Exosome-based liquid biopsies in cancer: opportunities and challenges

W Yu, J Hurley, D Roberts, S K Chakrabortty, D Enderle, M Noerholm, X O Breakefield, J K Skog, W Yu, J Hurley, D Roberts, S K Chakrabortty, D Enderle, M Noerholm, X O Breakefield, J K Skog

Abstract

Liquid biopsy in cancer has gained momentum in clinical research and is experiencing a boom for a variety of applications. There are significant efforts to utilize liquid biopsies in cancer for early detection and treatment stratification, as well as residual disease and recurrence monitoring. Although most efforts have used circulating tumor cells and circulating tumor DNA for this purpose, exosomes and other extracellular vesicles have emerged as a platform with potentially broader and complementary applications. Exosomes/extracellular vesicles are small vesicles released by cells, including cancer cells, into the surrounding biofluids. These exosomes contain tumor-derived materials such as DNA, RNA, protein, lipid, sugar structures, and metabolites. In addition, exosomes carry molecules on their surface that provides clues regarding their origin, making it possible to sort vesicle types and enrich signatures from tissue-specific origins. Exosomes are part of the intercellular communication system and cancer cells frequently use them as biological messengers to benefit their growth. Since exosomes are part of the disease process, they have become of tremendous interest in biomarker research. Exosomes are remarkably stable in biofluids, such as plasma and urine, and can be isolated for clinical evaluation even in the early stages of the disease. Exosome-based biomarkers have quickly become adopted in the clinical arena and the first exosome RNA-based prostate cancer test has already helped >50 000 patients in their decision process and is now included in the National Comprehensive Cancer Network guidelines for early prostate cancer detection. This review will discuss the advantages and challenges of exosome-based liquid biopsies for tumor biomarkers and clinical implementation in the context of circulating tumor DNA and circulating tumor cells.

Keywords: cell-free DNA (cfDNA); circulating tumor DNA (ctDNA); circulating tumor cell (CTC); exosome; extracellular vesicle; liquid biopsy.

Conflict of interest statement

Disclosure WY, JH, DR, SC, DE, MN, and JS are employees of Exosome Diagnostics, a Bio-Techne brand.

Copyright © 2021 The Author(s). Published by Elsevier Ltd.. All rights reserved.

Figures

Figure 1.. ctDNA, exosomes, and CTCs in…
Figure 1.. ctDNA, exosomes, and CTCs in circulation.
The molecular constituents from the tumor are released into biofluids at various stages of the tumor development through very different mechanisms and represent different biological entities. ctDNA is released from the dying tumor cells through apoptosis and/or necrosis, exosomes are actively released from cells at each stage of tumor formation, whereas CTCs are intact cancer cells disseminated from the tumor at later stages. Exosomes are involved in promoting the growth, vascularization, and even the dissemination and metastatic process of cancer. Elements are not drawn to scale. CTCs, circulating tumor cells; ctDNA, circulating tumor DNA.
Figure 2.. Exosomes enable cancer specific enrichment.
Figure 2.. Exosomes enable cancer specific enrichment.
Proteins overexpressed in a tumor are not necessarily cancer-specific when isolated from the blood or other biofluid. Non-cancerous tissues can produce the same marker and generate a variable amount of the marker in normal individuals. By enriching exosomes with a tissue-specific (or cancer-specific) marker, higher sensitivity and/or specificity can be achieved. There are also known neoantigens (for example EGFRv3) that can be used to isolate cancer-specific exosomes.,
Figure 3.. Multi-analyte liquid biopsy approach utilizing…
Figure 3.. Multi-analyte liquid biopsy approach utilizing exosomes for increased performance.
