Abstract
Extracellular vesicles (EVs), secreted by most living cells, encapsulate a diverse array of bioactive molecules from their parent cells, including proteins and nucleic acids. Recent studies underscore the potential of EVs as advanced biomarkers for the early diagnosis of a variety of clinical diseases. Nevertheless, traditional platforms for EVs separation and detection platforms working alone often involve multiple pieces of equipment and complex, multi-step protocols. This extends processing time and the likelihood of bioanalyte loss and cross-contamination, thereby impeding further EVs research. To date, few studies have effectively combined EVs separation, detection, and analysis functions into a single platform. Integrated microfluidic platforms present a compelling solution by enabling seamless progression from sample to result. These platforms can efficiently combine various separation and detection techniques, simplifying complex workflows and facilitating both efficient EVs separation and high-sensitivity detection. This review concentrates on integrated microfluidic platforms for EVs separation and detection, specifically examining whether the separation and detection units are fully integrated. Recent studies underscore the potential of EVs as promising biomarkers for early-stage diagnosis of diseases, including cancer and neurodegenerative disorders. Recent advances in EVs separation and analysis enable overcoming key translational barriers, accelerating their routine adoption in clinical diagnostics.
I. INTRODUCTION
A. EVs: Biogenesis and clinical potential
Extracellular vesicle (EVs; with diameters ranging from 30 to 150 nm) represent a specialized subtype of EVs, which are membrane-enclosed entities synthesized by living cells via the processes of endocytosis, fusion, and exocytosis.1 EVs are categorized into apoptotic vesicles (>1 μm), microvesicles (100 nm to 1 μm), and EVs (30 to 150 nm).2,3 EVs are instrumental in the stabilization and transfer of cellular cargo, encompassing specific nucleic acids, functional proteins, and lipids, thereby enhancing intercellular communication.4 The biogenesis of EVs is a multifaceted process: initially, the plasma membrane undergoes invagination to generate early sorting endosomes (ESEs). These ESEs subsequently mature into late sorting endosomes (LSEs), which further invaginate to form multivesicular bodies (MVBs).5,6 Ultimately, MVBs are released into the extracellular milieu, transforming into EVs7 (Fig. 1). EVs are abundantly found across various biological fluids, including blood, urine, saliva, breast milk, lymphatic fluid, and cerebrospinal fluid.8 Recent investigations reveal that EVs are significantly more prevalent in the blood of cancer patients compared to healthy individuals. They are implicated in pivotal processes such as oncogenesis, disease progression, metastasis, and therapeutic monitoring.9 For instance, pancreatic cancer patients show elevated levels of glypican-1 EVs in blood, correlating with tumor burden.10 Moreover, EVs have emerged as biomarkers for evaluating therapeutic responses in cancer treatment and serve as indicators in liquid biopsies,11 underscoring their potential as highly effective instruments for minimally invasive diagnostics and early screening in preventive healthcare.
FIG. 1.
Biogenesis process of EVs and schematic diagram of the molecular composition of EVs.
B. Limitations of conventional EVs analysis
A range of conventional techniques for the analysis of EVs, including separation, imaging/morphological characterization, molecular profiling, and clinical application, have been progressively developed. However, these methods still face notable limitations that hinder their broad clinical translation. Achieving high-purity and efficient EVs separation from complex biological samples remains a major challenge in the separation stage. Although ultracentrifugation is widely regarded as the gold standard,12 it is time-consuming and labor-intensive, requires large sample volumes and expensive equipment, and may inadvertently activate platelets or immune cells,13 thereby compromising EVs purity and downstream analyses. For imaging and morphological characterization, high-resolution techniques such as transmission electron microscopy (TEM) and scanning electron microscopy (SEM) require elaborate sample preparation and are unsuitable for high-throughput applications.14 Alternative methods such as dynamic light scattering (DLS) and nanoparticle tracking analysis (NTA) are commonly used to measure particle size and concentration, but they lack sufficient resolution to distinguish heterogeneous EVs populations.15 In the molecular analysis stage, EVs surface markers (e.g., CD9, CD63, and CD81) are typically identified using Western blotting or enzyme-linked immunosorbent assay (ELISA).16 Although these methods provide specificity, their sensitivity is limited, and quantitative performance remains suboptimal. In summary, although conventional EVs separation and detection methods are valuable for basic research, their reliance on complex instrumentation and multi-step workflows limits their suitability for clinical diagnostics, which require high throughput, speed, and cost-effectiveness.
C. Microfluidics as an integrated solution
Microfluidic technology, as an integrated and multifunctional platform, spans and optimizes the entire workflow of EVs analysis and is increasingly emerging as a powerful tool to overcome the limitations of conventional methods.17–19 These limitations underscore the need for integrated platforms that combine high purity, sensitivity, and scalability—criteria increasingly met by microfluidic technologies. In the separation stage, microfluidic devices can achieve high-throughput and highly specific EVs extraction through physical, chemical, or immunological mechanisms.20,21 For imaging and morphological characterization, microfluidic chips are typically fabricated from transparent materials and designed to be compatible with various microscopy systems, enabling in situ visualization and analysis of EVs.22 At the molecular analysis level, microfluidic platforms can integrate multiple sensing modules for the rapid and highly sensitive detection of functional biomolecules such as EV-associated proteins and RNAs.23,24 Finally, in clinical applications,25 microfluidic systems offer advantages such as operational simplicity, low sample volume requirements, and cost-effectiveness, demonstrating strong potential for clinical translation. While microfluidics offers high throughput and integration, challenges remain in standardization and manufacturing scalability. Nevertheless, its ability to combine separation (acoustics, immunoaffinity) with on-chip detection (electrochemical sensors) makes it uniquely suited for clinical translation.26 The ability of microfluidics to integrate separation, detection, and analysis on a single chip makes it a highly promising and practical platform for rapid EVs analysis, addressing the diverse needs of both fundamental research and clinical diagnostics. Some comprehensive reviews have outlined microfluidics-based technologies for EVs separation and detection.27–29 In this review, we place particular emphasis on the integrated microfluidic platforms designed for EVs analysis. We discuss distinct objects of EVs analysis, with a focus on comprehensive EVs analysis, detailed analyses of EVs' proteins and RNA are also provided, using the frameworks of the “combined microfluidic platform” and the “shared microfluidic platform” as foundational models.
II. MICROFLUIDIC EVs SEPARATION AND DETECTION STRATEGIES
Microfluidics entails the precise manipulation of microscale fluid volumes within intricately designed micro- and nanostructured channels.30 This advanced technology facilitates the seamless convergence of sample preparation, analytical detection, and diagnostic operations onto a singular microchip. Boasting key advantages such as minimal sample consumption, accelerated separation and purification processes, enhanced sensitivity, and superior separation efficiency,7,31 microfluidics has rapidly established itself as a formidable tool in the realm of biomedical research.
A. Microfluidic EVs separation strategies
1. Microfluidic EVs separation strategies
Microfluidics has made significant strides in EVs applications, praised for its efficiency in separation and detection.32 Yet, these claims often overlook challenges such as low throughput, inconsistent performance across biological samples, and scalability issues. Despite advantages like miniaturization and automation, microfluidics faces ongoing problems with reproducibility, complex integration, and the need for specialized expertise. Having said that, these microfluidic technologies effectively leverage both the physical characteristics of EVs, such as size and density, and their chemical features, including nucleic acids and surface proteins, by using microchip platforms to facilitate efficient EVs separation and detection.33,34 As a promising alternative to conventional methods, microfluidic-based approaches address current limitations through innovative integration strategies. The combination of techniques such as viscoelastic flow,35 acoustofluidics,36 dielectrophoresis,37 and magnetic field-based microfluidic systems is anticipated to greatly enhance the efficiency of EVs separation,38 enabling high-throughput, label-free separation of EVs directly from complex biological samples (Fig. 2). Table I summarizes various microfluidic-based strategies for exosome separation.
FIG. 2.
A schematic illustration of strategies for the separation and detection of EVs. The separation techniques, such as inertial focusing, viscoelastic fluid, filtration, acoustofluidics, dielectrophoresis (DEP), and magnetic field, are employed to separate EVs from various sample types. For detection, methods like fluorescence detection, surface plasmon resonance (SPR) detection, colorimetric detection, surface-enhanced Raman scattering (SERS), mass spectrometry-based detection, and electrochemical detection assays are utilized to further identify and analyze the separated EVs.
TABLE I.
Summary of microfluidic EVs separation strategies.
Method | Throughput (μl/min) | Recovery (%) | Scalability | Clinical feasibility | References |
---|---|---|---|---|---|
Inertial focusing | 1.67 | 52.8 ± 4.53 | High throughput and robustness; membrane-free system; ease of operation | Efficient submicrometer particle separation; integration with clinical testing | 39 |
Viscoelastic fluids | 3.34 | 87 | Suitable for large-scale production; progress steadily | High recovery and purity; low operational cost and ease of use; high repeatability | 40 |
Filtration | 2.4 | 81 | Low sample volume; high sensitivity and efficiency; quantification on a single chip | Direct EVs separation from blood; quantification of disease markers; fingertip-based testing | 41 |
Acoustofluidics | 10 | 99 | Automation and high reproducibility; continuous flow configuration; adjustable separation parameters | Speed and high efficiency; preservation of EVs integrity; minimal user intervention | 36 |
Dielectrophoresis (DEP) | 30–50 | NA | Rapid separation and recovery process; no sample dilution required; use of whole blood | Minimally invasive technology; on-chip biomarker detection; faster results for diagnostics | 42 |
Magnetic separation | 100 | NA | Enhanced separation efficiency; high sensitivity and low contamination; reversibility and expandability | Efficient separation of T-EVs; minimized contamination; integration with mass spectrometry | 43 |
Inertial focusing, a specialized form of hydrodynamic focusing, leverages the interplay between Dean forces and inertial lift forces exerted on particles of varying sizes within microfluidic channels under the influence of Newtonian fluids.44,45 The equilibrium of these forces generates distinct equilibrium positions along the curved channel, enabling size-dependent sorting of particles.46,47 Tay et al. developed a spiral microchannel system for the rapid purification of submicrometer particles directly from whole blood, achieving a recovery rate of 52.8 ± 4.53% for particles smaller than 1 μm, with a reported resolution approaching 1 μm.39 To further improve scalability, the authors later introduced an inertial microfluidic system based on previous work, enabling the direct separation of nanoscale EVs (exosomes, 50 to 200 nm) and medium-sized EVs (microvesicles, 200 nm to 1 μm) from whole blood.48 That said, its sensitivity to nanoscale particles is limited due to inherently weak inertial forces at that scale. As a result, effective separation of small EVs remains challenging and often requires extended channel lengths to generate adequate Dean flow.49 This trade-off between efficiency and system complexity raises concerns about scalability, particularly for clinical applications requiring rapid, high-throughput EVs processing.
Viscoelastic fluids enable particle separation by exploiting the elastic lift forces generated by the medium's viscoelastic properties,50–53 typically composed of synthetic polymers such as polyvinylpyrrolidone (PVP), polyethylene oxide (PEO), and polyacrylamide (PAA).54 Kim et al. first demonstrated the use of viscoelastic flow in straight microchannels to focus 500 and 200 nm particles, marking a key step toward EVs separation using this mechanism, despite the inability to focus 100 nm particles.55 Building on this, Meng et al. developed a cascaded viscoelastic microfluidic device that enabled high-purity (>97%) and high-recovery (>87%) separation of small extracellular vesicles (sEVs, <200 nm) directly from whole blood.40 The device achieved a 100 nm cutoff size and a throughput of 3000 μl/min, enabling rapid and nondestructive EVs extraction. While promising, viscoelastic microfluidics still faces challenges, including limited fluid stability, reduced reproducibility under high-throughput conditions, and poor scalability. Future efforts should aim to improve flow stability, control particle trajectories more precisely, and facilitate clinical translation through chip-level optimization.
Filtration leverages membranes with precisely defined pore sizes to selectively retain particles larger than the membrane pores, while permitting smaller particles, such as EVs, to pass through, thereby enabling size-based separation.56–58 This method can be classified into three primary types: membrane filtration, column filtration, and ultrafiltration (UF).59–61 Chen et al. developed a microfluidic platform for EVs separation from whole blood using dual membrane filtration, incorporating a 0.2 μm polycarbonate membrane to enhance performance.41 The system achieved over 99% sEV separation efficiency and an 81.1% recovery rate from just 2 μl of blood. Despite its high separation efficiency, the device faced limitations such as membrane clogging and potential EVs damage. To overcome these issues, Li et al. introduced a centrifugal microfluidic system combining dual nanofilters with pneumatic pulse filtration. This pulsatile flow reduced clogging and particle deposition, enabling EVs separation within 30 min and yielding 91% diagnostic accuracy for early breast cancer.62 Filtration-based methods offer simplicity and high purity but typically require external forces such as centrifugation, pressure, or vacuum to drive fluids through membrane pores. As samples concentrate, membrane fouling becomes more likely, reducing efficiency. Additionally, dependence on external actuation hinders integration into clinical workflows.
Acoustofluidics integrates external acoustic fields with microfluidic frameworks to actively sort particles by size,63–65 offering significant advantages in both precision and biocompatibility.66–68 Wu et al. developed an acoustofluidic device employing tilted surface acoustic waves (TSAWs) for continuous-flow exosome separation, achieving a recovery rate of approximately 90%.69 They later introduced a rapid, label-free two-module system, where the first module removed blood cells while maintaining an EVs recovery rate above 99%, and the second module purified EVs with a reported purity of 98.4%.36 An additional acoustic module was subsequently integrated to further improve performance. These results underscore the potential of acoustic fields for efficient, high-purity EVs separation. However, challenges remain, including frequent air bubble formation within the acoustic field and high system costs, which may hinder clinical translation. Improving device stability, reducing system complexity, and validating clinical applicability in large-scale studies are essential next steps.
Dielectrophoresis (DEP) refers to the migration of particles in a non-uniform electric field, where differential forces induce particle or cellular polarization, generating a net dipole moment on the particle's surface.70 This polarization allows the particles to move either along or counter to the electric field, contingent upon factors such as excitation frequency, particle size, and electrode configuration.37 Lewis et al. employed an alternating current electrokinetic (ACE) microarray chip to separate EVs via DEP. In this system, EVs were concentrated in high-field regions around circular microelectrodes, while larger cells were excluded into low-field zones. The device enabled direct EVs enrichment from 30 to 50 μl of whole blood without prior sample processing.42 It was later optimized for in situ immunofluorescent labeling and surface protein analysis on-chip.10 Although the method showed high enrichment efficiency, the recovery rate was not reported, raising concerns about overall yield and sample loss. DEP is appealing for its ability to selectively manipulate EVs using tunable electrical parameters, such as frequency and voltage. Nonetheless, it still faces challenges, including low throughput and the requirement for low-conductivity buffers, which may limit compatibility with biological fluids. Improving scalability and operational stability will be essential for translating DEP-based EVs systems into clinical use.
Magnetic separation involves the capture of EVs through an immunoaffinity reaction, wherein magnetic beads (MB) bind to antibodies. This process utilizes magnetic forces generated by a magnetic field to manipulate the particles effectively.71,72 Niu et al. developed the develop a fluid multivalent magnetic interface (FluidmagFace) in a microfluidic chip, a system that employs affinity magnetic beads (MB) for efficient EVs separation and proteomic analysis. Magnetic separation allows precise manipulation of MB-bound targets, improving separation efficiency by 13.9% compared to non-bivalent binding methods.43 This technique effectively and selectively captures EVs from plasma samples with high sensitivity. While magnetic separation offers excellent affinity and selectivity, its effectiveness can be affected by external interference, limiting reliability in complex biological samples. Although the technique demonstrates promising sensitivity and specificity, overcoming the impact of environmental factors remains a critical challenge for its broader clinical implementation.
