Next Article in Journal
Discordant and Converting Receptor Expressions in Brain Metastases from Breast Cancer: MRI-Based Non-Invasive Receptor Status Tracking
Next Article in Special Issue
The Current Landscape of Glioblastoma Biomarkers in Body Fluids
Previous Article in Journal
New Strategies in Diagnosis and Treatments for Brain Tumors
Previous Article in Special Issue
Cell-Free RNA from Plasma in Patients with Neuroblastoma: Exploring the Technical and Clinical Potential
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Importance of Detecting, Quantifying, and Characterizing Exosomes as a New Diagnostic/Prognostic Approach for Tumor Patients

by
Mariantonia Logozzi
1,
Nicola Salvatore Orefice
2,
Rossella Di Raimo
3,
Davide Mizzoni
3 and
Stefano Fais
1,*
1
Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy
2
Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
3
ExoLab Italia, Tecnopolo d’Abruzzo, 67100 L’Aquila, Italy
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(11), 2878; https://doi.org/10.3390/cancers15112878
Submission received: 6 April 2023 / Revised: 11 May 2023 / Accepted: 22 May 2023 / Published: 23 May 2023
(This article belongs to the Collection Cancer Biomarkers in Body Fluids)

Abstract

:

Simple Summary

Clinical oncology urgently needs more specific and helpful new biomarkers to improve the diagnosis and prognosis of cancer. Research of the last decade proposes extracellular vesicles, particularly exosomes, as a natural source of new biomarkers; since tumors massively release them, they circulate through the body and can be detected and characterized in plasma samples of tumor patients. After a decade of up-and-coming pre-clinical research, the results of the few clinical studies have provided some exciting data supporting the use of exosomes, at least in the follow-up of tumor patients. However, the most convincing data have taught us that, on the one hand, circulating exosomes deliver known tumor markers, such as PSA; on the other hand, the exosome plasmatic levels in tumor patients consistently exceed those of normal controls. This information will be extremely useful in the clinical management of tumor patients.

Abstract

Exosomes are extracellular vesicles (EVs) of nanometric size studied for their role in tumor pathogenesis and progression and as a new source of tumor biomarkers. The clinical studies have provided encouraging but probably unexpected results, including the exosome plasmatic levels’ clinical relevance and well-known biomarkers’ overexpression on the circulating EVs. The technical approach to obtaining EVs includes methods to physically purify EVs and characterize EVs, such as Nanosight Tracking Analysis (NTA), immunocapture-based ELISA, and nano-scale flow cytometry. Based on the above approaches, some clinical investigations have been performed on patients with different tumors, providing exciting and promising results. Here we emphasize data showing that exosome plasmatic levels are consistently higher in tumor patients than in controls and that plasmatic exosomes express well-known tumor markers (e.g., PSA and CEA), proteins with enzymatic activity, and nucleic acids. However, we also know that tumor microenvironment acidity is a key factor in influencing both the amount and the characteristics of the exosome released by tumor cells. In fact, acidity significantly increases exosome release by tumor cells, which correlates with the number of exosomes that circulate through the body of a tumor patient.

1. Introduction

Virtually any cell, during its lifespan (from embryonic development to senescence), releases extracellular vesicles (EVs). EVs range in size from 30 nm to 1 μm, and the size distinguishes microvesicles (from 200 nm to 1 μm) from exosomes (from 30 nm to 150 nm) under both physiological and pathological conditions [1,2,3,4,5,6].
Exosome generation processes include a membrane shedding-like phenomenon (for microvesicles) and multivesicular body (MVB) formation (for exosomes) [5,6,7,8]. Other mechanisms cannot be excluded and are currently under investigation worldwide. EVs, particularly nanovesicles (exosomes), are a natural delivery system for a wide array of substances. Exosomes travel the body through both hematic and lymphatic circulations. Between the molecules that may be detected in exosome preparations, there are housekeeping proteins, including tetraspanins (i.e., CD63, CD9, and CD81), heat shock proteins (such as HSP-70), members of the Rab family, as well as other proteins, including Tsg101 and Alix. These markers have been used to characterize and quantify exosomes [9,10]. However, exosomes, during their formation, involve internal cell structures and the plasma membrane; this may lead to the acquisition of markers of the cellular source [5,6,7,8].
Overall, exosomes purified from body fluids may contain typical tags that help distinguish exosomes from other particles and markers indicating the cellular source and often the body compartment from which they are released. The above reasons make EVs, particularly exosomes, a potential source of disease biomarkers with possible use as a liquid biopsy in clinical oncology. In addition, exosomes have been shown to contain a series of nucleic acids, including DNAs, mRNAs, and miRNAs, that may represent an additional source of disease biomarkers [5,6,11]. However, exosomes also remove unnecessary molecules poorly degraded by the lysosomal system [12,13], thus emphasizing the broad and complex function of these nanovesicles in our body [1,4,5,6,12,13,14,15,16,17,18,19,20].
Exosomes can be found in many biological fluids, including blood, urine, saliva, stools, cerebrospinal, epididymal, amniotic, serous fluids (including pleural, pericardial, and peritoneal fluids), bronchoalveolar lavage fluid, synovial fluid, and breast milk [6,21,22,23,24,25,26]. Exosomes are released in a paracrine way within tissues, from where they are spilled into the bloodstream, often ending in tissues of body compartments far from the production site. For example, scientific evidence has shown that exosomes containing a reporter gene are released from a tumor, found in the blood, and end in the germ line of the gonads, with the potential to transfer the acquired genetic material to the progeny [27]. It is, therefore, conceivable that exosomes may well participate in the continuous genome remodeling that occurs in our body. The matter of fact is that exosomes are considered a natural source of disease biomarkers [5,6,11,25,26,27,28,29,30,31,32,33,34,35,36,37]. A series of exciting molecules have been identified in the plasma of both patients and healthy donors [5,6,7,11,29,32,38]. Clinical studies, while still very few as compared to pre-clinical information, are providing exciting information while often not entirely fitting with the aim of the studies [11,39,40,41], challenging the use of these data in clinical settings. 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 [1,4,11,15]. Some unexpected but interesting findings propose the simple measurement of exosome plasmatic levels as a key prognostic value [41,42]. The clinical data show that, independently from the cancer histology (i.e., melanoma, prostate cancer, or oral cancers) and the technique used in the experimental protocol (e.g., immunocapture ELISA, nanoparticle tracking analysis (NTA), or nanoscale flow cytometry), patients displayed higher plasmatic exosome levels as compared to healthy donors [24,41,42,43]. Other interesting issues are the expression on plasmatic exosomes from tumor patients of acknowledged tumor markers (e.g., PSA and CEA) [44,45] and a series of surrogate tumor markers (e.g., Cav-1, HSP60, and carbonic anhydrase, such as CA IX) [24,43,46,47]. This review will introduce and discuss these issues to propose the best use of exosomes in clinical oncology.

2. A Technical Insight

A general discussion is given of the techniques used to purify and characterize exosomes from patient samples [5]. Currently, Nanoparticle Tracking Analysis (NTA) allows the determination of the number and size of the obtained EVs from either cell culture supernatant or body fluids. NTA acquires the Brownian movement of nanoparticles in a liquid suspension, analyzing the EVs’ concentration and size distribution in the sample. This is based on a single particle analysis with a serial correlation with the particle size [47,48]. The NTA analysis covers a broad range of particle sizes, ranging from 30 nm to 400–500 nm, thus distinguishing nanovesicles from microvesicles. NTA is, to date, considered the most reliable technique to analyze a mixed population of submicroscopical vesicles in human body fluids.
A preliminary analysis that might be performed on an EVS sample is transmission electron microscopy (TEM). While not allowing a quantitative evaluation, TEM is an integral approach to verifying whether samples under investigation contain submicroscopical vesicles and whether a round shape and the typical bilayer membrane are maintained after repeated centrifugation and ultracentrifugation. Moreover, vesicles may be phenotyped by immuno-TEM using immuno-gold-labeled antibodies. A disadvantage of TEM is that the samples undergo sequential rounds of fixing and dehydration before analysis, thus potentially inducing morphological damage [47,48]. However, it is advisable to evaluate exosomes by TEM analysis.
A rough evaluation of exosomes may also be performed by measuring the amount and type of exosomal proteins present in the sample. The last accepted guidelines (MISEV2018) have agreed on the following points that are required to establish that the sample under investigation contains exosomes: (i) enrichment in at least one transmembrane protein associated with the exosomal plasma membrane (e.g., tetraspanins CD9, CD63, CD81); (ii) enrichment in cytosolic proteins (e.g., TSG101, ALIX) [5,49]. The most commonly used techniques allowing this analysis are (i) Western blot, which is only a semi-quantitative approach not valid for the study of clinical samples. Moreover, it is expensive in terms of both the volumes required for the analysis and the time needed to obtain the results; it is undeniably a qualitative analysis, allowing the detection of many proteins at the same time; and (ii) flow cytometry allows simultaneous analysis of phenotyping (through labeling with fluorescent antibodies) and physical parameters (e.g., size and structure of particles). However, conventional cytometers could underestimate particles smaller than 300 nm, and a new generation of flow cytometers has been provided with both multi-angle lasers to improve particle resolution [50,51,52] and nanoscale equipment to include analysis of nanosized particles, also called nanoscale-flow cytometry, recently used in clinical studies [44,53].
A technical approach that allows us to simultaneously provide quantitative and qualitative data is the immunocapture-based ELISA. It was shown for the first time that immunocapture-based ELISA exosomes could be quantified and characterized from either cell culture supernatants or human plasma [24]. This technique was exploited in clinical investigations, including melanoma, prostate, and oral cancer patients [24,43,44]. This approach allows the analysis of the whole EV population, including exosomes. Fluorescence Activated Cell Sorter (FACS), while equipped with nanoscale flow cytometry, does not allow a broad spectrum of analysis or simultaneous analysis of different samples. Immunocapture-based ELISA looks ideal for this purpose since it will enable the detection and quantification of both exosome-specific antigens and tumor antigens on EVs isolated from small quantities of plasma simultaneously [24,43,44,53]. Recent data support the high level of versatility of the technique, with the identification of a series of housekeeping proteins, such as Rab5b, CD81, and CD63, and tumor-specific markers, such as PSA, but also surrogate tumor markers, such as Cav-1 and carbonic anhydrase [24,43,44,46,47,53].
Furthermore, this approach has been recently reported for characterizing urinary exosomes [25], thus representing a new approach for the follow-up of patients affected by urinary tract cancers. However, the goal will be to implement immunocapture-based ELISA with other methods, such as nanoscale flow cytometry (NFC) and NTA, as proposed in prostate cancer patients [41]. In the above study, statistical analysis of the results showed that immunocapture-based ELISA allows exosomal PSA detection and discriminates prostate cancer patients from both healthy subjects and benign prostate hypertrophy (BPH) patients with significantly higher sensitivity and specificity than serum PSA. Moreover, immunocapture-based ELISA allows for quantifying and characterizing several clinical samples simultaneously and in a broader population of EVs compared to nanoscale flow cytometry [53,54,55].

