Next Article in Journal
Quantitative ctDNA Profiling of RAS Mutations as a Prognostic Biomarker in Metastatic Colorectal Cancer
Previous Article in Journal
Donor- and Acceptor-Side Protection Against Photosystem I Photoinhibition in Arabidopsis thaliana
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Extracellular Vesicle-Derived microRNAs: Novel Non-Invasive Biomarkers for Gastrointestinal Malignancies

1
Department of Experimental Medicine, University of Rome “Sapienza”, Viale Regina Elena 324, 00161 Rome, Italy
2
Department of Life Science, Health, and Health Professions, Link Campus University, 00165 Rome, Italy
3
Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2026, 27(1), 10; https://doi.org/10.3390/ijms27010010
Submission received: 14 November 2025 / Revised: 9 December 2025 / Accepted: 16 December 2025 / Published: 19 December 2025
(This article belongs to the Section Biochemistry)

Abstract

Gastrointestinal (GI) cancers represent a heterogeneous group of malignant neoplasms arising from the digestive tract, including gastric, colorectal, hepatic, pancreatic, and biliary cancers. These tumors represent a major public health challenge due to their aggressive nature and poor prognosis. Although significant progress has been made in diagnostic imaging, endoscopy, and multimodal therapies, early detection remains difficult. Conventional serum biomarkers often lack sufficient sensitivity and specificity for reliable diagnosis, prompting a growing interest in identifying novel, minimally invasive biomarkers. In this context, liquid biopsy is emerging as a revolutionary tool in oncology. Among its components, extracellular vesicles (EVs) have gained increasing attention because they carry a wide range of molecular cargoes that reflect the biological state of their tumor of origin. In particular, EV-associated microRNAs (miRNAs) hold great promise as biomarkers for early cancer detection, real-time monitoring of disease progression, and assessment of therapeutic response. This review discusses the diagnostic and prognostic potential of EVs as novel biomarkers in GI cancers, emphasizing EV-contained miRNAs as a key resource for the development of personalized and precision medicine strategies.

1. Introduction

Gastrointestinal (GI) cancers are a group of neoplasms that arise in the GI tract and include malignant diseases of the stomach, intestines, liver, pancreas, and other parts of the digestive apparatus [1,2]. Given their highly aggressive potential and high mortality rates, corresponding to more than 35% of all cancer deaths, GI tumors represent a significant health problem [2,3,4]. For this reason, considerable efforts in recent years have focused on improving therapeutic outcomes through early diagnosis and multimodality treatment often involving a combination of surgery, chemotherapy, and radiation therapy [5]. Advances in imaging and endoscopic techniques have improved the ability to effectively diagnose and manage these tumors [6], and biomarkers are playing an increasingly important role. Glycoproteins, such as carcinoembryonic antigen (CEA), cancer antigen 19-9 (CA19-9), carbohydrate antigen 15–3 (CA 15–3), and carbohydrate antigen 12–5 (CA 12–5), are classical circulating biomarkers for GI tumors [7]. However, these biomarkers often lack the sensitivity and specificity requirements that would be needed for a reliable GI tumor diagnosis. Therefore, the identification of new sensitive and specific biomarkers is of crucial importance. In this context, the “liquid biopsy” concept has emerged as a transformative tool in the past two decades [8]. Liquid biopsy represents a key advancement in oncology, as it offers a minimally invasive and safely repeatable method to detect tumors and monitor their progression over time. Unlike traditional tissue biopsies, which require surgical procedures to obtain tumor samples, liquid biopsies consist of samples of blood or other body fluids that can be analyzed for their content of tumor-derived components. This approach enables early detection of tumors, continuous monitoring of disease progression, and dynamic assessment of therapeutic responses [8,9,10,11].
The content of a liquid biopsy mainly includes circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), extracellular vesicles (EVs), circulating cell-free RNA (cfRNA), including small RNAs/microRNAs (miRNAs), and various proteins and metabolites released from the tumor into the bloodstream [12]. The ctDNA fragments carry genetic and epigenetic signatures that reflect the mutational landscape of the tumor, and can provide information on both genetic alterations and resistance mechanisms. A similar role is played by cfRNAs [13,14]. CTCs, although rare, can be analyzed to obtain data on the tumor’s phenotypic and genotypic features. Among the tumor-derived components that can be found in liquid biopsies, EVs have attracted considerable attention in recent years. This is because their molecular cargo, derived from the cell of origin, simultaneously contains multiple biological analytes including nucleic acids, proteins, lipids and carbohydrates [13,14,15]. Therefore, the analysis of the molecules contained in liquid biopsies’ EVs can be quite helpful for the early detection of different types of cancer.
In this review, we highlight the central role of the liquid biopsy approach in monitoring the progression of GI tumors, with a focus on EVs and their cargo as emerging biomarkers with enhanced sensitivity, able to provide useful data for patient-tailored therapeutic strategies against GI tumors.

2. Extracellular Vesicles

EVs are small lipid membrane-enclosed structures that are released into the extracellular environment by almost every cell type and that can be easily detected in various body fluids, including blood, lymph, saliva, urine, cerebrospinal fluids, breast milk, tears and ascitic or pleural effusions [16,17,18].
In 1983, Pan and Johnstone were among the pioneers in discovering the characteristics of EVs during their studies of the maturation of reticulocytes into erythrocytes [19]. EVs were originally believed to be cellular debris, but later it was highlighted their important role in intercellular communication [20].
EVs can be categorized into three main types based on their biogenesis and size range: exosomes, microvesicles (MVs) and apoptotic bodies [21]. Exosomes are small membrane vesicles ranging from 30 to 150 nm in diameter, resulting from the exocytosis of intracellular multivesicular bodies [22,23]. MVs are larger membrane vesicles with diameters ranging from 150 to 1000 nm, which bud off from the plasma membrane [21,22]. It is noteworthy that every cell can simultaneously release both exosomes and MVs. Apoptotic bodies typically exhibit larger dimensions, ranging from 50 to 5000 nm, are formed during programmed cell death, and are identified by the inclusion of organelles within the vesicles [22,24].
Recently, attention has been focused on a population of cancer-derived EVs, known as “oncosomes”, that originate from the plasma membrane and present an unusually larger size (1–10 μm in diameter) compared to other EV types. Due to their atypical dimensions, oncosomes are thought to have distinctive properties in vivo, offering new potential tools for tumor profiling approaches [25].
In cancer, it is well known that EVs have a fundamental role in intercellular communication as vehicles that deliver bioactive cargo to recipient cells, thereby modulating cellular functions in both paracrine and autocrine manners [21,25,26,27].
By regulating cellular processes, such as proliferation, differentiation and apoptosis, EVs participate in maintaining tissue homeostasis, as well as in tissue repair and regeneration after injury [28,29]. Moreover, EVs contribute to angiogenesis and tissue healing by delivering pro-angiogenic factors to endothelial cells [30].
EVs are involved in immune regulation with non-univocal effects on immune cells: they can either (i) promote immune responses, as reported infectious processes, or (ii) suppress allergic, autoimmune and antitumor immune reactions, as it has been reported to occur in tolerance induction or immune evasion by cancer cells [31].
Overall, EVs represent a versatile mechanism for intercellular communication and exert significant effects on both physiological and pathological processes. Understanding their functions and mechanisms of action is a rapidly evolving area of research with implications for diagnostics, therapeutics, and regenerative medicine. Among these implications, the rapid isolation and characterization of liquid biopsy-derived EVs is becoming increasingly crucial in cancer diagnosis, prognosis, and therapy [32].
In addition, EVs have a distinctive biophysical property: exceptional stability in extracellular environments [33]. Their lipid bilayer encapsulation protects cargo from enzymatic degradation by RNases, DNases, and proteases, as well as mechanical stress, pH fluctuations, and other extracellular insults present in biological fluids. This protection preserves proteins, lipids, and nucleic acids, especially miRNAs, maintaining integrity during circulation and intercellular transfer [34,35]. Furthermore, their stability supports long-distance signaling and their reliability as liquid biopsy biomarkers [35,36].
For this reason, in the last decade, researchers have developed numerous methods for the isolation of EVs, including ultracentrifugation, size exclusion chromatography, immunoaffinity capture, polymeric precipitation and microfluidics methods [32,37]. These approaches can differentially influence EV size distribution and molecular cargo, potentially introducing methodological bias in biomarker detection and quantification. Therefore, in the context of EV isolation protocols, careful optimization, standardization, and cross-validation are essential to ensure the accuracy, reproducibility, and translational applicability of EV-miRNA profiling in clinical settings [38,39].
Importantly, many clinical trials involve cancer-derived EVs as a biomarker for cancer prediction, stage, prognosis, diagnosis and therapeutic treatment response [18,36], considering that tumor cells exhibit an increased and dysregulated release of EVs, caused by the activation of oncogenic signaling pathways or altered Rab GTPase activity (e.g., Rab27a/b) [40,41]. This determines a higher concentration of circulating EVs, which correlates with tumor burden and stage of disease.

2.1. Biogenesis of Extracellular Vesicles

EV biogenesis involves complex cellular processes and requires an accurate regulation of multiple intracellular trafficking pathways, shaping the composition of newly formed vesicles and influencing their production. As we previously mentioned, there are three main types of EVs, exosomes, MVs, and apoptotic bodies, and each has a distinct biogenesis pathway [21,22,24,28,42,43].
Even if exosomes and MVs are generated in different cellular compartments, their biogenesis involves common intracellular mechanisms and sorting machineries [42,44].
Exosomes originate from the endosomal system within the cell, starting with cargo endocytosis and subsequent formation of endocytic vesicles in specialized regions of the plasma membrane. These vesicles detach from the plasma membrane and fuse with the early endosomes, which then mature into late endosomes, also known as multivesicular bodies [28]. Late endosomes can undertake two different fates: typically, it fuses with the lysosome for degradation, but in some cases, they can fuse with the plasma membrane, releasing the intraluminal vesicles (ILVs), which are formed by endosomal membrane invagination in the organelle lumen, into the extracellular space as exosomes [17,21,28,45].
In contrast to exosomes’ biogenesis, MVs and apoptotic bodies originate by budding from the plasma membrane. MVs formation arises from the dynamic interplay between the redistribution of membrane phospholipids and the rearrangement of cytoskeletal proteins. The budding process involves the contraction of cytoskeletal structures through actin-myosin interactions, leading to the release of MVs into the extracellular space [22,42,43]. Apoptotic bodies are produced during programmed cell death (apoptosis). During apoptosis, cells undergo multiple structural changes, including DNA fragmentation, chromatin condensation, and membrane blebbing. These membrane-bound vesicles contain cellular organelles and fragmented DNA and are subsequently engulfed and cleared by phagocytic cells [24,42,43].
The biogenesis of EVs is regulated by various cellular processes and signaling pathways, including those involved in membrane trafficking, cytoskeletal dynamics, and cellular stress responses [42,43]. Understanding the mechanisms underlying EV biogenesis is crucial for elucidating their roles in physiological and pathological processes, including intercellular communication, immune regulation, and disease progression [42,43].

2.2. Uptake of Extracellular Vesicles

The growing interest in EVs is linked to their potential to induce changes in the physiology of recipient cells. The way EVs are captured by the recipient cells varies depending on the EV surface components and size [46].
There are several mechanisms by which EVs can communicate with recipient cells. Specific interactions between EV surface molecules (such as ligands) and receptors on recipient cells can facilitate EV uptake and activate downstream signal transduction pathways. For example, cellular integrins interact with ICAMs expressed on the surface of the exosome, or with integrins present on the exosome, through components of the extracellular matrix [47].
EVs can be internalized by recipient cells through various endocytic pathways such as clathrin-dependent or independent endocytosis, caveolin-mediated endocytosis, or micropinocytosis. In more detail, EVs can fuse directly with the plasma membrane of recipient cells by interacting with specific proteins and lipids and reach the lysosome, serving as a reservoir of metabolic materials; alternatively, they can avoid the fusion with the late endosome and release their content into the cytoplasm of the recipient cell, thereby influencing various cellular processes [48]. The docking between EVs and the target cell is facilitated by interactions involving tetraspanins, integrins, and other adhesion molecules present on both the EV surface and recipient cell membrane [46,49].

2.3. Cargo Selection

EVs can contain various molecules, including lipids, proteins, and nucleic acids (including DNA and the entire repertoire of RNAs). This content can vary depending on various factors such as their biogenesis, cell of origin, and the physiological or pathological conditions under which they are formed [50,51].
Since the specific cargo loaded into EVs can vary significantly from one EV to another and between EVs derived from different cell types, researchers have conducted extensive studies aimed at characterizing EVs’ contents and understanding the complexities of cargo selection mechanisms [50]. Moreover, understanding the mechanisms behind cargo sorting in EVs is crucial for devising strategies to utilize EVs for diagnostic and therapeutic applications.
EV cargo selection is a complex, dynamic, and highly regulated process that involves various mechanisms to selectively package specific biomolecules. The physiological state of cells can influence EV cargo selection. For example, cells undergoing apoptosis or cellular stress may release EVs containing specific biomolecules associated with these processes. Similarly, cell type, developmental stage, and changes in the microenvironment can also influence EV cargo composition [52].
During exosomes biogenesis, the Endosomal Sorting Complex Required for Transport (ESCRT) machinery allows, in fact, specific proteins and nucleic acids to be selectively incorporated into ILVs, contributing to the cargo of exosomes [53]. Furthermore, tetraspanins (such as CD9, CD63, and CD81) and lipid rafts, which are cholesterol-rich microdomains in the plasma membrane, play a crucial role in EVs biogenesis and cargo sorting because they can interact with specific cargo molecules and facilitate their packaging into EVs [54].
The selective packaging of RNAs into EVs is regulated by specific sorting machineries that involve RNA-binding proteins (RBPs) and their associated partners. RBPs, like heterogeneous nuclear ribonucleoproteins (hnRNPs) and members of the Argonaute (Ago) protein family, can bind to specific RNA sequences, target them to the sites of EV generation and protect them from degradation [55].

