Next Article in Journal
The Calcium-Dependent Protein Kinase TaCDPK7 Positively Regulates Wheat Resistance to Puccinia striiformis f. sp. tritici
Previous Article in Journal
Temporal Quantitative Proteomic and Phosphoproteomic Profiling of SH-SY5Y and IMR-32 Neuroblastoma Cells during All-Trans-Retinoic Acid-Induced Neuronal Differentiation
Previous Article in Special Issue
Damage-Associated Molecular Patterns, a Class of Potential Psoriasis Drug Targets
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

The Diagnostic and Prognostic Value of Plasmatic Exosome Count in Cancer Patients and in Patients with Other Pathologies

by
Stefano Fais
1,2,* and
Mariantonia Logozzi
1,*
1
Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy
2
ExoLab Italia, Tecnopolo d’Abruzzo, 67100 L’Aquila, Italy
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(2), 1049; https://doi.org/10.3390/ijms25021049
Submission received: 6 December 2023 / Revised: 3 January 2024 / Accepted: 5 January 2024 / Published: 15 January 2024
The extent of both scientific articles and reviews on extracellular vesicles (EVs) has grown impressively over the last few decades. The publications cover investigations of various kinds of EVs, from human EVs to animal and plant-derived EVs. A high degree of effort has been spent in proposing EVs, mostly those of a nanosized (i.e., exosomes), as a natural source of new disease biomarkers. It is a known fact that both normal and tumor cells release exosomes; after their paracrine release, exosomes are spilled over into the blood, thus circulating through the organism, often ending in organs and compartments far from the site of the original release. The first evidence of this phenomenon is that exosomes may be detected, characterized and quantified in plasma samples of both healthy individuals and tumor patients. Due to their size (30–150 nm), exosomes are invisible particles that may be analyzed through both electron microscopy and other techniques that hijack the peculiar make-up of these nanovesicles. Exosomes express on the surface markers of intracellular vesicles (e.g., endosomes, lysosomes, phagosomes) together with plasma membrane molecules, through which the cellular source of exosomes may be recognized. This peculiarity is due to the various processes of exosome generation, and is particularly due to the multivesicular body (MVB) formation, which is a process of repeated rounds of internal vesicle fusion that involves the plasma membrane as well [1,2,3,4,5]. This process causes exosomes to express an array of molecules (e.g., Tsg101, Alix, CD63, CD9, CD81, HSP-70, Rab5), rendering these nanovesicles phenotypically recognizable. In fact, all the above molecules have been exploited to set up immunocapture-based techniques that have allowed for exosome characterization and quantification [6]. The first clinical study, performed in 148 individuals, was exclusively based on the use of an immunocapture-based ELISA test, through which it was shown that melanoma patients had significantly higher CD63+ plasmatic exosomes compared to healthy individuals, but significantly higher Cav-1 positive exosomes as well, where Cav-1 is considered a surrogate tumor biomarker [7]. However, for the first time, this study supported a new finding that could play a key role in the future clinical management of tumors: melanoma patients present higher exosome levels in their plasma as compared to healthy individuals. In the same study, a preclinical investigation showed that higher plasmatic levels of exosomes correlated with the tumor mass [7]. The in vivo study was also supported by a series of reports showing that the microenvironmental acidity of tumors could exert a key role in determining an increased tumor exosome spill over into the blood stream inasmuch as, in vitro, a low pH condition induces an extensive exosome release by human tumor cells, independently from their histologies [8,9]. The increased low pH-dependent exosome release was consistent with both an increased expression of known tumor biomarkers (e.g., PSA) and a reduced exosome size [8]. In the same study, it was shown that the increased exosome release in acidic conditions correlated to the high plasmatic exosome levels as compared to controls [8]. It appears conceivable that the pH-dependent increase in exosome release may be dependent on one of the functions of the EVs, that is, to scavenge potentially toxic molecules as it has been shown for chemotherapeutics in tumor cells [10] and gold nanoparticles in human normal macrophages [11]. An interesting observation is that both chemical molecules and nanoparticles are released into the exosomes in their native/active form, further supporting the natural ability of exosomes to deliver functional molecules [10,11]. This advantage also includes the ability to deliver functional molecules with a full enzymatic function [12] and a cargo of protons [13]. A fascinating new issue is that exosomes deliver the pathologic prion protein (PrPC) as well [14], which is a glycoprotein anchored to the cell surface by glycosylphosphatidylinositol (GPI). Recent findings have shown the ectopic expression of PrPC in various cancers, including gastric, melanoma, breast, colorectal, pancreatic as well as rare cancers, where PrPC promotes cellular migration and invasion, tumor growth and metastasis [15]. Another topic of interest is that PrPC is delivered by exosomes in a model of prion-infected rodents and PrPC-associated exosomes can be purified in the plasma of the infected animals [15], suggesting that, in general, the identification of PrPC-associated proteins in the plasma of either tumor patients or patients with neurodegenerative diseases may represent a new valuable disease marker.