Building on previous research, the proposed platform demonstrates considerable promise for clinical applications, particularly in areas such as noninvasive liquid biopsy and personalized medicine. By enabling the detection and analysis of tumor-derived EVs (T-EVs) in blood, the platform facilitates early cancer diagnosis and continuous monitoring of treatment responses.43 Moreover, it offers the potential to identify biomarkers associated with drug resistance, thereby aiding in the customization of cancer treatments. Beyond oncology, the system's ability to separate and analyze circulating microparticles (MPs) provides a valuable diagnostic tool for assessing vascular health, offering insights into conditions such as atherosclerosis and diabetes-related cardiovascular risks.73,74
From a clinical adoption perspective, while acoustofluidics requires a higher initial investment due to the specialized equipment involved, it offers substantial long-term cost savings.75 Its automated, label-free operation reduces the need for consumables and manual labor, making it more cost-efficient over time compared to techniques like ultrafiltration, which are burdened by ongoing costs for membrane replacements and maintenance. As the system scales for high-throughput applications, the per-test cost of acoustofluidics becomes more favorable, further enhancing its potential for widespread clinical use. This cost comparison underscores the economic advantages of acoustofluidics, positioning it as a promising solution for clinical adoption in both oncology and vascular health diagnostics.76 Current comparisons favor older benchmarks; emerging alternatives require direct validation. Multicenter reproducibility studies remain imperative before cost advantages can be deemed generalizable.77,78
2. Purity metrics in EVs separation strategies
A significant challenge in microfluidic-based EVs separation is the lack of standardized metrics for assessing EVs purity, which hinders the ability to directly compare the performance of different platforms. Purity is typically quantified by evaluating the presence of contaminants, such as lipoproteins and soluble proteins, which may co-isolate with EVs during the separation process.79 However, the methods used to assess purity vary widely across studies, affecting the reported purity values and their interpretability. For instance, acoustofluidic separation techniques have reported a purity of 98.4%, typically by excluding lipoproteins from the analysis.36 This step is crucial for ensuring the specificity of the isolated EVs population. However, some studies fail to fully detail the specific methods employed to exclude lipoproteins and address the potential impact of other co-isolated components.80,81 A more thorough explanation of the purity quantification process, including the methods used to remove lipoproteins and non-EVs contaminants, is essential for ensuring accurate purity assessments.
In contrast, the viscoelastic flow-based method reports a purity of greater than 97% yet does not account for the potential co-isolation of soluble proteins.40 While soluble proteins are not directly associated with EVs, their presence can influence the overall purity of the isolated EVs.82 This issue complicates the interpretation of purity measurements and highlights the need to consider all potential contaminants when evaluating EVs separation techniques.
Microfluidic technologies have demonstrated significant potential in efficiently separating EVs from complex biological fluids.83 Nonetheless, a major challenge remains in achieving high purity of the separated EVs, particularly due to the co-separation of contaminants such as lipoproteins and soluble proteins. These contaminants can compromise the integrity and specificity of the EVs population, thus undermining the reliability of subsequent analyses. Therefore, addressing these sources of contamination is essential for enhancing the performance of microfluidic EVs separation techniques. Several microfluidic approaches, including acoustofluidics and viscoelastic flow-based methods, have been developed to improve EVs separation purity.26,84 Acoustofluidics can effectively minimize lipoprotein contamination by adjusting the acoustic field parameters to preferentially capture EVs while reducing the co-separation of larger lipid particles.85 However, despite achieving high purity by excluding lipoproteins, acoustofluidic systems may still face difficulties in separating EVs from soluble proteins, which share similar size and density characteristics.86 To further mitigate this issue, incorporating additional steps such as affinity-based capture or size-exclusion filtration may be necessary to reduce soluble protein contamination.
B. Microfluidic EVs detection strategies
We reviewed microfluidic strategies for EVs separation, emphasizing that accurate downstream detection relies on EVs being separated with high purity.87 Various EVs detection techniques have been used in disease prognosis and therapeutic monitoring. Microfluidic methods such as fluorescence assays, SPR, SERS, mass spectrometry, and electrochemical sensing offer benefits in miniaturization and integration (Fig. 2). Nevertheless, they often require expensive equipment and high reagent volumes and may lack sensitivity for detecting low-abundance biomarkers.88–92 Nonetheless, rapid advances in microfluidic technology are driving its use in EV-based diagnostics. These platforms show strong potential for high-throughput, sensitive detection from small sample volumes. Future work should improve robustness, sensitivity, and clinical adaptability while addressing standardization and reproducibility to enable broader clinical use.
Fluorescence detection, known for its high precision and sensitivity, offers a rapid and efficient method for EVs detection, particularly when combined with microfluidic platforms.87 EVs are typically captured on microfluidic chips and labeled with fluorescent dyes or quantum dots.93–95 The resulting signals are analyzed via fluorescence microscopy or flow cytometry, with their presence and intensity serving as diagnostic indicators.96 Chinnappan et al. developed an aptamer-based magnetic biosensor using magnetic nanobeads functionalized with anti-CD63 aptamers. By integrating immunomagnetic separation (IMS), the system enhanced fluorescence detection of EVs, with CD63 serving as the target marker. Carbon-coated magnetic beads adsorbed FAM (6-Carboxyfluorescein)-labeled ssDNA aptamers, resulting in a strong fluorescence signal and a detection limit of 1457 particles/ml.97 Fluorescence-based methods enable rapid and sensitive EVs detection and hold promise for in situ analysis of clinical samples. However, their clinical translation is limited by nonspecific binding, high cost, and complex procedures. Additionally, the small size and heterogeneity of EVs challenge labeling consistency and quantification. Quantum dots (QDs) offer excellent photostability but pose toxicity risks due to heavy metals like cadmium and lead, limiting their clinical use.98,99 Biodegradable alternatives, such as fluorescent nanoparticles from organic dyes or biocompatible polymers, provide similar optical properties with enhanced safety profiles. These materials can also be engineered to degrade in specific biological environments, improving biocompatibility.100 Exploring these options could lead to safer, more reliable biosensing platforms for applications like EVs analysis and biomarker detection.
Surface plasmon resonance (SPR) is an advanced optical biosensing technology that exploits the phenomenon of total internal reflection of light at the interface between a prism and a metal film.101 This interaction generates a diminishing wave within a photophobic medium, alongside a plasmonic wave present in the medium.102 Luo et al. developed a portable SPR-based EVs sensing platform that integrates microfluidics with a high-performance trilayer structure: a gold mirror, SiO2 spacer, and nanoporous gold sensing layer. The SiO2 layer served as an optical cavity, and the sensor surface was functionalized with CD63 or EpCAM aptamers for EVs detection. The average bulk sensitivity values for the Si/Au hole, Au/Au hole, and CIMH sensors were 316, 391, and 431 nm/RIU, with corresponding FOM values of 1.93, 12.1, and 29.2, respectively.103 This platform achieved a limit of detection of 6 × 105 particles/ml, demonstrating its potential for ultrasensitive EVs analysis. While SPR offers advantages in label-free, real-time detection for clinical diagnostics, its widespread use is hindered by the complexity, cost, and size of current systems. To unlock its full clinical potential, miniaturization, system integration, and simplified operation are critical.
Colorimetric detection is a method for identifying exosomes through a color change in solution, leveraging the simplicity of operation and signal visualization. This approach is integrated with a microfluidic platform to enhance its efficiency and practicality.104,105 Chen et al. developed a ZnO nanowire-coated scaffold chip for colorimetric detection of EVs using TMB, achieving a detection limit of 2.2 × 107 particles/ml.105 While simple, colorimetric detection often requires complementary methods to enhance sensitivity. Vaidyanathan et al. introduced a tunable AC hydrodynamic technique that utilizes nanoshear forces to improve exosome capture and detection, with a fivefold increase in efficiency, detecting over 2.76 × 106 particles/ml.106 That said, both methods face challenges, including reaction conditions, matrix interference, and nonspecific binding. Improvements in scalability, robustness, and reproducibility are crucial for clinical application, with future efforts focusing on high-throughput, clinical-grade platforms.
Surface-enhanced Raman scattering (SERS) is a sophisticated vibrational spectroscopy technique that amplifies Raman signals through plasmon excitation on irregular metal surfaces, predominantly gold (Au) or silver (Ag).107 SERS offers highly sensitive detection of low-concentration analytes,108–110 and can be seamlessly integrated into microfluidic systems for high-throughput and precise analysis.111 To achieve highly sensitive detection of EVs, Lee et al. developed an SERS substrate with gold nanopillars that generate hotspots, enabling precise miRNA detection of EVs at detection limits over 100 times lower than other methods; the detection sensitivity range is 1 aM to 100 nM.112 This was accomplished using a locked nucleic acid probe. While SERS offers exceptional sensitivity and label-free detection, challenges persist, including unstable signal enhancement, background noise, and high equipment costs. Future efforts should focus on improving signal stability, reducing noise, optimizing cost and scalability, and developing more robust substrates with better system integration for clinical applications.
As an integral component of proteomics, mass spectrometry offers several advantages in bioassays, including high throughput, exceptional specificity, remarkable sensitivity, and cost-effectiveness.113,114 Consequently, it has been extensively employed in the analysis of EVs' proteins and amino acids. Shan et al. developed a matrix comprising gold nanoparticles (AuNPs) and cellulose nanocrystals (CNCs) for the direct analysis of full proteins in serum-derived EVs, effectively addressing ion suppression caused by protein aggregation.115 This method reduced the detection limit to 0.01 mg/ml, enhancing sensitivity, automation, and cost-efficiency, and enabling the detection of a wide range of proteins with excellent reproducibility. The specificity of this detection method reaches 83.2%. However, the high cost of AuNPs, the complexity of sample preparation, and the challenges associated with data analysis hinder their broader clinical adoption. Future advancements should focus on streamlining sample preparation, lowering costs, and simplifying data analysis to facilitate more widespread clinical application.
Electrochemical detection relies on electrochemical reactions that convert sample concentration and structure into measurable electrochemical potentials.116,117 This method is straightforward, efficient, and highly selective, allowing for the quantification of EVs through alterations in electrochemical signals. Jiang et al. developed a poly(dopamine) (PDA)-assisted aptamer-based DNA microelectrode sensor for electrochemical EVs detection. The PDA deposition enhances the surface complexity, significantly improving both sensitivity (81.9%) and precision. Utilizing electrochemical impedance spectroscopy (EIS), the sensor achieves a detection limit of 1.39 × 102 particles/ml (approximately 14 EVs) within 180 min, enabling single-particle detection.118 While this method shows great promise for early cancer diagnosis, challenges remain, particularly with electrode lifespan and the potential economic burden of frequent replacements. Future research should focus on extending the electrode lifespan, enhancing system integration, and addressing reproducibility issues to facilitate broader clinical adoption.
Meanwhile, emerging techniques, such as digital ELISA and nanopore-based EVs counting, are transforming the field of EVs detection by providing exceptional sensitivity and resolution. Digital ELISA, with its attomolar sensitivity, enables the detection of low-abundance biomarkers within complex biological samples, making it particularly advantageous for early disease diagnosis and continuous monitoring.95,119–122 In contrast, nanopore-based EVs counting offers single-particle resolution, allowing for precise enumeration of individual EVs and the detailed characterization of their heterogeneity.123 The integration of these advanced technologies with microfluidic platforms has the potential to overcome current limitations in both sensitivity and scalability. By combining the high-throughput capabilities of microfluidics with the superior sensitivity of digital ELISA and the single-particle resolution of nanopore-based counting, it becomes possible to significantly enhance the efficiency and accuracy of EVs analysis.124,125 This integration could thus facilitate more reliable diagnostic tools and enable more effective personalized medicine applications.
III. INTEGRATED MICROFLUIDIC PLATFORM FOR EVs ANALYSIS
Building upon the previous systematic review of strategies for the separation and detection of EVs, significant advancements have been made in the methodologies employed for EVs separation and analysis. The microfluidic separation platform has evolved to enhance EVs extraction, achieving both high yield and exceptional purity, while detection modalities continue to diversify. Nevertheless, many studies tend to apply separation and detection techniques independently.126,127 This fragmented approach complicates experimental protocols and significantly increases both time and costs, ultimately hindering research efficiency and limiting practical applicability.128 Recently, many researchers have shifted their focus toward integrating separation and detection systems, leading to significant advancements in both sample preparation and direct analytical evaluation. As summarized in this review, current integration strategies can be primarily categorized into two paradigms: the “combined microfluidic platform” and the “shared microfluidic platform” (Fig. 3). The combined microfluidic platform uses a serial configuration of separation, enrichment, and detection units interconnected via microchannels or pipelines, facilitating the continuous operation of both enrichment and detection processes. In contrast, the shared microfluidic platform eliminates the need for an additional detection system, enabling enrichment and detection functionalities to operate on a single platform, supporting in situ and real-time EVs analysis. Furthermore, depending on the specific requirements for subsequent EVs analysis, detection methods across various integrated platforms can be further classified into comprehensive content analysis and bioactive substance profiling, which include the assessment of proteins and nucleic acids. Table II provides a comparison of combined microfluidic platforms and shared microfluidic platforms for EVs. Evaluating long-term operational costs of the combined and shared microfluidic platforms involves factors like consumables, maintenance, and scalability. The combined platform, with its modular design, incurs higher consumable and maintenance costs due to additional reagents and frequent calibration. This can limit its feasibility in resource-limited settings. In contrast, the shared platform has lower initial consumable costs but requires careful optimization to prevent process interference. Maintaining high performance over time may lead to higher maintenance costs.
FIG. 3.
A schematic representation of combined and shared microfluidic platforms built on integrated microfluidic systems. In the combined microfluidic platform, the separation and detection regions are mechanically linked via microchannels or tubing, enabling continuous operation. EVs are first separated in the separation region and then directed downstream to the detection region. In contrast, the shared microfluidic platform lacks a distinct detection area; here, EVs are both separated and captured within the separation region, where detectors are directly applied for in situ analysis of the EVs.
TABLE II.
Comparison of combined microfluidic platforms and shared microfluidic platforms for EVs.
Type | Merits | Demerits | Clinical suitability |
---|---|---|---|
Combined microfluidic platforms | High flexibility; multi-step optimization; precise analysis | Increased complexity; higher cost; slower throughput | Suitable for detailed; high-precision analyses; less ideal for rapid, point-of-care diagnostics |
Shared microfluidic platforms | Streamlined; cost-effective; real-time analysis | Lower flexibility; limited advanced detection options | Ideal for rapid; point-of-care diagnostics and real-time monitoring |
A. Combined microfluidic platforms for EVs analysis
Following the separation process of tumor-derived EVs, the integrated system typically evaluates these EVs based on three key biophysical and biochemical characteristics: quantification (EVs count) and the analysis of bioactive molecules. A prevalent method for counting EVs involves correlating their accumulation on the chip with the quantification of fluorescence intensity.95,129 Furthermore, individual EVs can be encapsulated and separated for subsequent counting analyses. The application of microfluidics technology in the detection of individual EVs holds significant promise, particularly when integrated with advanced analytical techniques such as digital droplet technology [including digital PCR (dPCR) and digital ELISA] and nanoarray technology.95,119–122 Over the past few years, numerous scholars have dedicated themselves to the development of combined microfluidic platforms.130–132 Table III provides an overview of various combined microfluidic platforms.
TABLE III.
Comparison of combined microfluidic platforms for EVs.
