in tumor tissues evokes potent tumor immunity with long-lasting memory. Proc. Natl. Acad. Sci. USA 2022, 119, e2114282119. [Google Scholar] [CrossRef]
- Zhang, A.; Ren, Z.; Tseng, K.F.; Liu, X.; Li, H.; Lu, C.; Cai, Y.; Minna, J.D.; Fu, Y.X. Dual targeting of CTLA-4 and CD47 on Treg cells promotes immunity against solid tumors. Sci. Transl. Med. 2021, 13, eabg8693. [Google Scholar] [CrossRef]
- Klages, K.; Mayer, C.T.; Lahl, K.; Loddenkemper, C.; Teng, M.W.; Ngiow, S.F.; Smyth, M.J.; Hamann, A.; Huehn, J.; Sparwasser, T. Selective depletion of Foxp3+ regulatory T cells improves effective therapeutic vaccination against established melanoma. Cancer Res. 2010, 70, 7788–7799. [Google Scholar] [CrossRef]
- Shitara, K.; Nishikawa, H. Regulatory T cells: A potential target in cancer immunotherapy. Ann. N. Y. Acad. Sci. 2018, 1417, 104–115. [Google Scholar] [CrossRef]
- Watanabe, T. Realization of Osteolysis, Angiogenesis, Immunosuppression, and Drug Resistance by Extracellular Vesicles: Roles of RNAs and Proteins in Their Cargoes and of Ectonucleotidases of the Immunosuppressive Adenosinergic Noncanonical Pathway in the Bone Marrow Niche of Multiple Myeloma. Cancers 2021, 13, 2969. [Google Scholar] [CrossRef] [PubMed]
- Zebrowska, A.; Widlak, P.; Whiteside, T.; Pietrowska, M. Signaling of Tumor-Derived sEV Impacts Melanoma Progression. Int. J. Mol. Sci. 2020, 21, 5066. [Google Scholar] [CrossRef] [PubMed]
- Clayton, A.; Mitchell, J.P.; Court, J.; Mason, M.D.; Tabi, Z. Human tumor-derived exosomes selectively impair lymphocyte responses to interleukin-2. Cancer Res. 2007, 67, 7458–7466. [Google Scholar] [CrossRef] [PubMed]
- Ning, T.; Li, J.; He, Y.; Zhang, H.; Wang, X.; Deng, T.; Liu, R.; Li, H.; Bai, M.; Fan, Q.; et al. Exosomal miR-208b related with oxaliplatin resistance promotes Treg expansion in colorectal cancer. Mol. Ther. 2021, 29, 2723–2736. [Google Scholar] [CrossRef]
- 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]
- 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]
- Ye, S.B.; Zhang, H.; Cai, T.T.; Liu, Y.N.; Ni, J.J.; He, J.; Peng, J.Y.; Chen, Q.Y.; Mo, H.Y.; Jun, C.; et al. Exosomal miR-24-3p impedes T-cell function by targeting FGF11 and serves as a potential prognostic biomarker for nasopharyngeal carcinoma. J. Pathol. 2016, 240, 329–340. [Google Scholar] [CrossRef]
- Szczepanski, M.J.; Szajnik, M.; Czystowska, M.; Mandapathil, M.; Strauss, L.; Welsh, A.; Foon, K.A.; Whiteside, T.L.; Boyiadzis, M. Increased frequency and suppression by regulatory T cells in patients with acute myelogenous leukemia. Clin. Cancer Res. 2009, 15, 3325–3332. [Google Scholar] [CrossRef]
- Swatler, J.; Turos-Korgul, L.; Brewinska-Olchowik, M.; De Biasi, S.; Dudka, W.; Le, B.V.; Kominek, A.; Cyranowski, S.; Pilanc, P.; Mohammadi, E.; et al. 4-1BBL-containing leukemic extracellular vesicles promote immunosuppressive effector regulatory T cells. Blood Adv. 2022, 6, 1879–1894. [Google Scholar] [CrossRef]
- Carlsten, M.; Jaras, M. Natural Killer Cells in Myeloid Malignancies: Immune Surveillance, NK Cell Dysfunction, and Pharmacological Opportunities to Bolster the Endogenous NK Cells. Front. Immunol. 2019, 10, 2357. [Google Scholar] [CrossRef]
- Myers, J.A.; Miller, J.S. Exploring the NK cell platform for cancer immunotherapy. Nat. Rev. Clin. Oncol. 2021, 18, 85–100. [Google Scholar] [CrossRef] [PubMed]
- Rea, D.; Henry, G.; Khaznadar, Z.; Etienne, G.; Guilhot, F.; Nicolini, F.; Guilhot, J.; Rousselot, P.; Huguet, F.; Legros, L.; et al. Natural killer-cell counts are associated with molecular relapse-free survival after imatinib discontinuation in chronic myeloid leukemia: The IMMUNOSTIM study. Haematologica 2017, 102, 1368–1377. [Google Scholar] [CrossRef] [PubMed]
- Hilpert, J.; Grosse-Hovest, L.; Grunebach, F.; Buechele, C.; Nuebling, T.; Raum, T.; Steinle, A.; Salih, H.R. Comprehensive analysis of NKG2D ligand expression and release in leukemia: Implications for NKG2D-mediated NK cell responses. J. Immunol. 2012, 189, 1360–1371. [Google Scholar] [CrossRef] [PubMed]
- Boissel, N.; Rea, D.; Tieng, V.; Dulphy, N.; Brun, M.; Cayuela, J.M.; Rousselot, P.; Tamouza, R.; Le Bouteiller, P.; Mahon, F.X.; et al. BCR/ABL oncogene directly controls MHC class I chain-related molecule A expression in chronic myelogenous leukemia. J. Immunol. 2006, 176, 5108–5116. [Google Scholar] [CrossRef] [PubMed]