However, in the last decade, a new technique called nanoparticle tracking analysis (NTA), which is used to determine exosome number and size in samples from both cell culture supernatants and body fluids, has come to support data obtained with immunocapture-based ELISA [16,17]. The NTA analyzes particles ranging from 30 nm to 400–500 nm, thus allowing for the distinguishing of nanovesicles from microvesicles. To date, NTA remains to be considered as the most reliable technique to analyze a mixed population of submicroscopical vesicles in human body fluids. A previous study compared the results obtained from NTA, immunocapture-based ELISA, and nanoscale flow cytometry in exosome preparations obtained from either cell culture supernatants or plasma samples [8]. The results clearly revealed a complete overlapping between the three techniques; however, the immunocapture-based ELISA was incapable of providing information on the EVs’ size, while the nanoscale flow cytometry allowed for the gating of EVs ranging from 100 to 300 nm. Nonetheless, the implementation of these techniques provided valuable information on the number, size, distribution and the phenotyping of EVs from a plasma sample of both tumor patients and either healthy or disease controls. On the basis of these preliminary results, the NTA was performed in plasma samples of patients with prostate cancer and were compared to healthy donors. The results clearly showed that prostate cancer patients had significantly higher exosome levels compared to healthy donors [16], thus strongly supporting the data obtained in the preliminary study [9]. An independent study performed in patients with glioblastoma reported comparable results in displaying higher exosome levels in the plasma of glioblastoma patients [17]. More recently, a longitudinal study performed using the NTA in patients with oral cancers has shown that high plasmatic levels of exosomes may be predictive of a recurrence after surgical treatment [18], supporting a previous investigation which revealed different exosome counts before and after surgical treatment [19].
The importance of these findings is increased by the growing evidence that EVs—particularly exosomes—have not been shown to deliver molecules with a tumor specificity despite being a potential source of disease biomarkers. Glypican-1 is a clear example; in fact, while it has been proposed to be a specific marker of pancreatic cancer, it has displayed a high level of expression in exosome samples obtained in plasma from patients with other cancers [20]. Nonetheless, it may be of some help when performed in combination with well-known tumor biomarkers [21,22]. It has in fact been demonstrated that plasmatic exosomes express high levels of acknowledged tumor markers such as PSA, which distinguishes prostate cancer from both healthy and inflammatory states [23]. Comparable studies should also be performed for other acknowledged tumor markers that have been demonstrated to be delivered by plasmatic exosomes (e.g., CEA) [24]. However, the increased exosome plasmatic levels have a double importance in clinical oncology. In fact, growing scientific evidence supports a key role of exosomes in tumor metastasis, [25,26,27]; on the other hand, the involvement in tumor metastasis increases the importance of exosome count in the plasma of tumor patients in further refining a prognostic evaluation. The circulating mass of tumor exosomes may represent a real danger for the patients’ body with regard to their potential to generate metastasis. However, it has been proposed that exosomes may shape the tumor microenvironment with different underlying mechanisms, depending on the exosome cargo [28]. Exosomes may be secreted within a tissue and can be found in the plasma, but they can also be released in many other biological fluids [29,30,31,32,33,34,35]. This means that the same approach of establishing differences in exosome counts between healthy individuals and those inflicted with a disease may be investigated using other body fluids.
There is also evidence that exosomes may actively contribute to the continuous genome remodeling during our lifetime. In fact, it has been shown that exosomes containing a reporter gene are released in vivo, circulating through blood and transferring the spermatozoa into the gonads, with a possible transfer of the acquired gene to the progeny [36].
The fact is that exosomes are considered a natural source of disease biomarkers [37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63]. The future goal of translational oncology is and will be to define the molecules’ cargo of body fluid-derived exosomes in tumor patients, also based on the evidence that tumor-released exosomes are involved in both tumor progression and metastasis [25,26,27,28,29,64]. However, to date, the data supporting the use of exosomes to identify new and valuable disease biomarkers have been below par. Several unexpected but interesting findings propose the simple measurement of exosome plasmatic levels as a key prognostic value [65,66,67]. The data also suggest that the increased exosome blood count is a hallmark of tumor patients as it is a common finding, regardless of tissue specificity [7,16,17,18,19].