Combining features from cfDNA and exosomes have clear synergies to achieve increased liquid biopsy performance. The exosome fraction contains RNA and DNA (exoDNA) that give increased copy numbers of mutations and enable splice variant analysis of RNA,– fusions,, and transcriptome analysis., Combining the ctDNA mutations with cfDNA methylation and/or exosome analysis can enable a more complete picture of the tumor-derived biomarkers and potentially confer a better diagnostic performance. Exosomes from various sources (exemplified by the colors) can also be enriched to increase tissue specificity of the biomarker. cfDNA, cell-free DNA; ctDNA, circulating tumor DNA.
Figure 4.
Figure 4.
(A) Exosome-based research publications. The interest in exosomes for diagnostics, therapeutics and as an intricate part of intercellular communication has significantly increased in recent years. (B) Overview of a total of 71 exosome-based clinical trials registered worldwide in ClinicalTrials.gov (as of 22 October 2020) in diagnostics and translational biomarkers for various cancers, manually curated through searches with keywords (exosomes, extracellular vesicle, EVs, and microvesicles) with the elimination of both duplicate hits and those for therapeutic applications.

References

    1. Kandoth C, McLellan MD, Vandin F, et al. Mutational landscape and significance across 12 major cancer types. Nature. 2013;502: 333–339.
    1. Gerlinger M, Rowan AJ, Horswell S, et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med. 2012;366:883–892.
    1. Ding L, Ley TJ, Larson DE, et al. Clonal evolution in relapsed acute myeloid leukaemia revealed by whole-genome sequencing. Nature. 2012;481:506–510.
    1. Driessens G, Beck B, Caauwe A, et al. Defining the mode of tumour growth by clonal analysis. Nature. 2012;488:527–530.
    1. Campbell PJ, Yachida S, Mudie LJ, et al. The patterns and dynamics of genomic instability in metastatic pancreatic cancer. Nature. 2010;467: 1109–1113.
    1. Navin N, Kendall J, Troge J, et al. Tumour evolution inferred by single-cell sequencing. Nature. 2011;472:90–94.
    1. Lokhandwala T, Bittoni MA, Dann RA, et al. Costs of diagnostic assessment for lung cancer: a medicare claims analysis. Clin Lung Cancer. 2017;18:e27–e34.
    1. Leighl NB, Page RD, Raymond VM, et al. Clinical utility of comprehensive cell-free DNA analysis to identify genomic biomarkers in patients with newly diagnosed metastatic non-small cell lung cancer. Clin Cancer Res. 2019;25:4691–4700.
    1. FDA. Summary of Safety and Effectiveness Data (SSED) P150047. cobas® EGFR Mutation Test v2. 2016.
    1. FDA. Summary of Safety and Effectiveness Data (SSED) P200010. Guardant360® CDx. 2020.
    1. FDA. Summary of Safety and Effectiveness Data (SSED) P190032. FoundationOne® Liquid CDx. 2020.
    1. Haque IS, Elemento O. Challenges in using ctDNA to achieve early detection of cancer. bioRxiv. 2017;237578.
    1. Bauml J, Levy B. Clonal hematopoiesis: a new layer in the liquid biopsy story in lung cancer. Clin Cancer Res. 2018;24:4352–4354.
    1. Jahn JL, Giovannucci EL, Stampfer MJ. The high prevalence of undiagnosed prostate cancer at autopsy: implications for epidemiology and treatment of prostate cancer in the prostate-specific antigen-era. Int J Cancer. 2015;137:1749–1757.
    1. Phallen J, Sausen M, Adleff V, et al. Direct detection of early-stage cancers using circulating tumor DNA. Sci Transl Med. 2017;9: eaan2415.
    1. Liu MC, Oxnard GR, Klein EA, et al. Sensitive and specific multi-cancer detection and localization using methylation signatures in cell-free DNA. Ann Oncol. 2020;31:745–759.
    1. Nassiri F, Chakravarthy A, Feng S, et al. Detection and discrimination of intracranial tumors using plasma cell-free DNA methylomes. Nat Med. 2020;26:1044–1047.