- Clayton, A.; Mitchell, J.P.; Court, J.; Linnane, S.; Mason, M.D.; Tabi, Z. Human tumor-derived exosomes down-modulate NKG2D expression. J. Immunol. 2008, 180, 7249–7258. [Google Scholar] [CrossRef]
- Ashiru, O.; Boutet, P.; Fernandez-Messina, L.; Aguera-Gonzalez, S.; Skepper, J.N.; Vales-Gomez, M.; Reyburn, H.T. Natural killer cell cytotoxicity is suppressed by exposure to the human NKG2D ligand MICA*008 that is shed by tumor cells in exosomes. Cancer Res. 2010, 70, 481–489. [Google Scholar] [CrossRef]
- Szczepanski, M.J.; Szajnik, M.; Welsh, A.; Whiteside, T.L.; Boyiadzis, M. Blast-derived microvesicles in sera from patients with acute myeloid leukemia suppress natural killer cell function via membrane-associated transforming growth factor-beta1. Haematologica 2011, 96, 1302–1309. [Google Scholar] [CrossRef]
- Hong, C.S.; Sharma, P.; Yerneni, S.S.; Simms, P.; Jackson, E.K.; Whiteside, T.L.; Boyiadzis, M. Circulating exosomes carrying an immunosuppressive cargo interfere with cellular immunotherapy in acute myeloid leukemia. Sci. Rep. 2017, 7, 14684. [Google Scholar] [CrossRef]
- Hong, C.S.; Muller, L.; Whiteside, T.L.; Boyiadzis, M. Plasma exosomes as markers of therapeutic response in patients with acute myeloid leukemia. Front. Immunol. 2014, 5, 160. [Google Scholar] [CrossRef]
- Hong, C.S.; Funk, S.; Muller, L.; Boyiadzis, M.; Whiteside, T.L. Isolation of biologically active and morphologically intact exosomes from plasma of patients with cancer. J. Extracell. Vesicles 2016, 5, 29289. [Google Scholar] [CrossRef]
- Hong, C.S.; Jeong, E.; Boyiadzis, M.; Whiteside, T.L. Increased small extracellular vesicle secretion after chemotherapy via upregulation of cholesterol metabolism in acute myeloid leukaemia. J. Extracell. Vesicles 2020, 9, 1800979. [Google Scholar] [CrossRef] [PubMed]
- Binder, H.M.; Maeding, N.; Wolf, M.; Cronemberger Andrade, A.; Vari, B.; Krisch, L.; Gomes, F.G.; Blochl, C.; Muigg, K.; Poupardin, R.; et al. Scalable Enrichment of Immunomodulatory Human Acute Myeloid Leukemia Cell Line-Derived Extracellular Vesicles. Cells 2021, 10, 3321. [Google Scholar] [CrossRef] [PubMed]
- Berchem, G.; Noman, M.Z.; Bosseler, M.; Paggetti, J.; Baconnais, S.; Le Cam, E.; Nanbakhsh, A.; Moussay, E.; Mami-Chouaib, F.; Janji, B.; et al. Hypoxic tumor-derived microvesicles negatively regulate NK cell function by a mechanism involving TGF-beta and miR23a transfer. Oncoimmunology 2016, 5, e1062968. [Google Scholar] [CrossRef] [PubMed]
- Graner, M.W.; Schnell, S.; Olin, M.R. Tumor-derived exosomes, microRNAs, and cancer immune suppression. Semin. Immunopathol. 2018, 40, 505–515. [Google Scholar] [CrossRef]
- Zhang, P.F.; Gao, C.; Huang, X.Y.; Lu, J.C.; Guo, X.J.; Shi, G.M.; Cai, J.B.; Ke, A.W. Cancer cell-derived exosomal circUHRF1 induces natural killer cell exhaustion and may cause resistance to anti-PD1 therapy in hepatocellular carcinoma. Mol. Cancer 2020, 19, 110. [Google Scholar] [CrossRef]
- Yang, Y.; Li, C.; Liu, T.; Dai, X.; Bazhin, A.V. Myeloid-Derived Suppressor Cells in Tumors: From Mechanisms to Antigen Specificity and Microenvironmental Regulation. Front. Immunol. 2020, 11, 1371. [Google Scholar] [CrossRef]
- Rodriguez, P.C.; Ochoa, A.C. Arginine regulation by myeloid derived suppressor cells and tolerance in cancer: Mechanisms and therapeutic perspectives. Immunol. Rev. 2008, 222, 180–191. [Google Scholar] [CrossRef]
- Tian, X.; Shen, H.; Li, Z.; Wang, T.; Wang, S. Tumor-derived exosomes, myeloid-derived suppressor cells, and tumor microenvironment. J. Hematol. Oncol. 2019, 12, 84. [Google Scholar] [CrossRef]
- Tucci, M.; Passarelli, A.; Mannavola, F.; Felici, C.; Stucci, L.S.; Cives, M.; Silvestris, F. Immune System Evasion as Hallmark of Melanoma Progression: The Role of Dendritic Cells. Front. Oncol. 2019, 9, 1148. [Google Scholar] [CrossRef]
- Wculek, S.K.; Cueto, F.J.; Mujal, A.M.; Melero, I.; Krummel, M.F.; Sancho, D. Dendritic cells in cancer immunology and immunotherapy. Nat. Rev. Immunol. 2020, 20, 7–24. [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]
- Kumari, N.; Choi, S.H. Tumor-associated macrophages in cancer: Recent advancements in cancer nanoimmunotherapies. J. Exp. Clin. Cancer Res. 2022, 41, 68. [Google Scholar] [CrossRef] [PubMed]
- Christofides, A.; Strauss, L.; Yeo, A.; Cao, C.; Charest, A.; Boussiotis, V.A. The complex role of tumor-infiltrating macrophages. Nat. Immunol. 2022, 23, 1148–1156. [Google Scholar] [CrossRef] [PubMed]
