Skip to main content
Top
Published in: Medical Oncology 11/2023

01-11-2023 | Metastasis | Review Article

Theranostic signature of tumor-derived exosomes in cancer

Authors: Samruti Kumar, Rajib Dhar, Lokesh Babu Sirkali Suresh Kumar, Gauresh Gurudas Shivji, Rama Jayaraj, Arikketh Devi

Published in: Medical Oncology | Issue 11/2023

Login to get access

Abstract

Cancer is the most challenging global health crisis. In the recent times, studies on extracellular vesicles (EVs) are adding a new chapter to cancer research and reports on EVs explores cancer in a new dimension. Exosomes are a group of subpopulations of EVs. It originates from the endosomes and carries biologically active molecules to the neighboring cells which in turn transforms the recipient cell activity. In general, it plays a role in cellular communication. The correlation between exosomes and cancer is fascinating. Tumor-derived exosomes (TEXs) play a dynamic role in cancer progression and are associated with uncontrolled cell growth, angiogenesis, immune suppression, and metastasis. Its molecular cargo is an excellent source of cancer biomarkers. Several advanced molecular profiling approaches assist in exploring the TEXs in depth. This paves the way for a strong foundation for identifying and detecting more specific and efficient biomarkers. TEXs are also gaining importance in scientific society for its role in cancer therapy and several clinical trials based on TEXs is a proof of its significance. In this review, we have highlighted the role of TEXs in mediating immune cell reprogramming, cancer development, metastasis, EMT, organ-specific metastasis, and its clinical significance in cancer theranostics. TEXs profiling is an effective method to understand the complications associated with cancer leading to good health and well-being of the individual and society as a whole.

Graphical abstract

Literature
3.
go back to reference Thery C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis and function. Nat Rev Immunol. 2002;2(8):569–79.PubMedCrossRef Thery C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis and function. Nat Rev Immunol. 2002;2(8):569–79.PubMedCrossRef
5.
go back to reference Baig MS, et al. Tumor-derived exosomes in the regulation of macrophage polarization. Inflamm Res. 2020;69(5):435–51.PubMedCrossRef Baig MS, et al. Tumor-derived exosomes in the regulation of macrophage polarization. Inflamm Res. 2020;69(5):435–51.PubMedCrossRef
7.
go back to reference Ghosh, S., et al., Clinical impact of exosomes in colorectal cancer metastasis. ACS Appl Bio Mater. 2023. Ghosh, S., et al., Clinical impact of exosomes in colorectal cancer metastasis. ACS Appl Bio Mater. 2023.
8.
go back to reference Mukherjee, S., et al., Exosomal miRNAs and breast cancer: a complex theranostics interlink with clinical significance. Biomarkers. 2023. 1–17. Mukherjee, S., et al., Exosomal miRNAs and breast cancer: a complex theranostics interlink with clinical significance. Biomarkers. 2023. 1–17.
9.
go back to reference Mathivanan, S., et al., ExoCarta 2012: database of exosomal proteins, RNA and lipids. Nucleic Acids Res, 2012. 40(Database issue): pp D1241–4. Mathivanan, S., et al., ExoCarta 2012: database of exosomal proteins, RNA and lipids. Nucleic Acids Res, 2012. 40(Database issue): pp D1241–4.
12.
go back to reference Dhar R, Devi A, Patil S, Tovani-Palone MR. Exosomes in cancer therapy: advances and current challenges. Electron J Gen Med. 2023;20(5):em524.CrossRef Dhar R, Devi A, Patil S, Tovani-Palone MR. Exosomes in cancer therapy: advances and current challenges. Electron J Gen Med. 2023;20(5):em524.CrossRef
15.
go back to reference Saleem SN, Abdel-Mageed AB. Tumor-derived exosomes in oncogenic reprogramming and cancer progression. Cell Mol Life Sci. 2015;72(1):1–10.PubMedCrossRef Saleem SN, Abdel-Mageed AB. Tumor-derived exosomes in oncogenic reprogramming and cancer progression. Cell Mol Life Sci. 2015;72(1):1–10.PubMedCrossRef
16.
go back to reference Colombo M, Raposo G, Thery C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu Rev Cell Dev Biol. 2014;30:255–89.PubMedCrossRef Colombo M, Raposo G, Thery C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu Rev Cell Dev Biol. 2014;30:255–89.PubMedCrossRef
18.
go back to reference Bhattacharya B, et al. Exosome DNA: An untold story of cancer. Clin Transl Disc. 2023;3: e218.CrossRef Bhattacharya B, et al. Exosome DNA: An untold story of cancer. Clin Transl Disc. 2023;3: e218.CrossRef
19.
go back to reference Shivji GG, et al. Role of exosomes and its emerging therapeutic applications in the pathophysiology of non-infectious diseases. Biomarkers. 2022;27(6):534–48.PubMedCrossRef Shivji GG, et al. Role of exosomes and its emerging therapeutic applications in the pathophysiology of non-infectious diseases. Biomarkers. 2022;27(6):534–48.PubMedCrossRef
20.
go back to reference Abels ER, Breakefield XO. Introduction to extracellular vesicles: biogenesis, RNA cargo selection, content, release, and uptake. Cell Mol Neurobiol. 2016;36(3):301–12.PubMedPubMedCentralCrossRef Abels ER, Breakefield XO. Introduction to extracellular vesicles: biogenesis, RNA cargo selection, content, release, and uptake. Cell Mol Neurobiol. 2016;36(3):301–12.PubMedPubMedCentralCrossRef
21.
go back to reference Tricarico C, Clancy J, D’Souza-Schorey C. Biology and biogenesis of shed microvesicles. Small GTPases. 2017;8(4):220–32.PubMedCrossRef Tricarico C, Clancy J, D’Souza-Schorey C. Biology and biogenesis of shed microvesicles. Small GTPases. 2017;8(4):220–32.PubMedCrossRef
22.
go back to reference Guo X, Tan W, Wang C. The emerging roles of exosomal circRNAs in diseases. Clin Transl Oncol. 2021;23(6):1020–33.PubMedCrossRef Guo X, Tan W, Wang C. The emerging roles of exosomal circRNAs in diseases. Clin Transl Oncol. 2021;23(6):1020–33.PubMedCrossRef
23.
go back to reference Dhar R, et al. Exosome and epithelial–mesenchymal transition: a complex secret of cancer progression. J Cell Mol Med. 2023. Dhar R, et al. Exosome and epithelial–mesenchymal transition: a complex secret of cancer progression. J Cell Mol Med. 2023.
24.
go back to reference Miller IV, Grunewald TG. Tumour-derived exosomes: tiny envelopes for big stories. Biol Cell. 2015;107(9):287–305.PubMedCrossRef Miller IV, Grunewald TG. Tumour-derived exosomes: tiny envelopes for big stories. Biol Cell. 2015;107(9):287–305.PubMedCrossRef
25.