Platform | Separation technique | Detection technique | Sample | Limit of detection (LOD) | Work time | References |
---|---|---|---|---|---|---|
ExoPCD chip | Y-shaped microcolumn array | Electrochemical detection | Serum | 4.39 × 103 particles/ml | 3.5 h | 133 |
Apta-magnetic biosensor | Immunomagnetic bead separation | Fluorescence detection | Cell culture supernatant | 1457 exosomes/ml | NA | 97 |
ExoELISA | Droplet-encapsulated single exosomes | Fluorescence counting | Cell culture supernatant | 10 exosomes/μl | NA | 95 |
EXID system | Serpentine microchannel | Microcolumn immunocapture assay | Blood | 10.76 exosomes/μl | 2 h | 134 |
EVLET | Vibrating membrane filtration (VMF) | Thermophoretic amplification | Plasma | 4.1 × 105 EVs | 100 min | 135 |
ExoSD chip | Immunomagnetic nanoparticles (IMNPs) separation | Fluorescence detection | Serum | NA | NA | 71 |
ExoDEP chip | Microsphere-mediated dielectrophoretic separation | Fluorescence detection | Cell line supernatant | 193 exosomes/ml | NA | 130 |
PS-ED chip | Inertial focusing | Antibody capture | Whole blood | 95 particles/μl | NA | 132 |
FEMC | Filtration | Electrochemical detection | Serum | 1 × 104 particles/ml | 1 h | 136 |
HiMEc | Inertial focusing | Electrochemical detection | Plasma | 1 × 104 to 1 × 108 EVs/ml | NA | 137 |
Nanomixing-microchip and SERS barcoding system | Nanomixing-microchip | SERS detection | Serum | NA | NA | 138 |
Microfluidic-SERS method | Filtration | SERS detection | Plasma | 2 EVs/μl | 5 h | 139 |
Nanochannel array membrane and immunocapture chip | Filtration | Immunocapture detection | Serum | NA | 1.5 h | 140 |
Indium-tin-oxide (ITO) sensor | Multi-orifice flow-fractionation (MOFF) channel | Electrochemical detection | Plasma | 15 EVs/μl | NA | 141 |
Double-filtration unit and photonic crystal (PC) | Filtration | Fluorescence detection | Serum | 8.9 × 103 EVs/ml | NA | 142 |
PCR-free integrated microfluidics platform | Acoustofluidics | Concentration/sensing detection | Plasma | 1 pM | 30 min | 24 |
Sample treatment and miRNA quantification module chip | Micromixer separation | dPCR-based miRNA quantification | Blood | 11 copies/ml | 4.3 h | 143 |
1. Combined platforms of overall EVs levels analysis
The comprehensive analysis of overall EVs levels is a crucial step in assessing the distribution and functionality of EVs in vivo. This analysis encompasses not only the absolute quantification of EVs but also their relative abundance and trends of variation across different biological samples.144–146 Commonly employed techniques include NTA, DLS, and FACS, all of which effectively provide insights into the size distribution and concentration of EVs. Microfluidic technologies have recently gained attention as complementary tools for EVs quantification, offering advantages such as low sample volume requirements, integration of multiple functions, and potential for automation.147 However, their sensitivity often varies with device design and surface chemistry, leading to inconsistent results. Furthermore, complex fabrication and operational requirements hinder standardization and reproducibility.
The ExoPCD chip developed by Xu et al. offers a promising strategy for integrated EVs separation and electrochemical detection, achieving a low sample requirement (30 μl) and a detection limit of 4.39 × 103 particles/ml for CD63-positive EVs. Its two-stage design, which combines Y-shaped microcolumns for enrichment with a downstream electrochemical sensing region, enables in situ analysis and improves workflow efficiency.133 Tim4-PS-based magnetic enrichment further enhances specificity by reducing nonspecific capture. Yet, the platform's reliance on antibody-functionalized magnetic beads and a custom chip structure may pose challenges in terms of fabrication, cost, and reproducibility. Its focus on single-marker detection also limits adaptability compared to multiplexed or label-free platforms. Future improvements should aim to reduce processing time, expand marker coverage, and simplify device architecture. Incorporating machine learning-based signal analysis could further enhance robustness and clinical applicability. Chinnappan et al. introduced an “apta magnetic biosensor” platform featuring anti-CD63 aptamers immobilized on the surface of magnetic nanobeads [Fig. 4(a)]. The described method combines flow-through magnetic separation with immunomagnetic separation (IMS) for EVs separation, using a rotating magnet assembly system (rMAS) and aptamer-based fluorescence detection.97 While the system achieves a detection limit of 1457 EVs/ml and its sensitivity is promising, there are concerns about scalability and reproducibility. Additionally, while the rMAS and microfluidic channels improve automation, the complexity and cost of the system hinder its clinical adoption. Future developments should focus on improving scalability, robustness, and specificity, while simplifying the system for broader clinical use.124,125 Liu et al. developed a droplet-based microfluidic platform, termed droplet digital ExoELISA, for ultrasensitive immunoassays and single-EV counting [Fig. 4(b)]. By encapsulating CD63 antibody–conjugated magnetic beads within droplets and labeling breast cancer-derived EVs with GPC-1, the system achieves a detection limit as low as 1 × 104 EVs/ml (∼10−17 M), demonstrating strong potential for early cancer diagnostics.95 Despite its high sensitivity, the platform faces notable challenges. The droplet generation process and magnetic bead manipulation introduce operational complexity and may affect reproducibility across laboratories. Additionally, reliance on antibody specificity can lead to cross-reactivity or inconsistent binding in heterogeneous samples. Compared to label-free or continuous-flow systems, droplet-based methods may also struggle with scalability due to precise flow control requirements. Future efforts should aim to streamline droplet handling, validate performance across diverse clinical samples, and incorporate multiplexing to broaden diagnostic applications. Lu et al. developed the EXID system, an integrated microfluidic platform combining serpentine channels and a microcolumn array for efficient EVs separation and detection [Fig. 4(c)]. Using anti-PD-L1-labeled immunomagnetic beads and quantum dot (QD)-based single-bead fluorescence analysis, the system enables multiplexed EVs profiling with a detection limit of 10.76 EVs/μl and completes analysis within 2 h.134 This approach shows strong potential for tumor subtype classification and guiding immunotherapy. Nonetheless, the device's structural complexity may hinder large-scale manufacturing and workflow integration. Additionally, QD-based detection, while enhancing signal resolution, requires careful calibration and poses potential concerns regarding photostability and biocompatibility. The system's reliance on antibody specificity may also limit its reproducibility across heterogeneous EVs populations. To sum up, without addressing these technical and translational challenges, the promising analytical capabilities of current microfluidic platforms risk remaining confined to research settings.
FIG. 4.
The combined platform was used for overall EVs content analysis. (a) An “apta magnetic biosensor” platform that utilizes anti-CD63 aptamers immobilized on the surfaces of magnetic nanobeads. Reprinted with permission from Chinnappan et al., Biosens. Bioelectron. 220, 114856 (2023). Copyright 2023 Elsevier.97 (b) The droplet digital ExoELISA microfluidic platform for single EVs counting and immunoassays. Reprinted with permission from Liu et al., Nano Lett. 18(7), 4226–4232 (2018). Copyright 2018 Authors, licensed under a Creative Commons Attribution License.95 (c) Immunomagnetic beads labeled with anti-PD-L1 fluorescent probes capture EVs, which are then separated within a serpentine channel before entering the microcolumn analysis area for single-bead analysis. Reprinted with permission from Lu et al., Biosens. Bioelectron. 204, 113879 (2022). Copyright 2022 Elsevier.134
2. Combined platforms of EVs bioactive substances analysis
The analysis of bioactive substances within EVs is a critical step in elucidating their roles in cellular communication, disease progression, and therapeutic interventions.148,149 The biomolecules transported by EVs, including proteins, nucleic acids (such as mRNA and miRNA), and glycans, participate in a myriad of physiological and pathological processes.150,151 Consequently, both quantitative and qualitative analyses of these molecules have become fundamental to understanding the functional dynamics of EVs. Currently, some research groups have utilized thermophoresis to target EVs glycans as detection objects, thereby breaking away from conventional methods of EVs detection and analysis [Fig. 5(a)].135
FIG. 5.
The combined platform was used for EVs bioactive substances analysis. (a) The lectin-based thermophoretic method (EVLET) streamlines the processes of vibrational membrane filtration (VMF) and thermophoretic amplification on a microfluidic platform for the analysis and detection of EVs glycans in cancer diagnostics. Reprinted with permission from Li et al., Nat. Commun. 15(1), 2292 (2024). Copyright 2024 Nature.135 (b) The dual-filter electrochemical microfluidic chip (FEMC) platform facilitates on-chip separation of EVs and continuous in situ electrochemical analysis of surface proteins. Reprinted with permission from Wang et al., Biosens. Bioelectron. 239, 115590 (2023). Copyright 2023 Elsevier.136 (c) An integrated platform for EVs separation and detection combining helical microfluidic channels with electrochemical devices (HiMEc). Reprinted with permission from Kwon et al., Biosens. Bioelectron. 267, 116792 (2025). Copyright 2025 Elsevier.137 (d) A microfluidic platform with integrated size filtration for SERS analysis of plasma from osteosarcoma patients. Reprinted with permission from Han et al., Biosens. Bioelectron. 217, 114709 (2022). Copyright 2022 Elsevier.139 (e) A microfluidic platform for EVs detection using integrated SAW lysis without PCR and chemical reagents. Reprinted with permission from Ramshani et al., Commun. Biol. 2(1), 1–9 (2019). Copyright 2019 Nature.24
a. EVs' protein analysis.
The in-depth investigation of EVs proteomics lays a foundational framework for the discovery of potential EVs assays, which can be employed for early disease diagnosis and therapeutic monitoring.152 By comparing the EVs' proteome of healthy individuals with those suffering from various diseases, researchers have identified characteristic proteins associated with disease progression, thereby informing the development of personalized treatment strategies.153
Wang et al. developed a dual-filter electrochemical microfluidic chip (FEMC) that integrates EVs separation and in situ detection into a streamlined workflow [Fig. 5(b)]. By combining dual filtration with multiplexed screen-printed electrodes (SPEs) functionalized with MB@UiO-66 and specific antibodies, the platform enables rapid detection of multiple tumor markers (PMSA, EGFR, CD81, and CEA) from blood samples.136 It achieves a detection limit of 1 × 104 particles/ml within 1 h, offering a 100-fold sensitivity improvement over traditional ELISA. Despite its advantages, the platform faces practical challenges. The use of multiple SPEs and pre-functionalized materials increases fabrication complexity and cost, potentially limiting scalability. Moreover, while MB@UiO-66 enhances signal output, it may introduce background noise if not properly controlled across diverse biological fluids. Although effective for protein markers, the platform's applicability to other biomolecules like RNA or lipids remains unverified, which could restrict its diagnostic versatility. Kwon et al. developed the HiMEc platform, integrating helical microfluidic channels with electrochemical sensors for efficient EVs separation and detection [Fig. 5(c)]. By combining size-based sorting with antibody-mediated lipoprotein capture, the system removes over 83% of lipoproteins and achieves an EVs recovery rate above 87%. Quantification of PD-L1 and PD-1 on EVs showed signal intensities 2.3× and 1.2× higher than those obtained via ultracentrifugation and SEC (size exclusion chromatography), respectively, with a detection range from 1 × 104 to 1 × 108 EVs/ml.137 Despite its strengths, HiMEc's structural complexity may limit scalability and standardization. Furthermore, its current design focuses on protein biomarkers, with limited validation for nucleic acid detection. While it offers enhanced purity and sensitivity compared to conventional methods, further optimization is needed to improve usability, broaden analytical targets, and ensure reproducibility across clinical samples. To enhance detection sensitivity and efficiency, integrated microfluidic platforms utilizing SERS offer notable advantages,108–110 enabling high-throughput and sensitive detection of low-abundance analytes.111 Wang et al. developed a nano-hybrid microchip integrated with SERS to detect four melanoma-related protein biomarkers directly from 5 μl of serum, without the need for extensive EVs pre-enrichment.138 The system employs a sandwich immunoassay format and achieves a signal amplification of 3.7- to 4.2-fold, enhancing both sensitivity and specificity while streamlining the workflow. Despite these advantages, SERS-based detection remains sensitive to substrate variability and hotspot reproducibility, potentially affecting consistency. Additionally, the platform's current application is limited to protein biomarkers, with unproven adaptability to other EVs contents such as miRNAs or lipids, which may constrain its broader diagnostic utility. Han et al. developed a SERS-integrated microfluidic platform with side filtration to quantify EVs biomarkers, CD63, vimentin (VIM), and EpCAM, in plasma from osteosarcoma patients [Fig. 5(d)]. The system achieved a detection limit of 2 EVs/μl using just 50 μl of plasma and completed analysis within 5 h, demonstrating strong diagnostic performance (100% sensitivity, 90% specificity, and 95% accuracy).139 These results highlight the platform's potential for accurate osteosarcoma classification and the utility of SERS in multiplexed EVs detection. Having said, that challenges remain for clinical translation. The reproducibility of Raman signal enhancement is limited by nanostructure uniformity and hotspot variability. Additionally, the high diagnostic accuracy was based on a small sample size and requires validation in broader patient populations. The use of multiple biomarkers also adds assay complexity, which may hinder standardization and scalability in clinical workflows. Gurudatt et al. developed an electrochemical microfluidic biochip that integrates a porous multi-orifice flow-fractionation (MOFF) channel with an indium-tin-oxide (ITO) sensor for rapid and sensitive detection of epithelial and mesenchymal markers on EVs, enabling evaluation of the epithelial–mesenchymal transition (EMT) index in pancreatic cancer.141 With a detection limit of 15 EVs/μl and a clinical quantification threshold of 50 EVs/μl, the platform delivers results within 5 min and effectively distinguishes IPMN (intraductal papillary mucinous neoplasm) from SCA samples, highlighting its diagnostic and commercialization potential. Yet, several limitations remain. EMT-based assessment, while biologically meaningful, can be affected by tumor heterogeneity and may not generalize across cancer types. The integration of MOFF and ITO components may complicate fabrication and hinder scalability. Moreover, the system's performance in complex clinical samples, particularly those with interfering biomolecules, requires further validation. These platforms often incorporate structurally complex components, such as dual filters or nanostructured substrates, which hinder scalability and increase production costs. Advancing clinical translation will require simplifying designs, improving manufacturability, and validating performance across diverse sample types.
b. EVs' RNA analysis.
In addition to the widespread utilization of proteins in the clinical evaluation of EVs, nucleic acids also play an indispensable role. The nucleic acids found within EVs, such as mRNA and miRNA, closely resemble the expression profiles of their originating cells, thereby providing valuable insights into the biological states and functions of those cells.131,154,155 Consequently, the transfer of nucleic acids between disparate cells via EVs' carriers is pivotal not only for intercellular communication but also significantly influences the onset and progression of various diseases.
Ramshani et al. developed a PCR- and chemical-free microfluidic platform integrating surface acoustic wave (SAW)-based lysis with an enrichment film sensor, enabling absolute quantification of EV-derived miRNAs (e.g., miR-21) in plasma or serum [Fig. 5(e)]. The system achieves a 1 pM detection limit from just 20 μl of sample within 30 min, with <10% uncertainty, offering high analytical precision and streamlined operation.24 Even though this approach represents a major advance in EVs RNA analysis due to its integration and speed, several limitations persist. The efficacy of SAW-based lysis may vary with sample composition, affecting reproducibility, and its utility across broader miRNA panels or more complex fluids remains to be fully assessed. Compared to fluorescence or thermophoretic systems, this method reduces procedural complexity and cost but may require further calibration to ensure consistent lysis across diverse EVs subtypes. Subsequently, Sung et al. developed an integrated microfluidic platform that automates EVs separation, miRNA-21 extraction, reverse transcription (RT), and digital PCR (dPCR) for ovarian cancer (OvCa) diagnostics.143 The system operates without an external pump, streamlining workflow and enhancing suitability for point-of-care use. It achieves a detection limit of 11 copies/ml with quantification inaccuracy under 12%, outperforming conventional qPCR in sensitivity and precision. Despite its analytical strength, the platform has limitations. Its reliance on dPCR demands specialized, costly equipment, which may limit clinical scalability. Additionally, the integration of multiple processing steps could introduce variability, affecting reproducibility across diverse samples. Compared to PCR-free systems like SAW-based platforms, this method offers greater accuracy but with increased complexity and resource demands. Future research should focus on miniaturizing dPCR components, reducing processing time, and validating performance across larger, heterogeneous cohorts.