- Cai, Z.; Yang, F.; Yu, L.; Yu, Z.; Jiang, L.; Wang, Q.; Yang, Y.; Wang, L.; Cao, X.; Wang, J. Activated T cell exosomes promote tumor invasion via Fas signaling pathway. J. Immunol. 2012, 188, 5954–5961. [Google Scholar] [CrossRef] [PubMed]
- Beccard, I.J.; Hofmann, L.; Schroeder, J.C.; Ludwig, S.; Laban, S.; Brunner, C.; Lotfi, R.; Hoffmann, T.K.; Jackson, E.K.; Schuler, P.J.; et al. Immune Suppressive Effects of Plasma-Derived Exosome Populations in Head and Neck Cancer. Cancers 2020, 12, 1997. [Google Scholar] [CrossRef] [PubMed]
- Chiou, N.T.; Kageyama, R.; Ansel, K.M. Selective Export into Extracellular Vesicles and Function of tRNA Fragments during T Cell Activation. Cell Rep. 2018, 25, 3356–3370.e4. [Google Scholar] [CrossRef]
- Neviani, P.; Wise, P.M.; Murtadha, M.; Liu, C.W.; Wu, C.H.; Jong, A.Y.; Seeger, R.C.; Fabbri, M. Natural Killer-Derived Exosomal miR-186 Inhibits Neuroblastoma Growth and Immune Escape Mechanisms. Cancer Res. 2019, 79, 1151–1164. [Google Scholar] [CrossRef]
- Fenselau, C.; Ostrand-Rosenberg, S. Molecular cargo in myeloid-derived suppressor cells and their exosomes. Cell Immunol. 2021, 359, 104258. [Google Scholar] [CrossRef]
- Rashid, M.H.; Borin, T.F.; Ara, R.; Piranlioglu, R.; Achyut, B.R.; Korkaya, H.; Liu, Y.; Arbab, A.S. Critical immunosuppressive effect of MDSCderived exosomes in the tumor microenvironment. Oncol. Rep. 2021, 45, 1171–1181. [Google Scholar] [CrossRef]
- Lan, J.; Sun, L.; Xu, F.; Liu, L.; Hu, F.; Song, D.; Hou, Z.; Wu, W.; Luo, X.; Wang, J.; et al. M2 Macrophage-Derived Exosomes Promote Cell Migration and Invasion in Colon Cancer. Cancer Res. 2019, 79, 146–158. [Google Scholar] [CrossRef]
- Wang, X.; Shen, H.; He, Q.; Tian, W.; Xia, A.; Lu, X.J. Exosomes derived from exhausted CD8+ T cells impaired the anticancer function of normal CD8+ T cells. J. Med. Genet. 2019, 56, 29–31. [Google Scholar] [CrossRef] [PubMed]
- Pu, J.; Xu, Z.; Nian, J.; Fang, Q.; Yang, M.; Huang, Y.; Li, W.; Ge, B.; Wang, J.; Wei, H. M2 macrophage-derived extracellular vesicles facilitate CD8+T cell exhaustion in hepatocellular carcinoma via the miR-21-5p/YOD1/YAP/beta-catenin pathway. Cell Death Discov. 2021, 7, 182. [Google Scholar] [CrossRef] [PubMed]
- Mi, X.; Xu, R.; Hong, S.; Xu, T.; Zhang, W.; Liu, M. M2 Macrophage-Derived Exosomal lncRNA AFAP1-AS1 and MicroRNA-26a Affect Cell Migration and Metastasis in Esophageal Cancer. Mol. Ther. Nucleic Acids 2020, 22, 779–790. [Google Scholar] [CrossRef]
- Hu, W.; Liu, C.; Bi, Z.-Y.; Zhou, Q.; Zhang, H.; Li, L.-L.; Zhang, J.; Zhu, W.; Song, Y.-Y.Y.; Zhang, F.; et al. Comprehensive landscape of extracellular vesicle-derived RNAs in cancer initiation, progression, metastasis and cancer immunology. Mol. Cancer 2020, 19, 102. [Google Scholar] [CrossRef]
- Yang, P.; Peng, Y.; Feng, Y.; Xu, Z.; Feng, P.; Cao, J.; Chen, Y.; Chen, X.; Cao, X.; Yang, Y.; et al. Immune Cell-Derived Extracellular Vesicles—New Strategies in Cancer Immunotherapy. Front. Immunol. 2021, 12, 771551. [Google Scholar] [CrossRef] [PubMed]
- Andrews, D.W.; Judy, K.D.; Scott, C.B.; Garcia, S.; Harshyne, L.A.; Kenyon, L.; Talekar, K.; Flanders, A.; Atsina, K.B.; Kim, L.; et al. Phase Ib Clinical Trial of IGV-001 for Patients with Newly Diagnosed Glioblastoma. Clin. Cancer Res. 2021, 27, 1912–1922. [Google Scholar] [CrossRef] [PubMed]
- Dionisi, M.; De Archangelis, C.; Battisti, F.; Rahimi Koshkaki, H.; Belleudi, F.; Zizzari, I.G.; Ruscito, I.; Albano, C.; Di Filippo, A.; Torrisi, M.R.; et al. Tumor-Derived Microvesicles Enhance Cross-Processing Ability of Clinical Grade Dendritic Cells. Front. Immunol. 2018, 9, 2481. [Google Scholar] [CrossRef]
- Ma, J.; Wei, K.; Zhang, H.; Tang, K.; Li, F.; Zhang, T.; Liu, J.; Xu, P.; Yu, Y.; Sun, W.; et al. Mechanisms by Which Dendritic Cells Present Tumor Microparticle Antigens to CD8(+) T Cells. Cancer Immunol. Res. 2018, 6, 1057–1068. [Google Scholar] [CrossRef]
- Dassler-Plenker, J.; Reiners, K.S.; van den Boorn, J.G.; Hansen, H.P.; Putschli, B.; Barnert, S.; Schuberth-Wagner, C.; Schubert, R.; Tuting, T.; Hallek, M.; et al. RIG-I activation induces the release of extracellular vesicles with antitumor activity. Oncoimmunology 2016, 5, e1219827. [Google Scholar] [CrossRef]
- 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]
- Ortiz-Bonilla, C.J.; Uccello, T.P.; Gerber, S.A.; Lord, E.M.; Messing, E.M.; Lee, Y.F. Bladder Cancer Extracellular Vesicles Elicit a CD8 T Cell-Mediated Antitumor Immunity. Int. J. Mol. Sci. 2022, 23, 2904. [Google Scholar] [CrossRef]
