Skip to main content
Erschienen in: Molecular Cancer 1/2022

Open Access 01.12.2022 | Review

Exosomes as a new frontier of cancer liquid biopsy

verfasst von: Dan Yu, Yixin Li, Maoye Wang, Jianmei Gu, Wenrong Xu, Hui Cai, Xinjian Fang, Xu Zhang

Erschienen in: Molecular Cancer | Ausgabe 1/2022

download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

Liquid biopsy, characterized by minimally invasive detection through biofluids such as blood, saliva, and urine, has emerged as a revolutionary strategy for cancer diagnosis and prognosis prediction. Exosomes are a subset of extracellular vesicles (EVs) that shuttle molecular cargoes from donor cells to recipient cells and play a crucial role in mediating intercellular communication. Increasing studies suggest that exosomes have a great promise to serve as novel biomarkers in liquid biopsy, since large quantities of exosomes are enriched in body fluids and are involved in numerous physiological and pathological processes. However, the further clinical application of exosomes has been greatly restrained by the lack of high-quality separation and component analysis methods. This review aims to provide a comprehensive overview on the conventional and novel technologies for exosome isolation, characterization and content detection. Additionally, the roles of exosomes serving as potential biomarkers in liquid biopsy for the diagnosis, treatment monitoring, and prognosis prediction of cancer are summarized. Finally, the prospects and challenges of applying exosome-based liquid biopsy to precision medicine are evaluated.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Cancer is a leading cause of death around the world [1]. Mounting evidence suggests that the development of cancer is a dynamic process and diverse components are involved, including tumor cells, stromal cells, and immune cells. Up to now, tissue biopsy has been considered as the most common method for cancer diagnosis [2]. However, the extracted small tissues fail to represent tumor heterogeneity or monitor dynamic tumor progression, and the potential of metastasis may be increased by this invasive method, finally leading to poor survival and prognosis [3]. Due to minimal invasion, liquid biopsy, which collects the specimen of biofluids such as blood and urine, has drawn widespread attention and generated more opportunities for cancer diagnosis as well as real-time monitoring [4]. Exosomes, with a diameter of 40-160 nm, are lipid bi-layer membrane vesicles that are actively released by most cells and stably circulate in body fluids [5, 6]. Originally underestimated as vehicles for disposal of cellular waste products, exosomes are now being recognized as important players in intercellular communication [4, 7]. Accumulating evidence suggests that a variety of bioactive molecules, including nucleic acids, proteins, and lipids, are enriched in exosomes and could be transferred from donor cells to recipient cells, leading to the intracellular transfer of information [4, 810]. The bioactive cargoes in exosomes may be uptaken by recipient cells, facilitating tumorigenesis and tumor progression. In addition, exosomes are involved in the formation of pre-metastatic niche, tumor angiogenesis, and tumor immune suppression. Moreover, exosomes could reflect the altered physiological and pathological state of their parental cells [1114]. These findings have led to the idea that analyzing the circulating exosomes and their derived cargoes may provide new opportunities for cancer liquid biopsy (Fig. 1), highlighting the potential of exosomes as biomarkers for cancer diagnosis, progression monitoring, and prognosis prediction.
Currently, circulating tumor cells (CTCs), circulating tumor DNA (ctDNA) and exosomes have become the three main branches of liquid biopsy [15, 16]. Compared with CTCs and ctDNA, exosomes have shown greater advantages in liquid biopsy. First, the presence of large amounts of exosomes (~ 109 particles/mL) in biofluids contributes to relatively easy obtaining of vesicles, while only several CTCs exist in 1 mL blood samples [14]. Second, exosomes are secreted by living cells and inherent abundant biological information from their parental cells. Therefore, exosome is more representative than ctDNA, which limitedly reflects the information of apoptotic or dead tumor cells [11, 14]. Third, exosomes are innately stable because of their lipid bilayers, and thus stably circulate in physiological conditions even in harsh tumor microenvironment. The high biological stability allows long-term storage of specimens for exosome isolation and detection [17]. Notably, one of the big challenges for the application of exosomes in liquid biopsy is isolation with high efficiency and purity, which arises from their nanoscale size and intrinsic heterogeneity [1820]. Moreover, since cancerous exosomes represent only a small fraction of all exosomes present in body fluids, ultrasensitive and specific detection is a prerequisite for the development of exosome-based cancer diagnostics. To date, a variety of methods have been developed for exosome isolation as well as the detection of exosomal proteins and nucleic acids [2126]. Although notable progress has been made, the limited sensitivity and specificity, low purity and throughput remain significant challenges for academic research and practical use [27]. Therefore, active research is needed to develop an easy-to-operate, high-sensitivity and high-purity platform for exosome separation and detection. In this review, we discussed about recent advance in the isolation and detection of exosomes as well as their clinical application with an emphasis on the newly developed techniques for exosome separation and detection. Furthermore, the application of exosomes as potential biomarkers in cancer liquid biopsy was summarized.

Exosome biogenesis and contents

Exosomes are a heterogeneous group of membrane-structured vesicles actively released by most cells and could be found in many human body fluids, such as blood, saliva, tear and urine [4]. The process of exosome biogenesis involves invagination of plasma membrane, formation of multivesicular bodies (MVBs) and exosome secretion [28]. MVBs are endocytic structures formed by the inward budding of endosomal membranes. Vesicles accumulating inside of MVBs, named intraluminal vesicles (ILVs), are released as exosomes by the fusion of MVBs with plasma membrane [4] (Fig. 2). Extensive studies suggest that donor cell-derived bioactive molecules are enriched in exosomes, which indicates the crucial role of exosomes in genetic information exchange [4, 28]. Notably, the specific RNA components in exosomes have shown great differences compared to those in their parental cells, which may be attributed to the unique process of cargo sorting during exosome formation [29]. The mechanisms of cargo sorting are still unclear, both endosomal sorting complex required for transport (ESCRT)-dependent manner [30, 31] and ESCRT-independent mechanism have been reported to participate in this process [32, 33]. Intriguingly, proteins such as tetraspanins (CD9, CD63 and CD81), heat shock proteins (HSP60, HSP70) and ESCRT-associated components (Alix and TSG101) have been confirmed to be present  in exosomes, which provides certain markers for their identification and detection [34].
Increasing studies have shown that various disease-related proteins and nucleic acids are loaded into exosomes and differentially expressed in tumors of different origin. Hoshino et al. indicated that plasma-derived exosomes could identify specific cancer types and distinguish tumor sources by proteomic analysis. They found that 51 and 19 plasma-derived exosome proteins were specifically identified in pancreatic and lung cancer, respectively [35]. In addition, exosomal CD63 was reported to be high in ovarian cancer while low in lung cancer [36]. CD317 and epidermal growth factor receptors (EGFR) were highly expressed on non-small cell lung cancer (NSCLC)-derived exosomes [37]. Compared with healthy subjects, glypican-1 (GPC-1) was found to significantly increase in serum exosome of patients with pancreatic cancer and could be used for the early detection of pancreatic cancer with 100% diagnostic specificity and sensitivity [38]. In addition to proteins, nucleic acids such as miRNA, mRNA, and lncRNA are differentially distributed in exosomes and could be used as specific cancer biomarkers. For instance, mutant EGFRvIII mRNA was detected in serum exosomes of glioblastoma patients [39]. Zhou et al. reported that the expression level of exosomal miR-15a-5p was 7-19 times higher in endometrial cancer than that in other cancer types [40]. Additionally, high expression of exosomal miR-1247-3p was positively associated with lung metastasis from liver cancer, which indicated a poor outcome [41]. Given that exosomes are secreted by living cells, the specific contents in exosomes could reflect the pathophysiological state of their parental cells, which makes them useful biomarkers for dynamic monitoring of disease progression [42]. In general, cargoes sorted into exosomes can not only supply additional characteristics for their identification, but also provide promising biomarkers for diagnosis, treatment monitoring, and prognosis prediction in patients with cancer, which offers a new tool for liquid biopsy.

Methods for exosome isolation and enrichment

Due to unique formation manner and specific cargo sorting process, exosomes are heterogeneous in size and molecular contents. Exosome separation and enrichment from complicated biological components is essential for basic study and clinical translation. Up to now, a number of methods have been developed that are significantly varied in the amount and purity of isolated exosomes.

Conventional isolation methods

Ultracentrifugation-based separation

As the gold standard for exosome separation, ultracentrifugation is the most commonly used method [43], including differential ultracentrifugation and gradient density ultracentrifugation. Conventional differential ultracentrifugation was first proposed by Johnstone et al. to isolate exosomes from culture medium of reticulocytes [44]. Typically, low-speed centrifugation (300 g) is first employed to remove cellular debris, while 20,000 g speed of centrifugation is utilized to eliminate other large vesicles. A high force (100,000 g) is finally utilized to sediment exosomes. However, this method needs costly instrument and has contamination with aggregated proteins. More centrifugation cycles may obtain a purer outcome but lead to lower recovery. Gradient density ultracentrifugation is a better alternative to obtain exosomes with higher purity [45]. During centrifugation, different sizes of particles from two or more solutions are separated into different layers, whose density increases from top to bottom. Based on this, the method has been applied to separate exosomes, which have been found to float with densities of 1.15 to 1.19 g/mL [45, 46]. Although with higher purity than ultracentrifugation, limitations such as time-consuming process and the requirement of large biofluid volume have largely restrained its use in clinical application.

Size-based separation

The feature of fixed-range diameters allows for the possibility to separate exosomes by size-based methods. Filtration is one of the size-based approaches for exosome separation by using membrane filters with specific pore sizes, which has the advantages of simple operation and effective purification but disadvantage of low yield [47]. Ultrafiltration is usually applied to concentrate exosomes from large amounts of original materials such as cell culture medium [48]. Currently, the combination of ultrafiltration with ultracentrifugation has been widely employed, in which filtration is used to remove cells and large vesicles while the purification of exosomes is achieved by ultracentrifugation [49]. Moreover, size-exclusion chromatography (SEC) could separate biomolecular components according to the size of sample and pore size of gel. During the separation, large molecules are eluted early, while small molecules or particles directly diffuse into the pores [50]. Anita N Böing et al. developed an SEC-based protocol by cross-linked sepharose CL-2B column, which could efficiently isolate exosomes with a diameter larger than 70 nm from platelet-free supernatant [50]. Guo et al. showed that CL-6B column had better performance than CL-2B column in particle yields and purity of exosomes [51]. Compared with centrifugation and filtration methods, SEC has the advantages of gentle processing and nondestructive outcomes [52]. Moreover, the combination of SEC with ultracentrifugation may have an improved recovery and purity [53].

Precipitation techniques

As for precipitation techniques, highly hydrophilic polymer is used to competitively bind to water molecules around the exosomal membrane, thereby reducing the solubility and finally achieving low input volume of exosome separation. To date, polyethylene glycol (PEG) is the most commonly used polymer for exosome separation [54]. An exosome purification method called ExtraPEG was proposed by Rider et al., which could rapidly enrich exosomes and obtain sufficient contents harvested from vesicles for downstream analysis [55]. Recently, many commercial kits that rely on precipitation techniques have been developed for exosome isolation and enrichment, such as ExoQuick™ and Total Exosome Isolation. Study from Ding et al. indicated that ExoQuick™ could generate a relatively high yield of exosomes compared with other kits [56]. However, this method has been often criticized for the high cost and contamination of coprecipitated protein aggregates.

New enrichment methods

The discoveries of exosome-specific markers and components provide a new avenue for separating exosomes as well as exosome subsets (Fig. 3). Through antibodies and aptamers that specifically target tumor-associated proteins such as GPC-1 and EpCAM (epithelial cell adhesion molecule), exosomes of cancer cell origin could be well distinguished from that of normal cell [57]. Moreover, the applications of microbeads, microfluid chip and thermophoresis enabled the rapid and convenient enrichment of exosomes. Herein, we summarized the advantages and limitations of new approaches for exosome enrichment (Table 1).
Table 1
The techniques for exosome separation
Techniques
Methods
Advantages
Disadvantages
Prominent examples
Ref.
Conventional techniques
 Ultracentrifugation-based Separation
Differential ultracentrifugation
High purity; established protocol;
Lengthy process; large sample volume; requires ultracentrifuge
Separation of EVs from reticulocyte culture medium
[44]
Gradient density ultracentrifugation
High purity;
Lengthy process; large sample volume; requires ultracentrifuge
Sucrose gradient-purified prostasomes
[46]
 Size-based Separation
Ultracentrifugation with ultrafiltration
High purity; high yield
Contamination of same-sized vesicles; lack specificity; difficulty in scaling
Separation of urinary exosomes
[49]
size-exclusion chromatography
High yield; gentle processing
Contamination of same-sized vesicles; lack specificity; difficulty in scaling
Isolation of EVs from platelet-free supernatant of platelet concentrates
[50]
 Precipitation
Polyethylene glycol precipitation
Simple; fast isolation
Lack specificity; much contamination; difficulty in scaling
Isolation of exosomes from plasma, cell culture supernatant
[49, 54]
Commercial kits
Simple; fast isolation
Lack specificity; much contamination; high price
Isolation of exosomes from serum and/or plasma
[56]
Novel techniques
 Immunoaffinity Enrichment
Antibody-conjugated platform
Simple; specificity
High-cost; marker dependent
Enrichment of exosomes from clinical samples
[20, 58, 70]
 Magnetic Separation
Antibody-modified magnetic beads
Convenient; high efficiency
High-cost; marker dependent
Separation of exosomes
[36, 7174]
 Physical Feature-based separation
Nanoscale lateral displacement
Reduced membrane blockage; gentle processing
Contamination of same-sized vesicles; lack specificity
On-chip sorting and quantification of exosomes
[75]
Membrane filter
Gentle processing
Contamination of same-sized vesicles; lack specificity
On-chip isolation of intact extracellular vesicles
[62, 76, 77]
Deterministic lateral displacement
Continuous accurate and precise separation
Low throughout and the requirement of high voltage
Efficient isolation of extracellular vesicles
[75, 78]
Size-exclusion chromatography
High yield; gentle processing
Contamination of same-sized vesicles; lack specificity
Efficient isolation of extracellular vesicles
[50, 52, 79]
 Lipid Mediated-Separation
Lipid nanoprobe/TiO2
Minimal damage
Contamination of other phospholipid membrane vesicles; lack specificity
Efficient isolation of extracellular vesicles
[66, 80]
 Acoustic-based microfluidics
Aacoustic radiation force (ARF) and dielectrophoretic (DEP)
Contact-free; high-throughput; continuous separation; wide range of particles
Design and fabrication finer gradations; finer-grade separation of subpopulations
Active sorting of extracellular vesicles
[67, 81]
 Thermophoretic Enrichment
Thermophoresis
Free from pre-isolation; simple; fast isolation
Contamination of same-sized vesicles; lack specificity
Efficient isolation of extracellular vesicles
[68, 69]

Immunoaffinity enrichment

Owing to the priority of simplicity and specificity, immunoaffinity isolation strategy has been used for exosome enrichment in many studies (Fig. 3A). For example, anti-CD81 functionalized microfluidic chip was fabricated by Zhang et al. to isolate exosomes from plasma samples [70]. Yang et al. reported an integrated microfluidic device for exosome separation through forming a sandwich structure of AuNC-exosome-AuR complexes [58] (Fig. 3Aa). This device achieved a yield of 5 × 109 particles from 5 mL urine sample in 30 min. Nevertheless, the dissociation of captured exosomes remains a big challenge, which arises from the strong affinity between antigen and antibody. To address this challenge, Kang et al. developed an exosome-specific dual-patterned immunofiltration (ExoDIF) device [59]. The biotinylated anti-CD63 antibody was immobilized on the surface of inner channel that sequentially pretreated with 3,3′-dithiobissulfosuccinimidylpropionate (DTSSP), biotinylated BSA, and avidin. As a result, nearly 87.1% of exosomes were captured from high dilution of cell culture media. Notably, the captured exosomes could be dissociated by DTT (dithiothreitol) through breaking the embedded disulfide bond of DTSSP (Fig. 3Ab).
Recently, EVs on demand chip (EVOD) was proposed by Kang et al., in which the capture of cancer-related exosome subpopulations was achieved by the reaction between tetrazine-conjugated anti-EpCAM/anti-EGFR antibody (TzAb) and TCO (trans-cyclooctene) functionalized microfluidic surface [82]. This chip was able to selectively isolate 76% more EGFR+ exosomes from cancer patients than that from healthy donors, which exhibited great potential for the early detection of NSCLC. Sun et al. designed an interesting covalent chemistry-mediated EV click chip to recognize, enrich, and recover hepatocellular carcinoma (HCC)-specific exosomes from plasma samples by multi-marker cocktail (anti-EpCAM, anti-ASGPR1 (asialoglycoprotein receptor 1), anti-CD147) [61] (Fig. 3Ac). The proposed exosome purification system achieved more than 81% of recovery yield and more than 85% of purity, providing a novel liquid biopsy tool to detect hepatocellular carcinoma. Despite great significance, these methods are high-cost and marker-dependent. In addition, more attention should be paid to the non-destructive release of captured exosomes.

Magnetic separation and enrichment

Magnetic bead-based immunoaffinity enrichment has attracted much attention in recent years due to the advantages of convenience and high efficiency (Fig. 3A). Generally, exosomes are captured by antibody-modified magnetic beads, which are then separated by magnetic force. For example, Fang et al. conducted CD63 antibody-conjugated magnetic nanoparticles to isolate exosomes [71]. Moreover, novel immuno-affinitive superparamagnetic nanoparticles (IS-NPs) were proved to possess high efficiency, which combined anti-CD63 antibodies with superparamagnetic nanoparticles through the interactions between β-cyclodextrin (β-CD) and 4-aminoazobenzene (AAB) [60] (Fig. 3Ad). α-CD, a competitive agent extracting AAB from the β-CD-AAB inclusion compound, was adapted for the elution of exosomes. As a result, the capture and release efficiency of exosomes from artificial model samples was as high as 80% and 86.5%, respectively. IS-NPs method exhibited higher yield, increased purity and well-retained structural and functional integrity of exosomes than conventional separation methods.
The isolation of exosome subsets, especially cancer-derived exosomes, could be achieved by coupling magnetic beads with antibodies targeting tumor-specific biomarkers. Luo et al. performed a rapid separation and capture of exosomes by immunoaffinity magnetic beads and DNA origami-based aptamer. The quantitative detection of exosomes could be achieved through the combination with DNA fluorescence probe [83]. Li et al. developed a homogenous magneto-fluorescent exosome (hMFEX) nanosensor to separate GPC-1 positive exosomes in 80 μL of plasma from breast cancer patients [84]. He et al. conducted a microfluidic platform to capture tumor-derived exosomes by mixing samples with anti-EpCAM or anti-CA125 antibodies-labeled magnetic beads [36]. In general, the immunomagnetic separation methods hold the potential to facilitate rapid separation and clinical implication of circulating exosomes in desired areas.

Physical feature-based separation

Size-based microfluidic chip was used to separate exosomes from large cell debris or other membranous vesicles [76, 85] (Fig. 3B). Liu et al. designed a size-based exosome total isolation chip (ExoTIC) in which exosomes ranging in 30-200 nm were enriched and purified by multiple nanoporous membranes [62] (Fig. 3Ba). Compared with ultracentrifugation and commercial PEG precipitation kits, ExoTIC obtained much higher yields of exosomes from small volumes of human plasma. However, the blockage of membrane pores greatly limited the continuous separation of exosomes. To overcome this problem, Chen et al. introduced an ultrafast-isolation system, EXODUS, which integrated double coupled harmonic oscillations into a dual membrane filter configuration. By periodic negative pressure and air pressure switching, periodic negative pressure oscillations were generated on the nanoporous anodic aluminum oxide membrane, allowing small particles (i.e., proteins and nucleic acids) and fluids to pass through, while larger exosomes remained in the central chamber. Moreover, two pairs of oscillators enabled the resuspension of particles into the liquid via transverse waves and acoustofluidic streaming, which effectively limited fouling and particle aggregation [86]. In addition, tangential flow filtration (TFF) is a technology that effectively reduces the potential of pore clogging due to the perpendicular state of flow direction and filtration direction. Sunkara et al. developed a microfluidic tangential flow filtration device, Exodisc, to separate exosomes from human plasma and urine, which showed better exosome yield compared to conventional methods [63] (Fig. 3Bb). Deterministic lateral displacement (DLD) has been used for exosome separation since particles with different sizes perform distinct trajectories in a platform with certain angle-displayed micropillars. Wunsch et al. proposed a nanoscale lateral displacement (nano-DLD) array, in which larger vesicles laterally displaced across the array and were collected at a side channel while smaller vesicles flew out of the array in a zigzag mode, finally achieving the collection of urine-derived exosomes [75]. However, DLD was limited by low throughout and the requirement of high voltage and the density and stiffness of vesicles may interfere with DLD-based exosome isolation.

Lipid-based separation

Lipid molecules on the surface of vesicles enable affinition-mediated exosome capture (Fig. 3C). Wan et al. reported a lipid nanoprobe for the rapid separation of exosomes from plasma [80]. The lipid bilayer of exosome was labeled with biotin-tagged 1,2-distearoyl-sn-glycero-3-phosphethanolamine-poly (ethylene glycol) (DSPE-PEG) probes in which DSPE was inserted into the exosome membrane by hydrophobic effect and PEG provided solubility in the aqueous phase. NeutrAvidin-coated magnetic sub-micrometer particles were used for the collection of exosomes through avidin-biotin affinity. As a result, it took only 15 min to isolate exosomes, which greatly shortened the isolation procedure [87]. Notably, cholesterol-PEG1000 of ~ 6.4 nm was considered to minimize steric hindrance in surface immobilization of lipid nanoprobes and thus achieved a much higher capture efficiency. Additionally, molecules absorbed to lipids on exosome membrane have also been applied to capture exosomes [88]. T-cell membrane protein 4 (Tim-4), which has high affinity to phosphatidylserine (PS), has been verified to facilitate simple exosome separation from serum samples, making it easier for rapid downstream analysis [65, 89] (Fig. 3Ca). In addition, the affinity interaction of TiO2 shell and phosphate groups of exosomes is a novel strategy for the enrichment of phosphorylated peptide. Pang et al. used Fe3O4@TiO2 nanoparticles to enrich and separate exosomes within 5 min with a capture efficiency of 96.5% [90]. The combination of Fe3O4@TiO2 nanoparticles and surface-enhanced Raman scattering (SERS) tags could enrich exosomes and achieve in-situ qualification of target miRNAs simultaneously [66] (Fig. 3Cb).

Acoustic-based isolation method

Acoustic-based microfluidics is a simple and efficient method for exosome separation. Typically, ultrasonic waves are applied to samples and the particles undergo different forces and are separated depending on their physical properties such as size and density [81] (Fig. 3D). Anson et al. utilized the ultrasonic waves scattering between micrometer-sized seeding particles and nanoparticles in a resonant cavity to enrich exosomes. The integrated acoustic device enabled fast operation, non-contact and continuous separation of exosomes from urine and plasma samples [91]. Wu et al. developed an acoustofluidic platform for direct, label-free, and contact-free enrichment of exosomes from whole blood. The device integrated two separation modules, one of which was able to remove particles larger than 1 μm in diameter and the other one could isolate exosomes from larger microvesicles and other particles [92]. Recently, Gu et al. reported an acoustically driven spinning droplets device, in which slanted interdigitated transducers (IDTs) were used to allow nanoparticles to move according to the sound waves of varying frequencies. Particles of fixed size could be specifically concentrated by placing two droplets of different sizes next to each other. As a result, the method could isolate exosomes from 5 μL samples in less than 1 min [93]. In addition, Tayebi et al. combined acoustic radiation force (ARF) with dielectrophoretic (DEP), in which high frequency (> 10 MHz) interdigital transducer was placed in the flow path to simultaneously generate ARF and dielectrophoretic force field. Particles in the medium presented lateral translation by the competition between fluid drag forces, ARF, and DEP force fields, resulting in an active separation of extracellular vesicles [67] (Fig. 3Da).

Thermophoretic enrichment

Thermophoresis refers to a phenomenon that particles migrate from space with high- temperature to low-temperature areas via a temperature gradient induced by localized laser heating. To address the challenge of time-consuming isolation and purification procedures, Liu et al. developed a sensitive thermophoretic method to enrich tumor-derived exosomes. By using aptamers that targeted tumor-specific markers, thermophoresis could achieve rapid isolation and enrichment of exosomes from other components without exosome pre-isolation (Fig. 3D). The accumulation of exosomes produced an amplified fluorescence signal of aptamers, which enabled profiling of surface biomarkers of exosomes as well as detection of miRNAs [68] (Fig. 3Db). Tian et al. performed a similar assay to analyze exosomes in 1 μL plasma, which offered a low-cost, sensitive method for liquid biopsy in metastatic breast cancer [69] (Fig. 3Dc).

Exosome characterization

As suggested in the Minimal information for studies of extracellular vesicles 2018 (MISEV2018), the identification of exosomes should include western blot verification of exosome-specific markers and at least two methods for characterization of single exosome [94, 95].

Visible characterization

Transmission electron microscopy (TEM) is considered as the common method to identify and characterize a single exosome with typically cup-shaped structure (Fig. 4A) [62]. For scanning electron microscopy (SEM), images are presented by collecting the electrons ejected from the samples (Fig. 4C) [96]. Notably, the morphology of exosomes may be affected by dehydration during sample handling procedures. On the contrary, cryo-electron microscopy (cryo-EM) is a better choice because it avoids sample fixation and dehydration. Exosomes are analyzed at a very low temperature and exhibit a round structure that is different from TEM images [97] (Fig. 4B). In addition, atomic force microscopy (AFM) could provide information on both surface morphology and material properties (stiffness, adhesion) by amplitude modulation and phase modulation [96] (Fig. 4D). Moreover, specific exosome membrane markers functionalized AFM tips permit the identification and detection of proteins in single exosome [96, 98].

Quantitative characterization

Dynamic light scattering (DLS) is a technique for measuring the distribution of exosome size and zeta potential. The changes of scattered light interfere and intensity can be identified by a sensor, allowing for estimating the size distribution of particles [100102] (Fig. 4E). However, the contamination of protein aggregates and large vesicles makes it challenging for DLS to distinguish them from exosomes [103]. Nanoparticle tracking analysis (NTA) is a widely used method for determining the concentration and size distribution of particles (Fig. 4F). Particles are illuminated by a beam of light and the scattering light signals are collected by optical microscope [100, 104]. Tunable resistive pulse sensing (TRPS) detects the electrical signals generated by changes in ion conductivity when exosomes pass through pores filled with conductive media [105] (Fig. 4G).

Techniques for detecting exosome contents

Conventional protein analysis

Both membrane and cytoplasmic proteins could be detected in exosomes [4]. Some membrane proteins on exosomes are involved in cancer development and progression and thus have been used as targets for exosome isolation and purification [28, 106]. Western blot and enzyme-linked immunosorbent assay (ELISA) are two regular approaches widely used to detect exosomal proteins [7, 107]. However, these methods are faced with complex procedure and low sensitivity problems [108].

New protein detection methods

Colorimetric detection

Up to now, multiply novel techniques have been developed to detect exosomal proteins (Table 2). Colorimetric detection is a method to determine the content of a component by comparing or measuring the color intensity of chromogenic substances (Fig. 5A). Similar to other nanomaterials like Fe3O4 NPs and AuNPs, DNA has the ability of significantly increasing the peroxidase activity of single-walled carbon nanotubes (s-SWCNTs). Xia et al. developed a visible and simple method for the detection of exosomes [109]. Briefly, CD63 aptamer improved the minic peroxidase activity of s-SWCNTs and effectively catalyzed oxidation of 3,3′,5,5′-tetramethylbenzidine (TMB), resulting in a colorless solution turning blue. On the contrary, the addition of exosomes induced the release of aptamer from the surface of nanotubes and the color of solution to turn from dark to light, which could be observed by naked eye or UV-visible spectrometry with a detection limit (LOD) of 5.2 × 105 particles/μL. As previously discussed, ZnO-chip was designed to effectively isolate exosomes. After incubation with primary antibody mixture (anti-CD9/CD63 antibody), exosomes were recognized by HRP (horseradish peroxidase)-labeled secondary antibody (Fig. 5Aa). Finally, a minimal detectable concentration of 2.2 × 104 particles/μL was obtained by UV-visible spectrometry or microplate analyzer [110]. Liang et al. designed a microfluidic system integrated with double membrane filter and ELISA to detect the content of exosomes in urine samples of bladder cancer patients [111]. In addition to common exosomal proteins, colorimetry could be applied to detect cancer-specific proteins as well. For instance, a PSA (prostate-specific antigen) aptamer-based sensor was used for the visual detection of prostate cancer-specific exosomes in 500 μL human plasma [72]. Moreover, Di et al. reported a nanozyme-assisted immunosorbent assay (NAISA), which enabled sensitive and rapid multiplex profiling of exosomal proteins [112] (Fig. 5Ab). The surface proteins of exosomes could be specifically captured by antibodies immobilized on the surface of a microplate and catalyzed a colorimetric reaction. Signal intensity obtained from microplate reader is proportional to the number of target proteins. As a result, NAISA allowed the rapid profiling of multiple exosomal proteins such as CD63, CEA (carcinoembryonic antigen), GPC-3 (Glypican-3), PD-L1 (programmed death-ligand 1), and HER2 (human epidermal growth factor receptor 2) from clinical samples.
Table 2
New technologies for exosomal protein detection
Methods
Exosome sources
Sample volume
Sensing mechanism
Sensing substances
Detection limit
Ref.
Colorimetric Detection
MCF-7 cells and breast cancer patient’s serum
 
H2O2-mediated oxidation of TMB
s-SWCNTs; CD63-specific aptamer
5.2 × 105 particles/μL
[109]
Cell-culture medium and prostate cancer patient’s plasma
500 μL
H2O2-mediated oxidation of TMB
Aptamer-capped Fe3O4 nanoparticles
3.58 × 106 particles/mL
[72]
Urine
100 mL
H2O2-mediated oxidation of TMB
Biotinylated anti-CD63 antibody; streptavidin-labeled HRP
35.0 AU/mL
[76]
MCF-7 cells and cancer patient’s serum
100 μL
H2O2-mediated oxidation of TMB
CD9, CD63 antibody mixture; HRP-labeled secondary antibody
2.2 × 104 particles/μL
[110]
BeWo cell
 
H2O2-mediated oxidation of TMB
Au-NP; Fe2O3NC
103 exosomes/mL
[148]
Fluorescence Detection
Plasma specimens from NSCLC and OVCA patients
30 μL
Chemifluorescence reagents
EpCAM, IGF-1R or CA125 antibodies; AP-conjugated secondary antibody, and the DiFMUP substrate
0.281 pg/mL; 0.383 pg/mL
[36]
SKOV3 cells and plasma of OVCA patients
10 μL
The reaction of SβG with FDG
Biotin conjugated detection antibodies and streptavidin conjugated SβG
21 exosomes/μL
[149]
MCF-7 and MDA-MB-231 cell culture medium
1 mL
Fluorescent carbocyanine dye (DiO)
CD63 antibody functionalized microbead and DIO labelling
 
[150]
Cell culture supernatant and serum from pancreatic cancer patients
 
Fluorescent carbocyanine dye (DiO)
CD63 antibody-functionalized EXOchip
 
[20]
MCF-7 cells and blood samples from cancer patients
100-300 μL
Fluorescent second antibody
Anti-EpCAM antibody and Alexafluor®647-conjugated secondary antibody
 
[59]
Cell-culture medium and plasma from breast cancer patients
 
Fluorescent second antibody
CD63 antibody-coated magnetic beads; fluorescent dye-conjugated antibodies
107 particles/μL
[151]
A549 cancer cell line and plasma samples of lung cancer patients
0.5 μL
Fluorescent aptamer
TMR-aptamer
500 particles/μL
[152]
Serum samples
 
Fluorescent aptamer
CD63 aptamer-modified magnetic beads; Cy3-labeled short sequence
1.0 × 105 particles/μL
[126]
Cancer cell line and plasma samples
500 μL
Fluorescent aptamer
TPE-TAs/aptamer complexes; graphene oxide surface
3.43 × 105 particles/μL
[99]
MDA-MB-231 cell-culture medium and plasma from breast cancer patients
80 μL
Fluorescence quenching
GPC-1 antibody coated magnetic beads; CD63 aptamer
6.56 × 104 particles/μL
[84]
Cancer cell line and serum samples
 
Fluorescence quenching
FAM-labeled aptamers; graphene oxide
1.6 × 105 particles/mL
[113]
Cancer cell line and blood samples
 
Fluorescence quenching
Anti-CD63-PE/MoS2–MWCNT
14.8 × 105 particles/mL
[153]
Electrochemical Detection
Ovarian cancer cell lines and plasma from patients with ovarian cancer
10 μL
Integrated magneto-electrochemical sensor
Immunomagnetic beads; HRP-labeled secondary antibody
3 × 104
[128]
Plasma samples
20 μL
Electrochemical biosensor
Immunomagnetic beads; probing antibodies
 
[129]
Cell-culture medium and blood samples from breast cancer patients
 
Electrochemical biosensor
Anti-PD-L1-linked DNA strand; PVP@HRP@ZIF-8
334 particles/mL
[114]
HepG2 cells and human serum of liver cancer patients
30 μL
Aptamer-based biosensors
CD63 aptamer and mimicking DNAzyme sequence
4.39 × 103 particles/mL
[65]
Culture medium of HepG2 cells
 
Aptamer-based biosensors
NTH-assisted aptasensor
2.09 × 104/mL
[154]
Cell-culture medium and serum
 
Aptamer-based biosensors
Aptamer-magnetic bead bioconjugates; electroactive Ru (NH3)63+
70 particles/μL
[155]
Cellular supernatant and human plasma samples
 
Aptamer-based biosensors
anti-CD63 antibody modified gold electrode and a gastric cancer exosome specific aptamer
9.54 × 102/mL
[156]
Human hepatoma cell lines MHCC97H/L and mouse melanoma cell lines B16-F1/10
 
Antibody microarray SPRi sensor
Anti-CD9, CD41b,21 and tyrosine kinase receptor MET8a antibodies immobilized gold-coated glass sensor chip
 
[157]
SPR Detection
MCF-7 breast cancer cells and MCF-10A normal breast cells
 
SPR-based aptasensor
Dual gold nanoparticle-assisted signal amplification
5 × 103 exosomes/mL
[158]
Human NSCLC cell lines, normal lung cell and plasma
1.5 mL
Bioaffinity interactions of antibodies and different recognition sites
Antibodies modified-gold chip and different recognition sites
104 particles/μL
[159]
Urine samples from lung cancer patients and controls
500 μL
SPR-induced improved scattering intensity
Anti CD81/LRG1 antibody modified nanoporous gold nanocluster membrane; second antibody-conjugated Au nanorod probes
< 1000 particles/mL
[58]
Breast cancer cell line and serum
250 μL
SPR-induced improved scattering intensity
Anti-HER2-functionalised SPR chip
8280 exosomes/μL
[160]
Cancer cells and serum and the CSF of an orthotopic mouse model
 
Strong localization of surface plasmon polaritons
TIC-AFM and TiN–NH-LSPR biosensors
5.29 × 10−1 μg/ml; 3.46 × 10 -3 μg /ml
[161]
Breast cancer cells and normal breast cells; plasma from HER2-positive breast cancer patients
 
Raman reporters
Gold-coated glass microscopy slide; QSY21-coated gold nanorods
2 × 106/mL
[134]
SERS
Plasma of cancer patients
400 μL
P-O bond signature
Beehives-like Au-coated TiO2 macroporous inverse opal
 
[117]
Cell-culture medium and serum sample
4 μL
MBA signature
Fe3O4@TiO2 nanoparticles; anti-PD-L1 antibody modified Au@Ag@MBA
1 PD-L1 exosome/μL
[90]
Normal and lung cancer cell lines; plasma
 
Deep learning
Deep learning model
 
[162]
Cell-culture medium; serum and plasma
< 1 μL
Enrichment of aptamer-bound EVs
Seven aptamers targeting specific proteins; machine-learning algorithm
3.3 × 103/μl
[68]
CRISPR/Cas-assisted detection
A549 cell-culture medium and serum from lung cancer patients
 
CRISPR/Cas12a
CD63 aptamer; CRISPR/Cas12a
Linear range of 3 × 103–6 × 107 particles/μL
[136]
SUNE2 cell-culture medium and serum from NPC patients
50 μL
CRISPR/Cas12a
Nucleolin and PD-L1 aptamers; CRISPR/Cas12a
102 particles/μL
[137]
SUNE2 cell-culture medium and serum from NPC patients
 
CRISPR/Cas12a
CD109 and EGFR aptamers; CRISPR/Cas12a
102 particles/μL
[138]
Single EV Analysis
Human serum
10 μL
Rolling circle amplification
ssDNA-assisted single EV detection platform
82 vesicles/μL
[139]
T3M4 pancreas cancer line and serum from PDAC patients
10 mL
Flow cytometry
Aldehyde/sulfate latex beads; anti-GPC-1 antibody and Alexa-488-tagged antibody
 
[38]
Breast cancer cell lines and serum of breast cancer patients
500 μL
Flow cytometry
Aldehyde/sulfate latex beads; anti-EpCAM or anti-HER2 antibody; Alexa-488- or − 594-tagged secondary antibodies
 
[141]
Human cell lines and serum of glioma patients
250 μL
Flow cytometry
Aldehyde/sulfate latex beads; anti-EGFR or anti-CXCR4 antibody
 
[118]
HCT15 cell-culture medium and plasma
500 μL
Nano-flow cytometry
  
[142]
OVCA Ovarian cancer, PGR Progesterone receptor, ESR1 Estrogen receptor 1, ERBB2 erb-b2 receptor tyrosine kinase 2

Fluorescence detection

Fluorescence spectrophotometry is a method for substance identification and content determination according to the positivity and intensity of fluorescence spectral line (Fig. 5B). He et al. conducted a microfluidic chip to detect surface and intravesicular biomarkers from 30 μL plasma samples. This chip obtained a markedly improved detection sensitivity of IGF-1R (type 1 insulin growth factor receptor), 0.281 pg/mL of IGF-1R and 0.383 pg/mL of p-IGF-1R, which was 100-fold higher than that achieved by ELISA [36] (Fig. 5Ba). Liu et al. developed an immunosorbent assay, in which immunomagnetic beads were utilized to capture exosomes, followed by the conjugation of an enzymatic reporter, which could produce a fluorescent signal for quantitation of GPC-1+ exosomes in the droplet microfluidic system [124]. Wei et al. proposed similar single molecule array (SiMoa) platform, by which universal exosomes and tumor-derived exosomes could be ultrasensitively detected with an LOD of 34 particles/μL and 25 particles/μL, respectively [125]. Yu et al. conducted a CD63 aptamer-based detection method. CD63 on exosomes could bind to aptamers modified on the surface of magnetic beads, resulting in the shedding of a Cy3-labeled short sequence into the supernatant. The fluorescence intensity in the supernatant was used to quantify exosomes in complex clinical samples [126].
Graphene oxide has the ability of quenching fluorescence via fluorescence resonance energy transfer (FRET) when conjugated with fluorescent dyes. For instance, the fluorescence of FAM-labeled aptamers was quenched when absorbed onto graphene oxide membranes, while target exosomes competitively bound to aptamers and re-exhibited fluorescent signals with an LOD of 1.6 × 105 particles/mL [113] (Fig. 5Bb). Li et al. developed a facile fluorescent aptasensor based on aggregation-induced emission luminogens (AIEgens). Graphene oxide absorbed (tetrafrylene-containing tertiary amine) TPE-TAS/aptamer complex allowed fluorescence quenching in the absence of exosomes. When target exosomes were introduced, the aptamer preferentially bound to its target, resulting in the separation of the TPE-TAS/aptamer complex from the graphene oxide surface, followed by a “turn-on” fluorescence signal. The linear range of tumor-derived exosomes was around 4.0 × 105-1.8 × 107 particles/μL under optimized conditions [99].

Electrochemical detection

Electrochemical detection is used to detect the analytes by measuring the electrochemical potential or current of the sample, which has the advantages of high sensitivity and wide measurement range (Fig. 5C). In recent years, electrochemical biosensors have been developed by researchers since the altered electrochemical signals could quantify exosomes when recognition elements such as antibody and aptamer specifically bind to exosomes [127, 128]. Cao et al. proposed an electrochemical biosensor to accurately detect PD-L1+ exosomes [114] (Fig. 5Ca). Exosomes were firstly captured by anti-CD63 functionalized magnetic beads and then bound with anti-PD-L1-linked DNA strand, introducing a hyperbranched rolling circle amplification (HRCA). The HRCA could decrease the environmental pH, leading to the decomposition of PVP@HRP@ZIF-8 and release of HRP, which resulted in amplified electrochemical responses and thus achieved the identification and detection of cancer-derived PD-L1+ exosomes. This biosensor displayed a wide dynamic range for PD-L1+ exosomes (1 × 103 to 1 × 1010 particles/mL) and the detection limit was 334 particles/mL. Jeongmin et al. reported a HiMEX approach which integrated magnetic exosome separation and electrochemical detection of exosome-bound proteins after enzymatic amplification. The combined detection of tumor biomarkers (EGFR, EpCAM, CD24 and GPA33) in exosomes from 20 μL plasma samples were helpful for the diagnosis and monitoring of colorectal cancer [129]. Electrochemical methods, especially aptamer-based biosensors have shown great potential in the detection and profiling of exosomal proteins [65]. Kashefi-Kheyrabadi et al. introduced a detachable microfluidic device implemented with an aptamer-based electrochemical biosensing method (DeMEA). In this system, aptamer targeting EpCAM was immobilized on the electrode surface that was pre-electroplated with gold nanostructures and microfluidic vortexes could increase the collision between exosomes and sensing surface (Fig. 5Cb). Consequently, DeMEA was able to quantify exosomes from plasma samples of breast cancer patients at different stages, which provides a highly sensitive and early detection of cancer-specific exosomes [115].

Surface plasmon resonance detection

Surface plasmon resonance (SPR) is a physically optical phenomenon caused by total reflection of light at the metal film/liquid level interface to analyze molecular interactions (Fig. 5D) [130, 131]. Im et al. developed a nano-plasmonic exosome (nPLEX) assay based on transmission SPR through periodic nanohole arrays (Fig. 5Da). With functionalized antibodies in each array, the nPLEX sensor displayed spectral shifts or intensity changes that were proportional to the levels of target exosomal proteins [116]. In another study, the markedly improved scattering wavelength shift and scattering intensity were observed on AuNC-Exosome-AuR due to the plasmon effect [58]. The differential expression of LRG1 (leucine rich alpha-2-glycoprotein 1) in urinary exosomes between lung cancer patients and healthy individuals was evaluated by anti-LRG1 antibody-conjugated AuR probes. Another surface plasmon resonance imaging (SPRi)-based biosensing assay was developed by Fan et al. The bioaffinity interactions between antibodies (anti-CD63/anti-EGFR/anti-EpCAM) modified-gold chip and different recognition sites permitted the multiplex characterization of NSCLC-derived exosomes. The LOD of this biosensor was estimated to be 104 particles/μL. Despite multiple merits, the broad applications of nanoplasmonic biosensors are restrained by the difficult fabrication of nanostructures. To address this challenge, Liu et al. developed an intensity-modulated SPR biosensor free from nanostructure. In this sensor, the reflection intensity and reference intensity of lasers were recorded by two photodetectors and were used to quantify the expression levels of exosomal proteins, which exhibited a higher detection sensitivity than ELISA [132].

Surface enhanced Raman scattering

Surface enhanced Raman scattering (SERS) is capable of enhancing the Raman signal of small molecules attached to the rough metal surface through electromagnetic and chemical mechanisms [133] (Fig. 5D). A new assay was reported for the real-time detection and protein profiling of exosomes. Generally, gold-plated slides combined with 3D-printed antibody arrays were fabricated to capture exosomes, and QSY21-coated gold nanorods were used as the label agent to quantitatively detect target proteins. The levels of plasma-derived exosomes of breast cancer patients were quantitatively determined by using this assay targeting HER2 and EpCAM. The proposed 3D-printed array template enabled cheap, portable, and easily available establishment of detection platform, providing a new strategy for the development of novel cancer liquid biopsy [134]. Dong et al. showed that the analysis of protein phosphorylation status may provide new possibilities for cancer diagnostics [117] (Fig. 5Db). A beehives-like Au-coated TiO2 macroporous inverse opal structure was developed to capture and analyze exosomes without any labeling process. The intensity of 1087 cm− 1 SERS peak referred to the P-O bond within the phosphoproteins of exosomes and the intensity of peak was at least two times from plasma of cancer patients than that from healthy donors. However, the above-mentioned two assays both require the pre-separation of exosomes, which greatly hinders the rapid analysis. Pang et al. presented a simple immunoassay to capture and analyze exosomal PD-L1 directly from serum samples [90]. Fe3O4@TiO2 nanoparticles were designed to isolate exosomes and anti-PD-L1 antibody modified Au@Ag@MBA SERS tags were applied for exosomal PD-L1 labeling and SERS detection. This assay was confirmed to quantify exosomal PD-L1 in 4 μL serum sample within 40 min.

CRISPR/Cas system-assisted detection

In CRISPR/Cas9 system, the Cas9 nuclease could efficiently shear the double-stranded DNA (dsDNA) sequence by recognizing specific complementary dsDNA containing protospacer adjacent motif (PAM) sequences with the help of guide RNA (gRNA) [135]. Through aptamers that specifically target exosomal proteins, the detection of proteins could be transformed into the quantification of nucleic acids. Recently, Zhao et al. reported a detection method which combined aptamer-based exosomal membrane protein recognition with CRISPR/Cas12-assisted fluorescence signal amplification [136]. In this system, CD63 aptamer specifically targeted exosome membrane proteins, triggering the conformational change of aptamer and release of blocker strands (with complementary sequences to aptamer). The released blocker was then recognized by CRISPR/Cas12a, resulting in the trans-cleavage toward TaqMan probe and the separation of fluorescence reporter group and quenching group, finally leading to the generation of amplified fluorescence signal. As a result, this method achieved a linear detection range of 103-107 particles/μL and was successfully applied in the direct detection of plasma exosomes without ultracentrifugation. Xing et al. developed an apta-HCR-CRISPR assay to detect circulating nucleolin+ or PD-L1+ exosomes from 50 μL serum of nasopharyngeal carcinoma cancer (NPC) patients [137]. Nucleolin or PD-L1-targeted aptamer was first amplified by HCR (hybridization chain reaction) to produce a long-repeated CRISPR-targetable DNA unit. Through collecting the fluorescence signal induced by collateral cleavage activities of CRISPR-Cas12a, the assay enabled a detection limit of 102 particles/μL. A similar assay was conducted by Li et al. for the ultrasensitive detection of CD109+ and EGFR+ exosomes [138]. CRISPR/Cas system is expected to be a sensitive tool for the identification and quantification of exosomal proteins and may contribute to the diagnosis and therapeutic monitoring of cancer.

Single exosome detection

Intrinsic heterogeneity is one of the main factors hindering exosome analysis in body fluids. Single exosome detection may provide more accurate information of tumor progression (Fig. 5E). Guo et al. presented an ssDNA-assisted single exosome detection platform. Rolling circle amplification (RCA) contributed to an amplified fluorescence signal from the surface protein, enabling easy visualization of individual exosomes with an LOD of 82 vesicles/μL [139]. In addition, Liu et al. developed a λ-DNA and aptamer-mediated approach, allowing for two-dimensional analysis of single exosome by size and tumor-associated marker expression [140].
Since nanosized exosomes cannot be sensitively identified by conventional flow cytometry, aldehyde/sulfate latex beads have been used to bind to vesicles, which are then stained with fluorescent antibodies and characterized for their protein markers. For example, anti-GPC-1 antibody and Alexa-488-tagged secondary antibody were introduced to the exosome-attached beads and the percentage of positive beads was therein referred as the percent of GPC-1+ exosomes [38]. In addition, the detection of expressed EpCAM, HER2 and EGFR in exosomes indicated their potential role in cancer diagnostics [118, 141] (Fig. 5Ea). Moreover, the development of nano-flow cytometry has provided a new option for multi-parameter analysis of single particle. The expression of CD9, CD63, CD81, CD235a, CD45, CD41a and CD144 of single exosome was measured via immunofluorescent labelling using nano-flow cytometry to evaluate the quality of exosome preparations isolated by six different methods. Liu et al. used nano-flow cytometry to analyze the expression of CD9, CD63, CD81, CD47, CD45, CD24, and EpCAM in tear-derived exosomes and found that the exosome concentration in tear fluid was approximately 100-fold higher than that of plasma exosomes [142].

Conventional nucleic acids analysis

In addition to protein cargoes, nucleic acids are encapsulated in exosomes as well. RNAs represent the major nucleic acid cargo of exosomes, which have shown the potential to be specific biomarkers for cancer diagnosis and prognosis prediction [2, 143, 144]. Nevertheless, the accuracy and feasibility of detecting exosomal nucleic acids are often hampered by their low abundance. To quantify the expression levels of exosomal nucleic acids, techniques such as qRT-PCR (real-time quantitative reverse transcription PCR), microarray, and next-generation sequencing (NGS) have been used. Despite high sensitivity, qRT-PCR can only be used to detect nucleic acids with known sequences [145]. NGS is beneficial for high-throughput discovery and quantitation of unknown exosomal RNA transcripts. However, shortcomings such as high cost, huge amount of data and complexity of building libraries need to be addressed [146]. Through the complementary combination of hybridized probes and target genes, microarrays can analyze thousands of nucleic acids in exosomes at one time but have the disadvantages of low sensitivity [147]. To overcome these limitations, more efforts are being devoted to developing highly sensitive and convenient methods for exosomal nucleic acid detection (Table 3).
Table 3
New technologies for exosomal nucleic acid detection
Methods
Exosome sources
Sample volume
Nucleic acids
Detection mechanism
Advantages
Disadvantages
Ref.
Droplet digital PCR
Urine
2 mL
miRNA; gene variation
Nucleic acid amplification of droplets in an oil emulsion
Absolute quantification; small sample volume; high accuracy and sensitivity;
High-cost; limited throughout; complex operation
[163]
Cerebrospinal fluid of GBM patients
1 mL
IDH1 mutation
[164]
Plasma samples of HCC patients and control cohorts
90 μL
HCC-specific mRNA
[61]
Cancer cell lines and patient plasma
2 μL
GAPDH mRNA
[165]
Human plasma
 
miR-15a-5p
[40]
Human plasma
 
PGR mRNA; ESR1 mRNA; ERBB2 mRNA
[120]
Clinical blood
1.5 mL
EV-lncRNA of SLC9A3-AS1 and PCAT6
[166]
Serum
100 μL
circHIPK3 and circSM ARCA5
[167]
Molecular beacons
Cancer cells and human serums
35 μL
miRNA-21
Fluorescent, enzyme-labeled oligonucleotide probes identifying and detecting nucleic acid with complementary sequences
High specificity, simplicity; low background fluorescence; rapid detection
High-cost; limited throughout
[122]
Breast cancer cell line and human plasma
 
miR-21; miR-375; and miR-27a
[121]
Prostate cancer cells and human urine
 
miRNA-375 and miRNA-574-3p
[168]
Human plasma
10 μL
miR-1246
[169]
RBC-derived EVs
 
miRNA-451a
[170]
PCA cell
 
miR-21
[151]
DNA tetrahedron probe
Serum
 
miR-21
Leverage localized reaction and cascade amplification
High specificity and sensitivity
High-cost
[171]
Plasma
1 mL
miR-1246; miR-221; miR-375; miR-21
[172]
SPR Detection
Pancreatic cancer cells and plasma
50 μL
miR-10b
The change of dielectric constant caused by molecule adsorption on the heavy metal film
High specificity and sensitivity; label-free
Nonspecific adsorption
[173]
Plasma
 
miRNA
[123]
Mouse serum
 
miR-10b
[174]
Single Vesicle Analysis
Serum
 
hsa-miRNA-21
Single-vesicle imaging
Direct visualization; acknowledgement of heterogeneity at the single-vesicle level
Nonspecific adsorption
[119]
Thermophoretic Detection
Serum
0.5 μL
miRNA
Nanoflare induced amplified fluorescence signal
Without the need for EV pre-isolation; high sensitivity; rapid detection; low cost
 
[175]
CRISPR/Cas-assisted detection
Plasma
500 μL
miRNA-21; miRNA-221; miRNA-222
CRISPR/Cas9
High sensitivity and specificity
 
[176]
PGR Progesterone receptor, ESR1 Estrogen receptor 1, ERBB2 erb-b2 receptor tyrosine kinase 2, PCAT6 Prostate cancer associated transcript 6

New nucleic acids detection technologies

ddPCR

Droplet digital PCR (ddPCR) is a technique in which the PCR reaction mixture is divided into tens of thousands of aqueous droplets in an oil emulsion (Fig. 5F). Each single droplet contains no more than one copy of target gene and is labeled as positive or negative according to the fluorescence amplitude. The concentration of target nucleic acids is then estimated by the Poisson distribution and ratio of the positive droplets [177]. The comparison between ddPCR and qPCR showed that ddPCR had higher accuracy and sensitivity in analyzing urinary exosomal miRNAs [163]. Chen et al. used ddPCR to identify IDH1 (isocitrate dehydrogenase 1) transcripts in exosomes derived from serum or cerebrospinal fluid (CSF) of glioblastoma (GBM) patients. Mutant IDH1 mRNA was identified in CSF-derived exosomes of patients bearing mutant IDH1 glioblastoma and higher level of IDH1 mRNA was found in exosomes from patients with tumors than healthy controls [164]. Sun et al. recently used ddPCR to quantify 10 HCC-specific mRNA from plasma samples of HCC patients (Fig. 5Fa). The diagnostic value of HCC exosome-derived mRNA signatures was evaluated by computing the digital scoring [61]. Moreover, exosomal miR-15a-5p was detected by ddPCR to distinguish endometrial cancer (EC) patients from healthy subjects [40]. Shen et al. reported that more copy numbers of lncRNAs RP11-77G23.5 and PHEX-AS1 were quantified by ddPCR in EpCAM-specific exosomes from malignant lung cancer patients compared to benign lung tumors [178].

Molecular beacons

Molecular beacon (MB) is a hairpin-like oligonucleotide labeled with fluorescent dye and quencher at two ends of the probe (Fig. 5G). MB is designed to spontaneously hybridize with the targeted sequence, thereby destroying the hairpin ring structure and inducing the appearance of fluorescence [179]. Lee et al. observed that high fluorescent signals were obtained by the hybridization of molecular beacons and miRNA-21 in exosomes of cancer cells and human serum [122] (Fig. 5Gb). Moreover, miR-375 and miR-574-3p were detected from exosomes of human urine by MB-based biosensors [168]. The expression levels of miR-21, miR-375, and miR-27a were detected in exosomes of human serum [121] (Fig. 5Ga). Oliveira et al. utilized CPP (cell-penetrating peptides) to deliver MB across the plasma membrane and then to detect miRNA-451a in red blood cell (RBC)-derived exosomes [170]. Chen et al. designed a 2′-O-methyl and phosphorothioate modified molecular beacon to quantitatively analyze exosomal miRNA-1246 from human plasma. After rupturing the exosome membrane with Triton X-100, the probe could specifically target miRNA-1246 inside and display quantitative fluorescence signals [169]. Zhang et al. developed an integrated exosome isolation and detection system, in which exosomes could be separated by microfluidic technology via using a little volume of samples. Meanwhile, nanopore detection technology effectively improved the detection efficiency of tumor-related miRNA without the need for amplification and fluorescence labeling of detected objects [180]. The molecular beacon-based biosensor has the priority of simple procedure, free from exosome pre-isolation and nucleic acid extraction, indicating their great potential in liquid biopsy for cancer diagnosis and prognosis.

DNA tetrahedron probe

DNA tetrahedron, a DNA nanoarchitecture with high controllability, can provide diverse amplified signal tags through chemical modification and DNA self-assembly [181]. Gao et al. conducted a DNA tetrahedron nanoprobe-based FRET sensing platform to sensitively detect miR-146b-5p in different cell lines [182]. Chen et al. reported a hairpin-tetrahedron nanoprobe for quantitative measurement of exosomal miR-21 in human serum [171]. In the absence of miR-21, the construction of fluorophore donor-quencher pairs resulted in a low FRET effect, while the presence of target miRNA induced the damage of stem-loop structure and occurrence of strong FRET. Consequently, the assay obtained a good linearity in the range from 1 × 10− 12 to 10 × 10− 9 M (mol/L) and a detection limit of 45.4 × 10− 15 M. Zhang et al. proposed a similarly electrochemical biosensor based on two multifunctional DNA tetrahedrons assisted catalytic hairpin assembly. By leverage localized reaction and cascade amplification, the sensor enabled sensitive detection of tumor-associated exo-miRNAs down to 7.2 aM in 30 min [172].

Localized surface plasmon resonance

Localized surface plasmon resonance (LSPR) occurs when the incident photon frequency matches the overall vibration frequency of precious metal nanoparticles or metal conducting electrons (Fig. 5H). Joshi et al. developed a biosensor based on LSPR for the label-free and nondestructive measurement of exosomal miR-10b [173]. Gold nanoprisms were chemically synthesized onto a silanized glass substrate and then functionalized with HS-C6-ssDNA and PEG6-SH. The direct hybridization of target miR-10b and HS-C6-ssDNA induced the formation of double-strand DNA which could increase the index of refraction of nanoprisms and change the wavelength of LSPR dipole peak (λLSPR). The concentration of miR-10b could be evaluated by ΔλLSPR. This platform was sensitive enough to distinguish between miR-10b and miR-10a (only one nucleotide difference) even in the subattomolar concentration range. Wu et al. developed an SPRi-based biosensor to detect multiple exosomal miRNAs for accurate diagnosis of NSCLC, in which Au-on-Ag heterostructure and DNA tetrahedral framework were utilized to enhance SPR signal and each exosomal miRNA could be ultrasensitively identified with different SPR signals [123] (Fig. 5Ha).

TIRF-based single-vesicle imaging

As a single-exosome analytic method, total internal reflection fluorescence (TIRF) has emerged for providing additional information about the heterogeneity of exosomes (Fig. 5E). He et al. proposed a TIRF-based single-vesicle imaging assay which delivered molecular beacon probes into exosomes and thus induced an amplified fluorescence of target miRNA (Fig. 5Eb). They performed a direct visualization of single vesicles and in-situ quantitative analysis of miR-21 in human serum samples and found that this assay showed better performance than conventional PCR assay in distinguishing cancer patients from healthy subjects [119].

Thermophoresis-assisted detection

Zhao et al. developed a thermophoretic sensor for in situ detection of exosomal miRNAs without the need of RNA extraction or target amplification (Fig. 5F). Aptamer modified in nanoflares could bind to target exosomal miRNA, inducing the appearance of fluorescence. The fluorescent signal became amplified after thermal electrophoretic accumulation, allowing the sensitive detection of 0.36 fM exosomal miRNA in a small volume of serum sample [175]. In addition, Han et al. proposed a DNA tetrahedron-based thermophoretic assay (DTTA) which achieved a detection limit of 14 aM mRNA in serum exosomes. After internalized by exosomes, the two fluorophore-labeled recognition sequences of DNA tetrahedron could bind to target mRNA, leading to an increase in FRET signal. Thermophoretic effect was applied to further amplify the FRET signal, which achieved a highly sensitive detection of PSA mRNA in exosomes. The DTTA assay showed that exosomal PSA mRNA performed better than serum PSA protein in discriminating prostate cancer from benign prostatic hyperplasia (AUC: 0.93 versus 0.74), providing a new approach for precise detection of prostate cancer [183].

CRISPR/Cas system-assisted detection

Recently, CRISPR/Cas system has offered new opportunities to develop analytical methods for the detection and quantification of nucleic acids in exosomes. The strategy of integrating nucleic acid amplification with CRISPR/Cas has been proposed to improve analytical specificity and sensitivity [184]. For instance, the platform of rolling circular amplification-assisted CRISPR/Cas9 cleavage (RACE) was conducted by Wang et al. to detect multiple exosomal miRNAs [176]. During RCA process, padlock probe recognized single base differences in the presence of HiFi Taq DNA ligase. The amplification product of long ssDNA, which consisted of a large number of repeated target sequences and PAM structures, was specifically recognized by Cas9 nuclease. Consequently, the TaqMan probe hybridized with ssDNA was completely cleaved by Cas9 protein, allowing “turn on” fluorescence change that could be conveniently measured by a spectroscopy. As a result, this RACE platform could be used for highly specific detection of single or multiple exosomal miRNAs from human plasma. However, the large number of naturally PAM structures in the genome may increase the risk of off-target effects, greatly impairing the specificity of analysis [185].

Machine learning

Machine learning refers to a technology that predicts and analyzes unknown data by building models of known data. As a crucial branch of artificial intelligence, machine learning has been widely used for the multiplex profiling of exosomal biomarkers. Algorithms such as linear discriminant analysis (LDA), principal component analysis (PCA), neural network (NN), support vector machine (SVM), and random forests (RF) have been developed to classify multivariable data into a typical classification model, which are then applied for the prediction and grouping of unknown biological data [186]. Kawakami et al. demonstrated that machine learning algorithms performed better than traditional logistic regression analysis to predict and diagnose epithelial ovarian cancer [187]. Wu et al. used the fluorescent signal of urine-derived exosomes as input data, and KNN (K-Nearest Neightbors) and SVM serving as machine learning models, were applied for exosomal biomarker analysis. By introducing machine learning algorithm, the diagnostic model could make an accurate diagnosis and classification of multiple diseases [188]. Liu et al. used LDA to determine a sum signature of seven exosomal biomarkers, which achieved a high accuracy in discriminating prostate cancer from benign disease [68]. Tian et al. analyzed a weighted sum of eight cancer-associated proteins of exosomes from 1 μL plasma via LDA [69]. PCA is an algorithm that transforms a group of probably-related variables into a series of linearly-unrelated variables through orthogonal transformation. Shin et al. used PCA to explore the features of cell exosomes and human plasma exosomes. As a result, the machine learning model could classify normal and lung cancer cell lines-derived exosomes with an accuracy of 95% and obtain an AUC of 0.912 in predicting lung cancer for the whole cohort [162]. Liu et al. employed RF, NN, and SVM to analyze multiple exosome-derived mRNAs of breast cancer patients, which exhibited improved diagnostic performance compared to a single marker [120]. These studies suggest the great potential of combining exosome analysis and machine learning in cancer liquid biopsy. Notably, large data sets are required for typical machine learning approaches, while a limited number of clinical samples and insufficient data may lead to poor accuracy and reliability. The emergence of more powerful machine learning algorithms will favor the analysis of exosomes for liquid biopsy.

The implication of exosomes in cancer liquid biopsy

Recent studies have shown that exosomes are superior to CTCs and ctDNA in liquid biopsy for early diagnosis, disease monitoring, and prognosis prediction [34, 189, 190]. Herein, we summarized the application of exosomes in liquid biopsy for various cancers (Fig. 6, Table 4).
Table 4
Exosomes as biomarkers for cancer liquid biopsy
Cancer types
Exosome sources
Sample volume
Exosomal biomarker
Clinical samples
Diagnostic performance
Clinical significance
Ref.
Gastric Cancer
Serum
 
lnc HOTTIP
126 GC patients; 120 healthy donors
AUC = 0.827
Early diagnosis
[191]
Serum
 
miR-15b-3p
108 GC patients; 108 healthy donors
AUC of 0.820; specificity of 80.6%; sensitivity of 74.1%
Early diagnosis
[192]
HCC
Plasma
100 μL
AFP; GPC3; ALB; APOH; FABP1; FGB; FGG; AHSG; RBP4; TF mRNA
36 HCC patients; 26 Cirrhosis
AUC of 0.87; sensitivity of 93.8%; specificity of 74.5%
Early diagnosis
[61]
Serum
500 μL
miRNA-21; lncRNA-A TB
72 HCC patients
Higher in HCC patients
Prognostic significance
[189]
Serum
 
miR-21
 
Higher in HCC patients
Early diagnosis
[193]
Serum
250 μL
miR-92b
28 non-HCC; 28 HCC patients without recurrence; 43 HCC patients with early recurrenc
Sensitivity of 85.7%; specificity of 86.0%; AUC = 0.925
Early recurrence diagnosis after LDLT
[194]
Serum
100 μL
CEA; GPC-3 and PD-L1
12 HCC patients; 12 hepatitis B; 6 healthy donors
Higher in HCC patients
Early diagnosis and progression monitoring
[112]
PDAC
Plasma
500 μL
miRNA-10b
3 PDAC Patients; 3 CP Patients; 3 healthy donors
Higher in PDAC patients
Early diagnosis and progression monitoring
[173]
Plasma
 
miRNA-10b
PDAC patients; CP patients and healthy donors
Higher in PDAC patients
Early diagnosis
[195]
Mouse plasma samples
 
miR-3970-5p
9 healthy donors; 9 PanIN patients; 9 PDAC patients
Accuracy of 65%
Early diagnosis
[196]
Serum
250 μL
Glypican1
192 patients; 100 healthy donors
Sensitivity of 100%; specificity of 100%; positive predictive value of 100%; negative predictive value of 100%; AUC of 1.0
Early diagnosis
[38]
Plasma
25 μL
Glypican1
20 PDAC patients; 7 benign pancreatic disease; 11 healthy donors
99% sensitivity and 82% specificity
Stage classification
[197]
Serum
5 μL
EpCAM, Glypican1
 
90% accuracy for pancreatic cancer or normal pancreatic epithelial cell lines; 87 and 90% predictive accuracy for HC and EPC individual samples
Early diagnosis
[198]
Serum
2 μL
MIF
4 patients at stage 1 ~ 2; 37 patients at stage 3
Discriminatory sensitivity of 95.7%
Stage classification
[199]
CRC
Serum
 
hsa-circ-0004771
179 patients; 45 healthy donors
AUC of 0.86, 0.88 to differentiate stage I/II CRC patients and CRC patients from HCs
Early diagnosis
[200]
Plasma
25 μL
Epcam-CD63
59 cancer patients; 20 healthy donors
AUC of 0.96
Early diagnosis; prognosis prediction
[125]
NSCLC
Plasma
 
miRNA-21; miRNA-139; miRNA-200; miRNA-378
5 patients; 5 healthy donors
Higher in NSCLC patients
Early diagnosis
[123]
Plasma
1 mL
miRNA-21
NSCLC patients; recurrence of NSCLC patients; healthy individuals
Higher in NSCLC patients
Early diagnosis and drug resistance in advanced cancers
[201]
Plasma
1.5 mL
CD63; EGFR; EpCAM
4 patients; 4 treated patients; 4 healthy donors
Higher in NSCLC patients
Early diagnosis and therapeutic effect evaluation
[159]
Serum
50 μL
PD-L1
5 patients; 4 healthy donors
Higher in NSCLC patients
Early diagnosis
[132]
Serum
4 μL
PD-L1
7 patients at stage 1 ~ 2; 10 patients at stage 3 ~ 4; 12 healthy controls
AUC of 0.97
Early diagnosis
[90]
Breast Cancer
Plasma
 
EpCAM
6 BC patients; 3 healthy donors
Higher in BC patients
Early diagnosis
[71]
Plasma
 
EpCAM; HER2
10 BC patients; 5 healthy donors
AUC of 1; AUC of 1
Early diagnosis
[134]
Serum
3.6 μL
EpCAM
20 BC patients; 10 healthy donors
AUC BC versus HD = 0.99; AUC HER2+ BC versus HER2– BC = 0.94
Cancer classification
[202]
Serum
 
PD-L1
7 patients with metastatic; 8 patients without metastatic; 6 healthy donors
Higher in BC patients
Prognosis prediction and progression monitoring
[114]
Blood
 
CA153
104 BC patients; 100 breast hyperplasia patients and 100 healthy controls
Higher in BC patients
Differential diagnosis
[203]
Serum
 
miR-21; miR-222; miR-200c
Luminal, HER2+, and TN breast cancer patients
Higher in BC patients
Classification of molecular subtypes of breast cancer
[204]
Plasma
1 μL
CA153; EpCAM
36 MBC patients before salvage treatment; 21 NMBC patients before surgical therapy; 66 age-matched healthy donors
AUPRC CA153 = 0.9286
Differential diagnosis of BC and healthy donors
[69]
AUPRC EpCAM = 0.9709
Plasma
1 μL
CA153; CA125; CEA; HER2; EGFR; PSMA; EpCAM; VEG
36 MBC patients before salvage treatment; 21 NMBC patients before surgical therapy; 66 age-matched healthy donors
AUPRC of 0.9826
Differential diagnosis of BC and healthy donors
[69]
Plasma
1 μL
CA153; CA125; CEA; HER2; EGFR; PSMA; EpCAM; VEG
36 MBC patients before salvage treatment; 21 NMBC patients before surgical therapy; 66 age-matched healthy donors
AUPRC of 0.8672
Differential diagnosis of MBC and NMBC
[69]
Plasma samples
 
EpCAM
Various breast cancer patients and healthy individuals
Higher in BC patients
Early diagnosis
[115]
Prostate Cancer
Urine
50-150 mL
miR-196a; miR-143-3p; miR-196-5p; miR-501-3p;
28 PCA patients; 19 healthy donors
AUC miR-196a = 0.92
Early diagnosis
[205]
AUC miR143-3p = 0.72
AUC miR196-5p = 0.73
AUC miR501-3p = 0.69
Plasma
750 μL
miR-217; miR-23b-3p
10 patients; 10 healthy donors
Higher in PCA patients
Early diagnosis
[206]
Serum
400 μL
EphrinA2
50 PCA patients; 21 BPH patients; 20 healthy donors
AUC of 0.7666
Early diagnosis; distinguish PCA from BPH patients
[207]
Serum
25 μL
EpCAM and PSMA
10 PCA patients; 5 healthy donors
Higher in PCA patients
Early diagnosis
[127]
Serum
 
TUBB3 mRNA
52 mCRPC patients
Higher in PCA patients
Prognosis
[208]
Ovarian Cancer
Ascites
 
EpCAM; CD24
20 patients; 10 healthy donors
Higher in OVCA patients
Early diagnosis
[116]
Plasma
2 mL
CA125; EpCAM; CD24
15 patients; 5 healthy donors
AUC CA125 = 1.0
Early diagnosis
[74]
AUC EpCAM = 1.0
AUC CD24 = 0.91
Plasma
20 μL
CD24; EpCAM; FRα
20 OVCA patients; 10 non-cancer controls
AUC CD24 = 1.0
Early diagnosis
[165]
AUC EpCAM = 1.0
AUC FRα = 0.995
Plasma
200 μL
miR-4732-5p
21 healthy controls and 34 epithelial ovarian cancer patients
AUC miR-4732-5p = 0.889
Early diagnosis
[209]
ALB Albumin, APOH Apolipoprotein H, AUPRC Area under the Precision-Recall Curves, FABP1 Fatty acid binding protein 1, FGB Fibrinogen beta chain, FGG Fibrinogen gamma chain, AHSG Alpha 2-HS glycoprotein, RBP4 Retinol binding protein 4, TF Transferrin, LDLT Living donor liver transplantation, CP Chronic pancreatitis, PanIN Pancreatic intraepithelial neoplasia, PanIN Pancreatic intraepithelial neoplasia, MIF Migration inhibitory factor

HCC

Hepatocellular cancer is the fourth cause of cancer-related death. Molecular signatures loaded into HCC-derived exosomes may be used to for diagnosis. Exosomal miR-21 was found to suppress the apoptosis of HCC cells and be upregulated in HCC patients [112]. Exosomal proteins such as CEA and GPC-3, were able to distinguish HCC patients from healthy subjects [193], which may serve as promising biomarkers for noninvasive cancer diagnosis. In a study containing 158 samples, plasma was collected for the detection of 10 HCC-specific genes, including alpha-fetoprotein (AFP), GPC3, albumin, apolipoprotein H, etc. [61]. It was observed that higher fluorescent signals were obtained in HCC cohort via ddPCR in comparison with noncancer cohorts. The 10 gene signatures were further computed by machine learning, which showed great potential to distinguish early-stage HCC from at-risk liver cirrhosis with an AUC of 0.93. Nakano et al. compared the expression levels of exosomal miR-92b and circulating AFP among HCC patients who received liver transplantation [194]. It was revealed that exosomal miR-92b could predict early recurrence of HCC with an AUC of 0.925, while the AUC of AFP was 0.651. Zhu et al. integrated plasma exosomal RNA-sequence, cell-free RNA-sequence and TCGA tissue RNA-sequence datasets to identify 5 noncoding RNAs (circ-0073052, circ-0080695, SNORD3B-1, LINC01226 and HULC) as potential biomarkers of liver cancer. In addition, a panel of SNORD3B-1, circ-0080695 and miR-122 showed the highest AUC (89.4%) to classify liver cancer patients from healthy donors in comparison with other marker panels. By the selected panel, 79.2% AFP-negative samples and 77.1% early-stage liver cancer samples were successfully detected in the testing and validation sets, which indicated the potential of exRNAs panel in the early diagnosis of liver cancer [210].

PDAC

As one of the most recognized indicators associated with pancreatic cancer progression, miRNA-10b is now being widely studied for the early diagnosis of pancreatic ductal adenocarcinoma (PDAC) [211]. Joshi et al. indicated that the levels of exosomal miR-10b were remarkably different among pancreatic cancer patients, at-risk patients with chronic pancreatitis (CP) and healthy individuals, which suggested the potential of miR-10b in diagnosing pancreatic cancer and predicting CP patients who may develop to PDAC [173]. The same conclusion was obtained by Pang’s group by using a dual-SERS biosensor for one-step detection of mRNAs in exosomes [195]. Melo et al. found that GPC-1+ circulating exosomes (GPC-1+ crExos) exhibited high specificity and sensitivity to identify PDAC patients from healthy individuals and chronic pancreatitis (AUC = 1.0), which was superior to CA199 (AUC = 0.739). Moreover, the levels of GPC-1+ crExos were correlated with tumor burden and survival in patients before and after surgery, suggesting the potential of GPC-1 serving as a reliable biomarker for treatment effect and prognostic monitoring [38]. In addition, the combination of GPC-1 and CD63 was demonstrated to show 99% sensitivity and 82% specificity in distinguishing PDAC patients from healthy subjects [197]. Furthermore, migration inhibitory factor (MIF) was reported to have better performance than GPC-1 and EGFR in the discrimination of different stages of pancreatic cancer [199].

CRC

Colorectal cancer (CRC) is one of the most common malignancies with high morbidity and mortality [1]. CircRNA in exosomes is related to the occurrence and development of cancer. Due to the circular structure, it provides a stable biomarker for cancer diagnosis. CircRNA-0004771 was found to be upregulated in serum exosomes of CRC patients [200]. The AUC of exosomal circ-0004771 to identify CRC from healthy controls was 0.88, and that to distinguish patients with stage I and II from other benign intestinal diseases was 0.816, which indicated that circ0004771 could be used as a potential diagnostic marker for colorectal cancer. Wei et al. explored novel exosome biomarkers for the early diagnosis and prognosis of colorectal cancer [125]. The detection of plasma samples revealed that the expression of CD9, CD63 and EpCAM were significantly higher in CRC patients compared with healthy and benign controls (with an AUC of 0.90 and 0.96, respectively). In addition, miR-15b, miR-21, and miR-31 were reported to be highly expressed in serum exosomes of CRC patients. ROC curves showed that AUC of miR-15b was 0.86 and the combined miR-15b, miR-21, and miR-31 panel exhibited 81.21% sensitivity and 81.03% specificity [212].

NSCLC

Non-small cell lung cancer accounts for over 80% of lung cancer-induced deaths [1]. High level of PD-L1 was detected in serum exosomes of NSCLC patients compared to normal individuals [132]. The diagnostic value of exosomal PD-L1 was further confirmed by Pang et al. in a larger cohort of patients with an AUC of 0.97 [90]. Ma et al. found that the expression of exosomal miR-21 was higher in patients with recurrent NSCLC compared with healthy individuals [201]. However, high levels of exosomal miR-21 were also identified in other cancers such as colorectal cancer, breast cancer, and liver cancer [123, 189, 204]. Therefore, the detection of multiple miRNAs may be more valuable for NSCLC diagnosis. Wu et al. demonstrated that miR-21, miR-378, miR-139, and miR-200 were differentially expressed between NSCLC patients and healthy donors, providing more optional biomarkers for the early diagnosis of NSCLC [123]. Notably, the experimental results showed that more amounts of exosomal RNAs were obtained in urine-derived exosomes of NSCLC patients than that from plasma and bronchoalveolar lavage fluid, which may provide a new direction of sample selection to analyze exosomes [62].

Breast cancer

Breast cancer (BC) is the most common cause of cancer-related death among females. Early diagnosis can effectively suppress the mortality and achieve better treatment outcomes. Despite as a widely used biomarker, CA15-3 (carbohydrate antigen 15-3) is not sensitive enough to diagnose primary and metastatic breast cancer (MBC) [203]. Lee et al. demonstrated that cancer cell-derived exosomal miR-21, miR-222, and miR-200c could be quantified in body fluids and used for breast cancer diagnosis [204]. Lu et al. reported that the combination of RDW (red blood cell distribution width), MPV (mean platelet volume), and CA15-3 showed better specificity and sensitivity to identify breast cancer than single biomarker [203]. In addition, an exosome signature was identified by computing weighted sum of eight biomarkers (CA15-3, CA125, CEA, HER2, EGFR, PSMA, EpCAM, and VEGF) through machine learning. Consequently, the signature showed a high discriminative accuracy to differentiate MBC from non-metastatic ones and age-matched healthy donors (91.1%). Moreover, the exosome signature was reported to accurately monitor MBC treatment response and serve as an independent prognostic factor for progression-free survival in MBC patients.

Prostate cancer

PSA is a widely used biomarker for the detection of prostate cancer. However, increased PSA was also identified in inflammatory diseases such as benign prostatic hyperplasia (BPH) [213]. Therefore, the development of sensitive and specific biomarker is urgently needed to diagnose patients at early stage and monitor the progression of cancer. Li et al. reported that exosomal ephrinA2 had superior capability to blood circulating PSA to differentiate prostate cancer patients from BPH patients with an AUC of 0.906 [207]. Moreover, increased EpCAM and PSMA were found in the serum exosomes of prostate cancer [127]. In addition, the expression of exosomal TUBB3 mRNA was reported to be associated with poor progression-free survival of abiraterone in metastatic castration-resistant prostate cancer patients [208].

Exosomes in clinical trial and use for cancer liquid biopsy

Due to the priority of living-cell secretion, large amounts and stable circulation compared to CTC and ctDNA, exosome-based liquid biopsy has been tested in clinical trials and several of them have been approved and reached the market. In 2016, Exosome Diagnostics proposed the first exosome-based liquid biopsy in the world, ExoDx™ Lung (ALK), for the isolation and analysis of exosomal RNA from blood samples. At CLIA-certified laboratory, ExoDx™ Lung (ALK) was proved to be an accurate, real-time tool to detect EML4-ALK mutations in NSCLC patients with 88% sensitivity and 100% specificity, which provides a more direct and sensitive method to detect gene fusions than cfDNA. In addition, the ExoDx Prostate IntelliScore (EPI) has been certified by FDA. Based on the detection of ERG, PCA3, and SPDEF RNA in exosomes, EPI provides a risk score to predict whether a patient with PSA from 2 to 10 ng/mL is likely to develop higher-grade prostate cancer [214]. According to ExoDx, 93% of sensitivity was achieved in prospective studies, and 26% of unnecessary needle biopsies were avoided when the EPI threshold was set at 15.6 [215]. Three independent, prospective, and multicenter clinical trials declared that EPI outperformed standard of care and could be used to assist in the early diagnosis of prostate cancer and eliminate unnecessary prostate biopsy [64]. Moreover, MedOncAlyzer 170 is a newly developed liquid biopsy system capable of detecting both exosomal RNA and ctDNA in a single trial. It can identify significant and functional mutations in multiple cancer types from small volumes (0.5 ml) of patient blood or plasma. Due to the unique formation manner of exosome and ctDNA, MedOncAlyzer 170 is accurate and highly sensitive to detect mutation at all stages of cancer progression and treatment. Although the clinical application value has been verified, larger clinical samples, populations and trials are still needed to confirm the role of exosome-based liquid biopsy in cancer diagnosis and treatment.

Conclusions and perspectives

At present, the limitation of tissue biopsy has been gradually recognized in the field of precision medicine. On the contrary, liquid biopsy has the advantages of minimal invasiveness, easy sample acquisition, and dynamic analysis. Exosomes have been confirmed to stably circulate in body fluids and contain diverse information that reflects the status of tumor progression [28, 33]. The potential of exosome serving as diagnostic and prognostic biomarkers has been investigated in a variety of cancers. However, the high heterogeneity and nano-size of exosomes have posed great technical challenges to the acquisition of their molecular information and interactions. In this review, we have summarized the advantages and drawbacks of conventional and novel techniques to isolate, characterize and detect exosomes. Techniques based on physical or biological characteristics are being widely developed for exosome separation, and the application of microfluidic devices holds great potential for the ultrafast separation of pure exosomes with high yields. Although revolutionary progress has been achieved, there is no standardized method for the high-throughput, high-purity, and minimal damage separation of exosomes from both cell culture medium and human body fluids. Diverse molecules contained in the circulating exosomes have highlighted the potential of exosomes in liquid biopsy. Faster and more convenient methods are required to validate the exosomal cargoes as biomarkers in the diagnosis of cancer. Most new detection platforms, although superior to conventional methods, still face the challenge of low sensitivity and high heterogeneity of different exosome subsets. The technology of single exosome detection and analysis may reveal the unique molecular profile of specific exosomes and provide a plausible strategy to obtain accurate cancer-related information. The further exploration in exosome heterogeneity will address many of the challenges in current exosome studies. Improvements in developing new strategies to isolate exosomes from body fluids and profile exosomal contents in a fast and sensitive way will facilitate the practical application of exosome-based liquid biopsy for cancer precision medicine.

Acknowledgements

We thank the members of the Zhang laboratory for helpful discussion and paper preparation.

Declarations

Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–49.CrossRefPubMed Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–49.CrossRefPubMed
2.
Zurück zum Zitat 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(11):2640–51.PubMed 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(11):2640–51.PubMed
3.
Zurück zum Zitat Sala M, Ros M, Saltel F. A complex and evolutive character: two face aspects of ECM in tumor progression. Front Oncol. 2020;10:1620.PubMedPubMedCentral Sala M, Ros M, Saltel F. A complex and evolutive character: two face aspects of ECM in tumor progression. Front Oncol. 2020;10:1620.PubMedPubMedCentral
4.
Zurück zum Zitat Kalluri R, LeBleu VS. The biology function and biomedical applications of exosomes. New York: Science; 2020. p. 367. Kalluri R, LeBleu VS. The biology function and biomedical applications of exosomes. New York: Science; 2020. p. 367.
5.
Zurück zum Zitat Caby M-P, Lankar D, Vincendeau-Scherrer C, Raposo G, Bonnerot C. Exosomal-like vesicles are present in human blood plasma. Int Immunol. 2005;17:879–87.PubMed Caby M-P, Lankar D, Vincendeau-Scherrer C, Raposo G, Bonnerot C. Exosomal-like vesicles are present in human blood plasma. Int Immunol. 2005;17:879–87.PubMed
6.
Zurück zum Zitat Street JM, Barran PE, Mackay CL, Weidt S, Balmforth C, Walsh TS, et al. Identification and proteomic profiling of exosomes in human cerebrospinal fluid. J Transl Med. 2012;10:5.PubMedPubMedCentral Street JM, Barran PE, Mackay CL, Weidt S, Balmforth C, Walsh TS, et al. Identification and proteomic profiling of exosomes in human cerebrospinal fluid. J Transl Med. 2012;10:5.PubMedPubMedCentral
7.
Zurück zum Zitat Shao H, Im H, Castro CM, Breakefield X, Weissleder R, Lee H. New technologies for analysis of extracellular vesicles. Chem Rev. 2018;118:1917–50.PubMedPubMedCentral Shao H, Im H, Castro CM, Breakefield X, Weissleder R, Lee H. New technologies for analysis of extracellular vesicles. Chem Rev. 2018;118:1917–50.PubMedPubMedCentral
8.
Zurück zum Zitat Lin S, Yu Z, Chen D, Wang Z, Miao J, Li Q, et al. Progress in microfluidics-based exosome separation and detection technologies for diagnostic applications. Small (Weinheim an der Bergstrasse, Germany). 2020;16:e1903916. Lin S, Yu Z, Chen D, Wang Z, Miao J, Li Q, et al. Progress in microfluidics-based exosome separation and detection technologies for diagnostic applications. Small (Weinheim an der Bergstrasse, Germany). 2020;16:e1903916.
9.
Zurück zum Zitat Zhang Y-C, Zhou Q, Wu Y-L. The emerging roles of NGS-based liquid biopsy in non-small cell lung cancer. J Hematol Oncol. 2017;10:167.PubMedPubMedCentral Zhang Y-C, Zhou Q, Wu Y-L. The emerging roles of NGS-based liquid biopsy in non-small cell lung cancer. J Hematol Oncol. 2017;10:167.PubMedPubMedCentral
10.
Zurück zum Zitat Becker A, Thakur BK, Weiss JM, Kim HS, Peinado H, Lyden D. Extracellular vesicles in cancer: cell-to-cell mediators of metastasis. Cancer Cell. 2016;30:836–48.PubMedPubMedCentral Becker A, Thakur BK, Weiss JM, Kim HS, Peinado H, Lyden D. Extracellular vesicles in cancer: cell-to-cell mediators of metastasis. Cancer Cell. 2016;30:836–48.PubMedPubMedCentral
11.
Zurück zum Zitat Hoshino A, Costa-Silva B, Shen T-L, Rodrigues G, Hashimoto A, Tesic Mark M, et al. Tumour exosome integrins determine organotropic metastasis. Nature. 2015;527:329–35.PubMedPubMedCentral Hoshino A, Costa-Silva B, Shen T-L, Rodrigues G, Hashimoto A, Tesic Mark M, et al. Tumour exosome integrins determine organotropic metastasis. Nature. 2015;527:329–35.PubMedPubMedCentral
12.
Zurück zum Zitat Kawamura S, Iinuma H, Wada K, Takahashi K, Minezaki S, Kainuma M, et al. Exosome-encapsulated microRNA-4525, microRNA-451a and microRNA-21 in portal vein blood is a high-sensitive liquid biomarker for the selection of high-risk pancreatic ductal adenocarcinoma patients. J Hepatobiliary Pancreat Sci. 2019;26:63–72.PubMed Kawamura S, Iinuma H, Wada K, Takahashi K, Minezaki S, Kainuma M, et al. Exosome-encapsulated microRNA-4525, microRNA-451a and microRNA-21 in portal vein blood is a high-sensitive liquid biomarker for the selection of high-risk pancreatic ductal adenocarcinoma patients. J Hepatobiliary Pancreat Sci. 2019;26:63–72.PubMed
13.
Zurück zum Zitat Nimir M, Ma Y, Jeffreys SA, Opperman T, Young F, Khan T, 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(7):688.PubMedCentral Nimir M, Ma Y, Jeffreys SA, Opperman T, Young F, Khan T, 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(7):688.PubMedCentral
14.
Zurück zum Zitat Cai X, Janku F, Zhan Q, Fan J-B. Accessing genetic information with liquid biopsies. Trends Genet. 2015;31:564–75.PubMed Cai X, Janku F, Zhan Q, Fan J-B. Accessing genetic information with liquid biopsies. Trends Genet. 2015;31:564–75.PubMed
15.
Zurück zum Zitat Ye Q, Ling S, Zheng S, Xu X. Liquid biopsy in hepatocellular carcinoma: circulating tumor cells and circulating tumor DNA. Mol Cancer. 2019;18:114.PubMedPubMedCentral Ye Q, Ling S, Zheng S, Xu X. Liquid biopsy in hepatocellular carcinoma: circulating tumor cells and circulating tumor DNA. Mol Cancer. 2019;18:114.PubMedPubMedCentral
16.
Zurück zum Zitat Vaidyanathan R, Soon RH, Zhang P, Jiang K, Lim CT. Cancer diagnosis: from tumor to liquid biopsy and beyond. Lab Chip. 2018;19:11–34.PubMed Vaidyanathan R, Soon RH, Zhang P, Jiang K, Lim CT. Cancer diagnosis: from tumor to liquid biopsy and beyond. Lab Chip. 2018;19:11–34.PubMed
17.
Zurück zum Zitat Yu W, Hurley J, Roberts D, Chakrabortty S, Enderle D, Noerholm M, et al. Exosome-based liquid biopsies in cancer: opportunities and challenges. Ann Oncol. 2021;32(4):466–77.PubMed Yu W, Hurley J, Roberts D, Chakrabortty S, Enderle D, Noerholm M, et al. Exosome-based liquid biopsies in cancer: opportunities and challenges. Ann Oncol. 2021;32(4):466–77.PubMed
18.
19.
Zurück zum Zitat He C, Zheng S, Luo Y, Wang B. Exosome theranostics: biology and translational medicine. Theranostics. 2018;8:237–55.PubMedPubMedCentral He C, Zheng S, Luo Y, Wang B. Exosome theranostics: biology and translational medicine. Theranostics. 2018;8:237–55.PubMedPubMedCentral
20.
Zurück zum Zitat Kanwar SS, Dunlay CJ, Simeone DM, Nagrath S. Microfluidic device (ExoChip) for on-chip isolation, quantification and characterization of circulating exosomes. Lab Chip. 2014;14:1891–900.PubMedPubMedCentral Kanwar SS, Dunlay CJ, Simeone DM, Nagrath S. Microfluidic device (ExoChip) for on-chip isolation, quantification and characterization of circulating exosomes. Lab Chip. 2014;14:1891–900.PubMedPubMedCentral
21.
Zurück zum Zitat Dobhal G, Datta A, Ayupova D, Teesdale-Spittle P, Goreham RV. Isolation, characterisation and detection of breath-derived extracellular vesicles. Sci Rep. 2020;10:17381.PubMedPubMedCentral Dobhal G, Datta A, Ayupova D, Teesdale-Spittle P, Goreham RV. Isolation, characterisation and detection of breath-derived extracellular vesicles. Sci Rep. 2020;10:17381.PubMedPubMedCentral
22.
Zurück zum Zitat Patel GK, Khan MA, Zubair H, Srivastava SK, Khushman M, Singh S, et al. Comparative analysis of exosome isolation methods using culture supernatant for optimum yield, purity and downstream applications. Sci Rep. 2019;9:5335.PubMedPubMedCentral Patel GK, Khan MA, Zubair H, Srivastava SK, Khushman M, Singh S, et al. Comparative analysis of exosome isolation methods using culture supernatant for optimum yield, purity and downstream applications. Sci Rep. 2019;9:5335.PubMedPubMedCentral
23.
Zurück zum Zitat An M, Wu J, Zhu J, Lubman DM. Comparison of an optimized ultracentrifugation method versus size-exclusion chromatography for isolation of exosomes from human serum. J Proteome Res. 2018;17:3599–605.PubMedPubMedCentral An M, Wu J, Zhu J, Lubman DM. Comparison of an optimized ultracentrifugation method versus size-exclusion chromatography for isolation of exosomes from human serum. J Proteome Res. 2018;17:3599–605.PubMedPubMedCentral
24.
Zurück zum Zitat Lobb RJ, Becker M, Wen SW, Wong CSF, Wiegmans AP, Leimgruber A, et al. Optimized exosome isolation protocol for cell culture supernatant and human plasma. J Extracellular Vesicles. 2015;4:27031. Lobb RJ, Becker M, Wen SW, Wong CSF, Wiegmans AP, Leimgruber A, et al. Optimized exosome isolation protocol for cell culture supernatant and human plasma. J Extracellular Vesicles. 2015;4:27031.
25.
Zurück zum Zitat Wang W, Luo J, Wang S. Recent progress in isolation and detection of extracellular vesicles for cancer diagnostics. Adv Healthc Mater. 2018;7:e1800484.PubMed Wang W, Luo J, Wang S. Recent progress in isolation and detection of extracellular vesicles for cancer diagnostics. Adv Healthc Mater. 2018;7:e1800484.PubMed
26.
Zurück zum Zitat Hu T, Wolfram J, Srivastava S. Extracellular vesicles in cancer detection: hopes and hypes. Trends Cancer. 2021;7(2):122–33.PubMed Hu T, Wolfram J, Srivastava S. Extracellular vesicles in cancer detection: hopes and hypes. Trends Cancer. 2021;7(2):122–33.PubMed
27.
Zurück zum Zitat Shao H, Chung J, Issadore D. Diagnostic technologies for circulating tumour cells and exosomes. Biosci Rep. 2015;36:e00292.PubMed Shao H, Chung J, Issadore D. Diagnostic technologies for circulating tumour cells and exosomes. Biosci Rep. 2015;36:e00292.PubMed
28.
Zurück zum Zitat Zhang X, Yuan X, Shi H, Wu L, Qian H, Xu W. Exosomes in cancer: small particle, big player. J Hematol Oncol. 2015;8:83.PubMedPubMedCentral Zhang X, Yuan X, Shi H, Wu L, Qian H, Xu W. Exosomes in cancer: small particle, big player. J Hematol Oncol. 2015;8:83.PubMedPubMedCentral
29.
Zurück zum Zitat Hu W, Liu C, Bi Z-Y, Zhou Q, Zhang H, Li L-L, et al. Comprehensive landscape of extracellular vesicle-derived RNAs in cancer initiation, progression, metastasis and cancer immunology. Mol Cancer. 2020;19:102.PubMedPubMedCentral Hu W, Liu C, Bi Z-Y, Zhou Q, Zhang H, Li L-L, et al. Comprehensive landscape of extracellular vesicle-derived RNAs in cancer initiation, progression, metastasis and cancer immunology. Mol Cancer. 2020;19:102.PubMedPubMedCentral
30.
Zurück zum Zitat Wollert T, Hurley JH. Molecular mechanism of multivesicular body biogenesis by ESCRT complexes. Nature. 2010;464:864–9.PubMedPubMedCentral Wollert T, Hurley JH. Molecular mechanism of multivesicular body biogenesis by ESCRT complexes. Nature. 2010;464:864–9.PubMedPubMedCentral
31.
Zurück zum Zitat Vietri M, Radulovic M, Stenmark H. The many functions of ESCRTs. Nat Rev Mol Cell Biol. 2020;21:25–42.PubMed Vietri M, Radulovic M, Stenmark H. The many functions of ESCRTs. Nat Rev Mol Cell Biol. 2020;21:25–42.PubMed
32.
Zurück zum Zitat van Niel G, Charrin S, Simoes S, Romao M, Rochin L, Saftig P, et al. The tetraspanin CD63 regulates ESCRT-independent and -dependent endosomal sorting during melanogenesis. Dev Cell. 2011;21:708–21.PubMedPubMedCentral van Niel G, Charrin S, Simoes S, Romao M, Rochin L, Saftig P, et al. The tetraspanin CD63 regulates ESCRT-independent and -dependent endosomal sorting during melanogenesis. Dev Cell. 2011;21:708–21.PubMedPubMedCentral
33.
Zurück zum Zitat van Niel G, D'Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol. 2018;19:213–28.PubMed van Niel G, D'Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol. 2018;19:213–28.PubMed
34.
Zurück zum Zitat Li W, Li C, Zhou T, Liu X, Liu X, Li X, et al. Role of exosomal proteins in cancer diagnosis. Mol Cancer. 2017;16:145.PubMedPubMedCentral Li W, Li C, Zhou T, Liu X, Liu X, Li X, et al. Role of exosomal proteins in cancer diagnosis. Mol Cancer. 2017;16:145.PubMedPubMedCentral
35.
Zurück zum Zitat Hoshino A, Kim HS, Bojmar L, Gyan KE, Cioffi M, Hernandez J, et al. Extracellular vesicle and particle biomarkers define multiple human cancers. Cell. 2020;182(4):1044–1061.e18.PubMedPubMedCentral Hoshino A, Kim HS, Bojmar L, Gyan KE, Cioffi M, Hernandez J, et al. Extracellular vesicle and particle biomarkers define multiple human cancers. Cell. 2020;182(4):1044–1061.e18.PubMedPubMedCentral
36.
Zurück zum Zitat He M, Crow J, Roth M, Zeng Y, Godwin AK. Integrated immunoisolation and protein analysis of circulating exosomes using microfluidic technology. Lab Chip. 2014;14:3773–80.PubMedPubMedCentral He M, Crow J, Roth M, Zeng Y, Godwin AK. Integrated immunoisolation and protein analysis of circulating exosomes using microfluidic technology. Lab Chip. 2014;14:3773–80.PubMedPubMedCentral
37.
Zurück zum Zitat Jakobsen KR, Paulsen BS, Bæk R, Varming K, Sorensen BS, Jørgensen MM. Exosomal proteins as potential diagnostic markers in advanced non-small cell lung carcinoma. J Extracellular Vesicles. 2015;4:26659. Jakobsen KR, Paulsen BS, Bæk R, Varming K, Sorensen BS, Jørgensen MM. Exosomal proteins as potential diagnostic markers in advanced non-small cell lung carcinoma. J Extracellular Vesicles. 2015;4:26659.
38.
Zurück zum Zitat Melo SA, Luecke LB, Kahlert C, Fernandez AF, Gammon ST, Kaye J, et al. Glypican-1 identifies cancer exosomes and detects early pancreatic cancer. Nature. 2015;523:177–82.PubMedPubMedCentral Melo SA, Luecke LB, Kahlert C, Fernandez AF, Gammon ST, Kaye J, et al. Glypican-1 identifies cancer exosomes and detects early pancreatic cancer. Nature. 2015;523:177–82.PubMedPubMedCentral
39.
Zurück zum Zitat Skog J, Würdinger T, van Rijn S, Meijer DH, Gainche L, Sena-Esteves M, et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol. 2008;10:1470–6.PubMedPubMedCentral Skog J, Würdinger T, van Rijn S, Meijer DH, Gainche L, Sena-Esteves M, et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol. 2008;10:1470–6.PubMedPubMedCentral
40.
Zurück zum Zitat Zhou L, Wang W, Wang F, Yang S, Hu J, Lu B, et al. Plasma-derived exosomal miR-15a-5p as a promising diagnostic biomarker for early detection of endometrial carcinoma. Mol Cancer. 2021;20:57.PubMedPubMedCentral Zhou L, Wang W, Wang F, Yang S, Hu J, Lu B, et al. Plasma-derived exosomal miR-15a-5p as a promising diagnostic biomarker for early detection of endometrial carcinoma. Mol Cancer. 2021;20:57.PubMedPubMedCentral
41.
Zurück zum Zitat Fang T, Lv H, Lv G, Li T, Wang C, Han Q, et al. Tumor-derived exosomal miR-1247-3p induces cancer-associated fibroblast activation to foster lung metastasis of liver cancer. Nat Commun. 2018;9:191.PubMedPubMedCentral Fang T, Lv H, Lv G, Li T, Wang C, Han Q, et al. Tumor-derived exosomal miR-1247-3p induces cancer-associated fibroblast activation to foster lung metastasis of liver cancer. Nat Commun. 2018;9:191.PubMedPubMedCentral
42.
Zurück zum Zitat Barile L, Vassalli G. Exosomes: therapy delivery tools and biomarkers of diseases. Pharmacol Ther. 2017;174:63–78.PubMed Barile L, Vassalli G. Exosomes: therapy delivery tools and biomarkers of diseases. Pharmacol Ther. 2017;174:63–78.PubMed
43.
Zurück zum Zitat Royo F, Théry C, Falcón-Pérez JM, Nieuwland R, Witwer KW. Methods for separation and characterization of extracellular vesicles: results of a worldwide survey performed by the ISEV rigor and standardization subcommittee. Cells. 2020;9(9):1955.PubMedCentral Royo F, Théry C, Falcón-Pérez JM, Nieuwland R, Witwer KW. Methods for separation and characterization of extracellular vesicles: results of a worldwide survey performed by the ISEV rigor and standardization subcommittee. Cells. 2020;9(9):1955.PubMedCentral
44.
Zurück zum Zitat Johnstone RM, Adam M, Hammond JR, Orr L, Turbide C. Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes). J Biol Chem. 1987;262:9412–20.PubMed Johnstone RM, Adam M, Hammond JR, Orr L, Turbide C. Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes). J Biol Chem. 1987;262:9412–20.PubMed
45.
Zurück zum Zitat Vidal M, Mangeat P, Hoekstra D. Aggregation reroutes molecules from a recycling to a vesicle-mediated secretion pathway during reticulocyte maturation. J Cell Sci. 1997;110(Pt 16):1867–77.PubMed Vidal M, Mangeat P, Hoekstra D. Aggregation reroutes molecules from a recycling to a vesicle-mediated secretion pathway during reticulocyte maturation. J Cell Sci. 1997;110(Pt 16):1867–77.PubMed
46.
Zurück zum Zitat Poliakov A, Spilman M, Dokland T, Amling CL, Mobley JA. Structural heterogeneity and protein composition of exosome-like vesicles (prostasomes) in human semen. Prostate. 2009;69:159–67.PubMed Poliakov A, Spilman M, Dokland T, Amling CL, Mobley JA. Structural heterogeneity and protein composition of exosome-like vesicles (prostasomes) in human semen. Prostate. 2009;69:159–67.PubMed
47.
Zurück zum Zitat Taylor DD, Shah S. Methods of isolating extracellular vesicles impact down-stream analyses of their cargoes. Methods (San Diego, Calif). 2015;87:3–10. Taylor DD, Shah S. Methods of isolating extracellular vesicles impact down-stream analyses of their cargoes. Methods (San Diego, Calif). 2015;87:3–10.
48.
Zurück zum Zitat Haraszti RA, Miller R, Stoppato M, Sere YY, Coles A, Didiot M-C, et al. Exosomes produced from 3D cultures of MSCs by tangential flow filtration show higher yield and improved activity. Mol Ther. 2018;26:2838–47.PubMedPubMedCentral Haraszti RA, Miller R, Stoppato M, Sere YY, Coles A, Didiot M-C, et al. Exosomes produced from 3D cultures of MSCs by tangential flow filtration show higher yield and improved activity. Mol Ther. 2018;26:2838–47.PubMedPubMedCentral
49.
Zurück zum Zitat Alvarez ML, Khosroheidari M, Kanchi Ravi R, DiStefano JK. Comparison of protein, microRNA, and mRNA yields using different methods of urinary exosome isolation for the discovery of kidney disease biomarkers. Kidney Int. 2012;82:1024–32.PubMed Alvarez ML, Khosroheidari M, Kanchi Ravi R, DiStefano JK. Comparison of protein, microRNA, and mRNA yields using different methods of urinary exosome isolation for the discovery of kidney disease biomarkers. Kidney Int. 2012;82:1024–32.PubMed
50.
Zurück zum Zitat Böing AN, van der Pol E, Grootemaat AE, Coumans FAW, Sturk A, Nieuwland R. Single-step isolation of extracellular vesicles by size-exclusion chromatography. J Extracellular Vesicles. 2014;3. Böing AN, van der Pol E, Grootemaat AE, Coumans FAW, Sturk A, Nieuwland R. Single-step isolation of extracellular vesicles by size-exclusion chromatography. J Extracellular Vesicles. 2014;3.
51.
Zurück zum Zitat Guo J, Wu C, Lin X, Zhou J, Zhang J, Zheng W, et al. Establishment of a simplified dichotomic size-exclusion chromatography for isolating extracellular vesicles toward clinical applications. J Extracellular Vesicles. 2021;10:e12145. Guo J, Wu C, Lin X, Zhou J, Zhang J, Zheng W, et al. Establishment of a simplified dichotomic size-exclusion chromatography for isolating extracellular vesicles toward clinical applications. J Extracellular Vesicles. 2021;10:e12145.
52.
Zurück zum Zitat Monguió-Tortajada M, Gálvez-Montón C, Bayes-Genis A, Roura S, Borràs FE. Extracellular vesicle isolation methods: rising impact of size-exclusion chromatography. Cell Mol Life Sci. 2019;76:2369–82.PubMed Monguió-Tortajada M, Gálvez-Montón C, Bayes-Genis A, Roura S, Borràs FE. Extracellular vesicle isolation methods: rising impact of size-exclusion chromatography. Cell Mol Life Sci. 2019;76:2369–82.PubMed
53.
Zurück zum Zitat Wei R, Zhao L, Kong G, Liu X, Zhu S, Zhang S, et al. Combination of size-exclusion chromatography and ultracentrifugation improves the proteomic profiling of plasma-derived small extracellular vesicles. Biol Proced Online. 2020;22:12.PubMedPubMedCentral Wei R, Zhao L, Kong G, Liu X, Zhu S, Zhang S, et al. Combination of size-exclusion chromatography and ultracentrifugation improves the proteomic profiling of plasma-derived small extracellular vesicles. Biol Proced Online. 2020;22:12.PubMedPubMedCentral
54.
Zurück zum Zitat Weng Y, Sui Z, Shan Y, Hu Y, Chen Y, Zhang L, et al. Effective isolation of exosomes with polyethylene glycol from cell culture supernatant for in-depth proteome profiling. Analyst. 2016;141:4640–6.PubMed Weng Y, Sui Z, Shan Y, Hu Y, Chen Y, Zhang L, et al. Effective isolation of exosomes with polyethylene glycol from cell culture supernatant for in-depth proteome profiling. Analyst. 2016;141:4640–6.PubMed
55.
Zurück zum Zitat Rider MA, Hurwitz SN, Meckes DG. ExtraPEG: a polyethylene glycol-based method for enrichment of extracellular vesicles. Sci Rep. 2016;6:23978.PubMedPubMedCentral Rider MA, Hurwitz SN, Meckes DG. ExtraPEG: a polyethylene glycol-based method for enrichment of extracellular vesicles. Sci Rep. 2016;6:23978.PubMedPubMedCentral
56.
Zurück zum Zitat Ding M, Wang C, Lu X, Zhang C, Zhou Z, Chen X, et al. Comparison of commercial exosome isolation kits for circulating exosomal microRNA profiling. Anal Bioanal Chem. 2018;410:3805–14.PubMed Ding M, Wang C, Lu X, Zhang C, Zhou Z, Chen X, et al. Comparison of commercial exosome isolation kits for circulating exosomal microRNA profiling. Anal Bioanal Chem. 2018;410:3805–14.PubMed
57.
Zurück zum Zitat Lin B, Tian T, Lu Y, Liu D, Huang M, Zhu L, et al. Tracing tumor-derived exosomal PD-L1 by dual-aptamer activated proximity-induced droplet digital PCR. Angewandte Chemie (International ed in English). 2021;60:7582–6. Lin B, Tian T, Lu Y, Liu D, Huang M, Zhu L, et al. Tracing tumor-derived exosomal PD-L1 by dual-aptamer activated proximity-induced droplet digital PCR. Angewandte Chemie (International ed in English). 2021;60:7582–6.
58.
Zurück zum Zitat Yang Q, Cheng L, Hu L, Lou D, Zhang T, Li J, et al. An integrative microfluidic device for isolation and ultrasensitive detection of lung cancer-specific exosomes from patient urine. Biosens Bioelectron. 2020;163:112290.PubMed Yang Q, Cheng L, Hu L, Lou D, Zhang T, Li J, et al. An integrative microfluidic device for isolation and ultrasensitive detection of lung cancer-specific exosomes from patient urine. Biosens Bioelectron. 2020;163:112290.PubMed
59.
Zurück zum Zitat Kang Y-T, Kim YJ, Bu J, Cho Y-H, Han S-W, Moon B-I. High-purity capture and release of circulating exosomes using an exosome-specific dual-patterned immunofiltration (ExoDIF) device. Nanoscale. 2017;9:13495–505.PubMed Kang Y-T, Kim YJ, Bu J, Cho Y-H, Han S-W, Moon B-I. High-purity capture and release of circulating exosomes using an exosome-specific dual-patterned immunofiltration (ExoDIF) device. Nanoscale. 2017;9:13495–505.PubMed
60.
Zurück zum Zitat Cai S, Luo B, Jiang P, Zhou X, Lan F, Yi Q, et al. Immuno-modified superparamagnetic nanoparticles via host-guest interactions for high-purity capture and mild release of exosomes. Nanoscale. 2018;10:14280–9.PubMed Cai S, Luo B, Jiang P, Zhou X, Lan F, Yi Q, et al. Immuno-modified superparamagnetic nanoparticles via host-guest interactions for high-purity capture and mild release of exosomes. Nanoscale. 2018;10:14280–9.PubMed
61.
Zurück zum Zitat Sun N, Lee Y-T, Zhang RY, Kao R, Teng P-C, Yang Y, et al. Purification of HCC-specific extracellular vesicles on nanosubstrates for early HCC detection by digital scoring. Nat Commun. 2020;11:4489.PubMedPubMedCentral Sun N, Lee Y-T, Zhang RY, Kao R, Teng P-C, Yang Y, et al. Purification of HCC-specific extracellular vesicles on nanosubstrates for early HCC detection by digital scoring. Nat Commun. 2020;11:4489.PubMedPubMedCentral
62.
Zurück zum Zitat Liu F, Vermesh O, Mani V, Ge TJ, Madsen SJ, Sabour A, et al. The exosome total isolation chip. ACS Nano. 2017;11:10712–23.PubMedPubMedCentral Liu F, Vermesh O, Mani V, Ge TJ, Madsen SJ, Sabour A, et al. The exosome total isolation chip. ACS Nano. 2017;11:10712–23.PubMedPubMedCentral
63.
Zurück zum Zitat Sunkara V, Kim C-J, Park J, Woo H-K, Kim D, Ha HK, et al. Fully automated, label-free isolation of extracellular vesicles from whole blood for cancer diagnosis and monitoring. Theranostics. 2019;9:1851–63.PubMedPubMedCentral Sunkara V, Kim C-J, Park J, Woo H-K, Kim D, Ha HK, et al. Fully automated, label-free isolation of extracellular vesicles from whole blood for cancer diagnosis and monitoring. Theranostics. 2019;9:1851–63.PubMedPubMedCentral
64.
Zurück zum Zitat Margolis E, Brown G, Partin A, Carter B, McKiernan J, Tutrone R, et al. Predicting high-grade prostate cancer at initial biopsy: clinical performance of the ExoDx (EPI) prostate Intelliscore test in three independent prospective studies. Prostate Cancer Prostatic Dis. 2021. https://doi.org/10.1038/s41391-021-00456-8. Margolis E, Brown G, Partin A, Carter B, McKiernan J, Tutrone R, et al. Predicting high-grade prostate cancer at initial biopsy: clinical performance of the ExoDx (EPI) prostate Intelliscore test in three independent prospective studies. Prostate Cancer Prostatic Dis. 2021. https://​doi.​org/​10.​1038/​s41391-021-00456-8.
65.
Zurück zum Zitat Xu H, Liao C, Zuo P, Liu Z, Ye B-C. Magnetic-based microfluidic device for on-chip isolation and detection of tumor-derived exosomes. Anal Chem. 2018;90:13451–8.PubMed Xu H, Liao C, Zuo P, Liu Z, Ye B-C. Magnetic-based microfluidic device for on-chip isolation and detection of tumor-derived exosomes. Anal Chem. 2018;90:13451–8.PubMed
66.
Zurück zum Zitat Jiang S, Li Q, Wang C, Pang Y, Sun Z, Xiao R. In Situ Exosomal MicroRNA determination by target-triggered SERS and FeO@TiO-based exosome accumulation. ACS Sensors. 2021;6:852–62.PubMed Jiang S, Li Q, Wang C, Pang Y, Sun Z, Xiao R. In Situ Exosomal MicroRNA determination by target-triggered SERS and FeO@TiO-based exosome accumulation. ACS Sensors. 2021;6:852–62.PubMed
67.
Zurück zum Zitat Tayebi M, Yang D, Collins DJ, Ai Y. Deterministic sorting of submicrometer particles and extracellular vesicles using a combined electric and acoustic field. Nano Lett. 2021;21:6835–42.PubMed Tayebi M, Yang D, Collins DJ, Ai Y. Deterministic sorting of submicrometer particles and extracellular vesicles using a combined electric and acoustic field. Nano Lett. 2021;21:6835–42.PubMed
68.
Zurück zum Zitat Liu C, Zhao J, Tian F, Cai L, Zhang W, Feng Q, et al. Low-cost thermophoretic profiling of extracellular-vesicle surface proteins for the early detection and classification of cancers. Nat Biomed Eng. 2019;3:183–93.PubMed Liu C, Zhao J, Tian F, Cai L, Zhang W, Feng Q, et al. Low-cost thermophoretic profiling of extracellular-vesicle surface proteins for the early detection and classification of cancers. Nat Biomed Eng. 2019;3:183–93.PubMed
69.
Zurück zum Zitat Tian F, Zhang S, Liu C, Han Z, Liu Y, Deng J, et al. Protein analysis of extracellular vesicles to monitor and predict therapeutic response in metastatic breast cancer. Nat Commun. 2021;12:2536.PubMedPubMedCentral Tian F, Zhang S, Liu C, Han Z, Liu Y, Deng J, et al. Protein analysis of extracellular vesicles to monitor and predict therapeutic response in metastatic breast cancer. Nat Commun. 2021;12:2536.PubMedPubMedCentral
70.
Zurück zum Zitat Zhang P, He M, Zeng Y. Ultrasensitive microfluidic analysis of circulating exosomes using a nanostructured graphene oxide/polydopamine coating. Lab Chip. 2016;16:3033–42.PubMedPubMedCentral Zhang P, He M, Zeng Y. Ultrasensitive microfluidic analysis of circulating exosomes using a nanostructured graphene oxide/polydopamine coating. Lab Chip. 2016;16:3033–42.PubMedPubMedCentral
71.
Zurück zum Zitat Fang S, Tian H, Li X, Jin D, Li X, Kong J, 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.PubMedPubMedCentral Fang S, Tian H, Li X, Jin D, Li X, Kong J, 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.PubMedPubMedCentral
72.
Zurück zum Zitat Chen J, Xu Y, Lu Y, Xing W. Isolation and visible detection of tumor-derived exosomes from plasma. Anal Chem. 2018;90:14207–15.PubMed Chen J, Xu Y, Lu Y, Xing W. Isolation and visible detection of tumor-derived exosomes from plasma. Anal Chem. 2018;90:14207–15.PubMed
73.
Zurück zum Zitat Wang Y, Li Q, Shi H, Tang K, Qiao L, Yu G, et al. Microfluidic Raman biochip detection of exosomes: a promising tool for prostate cancer diagnosis. Lab Chip. 2020;20:4632–7.PubMed Wang Y, Li Q, Shi H, Tang K, Qiao L, Yu G, et al. Microfluidic Raman biochip detection of exosomes: a promising tool for prostate cancer diagnosis. Lab Chip. 2020;20:4632–7.PubMed
74.
Zurück zum Zitat 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;16:489–96.PubMedPubMedCentral 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;16:489–96.PubMedPubMedCentral
75.
Zurück zum Zitat Wunsch BH, Smith JT, Gifford SM, Wang C, Brink M, Bruce RL, et al. Nanoscale lateral displacement arrays for the separation of exosomes and colloids down to 20 nm. Nat Nanotechnol. 2016;11:936–40.PubMed Wunsch BH, Smith JT, Gifford SM, Wang C, Brink M, Bruce RL, et al. Nanoscale lateral displacement arrays for the separation of exosomes and colloids down to 20 nm. Nat Nanotechnol. 2016;11:936–40.PubMed
76.
Zurück zum Zitat Liang L-G, Kong M-Q, Zhou S, Sheng Y-F, Wang P, Yu T, et al. An integrated double-filtration microfluidic device for isolation, enrichment and quantification of urinary extracellular vesicles for detection of bladder cancer. Sci Rep. 2017;7:46224.PubMedPubMedCentral Liang L-G, Kong M-Q, Zhou S, Sheng Y-F, Wang P, Yu T, et al. An integrated double-filtration microfluidic device for isolation, enrichment and quantification of urinary extracellular vesicles for detection of bladder cancer. Sci Rep. 2017;7:46224.PubMedPubMedCentral
77.
Zurück zum Zitat Davies RT, Kim J, Jang SC, Choi E-J, Gho YS, Park J. Microfluidic filtration system to isolate extracellular vesicles from blood. Lab Chip. 2012;12:5202–10.PubMed Davies RT, Kim J, Jang SC, Choi E-J, Gho YS, Park J. Microfluidic filtration system to isolate extracellular vesicles from blood. Lab Chip. 2012;12:5202–10.PubMed
78.
Zurück zum Zitat Hattori Y, Shimada T, Yasui T, Kaji N, Baba Y. Micro- and nanopillar chips for continuous separation of extracellular vesicles. Anal Chem. 2019;91:6514–21.PubMed Hattori Y, Shimada T, Yasui T, Kaji N, Baba Y. Micro- and nanopillar chips for continuous separation of extracellular vesicles. Anal Chem. 2019;91:6514–21.PubMed
79.
Zurück zum Zitat Dong L, Zieren RC, Horie K, Kim C-J, Mallick E, Jing Y, et al. Comprehensive evaluation of methods for small extracellular vesicles separation from human plasma, urine and cell culture medium. J Extracellular Vesicles. 2020;10:e12044. Dong L, Zieren RC, Horie K, Kim C-J, Mallick E, Jing Y, et al. Comprehensive evaluation of methods for small extracellular vesicles separation from human plasma, urine and cell culture medium. J Extracellular Vesicles. 2020;10:e12044.
80.
Zurück zum Zitat Wan Y, Cheng G, Liu X, Hao S-J, Nisic M, Zhu C-D, et al. Rapid magnetic isolation of extracellular vesicles via lipid-based nanoprobes. Nat Biomed Eng. 2017;1:0058.PubMedPubMedCentral Wan Y, Cheng G, Liu X, Hao S-J, Nisic M, Zhu C-D, et al. Rapid magnetic isolation of extracellular vesicles via lipid-based nanoprobes. Nat Biomed Eng. 2017;1:0058.PubMedPubMedCentral
81.
Zurück zum Zitat Lee K, Shao H, Weissleder R, Lee H. Acoustic purification of extracellular microvesicles. ACS Nano. 2015;9:2321–7.PubMedPubMedCentral Lee K, Shao H, Weissleder R, Lee H. Acoustic purification of extracellular microvesicles. ACS Nano. 2015;9:2321–7.PubMedPubMedCentral
82.
Zurück zum Zitat Kang Y-T, Hadlock T, Jolly S, Nagrath S. Extracellular vesicles on demand (EVOD) chip for screening and quantification of cancer-associated extracellular vesicles. Biosens Bioelectron. 2020;168:112535.PubMed Kang Y-T, Hadlock T, Jolly S, Nagrath S. Extracellular vesicles on demand (EVOD) chip for screening and quantification of cancer-associated extracellular vesicles. Biosens Bioelectron. 2020;168:112535.PubMed
83.
Zurück zum Zitat Luo Y, Yu X, Chen X, Chen H, Zhang L, Hu X. Method for detection and enrichment of extracellular vesicles. (CN202110614317.4) Luo Y, Yu X, Chen X, Chen H, Zhang L, Hu X. Method for detection and enrichment of extracellular vesicles. (CN202110614317.4)
84.
Zurück zum Zitat Li B, Pan W, Liu C, Guo J, Shen J, Feng J, et al. Homogenous magneto-fluorescent nanosensor for tumor-derived exosome isolation and analysis. ACS Sensors. 2020;5:2052–60.PubMed Li B, Pan W, Liu C, Guo J, Shen J, Feng J, et al. Homogenous magneto-fluorescent nanosensor for tumor-derived exosome isolation and analysis. ACS Sensors. 2020;5:2052–60.PubMed
85.
Zurück zum Zitat Rho J, Chung J, Im H, Liong M, Shao H, Castro CM, et al. Magnetic nanosensor for detection and profiling of erythrocyte-derived microvesicles. ACS Nano. 2013;7:11227–33.PubMedPubMedCentral Rho J, Chung J, Im H, Liong M, Shao H, Castro CM, et al. Magnetic nanosensor for detection and profiling of erythrocyte-derived microvesicles. ACS Nano. 2013;7:11227–33.PubMedPubMedCentral
86.
Zurück zum Zitat Chen Y, Zhu Q, Cheng L, Wang Y, Li M, Yang Q, et al. Exosome detection via the ultrafast-isolation system: EXODUS. Nat Methods. 2021;18:212–8.PubMed Chen Y, Zhu Q, Cheng L, Wang Y, Li M, Yang Q, et al. Exosome detection via the ultrafast-isolation system: EXODUS. Nat Methods. 2021;18:212–8.PubMed
87.
Zurück zum Zitat Wan Y, Maurer M, He H-Z, Xia Y-Q, Hao S-J, Zhang W-L, et al. Enrichment of extracellular vesicles with lipid nanoprobe functionalized nanostructured silica. Lab Chip. 2019;19:2346–55.PubMedPubMedCentral Wan Y, Maurer M, He H-Z, Xia Y-Q, Hao S-J, Zhang W-L, et al. Enrichment of extracellular vesicles with lipid nanoprobe functionalized nanostructured silica. Lab Chip. 2019;19:2346–55.PubMedPubMedCentral
88.
Zurück zum Zitat Kang Y-T, Purcell E, Palacios-Rolston C, Lo T-W, Ramnath N, Jolly S, et al. Isolation and profiling of circulating tumor-associated exosomes using extracellular vesicular lipid-protein binding affinity based microfluidic device. Small (Weinheim an der Bergstrasse, Germany). 2019;15:e1903600. Kang Y-T, Purcell E, Palacios-Rolston C, Lo T-W, Ramnath N, Jolly S, et al. Isolation and profiling of circulating tumor-associated exosomes using extracellular vesicular lipid-protein binding affinity based microfluidic device. Small (Weinheim an der Bergstrasse, Germany). 2019;15:e1903600.
89.
Zurück zum Zitat Nakai W, Yoshida T, Diez D, Miyatake Y, Nishibu T, Imawaka N, et al. A novel affinity-based method for the isolation of highly purified extracellular vesicles. Sci Rep. 2016;6:33935.PubMedPubMedCentral Nakai W, Yoshida T, Diez D, Miyatake Y, Nishibu T, Imawaka N, et al. A novel affinity-based method for the isolation of highly purified extracellular vesicles. Sci Rep. 2016;6:33935.PubMedPubMedCentral
90.
Zurück zum Zitat Pang Y, Shi J, Yang X, Wang C, Sun Z, Xiao R. Personalized detection of circling exosomal PD-L1 based on FeO@TiO isolation and SERS immunoassay. Biosens Bioelectron. 2020;148:111800.PubMed Pang Y, Shi J, Yang X, Wang C, Sun Z, Xiao R. Personalized detection of circling exosomal PD-L1 based on FeO@TiO isolation and SERS immunoassay. Biosens Bioelectron. 2020;148:111800.PubMed
91.
Zurück zum Zitat Ku A, Lim HC, Evander M, Lilja H, Laurell T, Scheding S, et al. Acoustic enrichment of extracellular vesicles from biological fluids. Anal Chem. 2018;90:8011–9.PubMedPubMedCentral Ku A, Lim HC, Evander M, Lilja H, Laurell T, Scheding S, et al. Acoustic enrichment of extracellular vesicles from biological fluids. Anal Chem. 2018;90:8011–9.PubMedPubMedCentral
92.
Zurück zum Zitat Wu M, Ouyang Y, Wang Z, Zhang R, Huang P-H, Chen C, et al. Isolation of exosomes from whole blood by integrating acoustics and microfluidics. Proc Natl Acad Sci U S A. 2017;114:10584–9.PubMedPubMedCentral Wu M, Ouyang Y, Wang Z, Zhang R, Huang P-H, Chen C, et al. Isolation of exosomes from whole blood by integrating acoustics and microfluidics. Proc Natl Acad Sci U S A. 2017;114:10584–9.PubMedPubMedCentral
93.
Zurück zum Zitat Gu Y, Chen C, Mao Z, Bachman H, Becker R, Rufo J, et al. Acoustofluidic centrifuge for nanoparticle enrichment and separation. Sci Adv. 2021;7(1):eabc0467.PubMedPubMedCentral Gu Y, Chen C, Mao Z, Bachman H, Becker R, Rufo J, et al. Acoustofluidic centrifuge for nanoparticle enrichment and separation. Sci Adv. 2021;7(1):eabc0467.PubMedPubMedCentral
94.
Zurück zum Zitat Gardiner C, Di Vizio D, Sahoo S, Théry C, Witwer KW, Wauben M, et al. Techniques used for the isolation and characterization of extracellular vesicles: results of a worldwide survey. J Extracellular Vesicles. 2016;5:32945. Gardiner C, Di Vizio D, Sahoo S, Théry C, Witwer KW, Wauben M, et al. Techniques used for the isolation and characterization of extracellular vesicles: results of a worldwide survey. J Extracellular Vesicles. 2016;5:32945.
95.
Zurück zum Zitat Théry C, Witwer KW, Aikawa E, Alcaraz MJ, Anderson JD, Andriantsitohaina R, 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 Extracellular Vesicles. 2018;7:1535750. Théry C, Witwer KW, Aikawa E, Alcaraz MJ, Anderson JD, Andriantsitohaina R, 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 Extracellular Vesicles. 2018;7:1535750.
96.
Zurück zum Zitat Sharma S, Rasool HI, Palanisamy V, Mathisen C, Schmidt M, Wong DT, et al. Structural-mechanical characterization of nanoparticle exosomes in human saliva, using correlative AFM, FESEM, and force spectroscopy. ACS Nano. 2010;4:1921–6.PubMedPubMedCentral Sharma S, Rasool HI, Palanisamy V, Mathisen C, Schmidt M, Wong DT, et al. Structural-mechanical characterization of nanoparticle exosomes in human saliva, using correlative AFM, FESEM, and force spectroscopy. ACS Nano. 2010;4:1921–6.PubMedPubMedCentral
97.
Zurück zum Zitat Tian Y, Ma L, Gong M, Su G, Zhu S, Zhang W, et al. Protein profiling and sizing of extracellular vesicles from colorectal cancer patients via flow cytometry. ACS Nano. 2018;12:671–80.PubMed Tian Y, Ma L, Gong M, Su G, Zhu S, Zhang W, et al. Protein profiling and sizing of extracellular vesicles from colorectal cancer patients via flow cytometry. ACS Nano. 2018;12:671–80.PubMed
98.
Zurück zum Zitat Sharma S, Gillespie BM, Palanisamy V, Gimzewski JK. Quantitative nanostructural and single-molecule force spectroscopy biomolecular analysis of human-saliva-derived exosomes. Langmuir. 2011;27:14394–400.PubMedPubMedCentral Sharma S, Gillespie BM, Palanisamy V, Gimzewski JK. Quantitative nanostructural and single-molecule force spectroscopy biomolecular analysis of human-saliva-derived exosomes. Langmuir. 2011;27:14394–400.PubMedPubMedCentral
99.
Zurück zum Zitat Li B, Liu C, Pan W, Shen J, Guo J, Luo T, et al. Facile fluorescent aptasensor using aggregation-induced emission luminogens for exosomal proteins profiling towards liquid biopsy. Biosens Bioelectron. 2020;168:112520.PubMed Li B, Liu C, Pan W, Shen J, Guo J, Luo T, et al. Facile fluorescent aptasensor using aggregation-induced emission luminogens for exosomal proteins profiling towards liquid biopsy. Biosens Bioelectron. 2020;168:112520.PubMed
100.
Zurück zum Zitat Street JM, Koritzinsky EH, Glispie DM, Star RA, Yuen PST. Urine exosomes: an emerging trove of biomarkers. Adv Clin Chem. 2017;78:103–22.PubMed Street JM, Koritzinsky EH, Glispie DM, Star RA, Yuen PST. Urine exosomes: an emerging trove of biomarkers. Adv Clin Chem. 2017;78:103–22.PubMed
101.
Zurück zum Zitat Carnino JM, Lee H, Jin Y. Isolation and characterization of extracellular vesicles from Broncho-alveolar lavage fluid: a review and comparison of different methods. Respir Res. 2019;20:240.PubMedPubMedCentral Carnino JM, Lee H, Jin Y. Isolation and characterization of extracellular vesicles from Broncho-alveolar lavage fluid: a review and comparison of different methods. Respir Res. 2019;20:240.PubMedPubMedCentral
102.
Zurück zum Zitat Kurian TK, Banik S, Gopal D, Chakrabarti S, Mazumder N. Elucidating methods for isolation and quantification of exosomes: a review. Mol Biotechnol. 2021;63:249–66.PubMedPubMedCentral Kurian TK, Banik S, Gopal D, Chakrabarti S, Mazumder N. Elucidating methods for isolation and quantification of exosomes: a review. Mol Biotechnol. 2021;63:249–66.PubMedPubMedCentral
103.
Zurück zum Zitat Božič D, Sitar S, Junkar I, Štukelj R, Pajnič M, Žagar E, et al. Viscosity of plasma as a key factor in assessment of extracellular vesicles by light scattering. Cells. 2019;8(9):1046.PubMedCentral Božič D, Sitar S, Junkar I, Štukelj R, Pajnič M, Žagar E, et al. Viscosity of plasma as a key factor in assessment of extracellular vesicles by light scattering. Cells. 2019;8(9):1046.PubMedCentral
104.
Zurück zum Zitat Bachurski D, Schuldner M, Nguyen P-H, Malz A, Reiners KS, Grenzi PC, et al. Extracellular vesicle measurements with nanoparticle tracking analysis - An accuracy and repeatability comparison between NanoSight NS300 and ZetaView. J Extracellular Vesicles. 2019;8:1596016. Bachurski D, Schuldner M, Nguyen P-H, Malz A, Reiners KS, Grenzi PC, et al. Extracellular vesicle measurements with nanoparticle tracking analysis - An accuracy and repeatability comparison between NanoSight NS300 and ZetaView. J Extracellular Vesicles. 2019;8:1596016.
105.
Zurück zum Zitat Coumans FAW, van der Pol E, Böing AN, Hajji N, Sturk G, van Leeuwen TG, et al. Reproducible extracellular vesicle size and concentration determination with tunable resistive pulse sensing. J Extracellular Vesicles. 2014;3:25922. Coumans FAW, van der Pol E, Böing AN, Hajji N, Sturk G, van Leeuwen TG, et al. Reproducible extracellular vesicle size and concentration determination with tunable resistive pulse sensing. J Extracellular Vesicles. 2014;3:25922.
106.
Zurück zum Zitat Jeppesen DK, Fenix AM, Franklin JL, Higginbotham JN, Zhang Q, Zimmerman LJ, et al. Reassessment of exosome composition. Cell. 2019;177(2):428–445.e18.PubMedPubMedCentral Jeppesen DK, Fenix AM, Franklin JL, Higginbotham JN, Zhang Q, Zimmerman LJ, et al. Reassessment of exosome composition. Cell. 2019;177(2):428–445.e18.PubMedPubMedCentral
107.
Zurück zum Zitat Ueda K, Ishikawa N, Tatsuguchi A, Saichi N, Fujii R, Nakagawa H. Antibody-coupled monolithic silica microtips for highthroughput molecular profiling of circulating exosomes. Sci Rep. 2014;4:6232.PubMedPubMedCentral Ueda K, Ishikawa N, Tatsuguchi A, Saichi N, Fujii R, Nakagawa H. Antibody-coupled monolithic silica microtips for highthroughput molecular profiling of circulating exosomes. Sci Rep. 2014;4:6232.PubMedPubMedCentral
108.
Zurück zum Zitat Yokoyama S, Takeuchi A, Yamaguchi S, Mitani Y, Watanabe T, Matsuda K, et al. Clinical implications of carcinoembryonic antigen distribution in serum exosomal fraction-measurement by ELISA. PLoS One. 2017;12:e0183337.PubMedPubMedCentral Yokoyama S, Takeuchi A, Yamaguchi S, Mitani Y, Watanabe T, Matsuda K, et al. Clinical implications of carcinoembryonic antigen distribution in serum exosomal fraction-measurement by ELISA. PLoS One. 2017;12:e0183337.PubMedPubMedCentral
109.
Zurück zum Zitat Xia Y, Liu M, Wang L, Yan A, He W, Chen M, et al. A visible and colorimetric aptasensor based on DNA-capped single-walled carbon nanotubes for detection of exosomes. Biosens Bioelectron. 2017;92:8–15.PubMed Xia Y, Liu M, Wang L, Yan A, He W, Chen M, et al. A visible and colorimetric aptasensor based on DNA-capped single-walled carbon nanotubes for detection of exosomes. Biosens Bioelectron. 2017;92:8–15.PubMed
110.
Zurück zum Zitat Chen Z, Cheng S-B, Cao P, Qiu Q-F, Chen Y, Xie M, et al. Detection of exosomes by ZnO nanowires coated three-dimensional scaffold chip device. Biosens Bioelectron. 2018;122:211–6.PubMed Chen Z, Cheng S-B, Cao P, Qiu Q-F, Chen Y, Xie M, et al. Detection of exosomes by ZnO nanowires coated three-dimensional scaffold chip device. Biosens Bioelectron. 2018;122:211–6.PubMed
111.
Zurück zum Zitat Liang L, Kong M, Sheng Y, Wang S. An integrated detection method and detection chip for the separation, enrichment and detection of urine exosomes. (CN201610386872.5) Liang L, Kong M, Sheng Y, Wang S. An integrated detection method and detection chip for the separation, enrichment and detection of urine exosomes. (CN201610386872.5)
112.
Zurück zum Zitat Di H, Mi Z, Sun Y, Liu X, Liu X, Li A, et al. Nanozyme-assisted sensitive profiling of exosomal proteins for rapid cancer diagnosis. Theranostics. 2020;10:9303–14.PubMedPubMedCentral Di H, Mi Z, Sun Y, Liu X, Liu X, Li A, et al. Nanozyme-assisted sensitive profiling of exosomal proteins for rapid cancer diagnosis. Theranostics. 2020;10:9303–14.PubMedPubMedCentral
113.
Zurück zum Zitat Jin D, Yang F, Zhang Y, Liu L, Zhou Y, Wang F, et al. ExoAPP: exosome-oriented, aptamer nanoprobe-enabled surface proteins profiling and detection. Anal Chem. 2018;90:14402–11.PubMed Jin D, Yang F, Zhang Y, Liu L, Zhou Y, Wang F, et al. ExoAPP: exosome-oriented, aptamer nanoprobe-enabled surface proteins profiling and detection. Anal Chem. 2018;90:14402–11.PubMed
114.
Zurück zum Zitat Cao Y, Wang Y, Yu X, Jiang X, Li G, Zhao J. Identification of programmed death ligand-1 positive exosomes in breast cancer based on DNA amplification-responsive metal-organic frameworks. Biosens Bioelectron. 2020;166:112452.PubMed Cao Y, Wang Y, Yu X, Jiang X, Li G, Zhao J. Identification of programmed death ligand-1 positive exosomes in breast cancer based on DNA amplification-responsive metal-organic frameworks. Biosens Bioelectron. 2020;166:112452.PubMed
115.
Zurück zum Zitat Kashefi-Kheyrabadi L, Kim J, Chakravarty S, Park S, Gwak H, Kim S-I, et al. Detachable microfluidic device implemented with electrochemical aptasensor (DeMEA) for sequential analysis of cancerous exosomes. Biosens Bioelectron. 2020;169:112622.PubMed Kashefi-Kheyrabadi L, Kim J, Chakravarty S, Park S, Gwak H, Kim S-I, et al. Detachable microfluidic device implemented with electrochemical aptasensor (DeMEA) for sequential analysis of cancerous exosomes. Biosens Bioelectron. 2020;169:112622.PubMed
116.
Zurück zum Zitat Im H, Shao H, Park YI, Peterson VM, Castro CM, Weissleder R, et al. Label-free detection and molecular profiling of exosomes with a nano-plasmonic sensor. Nat Biotechnol. 2014;32:490–5.PubMedPubMedCentral Im H, Shao H, Park YI, Peterson VM, Castro CM, Weissleder R, et al. Label-free detection and molecular profiling of exosomes with a nano-plasmonic sensor. Nat Biotechnol. 2014;32:490–5.PubMedPubMedCentral
117.
Zurück zum Zitat Dong S, Wang Y, Liu Z, Zhang W, Yi K, Zhang X, et al. Beehive-inspired macroporous SERS probe for cancer detection through capturing and analyzing exosomes in plasma. ACS Appl Mater Interfaces. 2020;12:5136–46.PubMed Dong S, Wang Y, Liu Z, Zhang W, Yi K, Zhang X, et al. Beehive-inspired macroporous SERS probe for cancer detection through capturing and analyzing exosomes in plasma. ACS Appl Mater Interfaces. 2020;12:5136–46.PubMed
118.
Zurück zum Zitat Wang H, Jiang D, Li W, Xiang X, Zhao J, Yu B, et al. Evaluation of serum extracellular vesicles as noninvasive diagnostic markers of glioma. Theranostics. 2019;9:5347–58.PubMedPubMedCentral Wang H, Jiang D, Li W, Xiang X, Zhao J, Yu B, et al. Evaluation of serum extracellular vesicles as noninvasive diagnostic markers of glioma. Theranostics. 2019;9:5347–58.PubMedPubMedCentral
119.
Zurück zum Zitat He D, Wang H, Ho S-L, Chan H-N, Hai L, He X, et al. Total internal reflection-based single-vesicle in situ quantitative and stoichiometric analysis of tumor-derived exosomal microRNAs for diagnosis and treatment monitoring. Theranostics. 2019;9:4494–507.PubMedPubMedCentral He D, Wang H, Ho S-L, Chan H-N, Hai L, He X, et al. Total internal reflection-based single-vesicle in situ quantitative and stoichiometric analysis of tumor-derived exosomal microRNAs for diagnosis and treatment monitoring. Theranostics. 2019;9:4494–507.PubMedPubMedCentral
120.
Zurück zum Zitat Liu C, Li B, Lin H, Yang C, Guo J, Cui B, et al. Multiplexed analysis of small extracellular vesicle-derived mRNAs by droplet digital PCR and machine learning improves breast cancer diagnosis. Biosens Bioelectron. 2021;194:113615.PubMed Liu C, Li B, Lin H, Yang C, Guo J, Cui B, et al. Multiplexed analysis of small extracellular vesicle-derived mRNAs by droplet digital PCR and machine learning improves breast cancer diagnosis. Biosens Bioelectron. 2021;194:113615.PubMed
121.
Zurück zum Zitat Lee JH, Kim JA, Jeong S, Rhee WJ. Simultaneous and multiplexed detection of exosome microRNAs using molecular beacons. Biosens Bioelectron. 2016;86:202–10.PubMed Lee JH, Kim JA, Jeong S, Rhee WJ. Simultaneous and multiplexed detection of exosome microRNAs using molecular beacons. Biosens Bioelectron. 2016;86:202–10.PubMed
122.
Zurück zum Zitat Lee JH, Kim JA, Kwon MH, Kang JY, Rhee WJ. In situ single step detection of exosome microRNA using molecular beacon. Biomaterials. 2015;54:116–25.PubMed Lee JH, Kim JA, Kwon MH, Kang JY, Rhee WJ. In situ single step detection of exosome microRNA using molecular beacon. Biomaterials. 2015;54:116–25.PubMed
123.
Zurück zum Zitat Wu W, Yu X, Wu J, Wu T, Fan Y, Chen W, et al. Surface plasmon resonance imaging-based biosensor for multiplex and ultrasensitive detection of NSCLC-associated exosomal miRNAs using DNA programmed heterostructure of Au-on-Ag. Biosens Bioelectron. 2021;175:112835.PubMed Wu W, Yu X, Wu J, Wu T, Fan Y, Chen W, et al. Surface plasmon resonance imaging-based biosensor for multiplex and ultrasensitive detection of NSCLC-associated exosomal miRNAs using DNA programmed heterostructure of Au-on-Ag. Biosens Bioelectron. 2021;175:112835.PubMed
124.
Zurück zum Zitat Liu C, Xu X, Li B, Situ B, Pan W, Hu Y, et al. Single-exosome-counting immunoassays for cancer diagnostics. Nano Lett. 2018;18:4226–32.PubMed Liu C, Xu X, Li B, Situ B, Pan W, Hu Y, et al. Single-exosome-counting immunoassays for cancer diagnostics. Nano Lett. 2018;18:4226–32.PubMed
125.
Zurück zum Zitat Wei P, Wu F, Kang B, Sun X, Heskia F, Pachot A, et al. Plasma extracellular vesicles detected by single molecule array technology as a liquid biopsy for colorectal cancer. J Extracellular Vesicles. 2020;9:1809765. Wei P, Wu F, Kang B, Sun X, Heskia F, Pachot A, et al. Plasma extracellular vesicles detected by single molecule array technology as a liquid biopsy for colorectal cancer. J Extracellular Vesicles. 2020;9:1809765.
126.
Zurück zum Zitat Yu X, He L, Pentok M, Yang H, Yang Y, Li Z, et al. An aptamer-based new method for competitive fluorescence detection of exosomes. Nanoscale. 2019;11:15589–95.PubMed Yu X, He L, Pentok M, Yang H, Yang Y, Li Z, et al. An aptamer-based new method for competitive fluorescence detection of exosomes. Nanoscale. 2019;11:15589–95.PubMed
127.
Zurück zum Zitat Zhou Y-G, Mohamadi RM, Poudineh M, Kermanshah L, Ahmed S, Safaei TS, et al. Interrogating circulating microsomes and exosomes using metal nanoparticles. Small (Weinheim an der Bergstrasse, Germany). 2016;12:727–32. Zhou Y-G, Mohamadi RM, Poudineh M, Kermanshah L, Ahmed S, Safaei TS, et al. Interrogating circulating microsomes and exosomes using metal nanoparticles. Small (Weinheim an der Bergstrasse, Germany). 2016;12:727–32.
128.
Zurück zum Zitat Jeong S, Park J, Pathania D, Castro CM, Weissleder R, Lee H. Integrated magneto-electrochemical sensor for exosome analysis. ACS Nano. 2016;10:1802–9.PubMedPubMedCentral Jeong S, Park J, Pathania D, Castro CM, Weissleder R, Lee H. Integrated magneto-electrochemical sensor for exosome analysis. ACS Nano. 2016;10:1802–9.PubMedPubMedCentral
129.
Zurück zum Zitat Park J, Park JS, Huang C-H, Jo A, Cook K, Wang R, et al. An integrated magneto-electrochemical device for the rapid profiling of tumour extracellular vesicles from blood plasma. Nat Biomed Eng. 2021;5:678–89.PubMedPubMedCentral Park J, Park JS, Huang C-H, Jo A, Cook K, Wang R, et al. An integrated magneto-electrochemical device for the rapid profiling of tumour extracellular vesicles from blood plasma. Nat Biomed Eng. 2021;5:678–89.PubMedPubMedCentral
130.
Zurück zum Zitat Homola J. Surface plasmon resonance sensors for detection of chemical and biological species. Chem Rev. 2008;108:462–93.PubMed Homola J. Surface plasmon resonance sensors for detection of chemical and biological species. Chem Rev. 2008;108:462–93.PubMed
131.
Zurück zum Zitat Zeng S, Baillargeat D, Ho H-P, Yong K-T. Nanomaterials enhanced surface plasmon resonance for biological and chemical sensing applications. Chem Soc Rev. 2014;43:3426–52.PubMed Zeng S, Baillargeat D, Ho H-P, Yong K-T. Nanomaterials enhanced surface plasmon resonance for biological and chemical sensing applications. Chem Soc Rev. 2014;43:3426–52.PubMed
132.
Zurück zum Zitat Liu C, Zeng X, An Z, Yang Y, Eisenbaum M, Gu X, et al. Sensitive detection of exosomal proteins via a compact surface plasmon resonance biosensor for cancer diagnosis. ACS Sensors. 2018;3:1471–9.PubMedPubMedCentral Liu C, Zeng X, An Z, Yang Y, Eisenbaum M, Gu X, et al. Sensitive detection of exosomal proteins via a compact surface plasmon resonance biosensor for cancer diagnosis. ACS Sensors. 2018;3:1471–9.PubMedPubMedCentral
133.
Zurück zum Zitat Kneipp, Wang, Kneipp, Itzkan, Dasari, Feld. Population pumping of excited vibrational states by spontaneous surface-enhanced Raman scattering. Phys Rev Lett. 1996;76:2444–7.PubMed Kneipp, Wang, Kneipp, Itzkan, Dasari, Feld. Population pumping of excited vibrational states by spontaneous surface-enhanced Raman scattering. Phys Rev Lett. 1996;76:2444–7.PubMed
134.
Zurück zum Zitat Kwizera EA, O'Connor R, Vinduska V, Williams M, Butch ER, Snyder SE, et al. Molecular detection and analysis of exosomes using surface-enhanced Raman scattering gold nanorods and a miniaturized device. Theranostics. 2018;8:2722–38.PubMedPubMedCentral Kwizera EA, O'Connor R, Vinduska V, Williams M, Butch ER, Snyder SE, et al. Molecular detection and analysis of exosomes using surface-enhanced Raman scattering gold nanorods and a miniaturized device. Theranostics. 2018;8:2722–38.PubMedPubMedCentral
135.
Zurück zum Zitat Ran FA, Hsu PD, Wright J, Agarwala V, Scott DA, Zhang F. Genome engineering using the CRISPR-Cas9 system. Nat Protoc. 2013;8:2281–308.PubMedPubMedCentral Ran FA, Hsu PD, Wright J, Agarwala V, Scott DA, Zhang F. Genome engineering using the CRISPR-Cas9 system. Nat Protoc. 2013;8:2281–308.PubMedPubMedCentral
136.
Zurück zum Zitat Zhao X, Zhang W, Qiu X, Mei Q, Luo Y, Fu W. Rapid and sensitive exosome detection with CRISPR/Cas12a. Anal Bioanal Chem. 2020;412:601–9.PubMed Zhao X, Zhang W, Qiu X, Mei Q, Luo Y, Fu W. Rapid and sensitive exosome detection with CRISPR/Cas12a. Anal Bioanal Chem. 2020;412:601–9.PubMed
137.
Zurück zum Zitat Xing S, Lu Z, Huang Q, Li H, Wang Y, Lai Y, et al. An ultrasensitive hybridization chain reaction-amplified CRISPR-Cas12a aptasensor for extracellular vesicle surface protein quantification. Theranostics. 2020;10:10262–73.PubMedPubMedCentral Xing S, Lu Z, Huang Q, Li H, Wang Y, Lai Y, et al. An ultrasensitive hybridization chain reaction-amplified CRISPR-Cas12a aptasensor for extracellular vesicle surface protein quantification. Theranostics. 2020;10:10262–73.PubMedPubMedCentral
138.
Zurück zum Zitat Li H, Xing S, Xu J, He Y, Lai Y, Wang Y, et al. Aptamer-based CRISPR/Cas12a assay for the ultrasensitive detection of extracellular vesicle proteins. Talanta. 2021;221:121670.PubMed Li H, Xing S, Xu J, He Y, Lai Y, Wang Y, et al. Aptamer-based CRISPR/Cas12a assay for the ultrasensitive detection of extracellular vesicle proteins. Talanta. 2021;221:121670.PubMed
139.
Zurück zum Zitat Guo K, Li Z, Win A, Coreas R, Adkins GB, Cui X, et al. Calibration-free analysis of surface proteins on single extracellular vesicles enabled by DNA nanostructure. Biosens Bioelectron. 2021;192:113502.PubMed Guo K, Li Z, Win A, Coreas R, Adkins GB, Cui X, et al. Calibration-free analysis of surface proteins on single extracellular vesicles enabled by DNA nanostructure. Biosens Bioelectron. 2021;192:113502.PubMed
140.
Zurück zum Zitat Liu C, Zhao J, Tian F, Chang J, Zhang W, Sun J. λ-DNA- and aptamer-mediated sorting and analysis of extracellular vesicles. J Am Chem Soc. 2019;141:3817–21.PubMed Liu C, Zhao J, Tian F, Chang J, Zhang W, Sun J. λ-DNA- and aptamer-mediated sorting and analysis of extracellular vesicles. J Am Chem Soc. 2019;141:3817–21.PubMed
141.
Zurück zum Zitat Li W, Shao B, Liu C, Wang H, Zheng W, Kong W, et al. Liquid biospy: noninvasive diagnosis and molecular phenotyping of breast cancer through microbead-assisted flow cytometry detection of tumor-derived extracellular vesicles (small methods 11/2018). Small Methods. 2018;2:1800050. Li W, Shao B, Liu C, Wang H, Zheng W, Kong W, et al. Liquid biospy: noninvasive diagnosis and molecular phenotyping of breast cancer through microbead-assisted flow cytometry detection of tumor-derived extracellular vesicles (small methods 11/2018). Small Methods. 2018;2:1800050.
142.
Zurück zum Zitat Liu H, Yuan W, Pang Q, Xue C, Yan X. Single-particle analysis of tear fluid reveals abundant presence of tissue factor-exposing extracellular vesicles with strong coagulation activity. Talanta. 2021;239:123089.PubMed Liu H, Yuan W, Pang Q, Xue C, Yan X. Single-particle analysis of tear fluid reveals abundant presence of tissue factor-exposing extracellular vesicles with strong coagulation activity. Talanta. 2021;239:123089.PubMed
143.
Zurück zum Zitat Tan S, Xia L, Yi P, Han Y, Tang L, Pan Q, et al. Exosomal miRNAs in tumor microenvironment. J Exp Clin Cancer Res. 2020;39:67.PubMedPubMedCentral Tan S, Xia L, Yi P, Han Y, Tang L, Pan Q, et al. Exosomal miRNAs in tumor microenvironment. J Exp Clin Cancer Res. 2020;39:67.PubMedPubMedCentral
144.
Zurück zum Zitat Wang Y, Liu J, Ma J, Sun T, Zhou Q, Wang W, et al. Exosomal circRNAs: biogenesis, effect and application in human diseases. Mol Cancer. 2019;18:116.PubMedPubMedCentral Wang Y, Liu J, Ma J, Sun T, Zhou Q, Wang W, et al. Exosomal circRNAs: biogenesis, effect and application in human diseases. Mol Cancer. 2019;18:116.PubMedPubMedCentral
145.
Zurück zum Zitat Gandham S, Su X, Wood J, Nocera AL, Alli SC, Milane L, et al. Technologies and standardization in research on extracellular vesicles. Trends Biotechnol. 2020;38:1066–98.PubMedPubMedCentral Gandham S, Su X, Wood J, Nocera AL, Alli SC, Milane L, et al. Technologies and standardization in research on extracellular vesicles. Trends Biotechnol. 2020;38:1066–98.PubMedPubMedCentral
146.
Zurück zum Zitat Mateescu B, Kowal EJK, van Balkom BWM, Bartel S, Bhattacharyya SN, Buzás EI, et al. Obstacles and opportunities in the functional analysis of extracellular vesicle RNA - an ISEV position paper. J Extracellular Vesicles. 2017;6:1286095. Mateescu B, Kowal EJK, van Balkom BWM, Bartel S, Bhattacharyya SN, Buzás EI, et al. Obstacles and opportunities in the functional analysis of extracellular vesicle RNA - an ISEV position paper. J Extracellular Vesicles. 2017;6:1286095.
147.
Zurück zum Zitat Jaluria P, Konstantopoulos K, Betenbaugh M, Shiloach J. A perspective on microarrays: current applications, pitfalls, and potential uses. Microb Cell Factories. 2007;6:4. Jaluria P, Konstantopoulos K, Betenbaugh M, Shiloach J. A perspective on microarrays: current applications, pitfalls, and potential uses. Microb Cell Factories. 2007;6:4.
148.
Zurück zum Zitat Boriachek K, Masud MK, Palma C, Phan H-P, Yamauchi Y, Hossain MSA, et al. Avoiding pre-isolation step in exosome analysis: direct isolation and sensitive detection of exosomes using gold-loaded nanoporous ferric oxide nanozymes. Anal Chem. 2019;91:3827–34.PubMed Boriachek K, Masud MK, Palma C, Phan H-P, Yamauchi Y, Hossain MSA, et al. Avoiding pre-isolation step in exosome analysis: direct isolation and sensitive detection of exosomes using gold-loaded nanoporous ferric oxide nanozymes. Anal Chem. 2019;91:3827–34.PubMed
149.
Zurück zum Zitat Zhang P, Zhou X, Zeng Y. Multiplexed immunophenotyping of circulating exosomes on nano-engineered ExoProfile chip towards early diagnosis of cancer. Chem Sci. 2019;10:5495–504.PubMedPubMedCentral Zhang P, Zhou X, Zeng Y. Multiplexed immunophenotyping of circulating exosomes on nano-engineered ExoProfile chip towards early diagnosis of cancer. Chem Sci. 2019;10:5495–504.PubMedPubMedCentral
150.
Zurück zum Zitat Tayebi M, Zhou Y, Tripathi P, Chandramohanadas R, Ai Y. Exosome purification and analysis using a facile microfluidic hydrodynamic trapping device. Anal Chem. 2020;92:10733–42.PubMed Tayebi M, Zhou Y, Tripathi P, Chandramohanadas R, Ai Y. Exosome purification and analysis using a facile microfluidic hydrodynamic trapping device. Anal Chem. 2020;92:10733–42.PubMed
151.
Zurück zum Zitat Cho S, Yang HC, Rhee WJ. Simultaneous multiplexed detection of exosomal microRNAs and surface proteins for prostate cancer diagnosis. Biosens Bioelectron. 2019;146:111749.PubMed Cho S, Yang HC, Rhee WJ. Simultaneous multiplexed detection of exosomal microRNAs and surface proteins for prostate cancer diagnosis. Biosens Bioelectron. 2019;146:111749.PubMed
152.
Zurück zum Zitat Zhang Z, Tang C, Zhao L, Xu L, Zhou W, Dong Z, et al. Aptamer-based fluorescence polarization assay for separation-free exosome quantification. Nanoscale. 2019;11:10106–13.PubMed Zhang Z, Tang C, Zhao L, Xu L, Zhou W, Dong Z, et al. Aptamer-based fluorescence polarization assay for separation-free exosome quantification. Nanoscale. 2019;11:10106–13.PubMed
153.
Zurück zum Zitat Tayebi M, Tavakkoli Yaraki M, Yang HY, Ai Y. A MoS2–MWCNT based fluorometric nanosensor for exosome detection and quantification. Nanoscale Adv. 2019;1:2866–72. Tayebi M, Tavakkoli Yaraki M, Yang HY, Ai Y. A MoS2–MWCNT based fluorometric nanosensor for exosome detection and quantification. Nanoscale Adv. 2019;1:2866–72.
154.
Zurück zum Zitat Wang S, Zhang L, Wan S, Cansiz S, Cui C, Liu Y, et al. Aptasensor with expanded nucleotide using DNA nanotetrahedra for electrochemical detection of cancerous exosomes. ACS Nano. 2017;11:3943–9.PubMedPubMedCentral Wang S, Zhang L, Wan S, Cansiz S, Cui C, Liu Y, et al. Aptasensor with expanded nucleotide using DNA nanotetrahedra for electrochemical detection of cancerous exosomes. ACS Nano. 2017;11:3943–9.PubMedPubMedCentral
155.
Zurück zum Zitat Dong H, Chen H, Jiang J, Zhang H, Cai C, Shen Q. Highly sensitive electrochemical detection of tumor exosomes based on aptamer recognition-induced multi-DNA release and cyclic enzymatic amplification. Anal Chem. 2018;90:4507–13.PubMed Dong H, Chen H, Jiang J, Zhang H, Cai C, Shen Q. Highly sensitive electrochemical detection of tumor exosomes based on aptamer recognition-induced multi-DNA release and cyclic enzymatic amplification. Anal Chem. 2018;90:4507–13.PubMed
156.
Zurück zum Zitat Huang R, He L, Xia Y, Xu H, Liu C, Xie H, et al. A sensitive Aptasensor based on a hemin/G-Quadruplex-assisted signal amplification strategy for electrochemical detection of gastric cancer exosomes. Small (Weinheim an der Bergstrasse, Germany). 2019;15:e1900735. Huang R, He L, Xia Y, Xu H, Liu C, Xie H, et al. A sensitive Aptasensor based on a hemin/G-Quadruplex-assisted signal amplification strategy for electrochemical detection of gastric cancer exosomes. Small (Weinheim an der Bergstrasse, Germany). 2019;15:e1900735.
157.
Zurück zum Zitat Zhu L, Wang K, Cui J, Liu H, Bu X, Ma H, et al. Label-free quantitative detection of tumor-derived exosomes through surface plasmon resonance imaging. Anal Chem. 2014;86:8857–64.PubMedPubMedCentral Zhu L, Wang K, Cui J, Liu H, Bu X, Ma H, et al. Label-free quantitative detection of tumor-derived exosomes through surface plasmon resonance imaging. Anal Chem. 2014;86:8857–64.PubMedPubMedCentral
158.
Zurück zum Zitat Wang Q, Zou L, Yang X, Liu X, Nie W, Zheng Y, et al. Direct quantification of cancerous exosomes via surface plasmon resonance with dual gold nanoparticle-assisted signal amplification. Biosens Bioelectron. 2019;135:129–36.PubMed Wang Q, Zou L, Yang X, Liu X, Nie W, Zheng Y, et al. Direct quantification of cancerous exosomes via surface plasmon resonance with dual gold nanoparticle-assisted signal amplification. Biosens Bioelectron. 2019;135:129–36.PubMed
159.
Zurück zum Zitat Fan Y, Duan X, Zhao M, Wei X, Wu J, Chen W, et al. High-sensitive and multiplex biosensing assay of NSCLC-derived exosomes via different recognition sites based on SPRi array. Biosens Bioelectron. 2020;154:112066.PubMed Fan Y, Duan X, Zhao M, Wei X, Wu J, Chen W, et al. High-sensitive and multiplex biosensing assay of NSCLC-derived exosomes via different recognition sites based on SPRi array. Biosens Bioelectron. 2020;154:112066.PubMed
160.
Zurück zum Zitat Sina AAI, Vaidyanathan R, Wuethrich A, Carrascosa LG, Trau M. Label-free detection of exosomes using a surface plasmon resonance biosensor. Anal Bioanal Chem. 2019;411:1311–8.PubMed Sina AAI, Vaidyanathan R, Wuethrich A, Carrascosa LG, Trau M. Label-free detection of exosomes using a surface plasmon resonance biosensor. Anal Bioanal Chem. 2019;411:1311–8.PubMed
161.
Zurück zum Zitat Thakur A, Xu C, Li WK, Qiu G, He B, Ng S-P, et al. In vivo liquid biopsy for glioblastoma malignancy by the AFM and LSPR based sensing of exosomal CD44 and CD133 in a mouse model. Biosens Bioelectron. 2021;191:113476.PubMed Thakur A, Xu C, Li WK, Qiu G, He B, Ng S-P, et al. In vivo liquid biopsy for glioblastoma malignancy by the AFM and LSPR based sensing of exosomal CD44 and CD133 in a mouse model. Biosens Bioelectron. 2021;191:113476.PubMed
162.
Zurück zum Zitat Shin H, Oh S, Hong S, Kang M, Kang D, Ji Y-G, et al. Early-stage lung cancer diagnosis by deep learning-based spectroscopic analysis of circulating exosomes. ACS Nano. 2020;14:5435–44.PubMed Shin H, Oh S, Hong S, Kang M, Kang D, Ji Y-G, et al. Early-stage lung cancer diagnosis by deep learning-based spectroscopic analysis of circulating exosomes. ACS Nano. 2020;14:5435–44.PubMed
163.
Zurück zum Zitat Wang C, Ding Q, Plant P, Basheer M, Yang C, Tawedrous E, et al. Droplet digital PCR improves urinary exosomal miRNA detection compared to real-time PCR. Clin Biochem. 2019;67:54–9.PubMed Wang C, Ding Q, Plant P, Basheer M, Yang C, Tawedrous E, et al. Droplet digital PCR improves urinary exosomal miRNA detection compared to real-time PCR. Clin Biochem. 2019;67:54–9.PubMed
164.
Zurück zum Zitat Chen WW, Balaj L, Liau LM, Samuels ML, Kotsopoulos SK, Maguire CA, et al. BEAMing and droplet digital PCR analysis of mutant IDH1 mRNA in glioma patient serum and cerebrospinal fluid extracellular vesicles. Mol Ther Nucleic Acids. 2013;2:e109.PubMedPubMedCentral Chen WW, Balaj L, Liau LM, Samuels ML, Kotsopoulos SK, Maguire CA, et al. BEAMing and droplet digital PCR analysis of mutant IDH1 mRNA in glioma patient serum and cerebrospinal fluid extracellular vesicles. Mol Ther Nucleic Acids. 2013;2:e109.PubMedPubMedCentral
165.
Zurück zum Zitat Zhang P, Zhou X, He M, Shang Y, Tetlow AL, Godwin AK, et al. Ultrasensitive detection of circulating exosomes with a 3D-nanopatterned microfluidic chip. Nat Biomed Eng. 2019;3:438–51.PubMedPubMedCentral Zhang P, Zhou X, He M, Shang Y, Tetlow AL, Godwin AK, et al. Ultrasensitive detection of circulating exosomes with a 3D-nanopatterned microfluidic chip. Nat Biomed Eng. 2019;3:438–51.PubMedPubMedCentral
166.
Zurück zum Zitat Bai Y, Qu Y, Wu Z, Ren Y, Cheng Z, Lu Y, et al. Absolute quantification and analysis of extracellular vesicle lncRNAs from the peripheral blood of patients with lung cancer based on multi-colour fluorescence chip-based digital PCR. Biosens Bioelectron. 2019;142:111523.PubMed Bai Y, Qu Y, Wu Z, Ren Y, Cheng Z, Lu Y, et al. Absolute quantification and analysis of extracellular vesicle lncRNAs from the peripheral blood of patients with lung cancer based on multi-colour fluorescence chip-based digital PCR. Biosens Bioelectron. 2019;142:111523.PubMed
167.
Zurück zum Zitat Stella M, Falzone L, Caponnetto A, Gattuso G, Barbagallo C, Battaglia R, et al. Serum extracellular vesicle-derived circHIPK3 and circSMARCA5 are two novel diagnostic biomarkers for glioblastoma multiforme. Pharmaceuticals (Basel, Switzerland). 2021;14(7):618. Stella M, Falzone L, Caponnetto A, Gattuso G, Barbagallo C, Battaglia R, et al. Serum extracellular vesicle-derived circHIPK3 and circSMARCA5 are two novel diagnostic biomarkers for glioblastoma multiforme. Pharmaceuticals (Basel, Switzerland). 2021;14(7):618.
168.
Zurück zum Zitat Lee J, Kwon MH, Kim JA, Rhee WJ. Detection of exosome miRNAs using molecular beacons for diagnosing prostate cancer. Artif Cells Nanomed Biotechnol. 2018;46:S52–63.PubMed Lee J, Kwon MH, Kim JA, Rhee WJ. Detection of exosome miRNAs using molecular beacons for diagnosing prostate cancer. Artif Cells Nanomed Biotechnol. 2018;46:S52–63.PubMed
169.
Zurück zum Zitat Chen Y, Zhai L-Y, Zhang L-M, Ma X-S, Liu Z, Li M-M, et al. Breast cancer plasma biopsy by in situ determination of exosomal microRNA-1246 with a molecular beacon. Analyst. 2021;146:2264–76.PubMed Chen Y, Zhai L-Y, Zhang L-M, Ma X-S, Liu Z, Li M-M, et al. Breast cancer plasma biopsy by in situ determination of exosomal microRNA-1246 with a molecular beacon. Analyst. 2021;146:2264–76.PubMed
170.
Zurück zum Zitat Oliveira GP, Zigon E, Rogers G, Davodian D, Lu S, Jovanovic-Talisman T, et al. Detection of extracellular vesicle RNA using molecular beacons. iScience. 2020;23:100782.PubMed Oliveira GP, Zigon E, Rogers G, Davodian D, Lu S, Jovanovic-Talisman T, et al. Detection of extracellular vesicle RNA using molecular beacons. iScience. 2020;23:100782.PubMed
171.
Zurück zum Zitat Chen X, Jia M, Liu L, Qiu X, Zhang H, Yu X, et al. High-fidelity determination and tracing of small extracellular vesicle cargoes. Small (Weinheim an der Bergstrasse, Germany). 2020;16:e2002800. Chen X, Jia M, Liu L, Qiu X, Zhang H, Yu X, et al. High-fidelity determination and tracing of small extracellular vesicle cargoes. Small (Weinheim an der Bergstrasse, Germany). 2020;16:e2002800.
172.
Zurück zum Zitat Zhang Y, Zhang X, Situ B, Wu Y, Luo S, Zheng L, et al. Rapid electrochemical biosensor for sensitive profiling of exosomal microRNA based on multifunctional DNA tetrahedron assisted catalytic hairpin assembly. Biosens Bioelectron. 2021;183:113205.PubMed Zhang Y, Zhang X, Situ B, Wu Y, Luo S, Zheng L, et al. Rapid electrochemical biosensor for sensitive profiling of exosomal microRNA based on multifunctional DNA tetrahedron assisted catalytic hairpin assembly. Biosens Bioelectron. 2021;183:113205.PubMed
173.
Zurück zum Zitat Joshi GK, Deitz-McElyea S, Liyanage T, Lawrence K, Mali S, Sardar R, et al. Label-free Nanoplasmonic-based short noncoding RNA sensing at Attomolar concentrations allows for quantitative and highly specific assay of MicroRNA-10b in biological fluids and circulating exosomes. ACS Nano. 2015;9:11075–89.PubMedPubMedCentral Joshi GK, Deitz-McElyea S, Liyanage T, Lawrence K, Mali S, Sardar R, et al. Label-free Nanoplasmonic-based short noncoding RNA sensing at Attomolar concentrations allows for quantitative and highly specific assay of MicroRNA-10b in biological fluids and circulating exosomes. ACS Nano. 2015;9:11075–89.PubMedPubMedCentral
174.
Zurück zum Zitat Thakur A, Qiu G, Ng S-P, Guan J, Yue J, Lee Y, et al. Direct detection of two different tumor-derived extracellular vesicles by SAM-AuNIs LSPR biosensor. Biosens Bioelectron. 2017;94:400–7.PubMed Thakur A, Qiu G, Ng S-P, Guan J, Yue J, Lee Y, et al. Direct detection of two different tumor-derived extracellular vesicles by SAM-AuNIs LSPR biosensor. Biosens Bioelectron. 2017;94:400–7.PubMed
175.
Zurück zum Zitat Zhao J, Liu C, Li Y, Ma Y, Deng J, Li L, et al. Thermophoretic detection of exosomal microRNAs by nanoflares. J Am Chem Soc. 2020;142:4996–5001.PubMed Zhao J, Liu C, Li Y, Ma Y, Deng J, Li L, et al. Thermophoretic detection of exosomal microRNAs by nanoflares. J Am Chem Soc. 2020;142:4996–5001.PubMed
176.
Zurück zum Zitat Wang R, Zhao X, Chen X, Qiu X, Qing G, Zhang H, et al. Rolling circular amplification (RCA)-assisted CRISPR/Cas9 cleavage (RACE) for highly specific detection of multiple extracellular vesicle MicroRNAs. Anal Chem. 2020;92:2176–85.PubMed Wang R, Zhao X, Chen X, Qiu X, Qing G, Zhang H, et al. Rolling circular amplification (RCA)-assisted CRISPR/Cas9 cleavage (RACE) for highly specific detection of multiple extracellular vesicle MicroRNAs. Anal Chem. 2020;92:2176–85.PubMed
177.
Zurück zum Zitat Olmedillas-López S, García-Arranz M, García-Olmo D. Current and emerging applications of droplet digital PCR in oncology. Mol Diagn Ther. 2017;21:493–510.PubMed Olmedillas-López S, García-Arranz M, García-Olmo D. Current and emerging applications of droplet digital PCR in oncology. Mol Diagn Ther. 2017;21:493–510.PubMed
178.
Zurück zum Zitat Shen X, Yang Y, Chen Y, Zhou C, Zhao X, Li N, et al. Evaluation of EpCAM-specific exosomal lncRNAs as potential diagnostic biomarkers for lung cancer using droplet digital PCR. J Mol Med (Berlin, Germany). 2022;100(1):87–100. Shen X, Yang Y, Chen Y, Zhou C, Zhao X, Li N, et al. Evaluation of EpCAM-specific exosomal lncRNAs as potential diagnostic biomarkers for lung cancer using droplet digital PCR. J Mol Med (Berlin, Germany). 2022;100(1):87–100.
179.
Zurück zum Zitat Rhee WJ, Jeong S. Extracellular vesicle miRNA detection using molecular beacons. Methods Mol Biol (Clifton, NJ). 2017;1660:287–94. Rhee WJ, Jeong S. Extracellular vesicle miRNA detection using molecular beacons. Methods Mol Biol (Clifton, NJ). 2017;1660:287–94.
180.
Zurück zum Zitat Zhang Y, Zhao J, Dong J, Chen Y, Si W, Sha J. A system and method for isolation and detection of exotic tumor marker miRNA. (CN201910064022.7) Zhang Y, Zhao J, Dong J, Chen Y, Si W, Sha J. A system and method for isolation and detection of exotic tumor marker miRNA. (CN201910064022.7)
181.
Zurück zum Zitat Ozhalici-Unal H, Armitage BA. Fluorescent DNA nanotags based on a self-assembled DNA tetrahedron. ACS Nano. 2009;3:425–33.PubMed Ozhalici-Unal H, Armitage BA. Fluorescent DNA nanotags based on a self-assembled DNA tetrahedron. ACS Nano. 2009;3:425–33.PubMed
182.
Zurück zum Zitat Gao J, Zhang H, Wang Z. A DNA tetrahedron nanoprobe-based fluorescence resonance energy transfer sensing platform for intracellular tumor-related miRNA detection. Analyst. 2020;145:3535–42.PubMed Gao J, Zhang H, Wang Z. A DNA tetrahedron nanoprobe-based fluorescence resonance energy transfer sensing platform for intracellular tumor-related miRNA detection. Analyst. 2020;145:3535–42.PubMed
183.
Zurück zum Zitat Han Z, Wan F, Deng J, Zhao J, Sun J. Ultrasensitive detection of mRNA in extracellular vesicles using DNA tetrahedron-based thermophoretic assay. Nano Today. 2021;38:101203. Han Z, Wan F, Deng J, Zhao J, Sun J. Ultrasensitive detection of mRNA in extracellular vesicles using DNA tetrahedron-based thermophoretic assay. Nano Today. 2021;38:101203.
184.
Zurück zum Zitat Feng W, Newbigging AM, Tao J, Cao Y, Peng H, Le C, et al. CRISPR technology incorporating amplification strategies: molecular assays for nucleic acids, proteins, and small molecules. Chem Sci. 2021;12:4683–98.PubMedPubMedCentral Feng W, Newbigging AM, Tao J, Cao Y, Peng H, Le C, et al. CRISPR technology incorporating amplification strategies: molecular assays for nucleic acids, proteins, and small molecules. Chem Sci. 2021;12:4683–98.PubMedPubMedCentral
185.
Zurück zum Zitat Duan J, Lu G, Xie Z, Lou M, Luo J, Guo L, et al. Genome-wide identification of CRISPR/Cas9 off-targets in human genome. Cell Res. 2014;24:1009–12.PubMedPubMedCentral Duan J, Lu G, Xie Z, Lou M, Luo J, Guo L, et al. Genome-wide identification of CRISPR/Cas9 off-targets in human genome. Cell Res. 2014;24:1009–12.PubMedPubMedCentral
186.
Zurück zum Zitat LeCun Y, Bengio Y, Hinton G. Deep learning. Nature. 2015;521:436–44.PubMed LeCun Y, Bengio Y, Hinton G. Deep learning. Nature. 2015;521:436–44.PubMed
187.
Zurück zum Zitat Kawakami E, Tabata J, Yanaihara N, Ishikawa T, Koseki K, Iida Y, et al. Application of artificial intelligence for preoperative diagnostic and prognostic prediction in epithelial ovarian Cancer based on blood biomarkers. Clin Cancer Res. 2019;25:3006–15.PubMed Kawakami E, Tabata J, Yanaihara N, Ishikawa T, Koseki K, Iida Y, et al. Application of artificial intelligence for preoperative diagnostic and prognostic prediction in epithelial ovarian Cancer based on blood biomarkers. Clin Cancer Res. 2019;25:3006–15.PubMed
188.
Zurück zum Zitat Wu N, Zhang X-Y, Xia J, Li X, Yang T, Wang J-H. Ratiometric 3D DNA machine combined with machine learning algorithm for ultrasensitive and high-precision screening of early urinary diseases. ACS Nano. 2021;15(12):19522–34.PubMed Wu N, Zhang X-Y, Xia J, Li X, Yang T, Wang J-H. Ratiometric 3D DNA machine combined with machine learning algorithm for ultrasensitive and high-precision screening of early urinary diseases. ACS Nano. 2021;15(12):19522–34.PubMed
189.
Zurück zum Zitat Lee YR, Kim G, Tak WY, Jang SY, Kweon YO, Park JG, et al. Circulating exosomal noncoding RNAs as prognostic biomarkers in human hepatocellular carcinoma. Int J Cancer. 2019;144:1444–52.PubMed Lee YR, Kim G, Tak WY, Jang SY, Kweon YO, Park JG, et al. Circulating exosomal noncoding RNAs as prognostic biomarkers in human hepatocellular carcinoma. Int J Cancer. 2019;144:1444–52.PubMed
190.
Zurück zum Zitat Vasconcelos MH, Caires HR, Ābols A, Xavier CPR, Linē A. Extracellular vesicles as a novel source of biomarkers in liquid biopsies for monitoring cancer progression and drug resistance. Drug Resist Updates. 2019;47:100647. Vasconcelos MH, Caires HR, Ābols A, Xavier CPR, Linē A. Extracellular vesicles as a novel source of biomarkers in liquid biopsies for monitoring cancer progression and drug resistance. Drug Resist Updates. 2019;47:100647.
191.
Zurück zum Zitat Zhao R, Zhang Y, Zhang X, Yang Y, Zheng X, Li X, et al. Exosomal long noncoding RNA HOTTIP as potential novel diagnostic and prognostic biomarker test for gastric cancer. Mol Cancer. 2018;17:68.PubMedPubMedCentral Zhao R, Zhang Y, Zhang X, Yang Y, Zheng X, Li X, et al. Exosomal long noncoding RNA HOTTIP as potential novel diagnostic and prognostic biomarker test for gastric cancer. Mol Cancer. 2018;17:68.PubMedPubMedCentral
192.
Zurück zum Zitat Wei S, Peng L, Yang J, Sang H, Jin D, Li X, et al. Exosomal transfer of miR-15b-3p enhances tumorigenesis and malignant transformation through the DYNLT1/Caspase-3/Caspase-9 signaling pathway in gastric cancer. J Exp Clin Cancer Res. 2020;39:32.PubMedPubMedCentral Wei S, Peng L, Yang J, Sang H, Jin D, Li X, et al. Exosomal transfer of miR-15b-3p enhances tumorigenesis and malignant transformation through the DYNLT1/Caspase-3/Caspase-9 signaling pathway in gastric cancer. J Exp Clin Cancer Res. 2020;39:32.PubMedPubMedCentral
193.
Zurück zum Zitat Cao L-Q, Yang X-W, Chen Y-B, Zhang D-W, Jiang X-F, Xue P. Correction to: Exosomal miR-21 regulates the TETs/PTENp1/PTEN pathway to promote hepatocellular carcinoma growth. Mol Cancer. 2020;19:59.PubMedPubMedCentral Cao L-Q, Yang X-W, Chen Y-B, Zhang D-W, Jiang X-F, Xue P. Correction to: Exosomal miR-21 regulates the TETs/PTENp1/PTEN pathway to promote hepatocellular carcinoma growth. Mol Cancer. 2020;19:59.PubMedPubMedCentral
194.
Zurück zum Zitat Nakano T, Chen IH, Wang C-C, Chen P-J, Tseng H-P, Huang K-T, et al. Circulating exosomal miR-92b: its role for cancer immunoediting and clinical value for prediction of posttransplant hepatocellular carcinoma recurrence. Am J Transplant Off J Am Soc Transplant Am Soc Transplant Surg. 2019;19:3250–62. Nakano T, Chen IH, Wang C-C, Chen P-J, Tseng H-P, Huang K-T, et al. Circulating exosomal miR-92b: its role for cancer immunoediting and clinical value for prediction of posttransplant hepatocellular carcinoma recurrence. Am J Transplant Off J Am Soc Transplant Am Soc Transplant Surg. 2019;19:3250–62.
195.
Zurück zum Zitat Pang Y, Wang C, Lu L, Wang C, Sun Z, Xiao R. Dual-SERS biosensor for one-step detection of microRNAs in exosome and residual plasma of blood samples for diagnosing pancreatic cancer. Biosens Bioelectron. 2019;130:204–13.PubMed Pang Y, Wang C, Lu L, Wang C, Sun Z, Xiao R. Dual-SERS biosensor for one-step detection of microRNAs in exosome and residual plasma of blood samples for diagnosing pancreatic cancer. Biosens Bioelectron. 2019;130:204–13.PubMed
196.
Zurück zum Zitat Ko J, Bhagwat N, Yee SS, Ortiz N, Sahmoud A, Black T, et al. Combining machine learning and nanofluidic technology to diagnose pancreatic cancer using exosomes. ACS Nano. 2017;11:11182–93.PubMed Ko J, Bhagwat N, Yee SS, Ortiz N, Sahmoud A, Black T, et al. Combining machine learning and nanofluidic technology to diagnose pancreatic cancer using exosomes. ACS Nano. 2017;11:11182–93.PubMed
197.
Zurück zum Zitat Lewis JM, Vyas AD, Qiu Y, Messer KS, White R, Heller MJ. Integrated analysis of exosomal protein biomarkers on alternating current electrokinetic chips enables rapid detection of pancreatic cancer in patient blood. ACS Nano. 2018;12:3311–20.PubMed Lewis JM, Vyas AD, Qiu Y, Messer KS, White R, Heller MJ. Integrated analysis of exosomal protein biomarkers on alternating current electrokinetic chips enables rapid detection of pancreatic cancer in patient blood. ACS Nano. 2018;12:3311–20.PubMed
198.
Zurück zum Zitat Carmicheal J, Hayashi C, Huang X, Liu L, Lu Y, Krasnoslobodtsev A, et al. Label-free characterization of exosome via surface enhanced Raman spectroscopy for the early detection of pancreatic cancer. Nanomedicine. 2019;16:88–96.PubMed Carmicheal J, Hayashi C, Huang X, Liu L, Lu Y, Krasnoslobodtsev A, et al. Label-free characterization of exosome via surface enhanced Raman spectroscopy for the early detection of pancreatic cancer. Nanomedicine. 2019;16:88–96.PubMed
199.
Zurück zum Zitat Li T-D, Zhang R, Chen H, Huang Z-P, Ye X, Wang H, et al. An ultrasensitive polydopamine bi-functionalized SERS immunoassay for exosome-based diagnosis and classification of pancreatic cancer. Chem Sci. 2018;9:5372–82.PubMedPubMedCentral Li T-D, Zhang R, Chen H, Huang Z-P, Ye X, Wang H, et al. An ultrasensitive polydopamine bi-functionalized SERS immunoassay for exosome-based diagnosis and classification of pancreatic cancer. Chem Sci. 2018;9:5372–82.PubMedPubMedCentral
200.
Zurück zum Zitat Pan B, Qin J, Liu X, He B, Wang X, Pan Y, et al. Identification of serum exosomal hsa-circ-0004771 as a novel diagnostic biomarker of colorectal cancer. Front Genet. 2019;10:1096.PubMedPubMedCentral Pan B, Qin J, Liu X, He B, Wang X, Pan Y, et al. Identification of serum exosomal hsa-circ-0004771 as a novel diagnostic biomarker of colorectal cancer. Front Genet. 2019;10:1096.PubMedPubMedCentral
201.
Zurück zum Zitat Ma D, Huang C, Zheng J, Tang J, Li J, Yang J, et al. Quantitative detection of exosomal microRNA extracted from human blood based on surface-enhanced Raman scattering. Biosens Bioelectron. 2018;101:167–73.PubMed Ma D, Huang C, Zheng J, Tang J, Li J, Yang J, et al. Quantitative detection of exosomal microRNA extracted from human blood based on surface-enhanced Raman scattering. Biosens Bioelectron. 2018;101:167–73.PubMed
202.
Zurück zum Zitat Li Y, Deng J, Han Z, Liu C, Tian F, Xu R, et al. Molecular identification of tumor-derived extracellular vesicles using thermophoresis-mediated DNA computation. J Am Chem Soc. 2021;143:1290–5.PubMed Li Y, Deng J, Han Z, Liu C, Tian F, Xu R, et al. Molecular identification of tumor-derived extracellular vesicles using thermophoresis-mediated DNA computation. J Am Chem Soc. 2021;143:1290–5.PubMed
203.
Zurück zum Zitat Lu F, Pan S, Qi Y, Li X, Wang J. The clinical application value of RDW, CA153, and MPV in breast cancer. Clin Lab. 2021;67(2). Lu F, Pan S, Qi Y, Li X, Wang J. The clinical application value of RDW, CA153, and MPV in breast cancer. Clin Lab. 2021;67(2).
204.
Zurück zum Zitat Lee JU, Kim WH, Lee HS, Park KH, Sim SJ. Quantitative and specific detection of exosomal miRNAs for accurate diagnosis of breast cancer using a surface-enhanced Raman scattering sensor based on plasmonic head-flocked gold nanopillars. Small (Weinheim an der Bergstrasse, Germany). 2019;15:e1804968. Lee JU, Kim WH, Lee HS, Park KH, Sim SJ. Quantitative and specific detection of exosomal miRNAs for accurate diagnosis of breast cancer using a surface-enhanced Raman scattering sensor based on plasmonic head-flocked gold nanopillars. Small (Weinheim an der Bergstrasse, Germany). 2019;15:e1804968.
205.
Zurück zum Zitat Rodríguez M, Bajo-Santos C, Hessvik NP, Lorenz S, Fromm B, Berge V, et al. Identification of non-invasive miRNAs biomarkers for prostate cancer by deep sequencing analysis of urinary exosomes. Mol Cancer. 2017;16:156.PubMedPubMedCentral Rodríguez M, Bajo-Santos C, Hessvik NP, Lorenz S, Fromm B, Berge V, et al. Identification of non-invasive miRNAs biomarkers for prostate cancer by deep sequencing analysis of urinary exosomes. Mol Cancer. 2017;16:156.PubMedPubMedCentral
206.
Zurück zum Zitat Zhou C, Chen Y, He X, Zheng Z, Xue D. Functional implication of exosomal miR-217 and miR-23b-3p in the progression of prostate cancer. Onco Targets Ther. 2020;13:11595–606.PubMedPubMedCentral Zhou C, Chen Y, He X, Zheng Z, Xue D. Functional implication of exosomal miR-217 and miR-23b-3p in the progression of prostate cancer. Onco Targets Ther. 2020;13:11595–606.PubMedPubMedCentral
207.
Zurück zum Zitat Li S, Zhao Y, Chen W, Yin L, Zhu J, Zhang H, et al. Exosomal ephrinA2 derived from serum as a potential biomarker for prostate cancer. J Cancer. 2018;9:2659–65.PubMedPubMedCentral Li S, Zhao Y, Chen W, Yin L, Zhu J, Zhang H, et al. Exosomal ephrinA2 derived from serum as a potential biomarker for prostate cancer. J Cancer. 2018;9:2659–65.PubMedPubMedCentral
208.
Zurück zum Zitat Zhu S, Ni Y, Sun G, Wang Z, Chen J, Zhang X, et al. Exosomal TUBB3 mRNA expression of metastatic castration-resistant prostate cancer patients: association with patient outcome under abiraterone. Cancer Med. 2021;10(18):6282–90.PubMedPubMedCentral Zhu S, Ni Y, Sun G, Wang Z, Chen J, Zhang X, et al. Exosomal TUBB3 mRNA expression of metastatic castration-resistant prostate cancer patients: association with patient outcome under abiraterone. Cancer Med. 2021;10(18):6282–90.PubMedPubMedCentral
209.
Zurück zum Zitat Liu J, Yoo J, Ho JY, Jung Y, Lee S, Hur SY, et al. Plasma-derived exosomal miR-4732-5p is a promising noninvasive diagnostic biomarker for epithelial ovarian cancer. J Ovarian Res. 2021;14:59.PubMedPubMedCentral Liu J, Yoo J, Ho JY, Jung Y, Lee S, Hur SY, et al. Plasma-derived exosomal miR-4732-5p is a promising noninvasive diagnostic biomarker for epithelial ovarian cancer. J Ovarian Res. 2021;14:59.PubMedPubMedCentral
210.
Zurück zum Zitat Zhu Y, Wang S, Xi X, Zhang M, Liu X, Tang W, et al. Integrative analysis of long extracellular RNAs reveals a detection panel of noncoding RNAs for liver cancer. Theranostics. 2021;11:181–93.PubMedPubMedCentral Zhu Y, Wang S, Xi X, Zhang M, Liu X, Tang W, et al. Integrative analysis of long extracellular RNAs reveals a detection panel of noncoding RNAs for liver cancer. Theranostics. 2021;11:181–93.PubMedPubMedCentral
211.
Zurück zum Zitat Preis M, Gardner TB, Gordon SR, Pipas JM, Mackenzie TA, Klein EE, et al. MicroRNA-10b expression correlates with response to neoadjuvant therapy and survival in pancreatic ductal adenocarcinoma. Clin Cancer Res. 2011;17:5812–21.PubMedPubMedCentral Preis M, Gardner TB, Gordon SR, Pipas JM, Mackenzie TA, Klein EE, et al. MicroRNA-10b expression correlates with response to neoadjuvant therapy and survival in pancreatic ductal adenocarcinoma. Clin Cancer Res. 2011;17:5812–21.PubMedPubMedCentral
212.
Zurück zum Zitat Han L, Shi W-J, Xie Y-B, Zhang Z-G. Diagnostic value of four serum exosome microRNAs panel for the detection of colorectal cancer. World J Gastrointest Oncol. 2021;13:970–9.PubMedPubMedCentral Han L, Shi W-J, Xie Y-B, Zhang Z-G. Diagnostic value of four serum exosome microRNAs panel for the detection of colorectal cancer. World J Gastrointest Oncol. 2021;13:970–9.PubMedPubMedCentral
213.
Zurück zum Zitat Aidoo-Brown J, Moschou D, Estrela P. Multiplexed prostate cancer companion diagnostic devices. Sensors (Basel, Switzerland). 2021;21(15):5023. Aidoo-Brown J, Moschou D, Estrela P. Multiplexed prostate cancer companion diagnostic devices. Sensors (Basel, Switzerland). 2021;21(15):5023.
214.
Zurück zum Zitat McKiernan J, Donovan MJ, O'Neill V, Bentink S, Noerholm M, Belzer S, et al. A novel urine exosome gene expression assay to predict high-grade prostate cancer at initial biopsy. JAMA Oncol. 2016;2:882–9.PubMed McKiernan J, Donovan MJ, O'Neill V, Bentink S, Noerholm M, Belzer S, et al. A novel urine exosome gene expression assay to predict high-grade prostate cancer at initial biopsy. JAMA Oncol. 2016;2:882–9.PubMed
215.
Zurück zum Zitat McKiernan J, Donovan MJ, Margolis E, Partin A, Carter B, Brown G, 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 2-10ng/ml at initial biopsy. Eur Urol. 2018;74:731–8.PubMed McKiernan J, Donovan MJ, Margolis E, Partin A, Carter B, Brown G, 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 2-10ng/ml at initial biopsy. Eur Urol. 2018;74:731–8.PubMed
Metadaten
Titel
Exosomes as a new frontier of cancer liquid biopsy
verfasst von
Dan Yu
Yixin Li
Maoye Wang
Jianmei Gu
Wenrong Xu
Hui Cai
Xinjian Fang
Xu Zhang
Publikationsdatum
01.12.2022
Verlag
BioMed Central
Erschienen in
Molecular Cancer / Ausgabe 1/2022
Elektronische ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-022-01509-9

Weitere Artikel der Ausgabe 1/2022

Molecular Cancer 1/2022 Zur Ausgabe

Alphablocker schützt vor Miktionsproblemen nach der Biopsie

16.05.2024 alpha-1-Rezeptorantagonisten Nachrichten

Nach einer Prostatabiopsie treten häufig Probleme beim Wasserlassen auf. Ob sich das durch den periinterventionellen Einsatz von Alphablockern verhindern lässt, haben australische Mediziner im Zuge einer Metaanalyse untersucht.

Antikörper-Wirkstoff-Konjugat hält solide Tumoren in Schach

16.05.2024 Zielgerichtete Therapie Nachrichten

Trastuzumab deruxtecan scheint auch jenseits von Lungenkrebs gut gegen solide Tumoren mit HER2-Mutationen zu wirken. Dafür sprechen die Daten einer offenen Pan-Tumor-Studie.

Mammakarzinom: Senken Statine das krebsbedingte Sterberisiko?

15.05.2024 Mammakarzinom Nachrichten

Frauen mit lokalem oder metastasiertem Brustkrebs, die Statine einnehmen, haben eine niedrigere krebsspezifische Mortalität als Patientinnen, die dies nicht tun, legen neue Daten aus den USA nahe.

Labor, CT-Anthropometrie zeigen Risiko für Pankreaskrebs

13.05.2024 Pankreaskarzinom Nachrichten

Gerade bei aggressiven Malignomen wie dem duktalen Adenokarzinom des Pankreas könnte Früherkennung die Therapiechancen verbessern. Noch jedoch klafft hier eine Lücke. Ein Studienteam hat einen Weg gesucht, sie zu schließen.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.