As EVs become increasingly important in clinical diagnostics, particularly for biomarker detection like miRNA and mRNA, efficient RNA analysis methods are essential.156,157 Microfluidic platforms offer significant advantages over traditional plate-based assays, including improved sensitivity, scalability, and operational efficiency.158 Unlike plate-based methods, which are limited by sample volume and long processing times, microfluidics enables rapid, high-throughput analysis with minimal sample waste, crucial in clinical settings. By integrating processes such as EVs separation, RNA extraction, and detection into a single, closed system, microfluidic platforms reduce contamination risks and human error.159 Their precise control over reaction conditions enhances RNA quantification accuracy, particularly for low-abundance biomarkers. Additionally, microfluidic devices are ideal for point-of-care applications, offering faster and more practical solutions than conventional laboratory assays. Overall, microfluidics provides superior sensitivity, automation, and efficiency, making it an excellent choice for scalable EVs RNA analysis in clinical diagnostics.
B. Shared microfluidic platforms for EVs analysis
Distinct from the previously summarized integrated platforms for separation and detection, the platforms in question merge the enrichment and detection units into a singular chip structure, eschewing the need for additional detection areas.130,160 These platforms primarily incorporate finely designed microchannels for the capture of EVs, typically employing immunoaffinity capture methods on the inner surfaces of these channels, thus enhancing both the accuracy and efficiency of early clinical detection. To simultaneously achieve enrichment and detection without an independent detection area, these integrated platforms leverage the unique characteristics of various material interfaces, introducing diverse measurement technologies that facilitate separation and enrichment while enabling timely in situ detection to address varying experimental requirements.161,162 Table IV presents a summary of various shared microfluidic platforms.
TABLE IV.
Comparison of shared microfluidic platforms for EVs.
Platform | Separation technique | Detection technique | Sample | Limit of detection | Work time | References |
---|---|---|---|---|---|---|
MoSERS microchip | MoS2 nanocavities | SERS | Blood | 1.23% | 30 min | 163 |
TIRF | Anti-CD63 capture | TIRF microscope fluorescence detection | Plasma | NA | NA | 164 |
PMMA and a nanoporous gold (Au) nanocluster | AuNC membrane capture | SPR | Urine | 1000 particles/ml | NA | 165 |
nPLEX | Nanopore capture | SPR | Ascites | 3000 exosomes (670 aM) | NA | 166 |
Exodisc | Filtration | ELISA | Urine | NA | 1 h | 104 |
ACE microarray chip | Microelectrode chip capture | Fluorescence detection | Blood | NA | 90 min | 10 |
DNA cage-based thermophoretic assay | DNA cage selective recognition | Thermoelectrophoresis | Serum | 2.05 fM | NA | 167 |
DTTA | RET-based DNA tetrahedron (FDT) label EVs | Thermoelectrophoresis | Serum | 14 aM | NA | 168 |
DNA three-way junction biosensor | Multicolor DNA biosensor label EVs | Fluorescence detection | Serum | 0.116 g/ml | NA | 169 |
Immunobiochip | Immunocapture | Fluorescence detection | Serum | NA | 4 h | 170 |
1. Shared platforms of overall EVs levels analysis
To facilitate the comprehensive counting of EVs and real-time monitoring of total content on the same platform, researchers are dedicated to developing integrated analytical systems aimed at enhancing detection efficiency and ensuring data consistency.171 Such systems not only provide absolute counts of EVs but also allow for the real-time monitoring of overall EVs' content within samples, thereby offering more precise and comprehensive information to support clinical diagnostics and research. Current overall EVs analysis predominantly emphasizes the total count of patient-specific EVs, alongside the evaluation of therapeutic efficacy and strategies for disease prevention related to these individual EVs.172 However, this approach may overlook the intricate complexities of EVs' composition and their diverse functional roles in various biological contexts.173 A more refined strategy that incorporates both qualitative and quantitative assessments of EVs could yield deeper insights into their involvement in disease mechanisms and therapeutic responses, ultimately advancing personalized treatment approaches. Single EVs analysis relies on detection methods focused on individual EVs.174 Microfluidic technology is particularly well suited for this purpose due to its high efficiency, accuracy, and minimal sample requirements.123
To enhance the sensitivity and efficiency of single-EV detection, Jalali et al. developed a multi-channel microfluidic device embedded with MoSERS nanocavity microchips [Fig. 6(a)]. These chips, composed of silver, zinc oxide, and molybdenum disulfide (MoS2), enable precise EVs separation and SERS-based detection in small sample volumes (<10 μl).163 The system achieved 87% diagnostic accuracy and identified glioblastoma (GBM) mutations in blood samples from 12 patients, with sample preparation completed in just 30 min. Though the platform demonstrates strong potential for clinical diagnostics, especially in oncology, its complexity and reliance on nanomaterial fabrication may challenge scalability and routine clinical adoption. Although the MoSERS platform shows strong potential for single-EV diagnostics, particularly in GBM detection, challenges remain in reproducibility, fabrication complexity, and clinical integration. Future work should aim to improve material consistency, streamline device manufacturing, and validate performance across varied patient groups and EVs subtypes. He et al. developed a single-vesicle imaging platform using total internal reflection fluorescence (TIRF) microscopy for direct, quantitative analysis of tumor-derived EVs from as little as 1 μl of plasma [Fig. 6(b)].164 The system captures EVs via antibodies bound to activated aptamer probes, triggering a hybridization chain reaction (HCR) targeting PTK7-exon and amplifying fluorescence signals for single-EV detection. While the platform offers high spatial resolution and sensitivity, its clinical applicability is limited by the need for specialized TIRF equipment and reliance on specific gene targets, which may reduce its generalizability across cancer types. Compared to other single-EV approaches such as MoSERS or electrochemical systems, this method excels in imaging precision but falls short in throughput and scalability. Future improvements should focus on simplifying system integration, broadening biomarker compatibility, and enhancing usability in clinical settings. Yang et al. developed an integrated microfluidic device combining polymethyl methacrylate (PMMA) with nanoporous gold (Au) nanocluster films for EVs separation and in situ detection from patient urine samples [Fig. 6(c)]. EVs are first captured on the AuNC membrane via antibodies and then hybridized with gold nanorods (AuRs), forming a sandwich complex that amplifies plasmonic scattering signals. This system achieved a detection limit below 1000 particles/ml, demonstrating strong potential for noninvasive cancer diagnostics.165 Despite its sensitivity and compact integration, the device faces challenges related to the fabrication of nanostructured gold surfaces and the complexity of dual-antibody binding, which may affect reproducibility in heterogeneous fluids. Compared to TIRF-based or electrochemical methods, this plasmonic approach offers enhanced signal strength but may sacrifice scalability and operational simplicity. Future efforts should aim to simplify fabrication, improve reproducibility, and ensure compatibility with point-of-care applications, potentially through automation or AI-assisted analysis.
FIG. 6.
The shared platform was used for overall EVs content analysis. (a) The multi-channel fluid device combines embedded array nanocavity microchips (MoSERS microchips) with an SERS detection platform for analysis. Reprinted with permission from Jalali et al., ACS Nano 17(13), 12052–12071 (2023). Copyright 2023 Authors, licensed under a Creative Commons Attribution License.163 (b) A single-vesicle detection imaging platform based on total internal reflection fluorescence (TIRF) microscopy for the direct observation of tumor-derived EVs. Reprinted with permission from He et al., Anal. Chem. 91(4), 2768–2775 (2019). Copyright 2019 Authors, licensed under a Creative Commons Attribution License.164 (c) An integrated microfluidic apparatus for separation and in situ detection of polymethyl methacrylate (PMMA) and nanoporous gold (Au) nanocluster films modified with trapping antibodies was constructed. Reprinted with permission from Yang et al., Biosens. Bioelectron. 163, 112290 (2020). Copyright 2020 Elsevier.165
2. Shared platforms of overall EVs bioactive substances analysis
Recently, the advent of integrated analytical systems leveraging microfluidic platforms and nanotechnology has significantly enhanced the efficiency and accuracy of EVs' protein detection, making these systems particularly well suited for high-throughput screening of small-volume samples.175 Microfluidic EVs protein detection predominantly employs immunocapture methodologies. This approach involves the selective capture of target proteins via antibody-functionalized microchannels or microbeads, facilitating a streamlined “one-stop” operation encompassing EVs capture, separation, and protein detection.176
a. EVs' protein analysis.
To bolster the accuracy and repeatability of data derived from EVs protein analysis, Lee et al. introduced the nPLEX sensor, utilizing transmissive-based surface plasmon resonance (TSI) to enable sensitive, label-free EVs protein analysis through nanopore arrays. This approach improves accuracy and repeatability by tuning the cyclicality of nanopores and electromagnetic fields to detect distinct EVs proteins. While highly sensitive, its reliance on nanopore fine-tuning may limit scalability and application in complex biological matrices.166 In a departure from traditional assays like ELISA, Woo et al. presented the Exodisc microfluidic platform, integrating two nanofilter sets for high-sensitivity EVs protein analysis in bladder cancer urine samples [Fig. 7(a)]. With an efficient 30-min separation process, it demonstrates impressive 95% separation efficiency and a 100-fold concentration of target proteins like GAPDH and CD9.104 This platform stands out for its automation and potential in liquid biopsy applications, but its clinical adoption may face challenges related to sample variability and reproducibility in different patient populations. To separate and enrich EVs from whole blood for subsequent detection, Lewis et al. developed an AC electromotor (ACE) microarray chip device for separating EVs from small blood samples in just 20 min [Fig. 7(b)].10 This system achieved high sensitivity (99%) and decent specificity (82%) for biomarkers like glypican-1 and CD63. Nonetheless, it struggles with distinguishing EVs from other biomarkers, a limitation that may impact its clinical precision. The device exhibits limitations in its inability to effectively distinguish EVs from other high-content biomarkers, although it still facilitates a rapid workflow from a single drop of whole blood to the final detection result. Overall, these advancements show promise for liquid biopsy technologies, yet improvements in scalability, reproducibility, and specificity will be critical for broader clinical application.
FIG. 7.
The shared platform was used for EVs bioactive substances analysis. (a) Exodisc microfluidic platform for urinary EVs enrichment and separation in patients with bladder cancer. Reprinted with permission from Woo et al., ACS Nano 11(2), 1360–1370 (2017). Copyright 2017 Authors, licensed under a Creative Commons Attribution license.104 (b) The device based on an alternating current electric (ACE) microarray chip directly separates and detects EVs in the whole blood of patients with pancreatic and colon cancer. Reprinted with permission from Lewis et al., ACS Nano 12(4), 3311–3320 (2018). Copyright 2018 American Chemical Society.10 (c) A DNA cage-based thermophoretic method enables highly sensitive in situ detection of microRNA within EVs. Reprinted with permission from Zhao et al., Angew. Chem., Int. Ed. 62(24), e202303121 (2023). Copyright 2023 Wiley VCH GmbH.167 (d) The DNA Tetrahedron-Based Thermostrophy Assay platform enables in situ detection of EVs mRNA, facilitating the distinction between prostate cancer (PCa) patients and those with benign prostatic hyperplasia (BPH). Reprinted with permission from Han et al., Nano Today 38, 101203 (2021). Copyright 2021 Elsevier.168 (e) An immunobiochemical chip that selectively captures tumor-associated proteins (such as EGFR and PD-L1) using antibodies and visualizes the fluorescence intensity of magnetic beads (MB) with total internal reflection fluorescence (TIRF) microscopy. Reprinted with permission from Yang et al., ACS Appl. Mater. Interfaces 10(50), 43375–43386 (2018). Copyright 2018 Authors, licensed under a Creative Commons Attribution License.170
b. EVs' RNA analysis.
The shared microfluidic platform optimizes the operational workflow by effectively integrating the separation of EVs with the extraction and analysis of RNA, thereby significantly enhancing both the efficiency and accuracy of the analytical process. In contrast to traditional methodologies, the design of this shared integrated platform not only mitigates sample loss at each procedural step but also enhances the overall repeatability of the experiments through automated processing.
The identification of EVs' RNA mainly focuses on miRNAs and mRNA. Zhao et al. used molecular probes to detect miRNAs in EVs, advancing their role in cancer diagnostics [Fig. 7(c)].167 The integrated platform for mRNA analysis minimizes losses and reduces processing time. While this approach shows promise for early cancer diagnostics, its effectiveness depends on the specificity and sensitivity of the probes, which can vary across different miRNA targets and biological samples. Han et al. developed a DNA tetrahedron-based thermophoretic assay (DTTA) for in situ mRNA detection within EVs, achieving a detection limit of 14 aM [Fig. 7(d)].168 The platform combines FDT with thermal accumulation to differentiate between prostate cancer (PCa) and benign prostatic hyperplasia (BPH) based on PSA (prostate-specific antigen) mRNA expression in EVs. While it efficiently integrates EVs separation, FDT internalization, and signal assessment in a single microfluidic device, the reliance on FRET (förster resonance energy transfer) and thermophoretic techniques may pose challenges in scalability and reproducibility, limiting its broader clinical adoption. Wang et al. developed an integrated microdevice using a DNA three-way junction for in situ detection of multiple miRNAs in breast cancer patients. The device detects miR-21, miR-27a, and miR-375 with detection limits of 0.116, 0.125, and 0.287 μg/ml, respectively.169 By enabling simultaneous quantification of miRNAs without the need for EVs cleavage or complex RNA processing, the device offers a streamlined approach. However, its sensitivity may be limited by hybridization specificity and interference from abundant non-target RNAs in biological samples. Yang et al. developed an immunobiochip for in situ detection of EVs' miRNAs using total internal reflection fluorescence (TIRF) microscopy.170 The platform captures tumor-associated proteins (e.g., EGFR and PD-L1) and detects RNA via electrostatic interactions between a cationic lipid complex and EVs. This interaction activates molecular beacons (MB), allowing TIRF to measure RNA levels. While TIRF offers high sensitivity, its reliance on specialized equipment and challenges with complex biological matrices limit its practicality. While these platforms mark significant progress in EV-based RNA detection, their clinical application is limited by scalability, sensitivity, and methodological complexity. Future improvements should focus on simplifying the platforms, enhancing reproducibility, and optimizing performance in complex biological fluids. Additionally, integrating emerging technologies, like AI-driven data analysis, could address current analytical bottlenecks.
C. Challenges for clinical adoption
Despite the significant promise of combined and shared microfluidic platforms for EVs analysis, several challenges must be overcome to facilitate their clinical adoption. The combined microfluidic platform presents complexities in design due to its reliance on multiple interconnected units for separation, enrichment, and detection. This serial configuration can complicate system integration and increase costs, which may limit its scalability and practicality in clinical environments. Furthermore, ensuring consistent and reliable performance across all interconnected units, especially when processing diverse clinical samples, remains a substantial challenge. While the platform's flexibility allows for a broad range of analytical tasks, this versatility often comes at the cost of extended processing times, which could hinder its suitability for real-time, point-of-care applications. In contrast, the shared microfluidic platform offers distinct advantages, such as compactness and the ability to perform real-time, in situ analysis. Yet, it faces limitations in accommodating diverse sample types and handling complex workflows. The integration of both enrichment and detection steps within a single unit can compromise sensitivity, particularly when analyzing low-abundance biomarkers.160 Additionally, the need to optimize both processes simultaneously can introduce potential interference, negatively affecting the quality of detection. This highlights the trade-off between convenience and performance, which must be carefully managed to ensure clinical reliability.177
From a clinical perspective, both platforms must meet stringent requirements for accuracy, reproducibility, and user accessibility. To gain acceptance, these microfluidic systems must demonstrate sensitivity and specificity comparable to existing gold-standard methods, ensuring their reliability in real-world diagnostic settings.132 Moreover, challenges related to regulatory approvals, cost-effectiveness, and the ability to function effectively in point-of-care settings are critical considerations for their widespread adoption. Addressing these technical, logistical, and practical barriers is essential for unlocking the full potential of microfluidic technologies in clinical diagnostics.