2.4. EVs as microRNA Carrier

The therapeutic potential of EVs is attracting increasing interest. Indeed, EVs could be transformed into sophisticated carriers able to deliver therapeutic proteins, RNA molecules, and drugs into specific cells [42]. The ability of these natural vectors to perform cellular targeting, to easily penetrate cell membranes and cell organelles, their special physico-chemical characteristics, and their ability to circulate without being detected by the immune system are very attractive features [56,57].
In recent years, particular attention has been paid to miRNAs, short endogenous single-stranded noncoding RNA sequences of about 17–23 nucleotides that can regulate gene expression by binding to target messenger RNAs (mRNAs), leading to their degradation or post-transcriptional changes [58].
Conventionally, there are standardized guidelines for the identification and annotation of miRNAs in order to reliably distinguish miRNAs from other RNAs, such as small interfering RNAs [59].
It has been shown that all types of EVs can transport miRNAs, which modulate gene expression in target cells in both normal and pathological conditions. Although the mechanisms by which miRNAs are sorted into EVs or retained within the cell of origin are still largely unknown [57], it has been demonstrated that miRNAs contain sorting sequences, EXOmotifs and CELLmotifs, that determine their secretion into EVs or their retention, respectively [60]. This selective loading of miRNAs into EVs is mediated by RBP, such as hnRNPA2B1, Ago2 and YBX1, which recognize specific sequence motifs in the miRNAs, guiding their inclusion into exosomes and ensuring that the EV miRNA signature reflects the physiological or pathological state of the producing cell, including malignant transformation [61,62,63]. Furthermore, different cell types preferentially use specific sorting sequences, so that the EVs’ miRNA profiles could reveal both the originating cell type and the cell’s functional status [60].
Moreover, EVs serve as carriers that protect miRNAs from degradation by extracellular RNases. This protection ensures the stability and integrity of miRNAs during their transfer between cells and through various biological fluids [60,64,65], offering a clear mechanistic advantage compared to free miRNAs, giving EV-miRNAs superior stability in circulation and making them highly suitable for detection in liquid biopsies [66].
The transfer of miRNAs to recipient cells via EVs can have multiple functional implications. For example, the EV-mediated transfer of tumor-suppressive miRNAs from normal cells to cancer cells can inhibit tumor growth and metastasis. Conversely, cancer-derived EVs can transfer oncogenic miRNAs to neighboring cells, promoting tumor progression and metastasis [67,68]. In GI cancers, EV-derived miRNAs have been reported to regulate critical processes such as epithelial–mesenchymal transition (EMT), angiogenesis, chemoresistance, tumor-associated inflammation and the formation of the pre-metastatic niche [69]. Since EV release increases with tumor aggressiveness and metabolic stress, the abundance of specific EV-miRNAs frequently correlates with tumor stage, lymph node involvement, metastasis and prognosis [61,62].
However, the EV isolation protocol previously mentioned can differentially influence EV size distribution and miRNA cargo, potentially introducing methodological bias in biomarker detection and quantification. Therefore, in the context of EV isolation protocols, careful optimization, standardization, and cross-validation are essential to ensure the accuracy, reproducibility, and translational applicability of EV-miRNA profiling in a clinical perspective [38,39].

3. Role of EVs as Biomarkers in Gastrointestinal Cancer Progression

Recent studies have highlighted the pivotal role of EVs in the development and progression of solid tumors, due to their important role as well in communication between tumors and the microenvironment. In fact, tumor cells can modify the behavior of both nearby and distant normal cells by transferring the cargo of EVs, which in turn supports tumor growth and metastasis [15,70,71,72].
In GI tumors, EVs can contribute to tumor growth, immune evasion, and metastasis by modulating the behavior of surrounding cells and creating a microenvironment favorable to cancer progression [15]. Furthermore, EVs have emerged as promising tools for both early detection and targeted therapy, opening new avenues for improving the diagnosis and treatment of GI cancers [15,73,74].
The detection of mutated DNA within the EVs’ cargo may indicate the presence of specific cancer types. For example, KRAS gene mutations in EVs serve as a potential biomarker for pancreatic or colon cancers [75,76]. However, the overall amount of DNA in EVs released from cancer cells is relatively low, and some mutations can also be observed in EVs derived from healthy subjects. Therefore, the sensitivity and specificity of such mutated DNA may not be sufficient for clinical applications.
As well, noncoding RNAs, including miRNA, circular RNA (circRNA), long non-coding RNA (lncRNA) and Piwi-interacting RNA (piRNA), are enriched in EVs isolated from cancer patients, resulting in a high impact on disease progression, through the promotion of neoangiogenesis and metastasis formation [70,77]. Given that their high levels in EVs allow an easy detection and effective amplification by PCR, they represent more reliable diagnostic candidates [4,74,78]. Additionally, EVs can carry tumor-specific proteins, both encapsulated within their lumen and expressed on their surface, providing further opportunities for early cancer detection [79].
It has been thoroughly investigated that compared to free circulating and tissue miRNAs, those from EVs are preferable biomarkers due to their stability against degradation and enhanced specificity for the source cell [80,81]. As a matter of fact, tissue miRNAs directly reflect specific tissues but require invasive collection methods, whereas circulating miRNAs tend to be unstable and are often diluted by miRNAs from other cell types, such as platelets and red blood cells. In contrast, EV-derived miRNAs represent a non-invasive choice that preserves both stability and specificity, positioning them as a more promising diagnostic and prognostic biomarker [80,81].
EVs derived from GI tumor cells (GI-EVs) promote angiogenesis by delivering pro-angiogenic factors such as vascular endothelial growth factor (VEGF) and miRNAs that modulate endothelial cell behavior. Additionally, GI-EVs contribute to immune evasion through the transfer of immunosuppressive molecules like PD-L1 and TGF-β and, yet, may confer drug resistance by polarizing macrophages [4,82,83].
A crucial aspect of GI-EVs’ function is their role in metastasis. Tumor-derived EVs can prime distant organs for metastasis by conditioning the pre-metastatic niche and altering the local microenvironment to support the survival of metastatic cancer cells and their colonization at the site [84,85]. For example, pancreatic cancer-derived EVs have been found to carry integrins that direct them to specific organs, where they interact with resident cells to remodel the extracellular matrix and promote the formation of a metastatic niche [86]. Specific miRNA signatures in EVs have been associated with colorectal cancer (CRC) stages and therapeutic outcomes [87].
In summary, EVs are central to the pathophysiology of GI tumors, influencing angiogenesis, immune evasion, metastasis, and offering promise as diagnostic and therapeutic tools. Understanding their complex roles and mechanisms may unlock new possibilities for fighting GI cancers.
Since traditional diagnostic methods, such as endoscopy and biopsy, are invasive and often detect cancer at advanced stages, the use of EVs as diagnostic biomarkers is a rapidly evolving field and offers a non-invasive alternative.

3.1. Gastric Cancer

Gastric cancer (GC) originates from the stomach lining. It is the fifth most common cancer in the world and the third leading cause of cancer death [88]. Despite advances in medical treatments and even though nowadays there are a variety of treatment strategies for GC, there has been no significant improvement in the outcome of patients with advanced-stage disease, underscoring the need for further research and more effective screening methods [89,90].
miRNA-containing EVs play a key role in GC progression by promoting cell proliferation through the activation of PI3K/Akt and mitogen-activated protein kinase (MAPK) signaling pathways [91,92]. In fact, they are released by GC cells and significantly influence tumor behavior and the tumor microenvironment (TME) by promoting tumor growth, metastasis, and resistance to chemotherapy through the silencing of tumor suppressor genes and the activation of oncogenic pathways in recipient cells [93]. Specifically, miR-149-5p affects the expression level of AKT1 in several GC cell lines, thus promoting tumor growth [94]. It has also been shown that GC cell lines resistant to paclitaxel, one of the main chemotherapeutic agents used in GC, transfer miR-155p through EVs to sensitive cells, thereby inducing chemoresistance in the latter [95]. Other studies conducted on GC cell cultures underline the role of EV, through the upregulation of miR23a, in promoting angiogenesis via the repression of PTEN [96]. EVs also play an important role in the interactions between GC cells and immune cells within the TME. miR-519a-3p has a critical role in mediating the crosstalk between primary GC cells and intrahepatic macrophages through MAPK/ERK pathways stimulation, thus promoting angiogenesis and metastasis, and represents a potential therapeutic target for GC [97]. Likewise, exosomal miR-151-3p causes M2-like macrophage polarization, favoring tumor progression [98]. In turn, miR-21 contained in EVs from M2-polarized macrophages can promote chemoresistance by supporting cancer cell survival and proliferation [83,99].
Several studies report the pivotal role of miRNAs contained in EVs as diagnostic and prognostic biomarkers. miR-19b and miR-106a are associated with GC progression and metastasis. In more detail, studies conducted on liquid biopsies of GC patients revealed that exosome-derived miR-106a directly interacts with Smad7, inducing peritoneal metastasis in GC, while miR-19b, isolated from the serum of GC patients, exhibits an oncogenic role and is able to discriminate between healthy controls and GC patients. Accordingly, these miRNAs have promising characteristics as potential biomarkers [100,101]. EV-derived miRNAs that could be used as biomarkers of prognosis or drug resistance include the exosome-derived miR-451, which induces Th17 differentiation by increasing mammalian target of rapamycin (mTOR) activity, and miR-493, whose expression correlates with paclitaxel resistance in human GC cell lines [102,103].
The principal EV-derived miRNAs used as biomarkers in GC are listed in Table 1.

3.2. Colorectal Cancer

CRC is one of the most common cancers worldwide, for which treatment options usually include a combination of surgery, chemotherapy, radiation, and targeted therapies, depending on the stage and specific location of the tumor [3]. Even in CRC, EVs play a crucial role in tumor progression and TME modulation, and can serve as biomarkers for diagnosis, prognosis, and therapeutic response. For instance, EVs’ release results in increased tumor invasiveness by transferring tumor promoters such as mutant KRAS, Epidermal Growth Factor Receptor (EGFR) and integrins to tumor cells with wild-type KRAS [108].
Similarly, miRNAs contained in EVs have a key role, with some promoting tumor growth, invasion, and metastasis by targeting tumor suppressor genes or modulating signaling pathways [109]. For example, EVs carrying miR-19a can promote CRC progression by suppressing the expression of TIA1, a tumor suppressor gene [109]. Conversely, EVs containing miR-145 can inhibit CRC progression and metastatic spread by targeting genes involved in cell proliferation and migration [110,111]. Exosome-derived miR-183-5p, highly expressed in CRC cells, induces cell proliferation and migration by targeting the transcription factor forkhead box O1 (FOXO1) [112]. EV-derived miR-92a can enhance angiogenesis by targeting and downregulating integrin α5 [113,114,115]. Exosome-derived miRNAs, such as miR-106b, miR-146a and miR-155-5p, are able to trigger the EMT and promote metastasis through different mechanisms, including macrophage polarization, activation of the PI3Kγ/Akt/mTOR pathway, or targeting of the suppressor of cytokine signaling 1 (SOCS1) [116,117]. Other exosomal miRNAs involved in tumor progression and metastasis induction are miR-203a and miR-934, which target PTEN and induce macrophage polarization [118,119]. Furthermore, it has been reported that EVs derived from cancer-associated fibroblasts (CAFs) transport lncRNA19 to CRC cells, inducing stemness properties and drug resistance through the activation of Wnt and β-catenin pathways [120]. Finally, some miRNAs can influence the immune response by modulating the activity of immune cells. For example, miR-21 contained in EVs can suppress the immune response, promoting tumor immune evasion [121].
EV-derived miRNAs can also be used as biomarkers of CRC progression through the analysis of liquid biopsies; in fact, they can serve as diagnostic biomarkers, indicating the presence of CRC in the early stages, and provide prognostic information. For example, miR-21 and miR-23a can be detected in plasma-derived EVs from CRC patients, showing high sensitivity and specificity [114]. Indeed, miR-21 plasma levels could be used as a diagnostic biomarker since it is differentially expressed between CRC patients and healthy donors, likewise miR-23a and miR-1246 [114,122].
Furthermore, miR-21 carried by EVs has been identified as a potential therapeutic biomarker, as it contributes to 5-fluorouracil resistance by targeting the PTEN/PI3K/AKT pathway, which controls genes involved in drug metabolism, ultimately promoting chemoresistance [123,124].
MiR-92a could also represent a promising biomarker, since it has been demonstrated that its expression is high in the serum of stage I and stage II CRC patients [114,125]. Elevated levels of some miRNAs, such as miR-200c, are associated with poor prognosis and advanced stages of the disease [114]. High levels of serum EVs containing miR-181a-5p have been positively correlated with liver metastasis in CRC patients [126].
In liquid biopsies of CRC patients, EV-derived miR-320c expression is linked to lower overall survival and metastatic niche formation, so that it can be used as a biomarker for therapeutic monitoring [127].
Table 2 lists some of the principal EV-derived miRNAs investigated as biomarkers in CRC.

3.3. Pancreatic Cancer

Pancreatic cancer (PC) is a highly aggressive and often fatal disease [132]. Due to its nonspecific early symptoms, such as abdominal pain and weight loss, PC is frequently diagnosed at an advanced stage, making effective treatment challenging. Despite advances in medical research, the prognosis remains poor, with a five-year survival rate of around 10%, underscoring the critical need for early detection and improved therapeutic strategies [133]. Pancreatic ductal adenocarcinoma (PDAC) is the most common PC variant, characterized by a very low survival rate due to the high incidence of metastasis and late diagnosis [134].
In vitro studies have shown that EVs play an important role in PDAC chemoresistance. For instance, EVs released from gemcitabine-exposed CAFs increase the proliferation and survival of chemoresistant PDAC cell lines through the regulation of the transcription factors SNAIL and miR-146a [135,136]. Exosomal miR-106b, released from CAFs after TP53INP1-targeted treatment, also induces gemcitabine resistance [137]. In addition, EVs from gemcitabine-resistant PDAC stem cells can transfer drug resistance to gemcitabine-sensitive PDAC cells by carrying miR-210, which targets the mTOR signaling pathway [73].
Tumor progression toward an invasive phenotype through the EMT is also promoted by PDAC-derived EVs. In this regard, it has been reported that tumor EVs containing tenascin C induce Wnt/β-catenin signaling, EMT, and tumor progression [86,138].
miR-34a, which typically acts as a tumor suppressor, is downregulated in PC [139]. It can inhibit cancer cell proliferation, migration, and invasion by targeting several genes involved in EMT. Loss of miR-34a contributes to uncontrolled tumor growth and metastatic potential [139,140]. The delivery of a miR-34a coated with exosomes (exomiR-34a) to PC cells has been shown to significantly inhibit their growth [140]. Conversely, miR-1246 is known to be upregulated in PC exosomes and has been linked to EMT and chemoresistance [135]. By targeting and downregulating tumor suppressor genes, miR-1246 supports a more aggressive cancer phenotype.
As mentioned before, miRNAs can modulate the immune landscape, supporting immune evasion by cancer cells. For instance, EV-derived miR-203 can reduce the anti-tumor immune response by downregulating SOCS3 and promoting immunosuppressive environments [141]. miRNAs can also influence stromal cells within the TME. EVs containing miR-1246 can activate pancreatic stellate cells, thereby promoting desmoplasia and facilitating tumor growth [86].
Numerous studies have demonstrated how EVs from PC patients contain specific miRNAs that can serve as diagnostic and prognostic markers. For instance, elevated levels of miR-19b, miR-21, miR-27b, miR-210 and miR-221 are frequently found in EVs from PC patients compared to healthy donors and are associated with tumor progression, metastasis, and poor prognosis [138,142,143,144,145]. Moreover, exosome-derived miR-21, as well as miR-10b, miR-181 and miR-121, are usually upregulated in PDAC patients compared to healthy donors [142]. Elevated levels of miR-21 in EVs have also been correlated with poor prognosis and advanced disease, highlighting its potential as a prognostic biomarker [142]. Exosome-derived miR-19b is regarded as a promising diagnostic marker, allowing to discriminate between patients with PC and patients with chronic pancreatitis or healthy controls [146]. miR-301a-3p carried by exosomes promotes migration and invasion of PC cells by targeting PTEN and activating the PI3K signaling pathway, so that it could be a valid diagnostic and prognostic biomarker [147,148]. In addition, miR-221 targets tumor suppressor genes such as PTEN and PDCD4, thereby activating pathways that lead to increased tumor growth and resistance to apoptosis [138]. On the contrary, EVs containing miR-146a can suppress PC cell invasion and metastasis by downregulating the EGFR/ERK pathway [149].
The principal EV-derived miRNAs used as biomarkers in PC and PDAC are listed in Table 3.