    1. Cohen JD, Li L, Wang Y, et al. Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science. 2018;359: 926–930.
    1. Chalasani NP, Ramasubramanian TS, Bhattacharya A, et al. A novel blood-based panel of methylated DNA and protein markers for detection of early-stage hepatocellular carcinoma. Clin Gastroenterol Hepatol. 2020;S1542–3565:31224–31226.
    1. Parikh AR, Leshchiner I, Elagina L, et al. Liquid versus tissue biopsy for detecting acquired resistance and tumor heterogeneity in gastrointestinal cancers. Nat Med. 2019;25:1415–1421.
    1. Bidard FC, Peeters DJ, Fehm T, et al. Clinical validity of circulating tumour cells in patients with metastatic breast cancer: a pooled analysis of individual patient data. Lancet Oncol. 2014;15:406–414.
    1. Clifton GT, Sears AK, Patil R, et al. Monitoring of circulating tumor cell trends in a prospective, randomized, placebo-controlled HER2/neu peptide vaccine trial. J Clin Oncol. 2011;29:e11126.
    1. Cohen SJ, Punt CJ, Iannotti N, et al. Relationship of circulating tumor cells to tumor response, progression-free survival, and overall survival in patients with metastatic colorectal cancer. J Clin Oncol. 2008;26: 3213–3221.
    1. Cristofanilli M, Budd GT, Ellis MJ, et al. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N Engl J Med. 2004;351:781–791.
    1. Castro-Giner F, Aceto N. Tracking cancer progression: from circulating tumor cells to metastasis. Genome Med. 2020;12:31.
    1. Rodrigues P, Vanharanta S. Circulating tumor cells: come together, right now, over metastasis. Cancer Discov. 2018;9:22–24.
    1. Liu X, Taftaf R, Kawaguchi M, et al. Homophilic CD44 interactions mediate tumor cell aggregation and polyclonal metastasis in patient-derived breast cancer models. Cancer Discov. 2019;9:96–113.
    1. Dawson S-J, Tsui DW, Murtaza M, et al. Analysis of circulating tumor DNA to monitor metastatic breast cancer. N Engl J Med. 2013;368(13):1199–1209.
    1. Bettegowda C, Sausen M, Leary RJ, et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med. 2014;6(224):224ra24.
    1. FDA. 510(k) Summary. CellSearch™ Circulating tumor cell kit. 2007.
    1. Zhang L, Riethdorf S, Wu G, et al. Meta-analysis of the prognostic value of circulating tumor cells in breast cancer. Clin Cancer Res. 2012;18:5701–5710.
    1. Riethdorf S, Fritsche H, Müller V, et al. Detection of circulating tumor cells in peripheral blood of patients with metastatic breast cancer: a validation study of the CellSearch system. Clin Cancer Res. 2007;13: 920–928.
    1. de Bono JS, Scher HI, Montgomery RB, et al. Circulating tumor cells predict survival benefit from treatment in metastatic castration-resistant prostate cancer. Clin Cancer Res. 2008;14:6302–6309.
    1. Antonarakis ES, Lu C, Luber B, et al. Androgen receptor splice variant 7 and efficacy of taxane chemotherapy in patients with metastatic castration-resistant prostate cancer. JAMA Oncol. 2015;1:582–591.
    1. Scher HI, Jia X, de Bono JS, et al. Circulating tumour cells as prognostic markers in progressive, castration-resistant prostate cancer: a reanalysis of IMMC38 trial data. Lancet Oncol. 2009;10:233–239.
    1. Rossi E, Fabbri F. CTCs 2020: great expectations or unreasonable dreams. Cells. 2019;8:989.
    1. Marchetti A, Del Grammastro M, Felicioni L, et al. Assessment of EGFR mutations in circulating tumor cell preparations from NSCLC patients by next generation sequencing: toward a real-time liquid biopsy for treatment. PLoS One. 2014;9:e103883.