- Montfort, A.; Bertrand, F.; Rochotte, J.; Gilhodes, J.; Filleron, T.; Milhes, J.; Dufau, C.; Imbert, C.; Riond, J.; Tosolini, M.; et al. Neutral Sphingomyelinase 2 Heightens Anti-Melanoma Immune Responses and Anti-PD-1 Therapy Efficacy. Cancer Immunol. Res. 2021, 9, 568–582. [Google Scholar] [CrossRef] [PubMed]
- Chen, Z.; You, L.; Wang, L.; Huang, X.; Liu, H.; Wei, J.Y.; Zhu, L.; Qian, W. Dual effect of DLBCL-derived EXOs in lymphoma to improve DC vaccine efficacy in vitro while favor tumorgenesis in vivo. J. Exp. Clin. Cancer Res. 2018, 37, 190. [Google Scholar] [CrossRef]
- Scholl, J.N.; de Fraga Dias, A.; Pizzato, P.R.; Lopes, D.V.; Moritz, C.E.J.; Jandrey, E.H.F.; Souto, G.D.; Colombo, M.; Rohden, F.; Sevigny, J.; et al. Characterization and antiproliferative activity of glioma-derived extracellular vesicles. Nanomedicine 2020, 15, 1001–1018. [Google Scholar] [CrossRef] [PubMed]
- Seo, N.; Shirakura, Y.; Tahara, Y.; Momose, F.; Harada, N.; Ikeda, H.; Akiyoshi, K.; Shiku, H. Activated CD8(+) T cell extracellular vesicles prevent tumour progression by targeting of lesional mesenchymal cells. Nat. Commun. 2018, 9, 435. [Google Scholar] [CrossRef] [PubMed]
- Lu, Z.; Zuo, B.; Jing, R.; Gao, X.; Rao, Q.; Liu, Z.; Qi, H.; Guo, H.; Yin, H. Dendritic cell-derived exosomes elicit tumor regression in autochthonous hepatocellular carcinoma mouse models. J. Hepatol. 2017, 67, 739–748. [Google Scholar] [CrossRef] [PubMed]
- Enomoto, Y.; Li, P.; Jenkins, L.M.; Anastasakis, D.; Lyons, G.C.; Hafner, M.; Leonard, W.J. Cytokine-enhanced cytolytic activity of exosomes from NK Cells. Cancer Gene Ther. 2022, 29, 734–749. [Google Scholar] [CrossRef]
- Shoae-Hassani, A.; Hamidieh, A.A.; Behfar, M.; Mohseni, R.; Mortazavi-Tabatabaei, S.A.; Asgharzadeh, S. NK Cell-derived Exosomes From NK Cells Previously Exposed to Neuroblastoma Cells Augment the Antitumor Activity of Cytokine-activated NK Cells. J. Immunother. 2017, 40, 265–276. [Google Scholar] [CrossRef]
- Zhou, W.J.; Zhang, J.; Xie, F.; Wu, J.N.; Ye, J.F.; Wang, J.; Wu, K.; Li, M.Q. CD45RO(-)CD8(+) T cell-derived exosomes restrict estrogen-driven endometrial cancer development via the ERbeta/miR-765/PLP2/Notch axis. Theranostics 2021, 11, 5330–5345. [Google Scholar] [CrossRef]
- DeNardo, D.G.; Ruffell, B. Macrophages as regulators of tumour immunity and immunotherapy. Nat. Rev. Immunol. 2019, 19, 369–382. [Google Scholar] [CrossRef]
- Cianciaruso, C.; Beltraminelli, T.; Duval, F.; Nassiri, S.; Hamelin, R.; Mozes, A.; Gallart-Ayala, H.; Ceada Torres, G.; Torchia, B.; Ries, C.H.; et al. Molecular Profiling and Functional Analysis of Macrophage-Derived Tumor Extracellular Vesicles. Cell Rep. 2019, 27, 3062–3080.e11. [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]
- Hadla, M.; Palazzolo, S.; Corona, G.; Caligiuri, I.; Canzonieri, V.; Toffoli, G.; Rizzolio, F. Exosomes increase the therapeutic index of doxorubicin in breast and ovarian cancer mouse models. Nanomedicine 2016, 11, 2431–2441. [Google Scholar] [CrossRef] [PubMed]
- Zhang, D.X.; Vu, L.T.; Ismail, N.N.; Le, M.T.N.; Grimson, A. Landscape of extracellular vesicles in the tumour microenvironment: Interactions with stromal cells and with non-cell components, and impacts on metabolic reprogramming, horizontal transfer of neoplastic traits, and the emergence of therapeutic resistance. Semin. Cancer Biol. 2021, 74, 24–44. [Google Scholar] [CrossRef] [PubMed]
- Khani, A.T.; Sharifzad, F.; Mardpour, S.; Hassan, Z.M.; Ebrahimi, M. Tumor extracellular vesicles loaded with exogenous Let-7i and miR-142 can modulate both immune response and tumor microenvironment to initiate a powerful anti-tumor response. Cancer Lett. 2021, 501, 200–209. [Google Scholar] [CrossRef]
- Reale, A.; Khong, T.; Mithraprabhu, S.; Spencer, A. Translational Potential of RNA Derived From Extracellular Vesicles in Multiple Myeloma. Front. Oncol. 2021, 11, 718502. [Google Scholar] [CrossRef]
- Desantis, V.; Saltarella, I.; Lamanuzzi, A.; Melaccio, A.; Solimando, A.G.; Mariggio, M.A.; Racanelli, V.; Paradiso, A.; Vacca, A.; Frassanito, M.A. MicroRNAs-Based Nano-Strategies as New Therapeutic Approach in Multiple Myeloma to Overcome Disease Progression and Drug Resistance. Int. J. Mol. Sci. 2020, 21, 3084. [Google Scholar] [CrossRef]
- Burnett, J.C.; Rossi, J.J. RNA-based therapeutics: Current progress and future prospects. Chem. Biol. 2012, 19, 60–71. [Google Scholar] [CrossRef]