3.4. Hepatocarcinoma

Hepatocarcinoma (HCC) is the most common primary malignant tumor of the liver and accounts for about 75–85% of all liver cancers [151]. It typically arises in the context of chronic liver disease, particularly cirrhosis caused by hepatitis B or C infection, chronic alcohol abuse, or nonalcoholic fatty liver disease [151]. HCC has a poor prognosis because of its aggressive nature and late presentation in many patients. Treatment options, which are limited and differ according to the stage of the disease, include surgical resection, liver transplantation, and systemic therapies, such as targeted treatments and immunotherapy [152]. Early detection and surveillance in high-risk populations are critical to improving outcomes. EVs have a significant role in HCC progression and are involved in processes such as cell proliferation, EMT and immune response modulation. High levels of miR-21 in EVs have been correlated with tumor progression, by targeting PTEN and AKT-signaling, and with poor prognosis in HCC patients [153,154,155]. Moreover, exosome-derived miR-21 isolated from HCC cell lines contributes to tumor progression by promoting angiogenesis and EMT [155,156]. In addition, EV-derived miR-3129, isolated from patients’ plasma, enhances HCC growth and promotes metastasis formation [157]. On the other hand, the downregulation of exosomal miR-200b-3p in HCC cells promotes angiogenesis through the increased expression of endothelial ERG [158].
Different studies have identified specific miRNAs in serum-derived EVs that are associated with HCC, such as miR-21, miR-122, and miR-192, often dysregulated in cancer cells [82,153,159]. Among these miRNAs, miR-122 is a liver-specific miRNA that has been found to be significantly reduced in EVs from HCC patients, reflecting its role in liver homeostasis and its potential as a diagnostic marker [160,161]. It has also been demonstrated that EV-derived miR-210 targets SMAD4 and STAT6, inducing angiogenesis and tumor progression [162]. Other EV-derived miRNAs that could be used as diagnostic biomarkers due to their upregulation in HCC patients’ serum are miR-15b, miR-16, miR-150 and miR-26a [163].
It has been reported that exosomal miR-106a expression, increased in HCC patients’ serum, correlates with a poor prognosis. In fact, miR-106a regulates the MAPK and c-Jun N-terminal kinase (JNK) pathways, thus supporting tumor progression [164].
miRNAs of the miR-199 family are instead considered diagnostic biomarkers downregulated in HCC patients. Notably, the low expression of miR-199a/b-3p, the third most abundantly expressed miRNA in the liver, is associated with poor overall survival. miR-199a plays a pivotal role in controlling tumor aggressiveness by influencing E-cadherin regulation. In addition, overexpression of miR-199 family significantly suppresses HCC cell proliferation, migration, and invasion by inhibiting the Regulators of G-protein signaling (RGS) pathway. These findings suggest that restoring normal levels of miR-199a/b-3p could serve as a potential therapeutic strategy for HCC, able to target multiple pathways involved in HCC growth and aggressiveness [153].
Another miRNA that can act as a tumor suppressor in HCC is miR-214, which prevents tumor growth via the inhibition of β-catenin, a key player in cancer progression [165]. Exosomal miR-214 also inhibits tumor cell proliferation, migration, and metabolic activity by targeting Pyruvate Dehydrogenase Kinase Isoform 2 (PDK2) and PHD Finger Protein 6 (PHF6) in HCC cell cultures. In addition, miR-214 negatively regulates PVT1, an lncRNA linked to HCC development. Elevated levels of miR-214 have been associated with reduced PVT1 expression both in in vivo and in vitro studies, and PVT1 silencing through miR-214 or siRNA significantly reduced HCC cell viability and invasiveness [153,166].
A recent study by Boonkaew et al. showed that plasma EV-miRNAs could effectively discriminate HCC patients from healthy ones. In particular, miR-223 expression levels were significantly higher in patients with HCC compared with healthy controls [167]. Hence, it may potentially serve as a biomarker for HCC progression and patient outcomes [153,168].
The principal EV-derived miRNAs used as biomarkers in HCC are listed in Table 4.

3.5. Cholangiocarcinoma

Cholangiocarcinoma (CCA) is an aggressive and heterogeneous malignancy originating from the bile duct epithelium [169]. It is associated with an adverse prognosis and high mortality and, due to its increasing global incidence, there is an urgent need for effective early detection and treatment strategies [169]. EVs, including exosomes and MVs, determine and influence different cellular processes in CCA, such as proliferation, invasion, and metastasis [170], and their contents have recently emerged as ideal biomarkers for liquid biopsy-based diagnosis and prognosis of CCA [9,171].
In this regard, specific miRNAs within exosomes have been identified as key players in CCA tumorigenesis and metastasis. miR-21 is frequently upregulated in many GI cancers, including CCA, and is known for its role in promoting cell proliferation, invasion, and survival [172]. Furthermore, miRNAs carried by EVs from CCA patients have shown promise in distinguishing between malignant and benign biliary conditions. High-throughput analysis, for example, highlighted that miR-9, a well-known marker of biliary tract cancer [173], was increased in CCA patients. Moreover, EV-associated miR-21 and miR-222 are frequently upregulated in CCA and correlate with tumor progression and poor prognosis. These miRNAs were, in fact, overexpressed in either plasma or serum of CCA patients compared to healthy donors [174,175]. In addition, miR-221 promotes cell cycle progression and inhibits apoptosis by targeting and downregulating cell cycle inhibitors such as p27Kip1 and pro-apoptotic proteins [176]. Its overexpression is often correlated with poor prognosis and advanced disease stages. Members of the miR-200 family, including miR-200c and miR-141, are involved in regulating EMT in CCA by targeting ZEB1 and ZEB2, which are transcriptional repressors of E-cadherin [177]. Dysregulation of these miRNAs in exosomes can lead to enhanced metastatic capabilities of CCA cells [178]. These miRNAs within EVs not only reflect the biological behavior of CCA cells, but also actively contribute to modifying the TME and promoting disease progression.
Lastly, the upregulation of EV-derived miRNAs, such as miR-34c, miR-30e and miR-195, leads to CCA development and progression by targeting Snail and inducing EMT; for this reason, they could be used as potential diagnostic biomarkers [179,180,181]. Overall, their detection in bodily fluids like blood and bile offers a promising approach for non-invasive diagnostics and monitoring, while their specific pathways provide targets for potential therapeutic interventions. The principal EV-derived miRNAs used as biomarkers in CCA are listed in Table 5.
Figure 1 is a schematic representation of EV-derived miRNA in GI cancer.

4. Conclusions

Traditional biomarkers often fail to provide the necessary sensitivity and specificity for early cancer detection and monitoring therapy response. However, due to their stability in body fluids and their ability to protect, thanks to their lipid bilayer, their molecular cargo from degradation, EVs represent a novel opportunity for non-invasive biomarker assessment, highly suitable for liquid biopsy approaches. Liquid biopsies are less invasive than traditional tissue biopsies and can be repeatedly obtained with minimal discomfort to the patient. This enables continuous monitoring of disease progression and treatment efficacy. EV-associated miRNAs offer several advantages as biomarkers, since they are stable in circulation and protected from RNase degradation. Compared with circulating miRNAs, EV-derived miRNAs offer an improved specificity since EVs are actively released by tumor cells and therefore can reflect distinct cellular states. In addition, surface markers expressed on EVs can enable further isolation and characterization of their cellular origin [182]. Although tissue-derived miRNAs offer a direct and highly specific molecular profile of the tissue of interest, their collection requires biopsy, an invasive and often painful procedure associated with potential clinical risks. Consequently, tissue biopsies are not suitable for routine screening or repeated monitoring.
Furthermore, they can be detected with high sensitivity and specificity using advanced molecular techniques, such as quantitative PCR, microarrays, and next-generation sequencing. These methodological advances have enabled comprehensive profiling of EV-derived miRNAs, facilitating the identification of specific expression signatures linked to tumor type, stage, and even response to therapy. The potential role of EVs in early detection and monitoring of GI cancer is drawing increasing attention. Studies have shown that EVs from GI cancer patients often contain specific tumor-derived proteins, nucleic acids, and other biomolecules that can distinguish them from those of healthy individuals. These tumor-derived EVs can serve as a molecular signature of the underlying neoplasm, providing a rich source of information for early diagnosis and characterization of the disease. The use of liquid biopsy approaches in GI could revolutionize cancer diagnostics, while current imaging and histopathological techniques often fail to capture tumor heterogeneity or detect metastatic disease. In addition to early detection, this novel approach would allow for monitoring of disease progression and evaluation of treatment efficacy with minimal invasiveness, ultimately improving patient survival rates and quality of life. Among the various EV cargoes, EV-associated miRNAs have emerged as particularly interesting biomarker candidates. In the context of GI cancers, EV-associated miRNAs play a crucial role in proliferation, invasion, angiogenesis, and immune evasion. These miRNAs can serve as diagnostic and prognostic markers, and miRNA signatures can also be exploited for patient stratification, driving therapeutic choices and predicting response to specific treatments, including chemotherapy and targeted therapies. Consequently, they may play a key role in the development of personalized therapeutic strategies. In conclusion, the unique biological properties of EVs make them powerful tools for the next generation of cancer diagnostics and monitoring. In the field of GI cancers, where there is an urgent need for non-invasive, accurate, and dynamic biomarkers, EVs, particularly their miRNA cargo, offer a promising approach for improving clinical decision-making and patient outcomes.

Author Contributions

D.N., V.L., V.A., N.F., M.B., C.F., L.D.C., O.B., C.P., L.C., E.F., R.B. and L.M. contributed to the writing, reviewing, and editing of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Ministero della Salute, Piano Nazionale Complementare “D3 4 Health—Digital Driven Diagnostics, prognostics and therapeutics for sustainable Health care”. n. PNC0000001 (L.M.). PRIN 2022 PNRR n° P2022XZKBM to (L.C.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

D.N. and L.D.C. are recipient of the Sapienza Doctoral program in Molecular Medicine, N.F. is recipient of the Sapienza Doctoral program in Network Oncology And Precision Medicine.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
Argonaute(Ago)
Cancer antigen 19-9(CA19-9)
Cancer-associated fibroblasts(CAFs)
Carbohydrate antigen 12–5(CA 12-5)
Carbohydrate antigen 15–3(CA 15-3)
Carcinoembryonic antigen(CEA)
Cholangiocarcinoma(CCA)
Circular RNA(circRNA)
Circulating cell-free RNA(cfDNA)
Circulating tumor cells(CTCs)
Circulating tumor DNA(ctDNA)
C-Jun N-terminal kinase(JNK)
Colorectal cancer(CRC)
Endosomal Sorting Complex Required for Transport (ESCRT)
Epidermal growth factor receptor(EGFR)
Epithelial-mesenchymal transition(EMT)
Extracellular vesicles(EVs)
Extracellular vesicles derived from gastrointestinal(GI-Evs)
Factor forkhead box O1(FOXO1)
Gastric cancer(GC)
Gastrointestinal(GI)
Hepatocarcinoma(HCC)
Heterogeneous nuclear ribonucleoproteins(hnRNPs)
Intraluminal vesicles(ILVs)
Log non-coding RNAs(lncRNAs)
Mammalian target of rapamycin(mTOR)
Messenger RNAs(mRNAs)
Microvesicles(MVs)
mir-34a coated with exosomes(exomiR-34a)
Mitogen-activated protein kinase(MAPK)
Pancreatic cancer(PC)
Pancreatic ductal adenocarcinoma(PDAC)
Phd finger protein 6(PHF6)
Piwi-interacting RNA(piRNA)
Pyruvate dehydrogenase kinase isoform 2(PDK2)
Regulators of G-protein signaling(RGS)
RNA-binding proteins(RBPs)
Small RNAs / microRNAs(miRNA)
Suppressor of cytokine signaling 1(SOCS1)
Vascular endothelial growth factor(VEGF)
Transforming growth factor-beta(TGF-β)
Tumor microenvironment(TME)