Conclusions

An intriguing paper has introduced a new term “Vesiclemia” [68], which means the presence of measurable plasmatic levels of extracellular vesicles in tumor patients. This paper adds further support to a new strategy in the follow-up of cancer patients that will take into careful account the plasmatic EV cargo, rather than the potential biomarkers’ cargo. However, the extracellular vesicle count appears to have potential significance in disease conditions other than tumors. In fact, recently, extracellular vesicle count has been proposed as a valuable new tool in infectious diseases [69]. Similar findings were reported in postmenopausal women taking hormonal replacement therapy [70]. Of course, the other disease conditions need clinical validation at least comparable to what we have to date for tumor patients. This editorial has emphasized a bulk of clinical results supporting the use of a “plasmatic exosome count” as a new valuable tool in the follow-up of tumor patients [7,17,18,19,20]. The plasmatic exosome count may be implemented by analyzing other components, including: (i) the exosome size (that has been proven to be smaller in tumor patients than in controls) [16,67]; (ii) the expression of known tumor markers [23,24,25] and (iii) the intraluminal pH of circulating exosomes [13].
Multicenter clinical studies are of course mandatory in order to validate the existing data in higher patient numbers. However, to perform longitudinal studies in patients undergoing either surgical or/and medical treatment is also mandatory [18], with the aim to use the plasmatic exosome count as a new tool in the clinical follow-up of cancer patients. Another important point is to extend the exosome count to other body fluids, with the purpose to limit unnecessary invasive procedures and reduce public health costs. The existing data on urine and other body fluids are very promising [29,30,31,32,33,34,35], but with limited data on the exosome count. A key series of data has shown that a major cause of the increased exosome release from tumors is the microenvironmental acidity [71]. Additionally, considering and deliberating on new anti-tumor therapies targeted to the tumor microenvironment rather than tumor cells [72] may lead to a reduced exosome release with a reduced risk of tumor metastasis [24,25,26,27].

Author Contributions

S.F., conceptualization, writing—original draft preparation; writing—review and editing, supervision; M.L., conceptualization, writing—original draft preparation; writing—review and editing, supervision. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. 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] [PubMed]
  2. Zhang, Y.; Liu, Y.; Liu, H.; Tang, W.H. Exosomes: Biogenesis, Biologic Function and Clinical Potential. Cell Biosci. 2019, 9, 19. [Google Scholar] [CrossRef] [PubMed]
  3. Cocucci, E.; Meldolesi, J. Ectosomes and Exosomes: Shedding the Confusion between Extracellular Vesicles. Trends Cell Biol. 2015, 25, 364–372. [Google Scholar] [CrossRef] [PubMed]
  4. Johnstone, R.M.; Adam, M.; Hammond, J.R.; Orr, L.; Turbide, C. Vesicle Formation during Reticulocyte Maturation. Association of Plasma Membrane Activities with Released Vesicles (Exosomes). J. Biol. Chem. 1987, 262, 9412–9420. [Google Scholar] [CrossRef] [PubMed]
  5. Bobrie, A.; Colombo, M.; Raposo, G.; Théry, C. Exosome Secretion: Molecular Mechanisms and Roles in Immune Responses. Traffic 2011, 12, 1659–1668. [Google Scholar] [CrossRef]
  6. Logozzi, M.; Di Raimo, R.; Mizzoni, D.; Fais, S. Immunocapture-Based ELISA to Characterize and Quantify Exosomes in Both Cell Culture Supernatants and Body Fluids. Methods Enzymol. 2020, 645, 155–180. [Google Scholar] [CrossRef]
