Open Access

Exosomal non‑coding RNAs: Novel biomarkers with emerging clinical applications in gastric cancer (Review)

  • Authors:
    • Xu Lu
    • Yu Zhang
    • Guangfei Xie
    • Ye Ding
    • Hui Cong
    • Shihai Xuan
  • View Affiliations

  • Published online on: September 17, 2020     https://doi.org/10.3892/mmr.2020.11519
  • Pages: 4091-4100
  • Copyright: © Lu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Gastric cancer (GC) is one of the most common types of malignant tumor and it demonstrates high mortality rates. The majority of cases of GC are diagnosed at an advanced stage, which seriously endangers the health of the patient. Therefore, discovering a novel diagnostic method for GC is a current priority. Exosomes are 40 to 150‑nm‑diameter vesicles consisting of a lipid bilayer secreted by a variety of cells that exist in multiple different types of body fluids. Exosomes contain diverse types of active substances, including RNAs, proteins and lipids, and play important roles in tumor cell communication, metastasis and neovascularization, as well as tumor growth. Non‑coding RNAs (ncRNAs) do not code proteins, and instead have roles in a variety of genetic mechanisms, such as regulating the structure, expression and stability of RNAs, and modulating the translation and function of proteins. In recent years, exosomal ncRNAs have become a novel focus in research. An increasing number of studies have demonstrated that exosomal ncRNAs can be used in the prediction and treatment of GC. The present review briefly discusses the role of exosomal ncRNAs as a potential biomarker, and summarizes important regulatory genes involved in the development and progression of GC.

Introduction

Gastric cancr (GeC) is the fifth most common neoplasm and the third most deadly type of cancer according to Globocan 2018 data (1). It is characterized by high metastatic probability, and diagnosis is often made at the late stages of disease (2). The majority of patients with early GC have no overt clinical symptoms (3), though some individuals may experience nausea and vomiting or upper gastrointestinal symptoms similar to that seen with ulcers, all of which lack specificity for the diagnosis of GC (4). In addition, the majority of patients with GC are already in the advanced stage of disease at the time of confirmed diagnosis (5). There is still a lack of effective diagnostic indicators at present, and therefore it is important to determine effective diagnostic and therapeutic targets for early detection and treatment of GC.

Exosomes are a group of extracellular vesicles with a diameter of 30–100 nm released from various cell types into body fluids, including the blood, bile, urine and saliva (6,7). Exosomes were originally considered to be cell debris and were therefore underestimated (8). Over the past decade, increasing attention has been paid to the use of exosomes as a vessel for transferring proteins, lipids and diverse RNA molecules (9), or as a key regulator in the communication of these cargoes with their target cells (10). Accumulating evidence has demonstrated that exosomes play important roles in multiple biological events, such as cell-to-cell communication, cellular metabolism, tumor metastasis, angiogenesis and immune response (11).

Non-coding RNAs (ncRNAs) refer to RNAs that can be transcribed from the genome but with no protein coding capability, so they can function at their respective RNA levels (12). The majority of ncRNAs are functional, including small interfering RNAs (siRNAs), antisense RNAs, microRNAs (miRNAs) and long ncRNAs (lncRNAs) (13). Among them, miRNAs are a type of non-coding single-stranded RNA molecule with a length of 22 nucleotides encoded by endogenous genes (14). It specifically binds to the 3′untranslated region of the target mRNA, thereby causing degradation or translation inhibition of the target mRNA molecule in post-transcriptional gene expression regulation in both animals and plants (15). lncRNAs are ncRNAs that are >200 nucleotides in length. Previous studies have revealed that lncRNAs play important roles in a number of different life activities, such as dose-compensation effects, epigenetic regulation, cell cycle regulation and cell differentiation regulation, and are considered to be a leading topic in genetic research (16). A novel group of endogenous ncRNAs, circularRNAs (circRNAs), have gained increased attention in research (17). circRNAs are a type of ncRNA molecule that lack the 5′(cap) and 3′(polyadenylation) ends and form a ring structure with a covalent bond (18). An increasing number of studies have reported that circRNAs can play key roles in a variety of physiological or pathological processes, including epithelial-to-mesenchymal transition (EMT), angiogenesis, tumor proliferation and tumor metastasis (1921). siRNAs, which are occasionally known as short interfering RNA or silencing RNA, are double-stranded RNAs of 20 to 25 nucleotides in length. It is currently known that siRNA is primarily involved in the phenomenon of RNA interference (RNAi), which regulates gene expression in a specific manner (22). Antisense RNA (asRNA) is a single-stranded RNA complementary to the transcription product mRNA. asRNAs can inhibit translation by binding to mRNA (23). In recent years, ncRNAs, especially miRNAs, lncRNAs and circRNAs, have been suggested to serve as a novel type of biomarker, which differ from the more conventional markers, in addition to participating in the development and progression of different types of cancer (24,25). Thus, ncRNAs have broad application prospects in the diagnosis and treatment of diseases.

In 2007, Valadi et al (26) discovered that exosomes secreted by mouse mastocytosis cells can be captured by human mastocytosis. They were found to be biologically active and could be absorbed by recipient cells, affecting the expression of recipient cells. Taylor and Gercel-Taylor (27) revealed that exosomal miRNAs could be used as markers for cancer diagnosis. They analyzed epithelial cell adhesion molecule-positive exosomes isolated from patients with ovarian cancer and non-cancer sera. By analyzing the differences in miRNA content in exosomes, it was revealed that eight miRNAs could be used as diagnostic markers for various stages in ovarian cancer, thus opening the application of nucleic acid markers in cancer diagnosis. The expression levels of ncRNAs in the serum and tissue has been widely applied in clinical practice, knowing that it is closely associated with the diagnosis, progression and prognosis of various diseases, and could be used as a biomarker for disease detection (28). Compared with serum, exosomal ncRNAs in exosomes derived from serum are more resistant to degradation by lipid bilayer membrane protection and are not easily interfered with by complex array of components found in the serum (29). Therefore, serum exosomal ncRNAs have a useful application in medicine. Recent studies have found that serum exosomal ncRNAs could serve as potential biomarkers in GC (30,31). The present review summarized the diagnostic value and clinical application of exosomal ncRNAs in GC.

Biology and characterization of exosomes

Exosomes are small membrane vesicles containing complex RNAs and proteins; specifically, they are discoid vesicles with a diameter between 30–100 nm (32). They were first found in sheep reticulocytes in 1983 (33), and then named ‘exosome’ by Johnstone et al in 1987 (34). A variety of cells can secrete exosomes under normal and pathological conditions (35). Exosomes are formed by the release of multivesicular bodies, which are produced as intraluminal vesicles (ILVs). ILV sorting and final formation process requires the participation of the endosomal sorting complex required for transport. In addition, two tetraspanins, CD9 and CD36, have also been demonstrated to serve regulatory roles in sorting transmembrane proteins into ILVs, thus promoting its secretion from the cell. They are the most commonly used exosome-identification proteins (6).

When exosomes were first discovered in the 1980s (36,37), they were hypothesized to be a mechanism for cell waste excretion. With in-depth research on the biological origin of exosomes in recent years (38), a variety of properties have been identified, including their material composition and transport (39), their role in the transduction of intracellular signals (40) and their distribution in body fluids (41). Their functions depend on the type of cells from which they originate (42). They can participate in immune responses (43), antigen presentation (44), cell migration (45), cell differentiation (46) and tumor invasion (47). Exosomes can not only be used as markers for early diagnosis of various diseases, but also as carriers of targeted drugs for disease treatment in multiple body fluids, including endothelial cells, immune cells, platelets, and smooth muscle cells (48,49). When exosomes are secreted into recipient cells from host cells, they regulate the biological activity of recipient cells via proteins, nucleic acids and lipids (Fig. 1) (50,51). Tumor immune escape is an important mechanism underlying malignant tumor progression. As a carrier of molecules released by cells, exosomes can not only mediate the interaction between cancer cells and immune cells, but can also inhibit the function of immune cells and promote the proliferation of cancer cells through different mechanisms. This plays a pivotal role in cancer immune surveillance and tumor escape (52). A previous study also confirmed that exosomes can participate in the transmission of immune mediators, such as cytokines and chemokines, and thus participate in the regulation of the tumor microenvironment (53). The extracellular communication mediated by exosomes is primarily established in the following three ways (54). First, exosomal membrane proteins can activate signaling pathways in the target cells by binding to the target cell membrane protein. Secondly, exosomal membrane proteins can be clipped by proteases in the extracellular matrix, and the clipped fragments can be used as ligands to bind to receptors on the cell membrane, thus activating the intracellular signaling pathway (55). It was reported that numerous exosomal membrane proteins could be detected on the cell membrane of their origin (56). Finally, the exosomal membrane could non-selectively release proteins, mRNAs and miRNAs by directly fusing with the target cell membrane (57).

