Next Article in Journal
The Prevalence of TET2 Gene Mutations in Patients with BCR-ABL-Negative Myeloproliferative Neoplasms (MPN): A Systematic Review and Meta-Analysis
Next Article in Special Issue
The TGF-β Pathway: A Pharmacological Target in Hepatocellular Carcinoma?
Previous Article in Journal
Targeted Therapies and Immune Checkpoint Inhibitors in Primary CNS Lymphoma
Previous Article in Special Issue
Evolving Treatment of Advanced Hepatocellular Carcinoma in the Asia–Pacific Region: A Review and Multidisciplinary Expert Opinion
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Extracellular Vesicles in Disease Progression and Detection of Hepatocellular Carcinoma

1
California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, CA 90095, USA
2
Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
3
Department of Surgery, University of California, Los Angeles, CA 90095, USA
4
Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90048, USA
5
Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
6
Division of Cancer Biology and Therapeutics, Departments of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
7
Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
8
Comprehensive Transplant Center Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
*
Author to whom correspondence should be addressed.
Cancers 2021, 13(12), 3076; https://doi.org/10.3390/cancers13123076
Submission received: 1 June 2021 / Revised: 15 June 2021 / Accepted: 17 June 2021 / Published: 20 June 2021

Abstract

:

Simple Summary

Extracellular vesicles (EVs) are particles naturally released from cells and mediate intercellular communication. Recently, emerging studies have shown that EVs play a crucial role in regulating progression of hepatocellular carcinoma (HCC), which is one of the leading causes of cancer-related death worldwide. With the advances of technologies in isolating EVs from patients’ blood, EVs are regarded as promising biomarkers for detecting HCC at an earlier stage. This review provides an overview of the current EVs isolation methods, the biological roles of EVs in mediating disease progression, and the feasibility of EVs’ use for detection of HCC.

Abstract

Hepatocellular carcinoma (HCC) is the most common primary liver malignancy and one of the leading causes of cancer-related death worldwide. Despite the improvements in surveillance and treatment, the prognosis of HCC remains poor. Extracellular vesicles (EVs) are a heterogeneous group of phospholipid bilayer-enclosed particles circulating in the bloodstream and mediating intercellular communication. Emerging studies have shown that EVs play a crucial role in regulating the proliferation, immune escape, and metastasis of HCC. In addition, because EVs are present in the circulation at relatively early stages of disease, they are getting attention as an attractive biomarker for HCC detection. Over the past decade, dedicated efforts have been made to isolate EVs more efficiently and make them useful tools in different clinical settings. In this review article, we provide an overview of the EVs isolation methods and highlight the role of EVs as mediators in the pathogenesis and progression of HCC. Lastly, we summarize the potential applications of EVs in early-stage HCC detection.

1. Introduction

Hepatocellular carcinoma (HCC) is the most common primary liver malignancy (>80% cases) and ranks sixth for cancer incidence and third for cancer-related death worldwide [1,2]. Main risk factors for hepatocarcinogenesis include chronic hepatitis B virus (HBV) and hepatitis C virus (HCV) infection, fatty liver disease and diabetes, alcohol consumption, and liver cirrhosis caused by any disease [2]. Despite the improvements in surveillance programs and treatment algorithms, the overall survival of patients with HCC remains dismal, with a 5-year overall survival rate of approximately 20% [3]. In light of this, biomarkers that could sensitively detect early-stage HCC have been under investigation [4]. In parallel, significant research efforts are undergoing to identify the mechanisms involved in HCC pathogenesis to prevent tumor progression and metastasis [5].
Extracellular vesicles (EVs) are a heterogeneous group of phospholipid bilayer-enclosed particles that are released by both tumor and normal cells [6]. Among the three major tumor liquid biopsy approaches, i.e., circulating tumor DNA [7], circulating tumor cells [8,9], and EVs [10], EVs are present in circulation at relatively early stages of disease and persist across all its stages [11]. Furthermore, the quantity of EVs is 2.3- to 3.0-fold higher in HCC cases than in cirrhosis controls [12,13], making them ideal biomarkers for non-invasive diagnosis of liver cancer. In addition, EVs are crucial mediators of cell-to-cell communications through transfer of functional cargoes from one cell to another [11]. As such, profiling the distinctive compositions of proteins, lipids, and nucleic acid in EVs may provide valuable information for understanding the pathological process of cancer.
Over the past decade, there have been emerging studies investigating the roles of EVs in HCC tumorigenesis. In this review, we summarize the EVs detection technology, role of EVs in cancer cell proliferation, angiogenesis, and metastasis of HCC, and the feasibility of EVs’ use as a diagnostic biomarker for HCC.

2. Nomenclature

Endorsed by the International Society for Extracellular Vesicles (ISEV), EVs are defined as lipid bilayer-delimited particles naturally released from cells that do not contain a functional nucleus for replication [6]. EVs are present in almost all the main body fluids and tissues and contain proteins, lipids, DNAs, messenger RNAs (mRNAs), microRNAs (miRNAs), and other non-coding RNAs, such as circular RNAs (circRNAs) and long non-coding RNAs (lncRNA) for cell-to-cell communication [11]. Historically, EVs are categorized into several subtypes, such as exomeres (size: ~35 nm), exosomes (size: 60–20 nm), microvesicles (size: 100 nm–1 µm), and large oncosomes (size: 1–10 µm), by size and biogenesis [14]. However, according to Minimal Information for Studies of Extracellular Vesicles 2018 (MISEV 2018) guidelines, these formerly used terms should be avoided due to their inaccurate definitions of size or biogenesis [6]. Currently, it is recommended to classify EVs by (1) physical characteristics, such as size or density with specific ranges defined, e.g., <200 nm (small EVs), or >200 nm (medium/large EVs); (2) biochemical and molecular components, e.g., epithelial cell adhesion molecule (EpCAM)+/CD147+ EVs; and (3) descriptions of conditions or cell of origin, e.g., HCC-derived EVs [6].
After being released from parental cells, EVs migrate to recipient cells for subsequent intercellular communication [11]. Numerous studies have shown that EVs are involved in the pathogenesis, differentiation, proliferation, and metastasis of HCC. Investigation of the molecular contents in EVs from different disease states would therefore be helpful to understand the landscape of HCC.

3. EVs isolation method

In general, there are at least six different types of EVs Isolation Methods (Figure 1): (1) ultracentrifugation, (2) filtration, (3) size exclusion chromatography, (4) precipitation, (5) immunoaffinity capture, and (6) microfluidics. There is no single optimal method for EVs isolation [6]. These methods can be used either alone or in combination to achieve the desired recovery and purity for downstream applications.

3.1. Ultracentrifugation

Ultracentrifugation is the most commonly used method for EVs isolation [15]. EVs are isolated by sequential centrifugations at increasing centrifugal forces, based on size and density. Using low-speed centrifugation, cells, platelets, apoptotic bodies, and cell debris are first eliminated from the EVs. Larger EVs and smaller EVs are then separated commonly at the speeds of 10,000–20,000 g and 100,000–120,000 g, respectively [16,17,18]. During ultracentrifugation, contaminants, including protein aggregates and lipoprotein particles, are also sedimented [17,18]. Additional steps of density gradient ultracentrifugation could be adopted to further purify the EVs [19,20,21]. However, ultracentrifugation is time-consuming and laborious and thus is unlikely to be applicable for routine use in clinical applications [16,17,18].

3.2. Filtration

Size-based separation of EVs from other non-EV particles can be achieved by filtration and size exclusion chromatography. The filter allows particles smaller than the pore to pass through while particles larger than the pore remain on the filter. Using a series of membrane filters with different pore diameters enables isolation of EVs fractions with specified sizes [22]. Filtration is amenable to clinical applications as it is less time-consuming and requires no special instrumentation. However, clogging and trapping of vesicles on the filter may lead to loss of EVs. Applying forces on particles to pass through filters can reduce the time of EVs isolation but may lead to deformation and breakup of large vesicles [23,24].

3.3. Size Exclusion Chromatography

Size exclusion chromatography, also known as gel filtration, separates molecules based on their size as they pass through a resin packed in a column [23,25]. The resin consists of porous beads with pores of a specific size that only allow smaller particles, such as proteins, to enter and thereby slow down their movement through the column due to increased path length. EVs are larger than the pore size and, therefore, flow through the column more quickly than soluble proteins. Therefore, the method can distinctly separate most protein contaminants from EVs [23,25]. Due to little pressure applied during the process, it preserves the structure, integrity, and biological functions of EVs [25,26,27,28]. However, there are some limitations: (1) The throughput is low, and (2) the resulting EV fraction is diluted and may require additional steps of concentration [25,29]. Some automated platforms have been developed to enable rapid and less laborious EVs isolation suitable for clinical application [30].

3.4. Precipitation

Precipitation of EVs in polyethylene glycol (PEG) solutions is, after ultracentrifugation, the second most common method for EVs isolation [31]. PEG is a superhydrophilic polymer, which occupies the water and concentrates other, less soluble, particles, including EVs, to the point of exceeding their solubility limit and precipitating [32]. The precipitate can then be pelleted by low-speed centrifugation [24]. Several commercial products have been developed based on precipitation to isolate EVs from biofluids, e.g., ExoQuick-TC™ Exosome Precipitation Solution (System Biosciences, Palo Alto, CA, USA), and Total Exosome Isolation Kit/Reagent (Invitrogen, Waltham, MA, USA). The method is quick, simple and allows for concurrent processing of numerous samples [31,32]. However, the major concern of this method is that non-EV proteins, such as lipoproteins, are co-precipitated along with EVs [33,34]. In addition, the presence of the polymer in purified EVs may interfere with down-stream analyses [33,34]. Therefore, this application is appropriate in samples thought to have sufficiently abundant EVs to reduce bias from contaminants. Additional steps before and after precipitation, including ultracentrifugation, filtration, or size exclusion chromatography, can be incorporated to reduce non-EV contaminants and PEG polymer [23,31].

3.5. Immunoaffinity Capture

Immunoaffinity capture isolates EVs based on the interaction between antibodies and surface proteins on EVs. Antibodies targeting surface proteins present on EVs are utilized to positively or negatively select specific subpopulations of EVs. These antibodies can be attached to magnetic beads [35,36,37] or microfluidic devices [38,39] (Section 3.6.), according to the desired downstream analysis. Therefore, the major benefit of this method is higher specificity and purity than those of the methods relying on physical properties [40,41]. However, selectivity can lead to lower yields [18]. Additionally, the cost of antibodies may be considerable.

3.6. Microfluidics

Microfluidic devices are designed as a network of microchannels to purify EVs by immunoaffinity and/or by physical characteristics [42]. Compared with traditional isolation methods, microfluidic-based isolation is more rapid (30 min–2 h), requires lower amounts of samples (50 μL–500 μL), and yields high-purity EVs in general [31,42]. However, some devices only allow low sample input and may therefore limit the applications of downstream analysis, such as massive sequencing [31,39,42,43,44]. Recently, our research team developed a microfluidic device named EVClick Chip [38], which synergistically integrates (1) multimarker antibody cocktails for targeting HCC-derived EVs, (2) nanostructured substrates for increasing the surface interacting with circulating EVs, and (3) click chemistry-mediated EVs capture and disulfide cleavage-driven EVs release, to achieve purification of HCC-derived EVs with high recovery yield (82.7%) and excellent purity (90.2%). Most importantly, we demonstrated the potential application of EV Click Chip combined with downstream reverse-transcription droplet digital polymerase chain reaction (RT-ddPCR) analysis in detection of early-stage HCC.

4. EVs as Mediators of Chronic Liver Disease and HCC Progression

The majority of HCC diagnoses arise in patients with underlying cirrhosis, with up to 20% of HCC cases found in non-cirrhotic patients [2,45]. In a healthy liver, EVs are critical in mediating numerous signals among hepatocytes, stellate cells, and various immune cells (Kupffer cells, T and B cells, natural killer—NK—cells) to perform important functions and maintain a homeostatic state. Previous studies have demonstrated EVs play a role in the development of these predisposing liver diseases and, subsequently, in the development of HCC. EVs mediate the pathogenesis and progression process of HCC by regulating the microenvironment and multiple signaling pathways in both cancer and surrounding normal cells [46,47,48]. The studies investigating EVs’ functions in HCC progression are summarized in Table 1 and Figure 2.