  7. Logozzi, M.; De Milito, A.; Lugini, L.; Borghi, M.; Calabrò, L.; Spada, M.; Perdicchio, M.; Marino, M.L.; Federici, C.; Iessi, E.; et al. High Levels of Exosomes Expressing CD63 and Caveolin-1 in Plasma of Melanoma Patients. PLoS ONE 2009, 4, e5219. [Google Scholar] [CrossRef]
  8. Logozzi, M.; Angelini, D.F.; Iessi, E.; Mizzoni, D.; Di Raimo, R.; Federici, C.; Lugini, L.; Borsellino, G.; Gentilucci, A.; Pierella, F.; et al. Increased PSA Expression on Prostate Cancer Exosomes in in Vitro Condition and in Cancer Patients. Cancer Lett. 2017, 403, 318–329. [Google Scholar] [CrossRef]
  9. Logozzi, M.; Mizzoni, D.; Angelini, D.; Di Raimo, R.; Falchi, M.; Battistini, L.; Fais, S. Microenvironmental PH and Exosome Levels Interplay in Human Cancer Cell Lines of Different Histotypes. Cancers 2018, 10, 370. [Google Scholar] [CrossRef]
  10. Federici, C.; Petrucci, F.; Caimi, S.; Cesolini, A.; Logozzi, M.; Borghi, M.; D’Ilio, S.; Lugini, L.; Violante, N.; Azzarito, T.; et al. Exosome Release and Low PH Belong to a Framework of Resistance of Human Melanoma Cells to Cisplatin. PLoS ONE 2014, 9, e88193. [Google Scholar] [CrossRef]
  11. Logozzi, M.; Mizzoni, D.; Bocca, B.; Di Raimo, R.; Petrucci, F.; Caimi, S.; Alimonti, A.; Falchi, M.; Cappello, F.; Campanella, C.; et al. Human Primary Macrophages Scavenge AuNPs and Eliminate It through Exosomes. A Natural Shuttling for Nanomaterials. Eur. J. Pharm. Biopharm. 2019, 137, 23–36. [Google Scholar] [CrossRef] [PubMed]
  12. Logozzi, M.; Capasso, C.; Di Raimo, R.; Del Prete, S.; Mizzoni, D.; Falchi, M.; Supuran, C.T.; Fais, S. Prostate Cancer Cells and Exosomes in Acidic Condition Show Increased Carbonic Anhydrase IX Expression and Activity. J. Enzym. Inhib. Med. Chem. 2019, 34, 272–278. [Google Scholar] [CrossRef]
  13. Logozzi, M.; Mizzoni, D.; Capasso, C.; Del Prete, S.; Di Raimo, R.; Falchi, M.; Angelini, D.F.; Sciarra, A.; Maggi, M.; Supuran, C.T.; et al. Plasmatic Exosomes from Prostate Cancer Patients Show Increased Carbonic Anhydrase IX Expression and Activity and Low PH. J. Enzym. Inhib. Med. Chem. 2020, 35, 280–288. [Google Scholar] [CrossRef] [PubMed]
  14. Properzi, F.; Logozzi, M.; Abdel-Haq, H.; Federici, C.; Lugini, L.; Azzarito, T.; Cristofaro, I.; di Sevo, D.; Ferroni, E.; Cardone, F.; et al. Detection of exosomal prions in blood by immunochemistry techniques. J. Gen. Virol. 2015, 96, 1969–1974. [Google Scholar] [CrossRef]
  15. Abi Nahed, R.; Safwan-Zaiter, H.; Gemy, K.; Lyko, C.; Boudaud, M.; Desseux, M.; Marquette, C.; Barjat, T.; Alfaidy, N.; Benharouga, M. The Multifaceted Functions of Prion Protein (PrPC) in Cancer. Cancers 2023, 15, 4982. [Google Scholar] [CrossRef] [PubMed]
  16. Logozzi, M.; Mizzoni, D.; Di Raimo, R.; Giuliani, A.; Maggi, M.; Sciarra, A.; Fais, S. Plasmatic Exosome Number and Size Distinguish Prostate Cancer Patients from Healthy Individuals: A Prospective Clinical Study. Front. Oncol. 2021, 11, 727317. [Google Scholar] [CrossRef] [PubMed]
  17. Osti, D.; Del Bene, M.; Rappa, G.; Santos, M.; Matafora, V.; Richichi, C.; Faletti, S.; Beznoussenko, G.V.; Mironov, A.; Bachi, A.; et al. Clinical Significance of Extracellular Vesicles in Plasma from Glioblastoma Patients. Clin. Cancer Res. 2019, 25, 266–276. [Google Scholar] [CrossRef]
  18. Rodríguez-Zorrilla, S.; Lorenzo-Pouso, A.I.; Fais, S.; Logozzi, M.; Mizzoni, D.; Di Raimo, R.; Giuliani, A.; García-García, A.; Pérez-Jardón, A.; Ortega, K.L.; et al. Increased Plasmatic Levels of Exosomes Are Significantly Related to Relapse Rate in Patients with Oral Squamous Cell Carcinoma: A Cohort Study. Cancers 2023, 15, 5693. [Google Scholar] [CrossRef]