3. A Role of Exosomes in Cancer: From Preclinical to Clinical Data

Scientific evidence is accumulating that exosomes have a crucial role in tumor metastasis, passing through either the generation of a metastatic niche or a tumor-like transformation of mesenchymal stem cells in organs that are targets of metastasis [4,15,56,57,58]. However, the acidic pH of the tumor microenvironment plays a determinant role in at least three essential features: (i) the increased exosome release by tumor cells; (ii) determining the exosome cargo, including some tumor biomarkers [2,46,53]; and (iii) it is associated with a reduced size as compared to the heterogeneous size of those released at physiological pH [2,53]. The increased exosome release in acidic conditions correlates to the high plasmatic exosome levels compared to controls [44,53]. The reason why tumor cells increase the release of exosomes in acidic conditions may be related to the attempt to eliminate toxic molecules that tend to accumulate in the tumor microenvironment; the molecules to stop include antitumor drugs such as cisplatin [59]. This is further supported by the observation that antitumor medications contained in the exosomes released by tumors are in their native/active form, thus potentially being released into the bloodstream and getting into unaffected organs, contributing to the heavy side effects that sadly often occur in cancer patients. Between the molecules delivered by tumor exosomes, there are ion transporters (e.g., CAIX) that, together, are significantly increased in exosomes released in acidic conditions and conserve their full enzymatic function [46]. The CA has also been shown in the plasmatic exosomes of cancer patients; the same plasmatic exosomes have shown increased acidity compared to healthy subjects [47].
Another hurdle was the claim for the specificity of some markers identified on circulating exosomes of tumor patients that turned out not to be so specific for a given tumor. One example is glypican-1, which has been proposed as a specific marker of pancreatic cancer but also showed a high expression level in exosome purification from other cancers [56]. Too often, the specificity of an exosome-related tumor biomarker was not tested by comparing different cancer patients [60].

4. Exosomes Deliver Enzymatic Activity

One of the most effective mechanisms by which exosomes may up-load their content into target cells is the fusion between their membrane and the plasma membrane of a target cell [61]. Through the above mechanism, exosomes released by a primary tumor may contribute to the metastatic process once they get to a metastatic organ via the bloodstream [15,58]. This is further supported by a recent report showing that exosomes obtained from cancer patients’ plasma deliver proteins and molecules with evident enzymatic activity and an intraluminal pH suitable for enzyme activation [47]. Notably, it was also shown that in vitro, the acidic condition increases the expression of exosomes and proteins with enzymatic activity, such as carbonic anhydrase [46]. This information, on the one hand, further highlights the importance of exosomes as a natural delivery system for a broad array of molecules; on the other hand, it suggests that the research of disease biomarkers should also be directed to functional molecules rather than the mere expression of a protein.

5. Exosomes Deliver Nucleic Acids

At the time, exosomes were considered vesicles released by the cells with a significant commitment to scavenging cells from either toxic or unwanted material. Of course, this remains a function of extracellular vesicles, as witnessed by EVs in the stools and urine [6]. However, the discovery that EVs deliver nucleic acids has changed how these vesicles have been considered [19]. It has been shown that EVs, purified from either cell culture supernatant or human body fluids, contain mRNA, miRNA, long non-coding RNA (lncRNA), and DNA [11,62,63]. Most clinical studies reporting the nucleic acid cargo of body fluid-derived exosomes have been performed in tumor patients. The results suggest significant differences exist between tumor patients and healthy individuals, particularly in exosomal miRNA composition [32,64,65,66,67,68,69,70,71]. Currently, there is some inconsistency primarily due to technical and analytical issues, which too often create inhomogeneity between the samples, which in turn affects miRNA’s yield, integrity, and purity [5]. One important issue is the evidence that miRNAs are not always associated with exosomes, often being associated with either RNA-binding proteins (e.g., Argonaute 2) or lipoproteins (e.g., HDL and LDL) [5,65,66]. More recently, a commercially available isolation kit (MACS Exosome Isolation Kit, Miltenyi Biotec, Germany) is starting to be exploited to obtain a more purified exosome population, thus providing a more certain exosome-associated miRNA yield [72]. Comparably to the MACS method, immunocapture-based exosome purification may greatly help in obtaining exosomes from the ultracentrifuged material using antibodies directed against the proteins that are overexpressed on the exosome membrane (e.g., CD9, CD63, CD81, ALIX). The same approach may be exploited using plastic wells and magnetic beads as primary substrates [73]. This approach allows us to obtain a highly enriched exosome preparation, thus analyzing only the vesicles captured by the antibodies in terms of characterization of either miRNAs or RNAs present in the immunocaptured material. The immunocapture-based methodology has also been described and used in clinical trials [53,54,55]. However, it needs to be extensively exploited in analyzing the presence of exosome-associated nucleic acids in clinical samples using different approaches [74]. Another interesting area is related to the analysis of the presence of genomic DNA mutations in exosomes purified from clinical samples. DNA mutations are involved in many tumor advantages, most notably resistance to therapies, and represent a potential tumor biomarker [71]. Detecting exosomal DNA in clinical samples is receiving a large consensus in cancer patients [75,76,77,78] and other diseases, including viral-related pathological conditions [79]. In addition, recent reports have shown that exosomes purified and concentrated from body fluids, such as ascites, may express high levels of protein glycosylation [80]. While the data reporting critical roles of exosome associated RNAs is becoming bulky, we need more convincing evidence that they may represent helpful and reliable tumor biomarkers to be diffusely used in oncology laboratories worldwide. Therefore, it appears mandatory that it should need central management of the available data to get to a conclusive analysis.

6. Conclusions

To date, we have considerable data supporting the use of exosomes and EVs for the clinical management of tumor patients (Table 1). However, of course, it needs clinical validation to be considered an accurate diagnostic/prognostic tool in clinical oncology. What was an exciting hypothesis for the scientists involved in the field a decade ago is now scientific evidence that exosomes are a source of new biomarkers. However, while the discovery of new biomarkers still needs time to be translated into the clinic, some unexpected findings promise to need a shorter path to clinical use: (1) The evaluation of the number of circulating exosomes that are proven to be higher in patients with cancer as compared to healthy controls; (2) Plasmatic exosomes hyperexpress known tumor biomarkers (e.g., PSA, CEA).
Additional information is that plasmatic exosomes are smaller in tumor patients than in healthy and diseased controls and more acidic in tumor patients than controls. Thus, quantifying and characterizing exosomes in human body fluids represents a new tool for clinical oncologists and a non-invasive diagnostic/prognostic approach.
We have three methods that, when implemented, may offer a solid approach to using these methods together to quantify and characterize exosomes: Nanoparticle Tracking Analysis (NTA), immunocapture-based ELISA, and nanoscale flow cytometry (NFC). Using all these methodologies to describe exosome purification in clinical samples may represent a real advance in the clinical management of tumor patients. Another interesting approach is to use immunocapture of exosomes to optimize the detection of tumor biomarkers, particularly in detecting and validating tumor-specific miRNA. Possible future directions could be: (i) to identify physical-chemical properties of exosomes associated with some tumor phenotypes (e.g., intraluminal pH); (ii) to include the expression of active molecules within exosomes (e.g., carbonic anhydrase). Clinical studies are also needed to validate the existing data in a broader range of body fluids, with considerable advantages for patients by avoiding or limiting unnecessary invasive procedures and hopefully significantly reducing public health costs. In this sense, the data from studies performed in the urines of patients look very promising [25,26,64,80,81,82,83]. Most of all, we need to tidy up the increasing amount of clinical and pre-clinical data supporting the use of exosomes as a source of tumor biomarkers, using too often different technologies and different ways to obtain exosomes from other body fluids [84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111]. This review asks for a more strategic approach to obtaining data on exosomes from clinical samples of tumor patients. As challenging news, it has been recently reported that exosomes may deliver therapeutic antibodies that have been shown to maintain their full activity when expressed on exosomes [112]. This finding might be of paramount importance not only for therapeutic use but also for its potential as a new family of biomarkers for both the diagnosis and prognosis of cancer patients. Table 2 summarizes the ongoing clinical trials using exosomes as diagnostic/prognostic tumor biomarkers. It is straightforward from the table that the number of clinical trials is increasing, and the current number is awe-inspiring, up to 65. This means that in the following years, we will have more data to reason about the future directions of clinical research on exosomes. The current clinical research covers a broad panel of exosome-associated potential tumor biomarkers that will hopefully represent a promising future for clinical oncology.
Table 1. Data from clinical investigations on extracellular vesicles.
Table 1. Data from clinical investigations on extracellular vesicles.
TumorBiomarkersSourceReferences
Breast cancerBreast cancer resistance protein (BCRP)Plasma[113]
Her2Plasma
Serum
[114,115]
Glypican-1Serum[56]
FibronectinPlasma[116]
PeriostinPlasma[117]
Del-1Plasma[118,119]
miR-101, miR-372, and miR-373Serum[84]
miR-1246 and miR-21Plasma[85]
Colorectal cancerHsp60Plasma[38]
TSAP6/CEAPlasma[86]
Glypican-1Plasma[28]
CEASerum[45,87]
CD147Serum[87]
Plasma[89]
let-7a, miR-1229, miR-1246, miR-150, miR-21, miR-223, and miR-23aSerum[90]
miR-19Serum[91]
miR-4772-3pSerum[92]
miR-21Serum[87]
miR-221Serum[94]
Esophageal squamous sell sarcinomamiR-21Serum[95]
Gastric cancerGKN1Serum[96]
TGF-β1Plasma[97]
RNABile[98]
miR-423-5pSerum[99]
Hematological tumorsCD9, CD13, CD19, CD30, CD38, and CD63Serum[100]
Hepatocellular carcinomamiR-18a, miR-221, miR-222, and miR-224Serum[101]
miR-718Serum[102]
Laryngeal squamous cell carcinomamiR-21 and HOTAIR (lncRNA)Serum[103]
Lung cancerNY-ESO-1Plasma[104]
miR-125a-5p, miR-145, and miR-146aSerum[105]
miR-151a-5p, miR-30a-3p, miR-200b-5p, miR-629, miR-100, and miR-154-3pPlasma[106]
MelanomaCaveolin-1Plasma[24]
HSP70 and HSP90Plasma[120]
MIA and S100BSerum[121]
Oral squamous cell carcinomaCAV-1Plasma[43]
Ovarian cancerEpCAM, CD24, and
CA-125
Plasma[122,123,124]
TGF-beta1 and MAGE3/6,Plasma[125]
miR-21, miR-214, miR-200a, miR-200b, miR-200c, miR-203, miR-205, and miR-141Serum[126]
miR-21, miR-100, miR-200, miR-320, and
miR373
Serum[107]
Pancreatic cancerCD44v6, Tspan 8, EpCAM, and CD104
miR-1246
miR-3976
miR-4306
miR-4644
Serum
Urine
[64]
KRAS
P53 mutations
Serum[71]
miR-17-5p and miR-21Serum[108]
miR-10b, miR-21, miR-30c, miR-181a, and miR-let7aSerum[127]
Glypican-1Plasma[109]
miR-191, miR-21, and miR-451aSerum[110]
miR-451aPlasma[111]
Prostate cancer (PCa)PSAPlasma[44,53]
Urine[25]
CA IXPlasma[47]
SurvivinPlasma[128]
Exosome levelsPlasma[41]
PTENPlasma[129]
miR-141 and miR-375Serum[130]
miR-1290 and miR-375Plasma[131]
miR-141Serum[132]
Table 2. Ongoing clinical trials using exosomes in tumor diagnosis.
Table 2. Ongoing clinical trials using exosomes in tumor diagnosis.
NCT NumberStatusDiseaseCharacteristicsRef.
NCT03235687Active, not recitingProstate CancerYear: 2017
Population: n = 1000; Age: 50 years and older; Sex: male
Phase: Not applicable
[133]
NCT03974204WithdrawnBreast Cancer
Leptomeningeal Metastasis
Year: 2019
Population: n = 0; Age: 18 years and older; Sex:
female
Phase: Not applicable
[134]
NCT05286684RecruitingBreast CancerYear: 2023
Population: n = 30; Age: 18 years and older; Sex: female
Phase: Not applicable
[135]
NCT04781062Active, not recruitingBreast CancerYear: 2021
Population: n = 367; Age: 18 years and older; Sex: female
Phase: Not applicable
[136]
NCT02662621CompletedCancer (Solid Tumors)Year: 2015
Population: n = 71; Age: 18 years and older; Sex: all
Phase: Not applicable
[137]
NCT04530890RecruitingBreast Cancer
Digestive Cancer
Gynecologic Cancer
Circulating Tumor DNA
Exosomes
Year: 2021
Population: n = 1000; Age: 18 years and older; Sex: all
Phase: Not applicable
[138]
NCT04258735RecruitingMetastatic Breast CancerYear: 2019
Population: n = 300; Age: 18 years and older; Sex: all
Phase: Not applicable
[139]
NCT04556916RecruitingProstate CancerYear: 2021
Population: n = 320; Age: 40 years and older; Sex: male
Phase: Not applicable
[140]
NCT03711890RecruitingPancreatic Carcinoma
Pancreatic Intraductal Papillary Mucinous Neoplasm, Pancreatobiliary Type
Year: 2019
Population: n = 75; Age: 18 years and older; Sex: all
Phase: Not applicable
[141]
NCT02507583CompletedMalignant Glioma
Neoplasms
Year: 2015
Population: n = 33; Age: 18 years and older; Sex: all
Phase: Phase 1
[142]
NCT05218759Not yet recruitingNon-Small Cell Lung CancerYear: 2022
Population: n = 30; Age: 18 to 75 years; Sex: all
Phase: Not applicable
[143]
NCT04427475Unknown statusNSCLC PatientsYear: 2020
Population: n = 200; Age: 18 years and older; Sex: all
Phase: Not applicable
[144]
NCT04636788Unknown statusPancreas AdenocarcinomaYear: 2020
Population: n = 102; Age: 18 years and older; Sex: all
Phase: Not applicable
[145]
NCT03542253Unknown statusEarly Lung CancerYear: 2018
Population: n = 80; Age: child, adult, and older adult; Sex: all
Phase: not reported
[146]
NCT04529915Active, not recruitingLung CancerYear: 2020
Population: n = 470; Age: 40 years and older; Sex: all
Phase: not reported
[147]
NCT03821909Unknown statusPancreatic CancerYear: 2018
Population: n = 30; Age: 18 to 80 years; Sex: all
Phase: not repoted
[148]
NCT03830619CompletedLung Cancer (Diagnosis)Year: 2017
Population: n = 1000; Age: 18 to 75 years; Sex: all
Phase: not reported
[149]
NCT04394572CompletedColorectal CancerYear: 2021
Population: n = 80; Age: 18 years and older; Sex: all
Phase: not reported
[150]
NCT04155359RecruitingBladder CancerYear: 2020
Population: n = 3000; Age: 45 to 85 years; Sex: all
Phase: not reported
[151]
NCT01344109WithdrawnBreast NeoplasmsYear: 2011
Population: n = 0; Age: 18 years and older; Sex: female
Phase: not reported
[152]
NCT05587114RecruitingLung Cancer
Diagnosis
Year: 2022
Population: n = 150; Age: 40 years and older; Sex: all
Phase: not reported
[153]
NCT05270174Not yet recruitingExplore Whether lncRNA-ElNAT1 in Urine Exosomes Can be Used as a New Target for Preoperative
Diagnosis of Lymph Node Metastasis
Year: 2023
Population: n = 75; Age: 18 years and older; Sex: all
Phase: not reported
[154]
NCT03032913CompletedPancreatic Ductal Adenocarcinoma (PDAC)Year: 2017
Population: n = 52; Age: 18 years and older; Sex: all
Phase: not reported
[155]
NCT02702856CompletedProstate CancerYear: 2014
Population: n = 2000; Age: 50 years and older; Sex: male
Phase: not reported
[156]
NCT04523389Unknown statusColorectal CancerYear: 2020
Population: n = 172; Age: 18 years and older; Sex: all
Phase: not reported
[157]
NCT03694483SuspendedProstate CancerYear: 2018
Population: n = 600; Age: 18 years and older; Sex: male
Phase: not reported
[158]
NCT04661176Active, not recruitingProstate CancerYear: 2020
Population: n = 500; Age: 22 years and older; Sex: male
Phase: not reported
[159]
NCT02393703RecruitingPancreatic Cancer
Benign Pancreatic Disease
Year: 2015
Population: n = 111; Age: 18 years and older; Sex: all
Phase: not reported
[160]
NCT01779583Unknown statusGastric CancerYear: 2013
Population: n = 80; Age: 18 years and older; Sex: all
Phase: not reported
[161]
NCT04081194Unknown statusNew Tumor Diagnostics From Human Plasma SamplesYear: 2016
Population: n = 15; Age: 50 to 90 years; Sex: all
Phase: not reported
[162]
NCT03236688SuspendedMetastatic Castrate-Resistant Prostate CancerYear: 2016
Population: n = 30; Age: 18 years and older; Sex: male
Phase: not reported
[163]
NCT04629079RecruitingLung CancerYear: 2020
Population: n = 800; Age: 18 years and older; Sex: all
Phase: not reported
[164]
NCT04939324Active, not recruitingLung Cancer
Exosomes
Non-Small Cell Lung Cancer
Year: 2021
Population: n = 30; Age: 18 years and older; Sex: all
Phase: Not Applicable
[165]
NCT04288141RecruitingHER2-positive Breast CancerYear: 2019
Population: n = 40; Age: 18 years and older; Sex: all
Phase: not reported
[166]
NCT03874559Unknown statusRectal CancerYear: 2018
Population: n = 30; Age: 18 years and older; Sex: all
Phase: not reported
[167]
NCT03738319Unknown statusHigh-Grade Serous Carcinoma
Ovarian Cancer
Exosomes
Prognosis
Early Diagnosis
Year: 2018
Population: n = 160; Age: 18 years and older; Sex: female
Phase: not reported
[168]
NCT04720599CompletedUrologic CancerYear: 2020
Population: n = 120; Age: 50 years and older; Sex: male
Phase: not reported
[169]
NCT05101655CompletedOsteosarcoma
Pulmonary Metastases
Year: 2020
Population: n = 60; Age: 12 to 60 years; Sex: all
Phase: not reported
[170]
NCT04315753Unknown statusLung CancerYear: 2018
Population: n = 2000; Age: 55 years and older; Sex: all
Phase: not reported
[171]
NCT03895216CompletedBone MetastasesYear: 2018
Population: n = 34; Age: 18 years and older; Sex: all
Phase: not reported
[172]
NCT04960956TerminatedProstate Cancer
Urothelial Carcinoma
Year: 2016
Population: n = 13; Age: 18 years and older; Sex: male
Phase: not reported
[173]
NCT03911999CompletedProstate CancerYear: 2018
Population: n = 180; Age: 45 years and older; Sex: male
Phase: not reported
[174]
NCT05572099RecruitingProstate CancerYear: 2018
Population: n = 750; Age: 45 years and older; Sex: male
Phase: not reported
[175]
NCT04323579Unknown statusLung CancerYear: 2018
Population: n = 2000; Age: 55 years and older; Sex: all
Phase: not reported
[176]
NCT04357717TerminatedProstate CancerYear: 2020
Population: n = 150; Age: 50 years and older; Sex: male
Phase: not reported
[177]
NCT04100811RecruitingProstate CancerYear: 2020
Population: n = 4000; Age: 45 years and older; Sex: male
Phase: not reported
[178]
NCT05463107Not yet recruitingThyroid Cancer
Follicular Thyroid Cancer
Year: 2022
Population: n = 50; Age: 20 to 80 years; Sex: all
Phase: not reported
[179]
NCT04653740RecruitingAdvanced Breast CancerYear: 2020
Population: n = 25; Age: 18 years and older; Sex: female
Phase: Not applicable
[180]
NCT02147418RecruitingOropharyngeal CancerYear: 2015
Population: n = 30; Age: 18 years and older; Sex: all
Phase: Not reported
[181]
NCT03432806RecruitingColon Cancer
Liver Tumors
Year: 2017
Population: n = 80; Age: 18 years and older; Sex: all
Phase: Not reported
[182]
NCT05397548RecruitingGastric CancerYear: 2022
Population: n = 700; Age: 18 to 80 years; Sex: all
Phase: Not reported
[183]
NCT03811600CompletedSleep Apnea Syndromes, Obstructive CancerYear: 2019
Population: n = 90; Age: 18 years and older; Sex: all
Phase: not reported
[184]
NCT03108677Active, not recruitingLung Metastases
Osteosarcoma
Year: 2017
Population: n = 90; Age: 12 to 60 years; Sex: all
Phase: not reported
[185]
NCT04499794RecruitingUntreated Advanced NSCLC Patients
FISH-Identified ALK Fusion (Positive or Negative)
Year: 2020
Population: n = 75; Age: 18 years and older; Sex: all
Phase: not reported
[186]
NCT04182893Unknown statusPulmonary NodulesYear: 2019
Population: n = 400; Age: 18 years and older; Sex: all
Phase: not reported
[187]
NCT02464930Unknown statusBarrett’s Esophagus
Gastroesophageal Reflux
Esophageal Adenocarcinoma
Year: 2015
Population: n = 220; Age: 18 years and older; Sex: all
Phase: not reported
[188]
NCT05625529Not yet recruitingPancreas Cancer
Exosomes
Extracellular Vesicles
Pancreatic Neoplasms
Year: 2022
Population: n = 1000; Age: 18 years and older; Sex: all
Phase: not reported
[189]
NCT03581435Unknown statusProteinosis
Gallbladder Carcinoma
Year: 2018
Population: n = 50; Age: 18 years and older; Sex: all
Phase: not reported
[190]
NCT03102268Unknown statusCholangiocarcinoma
Benign Biliary Stricture
Year: 2017
Population: n = 80; Age: 18 years and older; Sex: all
Phase: not reported
[191]
NCT05705583RecruitingRenal Cell CarcinomaYear: 2023
Population: n = 100; Age: 18 years and older; Sex: all
Phase: not reported
[192]
NCT03334708RecruitingPancreatic Cancer
Pancreatic Diseases
Pancreatitis
Pancreatic Cyst
Year: 2017
Population: n = 700; Age: 18 years and older; Sex: all
Phase: not reported
[193]
NCT03800121RecruitingSarcomaYear: 2018
Population: n = 30; Age: 18 years and older; Sex: all
Phase: not reported
[194]
NCT05744076Active, not recruitingMelanomaYear: 2019
Population: n = 150; Age: 18 years and older; Sex: all
Phase: not reported
[195]
NCT04053855RecruitingClear Cell Renal Cell CarcinomaYear: 2020
Population: n = 100; Age: 18 years and older; Sex: all
Phase: not reported
[196]

Author Contributions

Conceptualization, M.L., N.S.O. and S.F.; formal analysis, M.L., N.S.O. and S.F.; investigation, M.L., N.S.O. and S.F.; writing—original draft preparation, N.S.O. and S.F.; writing—review and editing, R.D.R. and D.M.; visualization, R.D.R. and D.M.; supervision, S.F.; project administration, N.S.O. and S.F.; funding acquisition, S.F. All authors have read and agreed to the published version of the manuscript.

Funding

The research leading to this review was supported by a project of the Italian Ministry of Health.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

All the authors have no conflict of interest to declare.

References

  1. Logozzi, M.; Spugnini, E.; Mizzoni, D.; Di Raimo, R.; Fais, S. Extracellular Acidity and Increased Exosome Release as Key Phenotypes of Malignant Tumors. Cancer Metastasis Rev. 2019, 38, 93–101. [Google Scholar] [CrossRef] [PubMed]
  2. 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] [PubMed]
  3. 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] [PubMed]
  4. 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]
  5. 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]
  6. Yáñez-Mó, M.; Siljander, P.R.-M.; Andreu, Z.; Bedina Zavec, A.; Borràs, F.E.; Buzas, E.I.; Buzas, K.; Casal, E.; Cappello, F.; Carvalho, J.; et al. Biological Properties of Extracellular Vesicles and Their Physiological Functions. J. Extracell. Vesicles 2015, 4, 27066. [Google Scholar] [CrossRef]
  7. Zhang, Y.; Liu, Y.; Liu, H.; Tang, W.H. Exosomes: Biogenesis, Biologic Function and Clinical Potential. Cell Biosci. 2019, 9, 19. [Google Scholar] [CrossRef]
  8. Cocucci, E.; Meldolesi, J. Ectosomes and Exosomes: Shedding the Confusion between Extracellular Vesicles. Trends Cell Biol. 2015, 25, 364–372. [Google Scholar] [CrossRef]
  9. 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]
  10. 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]
  11. Fais, S.; O’Driscoll, L.; Borras, F.E.; Buzas, E.; Camussi, G.; Cappello, F.; Carvalho, J.; Cordeiro da Silva, A.; Del Portillo, H.; El Andaloussi, S.; et al. Evidence-Based Clinical Use of Nanoscale Extracellular Vesicles in Nanomedicine. ACS Nano. 2016, 10, 3886–3899. [Google Scholar] [CrossRef]
  12. 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]
  13. 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]
  14. Andreola, G.; Rivoltini, L.; Castelli, C.; Huber, V.; Perego, P.; Deho, P.; Squarcina, P.; Accornero, P.; Lozupone, F.; Lugini, L.; et al. Induction of Lymphocyte Apoptosis by Tumor Cell Secretion of FasL-Bearing Microvesicles. J. Exp. Med. 2002, 195, 1303–1316. [Google Scholar] [CrossRef] [PubMed]
  15. Zhao, H.; Achreja, A.; Iessi, E.; Logozzi, M.; Mizzoni, D.; Di Raimo, R.; Nagrath, D.; Fais, S. The Key Role of Extracellular Vesicles in the Metastatic Process. Biochim. Biophys. Acta Rev. Cancer 2018, 1869, 64–77. [Google Scholar] [CrossRef]
  16. 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]
  17. Raposo, G.; Stoorvogel, W. Extracellular Vesicles: Exosomes, Microvesicles, and Friends. J. Cell Biol. 2013, 200, 373–383. [Google Scholar] [CrossRef]
  18. Nolte-’t Hoen, E.N.M.; Buermans, H.P.J.; Waasdorp, M.; Stoorvogel, W.; Wauben, M.H.M.; ’t Hoen, P.A.C. 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. [Google Scholar] [CrossRef]
  19. Camussi, G.; Deregibus, M.-C.; Bruno, S.; Grange, C.; Fonsato, V.; Tetta, C. Exosome/Microvesicle-Mediated Epigenetic Reprogramming of Cells. Am. J. Cancer Res. 2011, 1, 98–110. [Google Scholar]
  20. Valadi, H.; Ekström, K.; Bossios, A.; Sjöstrand, M.; Lee, J.J.; Lötvall, J.O. Exosome-Mediated Transfer of MRNAs and MicroRNAs Is a Novel Mechanism of Genetic Exchange between Cells. Nat. Cell Biol. 2007, 9, 654–659. [Google Scholar] [CrossRef]
  21. Fais, S.; Logozzi, M.; Lugini, L.; Federici, C.; Azzarito, T.; Zarovni, N.; Chiesi, A. Exosomes: The Ideal Nanovectors for Biodelivery. Biol. Chem. 2013, 394, 1–15. [Google Scholar] [CrossRef] [PubMed]
  22. Properzi, F.; Logozzi, M.; Fais, S. Exosomes: The Future of Biomarkers in Medicine. Biomark. Med. 2013, 7, 769–778. [Google Scholar] [CrossRef] [PubMed]
  23. Alvarez-Erviti, L.; Seow, Y.; Yin, H.; Betts, C.; Lakhal, S.; Wood, M.J.A. Delivery of SiRNA to the Mouse Brain by Systemic Injection of Targeted Exosomes. Nat. Biotechnol. 2011, 29, 341–345. [Google Scholar] [CrossRef] [PubMed]
  24. 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] [PubMed]
  25. 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]
  26. 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] [PubMed]
  27. 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]
  28. Li, J.; Chen, Y.; Guo, X.; Zhou, L.; Jia, Z.; Peng, Z.; Tang, Y.; Liu, W.; Zhu, B.; Wang, L.; et al. GPC1 Exosome and Its Regulatory MiRNAs Are Specific Markers for the Detection and Target Therapy of Colorectal Cancer. J. Cell. Mol. Med. 2017, 21, 838–847. [Google Scholar] [CrossRef] [PubMed]
  29. Soung, Y.H.; Ford, S.; Zhang, V.; Chung, J. Exosomes in Cancer Diagnostics. Cancers 2017, 9, 8. [Google Scholar] [CrossRef]
  30. Zocco, D.; Ferruzzi, P.; Cappello, F.; Kuo, W.P.; Fais, S. Extracellular Vesicles as Shuttles of Tumor Biomarkers and Anti-Tumor Drugs. Front. Oncol. 2014, 4, 267. [Google Scholar] [CrossRef]
  31. Campanella, C.; Caruso Bavisotto, C.; Logozzi, M.; Marino Gammazza, A.; Mizzoni, D.; Cappello, F.; Fais, S. On the Choice of the Extracellular Vesicles for Therapeutic Purposes. Int. J. Mol. Sci. 2019, 20, 236. [Google Scholar] [CrossRef] [PubMed]
  32. 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]
  33. Maugeri, M.; Nawaz, M.; Papadimitriou, A.; Angerfors, A.; Camponeschi, A.; Na, M.; Hölttä, M.; Skantze, P.; Johansson, S.; Sundqvist, M.; et al. Linkage between Endosomal Escape of LNP-MRNA and Loading into EVs for Transport to Other Cells. Nat. Commun. 2019, 10, 4333. [Google Scholar] [CrossRef] [PubMed]
  34. 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]
  35. 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]
  36. Cappello, F.; Logozzi, M.; Campanella, C.; Bavisotto, C.C.; Marcilla, A.; Properzi, F.; Fais, S. Exosome Levels in Human Body Fluids: A Tumor Marker by Themselves? Eur. J. Pharm. Sci. 2017, 96, 93–98. [Google Scholar] [CrossRef]
  37. Caruso Bavisotto, C.; Cappello, F.; Macario, A.J.L.; Conway de Macario, E.; Logozzi, M.; Fais, S.; Campanella, C. Exosomal HSP60: A Potentially Useful Biomarker for Diagnosis, Assessing Prognosis, and Monitoring Response to Treatment. Expert Rev. Mol. Diagn. 2017, 17, 815–822. [Google Scholar] [CrossRef] [PubMed]
  38. 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]
  39. Li, X.; Corbett, A.L.; Taatizadeh, E.; Tasnim, N.; Little, J.P.; Garnis, C.; Daugaard, M.; Guns, E.; Hoorfar, M.; Li, I.T.S. Challenges and Opportunities in Exosome Research-Perspectives from Biology, Engineering, and Cancer Therapy. APL Bioeng. 2019, 3, 011503. [Google Scholar] [CrossRef] [PubMed]
  40. Ludwig, N.; Whiteside, T.L.; Reichert, T.E. Challenges in Exosome Isolation and Analysis in Health and Disease. Int. J. Mol. Sci. 2019, 20, 4684. [Google Scholar] [CrossRef] [PubMed]
  41. 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]
  42. 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] [PubMed]
  43. 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] [PubMed]
  44. 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]
  45. Huber, V.; Fais, S.; Iero, M.; Lugini, L.; Canese, P.; Squarcina, P.; Zaccheddu, A.; Colone, M.; Arancia, G.; Gentile, M.; et al. Human Colorectal Cancer Cells Induce T-Cell Death through Release of Proapoptotic Microvesicles: Role in Immune Escape. Gastroenterology 2005, 128, 1796–1804. [Google Scholar] [CrossRef]
  46. 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] [PubMed]
  47. 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]
  48. Gurunathan, S.; Kang, M.-H.; Jeyaraj, M.; Qasim, M.; Kim, J.-H. Review of the Isolation, Characterization, Biological Function, and Multifarious Therapeutic Approaches of Exosomes. Cells 2019, 8, 307. [Google Scholar] [CrossRef]
  49. Szatanek, R.; Baj-Krzyworzeka, M.; Zimoch, J.; Lekka, M.; Siedlar, M.; Baran, J. The Methods of Choice for Extracellular Vesicles (EVs) Characterization. IJMS 2017, 18, 1153. [Google Scholar] [CrossRef]
  50. 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]
  51. Chandler, W.L.; Yeung, W.; Tait, J.F. A New Microparticle Size Calibration Standard for Use in Measuring Smaller Microparticles Using a New Flow Cytometer. J. Thromb. Haemost. 2011, 9, 1216–1224. [Google Scholar] [CrossRef] [PubMed]
  52. Morales-Kastresana, A.; Telford, B.; Musich, T.A.; McKinnon, K.; Clayborne, C.; Braig, Z.; Rosner, A.; Demberg, T.; Watson, D.C.; Karpova, T.S.; et al. Labeling Extracellular Vesicles for Nanoscale Flow Cytometry. Sci. Rep. 2017, 7, 1878. [Google Scholar] [CrossRef]
  53. 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] [PubMed]
  54. Yu, L.-L.; Zhu, J.; Liu, J.-X.; Jiang, F.; Ni, W.-K.; Qu, L.-S.; Ni, R.-Z.; Lu, C.-H.; Xiao, M.-B. A Comparison of Traditional and Novel Methods for the Separation of Exosomes from Human Samples. BioMed Res. Int. 2018, 2018, 3634563. [Google Scholar] [CrossRef] [PubMed]
  55. 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] [PubMed]
  56. 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]
  57. 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]
  58. 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] [PubMed]
  59. 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]
  60. 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]
  61. Parolini, I.; Federici, C.; Raggi, C.; Lugini, L.; Palleschi, S.; De Milito, A.; Coscia, C.; Iessi, E.; Logozzi, M.; Molinari, A.; et al. Microenvironmental PH Is a Key Factor for Exosome Traffic in Tumor Cells. J. Biol. Chem. 2009, 284, 34211–34222. [Google Scholar] [CrossRef]
  62. O’Driscoll, L.; Stoorvogel, W.; Théry, C.; Buzas, E.; Nazarenko, I.; Siljander, P.; Yáñez-Mó, M.; Fais, S.; Giebel, B.; Yliperttula, M. European Network on Microvesicles and Exosomes in Health and Disease (ME-HaD). Eur. J. Pharm. Sci. 2017, 98, 1–3. [Google Scholar] [CrossRef] [PubMed]
  63. Lener, T.; Gimona, M.; Aigner, L.; Börger, V.; Buzas, E.; Camussi, G.; Chaput, N.; Chatterjee, D.; Court, F.A.; del Portillo, H.A.; et al. Applying Extracellular Vesicles Based Therapeutics in Clinical Trials—An ISEV Position Paper. J. Extracell. Vesicles 2015, 4, 30087. [Google Scholar] [CrossRef]
  64. Lapitz, A.; Arbelaiz, A.; O’Rourke, C.J.; Lavin, J.L.; Casta, A.L.; Ibarra, C.; Jimeno, J.P.; Santos-Laso, A.; Izquierdo-Sanchez, L.; Krawczyk, M.; et al. Patients with Cholangiocarcinoma Present Specific RNA Profiles in Serum and Urine Extracellular Vesicles Mirroring the Tumor Expression: Novel Liquid Biopsy Biomarkers for Disease Diagnosis. Cells 2020, 9, 721. [Google Scholar] [CrossRef] [PubMed]
  65. Mateescu, B.; Kowal, E.J.K.; van Balkom, B.W.M.; Bartel, S.; Bhattacharyya, S.N.; Buzás, E.I.; Buck, A.H.; de Candia, P.; Chow, F.W.N.; Das, S.; et al. Obstacles and Opportunities in the Functional Analysis of Extracellular Vesicle RNA—An ISEV Position Paper. J. Extracell. Vesicles 2017, 6, 1286095. [Google Scholar] [CrossRef]
  66. Arroyo, J.D.; Chevillet, J.R.; Kroh, E.M.; Ruf, I.K.; Pritchard, C.C.; Gibson, D.F.; Mitchell, P.S.; Bennett, C.F.; Pogosova-Agadjanyan, E.L.; Stirewalt, D.L.; et al. Argonaute2 Complexes Carry a Population of Circulating MicroRNAs Independent of Vesicles in Human Plasma. Proc. Natl. Acad. Sci. USA 2011, 108, 5003–5008. [Google Scholar] [CrossRef] [PubMed]
  67. Gu, X.; Wang, C.; Deng, H.; Qing, C.; Liu, R.; Liu, S.; Xue, X. Exosomal PiRNA Profiling Revealed Unique Circulating PiRNA Signatures of Cholangiocarcinoma and Gallbladder Carcinoma. Acta Biochim. Biophys. Sin. 2020, 52, 475–484. [Google Scholar] [CrossRef]
  68. Kitagawa, T.; Taniuchi, K.; Tsuboi, M.; Sakaguchi, M.; Kohsaki, T.; Okabayashi, T.; Saibara, T. Circulating Pancreatic Cancer Exosomal RNAs for Detection of Pancreatic Cancer. Mol. Oncol. 2019, 13, 212–227. [Google Scholar] [CrossRef]
  69. Pan, C.; Stevic, I.; Müller, V.; Ni, Q.; Oliveira-Ferrer, L.; Pantel, K.; Schwarzenbach, H. Exosomal MicroRNAs as Tumor Markers in Epithelial Ovarian Cancer. Mol. Oncol. 2018, 12, 1935–1948. [Google Scholar] [CrossRef]
  70. Shi, M.; Jiang, Y.; Yang, L.; Yan, S.; Wang, Y.-G.; Lu, X.-J. Decreased Levels of Serum Exosomal MiR-638 Predict Poor Prognosis in Hepatocellular Carcinoma. J. Cell. Biochem. 2018, 119, 4711–4716. [Google Scholar] [CrossRef]
  71. Kahlert, C.; Melo, S.A.; Protopopov, A.; Tang, J.; Seth, S.; Koch, M.; Zhang, J.; Weitz, J.; Chin, L.; Futreal, 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. [Google Scholar] [CrossRef] [PubMed]
  72. Ciftci, E.; Bozbeyoglu, N.; Gursel, I.; Korkusuz, F.; Bakan Misirlioglu, F.; Korkusuz, P. Comparative Analysis of Magnetically Activated Cell Sorting and Ultracentrifugation Methods for Exosome Isolation. PLoS ONE 2023, 18, e0282238. [Google Scholar] [CrossRef] [PubMed]
  73. Brambilla, D.; Sola, L.; Ferretti, A.M.; Chiodi, E.; Zarovni, N.; Fortunato, D.; Criscuoli, M.; Dolo, V.; Giusti, I.; Murdica, V.; et al. EV Separation: Release of Intact Extracellular Vesicles Immunocaptured on Magnetic Particles. Anal. Chem. 2021, 93, 5476–5483. [Google Scholar] [CrossRef] [PubMed]
  74. Pallares-Rusiñol, A.; Bernuz, M.; Moura, S.L.; Fernández-Senac, C.; Rossi, R.; Martí, M.; Pividori, M.I. Advances in Exosome Analysis. In Advances in Clinical Chemistry; Elsevier: Amsterdam, The Netherlands, 2023; Volume 112, pp. 69–117. ISBN 978-0-443-19284-5. [Google Scholar]
  75. Wen, X.; Pu, H.; Liu, Q.; Guo, Z.; Luo, D. Circulating Tumor DNA-A Novel Biomarker of Tumor Progression and Its Favorable Detection Techniques. Cancers 2022, 14, 6025. [Google Scholar] [CrossRef]
  76. Soltész, B.; Pös, O.; Wlachovska, Z.; Budis, J.; Hekel, R.; Strieskova, L.; Liptak, J.B.; Krampl, W.; Styk, J.; Németh, N.; et al. Mitochondrial DNA Copy Number Changes, Heteroplasmy, and Mutations in Plasma-Derived Exosomes and Brain Tissue of Glioblastoma Patients. Mol. Cell. Probes 2022, 66, 101875. [Google Scholar] [CrossRef]
  77. Choi, J.; Cho, H.Y.; Jeon, J.; Kim, K.-A.; Han, Y.D.; Ahn, J.B.; Wortzel, I.; Lyden, D.; Kim, H.S. Detection of Circulating KRAS Mutant DNA in Extracellular Vesicles Using Droplet Digital PCR in Patients with Colon Cancer. Front. Oncol. 2022, 12, 1067210. [Google Scholar] [CrossRef]
  78. Zhang, X.; Hou, H.; Jiang, M.; Zhang, X. Aberrant Circulating Tumor DNA Methylation and Exosomal MicroRNA Biomarkers for Early Detection of Colorectal Cancer. Mol. Biol. Rep. 2023, 50, 2743–2750. [Google Scholar] [CrossRef]
  79. Xu, X.; Zhang, L.; Liu, J.; Kong, X.; Yin, Y.; Jia, Z.; Zhang, X.; Peng, B.; Ji, M.; Pan, W. Exosomal HBV-DNA for Diagnosis and Treatment Monitoring of Chronic Hepatitis B. Open Life Sci. 2023, 18, 20220585. [Google Scholar] [CrossRef]
  80. 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]
  81. 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]
  82. 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]
  83. 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]
  84. Eichelser, C.; Stückrath, I.; Müller, V.; Milde-Langosch, K.; Wikman, H.; Pantel, K.; Schwarzenbach, H. Increased Serum Levels of Circulating Exosomal MicroRNA-373 in Receptor-Negative Breast Cancer Patients. Oncotarget 2014, 5, 9650–9663. [Google Scholar] [CrossRef] [PubMed]
  85. Hannafon, B.N.; Trigoso, Y.D.; Calloway, C.L.; Zhao, Y.D.; Lum, D.H.; Welm, A.L.; Zhao, Z.J.; Blick, K.E.; Dooley, W.C.; Ding, W.Q. Plasma Exosome MicroRNAs Are Indicative of Breast Cancer. Breast Cancer Res. 2016, 18, 90. [Google Scholar] [CrossRef]
  86. 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]
  87. 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] [PubMed]
  88. 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] [PubMed]
  89. 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]
  90. 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]
  91. 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]
  92. Liu, C.; Eng, C.; Shen, J.; Lu, Y.; Takata, Y.; Mehdizadeh, A.; Chang, G.J.; Rodriguez-Bigas, M.A.; Li, Y.; Chang, P.; et al. Serum Exosomal MiR-4772-3p Is a Predictor of Tumor Recurrence in Stage II and III Colon Cancer. Oncotarget 2016, 7, 76250–76260. [Google Scholar] [CrossRef]
  93. Tsukamoto, M.; Iinuma, H.; Yagi, T.; Matsuda, K.; Hashiguchi, Y. Circulating Exosomal MicroRNA-21 as a Biomarker in Each Tumor Stage of Colorectal Cancer. Oncology 2017, 92, 360–370. [Google Scholar] [CrossRef] [PubMed]
  94. Liu, L.; Meng, T.; Yang, X.-H.; Sayim, P.; Lei, C.; Jin, B.; Ge, L.; Wang, H.-J. Prognostic and Predictive Value of Long Non-Coding RNA GAS5 and MircoRNA-221 in Colorectal Cancer and Their Effects on Colorectal Cancer Cell Proliferation, Migration and Invasion. Cancer Biomark. 2018, 22, 283–299. [Google Scholar] [CrossRef] [PubMed]
  95. Tanaka, Y.; Kamohara, H.; Kinoshita, K.; Kurashige, J.; Ishimoto, T.; Iwatsuki, M.; Watanabe, M.; Baba, H. Clinical Impact of Serum Exosomal MicroRNA-21 as a Clinical Biomarker in Human Esophageal Squamous Cell Carcinoma. Cancer 2013, 119, 1159–1167. [Google Scholar] [CrossRef]
  96. Yoon, J.H.; Ham, I.-H.; Kim, O.; Ashktorab, H.; Smoot, D.T.; Nam, S.W.; Lee, J.Y.; Hur, H.; Park, W.S. Gastrokine 1 Protein Is a Potential Theragnostic Target for Gastric Cancer. Gastric Cancer 2018, 21, 956–967. [Google Scholar] [CrossRef] [PubMed]
  97. Kalniņa, Z.; Meistere, I.; Kikuste, I.; Tolmanis, I.; Zayakin, P.; Linē, A. Emerging Blood-Based Biomarkers for Detection of Gastric Cancer. World J. Gastroenterol. 2015, 21, 11636–11653. [Google Scholar] [CrossRef] [PubMed]
  98. 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]
  99. Yang, H.; Fu, H.; Wang, B.; Zhang, X.; Mao, J.; Li, X.; Wang, M.; Sun, Z.; Qian, H.; Xu, W. Exosomal MiR-423-5p Targets SUFU to Promote Cancer Growth and Metastasis and Serves as a Novel Marker for Gastric Cancer. Mol. Carcinog. 2018, 57, 1223–1236. [Google Scholar] [CrossRef]
  100. 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]
  101. Sohn, W.; Kim, J.; Kang, S.H.; Yang, S.R.; Cho, J.-Y.; Cho, H.C.; Shim, S.G.; Paik, Y.-H. Serum Exosomal MicroRNAs as Novel Biomarkers for Hepatocellular Carcinoma. Exp. Mol. Med. 2015, 47, e184. [Google Scholar] [CrossRef]
  102. 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]
  103. Wang, J.; Zhou, Y.; Lu, J.; Sun, Y.; Xiao, H.; Liu, M.; Tian, L. Combined Detection of Serum Exosomal MiR-21 and HOTAIR as Diagnostic and Prognostic Biomarkers for Laryngeal Squamous Cell Carcinoma. Med. Oncol. 2014, 31, 148. [Google Scholar] [CrossRef] [PubMed]
  104. 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]
  105. Wang, R.-J.; Zheng, Y.-H.; Wang, P.; Zhang, J.-Z. Serum MiR-125a-5p, MiR-145 and MiR-146a as Diagnostic Biomarkers in Non-Small Cell Lung Cancer. Int. J. Clin. Exp. Pathol. 2015, 8, 765–771. [Google Scholar]
  106. Cazzoli, R.; Buttitta, F.; Di Nicola, M.; Malatesta, S.; Marchetti, A.; Rom, W.N.; Pass, H.I. MicroRNAs Derived from Circulating Exosomes as Noninvasive Biomarkers for Screening and Diagnosing Lung Cancer. J. Thorac. Oncol. 2013, 8, 1156–1162. [Google Scholar] [CrossRef]
  107. Meng, X.; Müller, V.; Milde-Langosch, K.; Trillsch, F.; Pantel, K.; Schwarzenbach, H. Diagnostic and Prognostic Relevance of Circulating Exosomal MiR-373, MiR-200a, MiR-200b and MiR-200c in Patients with Epithelial Ovarian Cancer. Oncotarget 2016, 7, 16923–16935. [Google Scholar] [CrossRef] [PubMed]
  108. Madhavan, B.; Yue, S.; Galli, U.; Rana, S.; Gross, W.; Müller, M.; Giese, N.A.; Kalthoff, H.; Becker, T.; Büchler, M.W.; et al. Combined Evaluation of a Panel of Protein and MiRNA Serum-Exosome Biomarkers for Pancreatic Cancer Diagnosis Increases Sensitivity and Specificity. Int. J. Cancer 2015, 136, 2616–2627. [Google Scholar] [CrossRef]
  109. 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]
  110. Goto, T.; Fujiya, M.; Konishi, H.; Sasajima, J.; Fujibayashi, S.; Hayashi, A.; Utsumi, T.; Sato, H.; Iwama, T.; Ijiri, M.; et al. An Elevated Expression of Serum Exosomal MicroRNA-191, - 21, -451a of Pancreatic Neoplasm Is Considered to Be Efficient Diagnostic Marker. BMC Cancer 2018, 18, 116. [Google Scholar] [CrossRef]
  111. Takahasi, K.; Iinuma, H.; Wada, K.; Minezaki, S.; Kawamura, S.; Kainuma, M.; Ikeda, Y.; Shibuya, M.; Miura, F.; Sano, K. Usefulness of Exosome-Encapsulated MicroRNA-451a as a Minimally Invasive Biomarker for Prediction of Recurrence and Prognosis in Pancreatic Ductal Adenocarcinoma. J. Hepato-Biliary-Pancreat. Sci. 2018, 25, 155–161. [Google Scholar] [CrossRef]
  112. Logozzi, M.; Di Raimo, R.; Properzi, F.; Barca, S.; Angelini, D.F.; Mizzoni, D.; Falchi, M.; Battistini, L.; Fais, S. Nanovesicles Released by OKT3 Hybridoma Express Fully Active Antibodies. J. Enzym. Inhib. Med. Chem. 2021, 36, 175–182. [Google Scholar] [CrossRef] [PubMed]
  113. 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] [PubMed]
  114. Fang, S.; Tian, H.; Li, X.; Jin, D.; Li, X.; Kong, J.; Yang, C.; Yang, X.; Lu, Y.; Luo, Y.; et al. Clinical Application of a Microfluidic Chip for Immunocapture and Quantification of Circulating Exosomes to Assist Breast Cancer Diagnosis and Molecular Classification. PLoS ONE 2017, 12, e0175050. [Google Scholar] [CrossRef] [PubMed]
  115. 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]
  116. 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]
  117. 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]
  118. 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]
  119. 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]
  120. 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]
  121. 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] [PubMed]
  122. 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. [Google Scholar] [CrossRef] [PubMed]
  123. 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] [PubMed]
  124. 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]
  125. 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, 003. [Google Scholar] [CrossRef] [PubMed]
  126. 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]
  127. Que, R.; Ding, G.; Chen, J.; Cao, L. Analysis of Serum Exosomal MicroRNAs and Clinicopathologic Features of Patients with Pancreatic Adenocarcinoma. World J. Surg. Onc. 2013, 11, 219. [Google Scholar] [CrossRef] [PubMed]
  128. Khan, S.; Jutzy, J.M.S.; Valenzuela, M.M.A.; Turay, D.; Aspe, J.R.; Ashok, A.; Mirshahidi, S.; Mercola, D.; Lilly, M.B.; Wall, N.R. Plasma-Derived Exosomal Survivin, a Plausible Biomarker for Early Detection of Prostate Cancer. PLoS ONE 2012, 7, e46737. [Google Scholar] [CrossRef]
  129. Gabriel, K.; Ingram, A.; Austin, R.; Kapoor, A.; Tang, D.; Majeed, F.; Qureshi, T.; Al-Nedawi, K. Regulation of the Tumor Suppressor PTEN through Exosomes: A Diagnostic Potential for Prostate Cancer. PLoS ONE 2013, 8, e70047. [Google Scholar] [CrossRef]
  130. Bryant, R.J.; Pawlowski, T.; Catto, J.W.F.; Marsden, G.; Vessella, R.L.; Rhees, B.; Kuslich, C.; Visakorpi, T.; Hamdy, F.C. Changes in Circulating MicroRNA Levels Associated with Prostate Cancer. Br. J. Cancer 2012, 106, 768–774. [Google Scholar] [CrossRef]
  131. Huang, X.; Yuan, T.; Liang, M.; Du, M.; Xia, S.; Dittmar, R.; Wang, D.; See, W.; Costello, B.A.; Quevedo, F.; et al. Exosomal MiR-1290 and MiR-375 as Prognostic Markers in Castration-Resistant Prostate Cancer. Eur. Urol. 2015, 67, 33–41. [Google Scholar] [CrossRef]
  132. Li, Z.; Ma, Y.-Y.; Wang, J.; Zeng, X.-F.; Li, R.; Kang, W.; Hao, X.-K. Exosomal MicroRNA-141 Is Upregulated in the Serum of Prostate Cancer Patients. Onco Targets Ther. 2016, 9, 139–148. [Google Scholar] [CrossRef] [PubMed]
  133. Exosome Diagnostics, Inc. A Prospective, Randomized Blinded, Shared Decision Impact Trial of the ExoDx Prostate (IntelliScore), EPI Test, in Men Presenting for Initial Biopsy. 2022. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  134. Centre Oscar Lambret. Benefit of Analyzing Exosomes in the Cerebrospinal Fluid During the Medical Care of Breast Cancer Patients with Suspicion of Leptomeningeal Metastasis. 2021. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  135. Centre Oscar Lambret. Feasibility of Exosome Analysis in Cerebrospinal Fluid During the Diagnostic Workup of Metastatic Meningitis from Breast Cancer. 2023. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  136. Ospedale Policlinico San Martino. Development of a Horizontal Data Integration Classifier for Noninvasive Early Diagnosis of Breast Cancer. 2022. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  137. Centre Georges Francois Leclerc. Pilot Study with the Aim to Quantify a Stress Protein in the Blood and in the Urine for Early Diagnosis of Malgnant Solid Tumors. 2020. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  138. Poitiers University Hospital. Interest of Circulating Tumor DNA in Digestive and Gynecologic/Breast Cancer. 2023. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  139. Im, Y.-H. Genetic Characteristics of Metastatic Breast Cancer Patients. 2020. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  140. University Hospital. Montpellier Early Detection of Prostate Cancer by Liquid Biopsy. 2021. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  141. Dillhoff, M. Imaging and Detection of Micrometer Sized Early Stage Pancreatic Cancer by Using Endoscopic Ultra-High Resolution Optical Coherence Tomography (OCT) Using Resected Pancreatic Specimen, a Pilot Study. 2022. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  142. David, A. Phase I Study in Humans Evaluating the Safety of Rectus Sheath Implantation of Diffusion Chambers Encapsulating Autologous Malignant Glioma Cells Treated with Insulin-like Growth Factor Receptor-1 Antisense Oligodeoxynucleotide (IGF-1R/AS ODN) in 32 Patients with Newly Diagnosed Malignant Glioma. 2020. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  143. Gu, A. Exosomes Detection for the Prediction of the Efficacy and Adverse Reactions of Anlotinib in Patients with Advanced NSCLC. 2022. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  144. Wang, J. Study on the Prediction of Immunotherapeutic Effect of Advanced Non-Small Cell Lung Cancer by Detection of Plasma Exosomes. 2020. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  145. Cheng, B. Diagnostic and Prognostic Values of EUS-FNA Specimens and Circulating Exosomal Small RNA in Patients with Pancreatic Cancer. 2020. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  146. Gong, J. Combined Diagnosis of CT and Exosome in Early Lung Cancer. 2018. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  147. Kim, H.K. Multicenter Clinical Research for Early Diagnosis of Lung Cancer Using Blood Plasma. 2021. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  148. Lv, Y. Acquisition of Portal Venous Circulating Tumor Cells and Exosomes from Patients with Pancreatic Cancer by Endoscopic Ultrasound: A Prospective Study. 2019. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  149. Jin, Y. Serum Exosomal Long Noncoding RNAs as Potential Biomarkers for Lung Cancer Diagnosis. 2021. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  150. De Reims, C. Identification of New Diagnostic Protein Markers for Colorectal Cancer in Circulating Tumor Exosomes. 2022. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  151. miR Scientific LLC. Establishment of the Performance of the MiR SentinelTM BCa Test in Men and Women Presenting for Initial Diagnosis of Bladder Cancer and the MiR SentinelTM BCR Test to Identify Recurrent Bladder Cancer. 2020. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  152. Susan, E.; Leo, W. A Pilot Study of Tumor-Derived Exosomes as Diagnostic and Prognostic Markers in Breast Cancer Patients Receiving Neoadjuvant Chemotherapy. 2017. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  153. Kim, H.K. A Retrospective Study to Compare Biomarker Expression of Exosomes Derived from Peripheral Blood and Primary Lung Cancer Drainage Pulmonary Blood in Lung Cancer Patients. 2022. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  154. Sun, Y.-S. Memorial Hospital of Sun Yat-Sen University a Prospective, Multicenter Cohort Study of Urinary Exosome LncRNAs for Preoperative Diagnosis of Lymphatic Metastasis in Patients with Bladder Cancer. 2023. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  155. University Hospital. Bordeaux Diagnostic Accuracy of Circulating Tumor Cells (CTCs) and Onco-Exosome Quantification in the Diagnosis of Pancreatic Cancer-PANC-CTC. 2018. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  156. Exosome Diagnostics, Inc. Clinical Validation of a Urinary Exosome Gene Signature in Men Presenting for Suspicion of Prostate Cancer. 2016. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  157. Centre Hospitalier Universitaire Dijon. Contents of Circulating Extracellular Vesicles: Biomarkers in Colorectal Cancer Patients. 2020. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  158. Hackensack Meridian Health. Quantification and Purification of Circulating Prostasomes as Diagnostic Tool for Prostate Cancer Detection. 2023. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  159. miR Scientific LLC. Evaluation of the Training Plan and Impact on Acceptance of the SentinelTM Prostate Cancer Classifier Platform (SentinelTM PCC4 Assay) and Determination of Assay Performance Characteristics of Clinical Utility in a Puerto Rican Population. 2022. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  160. Memorial Sloan Kettering Cancer Center. Interrogation of Exosome-Mediated Intercellular Signaling in Patients with Pancreatic Cancer. 2022. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  161. Pazo-Cid, R.A. Circulating Exosomes as Potential Prognostic and Predictive Biomarkers in Advanced Gastric Cancer Patients: A Prospective Observational Study (“EXO-PPP Study”). 2015. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  162. Alameldin, S. Cell Free Circulating Nucleic Acids as New Tumor Diagnostics from Human Plasma Samples. 2019. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  163. Exosome Diagnostics, Inc. Detection of ARv7 in the Plasma of Men with Advanced Metastatic Castrate Resistant Prostate Cancer (MCRP). 2022. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  164. King’s College London. Lung Cancer Detection Using Blood Exosomes and HRCT- Improving the Early Detection of Lung Cancer by Combining Exosomal Analysis of Hypoxia with Standard of Care Imaging. 2020. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  165. University Hospital. Limoges Analyse Du Profil Moléculaire des Exosomes de La Veine Pulmonaire Dans Le Cancer Bronchique de Stade Précoce. 2023. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  166. King’s College London. HERdi PREDICT: A Pilot Study to Measure the Expression of the HER2-HER3 Dimer in Samples from Patients with HER2 Positive Breast Cancer Receiving HER2 Targeted Therapies. 2022. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  167. Hoover, A. Exosomal as Correlative Biomarker in Clinical Outcomes in Patients Undergoing Neoadjuvant Chemoradiation Therapy for Rectal Cancer. 2019. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  168. Li, L. Non-Coding RNA in the Exosome of the Epithelia Ovarian Cancer. 2018. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  169. Exosome Diagnostics, Inc. Clinical Evaluation of ExoDxTM Prostate (IntelliScore) in Men Presenting for Initial Prostate Biopsy. 2021. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  170. Ruijin Hospital. Construction and Clinical Application of Microfluidic Exosome Chip for Early Diagnosis of Pulmonary Metastasis of Osteosarcoma. 2022. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  171. Istituto Clinico Humanitas. Circulating and Imaging Biomarkers to Improve Lung Cancer Management and Early Detection. 2020. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  172. MD, G.G. Identification and Characterization of Predictive Factors of Onset of Bone Metastases in Cancer Patients. 2022. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  173. Exosome Diagnostics, Inc. Clinical Evaluation of the “ExoDx Prostate IntelliScore” in Men Presenting for Initial Biopsy; Additional Confirmation Study Including Impact on Decision-Making and Health Economics. 2020. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  174. Fai, C. To Investigate the Diagnostic Accuracy of Exosomal MicroRNA in Predicting the Aggressiveness of Prostate Cancer in Chinese Patients. 2021. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  175. Exosome Diagnostics, Inc. ExoDx Prostate Evaluation in Active Surveillance Patient Population. 2022. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  176. Istituto Clinico Humanitas. Validation of Multiparametric Models and Circulating and Imaging Biomarkers to Improve Lung Cancer EARLY Detection. 2020. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  177. Exosome Diagnostics, Inc. Clinical Evaluation of ExoDxTM Prostate (IntelliScore) in Men with Prior Negative Prostate Biopsy Presenting for a Repeat Biopsy. 2022. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  178. miR Scientific LLC. Validating the MiR Scientific SentinelTM Platform (Sentinel PCC4 Assay) in Men Undergoing Core Needle Biopsy Due to Suspicion of Prostate Cancer for Distinguishing Between No Cancer, Low-, Intermediate- and High-Risk Prostate Cancer. 2021. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  179. National Taiwan University Hospital. Correlation Between Various Urinary Exosomal Protein Biomarkers and Pathological Manifestation in Thyroid Follicular Neoplasm: Early and Pre-Operative Diagnosis of Follicular Thyroid Cancer. 2022. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  180. Centre Oscar Lambret. Omic Technologies to Track Resistance to Palbociclib in Metastatic Breast Cancer (OMERIC): A Cohort Study. 2020. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  181. New Mexico Cancer Care Alliance. An Observational, Single-Institution Pilot/Feasibility Study of Exosome Testing as a Screening Modality for Human Papillomavirus-Positive Oropharyngeal Squamous Cell Carcinoma. 2022. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  182. Memorial Sloan Kettering Cancer Center. Development of Novel Imaging and Laboratory Biomarkers to Monitor the Liver Pre-Metastatic Niche and Guide Treatment of Colon Cancer: A Pilot Study. 2023. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  183. Chen, L. Use of Circulating Exosomal LncRNA-GC1 as Blood Biomarker for Early Detection and Monitoring Gastric Cancer. 2022. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  184. University Hospital. Angers Exosomes Implication in PD1-PD-L1 Pathway Activation in Obstructive Sleep Apnea Syndrome. 2023. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  185. Shen, Y. A Pilot Study of Circulating Exosome RNA as Diagnostic and Prognostic Markers in Lung Metastases of Primary High-Grade Osteosarcoma. 2022. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  186. Liu, Y. The Study of Exosome EML4-ALK Fusion in NSCLC Clinical Diagnosis and Dynamic Monitoring. 2020. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  187. Sun, J. Clinical Study of CtDNA and Exosome Combined Detection to Identify Benign and Malignant Pulmonary Nodules. 2019. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  188. Bansal, A. Evaluation of MicroRNA Expression in Blood and Cytology Specimens as a Novel Method for Detecting Barrett’s Esophagus. 2015. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  189. Dhani, H. Exoluminate Study: Observational Registry Study to Assess Exo-PDAC Assay Performance for Detection of Pancreatic Adenocarcinoma (PDAC) in High-Risk or Clinically Suspicious Patients. 2022. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  190. Liu, Y. A Study of Circulating Exosome Proteomics in Gallbladder Carcinoma Patients. 2018. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  191. The Second Hospital of Nanjing Medical University. Exosomes-Derived NcRNAs as Biomarkers in Cholangiocarcinoma Patients. 2017. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  192. An, Z. A Companion Diagnostic Study to Develop Circulating Exosomes as Predictive Biomarkers for the Response to Immunotherapy in Renal Cell Carcinoma. 2023. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  193. Memorial Sloan Kettering Cancer Center. Development of Biomarkers for the Early Detection, Surveillance and Monitoring of Pancreatic Ductal Adenocarcinoma. 2023. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  194. Centre Georges Francois Leclerc. Study of Exosomes in Monitoring Patients with Sarcoma (EXOSARC). 2022. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  195. Centre Hospitalier Universitaire de Besancon. Analysis of Circulating Exosomes in Melanoma Patients. 2023. Available online: clinicaltrials.gov (accessed on 5 April 2023).
  196. Centre Hospitalier Universitaire de Saint Etienne. Evaluation of Urinary Exosomes Presence from Clear Cell Renal Cell Carcinoma. 2022. Available online: clinicaltrials.gov (accessed on 5 April 2023).
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

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. https://doi.org/10.3390/cancers15112878

AMA Style

Logozzi M, Orefice NS, 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(11):2878. https://doi.org/10.3390/cancers15112878

Chicago/Turabian Style

Logozzi, Mariantonia, Nicola Salvatore Orefice, Rossella Di Raimo, Davide Mizzoni, and Stefano Fais. 2023. "The Importance of Detecting, Quantifying, and Characterizing Exosomes as a New Diagnostic/Prognostic Approach for Tumor Patients" Cancers 15, no. 11: 2878. https://doi.org/10.3390/cancers15112878

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