26.
go back to reference Jabbari N, et al. Tumor-derived extracellular vesicles: insights into bystander effects of exosomes after irradiation. Lasers Med Sci. 2020;35(3):531–45.PubMedCrossRef Jabbari N, et al. Tumor-derived extracellular vesicles: insights into bystander effects of exosomes after irradiation. Lasers Med Sci. 2020;35(3):531–45.PubMedCrossRef
27.
go back to reference Jafari A, et al. Exosomes and cancer: from molecular mechanisms to clinical applications. Med Oncol. 2021;38(4):45.PubMedCrossRef Jafari A, et al. Exosomes and cancer: from molecular mechanisms to clinical applications. Med Oncol. 2021;38(4):45.PubMedCrossRef
29.
go back to reference Meehan K, Vella LJ. The contribution of tumour-derived exosomes to the hallmarks of cancer. Crit Rev Clin Lab Sci. 2016;53(2):121–31.PubMedCrossRef Meehan K, Vella LJ. The contribution of tumour-derived exosomes to the hallmarks of cancer. Crit Rev Clin Lab Sci. 2016;53(2):121–31.PubMedCrossRef
30.
32.
go back to reference Fontana S, et al. Contribution of proteomics to understanding the role of tumor-derived exosomes in cancer progression: state of the art and new perspectives. Proteomics. 2013;13(10–11):1581–94.PubMed Fontana S, et al. Contribution of proteomics to understanding the role of tumor-derived exosomes in cancer progression: state of the art and new perspectives. Proteomics. 2013;13(10–11):1581–94.PubMed
33.
go back to reference Rak J, Guha A. Extracellular vesicles–vehicles that spread cancer genes. BioEssays. 2012;34(6):489–97.PubMedCrossRef Rak J, Guha A. Extracellular vesicles–vehicles that spread cancer genes. BioEssays. 2012;34(6):489–97.PubMedCrossRef
35.
go back to reference Niu L, et al. Tumor-derived exosomal proteins as diagnostic biomarkers in non-small cell lung cancer. Cancer Sci. 2019;110(1):433–42.PubMedCrossRef Niu L, et al. Tumor-derived exosomal proteins as diagnostic biomarkers in non-small cell lung cancer. Cancer Sci. 2019;110(1):433–42.PubMedCrossRef
36.
go back to reference Sharma S, et al. Tumor-derived exosomes in ovarian cancer - liquid biopsies for early detection and real-time monitoring of cancer progression. Oncotarget. 2017;8(61):104687–703.PubMedPubMedCentralCrossRef Sharma S, et al. Tumor-derived exosomes in ovarian cancer - liquid biopsies for early detection and real-time monitoring of cancer progression. Oncotarget. 2017;8(61):104687–703.PubMedPubMedCentralCrossRef
37.
go back to reference Pashoutan Sarvar, D., K. Shamsasenjan, and P. Akbarzadehlaleh, Mesenchymal Stem Cell-Derived Exosomes: New Opportunity in Cell-Free Therapy. Adv Pharm Bull, 2016. 6(3): 293–299. Pashoutan Sarvar, D., K. Shamsasenjan, and P. Akbarzadehlaleh, Mesenchymal Stem Cell-Derived Exosomes: New Opportunity in Cell-Free Therapy. Adv Pharm Bull, 2016. 6(3): 293–299.
39.
go back to reference Kharaziha P, et al. Tumor cell-derived exosomes: a message in a bottle. Biochim Biophys Acta. 2012;1826(1):103–11.PubMed Kharaziha P, et al. Tumor cell-derived exosomes: a message in a bottle. Biochim Biophys Acta. 2012;1826(1):103–11.PubMed
41.
go back to reference Mathieu M, et al. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat Cell Biol. 2019;21(1):9–17.PubMedCrossRef Mathieu M, et al. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat Cell Biol. 2019;21(1):9–17.PubMedCrossRef
42.
go back to reference Admyre C, et al. Exosomes with immune modulatory features are present in human breast milk. J Immunol. 2007;179(3):1969–78.PubMedCrossRef Admyre C, et al. Exosomes with immune modulatory features are present in human breast milk. J Immunol. 2007;179(3):1969–78.PubMedCrossRef
43.
44.
go back to reference Cheng H, et al. The tumor microenvironment shapes the molecular characteristics of exhausted CD8(+) T cells. Cancer Lett. 2021;506:55–66.PubMedCrossRef Cheng H, et al. The tumor microenvironment shapes the molecular characteristics of exhausted CD8(+) T cells. Cancer Lett. 2021;506:55–66.PubMedCrossRef
45.
go back to reference Whiteside, T.L., B. Diergaarde, and C.S. Hong, Tumor-Derived Exosomes (TEX) and Their Role in Immuno-Oncology. Int J Mol Sci, 2021. 22(12). Whiteside, T.L., B. Diergaarde, and C.S. Hong, Tumor-Derived Exosomes (TEX) and Their Role in Immuno-Oncology. Int J Mol Sci, 2021. 22(12).
46.
48.
go back to reference Naito Y, et al. How cancer cells dictate their microenvironment: present roles of extracellular vesicles. Cell Mol Life Sci. 2017;74(4):697–713.PubMedCrossRef Naito Y, et al. How cancer cells dictate their microenvironment: present roles of extracellular vesicles. Cell Mol Life Sci. 2017;74(4):697–713.PubMedCrossRef
50.
go back to reference Malla RR, Shailender G, Kamal MA. Exosomes: critical mediators of tumour microenvironment reprogramming. Curr Med Chem. 2021;28(39):8182–202.PubMedCrossRef Malla RR, Shailender G, Kamal MA. Exosomes: critical mediators of tumour microenvironment reprogramming. Curr Med Chem. 2021;28(39):8182–202.PubMedCrossRef
51.
go back to reference Lan J, et al. M2 macrophage-derived exosomes promote cell migration and invasion in colon cancer. Cancer Res. 2019;79(1):146–58.PubMedCrossRef Lan J, et al. M2 macrophage-derived exosomes promote cell migration and invasion in colon cancer. Cancer Res. 2019;79(1):146–58.PubMedCrossRef
52.
go back to reference Milane L, et al. Exosome mediated communication within the tumor microenvironment. J Control Release. 2015;219:278–94.PubMedCrossRef Milane L, et al. Exosome mediated communication within the tumor microenvironment. J Control Release. 2015;219:278–94.PubMedCrossRef
55.
go back to reference Kara-Terki L et al. Critical Roles of Tumor Extracellular Vesicles in the Microenvironment of Thoracic Cancers. Int J Mol Sci, 2020. 21(17). Kara-Terki L et al. Critical Roles of Tumor Extracellular Vesicles in the Microenvironment of Thoracic Cancers. Int J Mol Sci, 2020. 21(17).
57.
go back to reference Desage AL et al. The Immune Microenvironment of Malignant Pleural Mesothelioma: A Literature Review. Cancers (Basel), 2021. 13(13) Desage AL et al. The Immune Microenvironment of Malignant Pleural Mesothelioma: A Literature Review. Cancers (Basel), 2021. 13(13)
59.
go back to reference Whiteside TL. Exosomes in cancer: another mechanism of tumor-induced immune suppression. Adv Exp Med Biol. 2017;1036:81–9.PubMedCrossRef Whiteside TL. Exosomes in cancer: another mechanism of tumor-induced immune suppression. Adv Exp Med Biol. 2017;1036:81–9.PubMedCrossRef
61.
go back to reference Maji S, et al. Exosomal Annexin II promotes angiogenesis and breast cancer metastasis. Mol Cancer Res. 2017;15(1):93–105.PubMedCrossRef Maji S, et al. Exosomal Annexin II promotes angiogenesis and breast cancer metastasis. Mol Cancer Res. 2017;15(1):93–105.PubMedCrossRef
62.