    1. Foroni C, Zarovni N, Bianciardi L, et al. When less is more: specific capture and analysis of tumor exosomes in plasma increases the sensitivity of liquid biopsy for comprehensive detection of multiple androgen receptor phenotypes in advanced prostate cancer patients. Biomedicines. 2020;8:131.
    1. Al-Nedawi K, Meehan B, Micallef B, et al. Intercellular transfer of the oncogenic receptor EGFRvIII by microvesicles derived from tumour cells. Nat Cell Biol. 2008;10:619–624.
    1. Manda SV, Kataria Y, Tatireddy BR, et al. Exosomes as a biomarker platform for detecting epidermal growth factor receptor-positive high-grade gliomas. J Neurosurg. 2018;128:1091–1101.
    1. Skog J, Würdinger T, van Rijn S, et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol. 2008;10:1470–1476.
    1. Limaye S, Patil D, Akolkar DB, et al. Multi-analyte liquid biopsies based treatment in advanced refractory cancers. J Clin Oncol. 2020;38:e15623.
    1. Krug AK, Enderle D, Karlovich C, et al. Improved EGFR mutation detection using combined exosomal RNA and circulating tumor DNA in NSCLC patient plasma. Ann Oncol. 2018;29:2143.
    1. Castellanos-Rizaldos E, Grimm DG, Tadigotla V, et al. Exosome-based detection of EGFR T790M in plasma from non-small cell lung cancer patients. Clin Cancer Res. 2018;24:2944–2950.
    1. Möhrmann L, Huang HJ, Hong DS, et al. Liquid biopsies using plasma exosomal nucleic acids and plasma cell-free DNA compared with clinical outcomes of patients with advanced cancers. Clin Cancer Res. 2018;24:181–188.
    1. Figueroa JM, Skog J, Akers J, et al. Detection of wild-type EGFR amplification and EGFRvIII mutation in CSF-derived extracellular vesicles of glioblastoma patients. Neuro Oncol. 2017;19:1494–1502.
    1. Nimir M, Ma Y, Jeffreys SA, et al. Detection of AR-V7 in liquid biopsies of castrate resistant prostate cancer patients: a comparison of AR-V7 analysis in circulating tumor cells, circulating tumor RNA and exosomes. Cells. 2019;8:688.
    1. Del Re M, Biasco E, Crucitta S, et al. The detection of androgen receptor splice variant 7 in plasma-derived exosomal RNA strongly predicts resistance to hormonal therapy in metastatic prostate cancer patients. Eur Urol. 2017;71:680–687.
    1. Cortot AB, Kherrouche Z, Descarpentries C, et al. Exon 14 deleted MET receptor as a new biomarker and target in cancers. J Natl Cancer Inst. 2017;109:djw262.
    1. Nilsson J, Skog J, Nordstrand A, et al. Prostate cancer-derived urine exosomes: a novel approach to biomarkers for prostate cancer. Br J Cancer. 2009;100:1603.
    1. Reclusa P, Laes J-F, Malapelle U, et al. EML4-ALK translocation identification in RNA exosomal cargo (ExoALK) in NSCLC patients: a novel role for liquid biopsy. Transl Cancer Res. 2019;8:S76–S78.
    1. Rodosthenous RS, Hutchins E, Reiman R, et al. Profiling extracellular long RNA transcriptome in human plasma and extracellular vesicles for biomarker discovery. iScience. 2020;23:101182.
    1. Everaert C, Helsmoortel H, Decock A, et al. Performance assessment of total RNA sequencing of human biofluids and extracellular vesicles. Sci Rep. 2019;9:17574.
    1. Cocucci E, Racchetti G, Meldolesi J. Shedding microvesicles: artefacts no more. Trends Cell Biol. 2009;19:43–51.
    1. Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol. 2013;200:373–383.
    1. Agarwal K, Saji M, Lazaroff SM, et al. Analysis of exosome release as a cellular response to MAPK pathway inhibition. Langmuir. 2015;31: 5440–5448.