References

  1. Ben-Aharon, I.; van Laarhoven, H.W.M.; Fontana, E.; Obermannova, R.; Nilsson, M.; Lordick, F. Early-Onset Cancer in the Gastrointestinal Tract Is on the Rise-Evidence and Implications. Cancer Discov. 2023, 13, 538–551. [Google Scholar] [CrossRef]
  2. Arnold, M.; Abnet, C.C.; Neale, R.E.; Vignat, J.; Giovannucci, E.L.; McGlynn, K.A.; Bray, F. Global Burden of 5 Major Types of Gastrointestinal Cancer. Gastroenterology 2020, 159, 335–349.e15. [Google Scholar] [CrossRef]
  3. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  4. Lucarini, V.; Nardozi, D.; Angiolini, V.; Benvenuto, M.; Focaccetti, C.; Carrano, R.; Besharat, Z.M.; Bei, R.; Masuelli, L. Tumor Microenvironment Remodeling in Gastrointestinal Cancer: Role of miRNAs as Biomarkers of Tumor Invasion. Biomedicines 2023, 11, 1761. [Google Scholar] [CrossRef] [PubMed]
  5. Petricevic, B.; Kabiljo, J.; Zirnbauer, R.; Walczak, H.; Laengle, J.; Bergmann, M. Neoadjuvant immunotherapy in gastrointestinal cancers—The new standard of care? Semin. Cancer Biol. 2022, 86, 834–850. [Google Scholar] [CrossRef] [PubMed]
  6. Li, Y.; Sui, S.; Goel, A. Extracellular vesicles associated microRNAs: Their biology and clinical significance as biomarkers in gastrointestinal cancers. Semin. Cancer Biol. 2024, 99, 5–23. [Google Scholar] [CrossRef]
  7. Zhang, J.; Quadri, S.; Wolfgang, C.L.; Zheng, L. New Development of Biomarkers for Gastrointestinal Cancers: From Neoplastic Cells to Tumor Microenvironment. Biomedicines 2018, 6, 87. [Google Scholar] [CrossRef]
  8. Parikh, A.R.; Leshchiner, I.; Elagina, L.; Goyal, L.; Levovitz, C.; Siravegna, G.; Livitz, D.; Rhrissorrakrai, K.; Martin, E.E.; Van Seventer, E.E.; et al. Liquid versus tissue biopsy for detecting acquired resistance and tumor heterogeneity in gastrointestinal cancers. Nat. Med. 2019, 25, 1415–1421, Erratum in Nat. Med. 2019, 25, 1949. [Google Scholar] [CrossRef]
  9. Lapitz, A.; Azkargorta, M.; Milkiewicz, P.; Olaizola, P.; Zhuravleva, E.; Grimsrud, M.M.; Schramm, C.; Arbelaiz, A.; O’Rourke, C.J.; La Casta, A.; et al. Liquid biopsy-based protein biomarkers for risk prediction, early diagnosis, and prognostication of cholangiocarcinoma. J. Hepatol. 2023, 79, 93–108. [Google Scholar] [CrossRef]
  10. Ma, S.; Zhou, M.; Xu, Y.; Gu, X.; Zou, M.; Abudushalamu, G.; Yao, Y.; Fan, X.; Wu, G. Clinical application and detection techniques of liquid biopsy in gastric cancer. Mol. Cancer 2023, 22, 7. [Google Scholar] [CrossRef]
  11. Zhou, H.; Zhu, L.; Song, J.; Wang, G.; Li, P.; Li, W.; Luo, P.; Sun, X.; Wu, J.; Liu, Y.; et al. Liquid biopsy at the frontier of detection, prognosis and progression monitoring in colorectal cancer. Mol. Cancer 2022, 21, 86. [Google Scholar] [CrossRef]
  12. Raza, A.; Khan, A.Q.; Inchakalody, V.P.; Mestiri, S.; Yoosuf, Z.; Bedhiafi, T.; El-Ella, D.M.A.; Taib, N.; Hydrose, S.; Akbar, S.; et al. Dynamic liquid biopsy components as predictive and prognostic biomarkers in colorectal cancer. J. Exp. Clin. Cancer Res. 2022, 41, 99. [Google Scholar] [CrossRef]
  13. Lone, S.N.; Nisar, S.; Masoodi, T.; Singh, M.; Rizwan, A.; Hashem, S.; El-Rifai, W.; Bedognetti, D.; Batra, S.K.; Haris, M.; et al. Liquid biopsy: A step closer to transform diagnosis, prognosis and future of cancer treatments. Mol. Cancer 2022, 21, 79. [Google Scholar] [CrossRef]
  14. Yu, W.; Hurley, J.; Roberts, D.; Chakrabortty, S.K.; Enderle, D.; Noerholm, M.; Breakefield, X.O.; Skog, J.K. Exosome-based liquid biopsies in cancer: Opportunities and challenges. Ann. Oncol. 2021, 32, 466–477. [Google Scholar] [CrossRef]
  15. Otsuka, M.; Kotani, A. Recent advances in extracellular vesicles in gastrointestinal cancer and lymphoma. Cancer Sci. 2023, 114, 2230–2237. [Google Scholar] [CrossRef] [PubMed]
  16. Hanayama, R. Emerging roles of extracellular vesicles in physiology and disease. J. Biochem. 2021, 169, 135–138. [Google Scholar] [CrossRef] [PubMed]
  17. Nicolini, A.; Ferrari, P.; Biava, P.M. Exosomes and Cell Communication: From Tumour-Derived Exosomes and Their Role in Tumour Progression to the Use of Exosomal Cargo for Cancer Treatment. Cancers 2021, 13, 822. [Google Scholar] [CrossRef] [PubMed]
  18. Urabe, F.; Kosaka, N.; Ito, K.; Kimura, T.; Egawa, S.; Ochiya, T. Extracellular vesicles as biomarkers and therapeutic targets for cancer. Am. J. Physiol. Cell Physiol. 2020, 318, C29–C39. [Google Scholar] [CrossRef]
  19. Pan, B.T.; Johnstone, R.M. Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: Selective externalization of the receptor. Cell 1983, 33, 967–978. [Google Scholar] [CrossRef]
  20. Berumen Sanchez, G.; Bunn, K.E.; Pua, H.H.; Rafat, M. Extracellular vesicles: Mediators of intercellular communication in tissue injury and disease. Cell Commun. Signal. 2021, 19, 104. [Google Scholar] [CrossRef]
  21. Maacha, S.; Bhat, A.A.; Jimenez, L.; Raza, A.; Haris, M.; Uddin, S.; Grivel, J.C. Extracellular vesicles-mediated intercellular communication: Roles in the tumor microenvironment and anti-cancer drug resistance. Mol. Cancer 2019, 18, 55. [Google Scholar] [CrossRef]
  22. Zhou, M.; Li, Y.J.; Tang, Y.C.; Hao, X.Y.; Xu, W.J.; Xiang, D.X.; Wu, J.Y. Apoptotic bodies for advanced drug delivery and therapy. J. Control. Release 2022, 351, 394–406. [Google Scholar] [CrossRef]
  23. He, C.; Zheng, S.; Luo, Y.; Wang, B. Exosome Theranostics: Biology and Translational Medicine. Theranostics 2018, 8, 237–255. [Google Scholar] [CrossRef]
  24. Santavanond, J.P.; Rutter, S.F.; Atkin-Smith, G.K.; Poon, I.K.H. Apoptotic Bodies: Mechanism of Formation, Isolation and Functional Relevance. Subcell Biochem. 2021, 97, 61–88. [Google Scholar] [CrossRef] [PubMed]
  25. Schmidtmann, M.; D’Souza-Schorey, C. Extracellular Vesicles: Biological Packages That Modulate Tumor Cell Invasion. Cancers 2023, 15, 5617. [Google Scholar] [CrossRef]
  26. Meldolesi, J. Exosomes and Ectosomes in Intercellular Communication. Curr. Biol. 2018, 28, R435–R444. [Google Scholar] [CrossRef]
  27. Dixson, A.C.; Dawson, T.R.; Di Vizio, D.; Weaver, A.M. Context-specific regulation of extracellular vesicle biogenesis and cargo selection. Nat. Rev. Mol. Cell Biol. 2023, 24, 454–476. [Google Scholar] [CrossRef] [PubMed]
  28. van Niel, G.; D’Angelo, G.; Raposo, G. Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell Biol. 2018, 19, 213–228. [Google Scholar] [CrossRef]
  29. Lee, Y.; Kim, J.H. The emerging roles of extracellular vesicles as intercellular messengers in liver physiology and pathology. Clin. Mol. Hepatol. 2022, 28, 706–724. [Google Scholar] [CrossRef]
  30. Hade, M.D.; Suire, C.N.; Mossell, J.; Suo, Z. Extracellular vesicles: Emerging frontiers in wound healing. Med. Res. Rev. 2022, 42, 2102–2125. [Google Scholar] [CrossRef] [PubMed]
  31. Buzas, E.I. The roles of extracellular vesicles in the immune system. Nat. Rev. Immunol. 2023, 23, 236–250. [Google Scholar] [CrossRef]
  32. Wang, X.; Xia, J.; Yang, L.; Dai, J.; He, L. Recent progress in exosome research: Isolation, characterization and clinical applications. Cancer Gene Ther. 2023, 30, 1051–1065. [Google Scholar] [CrossRef]
  33. Chen, Z.Q.; Tang, T.T.; Tang, R.N.; Zhang, Y.; Zhang, Y.L.; Yang, H.B.; Song, J.; Yang, Q.; Qin, S.F.; Chen, F.; et al. A comprehensive evaluation of stability and safety for HEK293F-derived extracellular vesicles as promising drug delivery vehicles. J. Control. Release 2025, 382, 113673. [Google Scholar] [CrossRef] [PubMed]
  34. Lyu, N.; Knight, R.; Robertson, S.Y.T.; Dos Santos, A.; Zhang, C.; Ma, C.; Xu, J.; Zheng, J.; Deng, S.X. Stability and Function of Extracellular Vesicles Derived from Immortalized Human Corneal Stromal Stem Cells: A Proof of Concept Study. AAPS J. 2022, 25, 8. [Google Scholar] [CrossRef] [PubMed]
  35. Jin, Y.; Chen, K.; Wang, Z.; Wang, Y.; Liu, J.; Lin, L.; Shao, Y.; Gao, L.; Yin, H.; Cui, C.; et al. DNA in serum extracellular vesicles is stable under different storage conditions. BMC Cancer 2016, 16, 753. [Google Scholar] [CrossRef]
  36. Pang, B.; Zhu, Y.; Ni, J.; Thompson, J.; Malouf, D.; Bucci, J.; Graham, P.; Li, Y. Extracellular vesicles: The next generation of biomarkers for liquid biopsy-based prostate cancer diagnosis. Theranostics 2020, 10, 2309–2326. [Google Scholar] [CrossRef]
  37. Sidhom, K.; Obi, P.O.; Saleem, A. A Review of Exosomal Isolation Methods: Is Size Exclusion Chromatography the Best Option? Int. J. Mol. Sci. 2020, 21, 6466. [Google Scholar] [CrossRef]
  38. Su, Y.; He, W.; Zheng, L.; Fan, X.; Hu, T.Y. Toward Clarity in Single Extracellular Vesicle Research: Defining the Field and Correcting Missteps. ACS Nano 2025, 19, 16193–16203. [Google Scholar] [CrossRef]
  39. Coumans, F.A.W.; Brisson, A.R.; Buzas, E.I.; Dignat-George, F.; Drees, E.E.E.; El-Andaloussi, S.; Emanueli, C.; Gasecka, A.; Hendrix, A.; Hill, A.F.; et al. Methodological Guidelines to Study Extracellular Vesicles. Circ. Res. 2017, 120, 1632–1648. [Google Scholar] [CrossRef] [PubMed]
  40. Schubert, A.; Boutros, M. Extracellular vesicles and oncogenic signaling. Mol. Oncol. 2021, 15, 3–26. [Google Scholar] [CrossRef]
  41. Hinger, S.A.; Abner, J.J.; Franklin, J.L.; Jeppesen, D.K.; Coffey, R.J.; Patton, J.G. Rab13 regulates sEV secretion in mutant KRAS colorectal cancer cells. Sci. Rep. 2020, 10, 15804. [Google Scholar] [CrossRef]
  42. Kang, T.; Atukorala, I.; Mathivanan, S. Biogenesis of Extracellular Vesicles. Subcell. Biochem. 2021, 97, 19–43. [Google Scholar] [CrossRef]
  43. Nazarenko, I. Extracellular Vesicles: Recent Developments in Technology and Perspectives for Cancer Liquid Biopsy. Recent Results Cancer Res. 2020, 215, 319–344. [Google Scholar] [CrossRef] [PubMed]
  44. Johnstone, R.M.; Adam, M.; Hammond, J.R.; Orr, L.; Turbide, C. Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes). J. Biol. Chem. 1987, 262, 9412–9420. [Google Scholar] [CrossRef]
  45. Huotari, J.; Helenius, A. Endosome maturation. EMBO J. 2011, 30, 3481–3500. [Google Scholar] [CrossRef] [PubMed]
  46. Mathieu, M.; Martin-Jaular, L.; Lavieu, G.; Thery, C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat. Cell Biol. 2019, 21, 9–17. [Google Scholar] [CrossRef] [PubMed]
  47. Montaldo, C.; Terri, M.; Riccioni, V.; Battistelli, C.; Bordoni, V.; D’Offizi, G.; Prado, M.G.; Trionfetti, F.; Vescovo, T.; Tartaglia, E.; et al. Fibrogenic signals persist in DAA-treated HCV patients after sustained virological response. J. Hepatol. 2021, 75, 1301–1311. [Google Scholar] [CrossRef]
  48. Chellini, L.; Palombo, R.; Riccioni, V.; Paronetto, M.P. Oncogenic Dysregulation of Circulating Noncoding RNAs: Novel Challenges and Opportunities in Sarcoma Diagnosis and Treatment. Cancers 2022, 14, 4677. [Google Scholar] [CrossRef]
  49. Bonsergent, E.; Grisard, E.; Buchrieser, J.; Schwartz, O.; Thery, C.; Lavieu, G. Quantitative characterization of extracellular vesicle uptake and content delivery within mammalian cells. Nat. Commun. 2021, 12, 1864. [Google Scholar] [CrossRef]
  50. Fonseka, P.; Mathivanan, S. Extracellular Vesicles Biogenesis, Cargo Sorting and Implications in Disease Conditions. Cells 2023, 12, 280. [Google Scholar] [CrossRef]
  51. Chen, A.; He, B.; Jin, H. Isolation of Extracellular Vesicles from Arabidopsis. Curr. Protoc. 2022, 2, e352. [Google Scholar] [CrossRef]
  52. Atay, S.; Wilkey, D.W.; Milhem, M.; Merchant, M.; Godwin, A.K. Insights into the Proteome of Gastrointestinal Stromal Tumors-Derived Exosomes Reveals New Potential Diagnostic Biomarkers. Mol. Cell. Proteom. 2018, 17, 495–515. [Google Scholar] [CrossRef]
  53. Gurunathan, S.; Kang, M.H.; Qasim, M.; Khan, K.; Kim, J.H. Biogenesis, Membrane Trafficking, Functions, and Next Generation Nanotherapeutics Medicine of Extracellular Vesicles. Int. J. Nanomedicine 2021, 16, 3357–3383. [Google Scholar] [CrossRef] [PubMed]
  54. Silva, A.M.; Lazaro-Ibanez, E.; Gunnarsson, A.; Dhande, A.; Daaboul, G.; Peacock, B.; Osteikoetxea, X.; Salmond, N.; Friis, K.P.; Shatnyeva, O.; et al. Quantification of protein cargo loading into engineered extracellular vesicles at single-vesicle and single-molecule resolution. J. Extracell. Vesicles 2021, 10, e12130. [Google Scholar] [CrossRef]
  55. Hinger, S.A.; Cha, D.J.; Franklin, J.L.; Higginbotham, J.N.; Dou, Y.; Ping, J.; Shu, L.; Prasad, N.; Levy, S.; Zhang, B.; et al. Diverse Long RNAs Are Differentially Sorted into Extracellular Vesicles Secreted by Colorectal Cancer Cells. Cell Rep. 2018, 25, 715–725.e14. [Google Scholar] [CrossRef]
  56. Herrmann, I.K.; Wood, M.J.A.; Fuhrmann, G. Extracellular vesicles as a next-generation drug delivery platform. Nat. Nanotechnol. 2021, 16, 748–759. [Google Scholar] [CrossRef]
  57. O’Grady, T.; Njock, M.S.; Lion, M.; Bruyr, J.; Mariavelle, E.; Galvan, B.; Boeckx, A.; Struman, I.; Dequiedt, F. Sorting and packaging of RNA into extracellular vesicles shape intracellular transcript levels. BMC Biol. 2022, 20, 72. [Google Scholar] [CrossRef]
  58. Xi, E.; Bai, J.; Zhang, K.; Yu, H.; Guo, Y. Genomic Variants Disrupt miRNA-mRNA Regulation. Chem. Biodivers 2022, 19, e202200623. [Google Scholar] [CrossRef] [PubMed]
  59. Ambros, V.; Bartel, B.; Bartel, D.P.; Burge, C.B.; Carrington, J.C.; Chen, X.; Dreyfuss, G.; Eddy, S.R.; Griffiths-Jones, S.; Marshall, M.; et al. A uniform system for microRNA annotation. RNA 2003, 9, 277–279. [Google Scholar] [CrossRef] [PubMed]
  60. Garcia-Martin, R.; Wang, G.; Brandao, B.B.; Zanotto, T.M.; Shah, S.; Kumar Patel, S.; Schilling, B.; Kahn, C.R. MicroRNA sequence codes for small extracellular vesicle release and cellular retention. Nature 2022, 601, 446–451. [Google Scholar] [CrossRef]