64.
go back to reference Ying X, et al. Epithelial ovarian cancer-secreted exosomal miR-222-3p induces polarization of tumor-associated macrophages. Oncotarget. 2016;7(28):43076–87.PubMedPubMedCentralCrossRef Ying X, et al. Epithelial ovarian cancer-secreted exosomal miR-222-3p induces polarization of tumor-associated macrophages. Oncotarget. 2016;7(28):43076–87.PubMedPubMedCentralCrossRef
65.
go back to reference Worbs T, Hammerschmidt SI, Forster R. Dendritic cell migration in health and disease. Nat Rev Immunol. 2017;17(1):30–48.PubMedCrossRef Worbs T, Hammerschmidt SI, Forster R. Dendritic cell migration in health and disease. Nat Rev Immunol. 2017;17(1):30–48.PubMedCrossRef
66.
go back to reference Yu S, et al. Tumor exosomes inhibit differentiation of bone marrow dendritic cells. J Immunol. 2007;178(11):6867–75.PubMedCrossRef Yu S, et al. Tumor exosomes inhibit differentiation of bone marrow dendritic cells. J Immunol. 2007;178(11):6867–75.PubMedCrossRef
67.
go back to reference Zhou M, et al. Pancreatic cancer derived exosomes regulate the expression of TLR4 in dendritic cells via miR-203. Cell Immunol. 2014;292(1–2):65–9.PubMedCrossRef Zhou M, et al. Pancreatic cancer derived exosomes regulate the expression of TLR4 in dendritic cells via miR-203. Cell Immunol. 2014;292(1–2):65–9.PubMedCrossRef
69.
go back to reference Reiners KS, et al. Soluble ligands for NK cell receptors promote evasion of chronic lymphocytic leukemia cells from NK cell anti-tumor activity. Blood. 2013;121(18):3658–65.PubMedPubMedCentralCrossRef Reiners KS, et al. Soluble ligands for NK cell receptors promote evasion of chronic lymphocytic leukemia cells from NK cell anti-tumor activity. Blood. 2013;121(18):3658–65.PubMedPubMedCentralCrossRef
70.
go back to reference Garcia-Iglesias T, et al. Low NKp30, NKp46 and NKG2D expression and reduced cytotoxic activity on NK cells in cervical cancer and precursor lesions. BMC Cancer. 2009;9:186.PubMedPubMedCentralCrossRef Garcia-Iglesias T, et al. Low NKp30, NKp46 and NKG2D expression and reduced cytotoxic activity on NK cells in cervical cancer and precursor lesions. BMC Cancer. 2009;9:186.PubMedPubMedCentralCrossRef
71.
go back to reference Clayton A, et al. Human tumor-derived exosomes down-modulate NKG2D expression. J Immunol. 2008;180(11):7249–58.PubMedCrossRef Clayton A, et al. Human tumor-derived exosomes down-modulate NKG2D expression. J Immunol. 2008;180(11):7249–58.PubMedCrossRef
72.
go back to reference Szczepanski MJ, et al. 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(9):1302–9.PubMedPubMedCentralCrossRef Szczepanski MJ, et al. 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(9):1302–9.PubMedPubMedCentralCrossRef
73.
go back to reference Berchem G, et al. Hypoxic tumor-derived microvesicles negatively regulate NK cell function by a mechanism involving TGF-beta and miR23a transfer. Oncoimmunology. 2016;5(4): e1062968.PubMedCrossRef Berchem G, et al. Hypoxic tumor-derived microvesicles negatively regulate NK cell function by a mechanism involving TGF-beta and miR23a transfer. Oncoimmunology. 2016;5(4): e1062968.PubMedCrossRef
74.
go back to reference Ye 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(1):145.PubMedPubMedCentralCrossRef Ye 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(1):145.PubMedPubMedCentralCrossRef
75.
go back to reference Schroeder JC et al. Circulating exosomes inhibit b cell proliferation and activity. Cancers (Basel), 2020. 12(8) Schroeder JC et al. Circulating exosomes inhibit b cell proliferation and activity. Cancers (Basel), 2020. 12(8)
77.
go back to reference Mao Y, et al. Circulating exosomes from esophageal squamous cell carcinoma mediate the generation of B10 and PD-1(high) Breg cells. Cancer Sci. 2019;110(9):2700–10.PubMedPubMedCentralCrossRef Mao Y, et al. Circulating exosomes from esophageal squamous cell carcinoma mediate the generation of B10 and PD-1(high) Breg cells. Cancer Sci. 2019;110(9):2700–10.PubMedPubMedCentralCrossRef
78.
go back to reference Wieckowski EU, et al. Tumor-derived microvesicles promote regulatory T cell expansion and induce apoptosis in tumor-reactive activated CD8+ T lymphocytes. J Immunol. 2009;183(6):3720–30.PubMedCrossRef Wieckowski EU, et al. Tumor-derived microvesicles promote regulatory T cell expansion and induce apoptosis in tumor-reactive activated CD8+ T lymphocytes. J Immunol. 2009;183(6):3720–30.PubMedCrossRef
79.
go back to reference Montes CL, et al. Tumor-induced senescent T cells with suppressor function: a potential form of tumor immune evasion. Cancer Res. 2008;68(3):870–9.PubMedCrossRef Montes CL, et al. Tumor-induced senescent T cells with suppressor function: a potential form of tumor immune evasion. Cancer Res. 2008;68(3):870–9.PubMedCrossRef
80.
go back to reference Zhang Y, et al. Interleukin-7 inhibits tumor-induced CD27-CD28- suppressor T cells: implications for cancer immunotherapy. Clin Cancer Res. 2011;17(15):4975–86.PubMedPubMedCentralCrossRef Zhang Y, et al. Interleukin-7 inhibits tumor-induced CD27-CD28- suppressor T cells: implications for cancer immunotherapy. Clin Cancer Res. 2011;17(15):4975–86.PubMedPubMedCentralCrossRef
82.
go back to reference Siegel RM, et al. The multifaceted role of Fas signaling in immune cell homeostasis and autoimmunity. Nat Immunol. 2000;1(6):469–74.CrossRef Siegel RM, et al. The multifaceted role of Fas signaling in immune cell homeostasis and autoimmunity. Nat Immunol. 2000;1(6):469–74.CrossRef
83.
go back to reference Kim JW, et al. Fas ligand-positive membranous vesicles isolated from sera of patients with oral cancer induce apoptosis of activated T lymphocytes. Clin Cancer Res. 2005;11(3):1010–20.PubMedCrossRef Kim JW, et al. Fas ligand-positive membranous vesicles isolated from sera of patients with oral cancer induce apoptosis of activated T lymphocytes. Clin Cancer Res. 2005;11(3):1010–20.PubMedCrossRef
84.