  19. Rodríguez Zorrilla, S.; Pérez-Sayans, M.; Fais, S.; Logozzi, M.; Gallas Torreira, M.; García García, A. A Pilot Clinical Study on the Prognostic Relevance of Plasmatic Exosomes Levels in Oral Squamous Cell Carcinoma Patients. Cancers 2019, 11, 429. [Google Scholar] [CrossRef]
  20. Melo, S.A.; Luecke, L.B.; Kahlert, C.; Fernandez, A.F.; Gammon, S.T.; Kaye, J.; LeBleu, V.S.; Mittendorf, E.A.; Weitz, J.; Rahbari, N.; et al. Glypican-1 Identifies Cancer Exosomes and Detects Early Pancreatic Cancer. Nature 2015, 523, 177–182. [Google Scholar] [CrossRef]
  21. Buscail, E.; Chauvet, A.; Quincy, P.; Degrandi, O.; Buscail, C.; Lamrissi, I.; Moranvillier, I.; Caumont, C.; Verdon, S.; Brisson, A.; et al. CD63-GPC1-Positive Exosomes Coupled with CA19-9 Offer Good Diagnostic Potential for Resectable Pancreatic Ductal Adenocarcinoma. Transl. Oncol. 2019, 12, 1395–1403. [Google Scholar] [CrossRef]
  22. Lai, X.; Wang, M.; McElyea, S.D.; Sherman, S.; House, M.; Korc, M. A MicroRNA Signature in Circulating Exosomes Is Superior to Exosomal Glypican-1 Levels for Diagnosing Pancreatic Cancer. Cancer Lett. 2017, 393, 86–93. [Google Scholar] [CrossRef] [PubMed]
  23. Logozzi, M.; Angelini, D.F.; Giuliani, A.; Mizzoni, D.; Di Raimo, R.; Maggi, M.; Gentilucci, A.; Marzio, V.; Salciccia, S.; Borsellino, G.; et al. Increased Plasmatic Levels of PSA-Expressing Exosomes Distinguish Prostate Cancer Patients from Benign Prostatic Hyperplasia: A Prospective Study. Cancers 2019, 11, 1449. [Google Scholar] [CrossRef] [PubMed]
  24. Yokoyama, S.; Takeuchi, A.; Yamaguchi, S.; Mitani, Y.; Watanabe, T.; Matsuda, K.; Hotta, T.; Shively, J.E.; Yamaue, H. Clinical Implications of Carcinoembryonic Antigen Distribution in Serum Exosomal Fraction-Measurement by ELISA. PLoS ONE 2017, 12, e0183337. [Google Scholar] [CrossRef]
  25. 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]
  26. Peinado, H.; Zhang, H.; Matei, I.R.; Costa-Silva, B.; Hoshino, A.; Rodrigues, G.; Psaila, B.; Kaplan, R.N.; Bromberg, J.F.; Kang, Y.; et al. Pre-Metastatic Niches: Organ-Specific Homes for Metastases. Nat. Rev. Cancer 2017, 17, 302–317. [Google Scholar] [CrossRef] [PubMed]
  27. Lugini, L.; Valtieri, M.; Federici, C.; Cecchetti, S.; Meschini, S.; Condello, M.; Signore, M.; Fais, S. Exosomes from Human Colorectal Cancer Induce a Tumor-like Behavior in Colonic Mesenchymal Stromal Cells. Oncotarget 2016, 7, 50086–50098. [Google Scholar] [CrossRef]
  28. Tan, S.; Yang, Y.; Yang, W.; Han, Y.; Huang, L.; Yang, R.; Hu, Z.; Tao, Y.; Liu, L.; Li, Y.; et al. Exosomal cargos-mediated metabolic reprogramming in tumor microenvironment. J. Exp. Clin. Cancer Res. 2023, 42, 59. [Google Scholar] [CrossRef]
  29. Sakaue, T.; Koga, H.; Iwamoto, H.; Nakamura, T.; Ikezono, Y.; Abe, M.; Wada, F.; Masuda, A.; Tanaka, T.; Fukahori, M.; et al. Glycosylation of Ascites-Derived Exosomal CD133: A Potential Prognostic Biomarker in Patients with Advanced Pancreatic Cancer. Med. Mol. Morphol. 2019, 52, 198–208. [Google Scholar] [CrossRef]
  30. García-Flores, M.; Sánchez-López, C.M.; Ramírez-Calvo, M.; Fernández-Serra, A.; Marcilla, A.; López-Guerrero, J.A. Isolation and Characterization of Urine Microvesicles from Prostate Cancer Patients: Different Approaches, Different Visions. BMC Urol. 2021, 21, 137. [Google Scholar] [CrossRef]
  31. Xu, Y.; Lou, J.; Yu, M.; Jiang, Y.; Xu, H.; Huang, Y.; Gao, Y.; Wang, H.; Li, G.; Wang, Z.; et al. Urinary Exosomes Diagnosis of Urological Tumors: A Systematic Review and Meta-Analysis. Front. Oncol. 2021, 11, 734587. [Google Scholar] [CrossRef] [PubMed]
  32. Wei, C.; Chen, X.; Ji, J.; Xu, Y.; He, X.; Zhang, H.; Mo, Z.; Wang, F. Urinary Exosomal Prostate-specific Antigen Is a Noninvasive Biomarker to Detect Prostate Cancer: Not Only Old Wine in New Bottles. Int. J. Cancer 2023, 152, 1719–1727. [Google Scholar] [CrossRef] [PubMed]