Multiple roles of exosomal ncRNAs in GC

Exosomal ncRNAs could be potential biomarkers for GC

Routine tumor marker screening may require tissue biopsy, puncture and other means to obtain living samples from patients (41); however, this method requires a solid tumor location and therefore is not appropriate for disease screening in healthy individuals. At the same time, tissue biopsy can be harmful to patients (58). Liquid biopsies have emerged as a non-invasive, rapid and reliable detection method that show great development potential and application value. There are broad applications for evaluating the progression of tumor cloning, the efficacy of chemotherapy, the presence of minimal residual disease and acquired resistance in real-time. Non-invasive biomarkers have value in real-time tumor molecular classification and personalized treatment (59). Tumor cell-derived exosomes are expected to replace previous tissue biopsy techniques as a new minimally invasive test (60). The potential utility of miRNAs as biomarkers in both tissues and blood to assess the response of 5-fluorouracil-based therapies and function as EGFR inhibitors has been extensively demonstrated in colorectal cancer (61). Several ncRNAs in exosomes have been demonstrated to be potential diagnostic and predictive biomarkers for GC (Table I). A previous study detected total RNA from plasma exosomes of 67 patients with GC and healthy controls, and revealed that the expression levels of four exosomal miRNAs were consistent with the serum levels (62). Among these, the expression of miR-217 showed a significant upward trend, suggesting that miR-217 may contribute to the occurrence of GC. Soon after this study, Zhao et al (63) reported high expression levels of lncRNA HOTTIP in exosomes isolated from serum samples of patients with GC. Compared with conventional GC biomarkers such as CEA, CA19-9 and CA72-4, exosomal HOTTIP is expected to become a potential novel target for the diagnosis and treatment of GC, with improved specificity and sensitivity. At present, there remains to be a lack of specific minimally invasive biomarkers to distinguish early-stage GC (EGC) and precancerous lesions. It has recently been proposed that EGC-specific exosomal lncUEGC1 and lncUEGC2 could function as non-invasive specific EGC biomarkers (64). A recent study performed exosomal long chain RNA sequencing of plasma specimens from five healthy individuals and 10 patients diagnosed with first-stage GC, as well as four primary gastric epithelial cells and four gastric cancer cells. Combining the sequencing results of plasma samples and culture medium, exosomal lncUEGC1 and lncUEGC2 showed significantly high expression levels and notable changes in expression (30). This provided a strong basis for the diagnosis of EGC using these lncRNAs.

Table I.

Exosomal ncRNAs as biomarkers for gastric cancer.

Table I.

Exosomal ncRNAs as biomarkers for gastric cancer.

A, miRNA

First author, yearMoleculesExosome originExtraction methodIdentification methodTest method(Refs.)
Li et al, 2018miR-217PlasmaDifferential centrifugationNot mentionedRT-qPCR(62)
Ren et al, 2019miR-107SerumCommercial kitTEM and western blottingRT-qPCR(126)
Wang et al, 2017miR-106a-5p and miR-19b-3pSerumCommercial kitTEM and western blottingRT-qPCR(127)
Pan et al, 2017miR-10b-5p, miR-195-5p, miR-20a-3p and miR-296-5pSerumCommercial kitNot mentionedmiRNA microarray and RT-qPCR(76)
Calatayud et al, 2017miR-221Peripheral bloodCommercial kitWestern blottingRT-qPCR(20)
Yang et al, 2018miR-423-5pSerumCommercial kitTEM, NTA and western blottingRT-qPCR(81)
Tokuhisa et al, 2015miR-21, miR-1225-5p, miR-320c and miR-1202Peritoneum lavage fluidDifferential centrifugationNot mentionedmiRNA microarray and RT-qPCR(73)
Ohshima et al, 2010Let-7Cell lineSuccessive centrifugation and ultrafiltrationTEM and western blottingmiRNA microarray and RT-qPCR(128)

B, lncRNA

First author, year MoleculesExosome originExtraction methodIdentification methodTest method(Refs.)

Zhao et al, 2018HOTTIPSerumDifferential centrifugationNot mentionedRT-qPCR   (63)
Cai et al, 2019LINC00152PlasmaCommercial kitTEMRT-qPCR(129)
Pan et al, 2017ZFAS1SerumCommercial kitTEM, NTA and western blottingRT-qPCR   (76)
Lin et al, 2018UEGC1 and UEGC2PlasmaSerial centrifugation and discontinuous iodixanol gradientTEM, NTA and western blottingRNA sequencing and RT-qPCR   (30)

C, circRNA
First author, year MoleculesExosome originExtraction methodIdentification methodTest method(Refs.)

Tang et al, 2018Circ KIAA1244PlasmaCommercial kitNot mentionedcircRNA microarray and RT-qPCR(67)

[i] miR/miRNA, microRNA; lncRNA, long non-coding RNA; circRNA, circular RNA; RT-qPCR, reverse transcription-quantitative RNA; TEM, transmission electron microscopy; NTA, nanoparticle tracking analysis.

In recent years, exosomal circRNAs have gained increasing attention (64,65). Through the use of RNA sequencing, Wang et al (64) analyzed the total RNA content of liver cancer cells and cell-derived exosomes, and discovered the existence of high levels of circRNAs in exosomes (66). This suggested that exosomal circRNAs may be potential biomarkers for tumor detection. Other studies have measured the content of circKIAA1244 in plasma and exosomes derived from plasma, and preliminarily confirmed that circRNAs can exist stably in plasma but they were encapsulated in exosomes (67). However, it is understood that the pathogenesis and clinical application of exosomal circRNAs in GC remains very limited, and needs further investigation.

In the future, it may be possible to combine exosomal ncRNAs with circulating tumor DNA (ctDNA) and circulating tumor cells (CTCs) to provide novel research strategies for liquid biopsies (29). In view of the smaller number of CTCs, ctDNA may become a more practical non-invasive biomarker. ctDNA shows higher accuracy than CTCs in terms of tumor burden, and can be used as both a diagnostic and prognostic biomarker. The application of molecular analysis and mutation identification methods also provide ctDNA with predictive potential in the evaluation of antitumor therapy (68).

Exosomal ncRNAs promote metastasis of GC

GC is a malignant tumor characterized by a high incidence, difficult treatment, and easy metastasis and pervasion (69). Direct spread is one of the main dispersion methods of GC (70). GC cells are often planted in the abdominal cavity and pelvic organs, such as the intestine, ovary, diaphragm, gallbladder and rectal surface, where they often form a local tumor, producing serous or plasma liquid ascites (71). Wang et al (72) found that high expression levels of GC cell-derived exosomal miR-27a could translate fibroblasts into cancer-associated fibroblasts by targeting its downstream target mRNA cysteine and glycine-rich protein 2. Accordingly, it can promote the malignant changes of GC, such as accelerating proliferation and metastasis. Besides blood, ascites can also be used as the primary medium for liquid biopsy in patients with GC. Tokuhisa et al (73) used RNA sequencing to analyze the expression profiles of exosomal miRNAs in the ascites of patients with GC both before and after intraperitoneal chemotherapy and patients with non-malignant disease. They reported that concomitant detection of exosomal miR-21 and miR-1225-5p in ascites could be used as a prognostic and diagnostic marker of peritoneal recurrence after surgery and chemotherapy of patients with GC. Subsequently, another previous study identified differential characteristics of exosomal miRNA profiles in ascites between patients with non-malignant disease and patients with GC before and after intraperitoneal chemotherapy (74). They suggested that miRNAs enclosed in exosomes derived from ascites may prove to be biomarkers for the prognosis of GC peritoneal chemotherapy, thus providing a novel candidate for the treatment of patients with peritoneal metastatic GC.