IV. CONCLUSIONS
EVs have gained significant attention. These technologies offer notable advantages, including high automation, minimal sample volume requirements, and rapid processing speeds, making them highly promising for early diagnosis and disease monitoring. When compared to traditional methods, microfluidic platforms demonstrate superior sensitivity and lower detection limits. Nonetheless, despite substantial progress, several challenges and limitations continue to hinder their clinical translation. One of the primary barriers to widespread adoption is the issue of standardization. Currently, the design and operational conditions of microfluidic platforms often depend on specific laboratory settings, resulting in poor reproducibility across different laboratories. Furthermore, while microfluidic technologies excel in small sample volumes, scalability remains a challenge, particularly when applied to large-scale, complex biological samples. Sensitivity at low EVs concentrations also needs further improvement, which directly impacts their effectiveness in early-stage disease detection. Finally, regulatory hurdles impede the clinical application of these platforms, as the lack of standardized protocols and certification processes restricts their broader adoption. To overcome these limitations, future research should prioritize the standardization and simplification of microfluidic platforms. Developing more versatile and user-friendly chips will improve both reproducibility and scalability. Additionally, enhancing sensitivity for detecting low-concentration EVs and improving the handling of complex biological samples will be key to advancing their practical application. Additionally, leveraging emerging technologies, particularly AI-driven analytical methods, can accelerate data processing and interpretation, helping to resolve current analytical bottlenecks. By addressing these challenges, integrated microfluidic technologies have the potential to evolve into more precise and efficient tools for clinical diagnostics.
ACKNOWLEDGMENTS
This work was financially supported by the Foundation for Outstanding Young Scientist in Shandong Province (No. ZR2024YQ064), Shandong Province Science and Technology Small and Medium Enterprises Innovation Capacity Improvement Project (No. 2024TSGC008), Shandong Provincial Key Research and Development Project (Nos. 2020CXGC011304 and 2022CXGC020206), TaiShan Scholars (No. tsqn202408256), and the Major Innovation Project for the Science Education Industry Integration Pilot Project of Qilu University of Technology (Shandong Academy of Sciences) under Grant (No. 2023JBZ03).
Contributor Information
Piwu Li, Email: mailto:piwuli@qlu.edu.cn.
Xiaowen Huang, Email: mailto:huangxiaowen2013@gmail.com.
AUTHOR DECLARATIONS
Conflict of Interest
The authors have no conflicts to disclose.
Ethics Approval
Ethics approval is not required.
Author Contributions
Yang Dai: Investigation (lead); Writing – original draft (lead); Writing – review & editing (lead). Yibo Cui: Investigation (equal); Writing – original draft (equal); Writing – review & editing (equal). Jinwen Li: Investigation (equal); Writing – original draft (equal); Writing – review & editing (equal). Piwu Li: Funding acquisition (equal); Resources (equal). Xiaowen Huang: Funding acquisition (equal); Resources (equal).
DATA AVAILABILITY
The data that support the findings of this study are available from the corresponding authors upon reasonable request.
References
- 1.Murk J. L. A. N., Humbel B. M., Ziese U., Griffith J. M., Posthuma G., Slot J. W., Koster A. J., Verkleij A. J., Geuze H. J., and Kleijmeer M. J., “Endosomal compartmentalization in three dimensions: Implications for membrane fusion,” Proc. Natl. Acad. Sci. U. S. A. 100(23), 13332–13337 (2003). 10.1073/pnas.2232379100 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Zhang Y., Wu W., Pan X., Wang Y., Wu C., Lu L., Yu X.-Y., and Li Y., “Extracellular vesicles as novel therapeutic targets and diagnosis markers,” Extracell. Vesicle 1, 100017 (2022). 10.1016/j.vesic.2022.100017 [DOI] [Google Scholar]
- 3.Vrablova V., Kosutova N., Blsakova A., Bertokova A., Kasak P., Bertok T., and Tkac J., “Glycosylation in extracellular vesicles: Isolation, characterization, composition, analysis and clinical applications,” Biotechnol. Adv. 67, 108196 (2023). 10.1016/j.biotechadv.2023.108196 [DOI] [PubMed] [Google Scholar]
- 4.Gurung S., Perocheau D., Touramanidou L., and Baruteau J., “The exosome journey: From biogenesis to uptake and intracellular signalling,” Cell Commun. Signal. 19(1), 1–19 (2021). 10.1186/s12964-021-00730-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Eitan E., Suire C., Zhang S., and Mattson M. P., “Impact of lysosome status on extracellular vesicle content and release,” Ageing Res. Rev. 32, 65–74 (2016). 10.1016/j.arr.2016.05.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Jeppesen D. K., Fenix A. M., Franklin J. L., Higginbotham J. N., Zhang Q., Zimmerman L. J., Liebler D. C., Ping J., Liu Q., Evans R., Fissell W. H., Patton J. G., Rome L. H., Burnette D. T., and Coffey R. J., “Reassessment of exosome composition,” Cell 177(2), 428–445.e18 (2019). 10.1016/j.cell.2019.02.029 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Ding L., Yang X., Gao Z., Effah C. Y., Zhang X., Wu Y., and Qu L., “A holistic review of the state-of-the-art microfluidics for exosome separation: An overview of the current status, existing obstacles, and future outlook,” Small 17(29), 1–19 (2021). 10.1002/smll.202007174 [DOI] [PubMed] [Google Scholar]
- 8.Cai S., Luo B., Jiang P., Zhou X., Lan F., Yi Q., and Wu Y., “Immuno-modified superparamagnetic nanoparticles via host-guest interactions for high-purity capture and mild release of exosomes,” Nanoscale 10(29), 14280–14289 (2018). 10.1039/C8NR02871K [DOI] [PubMed] [Google Scholar]
- 9.Kalluri R. and McAndrews K. M., “The role of extracellular vesicles in cancer,” Cell 186(8), 1610–1626 (2023). 10.1016/j.cell.2023.03.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Lewis J. M., Vyas A. D., Qiu Y., Messer K. S., White R., and Heller M. J., “Integrated analysis of exosomal protein biomarkers on alternating current electrokinetic chips enables rapid detection of pancreatic cancer in patient blood,” ACS Nano 12(4), 3311–3320 (2018). 10.1021/acsnano.7b08199 [DOI] [PubMed] [Google Scholar]
- 11.Keller S., Ridinger J., Rupp A. K., Janssen J. W. G., and Altevogt P., “Body fluid derived exosomes as a novel template for clinical diagnostics,” J. Transl. Med. 9, 1–9 (2011). 10.1186/1479-5876-9-86 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Momen-Heravi F., Balaj L., Alian S., Mantel P. Y., Halleck A. E., Trachtenberg A. J., Soria C. E., Oquin S., Bonebreak C. M., Saracoglu E., Skog J., and Kuo W. P., “Current methods for the isolation of extracellular vesicles,” Biol. Chem. 394(10), 1253–1262 (2013). 10.1515/hsz-2013-0141 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Zhang S., Liu Y., Wang X., Yang L., Li H., Wang Y., Liu M., Zhao X., Xie Y., Yang Y., Zhang S., Fan Z., Dong J., Yuan Z., Ding Z., Zhang Y., and Hu L., “SARS-CoV-2 binds platelet ACE2 to enhance thrombosis in COVID-19,” J. Hematol. Oncol. 13(1), 1–22 (2020). 10.1186/s13045-020-00954-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Sokolova V., Ludwig A. K., Hornung S., Rotan O., Horn P. A., Epple M., and Giebel B., “Characterisation of exosomes derived from human cells by nanoparticle tracking analysis and scanning electron microscopy,” Colloids Surf., B 87(1), 146–150 (2011). 10.1016/j.colsurfb.2011.05.013 [DOI] [PubMed] [Google Scholar]
- 15.Sunkara V., Woo H. K., and Cho Y. K., “Emerging techniques in the isolation and characterization of extracellular vesicles and their roles in cancer diagnostics and prognostics,” Analyst 141(2), 371–381 (2016). 10.1039/C5AN01775K [DOI] [PubMed] [Google Scholar]
- 16.Wang J., Huang X., Xie J., Han Y., Huang Y., and Zhang H., “Exosomal analysis: Advances in biosensor technology,” Clin. Chim. Acta 518, 142–150 (2021). 10.1016/j.cca.2021.03.026 [DOI] [PubMed] [Google Scholar]
- 17.Yang F., Liao X., Tian Y., and Li G., “Exosome separation using microfluidic systems: Size-based, immunoaffinity-based and dynamic methodologies,” Biotechnol. J. 12(4), 1–9 (2017). 10.1002/biot.201600699 [DOI] [PubMed] [Google Scholar]
- 18.Shirejini S. Z. and Inci F., “The Yin and Yang of exosome isolation methods: Conventional practice, microfluidics, and commercial kits,” Biotechnol. Adv. 54, 107814 (2022). 10.1016/j.biotechadv.2021.107814 [DOI] [PubMed] [Google Scholar]
- 19.Hyun K. A., Gwak H., Lee J., Kwak B., and Jung H. I., “Salivary exosome and cell-free DNA for cancer detection,” Micromachines 9(7), 340 (2018). 10.3390/mi9070340 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Mun B., Kim R., Jeong H., Kang B., Kim J., Son H. Y., Lim J., Rho H. W., Lim E. K., and Haam S., “An immuno-magnetophoresis-based microfluidic chip to isolate and detect HER2-Positive cancer-derived exosomes via multiple separation,” Biosens. Bioelectron. 239, 115592 (2023). 10.1016/j.bios.2023.115592 [DOI] [PubMed] [Google Scholar]
- 21.Yang D., Zhang W., Zhang H., Zhang F., Chen L., Ma L., Larcher L. M., Chen S., Liu N., Zhao Q., Tran P. H. L., Chen C., Veedu R. N., and Wang T., “Progress, opportunity, and perspective on exosome isolation—Efforts for efficient exosome-based theranostics,” Theranostics 10(8), 3684–3707 (2020). 10.7150/thno.41580 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Beekman P., Enciso-Martinez A., Rho H. S., Pujari S. P., Lenferink A., Zuilhof H., Terstappen L. W. M. M., Otto C., and Le Gac S., “Immuno-capture of extracellular vesicles for individual multi-modal characterization using AFM, SEM and Raman spectroscopy,” Lab Chip 19(15), 2526–2536 (2019). 10.1039/C9LC00081J [DOI] [PubMed] [Google Scholar]
- 23.Liao Z., Zhang Y., Li Y., Miao Y., Gao S., Lin F., Deng Y., and Geng L., “Microfluidic chip coupled with optical biosensors for simultaneous detection of multiple analytes: A review,” Biosens. Bioelectron. 126, 697–706 (2019). 10.1016/j.bios.2018.11.032 [DOI] [PubMed] [Google Scholar]
- 24.Ramshani Z., Zhang C., Richards K., Chen L., Xu G., Stiles B. L., Hill R., Senapati S., Go D. B., and Chang H. C., “Extracellular vesicle microRNA quantification from plasma using an integrated microfluidic device,” Commun. Biol. 2(1), 1–9 (2019). 10.1038/s42003-019-0435-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Yi K., Zhang Z., Chen P., Xi X., Zhao X., Rong Y., Long F., Zhang Q., Zhang Y., Gao M., Liu W., Liu B. F., Zhu Z., and Wang F., “Tidal microfluidic chip-based isolation and transcriptomic profiling of plasma extracellular vesicles for clinical monitoring of high-risk patients with hepatocellular carcinoma-associated precursors,” Biosens. Bioelectron. 276, 117228 (2025). 10.1016/j.bios.2025.117228 [DOI] [PubMed] [Google Scholar]
- 26.Wang Y. K., Bao Y. R., Liang Y. X., Chen Y. J., Huang W. H., and Xie M., “Current progress and prospect of microfluidic-based exosome investigation,” TrAC, Trends Anal. Chem. 168, 117310 (2023). 10.1016/j.trac.2023.117310 [DOI] [Google Scholar]
- 27.Zhang T., Zhou T., Cui Q., Feng X., Feng S., Li M., Yang Y., Hosokawa Y., Tian G., Shen A. Q., and Yalikun Y., “Active microfluidic platforms for particle separation and integrated sensing applications,” ACS Sensors. 10(8), 5299–5313 (2025). 10.1021/acssensors.5c01896 [DOI] [PubMed] [Google Scholar]
- 28.Raju D., Bathini S., Badilescu S., Ghosh A., and Packirisamy M., “Microfluidic platforms for the isolation and detection of exosomes: A brief review,” Micromachines 13(5), 730 (2022). 10.3390/mi13050730 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Zhou Y., Liu H., and Chen H., “Advancement in exosome isolation and label-free detection towards clinical diagnosis,” TrAC, Trends Anal. Chem. 179, 117874 (2024). 10.1016/j.trac.2024.117874 [DOI] [Google Scholar]
- 30.Whitesides G. M., “The origins and the future of microfluidics,” Nature 442(7101), 368–373 (2006). 10.1038/nature05058 [DOI] [PubMed] [Google Scholar]
- 31.Arduini F., Cinti S., Scognamiglio V., Moscone D., and Palleschi G., “How cutting-edge technologies impact the design of electrochemical (bio)sensors for environmental analysis. A review,” Anal. Chim. Acta 959, 15–42 (2017). 10.1016/j.aca.2016.12.035 [DOI] [PubMed] [Google Scholar]
- 32.Chen J., Chen D., Xie Y., Yuan T., and Chen X., “Progress of microfluidics for biology and medicine,” Nano-Micro Lett. 5(1), 66–80 (2013). 10.1007/BF03354852 [DOI] [Google Scholar]
- 33.Witwer K. W., Buzás E. I., Bemis L. T., Bora A., Lässer C., Lötvall J., Nolte-'t Hoen E. N., Piper M. G., Sivaraman S., Skog J., Théry C., Wauben M. H., and Hochberg F., “Standardization of sample collection, isolation and analysis methods in extracellular vesicle research,” J. Extracell. Vesicles 2(1), 1–25 (2013). 10.3402/jev.v2i0.20360 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Jeppesen D. K., Hvam M. L., Primdahl-Bengtson B., Boysen A. T., Whitehead B., Dyrskjøt L., Ørntoft T. F., Howard K. A., and Ostenfeld M. S., “Comparative analysis of discrete exosome fractions obtained by differential centrifugation,” J. Extracell. Vesicles 3(1), 1–16 (2014). 10.3402/jev.v3.25011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Nam J., Yoon J., Jee H., Jang W. S., and Lim C. S., “High-throughput separation of microvesicles from whole blood components using viscoelastic fluid,” Adv. Mater. Technol. 5(12), 1–10 (2020). 10.1002/admt.202000612 [DOI] [Google Scholar]
- 36.Wu M., Ouyang Y., Wang Z., Zhang R., Huang P. H., Chen C., Li H., Li P., Quinn D., Dao M., Suresh S., Sadovsky Y., and Huang T. J., “Isolation of exosomes from whole blood by integrating acoustics and microfluidics,” Proc. Natl. Acad. Sci. U. S. A. 114(40), 10584–10589 (2017). 10.1073/pnas.1709210114 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Tayebi M., Yang D., Collins D. J., and Ai Y., “Deterministic sorting of submicrometer particles and extracellular vesicles using a combined electric and acoustic field,” Nano Lett. 21(16), 6835–6842 (2021). 10.1021/acs.nanolett.1c01827 [DOI] [PubMed] [Google Scholar]