  61. Jiang, X.; Liu, B.; Nie, Z.; Duan, L.; Xiong, Q.; Jin, Z.; Yang, C.; Chen, Y. The role of m6A modification in the biological functions and diseases. Signal Transduct. Target. Ther. 2021, 6, 74. [Google Scholar] [CrossRef]
  62. Liu, R.; Zhong, Y.; Chen, R.; Chu, C.; Liu, G.; Zhou, Y.; Huang, Y.; Fang, Z.; Liu, H. m(6)A reader hnRNPA2B1 drives multiple myeloma osteolytic bone disease. Theranostics 2022, 12, 7760–7774. [Google Scholar] [CrossRef] [PubMed]
  63. Liu, X.M.; Ma, L.; Schekman, R. Selective sorting of microRNAs into exosomes by phase-separated YBX1 condensates. eLife 2021, 10, e71982. [Google Scholar] [CrossRef] [PubMed]
  64. Cabiati, M.; Salvadori, C.; Basta, G.; Del Turco, S.; Aretini, P.; Cecchettini, A.; Del Ry, S. miRNA and long non-coding RNA transcriptional expression in hepatocellular carcinoma cell line-secreted extracellular vesicles. Clin. Exp. Med. 2022, 22, 245–255. [Google Scholar] [CrossRef] [PubMed]
  65. Battaglia, R.; Palini, S.; Vento, M.E.; La Ferlita, A.; Lo Faro, M.J.; Caroppo, E.; Borzi, P.; Falzone, L.; Barbagallo, D.; Ragusa, M.; et al. Identification of extracellular vesicles and characterization of miRNA expression profiles in human blastocoel fluid. Sci. Rep. 2019, 9, 84. [Google Scholar] [CrossRef]
  66. Moloudizargari, M.; Hekmatirad, S.; Mofarahe, Z.S.; Asghari, M.H. Exosomal microRNA panels as biomarkers for hematological malignancies. Curr. Probl. Cancer 2021, 45, 100726. [Google Scholar] [CrossRef]
  67. Blaser, M.C.; Aikawa, E. Differential miRNA Loading Underpins Dual Harmful and Protective Roles for Extracellular Vesicles in Atherogenesis. Circ. Res. 2019, 124, 467–469. [Google Scholar] [CrossRef]
  68. Groven, R.V.M.; Greven, J.; Mert, U.; Horst, K.; Zhao, Q.; Blokhuis, T.J.; Huber-Lang, M.; Hildebrand, F.; van Griensven, M. Circulating miRNA expression in extracellular vesicles is associated with specific injuries after multiple trauma and surgical invasiveness. Front. Immunol. 2023, 14, 1273612. [Google Scholar] [CrossRef]
  69. Wang, S.; Jin, S.; Zhang, J.; Wang, X. Exosomal miRNAs: Key Regulators of the Tumor Microenvironment and Cancer Stem Cells. Int. J. Mol. Sci. 2025, 26, 9323. [Google Scholar] [CrossRef]
  70. Lucotti, S.; Kenific, C.M.; Zhang, H.; Lyden, D. Extracellular vesicles and particles impact the systemic landscape of cancer. EMBO J. 2022, 41, e109288. [Google Scholar] [CrossRef]
  71. Becker, A.; Thakur, B.K.; Weiss, J.M.; Kim, H.S.; Peinado, H.; Lyden, D. Extracellular Vesicles in Cancer: Cell-to-Cell Mediators of Metastasis. Cancer Cell 2016, 30, 836–848. [Google Scholar] [CrossRef]
  72. Sullivan, R.; Maresh, G.; Zhang, X.; Salomon, C.; Hooper, J.; Margolin, D.; Li, L. The Emerging Roles of Extracellular Vesicles As Communication Vehicles within the Tumor Microenvironment and Beyond. Front. Endocrinol. 2017, 8, 194. [Google Scholar] [CrossRef]
  73. Schneider, N.; Hermann, P.C.; Eiseler, T.; Seufferlein, T. Emerging Roles of Small Extracellular Vesicles in Gastrointestinal Cancer Research and Therapy. Cancers 2024, 16, 567. [Google Scholar] [CrossRef]
  74. Jiang, C.; Zhang, J.; Wang, W.; Shan, Z.; Sun, F.; Tan, Y.; Tong, Y.; Qiu, Y. Extracellular vesicles in gastric cancer: Role of exosomal lncRNA and microRNA as diagnostic and therapeutic targets. Front. Physiol. 2023, 14, 1158839. [Google Scholar] [CrossRef] [PubMed]
  75. Lucchetti, D.; Zurlo, I.V.; Colella, F.; Ricciardi-Tenore, C.; Di Salvatore, M.; Tortora, G.; De Maria, R.; Giuliante, F.; Cassano, A.; Basso, M.; et al. Mutational status of plasma exosomal KRAS predicts outcome in patients with metastatic colorectal cancer. Sci. Rep. 2021, 11, 22686. [Google Scholar] [CrossRef] [PubMed]
  76. Greening, D.W.; Xu, R.; Rai, A.; Suwakulsiri, W.; Chen, M.; Simpson, R.J. Clinical relevance of extracellular vesicles in cancer—therapeutic and diagnostic potential. Nat. Rev. Clin. Oncol. 2025, 22, 924–952. [Google Scholar] [CrossRef]
  77. Gallo, A.; Tandon, M.; Alevizos, I.; Illei, G.G. The majority of microRNAs detectable in serum and saliva is concentrated in exosomes. PLoS ONE 2012, 7, e30679. [Google Scholar] [CrossRef] [PubMed]
  78. Xu, Y.; Han, J.; Zhang, X.; Zhang, X.; Song, J.; Gao, Z.; Qian, H.; Jin, J.; Liang, Z. Exosomal circRNAs in gastrointestinal cancer: Role in occurrence, development, diagnosis and clinical application (Review). Oncol. Rep. 2024, 51, 19. [Google Scholar] [CrossRef]
  79. Tu, J.; Luo, X.; Liu, H.; Zhang, J.; He, M. Cancer spheroids derived exosomes reveal more molecular features relevant to progressed cancer. Biochem. Biophys. Rep. 2021, 26, 101026. [Google Scholar] [CrossRef]
  80. Wang, X.; Chen, W.; Zeng, W.; Feng, K.; Zheng, Y.; Wang, P.; Chen, F.; Zhang, W.; Di, L.; Wang, R. Extracellular vesicles as biomarkers and drug delivery systems for tumor. Acta Pharm. Sin. B 2025, 15, 3460–3486. [Google Scholar] [CrossRef]
  81. Zhang, L.; Shen, H.; Zhang, N. Advances in extracellular vesicle (EV) biomarkers for precision diagnosis and therapeutic in colorectal cancer. Front. Oncol. 2025, 15, 1581015. [Google Scholar] [CrossRef]
  82. Kumar, M.A.; Baba, S.K.; Sadida, H.Q.; Marzooqi, S.A.; Jerobin, J.; Altemani, F.H.; Algehainy, N.; Alanazi, M.A.; Abou-Samra, A.B.; Kumar, R.; et al. Extracellular vesicles as tools and targets in therapy for diseases. Signal Transduct. Target. Ther. 2024, 9, 27. [Google Scholar] [CrossRef] [PubMed]
  83. Zheng, P.; Chen, L.; Yuan, X.; Luo, Q.; Liu, Y.; Xie, G.; Ma, Y.; Shen, L. Exosomal transfer of tumor-associated macrophage-derived miR-21 confers cisplatin resistance in gastric cancer cells. J. Exp. Clin. Cancer Res. 2017, 36, 53. [Google Scholar] [CrossRef]
  84. Kosaka, N.; Iguchi, H.; Hagiwara, K.; Yoshioka, Y.; Takeshita, F.; Ochiya, T. Neutral sphingomyelinase 2 (nSMase2)-dependent exosomal transfer of angiogenic microRNAs regulate cancer cell metastasis. J. Biol. Chem. 2013, 288, 10849–10859. [Google Scholar] [CrossRef]
  85. Zeng, Z.; Li, Y.; Pan, Y.; Lan, X.; Song, F.; Sun, J.; Zhou, K.; Liu, X.; Ren, X.; Wang, F.; et al. Cancer-derived exosomal miR-25-3p promotes pre-metastatic niche formation by inducing vascular permeability and angiogenesis. Nat. Commun. 2018, 9, 5395. [Google Scholar] [CrossRef]
  86. Waldenmaier, M.; Seibold, T.; Seufferlein, T.; Eiseler, T. Pancreatic Cancer Small Extracellular Vesicles (Exosomes): A Tale of Short- and Long-Distance Communication. Cancers 2021, 13, 4844. [Google Scholar] [CrossRef] [PubMed]
  87. Irmer, B.; Chandrabalan, S.; Maas, L.; Bleckmann, A.; Menck, K. Extracellular Vesicles in Liquid Biopsies as Biomarkers for Solid Tumors. Cancers 2023, 15, 1307. [Google Scholar] [CrossRef] [PubMed]
  88. Alsina, M.; Arrazubi, V.; Diez, M.; Tabernero, J. Current developments in gastric cancer: From molecular profiling to treatment strategy. Nat. Rev. Gastroenterol. Hepatol. 2023, 20, 155–170. [Google Scholar] [CrossRef]
  89. Sexton, R.E.; Al Hallak, M.N.; Diab, M.; Azmi, A.S. Gastric cancer: A comprehensive review of current and future treatment strategies. Cancer Metastasis Rev. 2020, 39, 1179–1203. [Google Scholar] [CrossRef]
  90. Smyth, E.C.; Nilsson, M.; Grabsch, H.I.; van Grieken, N.C.; Lordick, F. Gastric cancer. Lancet 2020, 396, 635–648. [Google Scholar] [CrossRef]
  91. Li, C.; Liu, D.R.; Li, G.G.; Wang, H.H.; Li, X.W.; Zhang, W.; Wu, Y.L.; Chen, L. CD97 promotes gastric cancer cell proliferation and invasion through exosome-mediated MAPK signaling pathway. World J. Gastroenterol. 2015, 21, 6215–6228. [Google Scholar] [CrossRef] [PubMed]
  92. Lei, Y.; Cai, S.; Zhang, C.D.; Li, Y.S. The biological role of extracellular vesicles in gastric cancer metastasis. Front. Cell Dev. Biol. 2024, 12, 1323348. [Google Scholar] [CrossRef]
  93. Huang, T.; Song, C.; Zheng, L.; Xia, L.; Li, Y.; Zhou, Y. The roles of extracellular vesicles in gastric cancer development, microenvironment, anti-cancer drug resistance, and therapy. Mol. Cancer 2019, 18, 62. [Google Scholar] [CrossRef]
  94. Zhang, X.; Wang, S.; Wang, H.; Cao, J.; Huang, X.; Chen, Z.; Xu, P.; Sun, G.; Xu, J.; Lv, J.; et al. Circular RNA circNRIP1 acts as a microRNA-149-5p sponge to promote gastric cancer progression via the AKT1/mTOR pathway. Mol. Cancer 2019, 18, 20. [Google Scholar] [CrossRef]
  95. Wang, M.; Qiu, R.; Yu, S.; Xu, X.; Li, G.; Gu, R.; Tan, C.; Zhu, W.; Shen, B. Paclitaxel-resistant gastric cancer MGC-803 cells promote epithelial-to-mesenchymal transition and chemoresistance in paclitaxel-sensitive cells via exosomal delivery of miR-155-5p. Int. J. Oncol. 2019, 54, 326–338. [Google Scholar] [CrossRef]
  96. Du, J.; Liang, Y.; Li, J.; Zhao, J.M.; Lin, X.Y. Gastric Cancer Cell-Derived Exosomal microRNA-23a Promotes Angiogenesis by Targeting PTEN. Front. Oncol. 2020, 10, 326, Erratum in Front. Oncol. 2022, 11, 797657. [Google Scholar] [CrossRef]
  97. Qiu, S.; Xie, L.; Lu, C.; Gu, C.; Xia, Y.; Lv, J.; Xuan, Z.; Fang, L.; Yang, J.; Zhang, L.; et al. Gastric cancer-derived exosomal miR-519a-3p promotes liver metastasis by inducing intrahepatic M2-like macrophage-mediated angiogenesis. J. Exp. Clin. Cancer Res. 2022, 41, 296. [Google Scholar] [CrossRef] [PubMed]
  98. Yang, S.; Wei, S.; Wei, F. Extracellular vesicles mediated gastric cancer immune response: Tumor cell death or immune escape? Cell Death Dis. 2024, 15, 377. [Google Scholar] [CrossRef]
  99. Renovato-Martins, M.; Gomes, A.C.; Amorim, C.S.; Moraes, J.A. The Role of Macrophage-Derived Extracellular Vesicles in Gastrointestinal Cancers. In Gastrointestinal Cancers; Morgado-Diaz, J.A., Ed.; Springer Nature: Brisbane, Austrlia, 2022. [Google Scholar]
  100. Zhu, M.; Zhang, N.; He, S.; Lu, X. Exosomal miR-106a derived from gastric cancer promotes peritoneal metastasis via direct regulation of Smad7. Cell Cycle 2020, 19, 1200–1221. [Google Scholar] [CrossRef]
  101. Wang, N.; Wang, L.; Yang, Y.; Gong, L.; Xiao, B.; Liu, X. A serum exosomal microRNA panel as a potential biomarker test for gastric cancer. Biochem. Biophys. Res. Commun. 2017, 493, 1322–1328. [Google Scholar] [CrossRef] [PubMed]
  102. Liu, F.; Bu, Z.; Zhao, F.; Xiao, D. Increased T-helper 17 cell differentiation mediated by exosome-mediated microRNA-451 redistribution in gastric cancer infiltrated T cells. Cancer Sci. 2018, 109, 65–73. [Google Scholar] [CrossRef]
  103. Makinoya, M.; Miyatani, K.; Matsumi, Y.; Sakano, Y.; Shimizu, S.; Shishido, Y.; Hanaki, T.; Kihara, K.; Matsunaga, T.; Yamamoto, M.; et al. Exosomal miR-493 suppresses MAD2L1 and induces chemoresistance to intraperitoneal paclitaxel therapy in gastric cancer patients with peritoneal metastasis. Sci. Rep. 2024, 14, 10075. [Google Scholar] [CrossRef] [PubMed]
  104. Kahroba, H.; Samadi, N.; Mostafazadeh, M.; Hejazi, M.S.; Sadeghi, M.R.; Hashemzadeh, S.; Eftekhar Sadat, A.T.; Karimi, A. Evaluating the presence of deregulated tumoral onco-microRNAs in serum-derived exosomes of gastric cancer patients as noninvasive diagnostic biomarkers. Bioimpacts 2022, 12, 127–138. [Google Scholar] [CrossRef]
  105. Izumi, D.; Zhu, Z.; Chen, Y.; Toden, S.; Huo, X.; Kanda, M.; Ishimoto, T.; Gu, D.; Tan, M.; Kodera, Y.; et al. Assessment of the Diagnostic Efficiency of a Liquid Biopsy Assay for Early Detection of Gastric Cancer. JAMA Netw. Open 2021, 4, e2121129. [Google Scholar] [CrossRef]
  106. Zheng, G.D.; Xu, Z.Y.; Hu, C.; Lv, H.; Xie, H.X.; Huang, T.; Zhang, Y.Q.; Chen, G.P.; Fu, Y.F.; Cheng, X.D. Exosomal miR-590-5p in Serum as a Biomarker for the Diagnosis and Prognosis of Gastric Cancer. Front. Mol. Biosci. 2021, 8, 636566. [Google Scholar] [CrossRef]
  107. Shi, Y.; Wang, Z.; Zhu, X.; Chen, L.; Ma, Y.; Wang, J.; Yang, X.; Liu, Z. Exosomal miR-1246 in serum as a potential biomarker for early diagnosis of gastric cancer. Int. J. Clin. Oncol. 2020, 25, 89–99. [Google Scholar] [CrossRef] [PubMed]
  108. van Houdt, W.J.; Hoogwater, F.J.; de Bruijn, M.T.; Emmink, B.L.; Nijkamp, M.W.; Raats, D.A.; van der Groep, P.; van Diest, P.; Borel Rinkes, I.H.; Kranenburg, O. Oncogenic KRAS desensitizes colorectal tumor cells to epidermal growth factor receptor inhibition and activation. Neoplasia 2010, 12, 443–452. [Google Scholar] [CrossRef] [PubMed]
  109. Liu, Y.; Liu, R.; Yang, F.; Cheng, R.; Chen, X.; Cui, S.; Gu, Y.; Sun, W.; You, C.; Liu, Z.; et al. miR-19a promotes colorectal cancer proliferation and migration by targeting TIA1. Mol. Cancer 2017, 16, 53. [Google Scholar] [CrossRef]
  110. Qin, J.; Wang, F.; Jiang, H.; Xu, J.; Jiang, Y.; Wang, Z. MicroRNA-145 suppresses cell migration and invasion by targeting paxillin in human colorectal cancer cells. Int. J. Clin. Exp. Pathol. 2015, 8, 1328–1340. [Google Scholar]
  111. Niu, Y.; Zhang, J.; Tong, Y.; Li, J.; Liu, B. miR-145-5p restrained cell growth, invasion, migration and tumorigenesis via modulating RHBDD1 in colorectal cancer via the EGFR-associated signaling pathway. Int. J. Biochem. Cell Biol. 2019, 117, 105641. [Google Scholar] [CrossRef]
  112. Shang, A.; Wang, X.; Gu, C.; Liu, W.; Sun, J.; Zeng, B.; Chen, C.; Ji, P.; Wu, J.; Quan, W.; et al. Exosomal miR-183-5p promotes angiogenesis in colorectal cancer by regulation of FOXO1. Aging 2020, 12, 8352–8371. [Google Scholar] [CrossRef]
  113. Zhou, M.; Wang, S.; Liu, D.; Zhou, J. LINC01915 Facilitates the Conversion of Normal Fibroblasts into Cancer-Associated Fibroblasts Induced by Colorectal Cancer-Derived Extracellular Vesicles through the miR-92a-3p/KLF4/CH25H Axis. ACS Biomater. Sci. Eng. 2021, 7, 5255–5268. [Google Scholar] [CrossRef]
  114. Desmond, B.J.; Dennett, E.R.; Danielson, K.M. Circulating Extracellular Vesicle MicroRNA as Diagnostic Biomarkers in Early Colorectal Cancer-A Review. Cancers 2019, 12, 52. [Google Scholar] [CrossRef]