4.1. EVs in Promoting Progression of Chronic Liver Disease

The majority of HCC cases can be attributed to chronic viral hepatitis secondary to HBV and HCV. It has been shown that inter- and intra-cellular modulation via EVs in chronic hepatitis B and hepatitis C lead to viral propagation [49,50], fibrogenesis [51,52], and dysregulation of the immune system [50,53] through various mechanisms. For example, miR-19a in EVs derived from HCV-infected hepatocytes initiates fibrosis by activating surrounding hepatic stellate cells through the transforming growth factor beta (TGF-β) signaling pathway [51]. In addition, studies also indicated that HBV-infected hepatocytes produce EVs to suppress the expression of interleukin (IL)-21 in T cells [53] and downregulate nuclear factor kappa B (NF-κB) in NK cells [50]. This creates a microenvironment in the cirrhotic or chronically infected liver that is primed for genetic mutations and cellular dysregulation, giving the potential for the development of HCC.
There are numerous mechanisms by which EVs promote fibrosis and inflammatory processes in metabolic liver disease. At the cellular level, fatty liver disease is caused by hepatocyte dysfunction and death due to the toxic accumulation of intra-cellular lipids and the stimulation of inflammatory and fibrogenic pathways, with EVs playing a key role [54]. Hepatocytes exposed to elevated levels of lipid, such as palmitate, had increased secretion of EVs containing tumor necrosis factor (TNF)-related apoptosis inducing ligand [55], sphingosine 1-phosphate [56], miRNAs [57,58], C-X-C motif chemokine ligand 10 (CXCL10) [59], and ceramides [60,61], leading to activation of macrophages and chemotaxis. Lipotoxic conditions also release EVs containing integrin β1, which promotes monocyte adhesion to liver sinusoidal endothelial cells, resulting in inflammation and fibrosis [62]. These findings demonstrate the roles of EVs in the progression of metabolic liver disease through inflammation and fibrosis, ultimately leading to a liver microenvironment at risk for the development of HCC.

4.2. EVs in Regulating Proliferation of HCC

Several studies have demonstrated that EVs could directly regulate the growth of HCC through different pathways [63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79]. Gai et al. found that a serum protein marker involved in the tumorigenesis and metastasis of HCC, Golgi membrane protein 1 (GOLM1), was significantly enriched in HCC-derived EVs. [63] They observed that the EV-derived GOLM1 promoted HCC proliferation, migration, and invasion and activated the glycogen synthase kinase 3β (GSK-3β)/matrix metalloproteinase-1 and -9 (MMP-1 and MMP-9) of recipient cells. miRNAs also modulate essential processes in cell proliferation at the post-transcriptional level. For example, miR-93, miR-224, and miR-665 from HCC-derived EVs have been proven to promote HCC proliferation [67,71,72], while miR-9-3p, miR-638, miR-718, and miR-744 have the opposite effect [64,68,69,70]. In addition to the effect of HCC-derived EVs on HCC proliferation, it was reported that EVs secreted from HCC cells promoted proliferation and suppressed apoptosis of normal hepatocytes through transferring long intergenic non-protein coding RNA, regulator of reprogramming (linc-ROR) [80]. After being cocultured with HCC-derived EVs for more than 30 days, the expression of stem cell-related proteins, such as OCT4, NANOG, SRY-box 2 (SOX2), P53, and CD133, in hepatocytes notably increased and these hepatic cells could still be subcultured compared with those not cocultured with HCC-derived EVs [80]. These results indicated that HCC-derived EVs-induced stem cell-like phenotype of normal hepatocytes and may lead to disease progression.
Interestingly, Tian et al. first described that an acidic tumor microenvironment, attributed to the increased glycolysis in cancer cells [81], increases the levels of miR-10b and miR-21 in HCC-derived EVs compared with those produced at normal pH conditions [65]. In this study, miR-10b and miR-21 were proven to promote HCC proliferation and metastasis both in vitro and in vivo. These results highlight the role of EVs in tumor progression in response to the changing microenvironment.
EVs from surrounding stromal cells, such as tumor-associated macrophages (TAMs) [82] and cancer-associated fibroblasts (CAFs) [83], participate in the regulation of HCC progression. In a recent study, Wang et al. reported a significantly lower level of miR-125a/b in TAM-derived EVs. Subsequent functional studies showed miR-125a/b in TAM-derived EVs suppress proliferation and stem cell properties of HCC in vitro [82]. Additional studies indicated that compared with EVs derived from the para-cancer fibroblasts, the CAF-derived EVs from the same HCC patients had a significantly lower level of miR-320a [83]. miR-320a in these CAF-derived EVs acts as a suppressor of HCC proliferation and migration by directly downregulating the PBX3 oncogene.

4.3. EVs in Regulating Angiogenesis in HCC

In addition to tumor proliferation, EVs also modulate angiogenesis in HCC [84,85,86,87,88,89,90,91]. As a hypervascular tumor, HCC requires the formation of new blood vessels for growth. Revealing the mechanism of angiogenesis through EVs might thus help identify potential therapeutic targets to inhibit HCC progression. As proven by in vitro human umbilical vein endothelial cells (HUVECs) tube-formation assay, many molecular cargoes in EVs are key players in angiogenesis [84,85,86,87,88,89]. For example, lysyl oxidase like 4 (LOXL4) could promote angiogenesis and metastasis both in vitro and in vivo through activating the focal adhesion kinase (FAK)/Src pathway [84]. In cell line studies, miR-155, lncRNA-H19, and circRNA-100338 from HCC-derived EVs are associated with angiogenesis [85,87,88]. On the other hand, miR-200b-3p and miR-451a suppress angiogenesis by downregulating ERG and LPIN1, respectively [86,89]. Vascular endothelial growth factor (VEGF) is a signaling protein that directly induces the growth of hepatocytes, cancer cells, and epithelial cells and leads to abnormal vascular structures in HCC. Fu et al. demonstrated that in EVs secreted from a multidrug-resistant HCC cell line, Bel/5-FU, miR-32-5p was the most overexpressed miRNA [90]. miR-32-5p raised the level of VEGF in vitro and increased the microvascular density of xenograft tumors in vivo.