- 38.Sancho-Albero M., Sebastián V., Sesé J., Pazo-Cid R., Mendoza G., Arruebo M., Martín-Duque P., and Santamaría J., “Isolation of exosomes from whole blood by a new microfluidic device: Proof of concept application in the diagnosis and monitoring of pancreatic cancer,” J. Nanobiotechnol. 18(1), 1–15 (2020). 10.1186/s12951-020-00701-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Tay H. M., Kharel S., Dalan R., Chen Z. J., Tan K. K., Boehm B. O., Loo S. C. J., and Hou H. W., “Rapid purification of sub-micrometer particles for enhanced drug release and microvesicles isolation,” NPG Asia Mater. 9(9), e434 (2017). 10.1038/am.2017.175 [DOI] [Google Scholar]
- 40.Meng Y., Zhang Y., Bühler M., Wang S., Asghari M., Stürchler A., Mateescu B., Weiss T., Stavrakis S., and deMello A. J., “Direct isolation of small extracellular vesicles from human blood using viscoelastic microfluidics,” Sci. Adv. 9(40), eadi5296 (2023). 10.1126/sciadv.adi5296 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Chen Y. S., Chen C., Lai C. P. K., and Lee G. B., “Isolation and digital counting of extracellular vesicles from blood via membrane-integrated microfluidics,” Sens. Actuators, B 358, 131473 (2022). 10.1016/j.snb.2022.131473 [DOI] [Google Scholar]
- 42.Ibsen S. D., Wright J., Lewis J. M., Kim S., Ko S. Y., Ong J., Manouchehri S., Vyas A., Akers J., Chen C. C., Carter B. S., Esener S. C., and Heller M. J., “Rapid isolation and detection of exosomes and associated biomarkers from plasma,” ACS Nano 11(7), 6641–6651 (2017). 10.1021/acsnano.7b00549 [DOI] [PubMed] [Google Scholar]
- 43.Niu Q., Shu Y., Chen Y., Huang Z., Yao Z., Chen X., Lin F., Feng J., Huang C., Wang H., Ding H., Yang C., and Wu L., “A fluid multivalent magnetic interface for high-performance isolation and proteomic profiling of tumor-derived extracellular vesicles,” Angew. Chem., Int. Ed. 62(21), e202215337 (2023). 10.1002/anie.202215337 [DOI] [PubMed] [Google Scholar]
- 44.Zhang J., Yan S., Yuan D., Alici G., Nguyen N. T., Ebrahimi Warkiani M., and Li W., “Fundamentals and applications of inertial microfluidics: A review,” Lab Chip 16(1), 10–34 (2016). 10.1039/C5LC01159K [DOI] [PubMed] [Google Scholar]
- 45.Sackmann E. K., Fulton A. L., and Beebe D. J., “The present and future role of microfluidics in biomedical research,” Nature 507(7491), 181–189 (2014). 10.1038/nature13118 [DOI] [PubMed] [Google Scholar]
- 46.Chen Z., Zhao L., Wei L., Huang Z., Yin P., Huang X., Shi H., Hu B., and Tian J., “River meander-inspired cross-section in 3D-printed helical microchannels for inertial focusing and enrichment,” Sens. Actuators, B 301, 127125 (2019). 10.1016/j.snb.2019.127125 [DOI] [Google Scholar]
- 47.Huang D., Man J., Jiang D., Zhao J., and Xiang N., “Inertial microfluidics: Recent advances,” Electrophoresis 41(24), 2166–2187 (2020). 10.1002/elps.202000134 [DOI] [PubMed] [Google Scholar]
- 48.Tay H. M., Leong S. Y., Xu X., Kong F., Upadya M., Dalan R., Tay C. Y., Dao M., Suresh S., and Hou H. W., “Direct isolation of circulating extracellular vesicles from blood for vascular risk profiling in type 2 diabetes mellitus,” Lab Chip 21(13), 2511–2523 (2021). 10.1039/D1LC00333J [DOI] [PubMed] [Google Scholar]
- 49.Leong S. Y., Lok W. W., Goh K. Y., Ong H. B., Tay H. M., Su C., Kong F., Upadya M., Wang W., Radnaa E., Menon R., Dao M., Dalan R., Suresh S., Lim D. W. T., and Hou H. W., “High-throughput microfluidic extraction of platelet-free plasma for microRNA and extracellular vesicle analysis,” ACS Nano 18(8), 6623–6637 (2024). 10.1021/acsnano.3c12862 [DOI] [PubMed] [Google Scholar]
- 50.Kang K., Lee S. S., Hyun K., Lee S. J., and Kim J. M., “DNA-based highly tunable particle focuser,” Nat. Commun. 4, 2567 (2013). 10.1038/ncomms3567 [DOI] [PubMed] [Google Scholar]
- 51.Lu X. and Xuan X., “Continuous microfluidic particle separation via elasto-inertial pinched flow fractionation,” Anal. Chem. 87(12), 6389–6396 (2015). 10.1021/acs.analchem.5b01432 [DOI] [PubMed] [Google Scholar]
- 52.Liu C., Guo J., Tian F., Yang N., Yan F., Ding Y., Wei J., Hu G., Nie G., and Sun J., “field-free isolation of exosomes from extracellular vesicles by microfluidic viscoelastic flows,” ACS Nano 11(7), 6968–6976 (2017). 10.1021/acsnano.7b02277 [DOI] [PubMed] [Google Scholar]
- 53.Liu C., Ding B., Xue C., Tian Y., Hu G., and Sun J., “Sheathless focusing and separation of diverse nanoparticles in viscoelastic solutions with minimized shear thinning,” Anal. Chem. 88(24), 12547–12553 (2016). 10.1021/acs.analchem.6b04564 [DOI] [PubMed] [Google Scholar]
- 54.D'Avino G., Greco F., and Maffettone P. L., “Particle migration due to viscoelasticity of the suspending liquid and its relevance in microfluidic devices,” Annu. Rev. Fluid Mech. 49, 341–360 (2017). 10.1146/annurev-fluid-010816-060150 [DOI] [Google Scholar]
- 55.Kim J. Y., Ahn S. W., Lee S. S., and Kim J. M., “Lateral migration and focusing of colloidal particles and DNA molecules under viscoelastic flow,” Lab Chip 12(16), 2807–2814 (2012). 10.1039/c2lc40147a [DOI] [PubMed] [Google Scholar]
- 56.Taylor D. D. and Shah S., “Methods of isolating extracellular vesicles impact down-stream analyses of their cargoes,” Methods 87, 3–10 (2015). 10.1016/j.ymeth.2015.02.019 [DOI] [PubMed] [Google Scholar]
- 57.Meggiolaro A., Moccia V., Brun P., Pierno M., Mistura G., Zappulli V., and Ferraro D., “Microfluidic strategies for extracellular vesicle isolation: Towards clinical applications,” Biosensors 13(1), 50 (2022). 10.3390/bios13010050 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Yu D., Li Y., Wang M., Gu J., Xu W., Cai H., Fang X., and Zhang X., “Exosomes as a new frontier of cancer liquid biopsy,” Mol. Cancer 21(1), 1–33 (2022). 10.1186/s12943-022-01509-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Kim G., Park M. C., Jang S., Han D., Kim H., Kim W., Chun H., and Kim S., “Diffusion-based separation of extracellular vesicles by nanoporous membrane chip,” Biosensors 11(9), 347 (2021). 10.3390/bios11090347 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Böing A. N., van der Pol E., Grootemaat A. E., Coumans F. A. W., Sturk A., and Nieuwland R., “Single-step isolation of extracellular vesicles by size-exclusion chromatography,” J. Extracell. Vesicles 3(1), 23430 (2014). 10.3402/jev.v3.23430 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Nordin J. Z., Bostancioglu R. B., Corso G., and El Andaloussi S., “Tangential flow filtration with or without subsequent bind-elute size exclusion chromatography for purification of extracellular vesicles,” Methods Mol. Biol. 1953, 287–299 (2019). 10.1007/978-1-4939-9145-7_18 [DOI] [PubMed] [Google Scholar]
- 62.Li Z., Liu C., Cheng Y., Li Y., Deng J., Bai L., Qin L., Mei H., Zeng M., Tian F., Zhang S., and Sun J., “Cascaded microfluidic circuits for pulsatile filtration of extracellular vesicles from whole blood for early cancer diagnosis,” Sci. Adv. 9(16), 1–15 (2023). 10.1126/sciadv.ade2819 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Li P. and Huang T. J., “Applications of acoustofluidics in bioanalytical chemistry,” Anal. Chem. 91(1), 757–767 (2019). 10.1021/acs.analchem.8b03786 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Zhang P., Bachman H., Ozcelik A., and Huang T. J., “Acoustic microfluidics,” Annu. Rev. Anal. Chem. 13, 17–43 (2020). 10.1146/annurev-anchem-090919-102205 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Collins D. J., Neild A., deMello A., Liu A. Q., and Ai Y., “The Poisson distribution and beyond: Methods for microfluidic droplet production and single cell encapsulation,” Lab Chip 15(17), 3439–3459 (2015). 10.1039/C5LC00614G [DOI] [PubMed] [Google Scholar]
- 66.Ding X., Peng Z., Lin S. C. S., Geri M., Li S., Li P., Chen Y., Dao M., Suresh S., and Huang T. J., “Cell separation using tilted-angle standing surface acoustic waves,” Proc. Natl. Acad. Sci. U. S. A. 111(36), 12992–12997 (2014). 10.1073/pnas.1413325111 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Laurell T., Petersson F., and Nilsson A., “Chip integrated strategies for acoustic separation and manipulation of cells and particles,” Chem. Soc. Rev. 36(3), 492–506 (2007). 10.1039/B601326K [DOI] [PubMed] [Google Scholar]
- 68.Lee K., Shao H., Weissleder R., and Lee H., “Acoustic purification of extracellular microvesicles,” ACS Nano 9(3), 2321–2327 (2015). 10.1021/nn506538f [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Wu M., Mao Z., Chen K., Bachman H., Chen Y., Rufo J., Ren L., Li P., Wang L., and Huang T. J., “Acoustic separation of nanoparticles in continuous flow,” Adv. Funct. Mater. 27(14), 1606039 (2017). 10.1002/adfm.201606039 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Qian C., Huang H., Chen L., Li X., Ge Z., Chen T., Yang Z., and Sun L., “Dielectrophoresis for bioparticle manipulation,” Int. J. Mol. Sci. 15(10), 18281–18309 (2014). 10.3390/ijms151018281 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Yu Z., Lin S., Xia F., Liu Y., Zhang D., Wang F., Wang Y., Li Q., Niu J., Cao C., Cui D., Sheng N., Ren J., Wang Z., and Chen D., “ExoSD chips for high-purity immunomagnetic separation and high-sensitivity detection of gastric cancer cell-derived exosomes,” Biosens. Bioelectron. 194, 113594 (2021). 10.1016/j.bios.2021.113594 [DOI] [PubMed] [Google Scholar]
- 72.Yan S., Liu Y., Nguyen N. T., and Zhang J., “Magnetophoresis-enhanced elasto-inertial migration of microparticles and cells in microfluidics,” Anal. Chem. 96(9), 3925–3932 (2024). 10.1021/acs.analchem.3c05803 [DOI] [PubMed] [Google Scholar]
- 73.Olejarz W., Sadowski K., and Radoszkiewicz K., “Extracellular vesicles in atherosclerosis: State of the art,” Int. J. Mol. Sci. 25(1), 388 (2024). 10.3390/ijms25010388 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Cassiano B. A., Silveira A. L. P. A., Kim Y. J., do Amaral J. B., da Silva Nali L. H., Bachi A. L. L., Resende L. D., Fonseca F. A. H., de Oliveira Izar M. C., Tuleta I. D., Victor J. R., Pallos D., and França C. N., “Role of circulating microparticles and cytokines in periodontitis associated with diabetes,” Front. Med. 11, 1–9 (2024). 10.3389/fmed.2024.1394300 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Zhang J., Chen C., Becker R., Rufo J., Yang S., Mai J., Zhang P., Gu Y., Wang Z., Ma Z., Xia J., Hao N., Tian Z., Wong D. T. W., Sadovsky Y., Lee L. P., and Huang T. J., “A solution to the biophysical fractionation of extracellular vesicles: Acoustic Nanoscale Separation via Wave-pillar Excitation Resonance (ANSWER),” Sci. Adv. 8(47), 1–10 (2022). 10.1126/sciadv.ade0640 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Cheng C. H., Yatsuda H., Liu S. H., Tsai W. N., Cheng T. S., Chen S. Y., Huang C. Y. F., Chang H. C., and Kondoh J., “An approach for measuring extracellular vesicle size using the attenuation-velocity change ratio of SH-SAW biosensors,” Anal. Chem. 97, 15234 (2025). 10.1021/acs.analchem.5c01881 [DOI] [PubMed] [Google Scholar]
- 77.Zhang S. P., Lata J., Chen C., Mai J., Guo F., Tian Z., Ren L., Mao Z., Huang P. H., Li P., Yang S., and Huang T. J., “Digital acoustofluidics enables contactless and programmable liquid handling,” Nat. Commun. 9(1), 1–11 (2018). 10.1038/s41467-018-05297-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Wang Y., Tao X., Tao R., Zhou J., Zhang Q., Chen D., Jin H., Dong S., Xie J., and Fu Y. Q., “Acoustofluidics along inclined surfaces based on AlN/Si Rayleigh surface acoustic waves,” Sens. Actuators, A 306, 111967 (2020). 10.1016/j.sna.2020.111967 [DOI] [Google Scholar]
- 79.Singh M., Tiwari P. K., Kashyap V., and Kumar S., “Proteomics of extracellular vesicles: Recent updates, challenges and limitations,” Proteomes 13(1), 12–17 (2025). 10.3390/proteomes13010012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Sharma M., Sheth M., Poling H. M., Kuhnell D., Langevin S. M., and Esfandiari L., “Rapid purification and multiparametric characterization of circulating small extracellular vesicles utilizing a label-free lab-on-a-chip device,” Sci. Rep. 13(1), 1–12 (2023). 10.1038/s41598-023-45409-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Tsamchoe M., Petrillo S., Lazaris A., and Metrakos P., “Isolation of extracellular vesicles from human plasma samples: The importance of controls,” Biotechnol. J. 18(6), 2200575 (2023). 10.1002/biot.202200575 [DOI] [PubMed] [Google Scholar]
- 82.Ma C., Xu Z., Hao K., Fan L., Du W., Gao Z., Wang C., Zhang Z., Li N., Li Q., Gao Q., and Yu C., “Rapid isolation method for extracellular vesicles based on Fe3O4@ZrO2,” Front. Bioeng. Biotechnol. 12, 1–10 (2024). 10.3389/fbioe.2024.1399689 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Li P., Kaslan M., Lee S. H., Yao J., and Gao Z., “Progress in exosome isolation techniques,” Theranostics 7(3), 789–804 (2017). 10.7150/thno.18133 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Tao R., Hasan S. A., Wang H. Z., Zhou J., Luo J. T., McHale G., Gibson D., Canyelles-Pericas P., Cooke M. D., Wood D., Liu Y., Wu Q., Ng W. P., Franke T., and Fu Y. Q., “Bimorph material/structure designs for high sensitivity flexible surface acoustic wave temperature sensors,” Sci. Rep. 8(1), 9052 (2018). 10.1038/s41598-018-27324-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Han J., Hu H., Lei Y., Huang Q., Fu C., Gai C., and Ning J., “Optimization analysis of particle separation parameters for a standing surface acoustic wave acoustofluidic chip,” ACS Omega 8, 311 (2023). 10.1021/acsomega.2c04273 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Li S., Ren L., Huang P. H., Yao X., Cuento R. A., McCoy J. P., Cameron C. E., Levine S. J., and Huang T. J., “Acoustofluidic transfer of inflammatory cells from human sputum samples,” Anal. Chem. 88(11), 5655–5661 (2016). 10.1021/acs.analchem.5b03383 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Lin S., Yu Z., Chen D., Wang Z., Miao J., Li Q., Zhang D., Song J., and Cui D., “Progress in microfluidics-based exosome separation and detection technologies for diagnostic applications,” Small 16(9), 1903916 (2020). 10.1002/smll.201903916 [DOI] [PubMed] [Google Scholar]