    1. Cha DJ, Franklin JL, Dou Y, et al. KRAS-dependent sorting of miRNA to exosomes. Elife. 2015;4:e07197.
    1. Dou Y, Cha DJ, Franklin JL, et al. Circular RNAs are down-regulated in KRAS mutant colon cancer cells and can be transferred to exosomes. Sci Rep. 2016;6:37982.
    1. Balaj L, Lessard R, Dai L, et al. Tumour microvesicles contain retrotransposon elements and amplified oncogene sequences. Nat Commun. 2011;2:180.
    1. Di Vizio D, Kim J, Hager MH, et al. Oncosome formation in prostate cancer: association with a region of frequent chromosomal deletion in metastatic disease. Cancer Res. 2009;69:5601–5609.
    1. Minciacchi VR, You S, Spinelli C, et al. Large oncosomes contain distinct protein cargo and represent a separate functional class of tumor-derived extracellular vesicles. Oncotarget. 2015;6:11327–11341.
    1. Vagner T, Spinelli C, Minciacchi VR, et al. Large extracellular vesicles carry most of the tumour DNA circulating in prostate cancer patient plasma. J Extracell Vesicles. 2018;7:1505403.
    1. Xu R, Rai A, Chen M, et al. Extracellular vesicles in cancer – implications for future improvements in cancer care. Nat Rev Clin Oncol. 2018;15:617–638.
    1. Arroyo JD, Chevillet JR, Kroh EM, et al. Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma. Proc Natl Acad Sci U S A. 2011;108:5003–5008.
    1. Vickers KC, Palmisano BT, Shoucri BM, et al. MicroRNAs are transported in plasma and delivered to recipient cells by high-density lipoproteins. Nat Cell Biol. 2011;13:423–433.
    1. Valadi H, Ekström K, Bossios A, et al. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007;9:654–659.
    1. Baj-Krzyworzeka M, Szatanek R, Weglarczyk K, et al. Tumour-derived microvesicles carry several surface determinants and mRNA of tumour cells and transfer some of these determinants to monocytes. Cancer Immunol Immunother. 2006;55:808–818.
    1. Taylor DD, Gercel-Taylor C. MicroRNA signatures of tumor-derived exosomes as diagnostic biomarkers of ovarian cancer. Gynecol Oncol. 2008;110:13–21.
    1. Rabinowits G, Gerçel-Taylor C, Day JM, et al. Exosomal microRNA: a diagnostic marker for lung cancer. Clin Lung Cancer. 2009;10: 42–46.
    1. Mittelbrunn M, Gutiérrez-Vázquez C, Villarroya-Beltri C, et al. Unidirectional transfer of microRNA-loaded exosomes from T cells to antigen-presenting cells. Nat Commun. 2011;2:282.
    1. Cheng L, Sharples RA, Scicluna BJ, Hill AF. Exosomes provide a protective and enriched source of miRNA for biomarker profiling compared to intracellular and cell-free blood. J Extracell Vesicles. 2014;3:22743.
    1. Pigati L, Yaddanapudi SC, Iyengar R, et al. Selective release of microRNA species from normal and malignant mammary epithelial cells. PLoS One. 2010;5:e13515.
    1. Bruno S, Grange C, Collino F, et al. Microvesicles derived from mesenchymal stem cells enhance survival in a lethal model of acute kidney injury. PLoS One. 2012;7:e33115.
    1. Deregibus MC, Cantaluppi V, Calogero R, et al. Endothelial progenitor cell derived microvesicles activate an angiogenic program in endothelial cells by a horizontal transfer of mRNA. Blood. 2007;110:2440–2448.
    1. Batagov AO, Kurochkin IV. Exosomes secreted by human cells transport largely mRNA fragments that are enriched in the 3′-untranslated regions. Biol Direct. 2013;8:12.