Lymphatic metastasis is the main pathway of metastasis in GC, and the lymphatic metastasis rate of advanced GC is as high as 70%, which is positively correlated with the depth of tumor invasion (75). Thus, there is an urgent need to find novel therapeutic targets related to lymphatic metastasis in the diagnosis and treatment of GC. Pan et al (76) found that lncRNA ZFAS1 was a highly expressed exosomal ncRNA in the serum samples of patients with GC. In addition, it was also considered to be associated with age, Tumor-Node-Metastasis stage and lymphatic metastasis. The knockdown of ZFAS1 was associated with the increase of E-cadherin and the decrease of N-cadherin. Inhibition of EMT is an important mechanism in tumor invasion and metastasis. Exosomal ZFAS1 was demonstrated to promote the progress of EMT, which in turn promoted lymphatic metastasis in cancer (76).

EMT refers to the phenotypic transformation of epithelial cells to Leydig cells under certain physiological or pathological conditions (77). EMT is an important biological process underlying the migratory and invasive abilities of malignant tumor cells derived from epithelial cells, and is positively correlated with poor prognosis of malignant tumor cells (78). In recent years, EMT has been found to be closely associated with tumor metastasis, which has become the focus of current research (79). EMT is characterized by the downregulation of epithelial cell markers, such as E-cadherin and β-catenin, and the upregulation of mesenchymal phenotypic markers, including N-cadherin and vimentin (80). Exosomal miR-423-5p has been reported to promote the migration and proliferation of GC by restraining the suppressor of fused protein gene, so as to promote the development of EMT in GC cells (81). High expression levels of miR-191 and let-7a in exosomes also supports the promotion of GC by inducing EMT (81,82). EMT can not only enhance the ability of invasion and metastasis of tumor cells, but provides the characteristics of tumor stem cells and promotes the production of cancer stem-like cells (CSCs) (83). CSCs have been considered as the basis of tumor invasion and metastasis (84). The level of CSCs in patients can indicate the probability of recurrence following treatment (85,86). A previous study performed miRNA deep sequencing of exosomes derived from CSCs and screened their differential cells (DCs), which led to the identification of six upregulated miRNAs and five downregulated miRNAs (87). These studies observed significant differences in the type and quantity of miRNAs upregulated in the exosomes from CSCs and DCs. The data provided by this study can help improve the current understanding of the predictive role of CSC-derived exosomal ncRNAs in the development and metastasis of GC.

Exosomal ncRNAs participate in the regulation of GC angiogenesis

Angiogenesis refers to the growth of new capillary blood vessels derived from existing capillaries and post-capillary venules (88). Tumor angiogenesis is an extremely complex process, which generally includes vascular endothelial matrix degradation, endothelial cell migration, endothelial cell proliferation, formation of vascular rings, and formation of a new basement membrane. An increasing number of studies have demonstrated that benign tumors usually grow slowly and rarely exhibit angiogenesis, while the majority of malignant tumors exhibit dense angiogenesis and grow rapidly (89,90). Therefore, angiogenesis plays an important role in the development and metastasis of tumors, and the inhibition of this process will markedly prevent the growth, diffusion and metastasis of tumor tissue. A previous study demonstrated that tumor-derived exosomes are involved in the exchange of genetic information between tumor cells and basal cells, which leads to the formation of ample neovascularization and promotes the growth and invasion of tumors (91). In recent years, several reports have indicated that a variety of exosomal non-coding molecules derived from cancer serum and cells are major inducers of angiogenesis both in vivo and in vitro (9296). There are also associated angiogenic exosomal ncRNAs that have been reported in GC. For instance, high expression levels of exosomal miR-130a in patients with GC was identified to promote tumor proliferation, migration and tubular formation by targeting c-MYB directly in vivo and in vitro experiments (97). This provides a novel strategy for antiangiogenic therapy of GC.

Exosomal ncRNAs as a novel target of chemotherapy drug treatment

Chemotherapy is one of the most important methods in the treatment of malignant tumors (21). However, the drug resistance of tumor cells to chemotherapeutic drugs often leads to the failure of chemotherapy (98). As an important topic in recent years, exosomes have been demonstrated to mediate the drug resistance of tumor cells in a variety of ways (35). Exosomes exist as a cell-to-cell communication mediator in the tumor microenvironment to affect drug resistance (99). They can also participate in the uptake, metabolism and excretion of drugs, thus affecting the drug resistance of tumor cells (100). In addition, drug resistance of tumor cells can also be mediated by the proteins or associated genes in exosomes (101). In recent years, there is more recognition that exosomal miRNAs derived from tumor cells can play important delivery and regulatory functions in the process of chemotherapeutic resistance to diseases (102,103). It is currently speculated that exosomes partially affect the transmission of drug resistance between resistant cells and parental cells. Recurrent and metastatic advanced GC requires a chemotherapy-based comprehensive treatment. Combined use of novel drugs is a new technique in the treatment of advanced GC. Paclitaxel is considered to be the optimum natural anticancer drug found thus far, and has been widely used in the treatment of GC, breast cancer (104), ovarian cancer (105), partial head and neck cancer (106), and lung cancer (107). However, chemotherapy resistance of paclitaxel in patients with GC is an issue that needs to be addressed (108,109). Wang et al (110) identified the delivery mechanism of exosomal miR-155-5p, through which the drug resistance and EMT phenotypes could be observed by establishing a paclitaxel-resistant GC cell line, MGC803R. Adriamycin (ADR) is a member of the anthracycline family. It is often used in combination with certain traditional chemotherapeutic drugs, such as fluorouracil, cisplatin, paclitaxel and mitomycin to treat multiple malignant tumors including GC (111). However, drug resistance to doxorubicin remains to be an obstacle in the treatment of GC. It was found that miR-501 in exosomes secreted by ADR-resistant GC cell line SGC7901/ADR was higher than that in exosomes secreted by sensitive SGC7901 cells (112). It was also found that SGC7901 could ingest Cy3-labeled miR-501 in exosomes from SGC7901/ADR. These experiments on exosomal miR-501 in vitro and in vivo suggested that drug resistance of patients with GC to doxorubicin may be associated with enhanced transmission of exosomal miR-501 by downregulation of BH3-like motif-containing cell death inducer, and subsequent inactivation of caspase-9/-3 and activation of AKT phosphorylation. Several studies have revealed that MSC-derived exosomes have the ability to transmit certain proteins, including multidrug resistance-associated protein 2, (113), copper-transporting ATPase 1 and copper-transporting ATPase 2 (114), as well as certain types of miRNAs, including miR-100 (115), miR-222 (116), miR-30a (117) and miR-17 (118). These exosomal cargoes can activate apoptosis-escaping pathways in ways other than conventional CaM-Ks/Raf/MEK/ERK signaling pathways in order to regulate the cell cycle and alter cell apoptosis rates, thus decreasing the sensitivity of GC cells to 5-fluorouracil (113). Cisplatin is one of the most commonly used classical drugs for chemotherapy and in vitro drug sensitivity tests in patients with GC (119). Its antitumor toxicity and effectiveness have been confirmed, however, in previous years, the emergence of cisplatin resistance has decreased the efficacy of cisplatin, and even led to the failure of chemotherapy for GC, which limits its clinical application (120). A recent study (121) reported that miR-214 overexpression affected the invasion and metastasis of GC cells, resulting in poor prognosis and resistance to apoptosis. It was reported that drug sensitivity to cisplatin in patients with refractory GC could be restored by the mechanism of exosomal-anti-miR-214 delivery to GC cells (121). Studies have revealed that in SGC7901/DDP cells, c-Met siRNA delivered by exosomes reversed the resistance to cisplatin and increased the rate of apoptosis (122). Exosomal miR-21 derived from M2 macrophage is also involved in the mechanism of chemotherapy resistance in GC (123). Exosomal miR-21 can decrease the expression levels of PTEN mRNA and protein. By means of delivering exosomal miR-21, GC cell chemoresistance and the antiapoptotic ability can be enhanced through regulation of the PTEN/PI3K/AKT signaling pathway.