  33. Hiltbrunner, S.; Mints, M.; Eldh, M.; Rosenblatt, R.; Holmström, B.; Alamdari, F.; Johansson, M.; Veerman, R.E.; Winqvist, O.; Sherif, A.; et al. Urinary Exosomes from Bladder Cancer Patients Show a Residual Cancer Phenotype despite Complete Pathological Downstaging. Sci. Rep. 2020, 10, 5960. [Google Scholar] [CrossRef] [PubMed]
  34. Yan, I.K.; Berdah, V.X.; Patel, T. Isolation of Extracellular RNA from Bile. Methods Mol. Biol. 2018, 1740, 59–67. [Google Scholar] [CrossRef] [PubMed]
  35. Foglio, E.; Puddighinu, G.; Fasanaro, P.; D’Arcangelo, D.; Perrone, G.A.; Mocini, D.; Campanella, C.; Coppola, L.; Logozzi, M.; Azzarito, T.; et al. Exosomal clusterin, identified in the pericardial fluid, improves myocardial performance following MI through epicardial activation, enhanced arteriogenesis and reduced apoptosis. Int. J. Cardiol. 2015, 197, 333–347. [Google Scholar] [CrossRef] [PubMed]
  36. Cossetti, C.; Lugini, L.; Astrologo, L.; Saggio, I.; Fais, S.; Spadafora, C. Soma-to-Germline Transmission of RNA in Mice Xenografted with Human Tumour Cells: Possible Transport by Exosomes. PLoS ONE 2014, 9, e101629. [Google Scholar] [CrossRef] [PubMed]
  37. van der Pol, E.; Böing, A.N.; Harrison, P.; Sturk, A.; Nieuwland, R. Classification, Functions, and Clinical Relevance of Extracellular Vesicles. Pharmacol. Rev. 2012, 64, 676–705. [Google Scholar] [CrossRef] [PubMed]
  38. Canitano, A.; Venturi, G.; Borghi, M.; Ammendolia, M.G.; Fais, S. Exosomes Released in Vitro from Epstein-Barr Virus (EBV)-Infected Cells Contain EBV-Encoded Latent Phase MRNAs. Cancer Lett. 2013, 337, 193–199. [Google Scholar] [CrossRef]
  39. Li, S.; Yi, M.; Dong, B.; Tan, X.; Luo, S.; Wu, K. The Role of Exosomes in Liquid Biopsy for Cancer Diagnosis and Prognosis Prediction. Int. J. Cancer 2021, 148, 2640–2651. [Google Scholar] [CrossRef]
  40. Soung, Y.H.; Ford, S.; Zhang, V.; Chung, J. Exosomes in Cancer Diagnostics. Cancers 2017, 9, 8. [Google Scholar] [CrossRef]
  41. Huang, T.; Deng, C.-X. Current Progresses of Exosomes as Cancer Diagnostic and Prognostic Biomarkers. Int. J. Biol. Sci. 2019, 15, 1–11. [Google Scholar] [CrossRef] [PubMed]
  42. Meng, Y.; Sun, J.; Wang, X.; Hu, T.; Ma, Y.; Kong, C.; Piao, H.; Yu, T.; Zhang, G. Exosomes: A Promising Avenue for the Diagnosis of Breast Cancer. Technol. Cancer Res. Treat. 2019, 18, 1533033818821421. [Google Scholar] [CrossRef] [PubMed]
  43. Sumrin, A.; Moazzam, S.; Khan, A.A.; Ramzan, I.; Batool, Z.; Kaleem, S.; Ali, M.; Bashir, H.; Bilal, M. Exosomes as Biomarker of Cancer. Braz. Arch. Biol. Technol. 2018, 61. [Google Scholar] [CrossRef]
  44. Campanella, C.; Rappa, F.; Sciumè, C.; Marino Gammazza, A.; Barone, R.; Bucchieri, F.; David, S.; Curcurù, G.; Caruso Bavisotto, C.; Pitruzzella, A.; et al. Heat Shock Protein 60 Levels in Tissue and Circulating Exosomes in Human Large Bowel Cancer before and after Ablative Surgery. Cancer 2015, 121, 3230–3239. [Google Scholar] [CrossRef] [PubMed]
  45. Biggs, C.N.; Siddiqui, K.M.; Al-Zahrani, A.A.; Pardhan, S.; Brett, S.I.; Guo, Q.Q.; Yang, J.; Wolf, P.; Power, N.E.; Durfee, P.N.; et al. Prostate Extracellular Vesicles in Patient Plasma as a Liquid Biopsy Platform for Prostate Cancer Using Nanoscale Flow Cytometry. Oncotarget 2016, 7, 8839–8849. [Google Scholar] [CrossRef]
  46. Silva, J.; Garcia, V.; Rodriguez, M.; Compte, M.; Cisneros, E.; Veguillas, P.; Garcia, J.M.; Dominguez, G.; Campos-Martin, Y.; Cuevas, J.; et al. Analysis of Exosome Release and Its Prognostic Value in Human Colorectal Cancer. Genes. Chromosomes Cancer 2012, 51, 409–418. [Google Scholar] [CrossRef] [PubMed]