  115. Yamada, N.O.; Heishima, K.; Akao, Y.; Senda, T. Extracellular Vesicles Containing MicroRNA-92a-3p Facilitate Partial Endothelial-Mesenchymal Transition and Angiogenesis in Endothelial Cells. Int. J. Mol. Sci. 2019, 20, 4406. [Google Scholar] [CrossRef]
  116. Yang, C.; Dou, R.; Wei, C.; Liu, K.; Shi, D.; Zhang, C.; Liu, Q.; Wang, S.; Xiong, B. Tumor-derived exosomal microRNA-106b-5p activates EMT-cancer cell and M2-subtype TAM interaction to facilitate CRC metastasis. Mol. Ther. 2021, 29, 2088–2107. [Google Scholar] [CrossRef] [PubMed]
  117. Wang, D.; Wang, X.; Song, Y.; Si, M.; Sun, Y.; Liu, X.; Cui, S.; Qu, X.; Yu, X. Exosomal miR-146a-5p and miR-155-5p promote CXCL12/CXCR7-induced metastasis of colorectal cancer by crosstalk with cancer-associated fibroblasts. Cell Death Dis. 2022, 13, 380. [Google Scholar] [CrossRef] [PubMed]
  118. Pei, W.; Wei, K.; Wu, Y.; Qiu, Q.; Zhu, H.; Mao, L.; Shi, X.; Zhang, S.; Shi, Y.; Tao, S.; et al. Colorectal cancer tumor cell-derived exosomal miR-203a-3p promotes CRC metastasis by targeting PTEN-induced macrophage polarization. Gene 2023, 885, 147692. [Google Scholar] [CrossRef]
  119. Zhao, S.; Mi, Y.; Guan, B.; Zheng, B.; Wei, P.; Gu, Y.; Zhang, Z.; Cai, S.; Xu, Y.; Li, X.; et al. Tumor-derived exosomal miR-934 induces macrophage M2 polarization to promote liver metastasis of colorectal cancer. J. Hematol. Oncol. 2020, 13, 156, Erratum in J. Hematol. Oncol. 2021, 14, 33. [Google Scholar] [CrossRef] [PubMed]
  120. Ren, J.; Ding, L.; Zhang, D.; Shi, G.; Xu, Q.; Shen, S.; Wang, Y.; Wang, T.; Hou, Y. Carcinoma-associated fibroblasts promote the stemness and chemoresistance of colorectal cancer by transferring exosomal lncRNA H19. Theranostics 2018, 8, 3932–3948. [Google Scholar] [CrossRef]
  121. Navarro, G.; Gomez-Autet, M.; Morales, P.; Rebassa, J.B.; Llinas Del Torrent, C.; Jagerovic, N.; Pardo, L.; Franco, R. Homodimerization of CB(2) cannabinoid receptor triggered by a bivalent ligand enhances cellular signaling. Pharmacol. Res. 2024, 208, 107363. [Google Scholar] [CrossRef]
  122. Ogata-Kawata, H.; Izumiya, M.; Kurioka, D.; Honma, Y.; Yamada, Y.; Furuta, K.; Gunji, T.; Ohta, H.; Okamoto, H.; Sonoda, H.; et al. Circulating exosomal microRNAs as biomarkers of colon cancer. PLoS ONE 2014, 9, e92921. [Google Scholar] [CrossRef]
  123. Campos, A.; Sharma, S.; Obermair, A.; Salomon, C. Extracellular Vesicle-Associated miRNAs and Chemoresistance: A Systematic Review. Cancers 2021, 13, 4608. [Google Scholar] [CrossRef]
  124. Bautista-Sanchez, D.; Arriaga-Canon, C.; Pedroza-Torres, A.; De La Rosa-Velazquez, I.A.; Gonzalez-Barrios, R.; Contreras-Espinosa, L.; Montiel-Manriquez, R.; Castro-Hernandez, C.; Fragoso-Ontiveros, V.; Alvarez-Gomez, R.M.; et al. The Promising Role of miR-21 as a Cancer Biomarker and Its Importance in RNA-Based Therapeutics. Mol. Ther. Nucleic. Acids 2020, 20, 409–420. [Google Scholar] [CrossRef]
  125. Matsumura, T.; Sugimachi, K.; Iinuma, H.; Takahashi, Y.; Kurashige, J.; Sawada, G.; Ueda, M.; Uchi, R.; Ueo, H.; Takano, Y.; et al. Exosomal microRNA in serum is a novel biomarker of recurrence in human colorectal cancer. Br. J. Cancer 2015, 113, 275–281. [Google Scholar] [CrossRef]
  126. Zhao, S.; Mi, Y.; Zheng, B.; Wei, P.; Gu, Y.; Zhang, Z.; Xu, Y.; Cai, S.; Li, X.; Li, D. Highly-metastatic colorectal cancer cell released miR-181a-5p-rich extracellular vesicles promote liver metastasis by activating hepatic stellate cells and remodelling the tumour microenvironment. J. Extracell. Vesicles 2022, 11, e12186. [Google Scholar] [CrossRef] [PubMed]
  127. Yang, C.K.; Hsu, H.C.; Liu, Y.H.; Tsai, W.S.; Ma, C.P.; Chen, Y.T.; Tan, B.C.; Lai, Y.Y.; Chang, I.Y.; Yang, C.; et al. EV-miRome-wide profiling uncovers miR-320c for detecting metastatic colorectal cancer and monitoring the therapeutic response. Cell. Oncol. 2022, 45, 621–638. [Google Scholar] [CrossRef] [PubMed]
  128. Liu, C.; Eng, C.; Shen, J.; Lu, Y.; Takata, Y.; Mehdizadeh, A.; Chang, G.J.; Rodriguez-Bigas, M.A.; Li, Y.; Chang, P.; et al. Serum exosomal miR-4772-3p is a predictor of tumor recurrence in stage II and III colon cancer. Oncotarget 2016, 7, 76250–76260. [Google Scholar] [CrossRef] [PubMed]
  129. Siveen, K.S.; Raza, A.; Ahmed, E.I.; Khan, A.Q.; Prabhu, K.S.; Kuttikrishnan, S.; Mateo, J.M.; Zayed, H.; Rasul, K.; Azizi, F.; et al. The Role of Extracellular Vesicles as Modulators of the Tumor Microenvironment, Metastasis and Drug Resistance in Colorectal Cancer. Cancers 2019, 11, 746. [Google Scholar] [CrossRef]
  130. Senfter, D.; Holzner, S.; Kalipciyan, M.; Staribacher, A.; Walzl, A.; Huttary, N.; Krieger, S.; Brenner, S.; Jager, W.; Krupitza, G.; et al. Loss of miR-200 family in 5-fluorouracil resistant colon cancer drives lymphendothelial invasiveness in vitro. Hum. Mol. Genet. 2015, 24, 3689–3698. [Google Scholar] [CrossRef]
  131. Wu, Y.; Zhang, J.; Lin, F.; Zhao, Y.; Zheng, B.; Zhou, N.; Zhang, Z.; Li, G.; Xie, L. Exosomal miR-1470 is a diagnostic biomarker and promotes cell proliferation and metastasis in colorectal cancer. Cancer Med. 2024, 13, e7117. [Google Scholar] [CrossRef]
  132. Klein, A.P. Pancreatic cancer epidemiology: Understanding the role of lifestyle and inherited risk factors. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 493–502. [Google Scholar] [CrossRef]
  133. Hu, Z.I.; O’Reilly, E.M. Therapeutic developments in pancreatic cancer. Nat. Rev. Gastroenterol. Hepatol. 2024, 21, 7–24. [Google Scholar] [CrossRef] [PubMed]
  134. Wei, H.; Ren, H. Precision treatment of pancreatic ductal adenocarcinoma. Cancer Lett. 2024, 585, 216636. [Google Scholar] [CrossRef]
  135. Qiu, J.; Yang, G.; Feng, M.; Zheng, S.; Cao, Z.; You, L.; Zheng, L.; Zhang, T.; Zhao, Y. Extracellular vesicles as mediators of the progression and chemoresistance of pancreatic cancer and their potential clinical applications. Mol. Cancer 2018, 17, 2. [Google Scholar] [CrossRef]
  136. Capula, M.; Peran, M.; Xu, G.; Donati, V.; Yee, D.; Gregori, A.; Assaraf, Y.G.; Giovannetti, E.; Deng, D. Role of drug catabolism, modulation of oncogenic signaling and tumor microenvironment in microbe-mediated pancreatic cancer chemoresistance. Drug Resist. Updat. 2022, 64, 100864. [Google Scholar] [CrossRef]
  137. Fang, Y.; Zhou, W.; Rong, Y.; Kuang, T.; Xu, X.; Wu, W.; Wang, D.; Lou, W. Exosomal miRNA-106b from cancer-associated fibroblast promotes gemcitabine resistance in pancreatic cancer. Exp. Cell Res. 2019, 383, 111543. [Google Scholar] [CrossRef] [PubMed]
  138. Tiwari, P.K.; Shanmugam, P.; Karn, V.; Gupta, S.; Mishra, R.; Rustagi, S.; Chouhan, M.; Verma, D.; Jha, N.K.; Kumar, S. Extracellular Vesicular miRNA in Pancreatic Cancer: From Lab to Therapy. Cancers 2024, 16, 2179. [Google Scholar] [CrossRef]
  139. Hidalgo-Sastre, A.; Lubeseder-Martellato, C.; Engleitner, T.; Steiger, K.; Zhong, S.; Desztics, J.; Ollinger, R.; Rad, R.; Schmid, R.M.; Hermeking, H.; et al. Mir34a constrains pancreatic carcinogenesis. Sci. Rep. 2020, 10, 9654. [Google Scholar] [CrossRef]
  140. Zuo, L.; Tao, H.; Xu, H.; Li, C.; Qiao, G.; Guo, M.; Cao, S.; Liu, M.; Lin, X. Exosomes-Coated miR-34a Displays Potent Antitumor Activity in Pancreatic Cancer Both in vitro and in vivo. Drug Des. Dev. Ther. 2020, 14, 3495–3507. [Google Scholar] [CrossRef] [PubMed]
  141. Reale, A.; Khong, T.; Spencer, A. Extracellular Vesicles and Their Roles in the Tumor Immune Microenvironment. J. Clin. Med. 2022, 11, 6892. [Google Scholar] [CrossRef]
  142. Lai, X.; Wang, M.; McElyea, S.D.; Sherman, S.; House, M.; Korc, M. A microRNA signature in circulating exosomes is superior to exosomal glypican-1 levels for diagnosing pancreatic cancer. Cancer Lett. 2017, 393, 86–93. [Google Scholar] [CrossRef]
  143. Girolimetti, G.; Pelisenco, I.A.; Eusebi, L.H.; Ricci, C.; Cavina, B.; Kurelac, I.; Verri, T.; Calcagnile, M.; Alifano, P.; Salvi, A.; et al. Dysregulation of a Subset of Circulating and Vesicle-Associated miRNA in Pancreatic Cancer. Noncoding RNA 2024, 10, 29. [Google Scholar] [CrossRef]
  144. Marin, A.M.; Mattar, S.B.; Amatuzzi, R.F.; Chammas, R.; Uno, M.; Zanette, D.L.; Aoki, M.N. Plasma Exosome-Derived microRNAs as Potential Diagnostic and Prognostic Biomarkers in Brazilian Pancreatic Cancer Patients. Biomolecules 2022, 12, 769. [Google Scholar] [CrossRef]
  145. Nesteruk, K.; Levink, I.J.M.; de Vries, E.; Visser, I.J.; Peppelenbosch, M.P.; Cahen, D.L.; Fuhler, G.M.; Bruno, M.J. Extracellular vesicle-derived microRNAs in pancreatic juice as biomarkers for detection of pancreatic ductal adenocarcinoma. Pancreatology 2022, 22, 626–635. [Google Scholar] [CrossRef]
  146. Wang, L.; Wu, J.; Ye, N.; Li, F.; Zhan, H.; Chen, S.; Xu, J. Plasma-Derived Exosome MiR-19b Acts as a Diagnostic Marker for Pancreatic Cancer. Front. Oncol. 2021, 11, 739111. [Google Scholar] [CrossRef] [PubMed]
  147. Wang, X.; Luo, G.; Zhang, K.; Cao, J.; Huang, C.; Jiang, T.; Liu, B.; Su, L.; Qiu, Z. Hypoxic Tumor-Derived Exosomal miR-301a Mediates M2 Macrophage Polarization via PTEN/PI3Kgamma to Promote Pancreatic Cancer Metastasis. Cancer Res. 2018, 78, 4586–4598, Erratum in Cancer Res. 2020, 80, 922. [Google Scholar] [CrossRef]
  148. Nasimi Shad, A.; Fanoodi, A.; Maharati, A.; Akhlaghipour, I.; Moghbeli, M. Molecular mechanisms of microRNA-301a during tumor progression and metastasis. Pathol. Res. Pract. 2023, 247, 154538. [Google Scholar] [CrossRef]
  149. Li, Y.; Vandenboom, T.G., 2nd; Wang, Z.; Kong, D.; Ali, S.; Philip, P.A.; Sarkar, F.H. miR-146a suppresses invasion of pancreatic cancer cells. Cancer Res. 2010, 70, 1486–1495. [Google Scholar] [CrossRef] [PubMed]
  150. Chang, C.H.; Pauklin, S. Extracellular vesicles in pancreatic cancer progression and therapies. Cell Death Dis. 2021, 12, 973. [Google Scholar] [CrossRef] [PubMed]
  151. Forner, A.; Reig, M.; Bruix, J. Hepatocellular carcinoma. Lancet 2018, 391, 1301–1314. [Google Scholar] [CrossRef]
  152. Pinero, F.; Dirchwolf, M.; Pessoa, M.G. Biomarkers in Hepatocellular Carcinoma: Diagnosis, Prognosis and Treatment Response Assessment. Cells 2020, 9, 1370. [Google Scholar] [CrossRef]
  153. Huang, P.S.; Liao, C.J.; Huang, Y.H.; Yeh, C.T.; Chen, C.Y.; Tang, H.C.; Chang, C.C.; Lin, K.H. Functional and Clinical Significance of Dysregulated microRNAs in Liver Cancer. Cancers 2021, 13, 5361. [Google Scholar] [CrossRef]
  154. Tian, X.P.; Wang, C.Y.; Jin, X.H.; Li, M.; Wang, F.W.; Huang, W.J.; Yun, J.P.; Xu, R.H.; Cai, Q.Q.; Xie, D. Acidic Microenvironment Up-Regulates Exosomal miR-21 and miR-10b in Early-Stage Hepatocellular Carcinoma to Promote Cancer Cell Proliferation and Metastasis. Theranostics 2019, 9, 1965–1979, Erratum in Theranostics 2021, 11, 6522–6523. [Google Scholar] [CrossRef]
  155. Cao, L.Q.; Yang, X.W.; Chen, Y.B.; Zhang, D.W.; Jiang, X.F.; Xue, P. Exosomal miR-21 regulates the TETs/PTENp1/PTEN pathway to promote hepatocellular carcinoma growth. Mol. Cancer 2019, 18, 148, Erratum in Mol. Cancer 2020, 19, 59. [Google Scholar] [CrossRef] [PubMed]
  156. Zhou, Y.; Ren, H.; Dai, B.; Li, J.; Shang, L.; Huang, J.; Shi, X. Hepatocellular carcinoma-derived exosomal miRNA-21 contributes to tumor progression by converting hepatocyte stellate cells to cancer-associated fibroblasts. J. Exp. Clin. Cancer Res. 2018, 37, 324, Erratum in J. Exp. Clin. Cancer Res. 2022, 41, 359. [Google Scholar] [CrossRef] [PubMed]
  157. Yang, Y.; Mao, F.; Guo, L.; Shi, J.; Wu, M.; Cheng, S.; Guo, W. Tumor cells derived-extracellular vesicles transfer miR-3129 to promote hepatocellular carcinoma metastasis by targeting TXNIP. Dig. Liver Dis. 2021, 53, 474–485. [Google Scholar] [CrossRef]
  158. Moh-Moh-Aung, A.; Fujisawa, M.; Ito, S.; Katayama, H.; Ohara, T.; Ota, Y.; Yoshimura, T.; Matsukawa, A. Decreased miR-200b-3p in cancer cells leads to angiogenesis in HCC by enhancing endothelial ERG expression. Sci. Rep. 2020, 10, 10418. [Google Scholar] [CrossRef]
  159. Sorop, A.; Constantinescu, D.; Cojocaru, F.; Dinischiotu, A.; Cucu, D.; Dima, S.O. Exosomal microRNAs as Biomarkers and Therapeutic Targets for Hepatocellular Carcinoma. Int. J. Mol. Sci. 2021, 22, 4997. [Google Scholar] [CrossRef]
  160. Thakral, S.; Ghoshal, K. miR-122 is a unique molecule with great potential in diagnosis, prognosis of liver disease, and therapy both as miRNA mimic and antimir. Curr. Gene Ther. 2015, 15, 142–150. [Google Scholar] [CrossRef]
  161. Gramantieri, L.; Giovannini, C.; Piscaglia, F.; Fornari, F. MicroRNAs as Modulators of Tumor Metabolism, Microenvironment, and Immune Response in Hepatocellular Carcinoma. J. Hepatocell. Carcinoma 2021, 8, 369–385. [Google Scholar] [CrossRef] [PubMed]
  162. Lin, X.J.; Fang, J.H.; Yang, X.J.; Zhang, C.; Yuan, Y.; Zheng, L.; Zhuang, S.M. Hepatocellular Carcinoma Cell-Secreted Exosomal MicroRNA-210 Promotes Angiogenesis In Vitro and In Vivo. Mol. Ther. Nucleic Acids 2018, 11, 243–252. [Google Scholar] [CrossRef]
  163. Paluschinski, M.; Loosen, S.; Kordes, C.; Keitel, V.; Kuebart, A.; Brandenburger, T.; Scholer, D.; Wammers, M.; Neumann, U.P.; Luedde, T.; et al. Extracellular Vesicles as Markers of Liver Function: Optimized Workflow for Biomarker Identification in Liver Disease. Int. J. Mol. Sci. 2023, 24, 9631. [Google Scholar] [CrossRef] [PubMed]
  164. Xue, X.; Zhao, Y.; Wang, X.; Qin, L.; Hu, R. Development and validation of serum exosomal microRNAs as diagnostic and prognostic biomarkers for hepatocellular carcinoma. J. Cell. Biochem. 2019, 120, 135–142. [Google Scholar] [CrossRef] [PubMed]