go back to reference Abusamra AJ, et al. Tumor exosomes expressing Fas ligand mediate CD8+ T-cell apoptosis. Blood Cells Mol Dis. 2005;35(2):169–73.PubMedCrossRef Abusamra AJ, et al. Tumor exosomes expressing Fas ligand mediate CD8+ T-cell apoptosis. Blood Cells Mol Dis. 2005;35(2):169–73.PubMedCrossRef
85.
go back to reference Contini P, et al. Apoptosis of antigen-specific T lymphocytes upon the engagement of CD8 by soluble HLA class I molecules is Fas ligand/Fas mediated: evidence for the involvement of p56lck, calcium calmodulin kinase II, and Calcium-independent protein kinase C signaling pathways and for NF-kappaB and NF-AT nuclear translocation. J Immunol. 2005;175(11):7244–54.PubMedCrossRef Contini P, et al. Apoptosis of antigen-specific T lymphocytes upon the engagement of CD8 by soluble HLA class I molecules is Fas ligand/Fas mediated: evidence for the involvement of p56lck, calcium calmodulin kinase II, and Calcium-independent protein kinase C signaling pathways and for NF-kappaB and NF-AT nuclear translocation. J Immunol. 2005;175(11):7244–54.PubMedCrossRef
86.
87.
go back to reference Del Re M, et al. PD-L1 mRNA expression in plasma-derived exosomes is associated with response to anti-PD-1 antibodies in melanoma and NSCLC. Br J Cancer. 2018;118(6):820–4.PubMedPubMedCentralCrossRef Del Re M, et al. PD-L1 mRNA expression in plasma-derived exosomes is associated with response to anti-PD-1 antibodies in melanoma and NSCLC. Br J Cancer. 2018;118(6):820–4.PubMedPubMedCentralCrossRef
88.
go back to reference Theodoraki MN, et al. Clinical significance of PD-L1(+) exosomes in plasma of head and neck cancer patients. Clin Cancer Res. 2018;24(4):896–905.PubMedCrossRef Theodoraki MN, et al. Clinical significance of PD-L1(+) exosomes in plasma of head and neck cancer patients. Clin Cancer Res. 2018;24(4):896–905.PubMedCrossRef
89.
go back to reference Mincheva-Nilsson L, Baranov V. Cancer exosomes and NKG2D receptor-ligand interactions: impairing NKG2D-mediated cytotoxicity and anti-tumour immune surveillance. Semin Cancer Biol. 2014;28:24–30.PubMedCrossRef Mincheva-Nilsson L, Baranov V. Cancer exosomes and NKG2D receptor-ligand interactions: impairing NKG2D-mediated cytotoxicity and anti-tumour immune surveillance. Semin Cancer Biol. 2014;28:24–30.PubMedCrossRef
90.
go back to reference Skog J, et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol. 2008;10(12):1470–6.PubMedPubMedCentralCrossRef Skog J, et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol. 2008;10(12):1470–6.PubMedPubMedCentralCrossRef
91.
go back to reference Balaj L, et al. Tumour microvesicles contain retrotransposon elements and amplified oncogene sequences. Nat Commun. 2011;2:180.PubMedCrossRef Balaj L, et al. Tumour microvesicles contain retrotransposon elements and amplified oncogene sequences. Nat Commun. 2011;2:180.PubMedCrossRef
92.
go back to reference Ye SB, et al. Tumor-derived exosomes promote tumor progression and T-cell dysfunction through the regulation of enriched exosomal microRNAs in human nasopharyngeal carcinoma. Oncotarget. 2014;5(14):5439–52.PubMedPubMedCentralCrossRef Ye SB, et al. Tumor-derived exosomes promote tumor progression and T-cell dysfunction through the regulation of enriched exosomal microRNAs in human nasopharyngeal carcinoma. Oncotarget. 2014;5(14):5439–52.PubMedPubMedCentralCrossRef
94.
95.
go back to reference Diao J, et al. Exosomal Hsp70 mediates immunosuppressive activity of the myeloid-derived suppressor cells via phosphorylation of Stat3. Med Oncol. 2015;32(2):453.PubMedCrossRef Diao J, et al. Exosomal Hsp70 mediates immunosuppressive activity of the myeloid-derived suppressor cells via phosphorylation of Stat3. Med Oncol. 2015;32(2):453.PubMedCrossRef
96.
go back to reference Gobbo J et al. Restoring anticancer immune response by targeting tumor-derived exosomes with a HSP70 peptide aptamer. J Natl Cancer Inst; 2016. 108(3) Gobbo J et al. Restoring anticancer immune response by targeting tumor-derived exosomes with a HSP70 peptide aptamer. J Natl Cancer Inst; 2016. 108(3)
98.
99.
100.
go back to reference Roma-Rodrigues C, et al. Smuggling gold nanoparticles across cell types—a new role for exosomes in gene silencing. Nanomedicine. 2017;13(4):1389–98.PubMedCrossRef Roma-Rodrigues C, et al. Smuggling gold nanoparticles across cell types—a new role for exosomes in gene silencing. Nanomedicine. 2017;13(4):1389–98.PubMedCrossRef
101.
go back to reference Sung BH, et al. Directional cell movement through tissues is controlled by exosome secretion. Nat Commun. 2015;6:7164.PubMedCrossRef Sung BH, et al. Directional cell movement through tissues is controlled by exosome secretion. Nat Commun. 2015;6:7164.PubMedCrossRef
102.
go back to reference Koumangoye RB, et al. Detachment of breast tumor cells induces rapid secretion of exosomes which subsequently mediate cellular adhesion and spreading. PLoS ONE. 2011;6(9): e24234.PubMedPubMedCentralCrossRef Koumangoye RB, et al. Detachment of breast tumor cells induces rapid secretion of exosomes which subsequently mediate cellular adhesion and spreading. PLoS ONE. 2011;6(9): e24234.PubMedPubMedCentralCrossRef
104.
go back to reference Luga V, et al. Exosomes mediate stromal mobilization of autocrine Wnt-PCP signaling in breast cancer cell migration. Cell. 2012;151(7):1542–56.PubMedCrossRef Luga V, et al. Exosomes mediate stromal mobilization of autocrine Wnt-PCP signaling in breast cancer cell migration. Cell. 2012;151(7):1542–56.PubMedCrossRef
105.
go back to reference Paolillo M, Schinelli S Integrins and exosomes, a dangerous liaison in cancer progression. Cancers (Basel), 2017. 9(8) Paolillo M, Schinelli S Integrins and exosomes, a dangerous liaison in cancer progression. Cancers (Basel), 2017. 9(8)
106.
go back to reference Bai S, et al. Role of tumour-derived exosomes in metastasis. Biomed Pharmacother. 2022;147: 112657.PubMedCrossRef Bai S, et al. Role of tumour-derived exosomes in metastasis. Biomed Pharmacother. 2022;147: 112657.PubMedCrossRef
107.
go back to reference Webber J, et al. Cancer exosomes trigger fibroblast to myofibroblast differentiation. Cancer Res. 2010;70(23):9621–30.PubMedCrossRef Webber J, et al. Cancer exosomes trigger fibroblast to myofibroblast differentiation. Cancer Res. 2010;70(23):9621–30.PubMedCrossRef
108.