    1. Huang X, Yuan T, Tschannen M, et al. Characterization of human plasma-derived exosomal RNAs by deep sequencing. BMC Genomics. 2013;14:319.
    1. Kogure T, Yan IK, Lin WL, Patel T. Extracellular vesicle-mediated transfer of a novel long noncoding RNA TUC339: a mechanism of intercellular signaling in human hepatocellular cancer. Genes Cancer. 2013;4:261–272.
    1. Miranda KC, Bond DT, Levin JZ, et al. Massively parallel sequencing of human urinary exosome/microvesicle RNA reveals a predominance of non-coding RNA. PLoS One. 2014;9:e96094.
    1. Bellingham SA, Coleman BM, Hill AF. Small RNA deep sequencing reveals a distinct miRNA signature released in exosomes from prion-infected neuronal cells. Nucleic Acids Res. 2012;40:10937–10949.
    1. Dhahbi JM, Spindler SR, Atamna H, et al. Deep sequencing of serum small RNAs identifies patterns of 5′ tRNA half and yRNA fragment expression associated with breast cancer. Biomark Cancer. 2014;6:37–47.
    1. Dhahbi JM, Spindler SR, Atamna H, et al. 5′-YRNA fragments derived by processing of transcripts from specific YRNA genes and pseudogenes are abundant in human serum and plasma. Physiol Genomics. 2013;45:990–998.
    1. Freedman JE, Gerstein M, Mick E, et al. Diverse human extracellular RNAs are widely detected in human plasma. Nat Commun. 2016;7: 11106.
    1. Nolte-’t Hoen EN, Buermans HP, Waasdorp M, et al. Deep sequencing of RNA from immune cell-derived vesicles uncovers the selective incorporation of small non-coding RNA biotypes with potential regulatory functions. Nucleic Acids Res. 2012;40:9272–9285.
    1. Saugstad JA, Lusardi TA, Van Keuren-Jensen KR, et al. Analysis of extracellular RNA in cerebrospinal fluid. J Extracell Vesicles. 2017;6: 1317577.
    1. Conley A, Minciacchi VR, Lee DH, et al. High-throughput sequencing of two populations of extracellular vesicles provides an mRNA signature that can be detected in the circulation of breast cancer patients. RNA Biol. 2017;14:305–316.
    1. Wei Z, Batagov AO, Schinelli S, et al. Coding and noncoding landscape of extracellular RNA released by human glioma stem cells. Nat Commun. 2017;8:1145.
    1. Picardi E, D’Erchia AM, Gallo A, et al. Uncovering RNA editing sites in long non-coding RNAs. Front Bioeng Biotechnol. 2014;2:64.
    1. Barrett SP, Salzman J. Circular RNAs: analysis, expression and potential functions. Development. 2016;143:1838–1847.
    1. Brinkman K, Meyer L, Bickel A, et al. Extracellular vesicles from plasma have higher tumour RNA fraction than platelets. J Extracell Vesicles. 2020;9:1741176.
    1. Oksvold MP, Neurauter A, Pedersen KW. Magnetic bead-based isolation of exosomes. Methods Mol Biol. 2015;1218:465–481.
    1. Tauro BJ, Greening DW, Mathias RA, et al. Two distinct populations of exosomes are released from LIM1863 colon carcinoma cell-derived organoids. Mol Cell Proteomics. 2013;12:587–598.
    1. Laulagnier K, Motta C, Hamdi S, et al. Mast cell- and dendritic cell-derived exosomes display a specific lipid composition and an unusual membrane organization. Biochem J. 2004;380:161–171.
    1. Zaborowski MP, Cheah PS, Zhang X, et al. Membrane-bound Gaussia luciferase as a tool to track shedding of membrane proteins from the surface of extracellular vesicles. Sci Rep. 2019;9:17387.
    1. Tiana Y, Gonga M, Hua Y, et al. Quality and efficiency assessment of six extracellular vesicle isolation methods by nano-flow cytometry. J Extracell Vesicles. 2019;9:1697028.