Conclusion

Exosomes, as a nano-sized biological transport carrier, can protect the ncRNAs that they contain against external damage (124). They can promote the exchange of genetic material through the communication of ncRNAs between target cells. In the present review, the mechanisms of ncRNAs carried by exosomes from GC and stromal cells were briefly summarized, with the aim that the information provided herein is conducive to an improved understanding of the position and role of exosomes in the development and progression of GC. Studies on the activities of exosomal ncRNAs in the proliferation, metastasis, angiogenesis and drug resistance of GC are still under progress. These results provide novel research directions and potential therapeutic targets for tumorigenesis. In addition, the potential of exosomal ncRNAs as tumor markers for early liquid biopsies should not be underestimated (125). There is an urgent need to develop a standard method for the rapid, simple and specific separation of exosomes, and detect abnormal exosomes of ncRNA quickly and inexpensively. To the best of our knowledge, the role of known exosomal ncRNAs in GC has not yet been confirmed, and their value in clinical application remains to be investigated. With the gradual increase in research that focuses on exosomes, the application of exosomal ncRNAs in the research and treatment of GC may become a reality.

Acknowledgements

Not applicable.

Funding

The present review was supported by the Jiangsu Provincial Funds for Six Categories of Top Talents (grant no. WS-066); the Research project of Jiangsu Provincial Health and Family Planning Commission (grant no. H201526); and the Nantong Technology Project (grant no. MS12017008-1).

Availability of data and materials

Not applicable.

Authors' contributions

XL conceived, designed and wrote the manuscript. YZ, GX and YD were involved in writing and critically reviewing the manuscript, and also designed the figures. HC and SX checked and modified the language of the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Aichler M, Luber B, Lordick F and Walch A: Proteomic and metabolic prediction of response to therapy in gastric cancer. World J Gastroenterol. 20:13648–13657. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Barreto SG and Windsor JA: Redefining early gastric cancer. Surg Endosc. 30:24–37. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Choi JH, Kim ES, Lee YJ, Cho KB, Park KS, Jang BK, Chung WJ, Hwang JS and Ryu SW: Comparison of quality of life and worry of cancer recurrence between endoscopic and surgical treatment for early gastric cancer. Gastrointest Endosc. 82:299–307. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Jemal A, Siegel R, Ward E, Murray T, Xu J and Thun MJ: Cancer statistics, 2007. CA Cancer J Clin. 57:43–66. 2007. View Article : Google Scholar : PubMed/NCBI

6 

Bobrie A, Colombo M, Raposo G and Thery C: Exosome secretion: Molecular mechanisms and roles in immune responses. Traffic. 12:1659–1668. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Mathivanan S, Ji H and Simpson RJ: Exosomes: Extracellular organelles important in intercellular communication. J Proteomics. 73:1907–1920. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Guo W, Gao Y, Li N, Shao F, Wang C, Wang P, Yang Z, Li R and He J: Exosomes: New players in cancer (Review). Oncol Rep. 38:665–675. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Caruso S and Poon IKH: Apoptotic cell-derived extracellular vesicles: More Than just debris. Front Immunol. 9:14862018. View Article : Google Scholar : PubMed/NCBI

10 

Seo N, Akiyoshi K and Shiku H: Exosome-mediated regulation of tumor immunology. Cancer Sci. 109:2998–3004. 2018. View Article : Google Scholar : PubMed/NCBI

11 

Simons M and Raposo G: Exosomes-vesicular carriers for intercellular communication. Curr Opin Cell Biol. 21:575–581. 2009. View Article : Google Scholar : PubMed/NCBI

12 

Deng G and Sui G: Noncoding RNA in oncogenesis: A new era of identifying key players. Int J Mol Sci. 14:18319–18349. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Wang WT, Han C, Sun YM, Chen TQ and Chen YQ: Noncoding RNAs in cancer therapy resistance and targeted drug development. J Hematol Oncol. 12:552019. View Article : Google Scholar : PubMed/NCBI

14 

Iqbal MA, Arora S, Prakasam G, Calin GA and Syed MA: MicroRNA in lung cancer: Role, mechanisms, pathways and therapeutic relevance. Mol Aspects Med. 70:3–20. 2019. View Article : Google Scholar : PubMed/NCBI

15 

Michlewski G and Caceres JF: Post-transcriptional control of miRNA biogenesis. RNA. 25:1–16. 2019. View Article : Google Scholar : PubMed/NCBI

16 

Zhang XZ, Liu H and Chen SR: Mechanisms of long non-coding rnas in cancers and their dynamic regulations. Cancers (Basel). 12:12452020. View Article : Google Scholar

17 

Memczak S, Jens M, Elefsinioti A, Torti F, Krueger J, Rybak A, Maier L, Mackowiak SD, Gregersen LH, Munschauer M, et al: Circular RNAs are a large class of animal RNAs with regulatory potency. Nature. 495:333–338. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Ma Y, Liu Y and Jiang Z: CircRNAs: A new perspective of biomarkers in the nervous system. Biomed Pharmacother. 128:1102512020. View Article : Google Scholar : PubMed/NCBI

19 

Qiu M, Xia W, Chen R, Wang S, Xu Y, Ma Z, Xu W, Zhang E, Wang J, Fang T, et al: The Circular RNA circPRKCI promotes tumor growth in lung adenocarcinoma. Cancer Res. 78:2839–2851. 2018. View Article : Google Scholar : PubMed/NCBI

20 

Calatayud D, Dehlendorff C, Boisen MK, Hasselby JP, Schultz NA, Werner J, Immervoll H, Molven A, Hansen CP and Johansen JS: Tissue MicroRNA profiles as diagnostic and prognostic biomarkers in patients with resectable pancreatic ductal adenocarcinoma and periampullary cancers. Biomarker Res. 5:82017. View Article : Google Scholar

21 

Ho YJ and Yeh CK: Concurrent anti-vascular therapy and chemotherapy in solid tumors using drug-loaded acoustic nanodroplet vaporization. Acta Biomater. 49:472–485. 2017. View Article : Google Scholar : PubMed/NCBI

22 

Kesharwani P, Gajbhiye V and Jain NK: A review of nanocarriers for the delivery of small interfering RNA. Biomaterials. 33:7138–7150. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Lin CH, Tsai ZT and Wang D: Role of antisense RNAs in evolution of yeast regulatory complexity. Genomics. 102:484–490. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Wong CM, Tsang FH and Ng IO: Non-coding RNAs in hepatocellular carcinoma: Molecular functions and pathological implications. Nat Rev Gastroenterol Hepatol. 15:137–151. 2018. View Article : Google Scholar : PubMed/NCBI

25 

Peng JF, Zhuang YY, Huang FT and Zhang SN: Noncoding RNAs and pancreatic cancer. World J Gastroenterol. 22:801–814. 2016. View Article : Google Scholar : PubMed/NCBI

26 

Valadi H, Ekstrom K, Bossios A, Sjostrand M, Lee JJ and Lotvall JO: Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 9:654–659. 2007. View Article : Google Scholar : PubMed/NCBI

27 

Taylor DD and Gercel-Taylor C: MicroRNA signatures of tumor-derived exosomes as diagnostic biomarkers of ovarian cancer. Gynecol Oncol. 110:13–21. 2008. View Article : Google Scholar : PubMed/NCBI