go back to reference Zhang Q, Peng C. Cancer-associated fibroblasts regulate the biological behavior of cancer cells and stroma in gastric cancer. Oncol Lett. 2018;15(1):691–8.PubMed Zhang Q, Peng C. Cancer-associated fibroblasts regulate the biological behavior of cancer cells and stroma in gastric cancer. Oncol Lett. 2018;15(1):691–8.PubMed
110.
go back to reference Hicklin DJ, Ellis LM. Role of the vascular endothelial growth factor pathway in tumor growth and angiogenesis. J Clin Oncol. 2005;23(5):1011–27.PubMedCrossRef Hicklin DJ, Ellis LM. Role of the vascular endothelial growth factor pathway in tumor growth and angiogenesis. J Clin Oncol. 2005;23(5):1011–27.PubMedCrossRef
111.
go back to reference Spannuth WA, Sood AK, Coleman RL. Angiogenesis as a strategic target for ovarian cancer therapy. Nat Clin Pract Oncol. 2008;5(4):194–204.PubMedCrossRef Spannuth WA, Sood AK, Coleman RL. Angiogenesis as a strategic target for ovarian cancer therapy. Nat Clin Pract Oncol. 2008;5(4):194–204.PubMedCrossRef
112.
go back to reference Momeny M, et al. Anti-tumour activity of tivozanib, a pan-inhibitor of VEGF receptors, in therapy-resistant ovarian carcinoma cells. Sci Rep. 2017;7:45954.PubMedPubMedCentralCrossRef Momeny M, et al. Anti-tumour activity of tivozanib, a pan-inhibitor of VEGF receptors, in therapy-resistant ovarian carcinoma cells. Sci Rep. 2017;7:45954.PubMedPubMedCentralCrossRef
113.
go back to reference Katoh M. Therapeutics targeting angiogenesis: genetics and epigenetics, extracellular miRNAs and signaling networks (Review). Int J Mol Med. 2013;32(4):763–7.PubMedPubMedCentralCrossRef Katoh M. Therapeutics targeting angiogenesis: genetics and epigenetics, extracellular miRNAs and signaling networks (Review). Int J Mol Med. 2013;32(4):763–7.PubMedPubMedCentralCrossRef
115.
go back to reference Gesierich S et al. Systemic induction of the angiogenesis switch by the tetraspanin D6.1A/CO-029. Cancer Res, 2006; 66(14): 7083–94. Gesierich S et al. Systemic induction of the angiogenesis switch by the tetraspanin D6.1A/CO-029. Cancer Res, 2006; 66(14): 7083–94.
116.
118.
go back to reference Syn N, et al. Exosome-mediated metastasis: from epithelial-mesenchymal transition to escape from immunosurveillance. Trends Pharmacol Sci. 2016;37(7):606–17.PubMedCrossRef Syn N, et al. Exosome-mediated metastasis: from epithelial-mesenchymal transition to escape from immunosurveillance. Trends Pharmacol Sci. 2016;37(7):606–17.PubMedCrossRef
119.
go back to reference Sanchez CA, et al. Exosomes from bulk and stem cells from human prostate cancer have a differential microRNA content that contributes cooperatively over local and pre-metastatic niche. Oncotarget. 2016;7(4):3993–4008.PubMedCrossRef Sanchez CA, et al. Exosomes from bulk and stem cells from human prostate cancer have a differential microRNA content that contributes cooperatively over local and pre-metastatic niche. Oncotarget. 2016;7(4):3993–4008.PubMedCrossRef
120.
go back to reference Dhar R et al. Exosomal microRNAs (exoMIRs): micromolecules with macro impact in oral cancer. 3 Biotech. 2022(12):155. Dhar R et al. Exosomal microRNAs (exoMIRs): micromolecules with macro impact in oral cancer. 3 Biotech. 2022(12):155.
121.
go back to reference Geiger TR, Peeper DS. Metastasis mechanisms. Biochim Biophys Acta. 2009;1796(2):293–308.PubMed Geiger TR, Peeper DS. Metastasis mechanisms. Biochim Biophys Acta. 2009;1796(2):293–308.PubMed
122.
124.
go back to reference Whiteside TL. The role of tumor-derived exosomes in epithelial mesenchymal transition (EMT). Transl Cancer Res. 2017;6(Suppl 1):S90–2.CrossRef Whiteside TL. The role of tumor-derived exosomes in epithelial mesenchymal transition (EMT). Transl Cancer Res. 2017;6(Suppl 1):S90–2.CrossRef
125.
go back to reference Wang X, et al. Hypoxic tumor-derived exosomal miR-301a mediates M2 macrophage polarization via PTEN/PI3Kgamma to promote pancreatic cancer metastasis. Cancer Res. 2018;78(16):4586–98.CrossRef Wang X, et al. Hypoxic tumor-derived exosomal miR-301a mediates M2 macrophage polarization via PTEN/PI3Kgamma to promote pancreatic cancer metastasis. Cancer Res. 2018;78(16):4586–98.CrossRef
126.
127.
go back to reference Dhar R et al. Interrelation between extracellular vesicles miRNAs with chronic lung diseases. J Cell Physiol, 237(11):4021–4036. Dhar R et al. Interrelation between extracellular vesicles miRNAs with chronic lung diseases. J Cell Physiol, 237(11):4021–4036.
130.
go back to reference Ferlay J et al. Cancer statistics for the year 2020: an overview. Int J Cancer, 2021. Ferlay J et al. Cancer statistics for the year 2020: an overview. Int J Cancer, 2021.
131.
133.
go back to reference Seijo LM, et al. Biomarkers in lung cancer screening: achievements, promises, and challenges. J Thorac Oncol. 2019;14(3):343–57.PubMedCrossRef Seijo LM, et al. Biomarkers in lung cancer screening: achievements, promises, and challenges. J Thorac Oncol. 2019;14(3):343–57.PubMedCrossRef
134.
go back to reference Cheng N, et al. Recent advances in biosensors for detecting cancer-derived exosomes. Trends Biotechnol. 2019;37(11):1236–54.PubMedCrossRef Cheng N, et al. Recent advances in biosensors for detecting cancer-derived exosomes. Trends Biotechnol. 2019;37(11):1236–54.PubMedCrossRef
135.
go back to reference Gowri A, Ashwin Kumar N, Suresh Anand BS, Recent advances in nanomaterials based biosensors for point of care (PoC) diagnosis of Covid-19—a minireview. Trends Analyt Chem, 2021;137: 116205. Gowri A, Ashwin Kumar N, Suresh Anand BS, Recent advances in nanomaterials based biosensors for point of care (PoC) diagnosis of Covid-19—a minireview. Trends Analyt Chem, 2021;137: 116205.
136.
go back to reference Sardini E, Serpelloni M, Tonello S, Printed electrochemical biosensors: opportunities and metrological challenges. Biosensors (Basel), 2020. 10(11). Sardini E, Serpelloni M, Tonello S, Printed electrochemical biosensors: opportunities and metrological challenges. Biosensors (Basel), 2020. 10(11).