  165. Karimkhanloo, H.; Mohammadi-Yeganeh, S.; Hadavi, R.; Koochaki, A.; Paryan, M. Potential role of miR-214 in beta-catenin gene expression within hepatocellular carcinoma. Mol. Biol. Rep. 2020, 47, 7429–7437. [Google Scholar] [CrossRef]
  166. Xiong, X.; Yuan, J.; Zhang, N.; Zheng, Y.; Liu, J.; Yang, M. Silencing of lncRNA PVT1 by miR-214 inhibits the oncogenic GDF15 signaling and suppresses hepatocarcinogenesis. Biochem. Biophys. Res. Commun. 2020, 521, 478–484. [Google Scholar] [CrossRef] [PubMed]
  167. Boonkaew, B.; Satthawiwat, N.; Pinjaroen, N.; Chuaypen, N.; Tangkijvanich, P. Circulating Extracellular Vesicle-Derived microRNAs as Novel Diagnostic and Prognostic Biomarkers for Non-Viral-Related Hepatocellular Carcinoma. Int. J. Mol. Sci. 2023, 24, 16043. [Google Scholar] [CrossRef]
  168. Ye, D.; Zhang, T.; Lou, G.; Liu, Y. Role of miR-223 in the pathophysiology of liver diseases. Exp. Mol. Med. 2018, 50, 1–12. [Google Scholar] [CrossRef]
  169. Banales, J.M.; Marin, J.J.G.; Lamarca, A.; Rodrigues, P.M.; Khan, S.A.; Roberts, L.R.; Cardinale, V.; Carpino, G.; Andersen, J.B.; Braconi, C.; et al. Cholangiocarcinoma 2020: The next horizon in mechanisms and management. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 557–588. [Google Scholar] [CrossRef]
  170. Fabris, L.; Sato, K.; Alpini, G.; Strazzabosco, M. The Tumor Microenvironment in Cholangiocarcinoma Progression. Hepatology 2021, 73, 75–85. [Google Scholar] [CrossRef]
  171. Wang, J.; Shi, R.; Yin, Y.; Luo, H.; Cao, Y.; Lyu, Y.; Luo, H.; Zeng, X.; Wang, D. Clinical significance of small extracellular vesicles in cholangiocarcinoma. Front. Oncol. 2024, 14, 1334592. [Google Scholar] [CrossRef]
  172. Han, J.Y.; Ahn, K.S.; Kim, Y.H.; Kim, T.S.; Baek, W.K.; Suh, S.I.; Kang, K.J. Circulating microRNAs as biomarkers in bile-derived exosomes of cholangiocarcinoma. Ann. Surg. Treat. Res. 2021, 101, 140–150. [Google Scholar] [CrossRef]
  173. Shigehara, K.; Yokomuro, S.; Ishibashi, O.; Mizuguchi, Y.; Arima, Y.; Kawahigashi, Y.; Kanda, T.; Akagi, I.; Tajiri, T.; Yoshida, H.; et al. Real-time PCR-based analysis of the human bile microRNAome identifies miR-9 as a potential diagnostic biomarker for biliary tract cancer. PLoS ONE 2011, 6, e23584. [Google Scholar] [CrossRef]
  174. Prinz, C.; Frese, R.; Grams, M.; Fehring, L. Emerging Role of microRNA Dysregulation in Diagnosis and Prognosis of Extrahepatic Cholangiocarcinoma. Genes 2022, 13, 1479. [Google Scholar] [CrossRef]
  175. Olaizola, P.; Lee-Law, P.Y.; Arbelaiz, A.; Lapitz, A.; Perugorria, M.J.; Bujanda, L.; Banales, J.M. MicroRNAs and extracellular vesicles in cholangiopathies. Biochim. Biophys. Acta Mol. Basis Dis. 2018, 1864, 1293–1307. [Google Scholar] [CrossRef] [PubMed]
  176. Li, J.; Yao, L.; Li, G.; Ma, D.; Sun, C.; Gao, S.; Zhang, P.; Gao, F. miR-221 Promotes Epithelial-Mesenchymal Transition through Targeting PTEN and Forms a Positive Feedback Loop with beta-catenin/c-Jun Signaling Pathway in Extra-Hepatic Cholangiocarcinoma. PLoS ONE 2015, 10, e0141168. [Google Scholar] [CrossRef]
  177. Shen, L.; Chen, G.; Xia, Q.; Shao, S.; Fang, H. Exosomal miR-200 family as serum biomarkers for early detection and prognostic prediction of cholangiocarcinoma. Int. J. Clin. Exp. Pathol. 2019, 12, 3870–3876. [Google Scholar]
  178. Yang, F.; Ning, Z.; Ma, L.; Liu, W.; Shao, C.; Shu, Y.; Shen, H. Exosomal miRNAs and miRNA dysregulation in cancer-associated fibroblasts. Mol. Cancer 2017, 16, 148. [Google Scholar] [CrossRef]
  179. Cao, L.; Hong, J.; Wu, J. Potential of extracellular vesicles and exosomes as diagnostic markers for cholangiocarcinoma. Hepatobiliary Surg. Nutr. 2022, 11, 436–438. [Google Scholar] [CrossRef] [PubMed]
  180. Ota, Y.; Takahashi, K.; Otake, S.; Tamaki, Y.; Okada, M.; Aso, K.; Makino, Y.; Fujii, S.; Ota, T.; Haneda, M. Extracellular vesicle-encapsulated miR-30e suppresses cholangiocarcinoma cell invasion and migration via inhibiting epithelial-mesenchymal transition. Oncotarget 2018, 9, 16400–16417. [Google Scholar] [CrossRef]
  181. Li, L.; Piontek, K.; Ishida, M.; Fausther, M.; Dranoff, J.A.; Fu, R.; Mezey, E.; Gould, S.J.; Fordjour, F.K.; Meltzer, S.J.; et al. Extracellular vesicles carry microRNA-195 to intrahepatic cholangiocarcinoma and improve survival in a rat model. Hepatology 2017, 65, 501–514. [Google Scholar] [CrossRef] [PubMed]
  182. Mori, M.A.; Ludwig, R.G.; Garcia-Martin, R.; Brandao, B.B.; Kahn, C.R. Extracellular miRNAs: From Biomarkers to Mediators of Physiology and Disease. Cell Metab. 2019, 30, 656–673. [Google Scholar] [CrossRef] [PubMed]
Figure 1. List of EV-derived miRNAs up-(red) and downregulated (green) in GI cancers. Created in BioRender. (https://BioRender.com/p7c6ta2 accessed on 15 December 2025).
Figure 1. List of EV-derived miRNAs up-(red) and downregulated (green) in GI cancers. Created in BioRender. (https://BioRender.com/p7c6ta2 accessed on 15 December 2025).
Ijms 27 00010 g001
Table 1. Potential role of EV-derived miRNAs in gastric cancer.
Table 1. Potential role of EV-derived miRNAs in gastric cancer.
miRNAsClinical ApplicationBiological RoleSampleReferences
miR-10a-5pDiagnostic biomarkerCell proliferation, metastasis and apoptosis inhibitionSerum[104]
miR-18a-5pEarly diagnostic biomarkerCell proliferation and metastasisSerum[104,105]
miR-19bEarly diagnostic biomarkerHigh levels in GC; cancer progressionSerum[101,104]
miR-21Prognostic biomarkerCell proliferation, migration and apoptosis inhibitionCell culture[83,99]
miR-23aDiagnostic biomarkerPromotion of angiogenesis via the repression of PTENCell culture[96]
miR-106aEarly diagnostic biomarkerInduction of migration and apoptosis reduction via Smad7 Cell culture;
Serum
[100,101]
miR-149-5pDiagnostic and prognostic biomarkerPromotion of GC progression via the AKT1/mTOR pathwayPlasma[94]
miR-151-3pPrognostic biomarkerTumor cell proliferationCell culture[98]
miR-155-5pDrug resistancePromotion of chemoresistant phenotypeCell culture[95]
miR-181bEarly diagnostic biomarkerHigh levels in GC; cancer progressionSerum[105]
miR-215-5pDiagnostic biomarkerCell cycle, cell proliferation and metastasisSerum[104]
miR-335Early diagnostic biomarkerHigh levels in GC; cancer progressionSerum[105]
miR-451Prognostic biomarkerIncrease in Th17 polarization through exosome-mediated delivery to infiltrating T cellsSerum[102]
miR-493Prognostic biomarkerInduction of chemoresistance via MAD2L1 suppressionPeritoneal lavage fluid[103]
miR-519a-3pDiagnostic biomarkerMetastasis and angiogenesisCell culture[97]
miR-590-5pDiagnostic and Prognostic biomarkerHigh levels in GC; cancer progressionSerum[106]
miR-1246Early diagnostic biomarkerHigh levels in GC; cancer progressionSerum[107]
Table 2. Potential role of EV-derived miRNAs in CRC.
Table 2. Potential role of EV-derived miRNAs in CRC.
miRNAsClinical ApplicationBiological RoleSampleReferences
miR-19aDiagnostic and prognostic
biomarker
Cell proliferation and migration via TIA1Cancer tissue;
Serum
[109,128]
miR-21Diagnostic and drug resistance
biomarker
Induction of cell proliferation, metastasis and chemoresistance by targeting the PTEN/PI3K/AKTCell culture;
Serum
[114,121,122,123,124,129]
miR-23aDiagnostic biomarkerCell proliferation, invasion and metastasisSerum[114,122]
miR-29aDiagnostic biomarkerUpregulated in CRC and associated with disease progressionSerum[129]
miR-92aDiagnostic and prognostic
biomarker
High levels in stage I-II CRC patientsSerum; Plasma;
Cell culture
[113,114,115,125]
miR-106aPrognostic biomarkerInduction of metastasis through promotion of EMT and TAM2 infiltrationCell culture[116]
miR-145Diagnostic and prognostic
biomarker
Suppression of cell migration and invasion by targeting paxillinCell culture[110,111]
miR-146aPrognostic biomarkerPromotion of tumor metastasis through the CXCL12/CXCR7 axisCell culture[117]
miR-155-5pPrognostic biomarkerPromotion of tumor metastasis through the CXCL12/CXCR7 axisCell culture[117]
miR-181a-5pPrognostic biomarkerPromotion of tumor metastasis by activating hepatic stellate cells and remodelling TMEPlasma; Cell culture[126]
miR-183-5pDiagnostic and prognostic
biomarker
High levels in CRC; cancer progressionCell culture[112]
miR-200Diagnostic biomarkerPromotion of 5-fluorouracil resistantCell culture[130]
miR-203aPrognostic biomarkerInduction of metastasis by targeting PTEN-induced macrophage polarizationPlasma[118]
miR-320cPrognostic and therapeutic
biomarker
High levels in CRC; cancer progressionPlasma[127]
miR-934Prognostic biomarkerInduction of metastasis through macrophage M2 polarizationCancer tissue[119]
miR-1246Diagnostic biomarkerHigh levels in CRC; cancer progressionSerum; Plasma[114,122]
miR-1470Diagnostic biomarkerCell proliferation and metastasisSerum[131]
Table 3. Potential role of EV-derived miRNAs in PC.
Table 3. Potential role of EV-derived miRNAs in PC.
miRNAsClinical ApplicationBiological RoleSampleReferences
miR-10bDiagnostic biomarkerHigh levels in PC; cancer progressionPlasma[142]
miR-16Diagnostic biomarkerHigh levels in PC; cancer progressionPancreatic juice and serum[145]
miR-19bDiagnostic biomarkerHigh levels in PC; cancer progressionPlasma[144,146]
miR-21Diagnostic biomarkerTumor progression, metastasis, and poor prognosisPlasma; Pancreatic juice and serum[142,145]
miR-23b-2pDiagnostic biomarkerHigh levels in PC; cancer progressionPlasma; Serum[143]
miR-25Diagnostic biomarkerHigh levels in PC; cancer progressionPancreatic juice and serum[145]
miR-27a-3pDiagnostic biomarkerHigh levels in PC; cancer progressionPlasma; Serum[143]
miR-27bDiagnostic biomarkerHigh levels in PC; cancer progressionPlasma[144]
miR-30cDiagnostic biomarkerIncreased cell survival and proliferationPlasma[142]
miR-34aDiagnostic biomarkerSuppression cancer cell proliferation, migration and invasionCell culture[140]
miR-106bDrug resistanceInduction of gemcitabine resistanceCell culture[137]
miR-122-5pDiagnostic biomarkerHigh levels in PC; cancer progressionPlasma[142,144]
miR-146aDiagnostic and drug resistance
biomarker
Suppression of cancer cell invasionSerum;
Cell culture
[135,149]
miR-155Diagnostic biomarkerCell proliferation, invasion, and metastasisCell culture[150]
miR-181aDiagnostic biomarkerHigh levels in PC; cancer progressionPlasma[142]
miR-193a-3pDiagnostic biomarkerInhibition of cell proliferation and metastasisPlasma; Serum[143]
miR-203Diagnostic and prognostic
biomarker
Regulation of tumor cell proliferation via JAK-STATCell culture[141]
miR-205-5pDiagnostic biomarkerHigh levels in PC; cancer progressionPlasma[144]
miR-210Drug resistanceCell invasion and chemoresistanceCell culture[73]
miR-221Diagnostic biomarkerTumor growth and apoptosis resistance through PTEN and PDCD4Cell culture[138]
miR-221-3pDiagnostic biomarkerHigh levels in PC; cancer progressionPlasma; Serum[143,144]
miR-301a-3pPrognostic biomarkerPromotion of metastasis via the PTEN/PI3Kγ pathwayCell culture;[147,148]
miR-1246Diagnostic biomarkerHigh levels in PC; cancer progressionSerum; Saliva[86,135]
Table 4. Potential role of EV-derived miRNAs in HCC.
Table 4. Potential role of EV-derived miRNAs in HCC.
miRNAsClinical ApplicationBiological RoleSampleReferences
miR-10bDiagnostic biomarkerCell Proliferation and metastasisCell culture[154]
miR-15bDiagnostic biomarkerHigh levels in HCC; cancer progressionSerum[163]
miR-16Diagnostic biomarkerHigh levels in HCC; cancer progressionSerum[163]
miR-21Diagnostic and prognostic
biomarker
High levels in HCC; cancer progressionCell culture; Serum[153,154,155,156,159]
miR-26aDiagnostic biomarkerHigh levels in HCC; cancer progressionSerum[163]
miR-106aPrognostic biomarkerCell proliferation and invasion via c-Jun pathwaySerum[164]
miR-122Diagnostic biomarkerCell cycle progression, tumor invasionSerum[153,160,161];
miR-150Diagnostic biomarkerHigh levels in HCC; cancer progressionSerum[163]
miR-199Diagnostic biomarkerInhibition of cell proliferation and migrationCancer tissues[153]
miR-200b-3pDiagnostic biomarkerInhibition of HCC growth Cell culture[158]
miR-210Diagnostic biomarkerPromotion of angiogenesisCell culture[162]
miR-214Diagnostic biomarkerInhibition of cell proliferationCell culture[165,166]
miR-223Diagnostic biomarkerHigh levels in HCC; cancer progressionSerum; Plasma[167,168]
miR-3129Prognostic biomarkerPromotion of metastasis formation by targeting TXNIPPlasma[157]
Table 5. Potential role of EV-derived miRNAs in CCA.
Table 5. Potential role of EV-derived miRNAs in CCA.
miRNAsClinical ApplicationBiological RoleSampleReferences
miR-9Diagnostic biomarkerHigh levels in CCA; cancer progressionBile[173]
miR-21Diagnostic biomarkerTumor progression and poor prognosisBile; Serum[172,174]
miR-30eDiagnosis biomarkerCell proliferation and invasionCell culture[179,180]
miR-34cDiagnostic biomarkerCell growth, invasion, and resistanceCell culture[179]
miR-141Diagnostic biomarkerHigh levels in CCA; cancer progressionSerum[177]
miR-195Diagnostic biomarkerInhibition of EMTCell culture[181]
miR-200cDiagnostic biomarkerRegulation of EMT by targeting ZEB1 and ZEB2Serum[177,179]
miR-221Diagnostic biomarkerCell cycle progression and apoptosis inhibition by targeting p27Kip1 and pro-apoptotic proteinsCell culture[176]
miR-222Diagnostic biomarkerTumor progression and poor prognosisSerum[175]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Nardozi, D.; Lucarini, V.; Angiolini, V.; Feverati, N.; Benvenuto, M.; Focaccetti, C.; Del Conte, L.; Buccitti, O.; Palumbo, C.; Cifaldi, L.; et al. Extracellular Vesicle-Derived microRNAs: Novel Non-Invasive Biomarkers for Gastrointestinal Malignancies. Int. J. Mol. Sci. 2026, 27, 10. https://doi.org/10.3390/ijms27010010