    1. Choi D, Montermini L, Jeong H, et al. Mapping subpopulations of cancer cell-derived extracellular vesicles and particles by nano-flow cytometry. ACS Nano. 2019;13:10499–10511.
    1. Jones PS,Yekula A, Lansbury E, et al. Characterization of plasma-derived protoporphyrin-IX-positive extracellular vesicles following 5-ALA use in patients with malignant glioma. EBioMedicine. 2019;48:23–35.
    1. Choi D, Montermini L, Kim DK, et al. The impact of oncogenic EGFRvIII on the proteome of extracellular vesicles released from glioblastoma cells. Mol Cell Prot. 2018;17:1948–1964.
    1. Grasso L, Wyss R, Weidenauer L, et al. Molecular screening of cancer-derived exosomes by surface plasmon resonance spectroscopy. Anal Bioanal Chem. 2015;407:5425–5432.
    1. Kwizera EA, O’Connor R, Vinduska V, et al. Molecular detection and analysis of exosomes using surface-enhanced raman scattering gold nanorods and a miniaturized device. Theranostics. 2018;8:2722–2738.
    1. Vaidyanathan R, Naghibosadat M, Rauf S, et al. Detecting exosomes specifically: a multiplexed device based on alternating current electrohydrodynamic induced nanoshearing. Anal Chem. 2014;86:11125–11132.
    1. Yoshioka Y, Kosaka N, Konishi Y, et al. Ultra-sensitive liquid biopsy of circulating extracellular vesicles using ExoScreen. Nat Commun. 2014;5:3591.
    1. Zhao Z, Yang Y, Zeng Y, He M. A microfluidic ExoSearch chip for multiplexed exosome detection towards blood-based ovarian cancer diagnosis. Lab Chip. 2016;16:489–496.
    1. Im H, Shao H, Park YI, et al. Label-free detection and molecular profiling of exosomes with a nano-plasmonic sensor. Nat Biotechnol. 2014;32:490–495.
    1. Logozzi M, Angelini DF, Giuliani A, et al. Increased plasmatic levels of PSA-expressing exosomes distinguish prostate cancer patients from benign prostatic hyperplasia: a prospective study. Cancers (Basel). 2019;11:1449.
    1. Li W, Bell NA, Hernández-Ainsa S, et al. Single protein molecule detection by glass nanopores. ACS Nano. 2013;7:4129–4134.
    1. Schubert SM, Arendt LM, Zhou W, et al. Ultra-sensitive protein detection via single molecule arrays towards early stage cancer monitoring. Sci Rep. 2015;5:11034.
    1. Dhondt B, Geeurickx E, Tulkens J, et al. Unravelling the proteomic landscape of extracellular vesicles in prostate cancer by density-based fractionation of urine. J Extracell Vesicles. 2020;9:1736935.
    1. McKiernan J, Donovan MJ, O’Neill V, et al. A novel urine exosome gene expression assay to predict high-grade prostate cancer at initial biopsy. JAMA Oncol. 2016;2:882–889.
    1. McKiernan J, Donovan MJ, Margolis E, et al. A prospective adaptive utility trial to validate performance of a novel urine exosome gene expression assay to predict high-grade prostate cancer in patients with prostate-specific antigen 2–10 ng/ml at initial biopsy. Eur Urol. 2018;74:731–738.
    1. National Comprehensive Cancer Network. Prostate Cancer EarlyDetection, (Version 2.2020). Available at: . Accessed December 4, 2020.
    1. Kahlert C, Melo SA, Protopopov A, et al. Identification of double-stranded genomic DNA spanning all chromosomes with mutated KRAS and p53 DNA in the serum exosomes of patients with pancreatic cancer. J Biol Chem. 2014;289:3869–3875.