137.
go back to reference Moss JL, et al. Persistent poverty and cancer mortality rates: an analysis of county-level poverty designations. Cancer Epidemiol Biomarkers Prev. 2020;29(10):1949–54.PubMedPubMedCentralCrossRef Moss JL, et al. Persistent poverty and cancer mortality rates: an analysis of county-level poverty designations. Cancer Epidemiol Biomarkers Prev. 2020;29(10):1949–54.PubMedPubMedCentralCrossRef
138.
go back to reference Yoshida K, et al. Exploring designability of electrostatic complementarity at an antigen-antibody interface directed by mutagenesis, biophysical analysis, and molecular dynamics simulations. Sci Rep. 2019;9(1):4482.PubMedPubMedCentralCrossRef Yoshida K, et al. Exploring designability of electrostatic complementarity at an antigen-antibody interface directed by mutagenesis, biophysical analysis, and molecular dynamics simulations. Sci Rep. 2019;9(1):4482.PubMedPubMedCentralCrossRef
139.
go back to reference Xia Y, et al. A visible and colorimetric aptasensor based on DNA-capped single-walled carbon nanotubes for detection of exosomes. Biosens Bioelectron. 2017;92:8–15.PubMedCrossRef Xia Y, et al. A visible and colorimetric aptasensor based on DNA-capped single-walled carbon nanotubes for detection of exosomes. Biosens Bioelectron. 2017;92:8–15.PubMedCrossRef
140.
go back to reference Oliveira-Rodriguez M, et al. Development of a rapid lateral flow immunoassay test for detection of exosomes previously enriched from cell culture medium and body fluids. J Extracell Vesicles. 2016;5:31803.PubMedCrossRef Oliveira-Rodriguez M, et al. Development of a rapid lateral flow immunoassay test for detection of exosomes previously enriched from cell culture medium and body fluids. J Extracell Vesicles. 2016;5:31803.PubMedCrossRef
141.
go back to reference Chen X, et al. A paper-supported aptasensor based on upconversion luminescence resonance energy transfer for the accessible determination of exosomes. Biosens Bioelectron. 2018;102:582–8.PubMedCrossRef Chen X, et al. A paper-supported aptasensor based on upconversion luminescence resonance energy transfer for the accessible determination of exosomes. Biosens Bioelectron. 2018;102:582–8.PubMedCrossRef
142.
go back to reference He F, et al. Quantification of exosome based on a copper-mediated signal amplification strategy. Anal Chem. 2018;90(13):8072–9.PubMedCrossRef He F, et al. Quantification of exosome based on a copper-mediated signal amplification strategy. Anal Chem. 2018;90(13):8072–9.PubMedCrossRef
143.
144.
go back to reference Sina AA, et al. Label-free detection of exosomes using a surface plasmon resonance biosensor. Anal Bioanal Chem. 2019;411(7):1311–8.PubMedCrossRef Sina AA, et al. Label-free detection of exosomes using a surface plasmon resonance biosensor. Anal Bioanal Chem. 2019;411(7):1311–8.PubMedCrossRef
145.
go back to reference Kwizera EA, et al. Molecular detection and analysis of exosomes using surface-enhanced raman scattering gold nanorods and a miniaturized device. Theranostics. 2018;8(10):2722–38.PubMedPubMedCentralCrossRef Kwizera EA, et al. Molecular detection and analysis of exosomes using surface-enhanced raman scattering gold nanorods and a miniaturized device. Theranostics. 2018;8(10):2722–38.PubMedPubMedCentralCrossRef
146.
go back to reference Zong S, et al. SERS-fluorescence-superresolution triple-mode nanoprobe based on surface enhanced Raman scattering and surface enhanced fluorescence. J Mater Chem B. 2020;8(36):8459–66.PubMedCrossRef Zong S, et al. SERS-fluorescence-superresolution triple-mode nanoprobe based on surface enhanced Raman scattering and surface enhanced fluorescence. J Mater Chem B. 2020;8(36):8459–66.PubMedCrossRef
147.
go back to reference Wang Z, et al. Screening and multiple detection of cancer exosomes using an SERS-based method. Nanoscale. 2018;10(19):9053–62.PubMedCrossRef Wang Z, et al. Screening and multiple detection of cancer exosomes using an SERS-based method. Nanoscale. 2018;10(19):9053–62.PubMedCrossRef
148.
go back to reference Park J, et al. An integrated magneto-electrochemical device for the rapid profiling of tumour extracellular vesicles from blood plasma. Nat Biomed Eng. 2021;5(7):678–89.PubMedPubMedCentralCrossRef Park J, et al. An integrated magneto-electrochemical device for the rapid profiling of tumour extracellular vesicles from blood plasma. Nat Biomed Eng. 2021;5(7):678–89.PubMedPubMedCentralCrossRef
149.
go back to reference Song D, et al. Sandwich-type electrochemical immunosensor for CEA detection using magnetic hollow Ni/C@SiO(2) nanomatrix and boronic acid functionalized CPS@PANI@Au probe. Talanta. 2021;225: 122006.PubMedCrossRef Song D, et al. Sandwich-type electrochemical immunosensor for CEA detection using magnetic hollow Ni/C@SiO(2) nanomatrix and boronic acid functionalized CPS@PANI@Au probe. Talanta. 2021;225: 122006.PubMedCrossRef
150.
go back to reference Wang S, et al. Aptasensor with expanded nucleotide using dna nanotetrahedra for electrochemical detection of cancerous exosomes. ACS Nano. 2017;11(4):3943–9.PubMedPubMedCentralCrossRef Wang S, et al. Aptasensor with expanded nucleotide using dna nanotetrahedra for electrochemical detection of cancerous exosomes. ACS Nano. 2017;11(4):3943–9.PubMedPubMedCentralCrossRef
151.
go back to reference Azmi AS, Bao B, Sarkar FH. Exosomes in cancer development, metastasis, and drug resistance: a comprehensive review. Cancer Metastasis Rev. 2013;32(3–4):623–42.PubMedCrossRef Azmi AS, Bao B, Sarkar FH. Exosomes in cancer development, metastasis, and drug resistance: a comprehensive review. Cancer Metastasis Rev. 2013;32(3–4):623–42.PubMedCrossRef
152.
go back to reference Rahbarghazi R, et al. Tumor-derived extracellular vesicles: reliable tools for Cancer diagnosis and clinical applications. Cell Commun Signal. 2019;17(1):73.PubMedPubMedCentralCrossRef Rahbarghazi R, et al. Tumor-derived extracellular vesicles: reliable tools for Cancer diagnosis and clinical applications. Cell Commun Signal. 2019;17(1):73.PubMedPubMedCentralCrossRef
154.
go back to reference Filipazzi P, et al. Recent advances on the role of tumor exosomes in immunosuppression and disease progression. Semin Cancer Biol. 2012;22(4):342–9.PubMedCrossRef Filipazzi P, et al. Recent advances on the role of tumor exosomes in immunosuppression and disease progression. Semin Cancer Biol. 2012;22(4):342–9.PubMedCrossRef
155.