- Giacobino, C.; Canta, M.; Fornaguera, C.; Borros, S.; Cauda, V. Extracellular Vesicles and Their Current Role in Cancer Immunotherapy. Cancers 2021, 13, 2280. [Google Scholar] [CrossRef]
- Zhang, L.; Sun, M.; He, Z.; Sun, J.; Li, H.; Luo, Q. Multi-functional extracellular vesicles: Potentials in cancer immunotherapy. Cancer Lett. 2022, 551, 215934. [Google Scholar] [CrossRef]
- Huang, F.; Wan, J.; Hu, W.; Hao, S. Enhancement of Anti-Leukemia Immunity by Leukemia-Derived Exosomes Via Downregulation of TGF-beta1 Expression. Cell Physiol. Biochem. 2017, 44, 240–254. [Google Scholar] [CrossRef] [PubMed]
- Huang, F.; Wan, J.; Hao, S.; Deng, X.; Chen, L.; Ma, L. TGF-beta1-silenced leukemia cell-derived exosomes target dendritic cells to induce potent anti-leukemic immunity in a mouse model. Cancer Immunol. Immunother. 2017, 66, 1321–1331. [Google Scholar] [CrossRef] [PubMed]
- Dang, X.T.T.; Kavishka, J.M.; Zhang, D.X.; Pirisinu, M.; Le, M.T.N. Extracellular Vesicles as an Efficient and Versatile System for Drug Delivery. Cells 2020, 9, 2191. [Google Scholar] [CrossRef] [PubMed]
- Huang, L.; Rong, Y.; Tang, X.; Yi, K.; Qi, P.; Hou, J.; Liu, W.; He, Y.; Gao, X.; Yuan, C.; et al. Engineered exosomes as an in situ DC-primed vaccine to boost antitumor immunity in breast cancer. Mol. Cancer 2022, 21, 45. [Google Scholar] [CrossRef]
- Koh, E.; Lee, E.J.; Nam, G.H.; Hong, Y.; Cho, E.; Yang, Y.; Kim, I.S. Exosome-SIRPalpha, a CD47 blockade increases cancer cell phagocytosis. Biomaterials 2017, 121, 121–129. [Google Scholar] [CrossRef]
- Hong, Y.; Kim, Y.K.; Kim, G.B.; Nam, G.H.; Kim, S.A.; Park, Y.; Yang, Y.; Kim, I.S. 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 2019, 8, 1670893. [Google Scholar] [CrossRef]
- Cheng, Q.; Shi, X.; Han, M.; Smbatyan, G.; Lenz, H.J.; Zhang, Y. Reprogramming Exosomes as Nanoscale Controllers of Cellular Immunity. J. Am. Chem. Soc. 2018, 140, 16413–16417. [Google Scholar] [CrossRef]
- McAndrews, K.M.; Che, S.P.Y.; LeBleu, V.S.; Kalluri, R. Effective delivery of STING agonist using exosomes suppresses tumor growth and enhances antitumor immunity. J. Biol. Chem. 2021, 296, 100523. [Google Scholar] [CrossRef]
- Chen, S.; Lv, M.; Fang, S.; Ye, W.; Gao, Y.; Xu, Y. Poly(I:C) enhanced anti-cervical cancer immunities induced by dendritic cells-derived exosomes. Int. J. Biol. Macromol. 2018, 113, 1182–1187. [Google Scholar] [CrossRef]
- Zhao, Z.; McGill, J.; Gamero-Kubota, P.; He, M. Microfluidic on-demand engineering of exosomes towards cancer immunotherapy. Lab Chip 2019, 19, 1877–1886. [Google Scholar] [CrossRef]
- Lee, H.; Park, H.; Noh, G.J.; Lee, E.S. pH-responsive hyaluronate-anchored extracellular vesicles to promote tumor-targeted drug delivery. Carbohydr. Polym. 2018, 202, 323–333. [Google Scholar] [CrossRef] [PubMed]
- Zhu, L.; Kalimuthu, S.; Gangadaran, P.; Oh, J.M.; Lee, H.W.; Baek, S.H.; Jeong, S.Y.; Lee, S.W.; Lee, J.; Ahn, B.C. Exosomes Derived From Natural Killer Cells Exert Therapeutic Effect in Melanoma. Theranostics 2017, 7, 2732–2745. [Google Scholar] [CrossRef]
- Zhu, L.; Gangadaran, P.; Kalimuthu, S.; Oh, J.M.; Baek, S.H.; Jeong, S.Y.; Lee, S.W.; Lee, J.; Ahn, B.C. Novel alternatives to extracellular vesicle-based immunotherapy—Exosome mimetics derived from natural killer cells. Artif. Cells Nanomed. Biotechnol. 2018, 46, S166–S179. [Google Scholar] [CrossRef]
- Atai, N.A.; Balaj, L.; van Veen, H.; Breakefield, X.O.; Jarzyna, P.A.; Van Noorden, C.J.; Skog, J.; Maguire, C.A. Heparin blocks transfer of extracellular vesicles between donor and recipient cells. J. Neurooncol. 2013, 115, 343–351. [Google Scholar] [CrossRef] [PubMed]
- Wu, X.; Kang, M.; Wang, D.; Zhu, M.; Hu, Y.; Zhang, Y.; Deng, C.; Chen, J.; Teng, L. Heparan sulfate analogues regulate tumor-derived exosome formation that attenuates exosome functions in tumor processes. Int. J. Biol. Macromol. 2021, 187, 481–491. [Google Scholar] [CrossRef] [PubMed]
- Sento, S.; Sasabe, E.; Yamamoto, T. Application of a Persistent Heparin Treatment Inhibits the Malignant Potential of Oral Squamous Carcinoma Cells Induced by Tumor Cell-Derived Exosomes. PLoS ONE 2016, 11, e0148454. [Google Scholar] [CrossRef] [PubMed]
- Mithraprabhu, S.; Chen, M.; Savvidou, I.; Reale, A.; Spencer, A. Liquid biopsy: An evolving paradigm for the biological characterisation of plasma cell disorders. Leukemia 2021, 35, 2771–2783. [Google Scholar] [CrossRef]
- Ostheimer, C.; Gunther, S.; Bache, M.; Vordermark, D.; Multhoff, G. Dynamics of Heat Shock Protein 70 Serum Levels As a Predictor of Clinical Response in Non-Small-Cell Lung Cancer and Correlation with the Hypoxia-Related Marker Osteopontin. Front. Immunol. 2017, 8, 1305. [Google Scholar] [CrossRef]
- McNulty, S.; Colaco, C.A.; Blandford, L.E.; Bailey, C.R.; Baschieri, S.; Todryk, S. Heat-shock proteins as dendritic cell-targeting vaccines—Getting warmer. Immunology 2013, 139, 407–415. [Google Scholar] [CrossRef]
- Madden, E.C.; Gorman, A.M.; Logue, S.E.; Samali, A. Tumour Cell Secretome in Chemoresistance and Tumour Recurrence. Trends Cancer 2020, 6, 489–505. [Google Scholar] [CrossRef]
- Xu, R.; Greening, D.W.; Chen, M.; Rai, A.; Ji, H.; Takahashi, N.; Simpson, R.J. Surfaceome of Exosomes Secreted from the Colorectal Cancer Cell Line SW480: Peripheral and Integral Membrane Proteins Analyzed by Proteolysis and TX114. Proteomics 2019, 19, e1700453. [Google Scholar] [CrossRef] [PubMed]
- Veerman, R.E.; Teeuwen, L.; Czarnewski, P.; Gucluler Akpinar, G.; Sandberg, A.; Cao, X.; Pernemalm, M.; Orre, L.M.; Gabrielsson, S.; Eldh, M. Molecular evaluation of five different isolation methods for extracellular vesicles reveals different clinical applicability and subcellular origin. J. Extracell. Vesicles 2021, 10, e12128. [Google Scholar] [CrossRef] [PubMed]
- Crescitelli, R.; Lasser, C.; Lotvall, J. Isolation and characterization of extracellular vesicle subpopulations from tissues. Nat. Protoc. 2021, 16, 1548–1580. [Google Scholar] [CrossRef] [PubMed]
- Rai, A.; Fang, H.; Fatmous, M.; Claridge, B.; Poh, Q.H.; Simpson, R.J.; Greening, D.W. A Protocol for Isolation, Purification, Characterization, and Functional Dissection of Exosomes. Methods Mol. Biol. 2021, 2261, 105–149. [Google Scholar] [CrossRef] [PubMed]
- Erdbrugger, U.; Blijdorp, C.J.; Bijnsdorp, I.V.; Borras, F.E.; Burger, D.; Bussolati, B.; Byrd, J.B.; Clayton, A.; Dear, J.W.; Falcon-Perez, J.M.; et al. Urinary extracellular vesicles: A position paper by the Urine Task Force of the International Society for Extracellular Vesicles. J. Extracell. Vesicles 2021, 10, e12093. [Google Scholar] [CrossRef] [PubMed]
- Yu, D.; Li, Y.; Wang, M.; Gu, J.; Xu, W.; Cai, H.; Fang, X.; Zhang, X. Exosomes as a new frontier of cancer liquid biopsy. Mol. Cancer 2022, 21, 56. [Google Scholar] [CrossRef] [PubMed]
- Ludwig, N.; Hong, C.S.; Ludwig, S.; Azambuja, J.H.; Sharma, P.; Theodoraki, M.N.; Whiteside, T.L. Isolation and Analysis of Tumor-Derived Exosomes. Curr. Protoc. Immunol. 2019, 127, e91. [Google Scholar] [CrossRef] [PubMed]
- Claridge, B.; Lozano, J.; Poh, Q.H.; Greening, D.W. Development of Extracellular Vesicle Therapeutics: Challenges, Considerations, and Opportunities. Front. Cell Dev. Biol. 2021, 9, 734720. [Google Scholar] [CrossRef]
- Rohde, E.; Pachler, K.; Gimona, M. Manufacturing and characterization of extracellular vesicles from umbilical cord-derived mesenchymal stromal cells for clinical testing. Cytotherapy 2019, 21, 581–592. [Google Scholar] [CrossRef]
- Harrison, S.J.; Perrot, A.; Alegre, A.; Simpson, D.; Wang, M.C.; Spencer, A.; Delimpasi, S.; Hulin, C.; Sunami, K.; Facon, T.; et al. Subgroup analysis of ICARIA-MM study in relapsed/refractory multiple myeloma patients with high-risk cytogenetics. Br. J. Haematol. 2021, 194, 120–131. [Google Scholar] [CrossRef]
Cancer Type (EV-Source) | Functional Molecules | Recipient/Target Cells | Functional Effect in Recipient Cells | References |
---|---|---|---|---|
Small cell lung cancer | PD-L1 | T cells | Inhibition of T cell activation | [53] |
Ovarian cancer | ARG1 | T cells | Suppression of T cells proliferation | [48] |
Ovarian cancer | SPHK1 | T cells | Promotion of T cell exhaustion | [54] |
Breast cancer | TβRII | T cells | CD8+ T cell exhaustion via SMAD3 and TCF1 | [55] |
Melanoma | miR-3187-3p, miR-498, miR-122, miR149, miR-181a/b | T cells | Inhibition of T cell activation | [56] |
Leukemia | miR-19a-3p | T cells | Suppression of T cells immune function | [57] |
Melanoma and squamous cell carcinoma of head and neck | FasL | T cells | Apoptosis of CD8+ T cells and Treg expansion | [58] |
Head and neck cancer | PD-L1 | CD8+ T cells | Decreased CD8+ T cell activation | [59] |
Glioblastoma cancer | PD-L1 | T cells | Suppressed T cell activation and proliferation | [47] |
Breast cancer | PD-L1 | T cells | Impaired activation and cancer killing potential of T cells | [60] |
Prostate and melanoma | PD-L1 | CD8+ T cells | Suppressed T cell activity | [61] |