- 88.Vitale S. R., Helmijr J. A., Gerritsen M., Coban H., van Dessel L. F., Beije N., van der Vlugt-Daane M., Vigneri P., Sieuwerts A. M., Dits N., van Royen M. E., Jenster G., Sleijfer S., Lolkema M., Martens J. W. M., and Jansen M. P. H. M., “Detection of tumor-derived extracellular vesicles in plasma from patients with solid cancer,” BMC Cancer 21(1), 315 (2021). 10.1186/s12885-021-08007-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Rissin D. M., Kan C. W., Campbell T. G., Howes S. C., Fournier D. R., Song L., Piech T., Patel P. P., Chang L., Rivnak A. J., Ferrell E. P., Randall J. D., Provuncher G. K., Walt D. R., and Duffy D. C., “Single-molecule enzyme-linked immunosorbent assay detects serum proteins at subfemtomolar concentrations,” Nat. Biotechnol. 28(6), 595–599 (2010). 10.1038/nbt.1641 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Bordanaba-Florit G., Royo F., Kruglik S. G., and Falcón-Pérez J. M., “Using single-vesicle technologies to unravel the heterogeneity of extracellular vesicles,” Nat. Protoc. 16(7), 3163–3185 (2021). 10.1038/s41596-021-00551-z [DOI] [PubMed] [Google Scholar]
- 91.Hatch A. C., Fisher J. S., Tovar A. R., Hsieh A. T., Lin R., Pentoney S. L., Yang D. L., and Lee A. P., “1-Million droplet array with wide-field fluorescence imaging for digital PCR,” Lab Chip 11(22), 3838–3845 (2011). 10.1039/c1lc20561g [DOI] [PubMed] [Google Scholar]
- 92.Blicharz T. M., Siqueira W. L., Helmerhorst E. J., Oppenheim F. G., Wexler P. J., Little F. F., and Walt D. R., “Fiber-optic microsphere-based antibody array for the analysis of inflammatory cytokines in saliva,” Anal. Chem. 81(6), 2106–2114 (2009). 10.1021/ac802181j [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Li N., Jiang Y., Lv T., Li G., and Yang F., “Immunofluorescence analysis of breast cancer biomarkers using antibody-conjugated microbeads embedded in a microfluidic-based liquid biopsy chip,” Biosens. Bioelectron. 216, 114598 (2022). 10.1016/j.bios.2022.114598 [DOI] [PubMed] [Google Scholar]
- 94.Li P., Chen J., Chen Y., Song S., Huang X., Yang Y., Li Y., Tong Y., Xie Y., Li J., Li S., Wang J., Qian K., Wang C., and Du L., “Construction of exosome SORL1 detection platform based on 3D porous microfluidic chip and its application in early diagnosis of colorectal cancer,” Small 19(20), 2207381 (2023). 10.1002/smll.202207381 [DOI] [PubMed] [Google Scholar]
- 95.Liu C., Xu X., Li B., Situ B., Pan W., Hu Y., An T., Yao S., and Zheng L., “Single-exosome-counting immunoassays for cancer diagnostics,” Nano Lett. 18(7), 4226–4232 (2018). 10.1021/acs.nanolett.8b01184 [DOI] [PubMed] [Google Scholar]
- 96.Zhang P., Zhou X., He M., Shang Y., Tetlow A. L., Godwin A. K., and Zeng Y., “Ultrasensitive detection of circulating exosomes with a 3D-nanopatterned microfluidic chip,” Nat. Biomed. Eng. 3(6), 438–451 (2019). 10.1038/s41551-019-0356-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Chinnappan R., Ramadan Q., and Zourob M., “An integrated lab-on-a-chip platform for pre-concentration and detection of colorectal cancer exosomes using anti-CD63 aptamer as a recognition element,” Biosens. Bioelectron. 220, 114856 (2023). 10.1016/j.bios.2022.114856 [DOI] [PubMed] [Google Scholar]
- 98.Yan Y., Wu Y., Lu C., Wei Y., Wang J., Weng B., Huang W. Y., Zhang J. L., Yang K., and Lu K., “Electrostatic self-assembly of CdS quantum dots with Co9S8 hollow nanotubes for enhanced visible light photocatalytic H2 production,” Molecules 29(15), 3530 (2024). 10.3390/molecules29153530 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Lyu B., Bao X., Gao D., Guo X., Lu X., and Ma J., “Highly stable CsSnCl3 quantum dots grown in an ionic liquid/gelatin composite system through an in situ method,” Inorg. Chem. 61(14), 5672–5682 (2022). 10.1021/acs.inorgchem.2c00716 [DOI] [PubMed] [Google Scholar]
- 100.Sobhanan J., Rival J. V., Anas A., Sidharth Shibu E., Takano Y., and Biju V., “Luminescent quantum dots: Synthesis, optical properties, bioimaging and toxicity,” Adv. Drug Delivery Rev. 197, 114830 (2023). 10.1016/j.addr.2023.114830 [DOI] [PubMed] [Google Scholar]
- 101.Zeng S., Baillargeat D., Ho H. P., and Yong K. T., “Nanomaterials enhanced surface plasmon resonance for biological and chemical sensing applications,” Chem. Soc. Rev. 43(10), 3426–3452 (2014). 10.1039/c3cs60479a [DOI] [PubMed] [Google Scholar]
- 102.Wang C., Huang C. H., Gao Z., Shen J., He J., MacLachlan A., Ma C., Chang Y., Yang W., Cai Y., Lou Y., Dai S., Chen W., Li F., and Chen P., “Nanoplasmonic sandwich immunoassay for tumor-derived exosome detection and exosomal PD-L1 profiling,” ACS Sens. 6(9), 3308–3319 (2021). 10.1021/acssensors.1c01101 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Luo X., Yan S., Chen G., Wang Y., Zhang X., Lan J., Chen J., and Yao X., “A cavity induced mode hybridization plasmonic sensor for portable detection of exosomes,” Biosens. Bioelectron. 261, 116492 (2024). 10.1016/j.bios.2024.116492 [DOI] [PubMed] [Google Scholar]
- 104.Woo H. K., Sunkara V., Park J., Kim T. H., Han J. R., Kim C. J., Choi H. I., Kim Y. K., and Cho Y. K., “Exodisc for rapid, size-selective, and efficient isolation and analysis of nanoscale extracellular vesicles from biological samples,” ACS Nano 11(2), 1360–1370 (2017). 10.1021/acsnano.6b06131 [DOI] [PubMed] [Google Scholar]
- 105.Chen Z., Cheng S. B., Cao P., Qiu Q. F., Chen Y., Xie M., Xu Y., and Huang W. H., “Detection of exosomes by ZnO nanowires coated three-dimensional scaffold chip device,” Biosens. Bioelectron. 122, 211–216 (2018). 10.1016/j.bios.2018.09.033 [DOI] [PubMed] [Google Scholar]
- 106.Shiddiky M. J. A., Vaidyanathan R., Naghibosadat M., Rauf S., Korbie D., Carrascosa L. G., and Trau M., “Detecting exosomes specifically: A microfluidic approach based on alternating current electrohydrodynamic induced nanoshearing,” Anal. Chem. 86(22), 674–676 (2014). 10.1021/AC502082B [DOI] [PubMed] [Google Scholar]
- 107.Sharma B., Frontiera R. R., Henry A. I., Ringe E., and Van Duyne R. P., “SERS: Materials, applications, and the future,” Mater. Today 15(1–2), 16–25 (2012). 10.1016/S1369-7021(12)70017-2 [DOI] [Google Scholar]
- 108.Stremersch S., Marro M., Pinchasik B. E., Baatsen P., Hendrix A., De Smedt S. C., Loza-Alvarez P., Skirtach A. G., Raemdonck K., and Braeckmans K., “Identification of individual exosome-like vesicles by surface enhanced Raman spectroscopy,” Small 12(24), 3292–3301 (2016). 10.1002/smll.201600393 [DOI] [PubMed] [Google Scholar]
- 109.Dai Y., Bai S., Hu C., Chu K., Shen B., and Smith Z. J., “Combined morpho-chemical profiling of individual extracellular vesicles and functional nanoparticles without labels,” Anal. Chem. 92(7), 5585–5594 (2020). 10.1021/acs.analchem.0c00607 [DOI] [PubMed] [Google Scholar]
- 110.Zhang W., Jiang L., Diefenbach R. J., Campbell D. H., Walsh B. J., Packer N. H., and Wang Y., “Enabling sensitive phenotypic profiling of cancer-derived small extracellular vesicles using surface-enhanced Raman spectroscopy nanotags,” ACS Sens. 5(3), 764–771 (2020). 10.1021/acssensors.9b02377 [DOI] [PubMed] [Google Scholar]
- 111.Nie C., Shaw I., and Chen C., “Application of microfluidic technology based on surface-enhanced Raman scattering in cancer biomarker detection: A review,” J. Pharm. Anal. 13(12), 1429–1451 (2023). 10.1016/j.jpha.2023.08.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Lee J. U., Kim W. H., Lee H. S., Park K. H., and Sim S. J., “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 15(17), 1804968 (2019). 10.1002/smll.201804968 [DOI] [PubMed] [Google Scholar]
- 113.Xu L., Gimple R. C., Lau W. B., Lau B., Fei F., Shen Q., Liao X., Li Y., Wang W., He Y., Feng M., Bu H., Wang W., and Zhou S., “The present and future of the mass spectrometry-based investigation of the exosome landscape,” Mass Spectrom. Rev. 39(5–6), 745–762 (2020). 10.1002/mas.21635 [DOI] [PubMed] [Google Scholar]
- 114.Han Z., Peng C., Yi J., Zhang D., Xiang X., Peng X., Su B., Liu B., Shen Y., and Qiao L., “Highly efficient exosome purification from human plasma by tangential flow filtration based microfluidic chip,” Sens. Actuators, B 333, 129563 (2021). 10.1016/j.snb.2021.129563 [DOI] [Google Scholar]
- 115.Shan L., Qiao Y., Ma L., Zhang X., Chen C., Xu X., Li D., Qiu S., Xue X., Yu Y., Guo Y., Qian K., and Wang J., “AuNPs/CNC nanocomposite with a ‘dual dispersion’ effect for LDI-TOF MS analysis of intact proteins in NSCLC serum exosomes,” Adv. Sci. 11(12), 2307360 (2024). 10.1002/advs.202307360 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Hashkavayi A. B., Cha B. S., Lee E. S., and Park K. S., “Dual rolling circle amplification-enabled ultrasensitive multiplex detection of exosome biomarkers using electrochemical aptasensors,” Anal. Chim. Acta 1205, 339762 (2022). 10.1016/j.aca.2022.339762 [DOI] [PubMed] [Google Scholar]
- 117.Singh S., Numan A., and Cinti S., “Electrochemical nano biosensors for the detection of extracellular vesicles exosomes: From the benchtop to everywhere?,” Biosens. Bioelectron. 216, 114635 (2022). 10.1016/j.bios.2022.114635 [DOI] [PubMed] [Google Scholar]
- 118.Jiang B., Zhang T., Liu S., Sheng Y., and Hu J., “Polydopamine-assisted aptamer-carrying tetrahedral DNA microelectrode sensor for ultrasensitive electrochemical detection of exosomes,” J. Nanobiotechnol. 22(1), 55 (2024). 10.1186/s12951-024-02318-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Ko J., Wang Y., Carlson J. C. T., Marquard A., Gungabeesoon J., Charest A., Weitz D., Pittet M. J., and Weissleder R., “Single extracellular vesicle protein analysis using immuno-droplet digital polymerase chain reaction amplification,” Adv. Biosyst. 4(12), 1900307 (2020). 10.1002/adbi.201900307 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Pasini L., Notarangelo M., Vagheggini A., Burgio M. A., Crinò L., Chiadini E., Prochowski A. I., Delmonte A., Ulivi P., and D'Agostino V. G., “Unveiling mutational dynamics in non-small cell lung cancer patients by quantitative EGFR profiling in vesicular RNA,” Mol. Oncol. 15(9), 2423–2438 (2021). 10.1002/1878-0261.12976 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Ko J., Wang Y., Sheng K., Weitz D. A., and Weissleder R., “Sequencing-based protein analysis of single extracellular vesicles,” ACS Nano 15(3), 5631–5638 (2021). 10.1021/acsnano.1c00782 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Yokota S., Kuramochi H., Okubo K., Iwaya A., Tsuchiya S., and Ichiki T., “Extracellular vesicles nanoarray technology: Immobilization of individual extracellular vesicles on nanopatterned polyethylene glycol-lipid conjugate brushes,” PLoS One 14(10), e0224091 (2019). 10.1371/journal.pone.0224091 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Friedrich R., Block S., Alizadehheidari M., Heider S., Fritzsche J., Esbjörner E. K., Westerlund F., and Bally M., “A nano flow cytometer for single lipid vesicle analysis,” Lab Chip 17(5), 830–841 (2017). 10.1039/C6LC01302C [DOI] [PubMed] [Google Scholar]
- 124.Willms E., Cabañas C., Mäger I., Wood M. J. A., and Vader P., “Extracellular vesicle heterogeneity: Subpopulations, isolation techniques, and diverse functions in cancer progression,” Front. Immunol. 9, 738 (2018). 10.3389/fimmu.2018.00738 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Yang Y., Shen G., Wang H., Li H., Zhang T., Tao N., Ding X., and Yu H., “Interferometric plasmonic imaging and detection of single exosomes,” Proc. Natl. Acad. Sci. U. S. A. 115(41), 10275–10280 (2018). 10.1073/pnas.1804548115 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Li G., Tang W., and Yang F., “Cancer liquid biopsy using integrated microfluidic exosome analysis platforms,” Biotechnol. J. 15(5), 1900225 (2020). 10.1002/biot.201900225 [DOI] [PubMed] [Google Scholar]
- 127.Surappa S., Multani P., Parlatan U., Sinawang P. D., Kaifi J., Akin D., and Demirci U., “Integrated ‘lab-on-a-chip’ microfluidic systems for isolation, enrichment, and analysis of cancer biomarkers,” Lab Chip 23(13), 2942–2958 (2023). 10.1039/D2LC01076C [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Xu H. and Ye B. C., “Integrated microfluidic platforms for tumor-derived exosome analysis,” TrAC, Trends Anal. Chem. 158, 116860 (2023). 10.1016/j.trac.2022.116860 [DOI] [Google Scholar]
- 129.Logozzi M., Mizzoni D., Angelini D. F., Di Raimo R., Falchi M., Battistini L., and Fais S., “Microenvironmental pH and exosome levels interplay in human cancer cell lines of different histotypes,” Cancers 10(10), 370 (2018). 10.3390/cancers10100370 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Zhao W., Zhang L., Ye Y., Li Y., Luan X., Liu J., Cheng J., Zhao Y., Li M., and Huang C., “Microsphere mediated exosome isolation and ultra-sensitive detection on a dielectrophoresis integrated microfluidic device,” Analyst 146(19), 5962–5972 (2021). 10.1039/D1AN01061A [DOI] [PubMed] [Google Scholar]
- 131.Dwivedi M., Ghosh D., Saha A., Hasan S., Jindal D., Yadav H., Yadava A., and Dwivedi M., “Biochemistry of exosomes and their theranostic potential in human diseases,” Life Sci. 315, 121369 (2023). 10.1016/j.lfs.2023.121369 [DOI] [PubMed] [Google Scholar]
- 132.Zhou S., Hu T., Zhang F., Tang D., Li D., Cao J., Wei W., Wu Y., and Liu S., “Integrated microfluidic device for accurate extracellular vesicle quantification and protein markers analysis directly from human whole blood,” Anal. Chem. 92(1), 1574–1581 (2020). 10.1021/acs.analchem.9b04852 [DOI] [PubMed] [Google Scholar]
- 133.Xu H., Liao C., Zuo P., Liu Z., and Ye B. C., “Magnetic-based microfluidic device for on-chip isolation and detection of tumor-derived exosomes,” Anal. Chem. 90(22), 13451–13458 (2018). 10.1021/acs.analchem.8b03272 [DOI] [PubMed] [Google Scholar]