AMA Style

Nardozi D, Lucarini V, Angiolini V, Feverati N, Benvenuto M, Focaccetti C, Del Conte L, Buccitti O, Palumbo C, Cifaldi L, et al. Extracellular Vesicle-Derived microRNAs: Novel Non-Invasive Biomarkers for Gastrointestinal Malignancies. International Journal of Molecular Sciences. 2026; 27(1):10. https://doi.org/10.3390/ijms27010010

Chicago/Turabian Style

Nardozi, Daniela, Valeria Lucarini, Valentina Angiolini, Nicole Feverati, Monica Benvenuto, Chiara Focaccetti, Letizia Del Conte, Olga Buccitti, Camilla Palumbo, Loredana Cifaldi, and et al. 2026. "Extracellular Vesicle-Derived microRNAs: Novel Non-Invasive Biomarkers for Gastrointestinal Malignancies" International Journal of Molecular Sciences 27, no. 1: 10. https://doi.org/10.3390/ijms27010010

APA Style

Nardozi, D., Lucarini, V., Angiolini, V., Feverati, N., Benvenuto, M., Focaccetti, C., Del Conte, L., Buccitti, O., Palumbo, C., Cifaldi, L., Ferretti, E., Bei, R., & Masuelli, L. (2026). Extracellular Vesicle-Derived microRNAs: Novel Non-Invasive Biomarkers for Gastrointestinal Malignancies. International Journal of Molecular Sciences, 27(1), 10. https://doi.org/10.3390/ijms27010010

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

Article Metrics

Back to TopTop