go back to reference Sun Y, Liu J. Potential of cancer cell-derived exosomes in clinical application: a review of recent research advances. Clin Ther. 2014;36(6):863–72.PubMedCrossRef Sun Y, Liu J. Potential of cancer cell-derived exosomes in clinical application: a review of recent research advances. Clin Ther. 2014;36(6):863–72.PubMedCrossRef
156.
go back to reference Langevin S, et al. Comprehensive microRNA-sequencing of exosomes derived from head and neck carcinoma cells in vitro reveals common secretion profiles and potential utility as salivary biomarkers. Oncotarget. 2017;8(47):82459–74.PubMedPubMedCentralCrossRef Langevin S, et al. Comprehensive microRNA-sequencing of exosomes derived from head and neck carcinoma cells in vitro reveals common secretion profiles and potential utility as salivary biomarkers. Oncotarget. 2017;8(47):82459–74.PubMedPubMedCentralCrossRef
158.
go back to reference An T, et al. Exosomes serve as tumour markers for personalized diagnostics owing to their important role in cancer metastasis. J Extracell Vesicles. 2015;4:27522.PubMedCrossRef An T, et al. Exosomes serve as tumour markers for personalized diagnostics owing to their important role in cancer metastasis. J Extracell Vesicles. 2015;4:27522.PubMedCrossRef
159.
go back to reference Cheng G. Circulating miRNAs: roles in cancer diagnosis, prognosis and therapy. Adv Drug Deliv Rev. 2015;81:75–93.PubMedCrossRef Cheng G. Circulating miRNAs: roles in cancer diagnosis, prognosis and therapy. Adv Drug Deliv Rev. 2015;81:75–93.PubMedCrossRef
160.
go back to reference Liao J, et al. Exosome-shuttling microRNA-21 promotes cell migration and invasion-targeting PDCD4 in esophageal cancer. Int J Oncol. 2016;48(6):2567–79.PubMedCrossRef Liao J, et al. Exosome-shuttling microRNA-21 promotes cell migration and invasion-targeting PDCD4 in esophageal cancer. Int J Oncol. 2016;48(6):2567–79.PubMedCrossRef
161.
162.
go back to reference Krishnan A, et al. Salivary exosomes: a theranostics secret of oral cancer—correspondence. Int J Surg. 2022;108: 106990.PubMedCrossRef Krishnan A, et al. Salivary exosomes: a theranostics secret of oral cancer—correspondence. Int J Surg. 2022;108: 106990.PubMedCrossRef
163.
go back to reference Tanaka Y, et al. Clinical impact of serum exosomal microRNA-21 as a clinical biomarker in human esophageal squamous cell carcinoma. Cancer. 2013;119(6):1159–67.PubMedCrossRef Tanaka Y, et al. Clinical impact of serum exosomal microRNA-21 as a clinical biomarker in human esophageal squamous cell carcinoma. Cancer. 2013;119(6):1159–67.PubMedCrossRef
165.
go back to reference Ghorbanmehr N, et al. miR-21-5p, miR-141-3p, and miR-205-5p levels in urine-promising biomarkers for the identification of prostate and bladder cancer. Prostate. 2019;79(1):88–95.PubMedCrossRef Ghorbanmehr N, et al. miR-21-5p, miR-141-3p, and miR-205-5p levels in urine-promising biomarkers for the identification of prostate and bladder cancer. Prostate. 2019;79(1):88–95.PubMedCrossRef
166.
go back to reference Lin H, et al. Urinary exosomal miRNAs as biomarkers of bladder Cancer and experimental verification of mechanism of miR-93-5p in bladder Cancer. BMC Cancer. 2021;21(1):1293.PubMedPubMedCentralCrossRef Lin H, et al. Urinary exosomal miRNAs as biomarkers of bladder Cancer and experimental verification of mechanism of miR-93-5p in bladder Cancer. BMC Cancer. 2021;21(1):1293.PubMedPubMedCentralCrossRef
167.
go back to reference Josson S, et al. Stromal fibroblast-derived miR-409 promotes epithelial-to-mesenchymal transition and prostate tumorigenesis. Oncogene. 2015;34(21):2690–9.PubMedCrossRef Josson S, et al. Stromal fibroblast-derived miR-409 promotes epithelial-to-mesenchymal transition and prostate tumorigenesis. Oncogene. 2015;34(21):2690–9.PubMedCrossRef
168.
go back to reference Wolfers J, et al. Tumor-derived exosomes are a source of shared tumor rejection antigens for CTL cross-priming. Nat Med. 2001;7(3):297–303.PubMedCrossRef Wolfers J, et al. Tumor-derived exosomes are a source of shared tumor rejection antigens for CTL cross-priming. Nat Med. 2001;7(3):297–303.PubMedCrossRef
169.
go back to reference Verdi J, et al. Development and clinical application of tumor-derived exosomes in patients with cancer. Curr Stem Cell Res Ther. 2022;17(1):91–102.PubMedCrossRef Verdi J, et al. Development and clinical application of tumor-derived exosomes in patients with cancer. Curr Stem Cell Res Ther. 2022;17(1):91–102.PubMedCrossRef
170.
go back to reference Farhood B, Najafi M, Mortezaee K. CD8(+) cytotoxic T lymphocytes in cancer immunotherapy: a review. J Cell Physiol. 2019;234(6):8509–21.PubMedCrossRef Farhood B, Najafi M, Mortezaee K. CD8(+) cytotoxic T lymphocytes in cancer immunotherapy: a review. J Cell Physiol. 2019;234(6):8509–21.PubMedCrossRef
171.
go back to reference Buchbinder EI, Desai A. CTLA-4 and PD-1 pathways: similarities, differences, and implications of their inhibition. Am J Clin Oncol. 2016;39(1):98–106.PubMedPubMedCentralCrossRef Buchbinder EI, Desai A. CTLA-4 and PD-1 pathways: similarities, differences, and implications of their inhibition. Am J Clin Oncol. 2016;39(1):98–106.PubMedPubMedCentralCrossRef
172.
go back to reference Alsaab HO, et al. PD-1 and PD-L1 checkpoint signaling inhibition for cancer immunotherapy: mechanism, combinations, and clinical outcome. Front Pharmacol. 2017;8:561.PubMedPubMedCentralCrossRef Alsaab HO, et al. PD-1 and PD-L1 checkpoint signaling inhibition for cancer immunotherapy: mechanism, combinations, and clinical outcome. Front Pharmacol. 2017;8:561.PubMedPubMedCentralCrossRef
173.
go back to reference Cheng L, et al. Exosomes from melatonin treated hepatocellularcarcinoma cells alter the immunosupression status through STAT3 pathway in macrophages. Int J Biol Sci. 2017;13(6):723–34.PubMedPubMedCentralCrossRef Cheng L, et al. Exosomes from melatonin treated hepatocellularcarcinoma cells alter the immunosupression status through STAT3 pathway in macrophages. Int J Biol Sci. 2017;13(6):723–34.PubMedPubMedCentralCrossRef
175.
go back to reference Hou W, et al. A novel tetravalent bispecific antibody targeting programmed death 1 and tyrosine-protein kinase Met for treatment of gastric cancer. Invest New Drugs. 2019;37(5):876–89.PubMedCrossRef Hou W, et al. A novel tetravalent bispecific antibody targeting programmed death 1 and tyrosine-protein kinase Met for treatment of gastric cancer. Invest New Drugs. 2019;37(5):876–89.PubMedCrossRef
177.
go back to reference Gao J, et al. Expression profiles and clinical value of plasma exosomal Tim-3 and Galectin-9 in non-small cell lung cancer. Biochem Biophys Res Commun. 2018;498(3):409–15.PubMedCrossRef Gao J, et al. Expression profiles and clinical value of plasma exosomal Tim-3 and Galectin-9 in non-small cell lung cancer. Biochem Biophys Res Commun. 2018;498(3):409–15.PubMedCrossRef
178.
go back to reference Gu X, et al. Improved vaccine efficacy of tumor exosome compared to tumor lysate loaded dendritic cells in mice. Int J Cancer. 2015;136(4):E74-84.PubMedCrossRef Gu X, et al. Improved vaccine efficacy of tumor exosome compared to tumor lysate loaded dendritic cells in mice. Int J Cancer. 2015;136(4):E74-84.PubMedCrossRef
179.
go back to reference Rezaei R, et al. Tumor-derived exosomes enriched by miRNA-124 promote anti-tumor immune response in CT-26 tumor-bearing mice. Front Med (Lausanne). 2021;8: 619939.PubMedPubMedCentralCrossRef Rezaei R, et al. Tumor-derived exosomes enriched by miRNA-124 promote anti-tumor immune response in CT-26 tumor-bearing mice. Front Med (Lausanne). 2021;8: 619939.PubMedPubMedCentralCrossRef
180.
go back to reference Batista IA, Melo SA Exosomes and the future of immunotherapy in pancreatic cancer. Int J Mol Sci, 2019. 20(3) Batista IA, Melo SA Exosomes and the future of immunotherapy in pancreatic cancer. Int J Mol Sci, 2019. 20(3)
182.
go back to reference Dhar R, et al. Exosome-based cancer vaccine: a cutting-edge approach—correspondence. Int J Surg. 2022;108: 106993.PubMedCrossRef Dhar R, et al. Exosome-based cancer vaccine: a cutting-edge approach—correspondence. Int J Surg. 2022;108: 106993.PubMedCrossRef
183.
go back to reference Chen YS, et al. Exosomes in clinical trial and their production in compliance with good manufacturing practice. Ci Ji Yi Xue Za Zhi. 2019;32(2):113–20.PubMed Chen YS, et al. Exosomes in clinical trial and their production in compliance with good manufacturing practice. Ci Ji Yi Xue Za Zhi. 2019;32(2):113–20.PubMed
185.
go back to reference Huang Y, et al. Exosomes function in tumor immune microenvironment. Adv Exp Med Biol. 2018;1056:109–22.PubMedCrossRef Huang Y, et al. Exosomes function in tumor immune microenvironment. Adv Exp Med Biol. 2018;1056:109–22.PubMedCrossRef
186.
go back to reference Rezaie J, Feghhi M, Etemadi T. A review on exosomes application in clinical trials: perspective, questions, and challenges. Cell Commun Signal. 2022;20(1):145.PubMedPubMedCentralCrossRef Rezaie J, Feghhi M, Etemadi T. A review on exosomes application in clinical trials: perspective, questions, and challenges. Cell Commun Signal. 2022;20(1):145.PubMedPubMedCentralCrossRef
188.
go back to reference Brinton LT, et al. Formation and role of exosomes in cancer. Cell Mol Life Sci. 2015;72(4):659–71.PubMedCrossRef Brinton LT, et al. Formation and role of exosomes in cancer. Cell Mol Life Sci. 2015;72(4):659–71.PubMedCrossRef
189.
190.
go back to reference Dhar R et al. Decoding of exosome heterogeneity for cancer theranostics. Clin Transl Med. 2023;e1288. Dhar R et al. Decoding of exosome heterogeneity for cancer theranostics. Clin Transl Med. 2023;e1288.
191.
go back to reference Gould SJ, Raposo G As we wait: coping with an imperfect nomenclature for extracellular vesicles. J Extracell Vesicles, 2013. 2 Gould SJ, Raposo G As we wait: coping with an imperfect nomenclature for extracellular vesicles. J Extracell Vesicles, 2013. 2
193.
go back to reference Harding C, et al. Receptor-mediated endocytosis of transferrin and recycling of the transferrin receptor in rat reticulocytes. J Cell Biol. 1983;97(2):329–39.PubMedCrossRef Harding C, et al. Receptor-mediated endocytosis of transferrin and recycling of the transferrin receptor in rat reticulocytes. J Cell Biol. 1983;97(2):329–39.PubMedCrossRef
194.
go back to reference Martins VR, et al. Tumor-cell-derived microvesicles as carriers of molecular information in cancer. Curr Opin Oncol. 2013;25(1):66–75.PubMedCrossRef Martins VR, et al. Tumor-cell-derived microvesicles as carriers of molecular information in cancer. Curr Opin Oncol. 2013;25(1):66–75.PubMedCrossRef
195.
go back to reference Kar R, et al, Exosome-based smart drug delivery tool for cancer theranostics. ACS Biomater Sci Eng. 2023. Kar R, et al, Exosome-based smart drug delivery tool for cancer theranostics. ACS Biomater Sci Eng. 2023.
196.
go back to reference Wilson DH, et al. The simoa HD-1 analyzer: a novel fully automated digital immunoassay analyzer with single-molecule sensitivity and multiplexing. J Lab Autom. 2016;21(4):533–47.PubMedCrossRef Wilson DH, et al. The simoa HD-1 analyzer: a novel fully automated digital immunoassay analyzer with single-molecule sensitivity and multiplexing. J Lab Autom. 2016;21(4):533–47.PubMedCrossRef
197.
go back to reference Dhar R, et al. Exosome: a megastar of future cancer personalized and precision medicine. Clin Transl Disc. 2023;3: e208.CrossRef Dhar R, et al. Exosome: a megastar of future cancer personalized and precision medicine. Clin Transl Disc. 2023;3: e208.CrossRef
198.
go back to reference Huda MN, et al. Potential use of exosomes as diagnostic biomarkers and in targeted drug delivery: progress in clinical and preclinical applications. ACS Biomater Sci Eng. 2021;7(6):2106–49.PubMedPubMedCentralCrossRef Huda MN, et al. Potential use of exosomes as diagnostic biomarkers and in targeted drug delivery: progress in clinical and preclinical applications. ACS Biomater Sci Eng. 2021;7(6):2106–49.PubMedPubMedCentralCrossRef
Metadata
Title
Theranostic signature of tumor-derived exosomes in cancer
Authors
Samruti Kumar
Rajib Dhar
Lokesh Babu Sirkali Suresh Kumar
Gauresh Gurudas Shivji
Rama Jayaraj
Arikketh Devi
Publication date
01-11-2023
Publisher
Springer US
Published in
Medical Oncology / Issue 11/2023
Print ISSN: 1357-0560
Electronic ISSN: 1559-131X
DOI
https://doi.org/10.1007/s12032-023-02176-6

Other articles of this Issue 11/2023

Medical Oncology 11/2023 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.