  47. Yoshioka, Y.; Kosaka, N.; Konishi, Y.; Ohta, H.; Okamoto, H.; Sonoda, H.; Nonaka, R.; Yamamoto, H.; Ishii, H.; Mori, M.; et al. Ultra-Sensitive Liquid Biopsy of Circulating Extracellular Vesicles Using ExoScreen. Nat. Commun. 2014, 5, 3591. [Google Scholar] [CrossRef]
  48. Ogata-Kawata, H.; Izumiya, M.; Kurioka, D.; Honma, Y.; Yamada, Y.; Furuta, K.; Gunji, T.; Ohta, H.; Okamoto, H.; Sonoda, H.; et al. Circulating Exosomal MicroRNAs as Biomarkers of Colon Cancer. PLoS ONE 2014, 9, e92921. [Google Scholar] [CrossRef]
  49. Matsumura, T.; Sugimachi, K.; Iinuma, H.; Takahashi, Y.; Kurashige, J.; Sawada, G.; Ueda, M.; Uchi, R.; Ueo, H.; Takano, Y.; et al. Exosomal MicroRNA in Serum Is a Novel Biomarker of Recurrence in Human Colorectal Cancer. Br. J. Cancer 2015, 113, 275–281. [Google Scholar] [CrossRef]
  50. Caivano, A.; Laurenzana, I.; De Luca, L.; La Rocca, F.; Simeon, V.; Trino, S.; D’Auria, F.; Traficante, A.; Maietti, M.; Izzo, T.; et al. High Serum Levels of Extracellular Vesicles Expressing Malignancy-Related Markers Are Released in Patients with Various Types of Hematological Neoplastic Disorders. Tumour. Biol. 2015, 36, 9739–9752. [Google Scholar] [CrossRef]
  51. Sugimachi, K.; Matsumura, T.; Hirata, H.; Uchi, R.; Ueda, M.; Ueo, H.; Shinden, Y.; Iguchi, T.; Eguchi, H.; Shirabe, K.; et al. Identification of a Bona Fide MicroRNA Biomarker in Serum Exosomes That Predicts Hepatocellular Carcinoma Recurrence after Liver Transplantation. Br. J. Cancer 2015, 112, 532–538. [Google Scholar] [CrossRef] [PubMed]
  52. Sandfeld-Paulsen, B.; Aggerholm-Pedersen, N.; Bæk, R.; Jakobsen, K.R.; Meldgaard, P.; Folkersen, B.H.; Rasmussen, T.R.; Varming, K.; Jørgensen, M.M.; Sorensen, B.S. Exosomal Proteins as Prognostic Biomarkers in Non-Small Cell Lung Cancer. Mol. Oncol. 2016, 10, 1595–1602. [Google Scholar] [CrossRef]
  53. Chen, Y.; Wang, L.; Zhu, Y.; Chen, Z.; Qi, X.; Jin, L.; Jin, J.; Hua, D.; Ma, X. Breast Cancer Resistance Protein (BCRP)-Containing Circulating Microvesicles Contribute to Chemoresistance in Breast Cancer. Oncol. Lett. 2015, 10, 3742–3748. [Google Scholar] [CrossRef]
  54. 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]
  55. Moon, P.-G.; Lee, J.-E.; Cho, Y.-E.; Lee, S.J.; Chae, Y.S.; Jung, J.H.; Kim, I.-S.; Park, H.Y.; Baek, M.-C. Fibronectin on Circulating Extracellular Vesicles as a Liquid Biopsy to Detect Breast Cancer. Oncotarget 2016, 7, 40189–40199. [Google Scholar] [CrossRef] [PubMed]
  56. Vardaki, I.; Ceder, S.; Rutishauser, D.; Baltatzis, G.; Foukakis, T.; Panaretakis, T. Periostin Is Identified as a Putative Metastatic Marker in Breast Cancer-Derived Exosomes. Oncotarget 2016, 7, 74966–74978. [Google Scholar] [CrossRef]
  57. Moon, P.-G.; Lee, J.-E.; Cho, Y.-E.; Lee, S.J.; Jung, J.H.; Chae, Y.S.; Bae, H.-I.; Kim, Y.-B.; Kim, I.-S.; Park, H.Y.; et al. Identification of Developmental Endothelial Locus-1 on Circulating Extracellular Vesicles as a Novel Biomarker for Early Breast Cancer Detection. Clin. Cancer Res. 2016, 22, 1757–1766. [Google Scholar] [CrossRef]
  58. Lee, S.J.; Lee, J.; Jung, J.H.; Park, H.Y.; Moon, P.-G.; Chae, Y.S.; Baek, M.-C. Exosomal Del-1 as a Potent Diagnostic Marker for Breast Cancer: Prospective Cohort Study. Clin. Breast Cancer 2021, 21, e748–e756. [Google Scholar] [CrossRef]
  59. Alegre, E.; Zubiri, L.; Perez-Gracia, J.L.; González-Cao, M.; Soria, L.; Martín-Algarra, S.; González, A. Circulating Melanoma Exosomes as Diagnostic and Prognosis Biomarkers. Clin. Chim. Acta 2016, 454, 28–32. [Google Scholar] [CrossRef]
  60. An, T.; Qin, S.; Xu, Y.; Tang, Y.; Huang, Y.; Situ, B.; Inal, J.M.; Zheng, L. Exosomes Serve as Tumour Markers for Personalized Diagnostics Owing to Their Important Role in Cancer Metastasis. J. Extracell. Vesicles 2015, 4, 27522. [Google Scholar] [CrossRef]