Non small cell lung cancer | PD-L1 | CD8+ T cells | Immunosuppressive properties | [62] |
Melanoma | PD-L1 | T cells | Suppressed T cell functions | [52,63] |
Gastric cancer | PD-L1 | T cells | Apoptosis of T cells; reduced activation of peripheral blood mononuclear cells | [64] |
Hepatocellular carcinoma | 14-3-3ζ | T cells | Immunosuppressive phenotype | [65] |
Ovarian cancer | Ganglioside GD3 | T cells | Decreased T cell activation | [66] |
Ovarian cancer | T cells | Suppressed T cell functions | [67] | |
Melanoma | CD8+ T cells and NK cells | Enhanced apoptosis and suppressed proliferation and activation of CD8+ T cells; decreased NKG2D in NK cells | [68] | |
Head and neck cancer | T cells and NK cells | Enhanced apoptosis of CD8+ T cells; suppressed proliferation of CD4+ T cells; decreased NKG2D in NK cells | [46] | |
Melanoma cancer | miRNAs | CD4+ T cells | Enhanced apoptosis of CD4+ T cells | [69] |
Non-small cell lung cancer | mutant KRAS DNA | CD4+ T cells | Conversion of naïve CD4+ T cells into Treg-like cells | [70] |
Breast cancer | lncRNA SNHG16 | T cells | Induction of CD73+γδ1 Tregs | [71] |
Bladdre, colorectal, prostate, breast cancer | CD39, CD73 | T cells | Hydrolysis of ATP and generation of adenosine | [72] |
Hepatocellular carcinoma | HMGB1 | B cells | Expansion of Bregs via TLR2/4-MAPK signaling pathway | [73] |
Pancreatic cancer | TAAs | B cells | Inhibition of complement-dependent and antibody-dependent cell-mediated cytotoxicity | [74] |
Lung cancer | miR-214 | Treg | Suppression of PTEN; Treg expansion | [75] |
Colorectal cancer | miR-208b | Treg | Treg expansion by targeting PDCD4, tumour growth and drug resistance | [76] |
Breast cancer | lncRNA SNHG16 | Vδ1 T cells | Potentiation of the TGF-β1/SMAD5 pathway to upregulate CD73 expression in Vδ1 T cells | [77] |
Ovarian and head and neck squamous cancers | Treg | Increased FasL, IL-10, TGF-β1, CTLA-4, granzyme B and perforin expression and Treg-mediated stronger suppression of T cell proliferation | [78] | |
Nasopharyngeal carcinoma | miR-24-3p; miR-891a; miR-106a-5p; miR-20a-5p; miR-1908 | Treg | Downregulated ERK/STAT1/STAT3 phosphorylation with a shift of T cells towards Treg phenotype | [79] |
Leukemia | 4-1BBL/CD137L | Treg | Elevated expression of effector/tumour Treg markers (CD39, CCR8, CD30, TNFR2, CCR4, TIGIT, IL21R) | [80] |
Pancreatic ductal adenocarcinoma | TGF-β1 | NK cells | Down-regulation of NKG2D, CD107a, TNF-α, INF-γ, CD71, CD98; impaired glucose uptake ability; attenuated NK cell cytotoxic activity | [81] |
Hypoxic lung adenocarcinoma cells | miR-150-5p | NK cells | Down-regulation of CD226 and functional repression of NK cells | [82] |
Pancreatic cancer | miR-212-3p | DCs | Induced immune tolerance via RFXAP | [83] |
Pancreatic cancer | miR-203 | DCs | Inhibition of antigen presentation via TLR4/TNF-α/IL-12 | [84] |
Glioblastoma multiforme | LGALS9 | DCs | Inhibition of antigen recognition, processing and presentation | [85] |
Oral and oropharyngeal squamous cell carcinoma | miR-17-5p, miR-21, miR-16, miR-24, miR-181a, miR-23b | DCs | Impaired differentiation and maturation of mono-DCs | [86] |
Prostate cancer | PGE2 | DCs | Disrupted cytokine production with inhibition of T cell activation, and increased secretion of adenosine with impaired DC functions and direct pro-tumour effects | [87] |
Chronic lymphocytic leukaemia and breast cancer | DCs | Inhibited DC maturation | [88] | |
Melanoma | miR-155, miR-125b, miR-100, miR-146a, miR-146b, let-7e, miR-125a, and miR-99b | MDSCs | Induced immunosuppressive properties | [89] |
Melanoma | HSP86 | MDSCs | TLR4 and NFkB activation on MDSCs, generation of PD-L1+CD11b+Gr1+ MDSCs that suppress T cell functionality | [90] |
Thymoma, mammary carcinoma, and colon carcinoma | Hsp72 | MDSCs | Immunosuppressive signaling via TLR2/STAT3 axis | [76] |
Renal cancer | HSP70 | MDSCs | MDSC proliferation and activation via TLR2 signaling to promote tumour growth and immunosuppression | [77] |
Breast cancer | miR-9, miR-181a | MDSCs | Activated JAK/STAT signaling in eMDSCs through the targeting of SOCS3 and PIAS3 | [78] |
Breast cancer | TGF-β1, PGE2 | MDSCs | MDSC accumulation and accelerated tumour growth | [91] |
Hypoxic glioma cancer cells | miR-10a, miR-21 | MDSCs | Promoted MDSCs expansion and activation through miR-10a/Rora/IκBα/NF-κB and miR-21/Pten/PI3K/AKT pathways | [79] |
Oral squamous cell carcinoma | miR-21 | MDSCs and γδ T cells | Inhibited γδ T cell functions through MDSCs | [92] |