    1. Thakur BK, Zhang H, Becker A, et al. Double-stranded DNA in exosomes: a novel biomarker in cancer detection. Cell Res. 2014;24:766–769.
    1. San Lucas FA, Allenson K, Bernard V, et al. Minimally invasive genomic and transcriptomic profiling of visceral cancers by next-generation sequencing of circulating exosomes. Ann Oncol. 2015;27:635–641.
    1. Oldrini B, Vaquero-Siguero N, Mu Q, et al. MGMT genomic rearrangements contribute to chemotherapy resistance in gliomas. Nat Commun. 2020;11:3883.
    1. Lee DH, Yoon H, Park S, et al. Urinary exosomal and cell-free DNA detects somatic mutation and copy number alteration in urothelial carcinoma of bladder. Sci Rep. 2018;8:14707.
    1. Dusoswa SA, Horrevorts SK, Ambrosini M, et al. Glycan modification of glioblastoma-derived extracellular vesicles enhances receptor-mediated targeting of dendritic cells. J Extracell Vesicles. 2019;8: 1648995.
    1. Scott E, Munkley J. Glycans as biomarkers in prostate cancer. Int J Mol Sci. 2019;20:1389.
    1. Skotland T, Ekroos K, Kauhanen D, et al. Molecular lipid species in urinary exosomes as potential prostate cancer biomarkers. Eur J Cancer. 2017;70:122–132.
    1. Myung JH, Hong S. Microfluidic devices to enrich and isolate circulating tumor cells. Lab Chip. 2015;15:4500–4511.
    1. Contreras-Naranjo JC, Wu H, Ugaz VM. Microfluidics for exosome isolation and analysis: enabling liquid biopsy for personalized medicine. Lab Chip. 2017;17:3558–3577.
    1. Gregson AL, Hoji A, Injean P, et al. Altered exosomal RNA profiles in bronchoalveolar lavage from lung transplants with acute rejection. Am J Respir Crit Care Med. 2015;192:1490–1503.
    1. Sigdel TK, Ng YW, Lee S, et al. Perturbations in the urinary exosome in transplant rejection. Front Med (Lausanne). 2014;1:57.
    1. Das S, Halushka MK. Extracellular vesicle microRNA transfer in cardiovascular disease. Cardio Path. 2015;24:199–206.
    1. Stern RA, Tripodis Y, Baugh CM, et al. Preliminary study of plasma exosomal tau as a potential biomarker for chronic traumatic encephalopathy. J Alzheimers Dis. 2016;51:1099–1109.
    1. Vella LJ, Hill AF, Cheng L. Focus on extracellular vesicles: exosomes and their role in protein trafficking and biomarker potential in Alzheimer’s and Parkinson’s disease. Int J Mol Sci. 2016;17:173.
    1. Graham EM, Burd I, Everett AD, Northington FJ. Blood biomarkers for evaluation of perinatal encephalopathy. Front Pharmacol. 2016;7: 196.
    1. Coras R, Narasimhan R, Guma M. Liquid biopsies to guide therapeutic decisions in rheumatoid arthritis. Transl Res. 2018;201:1–12.
    1. Huang Y, Li R, Ye S, et al. Recent advances in the use of exosomes in Sjögren’s Syndrome. Front Immunol. 2020;11:1509.
    1. Matsumoto J, Stewart T, Banks WA, et al. The transport mechanism of extracellular vesicles at the blood-brain barrier. Curr Pharm Des. 2017;23:6206–6214.
    1. Enderle D, Spiel A, Coticchia CM, et al. Characterization of RNA from exosomes and other extracellular vesicles isolated by a novel spin column-based method. PLoS One. 2015;10:e0136133.
    1. Best MG, Sol N, Kooi I, et al. RNA-seq of tumor-educated platelets enables blood-based pan-cancer, multiclass, and molecular pathway cancer diagnostics. Cancer Cell. 2015;28:666–676.

Source: PubMed

3
Sottoscrivi