- 134.Lu Y., Ye L., Jian X., Yang D., Zhang H., Tong Z., Wu Z., Shi N., Han Y., and Mao H., “Integrated microfluidic system for isolating exosome and analyzing protein marker PD-L1,” Biosens. Bioelectron. 204, 113879 (2022). 10.1016/j.bios.2021.113879 [DOI] [PubMed] [Google Scholar]
- 135.Li Y., Zhang S., Liu C., Deng J., Tian F., Feng Q., Qin L., Bai L., Fu T., Zhang L., Wang Y., and Sun J., “Thermophoretic glycan profiling of extracellular vesicles for triple-negative breast cancer management,” Nat. Commun. 15(1), 2292 (2024). 10.1038/s41467-024-46557-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Wang Y., Gao W., Sun M., Feng B., Shen H., Zhu J., Chen X., and Yu S., “A filter-electrochemical microfluidic chip for multiple surface protein analysis of exosomes to detect and classify breast cancer,” Biosens. Bioelectron. 239, 115590 (2023). 10.1016/j.bios.2023.115590 [DOI] [PubMed] [Google Scholar]
- 137.Kwon Y. H., Park S., Jiang H., Gurudatt N. G., Lee K., Jeong H., Nie C., Shin J., Hyun K. A., and Jung H. I., “High-resolution spiral microfluidic channel integrated electrochemical device for isolation and detection of extracellular vesicles without lipoprotein contamination,” Biosens. Bioelectron. 267, 116792 (2025). 10.1016/j.bios.2024.116792 [DOI] [PubMed] [Google Scholar]
- 138.Wang J., Kao Y. C., Zhou Q., Wuethrich A., Stark M. S., Schaider H., Soyer H. P., Lin L. L., and Trau M., “An integrated microfluidic-SERS platform enables sensitive phenotyping of serum extracellular vesicles in early stage melanomas,” Adv. Funct. Mater. 32(3), 2010296 (2022). 10.1002/adfm.202010296 [DOI] [Google Scholar]
- 139.Han Z., Peng X., Yang Y., Yi J., Zhao D., Bao Q., Long S., Yu S. X., Xu X. X., Liu B., Liu Y. J., Shen Y., and Qiao L., “Integrated microfluidic-SERS for exosome biomarker profiling and osteosarcoma diagnosis,” Biosens. Bioelectron. 217, 114709 (2022). 10.1016/j.bios.2022.114709 [DOI] [PubMed] [Google Scholar]
- 140.Qiu J., Guo Q., Chu Y., Wang C., Xue H., Zhang Y., Liu H., Li G., and Han L., “Efficient EVs separation and detection by an alumina-nanochannel-array-membrane integrated microfluidic chip and an antibody barcode biochip,” Anal. Chim. Acta 1304, 342576 (2024). 10.1016/j.aca.2024.342576 [DOI] [PubMed] [Google Scholar]
- 141.Gurudatt N. G., Gwak H., Hyun K. A., Jeong S. E., Lee K., Park S., Chung M. J., Kim S. E., Jo J. H., and Jung H. I., “Electrochemical detection and analysis of tumor-derived extracellular vesicles to evaluate malignancy of pancreatic cystic neoplasm using integrated microfluidic device,” Biosens. Bioelectron. 226, 115124 (2023). 10.1016/j.bios.2023.115124 [DOI] [PubMed] [Google Scholar]
- 142.Dong X., Chi J., Zheng L., Ma B., Li Z., Wang S., Zhao C., and Liu H., “Efficient isolation and sensitive quantification of extracellular vesicles based on an integrated ExoID-Chip using photonic crystals,” Lab Chip 19(17), 2897–2904 (2019). 10.1039/C9LC00445A [DOI] [PubMed] [Google Scholar]
- 143.Sung C. Y., Huang C. C., Chen Y. S., Hsu K. F., and Lee G. B., “Isolation and quantification of extracellular vesicle-encapsulated microRNA on an integrated microfluidic platform,” Lab Chip 21(23), 4660–4671 (2021). 10.1039/D1LC00663K [DOI] [PubMed] [Google Scholar]
- 144.Tai Q., Yu H., Gao M., and Zhang X., “In situ capturing and counting device for the specific depletion and purification of cancer-derived exosomes,” Anal. Chem. 95(35), 13113–13122 (2023). 10.1021/acs.analchem.3c01670 [DOI] [PubMed] [Google Scholar]
- 145.Kowal J., Tkach M., and Théry C., “Biogenesis and secretion of exosomes,” Curr. Opin. Cell Biol. 29(1), 116–125 (2014). 10.1016/j.ceb.2014.05.004 [DOI] [PubMed] [Google Scholar]
- 146.Gurunathan S., Kang M. H., Jeyaraj M., and Kim J. H., “Platinum nanoparticles enhance exosome release in human lung epithelial adenocarcinoma cancer cells (A549): Oxidative stress and the ceramide pathway are key players,” Int. J. Nanomed. 16, 515–538 (2021). 10.2147/IJN.S291138 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Mukerjee N., Bhattacharya A., Maitra S., Kaur M., Ganesan S., Mishra S., Ashraf A., Rizwan M., Kesari K. K., Tabish T. A., and Thorat N. D., “Exosome isolation and characterization for advanced diagnostic and therapeutic applications,” Mater. Today Bio 31, 101613 (2025). 10.1016/j.mtbio.2025.101613 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Zhang Y., Tong X., Yang L., Yin R., Li Y., Zeng D., Wang X., and Deng K., “A herringbone mixer based microfluidic device HBEXO-chip for purifying tumor-derived exosomes and establishing miRNA signature in pancreatic cancer,” Sens. Actuators, B 332, 129511 (2021). 10.1016/j.snb.2021.129511 [DOI] [Google Scholar]
- 149.Kowal J., Arras G., Colombo M., Jouve M., Morath J. P., Primdal-Bengtson B., Dingli F., Loew D., Tkach M., and Théry C., “Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes,” Proc. Natl. Acad. Sci. U. S. A. 113(8), E968–E977 (2016). 10.1073/pnas.1521230113 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Mun B., Jeong H., Kim R., Gu B., Kim J., Son H. Y., Rho H. W., Lim E. K., and Haam S., “3D-Nanostructured microfluidic device arranged in a herringbone pattern for the highly effective capture of HER2-Positive cancer-derived exosomes in urine,” Chem. Eng. J. 482, 148851 (2024). 10.1016/j.cej.2024.148851 [DOI] [Google Scholar]
- 151.Niu Q., Gao J., Zhao K., Chen X., Lin X., Huang C., An Y., Xiao X., Wu Q., Cui L., Zhang P., Wu L., and Yang C., “Fluid nanoporous microinterface enables multiscale-enhanced affinity interaction for tumor-derived extracellular vesicle detection,” Proc. Natl. Acad. Sci. U. S. A. 119(44), e2213236119 (2022). 10.1073/pnas.2213236119 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Elmallah M. I. Y., Ortega-Deballon P., Hermite L., Pais-De-Barros J. P., Gobbo J., and Garrido C., “Lipidomic profiling of exosomes from colorectal cancer cells and patients reveals potential biomarkers,” Mol. Oncol. 16(14), 2710–2718 (2022). 10.1002/1878-0261.13223 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Wang L., Abdulla A., Wang A., Warden A. R., Ahmad K. Z., Xin Y., and Ding X., “Sickle-like inertial microfluidic system for online rare cell separation and tandem label-free quantitative proteomics (Orcs-Proteomics),” Anal. Chem. 94(15), 6026–6035 (2022). 10.1021/acs.analchem.2c00679 [DOI] [PubMed] [Google Scholar]
- 154.Van den Boorn J. G., Daßler J., Coch C., Schlee M., and Hartmann G., “Exosomes as nucleic acid nanocarriers,” Adv. Drug Delivery Rev. 65(3), 331–335 (2013). 10.1016/j.addr.2012.06.011 [DOI] [PubMed] [Google Scholar]
- 155.Ke W. and Afonin K. A., “Exosomes as natural delivery carriers for programmable therapeutic nucleic acid nanoparticles (NANPs),” Adv. Drug Delivery Rev. 176, 113835 (2021). 10.1016/j.addr.2021.113835 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Chisholm J., Haas-Neill S., Margetts P., and Al-Nedawi K., “Characterization of proteins, mRNAs, and miRNAs of circulating extracellular vesicles from prostate cancer patients compared to healthy subjects,” Front. Oncol. 12, 1–12 (2022). 10.3389/fonc.2022.895555 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Cheng J., Zhang K., Qu C., Peng J., and Yang L., “Non-Coding RNAs Derived from Extracellular Vesicles Promote Pre-Metastatic Niche Formation and Tumor Distant Metastasis,” Cancers (Basel). 15(7), 1–18 (2023). 10.3390/cancers15072158 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Tong Z., Yang D., Shen C., Li C., Xu X., Li Q., Wu Z., Ma H., Chen F., and Mao H., “Rapid automated extracellular vesicle isolation and miRNA preparation on a cost-effective digital microfluidic platform,” Anal. Chim. Acta 1296, 342337 (2024). 10.1016/j.aca.2024.342337 [DOI] [PubMed] [Google Scholar]
- 159.Tong Z., Xu X., Shen C., Yang D., Li Y., Li Q., Yang W., Xu F., Wu Z., Zhou L., Zhan C., and Mao H., “All-in-one multiple extracellular vesicle miRNA detection on a miniaturized digital microfluidic workstation,” Biosens. Bioelectron. 270, 116976 (2025). 10.1016/j.bios.2024.116976 [DOI] [PubMed] [Google Scholar]
- 160.Chen Y. S., Ma Y. D., Chen C., Shiesh S. C., and Lee G. B., “An integrated microfluidic system for on-chip enrichment and quantification of circulating extracellular vesicles from whole blood,” Lab Chip 19(19), 3305–3315 (2019). 10.1039/C9LC00624A [DOI] [PubMed] [Google Scholar]
- 161.Mross S., Pierrat S., Zimmermann T., and Kraft M., “Microfluidic enzymatic biosensing systems: A review,” Biosens. Bioelectron. 70, 376–391 (2015). 10.1016/j.bios.2015.03.049 [DOI] [PubMed] [Google Scholar]
- 162.Park J., Park J. S., Huang C. H., Jo A., Cook K., Wang R., Lin H. Y., Van Deun J., Li H., Min J., Wang L., Yoon G., Carter B. S., Balaj L., Choi G. S., Castro C. M., Weissleder R., and Lee H., “An integrated magneto-electrochemical device for the rapid profiling of tumour extracellular vesicles from blood plasma,” Nat. Biomed. Eng. 5(7), 678–689 (2021). 10.1038/s41551-021-00752-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Jalali M., del Real Mata C., Montermini L., Jeanne O., Hosseini I. I., Gu Z., Spinelli C., Lu Y., Tawil N., Guiot M. C., He Z., Wachsmann-Hogiu S., Zhou R., Petrecca K., Reisner W. W., Rak J., and Mahshid S., “MoS2-plasmonic nanocavities for Raman spectra of single extracellular vesicles reveal molecular progression in glioblastoma,” ACS Nano 17(13), 12052–12071 (2023). 10.1021/acsnano.2c09222 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.He D., Ho S. L., Chan H. N., Wang H., Hai L., He X., Wang K., and Li H. W., “Molecular-recognition-based DNA nanodevices for enhancing the direct visualization and quantification of single vesicles of tumor exosomes in plasma microsamples,” Anal. Chem. 91(4), 2768–2775 (2019). 10.1021/acs.analchem.8b04509 [DOI] [PubMed] [Google Scholar]
- 165.Yang Q., Cheng L., Hu L., Lou D., Zhang T., Li J., Zhu Q., and Liu F., “An integrative microfluidic device for isolation and ultrasensitive detection of lung cancer-specific exosomes from patient urine,” Biosens. Bioelectron. 163, 112290 (2020). 10.1016/j.bios.2020.112290 [DOI] [PubMed] [Google Scholar]
- 166.Im H., Shao H., Park Y. I., Peterson V. M., Castro C. M., Weissleder R., and Lee H., “Label-free detection and molecular profiling of exosomes with a nano-plasmonic sensor,” Nat. Biotechnol. 32(5), 490–495 (2014). 10.1038/nbt.2886 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Zhao S., Zhang S., Hu H., Cheng Y., Zou K., Song J., Deng J., Li L., Zhang X. B., Ke G., and Sun J., “Selective in situ analysis of mature microRNAs in extracellular vesicles using a DNA cage-based thermophoretic assay,” Angew. Chem., Int. Ed. 62(24), e202303121 (2023). 10.1002/anie.202303121 [DOI] [PubMed] [Google Scholar]
- 168.Han Z., Wan F., Deng J., Zhao J., Li Y., Yang Y., Jiang Q., Ding B., Liu C., Dai B., and Sun J., “Ultrasensitive detection of mRNA in extracellular vesicles using DNA tetrahedron-based thermophoretic assay,” Nano Today 38, 101203 (2021). 10.1016/j.nantod.2021.101203 [DOI] [Google Scholar]
- 169.Wang H., He D., Wan K., Sheng X., Cheng H., Huang J., Zhou X., He X., and Wang K., “In situ multiplex detection of serum exosomal microRNAs using an all-in-one biosensor for breast cancer diagnosis,” Analyst 145(9), 3289–3296 (2020). 10.1039/D0AN00393J [DOI] [PubMed] [Google Scholar]
- 170.Yang Y., Kannisto E., Yu G., Reid M. E., Patnaik S. K., and Wu Y., “An immuno-biochip selectively captures tumor-derived exosomes and detects exosomal RNAs for cancer diagnosis,” ACS Appl. Mater. Interfaces 10(50), 43375–43386 (2018). 10.1021/acsami.8b13971 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Shen M., Di K., He H., Xia Y., Xie H., Huang R., Liu C., Yang M., Zheng S., He N., and Li Z., “Progress in exosome associated tumor markers and their detection methods,” Mol. Biomed. 1(1), 1–25 (2020). 10.1186/s43556-020-00002-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Banijamali M., Höjer P., Nagy A., Hååg P., Gomero E. P., Stiller C., Kaminskyy V. O., Ekman S., Lewensohn R., Karlström A. E., Viktorsson K., and Ahmadian A., “Characterizing single extracellular vesicles by droplet barcode sequencing for protein analysis,” J. Extracell. Vesicles 11(11), e12277 (2022). 10.1002/jev2.12277 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Sun Z., Chen X., Niu R., Chen H., Zhu Y., Zhang C., Wang L., Mou H., Zhang H., and Luo Y., “Liposome fusogenic enzyme-free circuit enables high-fidelity determination of single exosomal RNA,” Mater. Today Bio 19, 100613 (2023). 10.1016/j.mtbio.2023.100613 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Panagopoulou M. S., Wark A. W., Birch D. J. S., and Gregory C. D., “Phenotypic analysis of extracellular vesicles: A review on the applications of fluorescence,” J. Extracell. Vesicles 9(1), 1710020 (2020). 10.1080/20013078.2019.1710020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Li K., Hong Z. P., Li Y. X., Li Y., and Yang J. L., “Clinical significance of CD151 expression in non-small cell lung cancer,” Chin. J. Cancer Prev. Treat. 21(1), 34–38 (2014). 10.47056/0365-9615-2022-174-12-756-760 [DOI] [Google Scholar]
- 176.Jeong S., Park J., Pathania D., Castro C. M., Weissleder R., and Lee H., “Integrated magneto-electrochemical sensor for exosome analysis,” ACS Nano 10(2), 1802–1809 (2016). 10.1021/acsnano.5b07584 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Movahedi S., Bahramian F., Ahmadi M., Pouyanfar N., Masoudifar R., Ghalkhani M., Hussain C. M., Keçili R., Siavashy S., and Ghorbani-Bidkorpeh F., “Next-generation microfluidics based on artificial intelligence: Applications for food sample analysis,” Microchem. J. 212, 113395 (2025). 10.1016/j.microc.2025.113395 [DOI] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
The data that support the findings of this study are available from the corresponding authors upon reasonable request.