4.4. EVs in Promoting Metastasis, Immune Escape, and Recurrence in HCC

Several molecules in EVs participate in epithelial–mesenchymal transition (EMT), extracellular matrix (ECM) remodeling, immune regulation, and cancer cell adhesion to promote HCC metastasis [66,74,75,90,92,93,94,95,96,97,98,99,100,101,102,103,104]. EMT is a process during which cells transform from a polarized, epithelial to a mesenchymal phenotype [105]. During EMT, cells lose polarity, decrease cell–cell and cell–ECM adhesions and therefore acquire increased motility and invasive properties [105]. miR-32-5p and miR-92a-3p in HCC-derived EVs suppress phosphatase and tensin homolog (PTEN) and activate the phosphoinositide 3-kinase (PI3K)/Akt (Protein kinase B, PKB) pathway to induce EMT and metastasis in vivo [66,90]. Similarly, other oncogenic proteins in HCC-derived EVs, MET and caveolins (CAV1 and CAV2), are also involved in the PI3K/Akt and mitogen-activated protein kinase (MAPK)/extracellular-signal-regulated kinase (ERK) pathways to promote migration and invasion of immortalized hepatocytes [98]. Higher expression of miR-1247-3p in HCC-derived EVs induces CAF activation and increases secretion of IL-6 and IL-8, thereby creating an inflammatory microenvironment [92]. Importantly, the authors demonstrated the activated CAF, in turn, further promoted EMT and metastasis of HCC in vitro and in vivo [92].
Previous studies have suggested the oncogenic roles of HCC-derived EVs in regulation of several immune cells [75,93,94,95,96,97]. The 14-3-3ζ protein is highly expressed in HCC, and impairs the anti-tumor activity of tumor-infiltrating T cells via HCC-derived EVs [93]. TAMs are one of the immune cells crucial in creation of the immunosuppressive tumor microenvironment [106]. miR-146a-5p and lnc-TUC339 are enriched in HCC-derived EVs and proven to promote M2-polarization of TAMs [75,96], which can further result in T cell exhaustion [96]. In addition, Ye et al. found that the high mobility group box 1 (HMGB1) protein in EVs promoted T cell immunoglobulin and mucin domain 1 (TIM-1)+ regulatory B cell expansion and suppressed CD8+ T cell proliferation as well [94]. Lastly, it was shown that circ-UHRF1 in HCC-derived EVs inhibited interferon gamma (IFN-γ) and TNF-α secretions from NK cells by suppressing miR-449c-5p and upregulating TIM-3 [97]. All these studies provide strong evidence indicating HCC-derived EVs could induce immune escape and promote metastasis.
Tumor intravasation represents a critical step for HCC metastasis and relies on the interaction of cancer and endothelial cells [107]. Of note, Fang et al. reported that miR-103 in HCC-derived EVs increased vascular permeability in vitro and in vivo by suppressing the expression of VE-cadherin, p120-catenin, and zonula occludens-1, which are endothelial adhesion molecules important in maintaining cell–cell junctions [101]. Interruption of the junction integrity eventually promotes liver and lung metastases. Similarly, miRNA-25-5p in HCC-derived EVs promotes trans-endothelial motility of HCC cells and causes tumor self-seeding in vivo [100]. Once cancer cells enter and survive in the circulation, attachment to the endothelial lining of microvasculature is essential for extravasation and consequent metastasis [108]. Fu et al. demonstrated SMAD family member 3 (SMAD3) in HCC-derived EVs-promoted adhesion of HCC cells to endothelial cells in vitro and observed a higher level of SMAD3 in EVs from patients with advanced stage HCC [99]. A non-coding RNA, lnc-H19, in EVs secreted from CD90+ HCC cells also facilitates adhesion of HCC cells to endothelium [87].
Lastly, evidence indicates EVs may be responsible for HCC recurrence after surgical treatment. By injecting HCC-derived EVs or phosphate-buffered saline (PBS) into mice of which engrafted tumors were completely resected, Chen et al. showed 100% of the mice (5/5) in the EVs injection group experienced intrahepatic recurrence, compared with the recurrence rate of only 40% (2/5) in the PBS injection group [109]. However, the molecules in EVs participating in the process of recurrence were not investigated. In another study, Nakano et al. isolated EVs from HCC patients receiving liver transplantation (LT) and demonstrated that the patients without posttransplant HCC recurrence had lower EVs-derived miR-92b at 1 month after LT compared with those with posttransplant HCC recurrence [95]. The authors also proved that miR-92b derived from HCC EVs suppresses the cytotoxicity of NK cells by, which may cause immune escape of HCC cells, followed by the induction of posttransplant recurrence [95].
Table 1. EVs as mediators of HCC progression.
Table 1. EVs as mediators of HCC progression.
Name of the Cargo in EVs.Cargo
Type
Level
in HCC EVs 1
EVs isolation Method 2Function of the CargoMechanism of the CargoRef
HCC cell proliferation
GOLM1ProteinDifferential ultracentrifugationPromotes HCC cell proliferation, migration, and invasion in vitroActivates the GSK-3β/MMP-1 and -9 pathway[63]
miR-9-3pmiRNADifferential ultracentrifugationSuppresses HCC cell proliferation in vitroSuppresses the ERK1/2 pathway and HBGF-5 expression[64]
miR-10b, miR-21
(cultured at acidic condition—pH 6.6)
miRNADifferential ultracentrifugation
Promotes HCC cell proliferation, migration, and invasion in vitro; promotes HCC growth and lung metastasis in vivo[65]
miR-92a-3pmiRNADifferential ultracentrifugationPromotes HCC cell proliferation, migration, invasion, and EMT in vitro, promotes EMT and metastasis in vivoSuppresses PTEN and activates the PI3K/AkT pathway[66]
miR-93miRNATotal Exosome Isolation Kit Promotes HCC cell proliferation and invasion in vitroSuppresses expression of TP53INP1, TIMP2, and CDKN1A[67]
miR-125a/b
(from TAM)
miRNA-ExoQuick™ Exosome Precipitation SolutionSuppresses HCC cell proliferation, migration, invasion, and stem cell properties in vitro Suppresses CD90 expression[82]
miR-224miRNATotal Exosome Isolation Kit Promotes HCC cell proliferation and invasion in vitroSuppresses GNMT expression[71]
miR-320a
(from CAF)
miRNA-Total Exosome Isolation Kit Suppresses HCC cells proliferation, migration and metastasis in vitro and in vivoSuppresses the PBX3/ERK1/2/CDK2 pathway[83]
miR-638miRNATotal Exosome Isolation Kit Suppresses HCC cell proliferation in vitro[68]
miR-665miRNADifferential ultracentrifugationPromotees HCC cell proliferation in vitro,
promotees HCC growth in vivo
Activates the MAPK/ERK pathway[72]
miR-718miRNADifferential ultracentrifugationSuppresseses HCC cell proliferation in vitro Suppresses HOXB8 expression[69]
miR-744miRNADifferential ultracentrifugationSuppresseses HCC cell proliferation and chemoresistance to sorafenib in vitroSuppresses PAX2 expression[70]
miR-1247-3pmiRNADifferential ultracentrifugationPromotes proliferation of CAF in vitro, the activated CAF further promotes HCC cell progression, migration, stem cell properties, EMT, and chemoresistance to sorafenib in vitro and in vivoSuppresses B4GALT3 to activate the NF-κB pathway[92]
lnc-EPC1-4lncRNADifferential ultracentrifugationSuppresses HCC cell proliferation and promotes HCC cell apoptosis[73]
lnc-FAL1lncRNAExoQuick-TC™ Exosome Precipitation SolutionPromotees HCC cell proliferation, migration, invasion, and EMT in vitroSuppresses miR-1236 to activate ZEB1 and AFP expression[74]
lnc-FAM72D-3lncRNADifferential ultracentrifugationPromotes HCC cell proliferation and suppresses HCC cell apoptosis[73]
lnc-TUC339lncRNADifferential ultracentrifugationPromotes proliferation and suppresses cell adhesion to extracellular matrix of HCC cell in vitro, suppresses phagocytic activity and promotes M2-polarization of macrophage in vitroMay be involved in several pathways to regulate macrophages[75]
SENP3-EIF4A1lncRNAExoQuick-TC™ Exosome Precipitation Solution Suppresses HCC cell proliferation and migration in vitro,
suppresses HCC growth in vivo
Suppresses miR-9-5p to activates ZFP36 expression[76]
circ-0051443circRNAExoQuick™ Exosome Precipitation SolutioSuppresses HCC cell proliferation and promotes HCC cell apoptosis in vitro, suppresses HCC growth in vivoActivates BAK1 expression[77]
circ-FBLIM1circRNADifferential ultracentrifugationPromotes HCC cell proliferation and glycolysis in vitro,
promotes HCC growth in vivo
Suppresses miR-338 to activate LRP6 expression[78]
circ-DB
(from adipocyte)
circRNA-Differential ultracentrifugationPromotes HCC cell proliferation and reduces DNA damage in vitro, promotes HCC growth in vivoSuppresses miR-34a and activates expression of USP7 and cyclin A2[79]
Angiogenesis
LOXL4ProteinDifferential ultracentrifugationPromotes angiogenesis, HCC cell migration and invasion in vitro, promotes liver and lung metastasis in vivoActivates the FAK/Src pathway[84]
miR-21miRNADifferential ultracentrifugationConverts hepatic stellate cells into to cancer-associated fibroblasts and promotes angiogenesis in vitro, promotes HCC growth and angiogenesis in vivoSuppresses PTEN and activates the PI3K/AkT pathway in hepatic stellate cells[91]
miR-32-5p
(from multidrug-resistant HCC cell line, Bel/5-FU)
miRNADifferential ultracentrifugationPromotes angiogenesis, HCC cell migration, invasion, and EMT, causes multidrug resistance in vitro, promotes angiogenesis and EMT, and causes 5-FU resistance in vivoSuppresses PTEN and activates the PI3K/Akt pathway[90]
miR-155
(cultured at hypoxic condition—1% O2)
miRNAExoQuick-TC™ Exosome Precipitation Solution Promotes angiogenesis in vitro
[85]
miR-200b-3pmiRNATotal Exosome Isolation KitSuppresses angiogenesis in vitroSuppresses ERG expression[86]
miR-451amiRNADifferential ultracentrifugationSuppresses cell proliferation and migration, promotes apoptosis of HCC cell and HUVEC in vitro, and suppresses angiogenesis in vitro and in vivo Suppresses LPIN1 expression[89]
lnc-H19
(from CD90+ HCC cell)
lncRNADifferential ultracentrifugationPromotes cell–cell adhesion of HCC cells and promotes angiogenesis in vitroActivates VEGF expression[87]
circ-100338circRNADifferential ultracentrifugationPromotes HCC cell invasion and angiogenesis in vitro,
promotes HCC growth, angiogenesis, and lung metastasis in vivo
[88]
Metastasis
14-3-3ζProteinDifferential ultracentrifugationSuppresses anti-tumor activity of tumor-infiltrating T lymphocytes [93]
CAV1, CAV2, MET ProteinDifferential ultracentrifugationPromotes migration and invasion of non-motile immortalized hepatocyte cells in vitroActivates the PI3K/AkT and MAPK/ERK pathways[98]
SMAD3Protein
ExoQuick™ Exosome Precipitation Solution Promotes HCC cells adhesion in vitro
Activates ROS expression[99]
HMGB1Protein
Differential ultracentrifugationPromotes TIM-1+ B cell expansion and suppresses CD8+ T cells activity in vitroActivates the TLR2/4-MAPK pathway[94]
miR-25-5pmiRNADifferential ultracentrifugationPromotes transendothelial migration of HCC cell in vitro,
promotes HCC tumor self-seeding in vivo
Suppresses LRRC7 expression[100]
miR-92bmiRNAExoQuick™ Exosome Precipitation Solution Promotes HCC cell migration and suppresses NK cells cytotoxicity in vitroMechanism regarding HCC migration is not mentioned
Suppresses CD69 on NK cells
[95]
miR-103miRNADifferential ultracentrifugationIncreases vascular permeability in vitro and in vivo,
promotes liver and lung metastasis in vivo
Suppresses expression of VE-cadherin, p120-catenin, and ZO-1[101]
miR-146a-5pmiRNADifferential ultracentrifugationPromotes M2-polarization of tumor-associated macrophages and suppresses T cells activity in vitro and in vivo[96]
miR-150-3p
(from CAF)
miRNATotal Exosome Isolation ReagentSuppresses HCC cell migration and invasion in vitro[102]
miR-490
(from mast cells)
miRNATotal Exosome Isolation ReagentSuppresses HCC cell migration and invasion in vitroSuppresses the EGFR/AkT/ERK1/2 pathway[103]
circ-PTGR1circRNAExoQuick-TC™ Exosome Precipitation SolutionPromotees HCC cell migration and invasion in vitro,
promotes mesenteric lymph node metastasis in vivo
Competes with MET and suppresses miR449a expression[104]
circ-UHRF1circRNAExoQuick™ Exosome Precipitation Solution Suppresses NK cell secretion of IFN-γ and TNF-α in vitro and in vivo, promotes metastasis in vivoSuppresses miR-449c-5p to upregulate TIM-3[97]
1 The label ↑ indicates that the expression level of the cargo in EVs derived from HCC cells is higher than which derived from normal hepatoctyes. The label ↓ indicates that the expression level of the cargo in EVs derived from HCC cells is lower than which derived from normal hepatoctyes. 2 ExoQuick™ Exosome Precipitation Solution and ExoQuick-TC™ Exosome Precipitation Solution are produced by System Biosciences, USA; Total Exosome Isolation Reagent and Kit are produced by Invitrogen, USA. 5-FU, 5-fluorouracil; Akt, AKT serine/threonine kinase 1; B4GALT3, β-1,4-galactosyltransferases III; BAK1, BCL2 antagonist/killer 1; CAF, cancer-associated fibroblasts; CAV, caveolins; CDK2, cyclin-dependent kinase 2; CDKN1A, cyclin-dependent kinase inhibitor 1A; EGFR, epidermal growth factor receptor; EMT, epithelial–mesenchymal transition; ERG, erythroblast transformation-specific-related gene; ERK, extracellular signal-regulated kinase; FAK, focal adhesion kinase; GOLM1, Golgi membrane protein 1; GSK3β, glycogen synthase kinase 3β; HBGF-5, human fibroblast growth factor 5; HCC, hepatocellular carcinoma; HUVEC, human umbilical vein endothelial cell; IFN-γ, interferon gamma; LRP6, LDL Receptor Related Protein 6; MAPK, mitogen activated protein kinase; MMP, matrix metalloproteinase; NK cell, natural killer cell; PBX2, pre-B-cell leukemia homeobox 3; PI3K, phosphatidylinositol-3 kinase; PTEN, phosphatase and tensin homolog; ROS, reactive oxygen species; TAM, tumor-associated macrophages; TIMP2, tissue inhibitor metalloproteinase-2; TLR, toll-like receptor; TNF-α, tumor necrosis factor alpha; TP53INP1, tumor protein 53-induced nuclear protein 1; USP7, ubiquitin specific peptidase 7; VE-cadherin, vascular endothelial cadherin; VEGF, vascular endothelial growth factor; ZEB1, zinc finger E-box binding homeobox 1; ZO-1, zonula occludens-1.

5. EVs as Biomarkers for Detection of HCC

Current tools for the detection and diagnosis of HCC include radiographic assessments (ultrasound for screening, computed tomography, and magnetic resonance imaging for diagnosis), and serum biomarkers (alpha-fetoprotein—AFP) [110]. Imaging techniques have limitations in identifying small tumors [111]. In cases of diagnostic uncertainty, invasive procedures, such as liver biopsy, may be necessary [110]. As a result of these limitations, development of novel diagnostic tools for the detection of HCC represents an unmet need. Currently available data suggest that EVs and their cargoes, including mRNAs, non-coding RNAs, and proteins, have the potential to serve as biomarkers for the detection of HCC (Table 2). These potential biomarkers can be isolated from plasma, serum, and, in some cases, bile.
Studies have demonstrated that measuring the amounts of EVs could be a strategy for the diagnosis of HCC [12,13]. By isolating EVs from peripheral blood using ultracentrifugation, Wang et al. found an increased quantity of EVs in HCC patients compared with those with liver cirrhosis [12]. The level of EVs was correlated to the tumor size and pathological classification of HCC and, most importantly, could be used to distinguishe early-stage (TNM Stage I) HCC from cirrhotic controls with an area under the curve (AUC) of 0.83 in receiver operating characteristic (ROC) analysis. Using the fluorescence-activated cell scanning, Julich-Haertel et al. identified a subgroup of EVs, the EpCAM+ asialoglycoprotein receptor 1+ (ASGPR1)+ EVs, which is capable of distinguishing HCC from cirrhosis with an AUC of 0.73 [13]. In both of these studies, blood samples after curative surgical treatment had significantly reduced levels of EVs, indicating the ability of these biomarkers to reflect tumor burden and monitor treatment response [12,13].

5.1. EV Protein for Detection of HCC

Proteomic analysis of EVs is a less explored avenue in the identification of novel biomarkers, with studies demonstrating different compositions of the proteomes of HCC, cirrhotic, and healthy control patients [112]. Arbelaiz et al. analyzed the EVs proteome profiles of intrahepatic cholangiocarcinoma (iCCA) and HCC and showed the differentially expressed proteins within these EVs could separate these two groups with an AUC of 0.89 [113]. This finding suggests EV-protein as a promising biomarker for characterizing an atypical intrahepatic lesion between HCC and iCCA. As one of the molecules mediating HCC metastasis, SMAD3, to has diagnostic power for HCC as well (AUC of 0.70 for distinguishing HCC from benign hepatoma and healthy controls) [99]. However, none of the EV-protein markers has been evaluated among at-risk patients with cirrhosis or chronic hepatitis B and patients with HCC, and further investigation is needed to assess their accuracy as a test for HCC surveillance.

5.2. EV miRNA for Detection of HCC

miRNAs in EVs have shown particular promise as biomarkers for the detection of HCC. Wang et al. examined the miRNA profile of EVs derived from HCC and cirrhotic patients and found that certain upregulated miRNAs (miR-122, miR-148a, and miR-1246) outperformered AFP in distinguishing HCC from cirrhosis. The final panel, comprising of miR-122, miR-148a, and AFP, resulted in an AUC of 0.93 [114]. It is noteworthy that the authors did not restrict the HCC cases to early-stage disease, thus likely overestimated the diagnostic power of the assay. In another study, Ghosh et al. identified four miRNAs, miR-10b-5p, miR-21-5p, miR-221-3p, and miR-223-3p, in liver-specific asialoglycoprotein receptor 2+ (ASGR2)+ EVs for HCC diagnosis. The combination of these four miRNAs exhibited good diagnostic power among patients with low AFP expression (<250 ng/mL), with an AUC of 0.80 [115]. Although Sohn et al. showed that clusters of miRNAs (miR-18a, miR-221, miR-222, miR-224, miR-101, miR-106b, miR-122, and miR-195) were differentially expressed among patients with chronic hepatitis B, cirrhosis and patients with HCC, no further analysis using ROC was performed to determine their diagnostic performance [116].
Table 2. EVs as biomarkers for detection of HCC.
Table 2. EVs as biomarkers for detection of HCC.
Biomarkers/
Diagnostic Model
Biomarker
Type
Expression Level in HCCEV isolation Method 1Number of PatientsSen/Spe
(%)
AUROCStudy TypeRestricts HCC to Early-Stage?Ref.
Amount of total EVsUltracentrifugation28 TNM stage I HCC vs. 40 cirrhosis63/890.83Case-controlYes [12]
Amount of AnnexinV+ EpCAM+ ASGPR1+ EVUltracentrifugation86 HCC vs. 49 cirrhosis81/470.73Case-controlNo[13]
FIBGProtein
(↑in iCCA)
Filtration and Ultracentrifugation29 HCC vs. 12 iCCA 83/900.89Case-controlNo[113]
SMAD3ProteinExoQuick™ Exosome Precipitation Solution29 HCC vs. 37 HD + benign hepatoma–/–0.70Case-controlNo[99]
A panel combining miR-122, miR-148a, and AFPmiRNA +
AFP
Ultracentrifugation, filtration, and precipitation50 HCC vs. 40 cirrhosis86/880.93Case-controlNo[114]
A panel combining miR-10b-5p, miR-221-3p, miR-223-3p, and miR-21-5pmiRNAExoEnrich™ instant exosome isolation kit and immunoaffinity capture (anti-ASGR2)38 HCC vs. 35 CH + 25 cirrhosis59/950.80Case-controlNo[115]
miR-18a, miR-101, miR-106b, miR-122, miR-195, miR-221, miR-222, miR-224miRNA↑(18a, 221, 222, 224)
↓(101, 106b, 122, 195)
Ultracentrifugation20 HCC vs. 20 cirrhosis vs. 20 CH B–/–Case-controlNo[116]
LINC00853lncRNAExoQuick™ Exosome Precipitation Solution32 early-stage HCC (single, <2 cm) vs. 28 CH + 35 cirrhosis94/850.96Case-controlYes[117]
Lnc85lncRNARibo™ Exosome Isolation Reagent122 HCC vs. 43 cirrhosis80/740.89Case-controlNo[118]
RN7SL1 S fragmentlncRNAUltracentrifugation and Filtration25 HCC vs. 25 healthy donors–/–0.75Case-controlNo[119]
A risk score panel combining AFP and
ENSG00000248932.1, ENST00000440688.1, ENST00000457302.2
lncRNA +
AFP
ExoQuick™ Exosome Precipitation SolutionTraining set: 20 HCC vs. 20 CH
Validation set: 180 HCC vs. 180 CH
–/–0.97
0.87
Case-controlNo[120]
A panel combining circ_0004001,
circ_0004123, and circ_0075792
circRNAUltracentrifugation71 HCC vs. 40 HD91/780.89Case-controlNo[121]
A panel combining 8 long RNAsexoRNeasy Maxi KitTraining set: 44 HCC vs. 78 HD
1st Validation set: 27 HCC vs. 53 HD
2nd Validation set: 33 HCC vs. 33 HD + 6 hepatic benign disorders
84/94
89/91
–/–
0.95
0.96
0.96
Case-controlNo[122]
LDHCmRNAexoRNeasy Midi Kit50 TNM stage I/II HCC vs. 100 HD88/930.95Case-controlYes[123]
A panel combining AFP, GPC3, ALB, APOH, FABP1, FGB, FGG, AHSG, RBP4,TFmRNAEV Click Chip (immunoaffinity + microfluidic device)36 BCLC stage 0-A HCC vs. 26 cirrhosis84/880.93Case-controlYes [38]
1 ExoEnrich™ instant exosome isolation kit is produced by ExoCan Healthcare Technologies Private Limited, India; ExoQuick™ Exosome Precipitation Solution is produced by System Biosciences, USA; exoRNeasy Midi/Maxi Kit is produced by Qiagen, Germany; Ribo™ Exosome Isolation Reagent is produced by RiboBio, China. AFP, alpha-fetoprotein; AHSG, alpha 2-HS glycoprotein; ALB, albumin; APOH, apolipoprotein H; ASGPR1, asialoglycoprotein receptor 1; ASGR 2, asialoglycoprotein receptor 2; AUROC, area under the receiver operating characteristic; BCLC, Barcelona Clinic liver cancer; CH, chronic hepatitis; CH B, chronic hepatitis B; circRNA, circular RNA; FABP1, fatty acid binding protein 1; EpCAM, epithelial cell adhesion molecule; EVs, extracellular vesicles; FIBG, fibrinogen gamma chain; FGB, fibrinogen beta chain; FGG, fibrinogen gamma cha; GPC3, glypican 3; HCC, hepatocellular carcinoma; HD, healthy donors; iCCA, intrahepatic cholangiocarcinoma; LDHC, actate dehydrogenase C; lncRNA, long non-coding RNA; miRNA, microRNA; RBP4, retinol binding protein 4; RN7SL1, RNA component of signal recognition particle 7SL1; SMAD3, SMAD family member 3; TF, transferrin.

5.3. EV lncRNA and EV circRNA for Detection of HCC

Along with the numerous promising miRNA biomarker targets, other non-coding RNAs packaged in EVs, such as lncRNAs, have shown promise in the early detection of HCC [117,118,119,120]. One study selected six upregulated lncRNAs from The Cancer Genome Atlas by comparing 371 HCC and 50 nontumor tissues and showed that LINC00853 in EVs was particularly promising for the identification of early-stage HCC [117]. Specifically, when setting a 14-fold increase as a cutoff for the expression of LINC00853 in EVs, it could discriminate patients with a single, <2cm HCC from those with chronic hepatitis or liver cirrhosis, with an AUC of 0.96 [117]. By combining three EV-derived lncRNA and AFP, Lu et al. established an HCC diagnostic model and validated it in an independent validation cohort [120]. In this large validation cohort (n = 360), they demonstrated the model could distinguish HCC from chronic hepatitis with an AUC of 0.87 [120]. Nevertheless, failure to restrict the HCC patients to those with an early-stage disease and failure to set noncirrhotic patients as the control group are the major limitations of this study.
EV-derived circRNAs are another example of potential biomarkers for HCC detection. One study identified three upregulated circRNAs, circ_0004001, circ_0004123, and circ_0075792, in EVs isolated from HCC patients compared with those from healthy donors [121]. These three circRNAs are associated with the VEGF, PI3K/Akt, mechanistic target of rapamycin (mTOR), and Wnt pathways and the authors found the combination of these circRNAs had a potential for detection of HCC with an AUC of 0.89 [121]. In 2019, Li et al. performed EVs’ long RNA sequencing in five cancers including HCC to identify biomarkers for cancer diagnosis [122]. A diagnostic model containing eight long RNAs was built and validated in two cohorts to detect HCC from healthy donors and patients with unspecified benign hepatic disorders [122]. Despite the promising results with AUCs of 0.96 in the validation cohorts, there is still a concern about overestimating the diagnostic power considering the cohort composition.

5.4. EV mRNA for Detection of HCC

A study compared the potential of LDHC mRNA level in serum vs. serum-derived EVs for HCC detection. Interestingly, the authors demonstrated the superior ability of EV-derived LDHC mRNA to distinguish the TNM stage I/II HCC patients from healthy controls compared with serum-only LDHC mRNA, with an AUC of 0.95 vs. 0.84, respectively [123]. This difference might be attributed to the fact that the mRNAs in EVs are more stable than the circulating ones due to the protection by a phospholipid bilayer.
Together, all these studies demonstrate the promising application of EVs as biomarkers for the detection of HCC. It should be noted that almost all the studies enriched total EVs rather than those specifically secreted by HCC or hepatocytes. As the HCC-derived EVs represent a small portion of the total EVs, disease-specific changes in these potential EVs biomarkers may be difficult to detect, considering elevated background noise. To address this issue, our research team has developed a streamlined HCC EVs digital scoring assay [38] that couples two very powerful technologies, i.e., EV Click Chip for purification of HCC-derived EVs and RT-ddPCR for quantification of a panel of 10 HCC-specific mRNA markers. Benefiting from the nanostructured substrates, antibody cocktails—including anti-EpCAM, anti-ASGPR1, and anti-CD147—and click chemistry-mediated EVs capture/release process, EV Click Chip enables rapid and efficient purification of HCC-derived EVs. Most important of all, thanks to quantifying the 10 HCC-specific mRNA markers [124] in these purified EVs, the resulting HCC EVs digital scores exhibited promising potential for distinguishing the Barcelona Clinic liver cancer (BCLC) stage 0-A HCC from at-risk cirrhotic patients, with an AUC of 0.93 [38].

6. Conclusion and Future Direction

EVs play a crucial role in intercellular communication and mediate the pathogenesis, proliferation, immune escape, and metastasis of HCC [46,47,48]. As such, EVs are regarded as potential therapeutic agents or vehicles for HCC treatment [125]. With emerging studies in the field, the EVs’ cargos, including functional proteins, non-coding RNAs, and mRNAs, are promising biomarkers for the detection of early-stage HCC [126]. In parallel, dedicated efforts have been made to isolate EVs more efficiently, to facilitate the adoption of this technology for clinical applications [31,127].
Despite these encouraging results, most of the studies on detecting HCC by using EVs are still in the preclinical phase and large prospective cohort studies are warranted to validate their diagnostic value. In addition, for case-control studies to accurately estimate the diagnostic performance of EVs, it is essential to restrict the cases to early-stage HCC and the controls to at-risk patients with liver cirrhosis or chronic hepatitis B, in line with the current clinical practice guidelines which define the at-risk population to whom it is recommended to undergo screening. With the guidance of a biomarker development framework provided by the International Liver Cancer Association [128], advents of more high-quality biomarker studies on EVs for detection of early-stage HCC are expected.

Author Contributions

Conceptualization, H.-R.T. and J.D.Y.; investigation, Y.-T.L., B.V.T., J.J.W., H.-R.T., and J.D.Y.; writing—original draft preparation, Y.-T.L., B.V.T., J.J.W., and I.Y.L.; writing—review and editing, Y.-T.L., B.V.T., J.J.W., I.Y.L., S.Y., Y.Z., V.G.A., H.-R.T., and J.D.Y.; visualization, Y.-T.L., B.V.T., J.J.W., and I.Y.L.; supervision, S.Y., Y.Z., V.G.A., H.-R.T., and J.D.Y.; project administration, J.D.Y.; funding acquisition, S.Y., Y.Z., V.G.A.,H.-R.T., and J.D.Y. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by American College of Gastroenterology, grant name “Junior Faculty Development Award”; United States Department of Defense, grant number CA191051; Cedars-Sinai Medical Center, grant name “Clinical Scholar award”; Huiying Foundation; and National Institutes of Health, grant number R01CA246304, R01CA253651, R01CA255727, R21CA235340, and R21CA240887.

Conflicts of Interest

H.-R.T. would like to disclose his financial interests in CytoLumina Technologies Corp. and Pulsar Therapeutics Corp., given his role as the co-founder of these companies. J.D.Y. provides a consulting service for Exact Sciences, Gilead, and Eisai. The funders had no role in the interpretation of data and in the writing of the manuscript.

References

  1. Global Burden of Disease Cancer Collaboration; Fitzmaurice, C.; Allen, C.; Barber, R.M.; Barregard, L.; Bhutta, Z.A.; Brenner, H.; Dicker, D.J.; Chimed-Orchir, O.; Dandona, R.; et al. Global, Regional, and National Cancer Incidence, Mortality, Years of Life Lost, Years Lived With Disability, and Disability-Adjusted Life-years for 32 Cancer Groups, 1990 to 2015: A Systematic Analysis for the Global Burden of Disease Study. JAMA Oncol. 2017, 3, 524–548. [Google Scholar] [CrossRef]
  2. Yang, J.D.; Hainaut, P.; Gores, G.J.; Amadou, A.; Plymoth, A.; Roberts, L.R. A global view of hepatocellular carcinoma: Trends, risk, prevention and management. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 589–604. [Google Scholar] [CrossRef] [PubMed]
  3. Lee, Y.T.; Wang, J.J.; Luu, M.; Noureddin, M.; Kosari, K.; Agopian, V.G.; Rich, N.E.; Lu, S.C.; Tseng, H.R.; Nissen, N.N.; et al. The Mortality and Overall Survival Trends of Primary Liver Cancer in the United States. J. Natl. Cancer Inst. 2021. [Google Scholar] [CrossRef]
  4. Parikh, N.D.; Mehta, A.S.; Singal, A.G.; Block, T.; Marrero, J.A.; Lok, A.S. Biomarkers for the Early Detection of Hepatocellular Carcinoma. Cancer Epidemiol. Biomark. Prev. 2020, 29, 2495–2503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Dhanasekaran, R.; Bandoh, S.; Roberts, L.R. Molecular pathogenesis of hepatocellular carcinoma and impact of therapeutic advances. F1000Research 2016, 5. [Google Scholar] [CrossRef] [PubMed]
  6. Thery, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 2018, 7, 1535750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Rossi, G.; Ignatiadis, M. Promises and Pitfalls of Using Liquid Biopsy for Precision Medicine. Cancer Res. 2019, 79, 2798–2804. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Dong, J.; Chen, J.F.; Smalley, M.; Zhao, M.; Ke, Z.; Zhu, Y.; Tseng, H.R. Nanostructured Substrates for Detection and Characterization of Circulating Rare Cells: From Materials Research to Clinical Applications. Adv. Mater. 2020, 32, e1903663. [Google Scholar] [CrossRef]
  9. Krebs, M.G.; Metcalf, R.L.; Carter, L.; Brady, G.; Blackhall, F.H.; Dive, C. Molecular analysis of circulating tumour cells-biology and biomarkers. Nat. Rev. Clin. Oncol. 2014, 11, 129–144. [Google Scholar] [CrossRef]
  10. Kosaka, N.; Kogure, A.; Yamamoto, T.; Urabe, F.; Usuba, W.; Prieto-Vila, M.; Ochiya, T. Exploiting the message from cancer: The diagnostic value of extracellular vesicles for clinical applications. Exp. Mol. Med. 2019, 51, 1–9. [Google Scholar] [CrossRef] [Green Version]
  11. 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]
  12. Wang, W.; Li, H.; Zhou, Y.; Jie, S. Peripheral blood microvesicles are potential biomarkers for hepatocellular carcinoma. Cancer Biomark. 2013, 13, 351–357. [Google Scholar] [CrossRef]
  13. Julich-Haertel, H.; Urban, S.K.; Krawczyk, M.; Willms, A.; Jankowski, K.; Patkowski, W.; Kruk, B.; Krasnodebski, M.; Ligocka, J.; Schwab, R.; et al. Cancer-associated circulating large extracellular vesicles in cholangiocarcinoma and hepatocellular carcinoma. J. Hepatol. 2017, 67, 282–292. [Google Scholar] [CrossRef] [PubMed]
  14. Zijlstra, A.; Di Vizio, D. Size matters in nanoscale communication. Nat. Cell Biol. 2018, 20, 228–230. [Google Scholar] [CrossRef]
  15. Gardiner, C.; Di Vizio, D.; Sahoo, S.; Thery, C.; Witwer, K.W.; Wauben, M.; Hill, A.F. Techniques used for the isolation and characterization of extracellular vesicles: Results of a worldwide survey. J. Extracell. Vesicles 2016, 5, 32945. [Google Scholar] [CrossRef] [PubMed]
  16. Raposo, G.; Nijman, H.W.; Stoorvogel, W.; Liejendekker, R.; Harding, C.V.; Melief, C.J.; Geuze, H.J. B lymphocytes secrete antigen-presenting vesicles. J. Exp. Med. 1996, 183, 1161–1172. [Google Scholar] [CrossRef] [PubMed]
  17. Thery, C.; Amigorena, S.; Raposo, G.; Clayton, A. Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Curr. Protoc. Cell Biol. 2006. [Google Scholar] [CrossRef]
  18. Witwer, K.W.; Buzas, E.I.; Bemis, L.T.; Bora, A.; Lasser, C.; Lotvall, J.; Nolte-’t Hoen, E.N.; Piper, M.G.; Sivaraman, S.; Skog, J.; et al. Standardization of sample collection, isolation and analysis methods in extracellular vesicle research. J. Extracell. Vesicles 2013, 2, 20360. [Google Scholar] [CrossRef]
  19. Poliakov, A.; Spilman, M.; Dokland, T.; Amling, C.L.; Mobley, J.A. Structural heterogeneity and protein composition of exosome-like vesicles (prostasomes) in human semen. Prostate 2009, 69, 159–167. [Google Scholar] [CrossRef] [PubMed]
  20. Keller, S.; Ridinger, J.; Rupp, A.K.; Janssen, J.W.; Altevogt, P. Body fluid derived exosomes as a novel template for clinical diagnostics. J. Transl. Med. 2011, 9, 86. [Google Scholar] [CrossRef] [Green Version]
  21. Cantin, R.; Diou, J.; Belanger, D.; Tremblay, A.M.; Gilbert, C. Discrimination between exosomes and HIV-1: Purification of both vesicles from cell-free supernatants. J. Immunol. Methods 2008, 338, 21–30. [Google Scholar] [CrossRef] [PubMed]
  22. Heinemann, M.L.; Ilmer, M.; Silva, L.P.; Hawke, D.H.; Recio, A.; Vorontsova, M.A.; Alt, E.; Vykoukal, J. Benchtop isolation and characterization of functional exosomes by sequential filtration. J. Chromatogr. A 2014, 1371, 125–135. [Google Scholar] [CrossRef] [PubMed]
  23. Taylor, D.D.; Shah, S. Methods of isolating extracellular vesicles impact down-stream analyses of their cargoes. Methods 2015, 87, 3–10. [Google Scholar] [CrossRef]
  24. Zeringer, E.; Barta, T.; Li, M.; Vlassov, A.V. Strategies for isolation of exosomes. Cold Spring Harb. Protoc. 2015, 2015, 319–323. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Gamez-Valero, A.; Monguio-Tortajada, M.; Carreras-Planella, L.; Franquesa, M.; Beyer, K.; Borras, F.E. Size-Exclusion Chromatography-based isolation minimally alters Extracellular Vesicles’ characteristics compared to precipitating agents. Sci. Rep. 2016, 6, 33641. [Google Scholar] [CrossRef] [Green Version]
  26. Mol, E.A.; Goumans, M.J.; Doevendans, P.A.; Sluijter, J.P.G.; Vader, P. Higher functionality of extracellular vesicles isolated using size-exclusion chromatography compared to ultracentrifugation. Nanomedicine 2017, 13, 2061–2065. [Google Scholar] [CrossRef] [PubMed]
  27. Takov, K.; Yellon, D.M.; Davidson, S.M. Comparison of small extracellular vesicles isolated from plasma by ultracentrifugation or size-exclusion chromatography: Yield, purity and functional potential. J. Extracell. Vesicles 2019, 8, 1560809. [Google Scholar] [CrossRef] [PubMed]
  28. Hong, C.S.; Funk, S.; Muller, L.; Boyiadzis, M.; Whiteside, T.L. Isolation of biologically active and morphologically intact exosomes from plasma of patients with cancer. J. Extracell. Vesicles 2016, 5, 29289. [Google Scholar] [CrossRef]
  29. Welton, J.L.; Webber, J.P.; Botos, L.A.; Jones, M.; Clayton, A. Ready-made chromatography columns for extracellular vesicle isolation from plasma. J. Extracell. Vesicles 2015, 4, 27269. [Google Scholar] [CrossRef]
  30. Vogel, R.; Coumans, F.A.; Maltesen, R.G.; Boing, A.N.; Bonnington, K.E.; Broekman, M.L.; Broom, M.F.; Buzas, E.I.; Christiansen, G.; Hajji, N.; et al. A standardized method to determine the concentration of extracellular vesicles using tunable resistive pulse sensing. J. Extracell. Vesicles 2016, 5, 31242. [Google Scholar] [CrossRef] [PubMed]
  31. Konoshenko, M.Y.; Lekchnov, E.A.; Vlassov, A.V.; Laktionov, P.P. Isolation of Extracellular Vesicles: General Methodologies and Latest Trends. Biomed. Res. Int. 2018, 2018, 8545347. [Google Scholar] [CrossRef]
  32. Rider, M.A.; Hurwitz, S.N.; Meckes, D.G., Jr. ExtraPEG: A Polyethylene Glycol-Based Method for Enrichment of Extracellular Vesicles. Sci. Rep. 2016, 6, 23978. [Google Scholar] [CrossRef] [PubMed]
  33. Van Deun, J.; Mestdagh, P.; Sormunen, R.; Cocquyt, V.; Vermaelen, K.; Vandesompele, J.; Bracke, M.; De Wever, O.; Hendrix, A. The impact of disparate isolation methods for extracellular vesicles on downstream RNA profiling. J. Extracell. Vesicles 2014, 3. [Google Scholar] [CrossRef] [Green Version]
  34. Lobb, R.J.; Becker, M.; Wen, S.W.; Wong, C.S.; Wiegmans, A.P.; Leimgruber, A.; Moller, A. Optimized exosome isolation protocol for cell culture supernatant and human plasma. J. Extracell. Vesicles 2015, 4, 27031. [Google Scholar] [CrossRef] [PubMed]
  35. Kim, G.; Yoo, C.E.; Kim, M.; Kang, H.J.; Park, D.; Lee, M.; Huh, N. Noble polymeric surface conjugated with zwitterionic moieties and antibodies for the isolation of exosomes from human serum. Bioconjug. Chem. 2012, 23, 2114–2120. [Google Scholar] [CrossRef] [PubMed]
  36. Yoo, C.E.; Kim, G.; Kim, M.; Park, D.; Kang, H.J.; Lee, M.; Huh, N. A direct extraction method for microRNAs from exosomes captured by immunoaffinity beads. Anal. Biochem. 2012, 431, 96–98. [Google Scholar] [CrossRef] [PubMed]
  37. Mathivanan, S.; Lim, J.W.; Tauro, B.J.; Ji, H.; Moritz, R.L.; Simpson, R.J. Proteomics analysis of A33 immunoaffinity-purified exosomes released from the human colon tumor cell line LIM1215 reveals a tissue-specific protein signature. Mol. Cell Proteom. 2010, 9, 197–208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Sun, N.; Lee, Y.T.; Zhang, R.Y.; Kao, R.; Teng, P.C.; Yang, Y.; Yang, P.; Wang, J.J.; Smalley, M.; Chen, P.J.; et al. Purification of HCC-specific extracellular vesicles on nanosubstrates for early HCC detection by digital scoring. Nat. Commun. 2020, 11, 4489. [Google Scholar] [CrossRef]
  39. Chen, C.; Skog, J.; Hsu, C.H.; Lessard, R.T.; Balaj, L.; Wurdinger, T.; Carter, B.S.; Breakefield, X.O.; Toner, M.; Irimia, D. Microfluidic isolation and transcriptome analysis of serum microvesicles. Lab Chip 2010, 10, 505–511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Tauro, B.J.; Greening, D.W.; Mathias, R.A.; Ji, H.; Mathivanan, S.; Scott, A.M.; Simpson, R.J. Comparison of ultracentrifugation, density gradient separation, and immunoaffinity capture methods for isolating human colon cancer cell line LIM1863-derived exosomes. Methods 2012, 56, 293–304. [Google Scholar] [CrossRef] [PubMed]
  41. Rana, S.; Yue, S.; Stadel, D.; Zoller, M. Toward tailored exosomes: The exosomal tetraspanin web contributes to target cell selection. Int. J. Biochem. Cell Biol. 2012, 44, 1574–1584. [Google Scholar] [CrossRef]
  42. He, M.; Zeng, Y. Microfluidic Exosome Analysis toward Liquid Biopsy for Cancer. J. Lab. Autom. 2016, 21, 599–608. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Jo, W.; Jeong, D.; Kim, J.; Cho, S.; Jang, S.C.; Han, C.; Kang, J.Y.; Gho, Y.S.; Park, J. Microfluidic fabrication of cell-derived nanovesicles as endogenous RNA carriers. Lab Chip 2014, 14, 1261–1269. [Google Scholar] [CrossRef]
  44. Davies, R.T.; Kim, J.; Jang, S.C.; Choi, E.J.; Gho, Y.S.; Park, J. Microfluidic filtration system to isolate extracellular vesicles from blood. Lab Chip 2012, 12, 5202–5210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Desai, A.; Sandhu, S.; Lai, J.P.; Sandhu, D.S. Hepatocellular carcinoma in non-cirrhotic liver: A comprehensive review. World J. Hepatol. 2019, 11, 1–18. [Google Scholar] [CrossRef] [PubMed]
  46. Rios-Colon, L.; Arthur, E.; Niture, S.; Qi, Q.; Moore, J.T.; Kumar, D. The Role of Exosomes in the Crosstalk between Adipocytes and Liver Cancer Cells. Cells 2020, 9, 1988. [Google Scholar] [CrossRef] [PubMed]
  47. Wang, H.; Lu, Z.; Zhao, X. Tumorigenesis, diagnosis, and therapeutic potential of exosomes in liver cancer. J. Hematol. Oncol. 2019, 12, 133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. He, R.; Wang, Z.; Shi, W.; Yu, L.; Xia, H.; Huang, Z.; Liu, S.; Zhao, X.; Xu, Y.; Yam, J.W.P.; et al. Exosomes in hepatocellular carcinoma microenvironment and their potential clinical application value. Biomed. Pharmacother. 2021, 138. [Google Scholar] [CrossRef]
  49. Ramakrishnaiah, V.; Thumann, C.; Fofana, I.; Habersetzer, F.; Pan, Q.; de Ruiter, P.E.; Willemsen, R.; Demmers, J.A.; Stalin Raj, V.; Jenster, G.; et al. Exosome-mediated transmission of hepatitis C virus between human hepatoma Huh7.5 cells. Proc. Natl. Acad. Sci. USA 2013, 110, 13109–13113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Yang, Y.; Han, Q.; Hou, Z.; Zhang, C.; Tian, Z.; Zhang, J. Exosomes mediate hepatitis B virus (HBV) transmission and NK-cell dysfunction. Cell Mol. Immunol. 2017, 14, 465–475. [Google Scholar] [CrossRef]
  51. Devhare, P.B.; Sasaki, R.; Shrivastava, S.; Di Bisceglie, A.M.; Ray, R.; Ray, R.B. Exosome-Mediated Intercellular Communication between Hepatitis C Virus-Infected Hepatocytes and Hepatic Stellate Cells. J. Virol. 2017, 91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Kapoor, N.R.; Chadha, R.; Kumar, S.; Choedon, T.; Reddy, V.S.; Kumar, V. The HBx gene of hepatitis B virus can influence hepatic microenvironment via exosomes by transferring its mRNA and protein. Virus Res. 2017, 240, 166–174. [Google Scholar] [CrossRef] [PubMed]
  53. Enomoto, Y.; Takagi, R.; Naito, Y.; Kiniwa, T.; Tanaka, Y.; Hamada-Tsutsumi, S.; Kawano, M.; Matsushita, S.; Ochiya, T.; Miyajima, A. Identification of the novel 3’ UTR sequences of human IL-21 mRNA as potential targets of miRNAs. Sci. Rep. 2017, 7, 7780. [Google Scholar] [CrossRef] [PubMed]
  54. Hernandez, A.; Arab, J.P.; Reyes, D.; Lapitz, A.; Moshage, H.; Banales, J.M.; Arrese, M. Extracellular Vesicles in NAFLD/ALD: From Pathobiology to Therapy. Cells 2020, 9, 817. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Hirsova, P.; Ibrahim, S.H.; Krishnan, A.; Verma, V.K.; Bronk, S.F.; Werneburg, N.W.; Charlton, M.R.; Shah, V.H.; Malhi, H.; Gores, G.J. Lipid-Induced Signaling Causes Release of Inflammatory Extracellular Vesicles From Hepatocytes. Gastroenterology 2016, 150, 956–967. [Google Scholar] [CrossRef] [Green Version]
  56. Liao, C.Y.; Song, M.J.; Gao, Y.; Mauer, A.S.; Revzin, A.; Malhi, H. Hepatocyte-Derived Lipotoxic Extracellular Vesicle Sphingosine 1-Phosphate Induces Macrophage Chemotaxis. Front. Immunol. 2018, 9, 2980. [Google Scholar] [CrossRef] [Green Version]
  57. Liu, X.L.; Pan, Q.; Cao, H.X.; Xin, F.Z.; Zhao, Z.H.; Yang, R.X.; Zeng, J.; Zhou, H.; Fan, J.G. Lipotoxic Hepatocyte-Derived Exosomal MicroRNA 192-5p Activates Macrophages Through Rictor/Akt/Forkhead Box Transcription Factor O1 Signaling in Nonalcoholic Fatty Liver Disease. Hepatology 2020, 72, 454–469. [Google Scholar] [CrossRef]
  58. Zhao, Z.; Zhong, L.; Li, P.; He, K.; Qiu, C.; Zhao, L.; Gong, J. Cholesterol impairs hepatocyte lysosomal function causing M1 polarization of macrophages via exosomal miR-122-5p. Exp. Cell Res. 2020, 387, 111738. [Google Scholar] [CrossRef]
  59. Ibrahim, S.H.; Hirsova, P.; Tomita, K.; Bronk, S.F.; Werneburg, N.W.; Harrison, S.A.; Goodfellow, V.S.; Malhi, H.; Gores, G.J. Mixed lineage kinase 3 mediates release of C-X-C motif ligand 10-bearing chemotactic extracellular vesicles from lipotoxic hepatocytes. Hepatology 2016, 63, 731–744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Dasgupta, D.; Nakao, Y.; Mauer, A.S.; Thompson, J.M.; Sehrawat, T.S.; Liao, C.Y.; Krishnan, A.; Lucien, F.; Guo, Q.; Liu, M.; et al. IRE1A Stimulates Hepatocyte-Derived Extracellular Vesicles That Promote Inflammation in Mice With Steatohepatitis. Gastroenterology 2020, 159, 1487–1503.e17. [Google Scholar] [CrossRef]
  61. Kakazu, E.; Mauer, A.S.; Yin, M.; Malhi, H. Hepatocytes release ceramide-enriched pro-inflammatory extracellular vesicles in an IRE1alpha-dependent manner. J. Lipid Res. 2016, 57, 233–245. [Google Scholar] [CrossRef] [Green Version]
  62. Guo, Q.; Furuta, K.; Lucien, F.; Gutierrez Sanchez, L.H.; Hirsova, P.; Krishnan, A.; Kabashima, A.; Pavelko, K.D.; Madden, B.; Alhuwaish, H.; et al. Integrin beta1-enriched extracellular vesicles mediate monocyte adhesion and promote liver inflammation in murine NASH. J. Hepatol. 2019, 71, 1193–1205. [Google Scholar] [CrossRef]
  63. Gai, X.; Tang, B.; Liu, F.; Wu, Y.; Wang, F.; Jing, Y.; Huang, F.; Jin, D.; Wang, L.; Zhang, H. mTOR/miR-145-regulated exosomal GOLM1 promotes hepatocellular carcinoma through augmented GSK-3beta/MMPs. J. Genet. Genom. 2019, 46, 235–245. [Google Scholar] [CrossRef] [PubMed]
  64. Tang, J.; Li, Y.; Liu, K.; Zhu, Q.; Yang, W.H.; Xiong, L.K.; Guo, D.L. Exosomal miR-9-3p suppresses HBGF-5 expression and is a functional biomarker in hepatocellular carcinoma. Minerva Med. 2018, 109, 15–23. [Google Scholar] [CrossRef] [PubMed]
  65. 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. [Google Scholar] [CrossRef] [PubMed]
  66. Yang, B.; Feng, X.; Liu, H.; Tong, R.; Wu, J.; Li, C.; Yu, H.; Chen, Y.; Cheng, Q.; Chen, J.; et al. High-metastatic cancer cells derived exosomal miR92a-3p promotes epithelial-mesenchymal transition and metastasis of low-metastatic cancer cells by regulating PTEN/Akt pathway in hepatocellular carcinoma. Oncogene 2020, 39, 6529–6543. [Google Scholar] [CrossRef] [PubMed]
  67. Xue, X.; Wang, X.; Zhao, Y.; Hu, R.; Qin, L. Exosomal miR-93 promotes proliferation and invasion in hepatocellular carcinoma by directly inhibiting TIMP2/TP53INP1/CDKN1A. Biochem. Biophys. Res. Commun. 2018, 502, 515–521. [Google Scholar] [CrossRef] [PubMed]
  68. Shi, M.; Jiang, Y.; Yang, L.; Yan, S.; Wang, Y.G.; Lu, X.J. Decreased levels of serum exosomal miR-638 predict poor prognosis in hepatocellular carcinoma. J. Cell Biochem. 2018, 119, 4711–4716. [Google Scholar] [CrossRef] [PubMed]
  69. Sugimachi, K.; Matsumura, T.; Hirata, H.; Uchi, R.; Ueda, M.; Ueo, H.; Shinden, Y.; Iguchi, T.; Eguchi, H.; Shirabe, K.; et al. Identification of a bona fide microRNA biomarker in serum exosomes that predicts hepatocellular carcinoma recurrence after liver transplantation. Br. J. Cancer 2015, 112, 532–538. [Google Scholar] [CrossRef] [PubMed]
  70. Wang, G.; Zhao, W.; Wang, H.; Qiu, G.; Jiang, Z.; Wei, G.; Li, X. Exosomal MiR-744 Inhibits Proliferation and Sorafenib Chemoresistance in Hepatocellular Carcinoma by Targeting PAX2. Med. Sci. Monit. 2019, 25, 7209–7217. [Google Scholar] [CrossRef]
  71. Cui, Y.; Xu, H.F.; Liu, M.Y.; Xu, Y.J.; He, J.C.; Zhou, Y.; Cang, S.D. Mechanism of exosomal microRNA-224 in development of hepatocellular carcinoma and its diagnostic and prognostic value. World J. Gastroenterol. 2019, 25, 1890–1898. [Google Scholar] [CrossRef] [PubMed]
  72. Qu, Z.; Wu, J.; Wu, J.; Ji, A.; Qiang, G.; Jiang, Y.; Jiang, C.; Ding, Y. Exosomal miR-665 as a novel minimally invasive biomarker for hepatocellular carcinoma diagnosis and prognosis. Oncotarget 2017, 8, 80666–80678. [Google Scholar] [CrossRef] [Green Version]
  73. Yao, Z.; Jia, C.; Tai, Y.; Liang, H.; Zhong, Z.; Xiong, Z.; Deng, M.; Zhang, Q. Serum exosomal long noncoding RNAs lnc-FAM72D-3 and lnc-EPC1-4 as diagnostic biomarkers for hepatocellular carcinoma. Aging (Albany NY) 2020, 12, 11843–11863. [Google Scholar] [CrossRef]
  74. Li, B.; Mao, R.; Liu, C.; Zhang, W.; Tang, Y.; Guo, Z. LncRNA FAL1 promotes cell proliferation and migration by acting as a CeRNA of miR-1236 in hepatocellular carcinoma cells. Life Sci. 2018, 197, 122–129. [Google Scholar] [CrossRef]
  75. Li, X.; Lei, Y.; Wu, M.; Li, N. Regulation of Macrophage Activation and Polarization by HCC-Derived Exosomal lncRNA TUC339. Int. J. Mol. Sci. 2018, 19, 2958. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Wang, J.; Pu, J.; Zhang, Y.; Yao, T.; Luo, Z.; Li, W.; Xu, G.; Liu, J.; Wei, W.; Deng, Y. Exosome-transmitted long non-coding RNA SENP3-EIF4A1 suppresses the progression of hepatocellular carcinoma. Aging (Albany NY) 2020, 12, 11550–11567. [Google Scholar] [CrossRef] [PubMed]
  77. Chen, W.; Quan, Y.; Fan, S.; Wang, H.; Liang, J.; Huang, L.; Chen, L.; Liu, Q.; He, P.; Ye, Y. Exosome-transmitted circular RNA hsa_circ_0051443 suppresses hepatocellular carcinoma progression. Cancer Lett. 2020, 475, 119–128. [Google Scholar] [CrossRef]
  78. Lai, Z.; Wei, T.; Li, Q.; Wang, X.; Zhang, Y.; Zhang, S. Exosomal circFBLIM1 Promotes Hepatocellular Carcinoma Progression and Glycolysis by Regulating the miR-338/LRP6 Axis. Cancer Biother. Radiopharm. 2020. [Google Scholar] [CrossRef] [PubMed]
  79. Zhang, H.; Deng, T.; Ge, S.; Liu, Y.; Bai, M.; Zhu, K.; Fan, Q.; Li, J.; Ning, T.; Tian, F.; et al. Exosome circRNA secreted from adipocytes promotes the growth of hepatocellular carcinoma by targeting deubiquitination-related USP7. Oncogene 2019, 38, 2844–2859. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. He, X.; Yu, J.; Xiong, L.; Liu, Y.; Fan, L.; Li, Y.; Chen, B.; Chen, J.; Xu, X. Exosomes derived from liver cancer cells reprogram biological behaviors of LO2 cells by transferring Linc-ROR. Gene 2019, 719, 144044. [Google Scholar] [CrossRef]
  81. Amann, T.; Hellerbrand, C. GLUT1 as a therapeutic target in hepatocellular carcinoma. Expert Opin. Ther. Targets 2009, 13, 1411–1427. [Google Scholar] [CrossRef] [PubMed]
  82. Wang, Y.; Wang, B.; Xiao, S.; Li, Y.; Chen, Q. miR-125a/b inhibits tumor-associated macrophages mediated in cancer stem cells of hepatocellular carcinoma by targeting CD90. J. Cell Biochem. 2019, 120, 3046–3055. [Google Scholar] [CrossRef] [PubMed]
  83. Zhang, Z.; Li, X.; Sun, W.; Yue, S.; Yang, J.; Li, J.; Ma, B.; Wang, J.; Yang, X.; Pu, M.; et al. Loss of exosomal miR-320a from cancer-associated fibroblasts contributes to HCC proliferation and metastasis. Cancer Lett. 2017, 397, 33–42. [Google Scholar] [CrossRef] [PubMed]
  84. Li, R.; Wang, Y.; Zhang, X.; Feng, M.; Ma, J.; Li, J.; Yang, X.; Fang, F.; Xia, Q.; Zhang, Z.; et al. Exosome-mediated secretion of LOXL4 promotes hepatocellular carcinoma cell invasion and metastasis. Mol. Cancer 2019, 18, 18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Matsuura, Y.; Wada, H.; Eguchi, H.; Gotoh, K.; Kobayashi, S.; Kinoshita, M.; Kubo, M.; Hayashi, K.; Iwagami, Y.; Yamada, D.; et al. Exosomal miR-155 Derived from Hepatocellular Carcinoma Cells Under Hypoxia Promotes Angiogenesis in Endothelial Cells. Dig. Dis. Sci. 2019, 64, 792–802. [Google Scholar] [CrossRef] [PubMed]
  86. 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]
  87. Conigliaro, A.; Costa, V.; Lo Dico, A.; Saieva, L.; Buccheri, S.; Dieli, F.; Manno, M.; Raccosta, S.; Mancone, C.; Tripodi, M.; et al. CD90+ liver cancer cells modulate endothelial cell phenotype through the release of exosomes containing H19 lncRNA. Mol. Cancer 2015, 14, 155. [Google Scholar] [CrossRef] [PubMed]
  88. Huang, X.Y.; Huang, Z.L.; Huang, J.; Xu, B.; Huang, X.Y.; Xu, Y.H.; Zhou, J.; Tang, Z.Y. Exosomal circRNA-100338 promotes hepatocellular carcinoma metastasis via enhancing invasiveness and angiogenesis. J. Exp. Clin. Cancer Res. 2020, 39, 20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Zhao, S.; Li, J.; Zhang, G.; Wang, Q.; Wu, C.; Zhang, Q.; Wang, H.; Sun, P.; Xiang, R.; Yang, S. Exosomal miR-451a Functions as a Tumor Suppressor in Hepatocellular Carcinoma by Targeting LPIN1. Cell Physiol. Biochem. 2019, 53, 19–35. [Google Scholar] [CrossRef] [Green Version]
  90. Fu, X.; Liu, M.; Qu, S.; Ma, J.; Zhang, Y.; Shi, T.; Wen, H.; Yang, Y.; Wang, S.; Wang, J.; et al. Exosomal microRNA-32-5p induces multidrug resistance in hepatocellular carcinoma via the PI3K/Akt pathway. J. Exp. Clin. Cancer Res. 2018, 37, 52. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Fang, T.; Lv, H.; Lv, G.; Li, T.; Wang, C.; Han, Q.; Yu, L.; Su, B.; Guo, L.; Huang, S.; et al. Tumor-derived exosomal miR-1247-3p induces cancer-associated fibroblast activation to foster lung metastasis of liver cancer. Nat. Commun. 2018, 9, 191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Wang, X.; Shen, H.; Zhangyuan, G.; Huang, R.; Zhang, W.; He, Q.; Jin, K.; Zhuo, H.; Zhang, Z.; Wang, J.; et al. 14-3-3zeta delivered by hepatocellular carcinoma-derived exosomes impaired anti-tumor function of tumor-infiltrating T lymphocytes. Cell Death Dis. 2018, 9, 159. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Ye, L.; Zhang, Q.; Cheng, Y.; Chen, X.; Wang, G.; Shi, M.; Zhang, T.; Cao, Y.; Pan, H.; Zhang, L.; et al. Tumor-derived exosomal HMGB1 fosters hepatocellular carcinoma immune evasion by promoting TIM-1(+) regulatory B cell expansion. J. Immunother. Cancer 2018, 6, 145. [Google Scholar] [CrossRef] [Green Version]
  95. Nakano, T.; Chen, I.H.; Wang, C.C.; Chen, P.J.; Tseng, H.P.; Huang, K.T.; Hu, T.H.; Li, L.C.; Goto, S.; Cheng, Y.F.; et al. Circulating exosomal miR-92b: Its role for cancer immunoediting and clinical value for prediction of posttransplant hepatocellular carcinoma recurrence. Am. J. Transplant. 2019, 19, 3250–3262. [Google Scholar] [CrossRef] [PubMed]
  96. Yin, C.; Han, Q.; Xu, D.; Zheng, B.; Zhao, X.; Zhang, J. SALL4-mediated upregulation of exosomal miR-146a-5p drives T-cell exhaustion by M2 tumor-associated macrophages in HCC. Oncoimmunology 2019, 8, 1601479. [Google Scholar] [CrossRef]
  97. Zhang, P.F.; Gao, C.; Huang, X.Y.; Lu, J.C.; Guo, X.J.; Shi, G.M.; Cai, J.B.; Ke, A.W. Cancer cell-derived exosomal circUHRF1 induces natural killer cell exhaustion and may cause resistance to anti-PD1 therapy in hepatocellular carcinoma. Mol. Cancer 2020, 19, 110. [Google Scholar] [CrossRef] [PubMed]
  98. He, M.; Qin, H.; Poon, T.C.; Sze, S.C.; Ding, X.; Co, N.N.; Ngai, S.M.; Chan, T.F.; Wong, N. Hepatocellular carcinoma-derived exosomes promote motility of immortalized hepatocyte through transfer of oncogenic proteins and RNAs. Carcinogenesis 2015, 36, 1008–1018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Fu, Q.; Zhang, Q.; Lou, Y.; Yang, J.; Nie, G.; Chen, Q.; Chen, Y.; Zhang, J.; Wang, J.; Wei, T.; et al. Primary tumor-derived exosomes facilitate metastasis by regulating adhesion of circulating tumor cells via SMAD3 in liver cancer. Oncogene 2018, 37, 6105–6118. [Google Scholar] [CrossRef] [Green Version]
  100. Liu, H.; Chen, W.; Zhi, X.; Chen, E.J.; Wei, T.; Zhang, J.; Shen, J.; Hu, L.Q.; Zhao, B.; Feng, X.H.; et al. Tumor-derived exosomes promote tumor self-seeding in hepatocellular carcinoma by transferring miRNA-25-5p to enhance cell motility. Oncogene 2018, 37, 4964–4978. [Google Scholar] [CrossRef] [PubMed]
  101. Fang, J.H.; Zhang, Z.J.; Shang, L.R.; Luo, Y.W.; Lin, Y.F.; Yuan, Y.; Zhuang, S.M. Hepatoma cell-secreted exosomal microRNA-103 increases vascular permeability and promotes metastasis by targeting junction proteins. Hepatology 2018, 68, 1459–1475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Yugawa, K.; Yoshizumi, T.; Mano, Y.; Itoh, S.; Harada, N.; Ikegami, T.; Kohashi, K.; Oda, Y.; Mori, M. Cancer-associated fibroblasts promote hepatocellular carcinoma progression through downregulation of exosomal miR-150-3p. Eur. J. Surg. Oncol. 2021, 47, 384–393. [Google Scholar] [CrossRef]
  103. Xiong, L.; Zhen, S.; Yu, Q.; Gong, Z. HCV-E2 inhibits hepatocellular carcinoma metastasis by stimulating mast cells to secrete exosomal shuttle microRNAs. Oncol. Lett. 2017, 14, 2141–2146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Wang, G.; Liu, W.; Zou, Y.; Wang, G.; Deng, Y.; Luo, J.; Zhang, Y.; Li, H.; Zhang, Q.; Yang, Y.; et al. Three isoforms of exosomal circPTGR1 promote hepatocellular carcinoma metastasis via the miR449a-MET pathway. EBioMedicine 2019, 40, 432–445. [Google Scholar] [CrossRef] [Green Version]
  105. Giannelli, G.; Koudelkova, P.; Dituri, F.; Mikulits, W. Role of epithelial to mesenchymal transition in hepatocellular carcinoma. J. Hepatol. 2016, 65, 798–808. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Capece, D.; Fischietti, M.; Verzella, D.; Gaggiano, A.; Cicciarelli, G.; Tessitore, A.; Zazzeroni, F.; Alesse, E. The inflammatory microenvironment in hepatocellular carcinoma: A pivotal role for tumor-associated macrophages. Biomed. Res. Int. 2013, 2013, 187204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Van Zijl, F.; Krupitza, G.; Mikulits, W. Initial steps of metastasis: Cell invasion and endothelial transmigration. Mutat. Res. 2011, 728, 23–34. [Google Scholar] [CrossRef] [PubMed]
  108. Winkler, J.; Abisoye-Ogunniyan, A.; Metcalf, K.J.; Werb, Z. Concepts of extracellular matrix remodelling in tumour progression and metastasis. Nat. Commun. 2020, 11, 5120. [Google Scholar] [CrossRef] [PubMed]
  109. Chen, L.; Guo, P.; He, Y.; Chen, Z.; Chen, L.; Luo, Y.; Qi, L.; Liu, Y.; Wu, Q.; Cui, Y.; et al. HCC-derived exosomes elicit HCC progression and recurrence by epithelial-mesenchymal transition through MAPK/ERK signalling pathway. Cell Death Dis. 2018, 9, 513. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Marrero, J.A.; Kulik, L.M.; Sirlin, C.B.; Zhu, A.X.; Finn, R.S.; Abecassis, M.M.; Roberts, L.R.; Heimbach, J.K. Diagnosis, Staging, and Management of Hepatocellular Carcinoma: 2018 Practice Guidance by the American Association for the Study of Liver Diseases. Hepatology 2018, 68, 723–750. [Google Scholar] [CrossRef] [Green Version]
  111. Roberts, L.R.; Sirlin, C.B.; Zaiem, F.; Almasri, J.; Prokop, L.J.; Heimbach, J.K.; Murad, M.H.; Mohammed, K. Imaging for the diagnosis of hepatocellular carcinoma: A systematic review and meta-analysis. Hepatology 2018, 67, 401–421. [Google Scholar] [CrossRef] [Green Version]
  112. Taleb, R.S.Z.; Moez, P.; Younan, D.; Eisenacher, M.; Tenbusch, M.; Sitek, B.; Bracht, T. Quantitative proteome analysis of plasma microparticles for the characterization of HCV-induced hepatic cirrhosis and hepatocellular carcinoma. Proteom. Clin. Appl. 2017, 11, 1700014. [Google Scholar] [CrossRef] [PubMed]
  113. Arbelaiz, A.; Azkargorta, M.; Krawczyk, M.; Santos-Laso, A.; Lapitz, A.; Perugorria, M.J.; Erice, O.; Gonzalez, E.; Jimenez-Aguero, R.; Lacasta, A.; et al. Serum extracellular vesicles contain protein biomarkers for primary sclerosing cholangitis and cholangiocarcinoma. Hepatology 2017, 66, 1125–1143. [Google Scholar] [CrossRef]
  114. Wang, Y.; Zhang, C.; Zhang, P.; Guo, G.; Jiang, T.; Zhao, X.; Jiang, J.; Huang, X.; Tong, H.; Tian, Y. Serum exosomal microRNAs combined with alpha-fetoprotein as diagnostic markers of hepatocellular carcinoma. Cancer Med. 2018, 7, 1670–1679. [Google Scholar] [CrossRef] [Green Version]
  115. Ghosh, S.; Bhowmik, S.; Majumdar, S.; Goswami, A.; Chakraborty, J.; Gupta, S.; Aggarwal, S.; Ray, S.; Chatterjee, R.; Bhattacharyya, S.; et al. The exosome encapsulated microRNAs as circulating diagnostic marker for hepatocellular carcinoma with low alpha-fetoprotein. Int. J. Cancer 2020, 147, 2934–2947. [Google Scholar] [CrossRef] [PubMed]
  116. Sohn, W.; Kim, J.; Kang, S.H.; Yang, S.R.; Cho, J.Y.; Cho, H.C.; Shim, S.G.; Paik, Y.H. Serum exosomal microRNAs as novel biomarkers for hepatocellular carcinoma. Exp. Mol. Med. 2015, 47, e184. [Google Scholar] [CrossRef]
  117. Kim, S.S.; Baek, G.O.; Ahn, H.R.; Sung, S.; Seo, C.W.; Cho, H.J.; Nam, S.W.; Cheong, J.Y.; Eun, J.W. Serum small extracellular vesicle-derived LINC00853 as a novel diagnostic marker for early hepatocellular carcinoma. Mol. Oncol. 2020, 14, 2646–2659. [Google Scholar] [CrossRef]
  118. Huang, X.; Sun, L.; Wen, S.; Deng, D.; Wan, F.; He, X.; Tian, L.; Liang, L.; Wei, C.; Gao, K.; et al. RNA sequencing of plasma exosomes revealed novel functional long noncoding RNAs in hepatocellular carcinoma. Cancer Sci. 2020, 111, 3338–3349. [Google Scholar] [CrossRef] [PubMed]
  119. Tan, C.; Cao, J.; Chen, L.; Xi, X.; Wang, S.; Zhu, Y.; Yang, L.; Ma, L.; Wang, D.; Yin, J.; et al. Noncoding RNAs Serve as Diagnosis and Prognosis Biomarkers for Hepatocellular Carcinoma. Clin. Chem. 2019, 65, 905–915. [Google Scholar] [CrossRef] [PubMed]
  120. Lu, Y.; Duan, Y.; Xu, Q.; Zhang, L.; Chen, W.; Qu, Z.; Wu, B.; Liu, W.; Shi, L.; Wu, D.; et al. Circulating exosome-derived bona fide long non-coding RNAs predicting the occurrence and metastasis of hepatocellular carcinoma. J. Cell Mol. Med. 2020, 24, 1311–1318. [Google Scholar] [CrossRef]
  121. Sun, X.H.; Wang, Y.T.; Li, G.F.; Zhang, N.; Fan, L. Serum-derived three-circRNA signature as a diagnostic biomarker for hepatocellular carcinoma. Cancer Cell Int. 2020, 20, 226. [Google Scholar] [CrossRef] [PubMed]
  122. Li, Y.; Zhao, J.; Yu, S.; Wang, Z.; He, X.; Su, Y.; Guo, T.; Sheng, H.; Chen, J.; Zheng, Q.; et al. Extracellular Vesicles Long RNA Sequencing Reveals Abundant mRNA, circRNA, and lncRNA in Human Blood as Potential Biomarkers for Cancer Diagnosis. Clin. Chem. 2019, 65, 798–808. [Google Scholar] [CrossRef] [PubMed]
  123. Cui, Z.; Li, Y.; Gao, Y.; Kong, L.; Lin, Y.; Chen, Y. Cancer-testis antigen lactate dehydrogenase C4 in hepatocellular carcinoma: A promising biomarker for early diagnosis, efficacy evaluation and prognosis prediction. Aging (Albany NY) 2020, 12, 19455–19467. [Google Scholar] [CrossRef] [PubMed]
  124. Kalinich, M.; Bhan, I.; Kwan, T.T.; Miyamoto, D.T.; Javaid, S.; LiCausi, J.A.; Milner, J.D.; Hong, X.; Goyal, L.; Sil, S.; et al. An RNA-based signature enables high specificity detection of circulating tumor cells in hepatocellular carcinoma. Proc. Natl. Acad. Sci. USA 2017, 114, 1123–1128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Zhang, X.; Zhang, H.; Gu, J.; Zhang, J.; Shi, H.; Qian, H.; Wang, D.; Xu, W.; Pan, J.; Santos, H.A. Engineered Extracellular Vesicles for Cancer Therapy. Adv. Mater. 2021, 33, e2005709. [Google Scholar] [CrossRef] [PubMed]
  126. Thietart, S.; Rautou, P.E. Extracellular vesicles as biomarkers in liver diseases: A clinician’s point of view. J. Hepatol. 2020, 73, 1507–1525. [Google Scholar] [CrossRef]
  127. Liangsupree, T.; Multia, E.; Riekkola, M.L. Modern isolation and separation techniques for extracellular vesicles. J. Chromatogr. A 2021, 1636, 461773. [Google Scholar] [CrossRef] [PubMed]
  128. Singal, A.G.; Hoshida, Y.; Pinato, D.J.; Marrero, J.; Nault, J.C.; Paradis, V.; Tayob, N.; Sherman, M.; Lim, Y.S.; Feng, Z.; et al. International Liver Cancer Association (ILCA) White Paper on Biomarker Development for Hepatocellular Carcinoma. Gastroenterology 2021. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic summary of the EVs isolation methods. (A) Ultracentrifugation. By centrifugation at the speeds of 10,000–20,000 g and 100,000–120,000 g, large and small EVs are separated, respectively. Density gradient ultracentrifugation can be used for further EVs purification. (B) Filtration. Using a series of membrane filters with different pore diameters enables isolation of EVs with a specified size. (C) Size exclusion chromatography. The chromatography column consists of porous beads only allowing smaller particles, such as proteins, to enter. EVs are larger than the pore size; therefore, they migrate at a higher speed than the smaller particles and are isolated. (D) Precipitation. Precipitants occupy the solution and make less soluble particles, including EVs, exceed their solubility limit and precipitate. (E) Immunoaffinity capture. Antibodies targeting surface proteins on EVs are used to positively or negatively select specific subpopulations of EVs. (F) Microfluidics. For example, EV Click Chip: (1) the multimarker antibody cocktails enable targeting HCC-derived EVs, (2) nanostructured substrates increase the surface interacting with EVs, and (3) click chemistry-mediated EVs capture (TCO/TZ interaction) and disulfide cleavage lead to DTT-driven EVs release, which results in isolation of HCC-derived EVs with high purity. ASGPR1, asialoglycoprotein receptor 1; DTT, 1,4-dithiothreitol; EpCAM, epithelial cell adhesion molecule; EVs, extracellular vesicles; HCC, hepatocellular carcinoma; TCO, trans-cyclooctene; Tz, tetrazine.
Figure 1. Schematic summary of the EVs isolation methods. (A) Ultracentrifugation. By centrifugation at the speeds of 10,000–20,000 g and 100,000–120,000 g, large and small EVs are separated, respectively. Density gradient ultracentrifugation can be used for further EVs purification. (B) Filtration. Using a series of membrane filters with different pore diameters enables isolation of EVs with a specified size. (C) Size exclusion chromatography. The chromatography column consists of porous beads only allowing smaller particles, such as proteins, to enter. EVs are larger than the pore size; therefore, they migrate at a higher speed than the smaller particles and are isolated. (D) Precipitation. Precipitants occupy the solution and make less soluble particles, including EVs, exceed their solubility limit and precipitate. (E) Immunoaffinity capture. Antibodies targeting surface proteins on EVs are used to positively or negatively select specific subpopulations of EVs. (F) Microfluidics. For example, EV Click Chip: (1) the multimarker antibody cocktails enable targeting HCC-derived EVs, (2) nanostructured substrates increase the surface interacting with EVs, and (3) click chemistry-mediated EVs capture (TCO/TZ interaction) and disulfide cleavage lead to DTT-driven EVs release, which results in isolation of HCC-derived EVs with high purity. ASGPR1, asialoglycoprotein receptor 1; DTT, 1,4-dithiothreitol; EpCAM, epithelial cell adhesion molecule; EVs, extracellular vesicles; HCC, hepatocellular carcinoma; TCO, trans-cyclooctene; Tz, tetrazine.
Cancers 13 03076 g001
Figure 2. EVs as mediators in progression of HCC. Studies have demonstrated that EVs regulate cancer cell proliferation, angiogenesis, epithelial–mesenchymal transition, immune escape, and intravasation and extravasation by their functional cargoes including proteins, miRNA, lncRNA, and circRNA.
Figure 2. EVs as mediators in progression of HCC. Studies have demonstrated that EVs regulate cancer cell proliferation, angiogenesis, epithelial–mesenchymal transition, immune escape, and intravasation and extravasation by their functional cargoes including proteins, miRNA, lncRNA, and circRNA.
Cancers 13 03076 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Lee, Y.-T.; Tran, B.V.; Wang, J.J.; Liang, I.Y.; You, S.; Zhu, Y.; Agopian, V.G.; Tseng, H.-R.; Yang, J.D. The Role of Extracellular Vesicles in Disease Progression and Detection of Hepatocellular Carcinoma. Cancers 2021, 13, 3076. https://doi.org/10.3390/cancers13123076

AMA Style

Lee Y-T, Tran BV, Wang JJ, Liang IY, You S, Zhu Y, Agopian VG, Tseng H-R, Yang JD. The Role of Extracellular Vesicles in Disease Progression and Detection of Hepatocellular Carcinoma. Cancers. 2021; 13(12):3076. https://doi.org/10.3390/cancers13123076

Chicago/Turabian Style

Lee, Yi-Te, Benjamin V. Tran, Jasmine J. Wang, Icy Y. Liang, Sungyong You, Yazhen Zhu, Vatche G. Agopian, Hsian-Rong Tseng, and Ju Dong Yang. 2021. "The Role of Extracellular Vesicles in Disease Progression and Detection of Hepatocellular Carcinoma" Cancers 13, no. 12: 3076. https://doi.org/10.3390/cancers13123076

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