Hypoxic pancreatic cancer cells | miR-301a | Macrophages | M2 polarization via PTEN/PI3Kgamma | [80] |
Lung cancer | miR-103a | Macrophages | M2 polarization | [93] |
Lung cancer | EGFR | Macrophages | Lower host innate antiviral immunity through MEKK2/IRF3 axis | [94] |
Melanoma, lung and squamous skin cancers | Let-7a | Macrophages | Increased OXPHOS activity and TAM via AKT/mTOR | [95] |
p53-mutant cancer cells | miR-1246 | Macrophages | Promoted TAM phenotype | [96] |
Melanoma | miR-125b-5p | Macrophages | Promoted survival via LIPA | [97] |
Hypoxic pancreatic cancer cells | miR-301a-3p | Macrophages | Induced M2 phenotype via PTEN/PI3K signaling | [80] |
Pancreatic cancer | Arachidonic acid | Macrophages | Promoted M2 phenotype | [98] |
Pancreatic cancer cells undergoing ferroptosis | KRASG12D protein | Macrophages | Promoted M2 polarization through STAT3-dependent fatty acid oxidation | [99] |
Pancreatic cancer | miR-155-5p | Macrophages | TAM formation via EHF/Akt/NF-kB axis | [100] |
Metastatic osteosarcoma cells | Macrophages | Induced M2 polarization and impaired phagocytosis, efferocytosis, and macrophage-dependent tumour cell killing | [101] | |
Non-small cell lung cancer | Macrophages | Induced M2 polarization and via PD-L1/HIF1α | [102] | |
Colorectal cancer | miR-25-3p, miR-130b-3p, miR-425-5p | Macrophages | Induced M2 polarization through suppression of PTEN and activation of PI3K/Akt signaling and contributed to the establishment of liver metastasis | [103] |
Colorectal cancer | Macrophages | Mixed M1/M2 secretion pattern | [104] | |
Breast cancer | gp130 | Macrophages | Promoted activation of STAT3 signaling, and enhanced the levels of protumourigenic cytokines and the survival of macrophages | [105] |
Liver cancer cells undergoing ER stress | Macrophages | Promoted secretion of IL-6, MCP-1, IL-10 and TNF-α in macrophages through STAT3 signaling | [106] | |
Breast cancer cells undergoing ER stress | miR-27a-3p | Macrophages | Increase in PD-L1 expression in macrophages | [107] |
Breast cancer | Macrophages | Activation of NF-κB pathway in macrophages, and enhanced levels of IL-6, TNF-α, GCSF, and CCL2 in a TLR2-dependent manner | [108] | |
Gastric cancer | Macrophages | Activation of NF-κB and expression of the proinflammatory factors IL-6, TNF-α, and CCL2 in macrophages to promote tumour progression | [109] | |
Lung cancer | miR-21, miR-29a | Macrophages | Pro-metastatic inflammatory response by serving as ligands of TLR receptors in macrophages; NF-κB activation and increasing the secretion of IL-6 and TNF-α | [110] |
Hypoxic ovarian cancer cells | miR-21-3p, miR-125b-5p, miR-181d-5p | Macrophages | Differentiation into TAM | [111] |
Chronic lymphocytic leukemia (CLL) | Non coding Y RNA hY4 | Monocytes | Increased PD-L1 | [49] |
Pancreatic cancer | Monocytes | Decreased HLA-DR expression, induced arginase and ROS | [112] | |
Snail-expressing head and neck squamous cell carcinoma | miR-21 | Monocytes | M2 polarization | [113] |
Glioblastoma stem cells | Monocytes | M2 polarization and enhanced PD-L1 expression | [114] | |
Gastric cancer | Monocytes | Monocyte differentiation into PD-1+ TAMs, which suppress CD8+ T cell functions | [115] | |
Multiple myeloma | HDGF | Monocytes | MDSCs with suppressive functions | [116] |
Neuroblastoma | miR-21 | Monocytes | Trigger TLR8 with increased miR-155 which transfer to tumour cells leads to drug resistance | [117] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 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 (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Reale, A.; Khong, T.; Spencer, A. Extracellular Vesicles and Their Roles in the Tumor Immune Microenvironment. J. Clin. Med. 2022, 11, 6892. https://doi.org/10.3390/jcm11236892
Reale A, Khong T, Spencer A. Extracellular Vesicles and Their Roles in the Tumor Immune Microenvironment. Journal of Clinical Medicine. 2022; 11(23):6892. https://doi.org/10.3390/jcm11236892
Chicago/Turabian StyleReale, Antonia, Tiffany Khong, and Andrew Spencer. 2022. "Extracellular Vesicles and Their Roles in the Tumor Immune Microenvironment" Journal of Clinical Medicine 11, no. 23: 6892. https://doi.org/10.3390/jcm11236892
APA StyleReale, A., Khong, T., & Spencer, A. (2022). Extracellular Vesicles and Their Roles in the Tumor Immune Microenvironment. Journal of Clinical Medicine, 11(23), 6892. https://doi.org/10.3390/jcm11236892