28 

Xie Y, Dang W, Zhang S, Yue W, Yang L, Zhai X, Yan Q and Lu J: The role of exosomal noncoding RNAs in cancer. Mol Cancer. 18:372019. View Article : Google Scholar : PubMed/NCBI

29 

Skotland T, Sandvig K and Llorente A: Lipids in exosomes: Current knowledge and the way forward. Prog Lipid Res. 66:30–41. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Lin LY, Yang L, Zeng Q, Wang L, Chen ML, Zhao ZH, Ye GD, Luo QC, Lv PY, Guo QW, et al: Tumor-originated exosomal lncUEGC1 as a circulating biomarker for early-stage gastric cancer. Mol Cancer. 17:842018. View Article : Google Scholar : PubMed/NCBI

31 

Zhang X, Wang S, Wang H, Cao J, Huang X, Chen Z, Xu P, Sun G, Xu J, Lv J and Xu Z: Circular RNA circNRIP1 acts as a microRNA-149-5p sponge to promote gastric cancer progression via the AKT1/mTOR pathway. Mol Cancer. 18:202019. View Article : Google Scholar : PubMed/NCBI

32 

Rajagopal C and Harikumar KB: The origin and functions of exosomes in cancer. Front Oncol. 8:662018. View Article : Google Scholar : PubMed/NCBI

33 

Pan BT and Johnstone RM: Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: Selective externalization of the receptor. Cell. 33:967–978. 1983. View Article : Google Scholar : PubMed/NCBI

34 

Johnstone RM, Adam M, Hammond JR, Orr L and Turbide C: Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes). J Biol Chem. 262:9412–9420. 1987.PubMed/NCBI

35 

Milman N, Ginini L and Gil Z: Exosomes and their role in tumorigenesis and anticancer drug resistance. Drug Resist Updat. 45:1–12. 2019. View Article : Google Scholar : PubMed/NCBI

36 

Harding C, Heuser J and Stahl P: Receptor-mediated endocytosis of transferrin and recycling of the transferrin receptor in rat reticulocytes. J Cell Biol. 97:329–339. 1983. View Article : Google Scholar : PubMed/NCBI

37 

Pan BT, Teng K, Wu C, Adam M and Johnstone RM: Electron microscopic evidence for externalization of the transferrin receptor in vesicular form in sheep reticulocytes. J Cell Biol. 101:942–948. 1985. View Article : Google Scholar : PubMed/NCBI

38 

Jan AT, Rahman S, Khan S, Tasduq SA and Choi I: Biology, pathophysiological role, and clinical implications of exosomes: A Critical Appraisal. Cells. 8:992019. View Article : Google Scholar

39 

Ferguson SW and Nguyen J: Exosomes as therapeutics: The implications of molecular composition and exosomal heterogeneity. J Control Release. 228:179–190. 2016. View Article : Google Scholar : PubMed/NCBI

40 

Hannafon BN, Carpenter KJ, Berry WL, Janknecht R, Dooley WC and Ding WQ: Exosome-mediated microRNA signaling from breast cancer cells is altered by the anti-angiogenesis agent docosahexaenoic acid (DHA). Mol Cancer. 14:1332015. View Article : Google Scholar : PubMed/NCBI

41 

Wu Z, Yang Z, Dai Y, Zhu Q and Chen LA: Update on liquid biopsy in clinical management of non-small cell lung cancer. Onco Targets Ther. 12:5097–5109. 2019. View Article : Google Scholar : PubMed/NCBI

42 

Sancho-Albero M, Navascues N, Mendoza G, Sebastián V, Arruebo M, Martín-Duque P and Santamaría J: Exosome origin determines cell targeting and the transfer of therapeutic nanoparticles towards target cells. J Nanobiotechnology. 17:162019. View Article : Google Scholar : PubMed/NCBI

43 

Bai J, Xie X, Lei Y, An G, He L and Chen R: Consideration of dual characters of exosomes in the tumour immune response. Cell Biol Int. 38:538–545. 2014. View Article : Google Scholar : PubMed/NCBI

44 

Arima Y, Liu W, Takahashi Y, Nishikawa M and Takakura Y: Effects of localization of antigen proteins in antigen-loaded exosomes on efficiency of antigen presentation. Mol Pharm. 16:2309–2314. 2019. View Article : Google Scholar : PubMed/NCBI

45 

Zhang X, Shi H, Yuan X, Jiang P, Qian H and Xu W: Tumor-derived exosomes induce N2 polarization of neutrophils to promote gastric cancer cell migration. Mol Cancer. 17:1462018. View Article : Google Scholar : PubMed/NCBI

46 

Lloret-Llinares M, Karadoulama E, Chen Y, Wojenski LA, Villafano GJ, Bornholdt J, Andersson R, Core L, Sandelin A and Jensen TH: The RNA exosome contributes to gene expression regulation during stem cell differentiation. Nucleic Acids Res. 46:11502–11513. 2018. View Article : Google Scholar : PubMed/NCBI

47 

Singh R, Pochampally R, Watabe K, Lu Z and Mo YY: Exosome-mediated transfer of miR-10b promotes cell invasion in breast cancer. Mol Cancer. 13:2562014. View Article : Google Scholar : PubMed/NCBI

48 

Di C, Zhang Q, Wang Y, Wang F, Chen Y, Gan L, Zhou R, Sun C, Li H, Zhang X, et al: Exosomes as drug carriers for clinical application. Artif Cells Nanomed Biotechnol. 46 (sup3):S564–S570. 2018. View Article : Google Scholar : PubMed/NCBI

49 

Izadpanah M, Seddigh A, Ebrahimi Barough S, Fazeli SAS and Ai J: Potential of extracellular vesicles in neurodegenerative diseases: Diagnostic and therapeutic indications. J Mol Neurosci. 66:172–179. 2018. View Article : Google Scholar : PubMed/NCBI

50 

Dhondt B, Van Deun J, Vermaerke S, de Marco A, Lumen N, De Wever O and Hendrix A: Urinary extracellular vesicle biomarkers in urological cancers: From discovery towards clinical implementation. Int J Biochem Cell Biol. 99:236–256. 2018. View Article : Google Scholar : PubMed/NCBI

51 

Roy S, Hochberg FH and Jones PS: Extracellular vesicles: The growth as diagnostics and therapeutics; a survey. J Extracell Vesicles. 7:14387202018. View Article : Google Scholar : PubMed/NCBI

52 

Akers JC, Gonda D, Kim R, Carter BS and Chen CC: Biogenesis of extracellular vesicles (EV): Exosomes, microvesicles, retrovirus-like vesicles, and apoptotic bodies. J Neurooncol. 113:1–11. 2013. View Article : Google Scholar : PubMed/NCBI

53 

Wang X, Yao X, Xie T, Chang Z, Guo Y and Ni H: Exosome-derived uterine miR-218 isolated from cows with endometritis regulates the release of cytokines and chemokines. Microb Biotechnol. 13:1103–1117. 2020. View Article : Google Scholar : PubMed/NCBI

54 

Mathieu M, Martin-Jaular L, Lavieu G and Thery C: Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Cell Biol. 21:9–17. 2019.

55 

Klingeborn M, Dismuke WM, Bowes Rickman C and Stamer WD: Roles of exosomes in the normal and diseased eye. Prog Retin Eye Res. 59:158–177. 2017. View Article : Google Scholar : PubMed/NCBI

56 

Xitong D and Xiaorong Z: Targeted therapeutic delivery using engineered exosomes and its applications in cardiovascular diseases. Gene. 575:377–384. 2016. View Article : Google Scholar : PubMed/NCBI

57 

Yang XX, Sun C, Wang L and Guo XL: New insight into isolation, identification techniques and medical applications of exosomes. J Control Release. 308:119–129. 2019. View Article : Google Scholar : PubMed/NCBI

58 

Lopez-Beltran A, Cheng L, Gevaert T, Blanca A, Cimadamore A, Santoni M, Massari F, Scarpelli M, Raspollini MR and Montironi R: Current and emerging bladder cancer biomarkers with an emphasis on urine biomarkers. Expert Rev Mol Diagn. 20:231–243. 2020. View Article : Google Scholar : PubMed/NCBI

59 

Normanno N, Cervantes A, Ciardiello F, De Luca A and Pinto C: The liquid biopsy in the management of colorectal cancer patients: Current applications and future scenarios. Cancer Treat Rev. 70:1–8. 2018. View Article : Google Scholar : PubMed/NCBI

60 

Buscail E, Maulat C, Muscari F, Chiche L, Cordelier P, Dabernat S, Alix-Panabières C and Buscail L: Liquid biopsy approach for pancreatic ductal adenocarcinoma. Cancers. 11:8522019. View Article : Google Scholar

61 

Boussios S, Ozturk MA, Moschetta M, Karathanasi A, Zakynthinakis-Kyriakou N, Katsanos KH, Christodoulou DK and Pavlidis N: The developing story of predictive biomarkers in colorectal cancer. J Pers Med. 9:122019. View Article : Google Scholar

62 

Li W and Gao YQ: MiR-217 is involved in the carcinogenesis of gastric cancer by down-regulating CDH1 expression. Kaohsiung J Med Sci. 34:377–384. 2018. View Article : Google Scholar : PubMed/NCBI

63 

Zhao R and Zhang Y, Zhang X, Yang Y, Zheng X, Li X, Liu Y and Zhang Y: Exosomal long noncoding RNA HOTTIP as potential novel diagnostic and prognostic biomarker test for gastric cancer. Mol Cancer. 17:682018. View Article : Google Scholar : PubMed/NCBI

64 

Wang G, Liu W, Zou Y, Wang G, Deng Y, Luo J, Zhang Y, Li H, Zhang Q, Yang Y and Chen G: Three isoforms of exosomal circPTGR1 promote hepatocellular carcinoma metastasis via the miR449a-MET pathway. EBioMedicine. 40:432–445. 2019. View Article : Google Scholar : PubMed/NCBI

65 

Li J, Li Z, Jiang P, Peng M, Zhang X, Chen K, Liu H, Bi H, Liu X and Li X: Circular RNA IARS (circ-IARS) secreted by pancreatic cancer cells and located within exosomes regulates endothelial monolayer permeability to promote tumor metastasis. J Exp Clin Cancer Res. 37:1772018. View Article : Google Scholar : PubMed/NCBI

66 

Li Y, Zheng Q, Bao C, Li S, Guo W, Zhao J, Chen D, Gu J, He X and Huang S: Circular RNA is enriched and stable in exosomes: A promising biomarker for cancer diagnosis. Cell Res. 25:981–984. 2015. View Article : Google Scholar : PubMed/NCBI

67 

Tang W, Fu K, Sun H, Rong D, Wang H and Cao H: CircRNA microarray profiling identifies a novel circulating biomarker for detection of gastric cancer. Mol Cancer. 17:1372018. View Article : Google Scholar : PubMed/NCBI

68 

Zarkavelis G, Boussios S, Papadaki A, Katsanos KH, Christodoulou DK and Pentheroudakis G: Current and future biomarkers in colorectal cancer. Ann Gastroenterol. 30:613–621. 2017.PubMed/NCBI

69 

Vedeld HM, Goel A and Lind GE: Epigenetic biomarkers in gastrointestinal cancers: The current state and clinical perspectives. Semin Cancer Biol. 51:36–49. 2018. View Article : Google Scholar : PubMed/NCBI

70 

Shah M, Prasad A, Rajan D, Tan CB, Shah M, Raghavan P and Mustacchia P: Direct liver invasion from a gastric adenocarcinoma as an initial presentation of extranodal tumor spread. Case Rep Med. 2012:6512322012. View Article : Google Scholar : PubMed/NCBI

71 

Ruggieri V, Russi S, Zoppoli P, La Rocca F, Angrisano T, Falco G, Calice G and Laurino S: The Role of MicroRNAs in the regulation of gastric cancer stem cells: A meta-analysis of the current status. J Clin Med. 8:6392019. View Article : Google Scholar

72 

Wang J, Guan X, Zhang Y, Ge S, Zhang L, Li H, Wang X, Liu R, Ning T, Deng T, et al: Exosomal miR-27a derived from gastric cancer cells regulates the transformation of fibroblasts into cancer-associated fibroblasts. Cell Physiol Biochem. 49:869–883. 2018. View Article : Google Scholar : PubMed/NCBI

73 

Tokuhisa M, Ichikawa Y, Kosaka N, Ochiya T, Yashiro M, Hirakawa K, Kosaka T, Makino H, Akiyama H, Kunisaki C and Endo I: Exosomal miRNAs from peritoneum lavage fluid as potential prognostic biomarkers of peritoneal metastasis in gastric cancer. PLoS One. 10:e01304722015. View Article : Google Scholar : PubMed/NCBI

74 

Hu Y, Qi C, Liu X, Zhang C, Gao J, Wu Y, Yang J, Zhao Q, Li J, Wang X and Shen L: Malignant ascites-derived exosomes promote peritoneal tumor cell dissemination and reveal a distinct miRNA signature in advanced gastric cancer. Cancer Lett. 457:142–150. 2019. View Article : Google Scholar : PubMed/NCBI

75 

Wang ZK, Lin JX, Li P, Xie JW, Wang JB, Lu J, Chen QY, Cao LL, Lin M, Tu RH, et al: Higher risk of lymph node metastasis in young patients with early gastric cancer. J Cancer. 10:4389–4396. 2019. View Article : Google Scholar : PubMed/NCBI

76 

Pan L, Liang W, Fu M, Huang ZH, Li X, Zhang W, Zhang P, Qian H, Jiang PC, Xu WR and Zhang X: Exosomes-mediated transfer of long noncoding RNA ZFAS1 promotes gastric cancer progression. J Cancer Res Clin Oncol. 143:991–1004. 2017. View Article : Google Scholar : PubMed/NCBI

77 

Bagger SO, Hopkinson BM, Pandey DP, Bak M, Brydholm AV, Villadsen R, Helin K, Rønnov-Jessen L, Petersen OW and Kim J: Aggressiveness of non-EMT breast cancer cells relies on FBXO11 activity. Mol Cancer. 17:1712018. View Article : Google Scholar : PubMed/NCBI

78 

Colella B, Faienza F and Di Bartolomeo S: EMT regulation by autophagy: A new perspective in glioblastoma biology. Cancers. 11:3122019. View Article : Google Scholar

79 

Aiello NM, Maddipati R, Norgard RJ, Balli D, Li J, Yuan S, Yamazoe T, Black T, Sahmoud A, Furth EE, et al: EMT subtype influences epithelial plasticity and mode of cell migration. Dev Cell. 45:681–695.e684. 2018. View Article : Google Scholar : PubMed/NCBI

80 

Pastushenko I and Blanpain C: EMT Transition States during Tumor Progression and Metastasis. Trends Cell Biol. 29:212–226. 2019. View Article : Google Scholar : PubMed/NCBI

81 

Yang H, Fu H, Wang B, Zhang X, Mao J, Li X, Wang M, Sun Z, Qian H and Xu W: Exosomal miR-423-5p targets SUFU to promote cancer growth and metastasis and serves as a novel marker for gastric cancer. Mol Carcinog. 57:1223–1236. 2018. View Article : Google Scholar : PubMed/NCBI

82 

Tian W, Liu S and Li B: Potential role of exosomes in cancer metastasis. Biomed Res Int. 2019:46497052019. View Article : Google Scholar : PubMed/NCBI

83 

Rodriguez-Aznar E, Wiesmuller L, Sainz B Jr and Hermann PC: EMT and stemness-key players in pancreatic cancer stem cells. Cancers. 11:11362019. View Article : Google Scholar

84 

Lee IC, Fadera S and Liu HL: Strategy of differentiation therapy: effect of dual-frequency ultrasound on the induction of liver cancer stem-like cells on a HA-based multilayer film system. J Mater Chem B. 7:5401–5411. 2019. View Article : Google Scholar : PubMed/NCBI

85 

Park SY, Choi JH and Nam JS: Targeting cancer stem cells in triple-negative breast cancer. Cancers (Basel). 11:9652019. View Article : Google Scholar

86 

Peitzsch C, Tyutyunnykova A, Pantel K and Dubrovska A: Cancer stem cells: The root of tumor recurrence and metastases. Semin Cancer Biol. 44:10–24. 2017. View Article : Google Scholar : PubMed/NCBI

87 

Sun ZP, Li AQ, Jia WH, Ye S, Van Eps G, Yu JM and Yang WJ: MicroRNA expression profiling in exosomes derived from gastric cancer stem-like cells. Oncotarget. 8:93839–93855. 2017. View Article : Google Scholar : PubMed/NCBI

88 

Viallard C and Larrivee B: Tumor angiogenesis and vascular normalization: alternative therapeutic targets. Angiogenesis. 20:409–426. 2017. View Article : Google Scholar : PubMed/NCBI

89 

Mao G, Liu Y, Fang X, Liu Y, Fang L, Lin L, Liu X and Wang N: Tumor-derived microRNA-494 promotes angiogenesis in non-small cell lung cancer. Angiogenesis. 18:373–382. 2015. View Article : Google Scholar : PubMed/NCBI

90 

Dong H, Weng C, Bai R, Sheng J, Gao X, Li L and Xu Z: The regulatory network of miR-141 in the inhibition of angiogenesis. Angiogenesis. 22:251–262. 2019. View Article : Google Scholar : PubMed/NCBI

91 

Bikfalvi A: History and conceptual developments in vascular biology and angiogenesis research: A personal view. Angiogenesis. 20:463–478. 2017. View Article : Google Scholar : PubMed/NCBI

92 

Zhao S, Li J, Zhang G, Wang Q, Wu C, Zhang Q, Wang H, Sun P, Xiang R and Yang S: Exosomal miR-451a functions as a tumor suppressor in hepatocellular carcinoma by targeting LPIN1. Cell Physiol Biochem. 53:19–35. 2019. View Article : Google Scholar : PubMed/NCBI

93 

Wang ZF, Liao F, Wu H and Dai J: Glioma stem cells-derived exosomal miR-26a promotes angiogenesis of microvessel endothelial cells in glioma. J Exp Clin Cancer Res. 38:2012019. View Article : Google Scholar : PubMed/NCBI

94 

Wu XG, Zhou CF, Zhang YM, Yan RM, Wei WF, Chen XJ, Yi HY, Liang LJ, Fan LS, Liang L, et al: Cancer-derived exosomal miR-221-3p promotes angiogenesis by targeting THBS2 in cervical squamous cell carcinoma. Angiogenesis. 22:397–410. 2019. View Article : Google Scholar : PubMed/NCBI

95 

Hu HY, Yu CH, Zhang HH, Zhang SZ, Yu WY, Yang Y and Chen Q: Exosomal miR-1229 derived from colorectal cancer cells promotes angiogenesis by targeting HIPK2. Int J Biol Macromol. 132:470–477. 2019. View Article : Google Scholar : PubMed/NCBI

96 

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. 64:792–802. 2019. View Article : Google Scholar : PubMed/NCBI

97 

Yang H, Zhang H, Ge S, Ning T, Bai M, Li J, Li S, Sun W, Deng T, Zhang L, et al: Exosome-Derived miR-130a activates angiogenesis in gastric cancer by targeting C-MYB in vascular endothelial cells. Mol Ther. 26:2466–2475. 2018. View Article : Google Scholar : PubMed/NCBI

98 

Hida K, Kikuchi H, Maishi N and Hida Y: ATP-binding cassette transporters in tumor endothelial cells and resistance to metronomic chemotherapy. Cancer Lett. 400:305–310. 2017. View Article : Google Scholar : PubMed/NCBI

99 

Zhang HD, Jiang LH, Hou JC, Zhong SL, Zhu LP, Wang DD, Zhou SY, Yang SJ, Wang JY, Zhang Q, et al: Exosome: A novel mediator in drug resistance of cancer cells. Epigenomics. 10:1499–1509. 2018. View Article : Google Scholar : PubMed/NCBI

100 

Qu Z, Wu J, Wu J, Luo D, Jiang C and Ding Y: Exosomes derived from HCC cells induce sorafenib resistance in hepatocellular carcinoma both in vivo and in vitro. J Exp Clin Cancer Res. 35:1592016. View Article : Google Scholar : PubMed/NCBI

101 

Zhang W, Cai X, Yu J, Lu X, Qian Q and Qian W: Exosome-mediated transfer of lncRNA RP11838N2.4 promotes erlotinib resistance in non-small cell lung cancer. Int J Oncol. 53:527–538. 2018.PubMed/NCBI

102 

Wei F, Ma C, Zhou T, Dong X, Luo Q, Geng L, Ding L, Zhang Y, Zhang L, Li N, et al: Exosomes derived from gemcitabine-resistant cells transfer malignant phenotypic traits via delivery of miRNA-222-3p. Mol Cancer. 16:1322017. View Article : Google Scholar : PubMed/NCBI

103 

Qin X, Yu S, Zhou L, Shi M, Hu Y, Xu X, Shen B, Liu S, Yan D and Feng J: Cisplatin-resistant lung cancer cell-derived exosomes increase cisplatin resistance of recipient cells in exosomal miR-100-5p-dependent manner. Int J Nanomedicine. 12:3721–3733. 2017. View Article : Google Scholar : PubMed/NCBI

104 

Xie F, Chen R, Zhang L, Yin Z, Zhu Q, You S, Jiang C, Li Y, Li S, Zha X and Wang J: Efficacy of two-weekly nanoparticle albumin-bound paclitaxel as neoadjuvant chemotherapy for breast cancer. Nanomedicine (Lond). 14:1595–1603. 2019. View Article : Google Scholar : PubMed/NCBI

105 

Vergote I, Scambia G, O'Malley DM, Van Calster B, Park SY, Del Campo JM, Meier W, Bamias A, Colombo N, Wenham RM, et al: Trebananib or placebo plus carboplatin and paclitaxel as first-line treatment for advanced ovarian cancer (TRINOVA-3/ENGOT-ov2/GOG-3001): A randomised, double-blind, phase 3 trial. Lancet Oncol. 20:862–876. 2019. View Article : Google Scholar : PubMed/NCBI

106 

Colevas AD: Systemic therapy for metastatic or recurrent squamous cell carcinoma of the head and neck. J Natl Compr Canc Netw. 13:e37–e48. 2015. View Article : Google Scholar : PubMed/NCBI

107 

Socinski MA, Okamoto I, Hon JK, Hirsh V, Dakhil SR, Page RD, Orsini J, Yamamoto N, Zhang H and Renschler MF: Safety and efficacy analysis by histology of weekly nab-paclitaxel in combination with carboplatin as first-line therapy in patients with advanced non-small-cell lung cancer. Ann Oncol. 24:2390–2396. 2013. View Article : Google Scholar : PubMed/NCBI

108 

Guo Z, Wang X, Lin R, Chen L, Fan N, Chen Y, Lin J and Yu J: Paclitaxel-based regimens as first-line treatment in advanced gastric cancer. J Chemother. 27:94–98. 2015. View Article : Google Scholar : PubMed/NCBI

109 

Zhang D and Fan D: Multidrug resistance in gastric cancer: Recent research advances and ongoing therapeutic challenges. Expert Rev Anticancer Ther. 7:1369–1378. 2007. View Article : Google Scholar : PubMed/NCBI

110 

Wang M, Qiu R, Yu S, Xu X, Li G, Gu R, Tan C, Zhu W and Shen B: Paclitaxelresistant gastric cancer MGC803 cells promote epithelialtomesenchymal transition and chemoresistance in paclitaxelsensitive cells via exosomal delivery of miR1555p. Int J Oncol. 54:326–338. 2019.PubMed/NCBI

111 

Hultman B, Mahteme H, Sundbom M, Ljungman M, Larsson R and Nygren P: Benchmarking of gastric cancer sensitivity to anti-cancer drugs ex vivo as a basis for drug selection in systemic and intraperitoneal therapy. J Exp Clin Cancer Res. 33:1102014. View Article : Google Scholar : PubMed/NCBI

112 

Liu X, Lu Y, Xu Y, Hou S, Huang J, Wang B, Zhao J, Xia S, Fan S, Yu X, et al: Exosomal transfer of miR-501 confers doxorubicin resistance and tumorigenesis via targeting of BLID in gastric cancer. Cancer Lett. 459:122–134. 2019. View Article : Google Scholar : PubMed/NCBI

113 

Ji R, Zhang B, Zhang X, Xue J, Yuan X, Yan Y, Wang M, Zhu W, Qian H and Xu W: Exosomes derived from human mesenchymal stem cells confer drug resistance in gastric cancer. Cell Cycle. 14:2473–2483. 2015. View Article : Google Scholar : PubMed/NCBI

114 

Panfoli I, Ravera S, Podestà M, Cossu C, Santucci L, Bartolucci M, Bruschi M, Calzia D, Sabatini F, Bruschettini M, et al: Exosomes from human mesenchymal stem cells conduct aerobic metabolism in term and preterm newborn infants. FASEB J. 30:1416–1424. 2016. View Article : Google Scholar : PubMed/NCBI

115 

Pakravan K, Babashah S, Sadeghizadeh M, Mowla SJ, Mossahebi-Mohammadi M, Ataei F, Dana N and Javan M: MicroRNA-100 shuttled by mesenchymal stem cell-derived exosomes suppresses in vitro angiogenesis through modulating the mTOR/HIF-1α/VEGF signaling axis in breast cancer cells. Cell Oncol (Dordr). 40:457–470. 2017. View Article : Google Scholar : PubMed/NCBI

116 

Bliss SA, Sinha G, Sandiford OA, Williams LM, Engelberth DJ, Guiro K, Isenalumhe LL, Greco SJ, Ayer S, Bryan M, et al: Mesenchymal stem cell-derived exosomes stimulate cycling quiescence and early breast cancer dormancy in bone marrow. Cancer Res. 76:5832–5844. 2016. View Article : Google Scholar : PubMed/NCBI

117 

Zhang H, Wang Y, Yang G, Yu H, Zhou Z and Tang M: MicroRNA-30a regulates chondrogenic differentiation of human bone marrow-derived mesenchymal stem cells through targeting Sox9. Exp Ther Med. 18:4689–4697. 2019.PubMed/NCBI

118 

Xin H, Liu Z, Buller B, Li Y, Golembieski W, Gan X, Wang F, Lu M, Ali MM, Zhang ZG and Chopp M: MiR-17-92 enriched exosomes derived from multipotent mesenchymal stromal cells enhance axon-myelin remodeling and motor electrophysiological recovery after stroke. J Cereb Blood Flow Metab. 2020.(Ahead of print). View Article : Google Scholar

119 

Cheng Q, Li X and Liu J, Ye Q, Chen Y, Tan S and Liu J: Multiple myeloma-derived exosomes regulate the functions of mesenchymal stem cells partially via modulating miR-21 and miR-146a. Stem Cells Int. 2017:90121522017. View Article : Google Scholar : PubMed/NCBI

120 

Ho GY, Woodward N and Coward JI: Cisplatin versus carboplatin: comparative review of therapeutic management in solid malignancies. Crit Rev Oncol Hematol. 102:37–46. 2016. View Article : Google Scholar : PubMed/NCBI

121 

Wang X, Zhang H, Bai M, Ning T, Ge S, Deng T, Liu R, Zhang L, Ying G and Ba Y: Exosomes serve as nanoparticles to deliver anti-miR-214 to reverse chemoresistance to cisplatin in gastric cancer. Mol Ther. 26:774–783. 2018. View Article : Google Scholar : PubMed/NCBI

122 

Zhang Q, Zhang H, Ning T, Liu D, Deng T, Liu R, Bai M, Zhu K, Li J, Fan Q, et al: Exosome-delivered c-met sirna could reverse chemoresistance to cisplatin in gastric cancer. Int J Nanomedicine. 15:2323–2335. 2020. View Article : Google Scholar : PubMed/NCBI

123 

Zheng P, Chen L, Yuan X, Luo Q, Liu Y, Xie G, Ma Y and Shen L: Exosomal transfer of tumor-associated macrophage-derived miR-21 confers cisplatin resistance in gastric cancer cells. J Exp Clin Cancer Res. 36:532017. View Article : Google Scholar : PubMed/NCBI

124 

van der Pol E, Boing AN, Harrison P, Sturk A and Nieuwland R: Classification, functions, and clinical relevance of extracellular vesicles. Pharmacol Rev. 64:676–705. 2012. View Article : Google Scholar : PubMed/NCBI

125 

Palmirotta R, Lovero D, Cafforio P, Felici C, Mannavola F, Pellè E, Quaresmini D, Tucci M and Silvestris F: Liquid biopsy of cancer: A multimodal diagnostic tool in clinical oncology. Ther Adv Med Oncol. 10:17588359187946302018. View Article : Google Scholar : PubMed/NCBI

126 

Ren W, Zhang X, Li W, Feng Q, Feng H, Tong Y, Rong H, Wang W, Zhang D, Zhang Z, et al: Exosomal miRNA-107 induces myeloid-derived suppressor cell expansion in gastric cancer. Cancer Manag Res. 11:4023–4040. 2019. View Article : Google Scholar : PubMed/NCBI

127 

Wang N, Wang L, Yang Y, Gong L, Xiao B and Liu X: A serum exosomal microRNA panel as a potential biomarker test for gastric cancer. Biochem Biophys Res Commun. 493:1322–1328. 2017. View Article : Google Scholar : PubMed/NCBI

128 

Ohshima K, Inoue K, Fujiwara A, Hatakeyama K, Kanto K, Watanabe Y, Muramatsu K, Fukuda Y, Ogura S, Yamaguchi K and Mochizuki T: Let-7 microRNA family is selectively secreted into the extracellular environment via exosomes in a metastatic gastric cancer cell line. PLoS One. 5:e132472010. View Article : Google Scholar : PubMed/NCBI

129 

Cai C, Zhang H, Zhu Y, Zheng P, Xu Y, Sun J, Zhang M, Lan T, Gu B, Li S and Ma P: Serum exosomal long noncoding RNA pcsk2-2:1 as a potential novel diagnostic biomarker for gastric cancer. Onco Targets Ther. 12:10035–10041. 2019. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2020
Volume 22 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lu X, Zhang Y, Xie G, Ding Y, Cong H and Xuan S: Exosomal non‑coding RNAs: Novel biomarkers with emerging clinical applications in gastric cancer (Review). Mol Med Rep 22: 4091-4100, 2020
APA
Lu, X., Zhang, Y., Xie, G., Ding, Y., Cong, H., & Xuan, S. (2020). Exosomal non‑coding RNAs: Novel biomarkers with emerging clinical applications in gastric cancer (Review). Molecular Medicine Reports, 22, 4091-4100. https://doi.org/10.3892/mmr.2020.11519
MLA
Lu, X., Zhang, Y., Xie, G., Ding, Y., Cong, H., Xuan, S."Exosomal non‑coding RNAs: Novel biomarkers with emerging clinical applications in gastric cancer (Review)". Molecular Medicine Reports 22.5 (2020): 4091-4100.
Chicago
Lu, X., Zhang, Y., Xie, G., Ding, Y., Cong, H., Xuan, S."Exosomal non‑coding RNAs: Novel biomarkers with emerging clinical applications in gastric cancer (Review)". Molecular Medicine Reports 22, no. 5 (2020): 4091-4100. https://doi.org/10.3892/mmr.2020.11519