  61. Beach, A.; Zhang, H.-G.; Ratajczak, M.Z.; Kakar, S.S. Exosomes: An Overview of Biogenesis, Composition and Role in Ovarian Cancer. J. Ovarian Res. 2014, 7, 14. [Google Scholar] [CrossRef]
  62. Magdalena Derbis, M.S. Exosomes in Plasma of Patients with Ovarian Carcinoma: Potential Biomarkers of Tumor Progression and Response to Therapy. Gynecol. Obstet. 2012, 4, 3. [Google Scholar] [CrossRef] [PubMed]
  63. Taylor, D.D.; Gercel-Taylor, C. MicroRNA Signatures of Tumor-Derived Exosomes as Diagnostic Biomarkers of Ovarian Cancer. Gynecol. Oncol. 2008, 110, 13–21. [Google Scholar] [CrossRef] [PubMed]
  64. Nogués, L.; Benito-Martin, A.; Hergueta-Redondo, M.; Peinado, H. The Influence of Tumour-Derived Extracellular Vesicles on Local and Distal Metastatic Dissemination. Mol. Asp. Med. 2018, 60, 15–26. [Google Scholar] [CrossRef]
  65. Spugnini, E.P.; Logozzi, M.; Di Raimo, R.; Mizzoni, D.; Fais, S. A Role of Tumor-Released Exosomes in Paracrine Dissemination and Metastasis. Int. J. Mol. Sci. 2018, 19, 3968. [Google Scholar] [CrossRef] [PubMed]
  66. Logozzi, M.; Orefice, N.S.; Di Raimo, R.; Mizzoni, D.; Fais, S. The Importance of Detecting, Quantifying, and Characterizing Exosomes as a New Diagnostic/Prognostic Approach for Tumor Patients. Cancers 2023, 15, 2878. [Google Scholar] [CrossRef]
  67. Cappello, F.; Fais, S. Extracellular vesicles in cancer pros and cons: The importance of the evidence-based medicine. Semin. Cancer Biol. 2022, 86, 4–12. [Google Scholar] [CrossRef]
  68. Sabbagh, Q.; Andre-Gregoire, G.; Guevel, L.; Gavard, J. Vesiclemia: Counting on extracellular vesicles for glioblastoma patients. Oncogene 2020, 39, 6043–6052. [Google Scholar] [CrossRef]
  69. Bonifay, A.; Robert, S.; Champagne, B.; Petit, P.R.; Eugène, A.; Chareyre, C.; Duchez, A.C.; Vélier, M.; Fritz, S.; Vallier, L.; et al. A new strategy to count and sort neutrophil-derived extracellular vesicles: Validation in infectious disorders. J. Extracell Vesicles 2022, 11, e12204. [Google Scholar] [CrossRef]
  70. Rank, A.; Nieuwland, R.; Nikolajek, K.; Rösner, S.; Wallwiener, L.M.; Hiller, E.; Toth, B. Hormone replacement therapy leads to increased plasma levels of platelet derived microparticles in postmenopausal women. Arch. Gynecol. Obstet. 2012, 285, 1035–1041. [Google Scholar] [CrossRef]
  71. Tian, Y.; Ma, L.; Gong, M.; Su, G.; Zhu, S.; Zhang, W.; Wang, S.; Li, Z.; Chen, C.; Li, L.; et al. Protein Profiling and Sizing of Extracellular Vesicles from Colorectal Cancer Patients via Flow Cytometry. ACS Nano 2018, 12, 671–680. [Google Scholar] [CrossRef] [PubMed]
  72. Giuliani, A.; Fais, S. Proposal to Consider Chemical/Physical Microenvironment as a New Therapeutic Off-Target Approach. Pharmaceutics 2022, 14, 2084. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Fais, S.; Logozzi, M. The Diagnostic and Prognostic Value of Plasmatic Exosome Count in Cancer Patients and in Patients with Other Pathologies. Int. J. Mol. Sci. 2024, 25, 1049. https://doi.org/10.3390/ijms25021049

AMA Style

Fais S, Logozzi M. The Diagnostic and Prognostic Value of Plasmatic Exosome Count in Cancer Patients and in Patients with Other Pathologies. International Journal of Molecular Sciences. 2024; 25(2):1049. https://doi.org/10.3390/ijms25021049

Chicago/Turabian Style

Fais, Stefano, and Mariantonia Logozzi. 2024. "The Diagnostic and Prognostic Value of Plasmatic Exosome Count in Cancer Patients and in Patients with Other Pathologies" International Journal of Molecular Sciences 25, no. 2: 1049. https://doi.org/10.3390/ijms25021049

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop