Skip to main content

REVIEW article

Front. Cell Dev. Biol., 05 November 2021
Sec. Cellular Biochemistry
This article is part of the Research Topic Biology and Pharmacological Effects of Extracellular Vesicles in Cancer View all 16 articles

The Biology and Function of Extracellular Vesicles in Cancer Development

Xinyi Zhang&#x;Xinyi Zhang1Dianfeng Liu&#x;Dianfeng Liu1Yongjian GaoYongjian Gao2Chao Lin,Chao Lin3,4Qingwu AnQingwu An1Ye FengYe Feng2Yangyang LiuYangyang Liu3Da LiuDa Liu3Haoming Luo
Haoming Luo3*Dongxu Wang
Dongxu Wang1*
  • 1Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
  • 2Department of Hepatobiliary and Pancreas Surgery, China–Japan Union Hospital of Jilin University, Changchun, China
  • 3Department of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
  • 4School of Grain Science and Technology, Jilin Business and Technology College, Changchun, China

Extracellular vesicles (EVs) exert their biological functions by delivering proteins, metabolites, and nucleic acids to recipient cells. EVs play important roles in cancer development. The anti-tumor effect of EVs is by their cargos carrying proteins, metabolites, and nucleic acids to affect cell-to-cell communication. The characteristics of cell-to-cell communication can potentially be applied for the therapy of cancers, such as gastric cancer. In addition, EVs can be used as an effective cargos to deliver ncRNAs, peptides, and drugs, to target tumor tissues. In addition, EVs have the ability to regulate cell apoptosis, autophagy, proliferation, and migration of cancer cells. The ncRNA and peptides that were engaged with EVs were associated with cell signaling pathways in cancer development. This review focuses on the composition, cargo, function, mechanism, and application of EVs in cancers.

Introduction

EVs are 40–100 nm extracellular vesicles that are released by cells (Kahlert and Kalluri, 2013). EVs were initially observed in sheep reticulocytes in the 1980s (Raposo and Stoorvogel, 2013). Recently, studies have focused on the source of their endocytosis and on distinguishing them from micro-vesicles (Théry et al., 2002). EVs have anti-tumor functions associated with the development of a variety of cancers, such as breast, stomach, liver, and lung cancers (Table 1).

TABLE 1
www.frontiersin.org

TABLE 1. The function of EVs in cancers.

The Biogenesis and Composition of EVs

Mammalian cell, EVs are highly heterogeneous. They contain lipid membranes, proteins, RNAs, and DNAs (Kowal et al., 2016). The lipid membrane of EVs carries the ligands and receptors from the source cells and has a role in cell-to-cell communication (Valadi et al., 2007; Kahlert et al., 2014). Due to the specificity of the lipid membrane, EVs can invade target cells through biogenesis (Balaj et al., 2011). The components on the membrane also play a key role in cell-to-cell communication (Wu et al., 2021). EVs use lipid membranes to enter recipient cells to release cargo and affect recipient cells. These characteristics indicate that EVs have potential applications in regulating cancer development.

The Formation of EVs

Many EVs formed from normal and pathological cells. In contrast to micro-vesicles, EVs are mainly derived from multivesicular bodies (MVBs) that are formed by intracellular lysosomal particles. EVs are released into the extracellular matrix through the fusion of the outer membrane of the MVBs with the membrane of source cells (Figure 1). Specifically, EVs are formed through the endosomal pathway. First, the endosome is formed by the invasion of the plasma membrane during cell maturation process (Harding et al., 1983). The endosome is a membrane-encapsulated vesicular structure and includes both early and late endosomes. Early endosomes are usually located outside of the cytoplasm. In contrast, late endosomes are located inside of the cytoplasm, near the nucleus. Endosomes are acidic vesicles without lysosomal enzymes (Bainton and Farquhar, 1968). The invasion of endosomes produces MVBs which contain 40–150 nm vesicles. The inner membrane forms intraluminal vesicles (ILV). Finally, the late lysosome melts or fuses with the plasma membrane of the source cell and degrades MVBS to release EVs (Harding et al., 1983). This process is known as EV biogenesis and is different from apoptotic bodies (Taylor and Gercel-Taylor, 2008). EVs are widely observed in tumor cells, mesenchymal stem cells, fibroblasts, neurons, endothelial cells (ECs), and epithelial cells (Kalluri, 2016). Previous reports have suggested that the tumor cells can specifically absorb their own secreted EVs (Kahlert and Kalluri, 2013). This implies that during the formation of EVs, specific biomarkers are formed on the surface of the EVs. These biomarkers are the cues that render EVs to be absorbed by specific cells.

FIGURE 1
www.frontiersin.org

FIGURE 1. Formation of EVs.

EVs Cargo

Nucleic acids such as DNAs or RNAs, proteins, or drugs can be carried in EVs as cargo to be delivered for cell-to-cell communication (Figure 2). In the past decades, miRNAs and mRNAs have been found to be major components of EVs. The improvement of EV detection techniques has allowed more RNA species, including transfer RNAs (tRNAs), long non-coding RNAs (lncRNAs), and viral RNAs, to be observed (Valadi et al., 2007; Su et al., 2021). An increasing amount of data suggests that these RNAs, such as lncRNA, have crucial functions that affect the development of cancer cells (Gusachenko et al., 2013). Moreover, numerous studies have demonstrated that the abnormal expressions of miRNAs, lncRNAs, and mRNAs are associated with cancer development (Chan and Tay, 2018; Huang et al., 2020). Hence, these RNAs, that are contained within EVs, can either preserve or degrade their target genes.

FIGURE 2
www.frontiersin.org

FIGURE 2. The contents of EVs.

Cancers develop because of the expression and interaction of numerous genes or proteins. EVs can express proteins through genetic engineering (Silva et al., 2021). The EVs were obtained from the source cells that were transfected with the target gene plasmids. These EVs contain the synthesized proteins or peptides through cell culture (Perin et al., 2011). There is evidence that fusing the exosomally-enriched membrane protein (Lamp 2b) with the ischemic myocardium‐targeting peptide (IMTP) can be used to inhibit cancer development by molecular cloning lentiviral packaging protocols (Fernández et al., 2002). EVs secreted by tumor cells can be taken up by the same tumor cell with specificity. Some molecules (such as Let-7a) can be easily introduced to donor cells through EVs, and tumor targeting EVs carrying these molecules can be used for cancer treatment (Wu et al., 2021). In addition, EVs can carry various chemotherapeutic drugs and materials for targeted treatment of cancers (Wang et al., 2019a).

EVs can Decide Cell Fate

The function of EVs depends on the source cells, such as tumor cells or stem cells (Draganov et al., 2019; Dzobo et al., 2020). The EVs released from these source cells can affect the apoptosis, growth, cell cycle, migration, invasion, and differentiation of recipient cells. Previous studies have indicated that tumor-released EVs could deliver genetic information to the recipient cells for cell-to-cell communication (Valadi et al., 2007). This process promotes cell growth, invasion, and active angiogenesis in a tumor microenvironment (Figure 3).

FIGURE 3
www.frontiersin.org

FIGURE 3. EVs decide cell fate.

Initially, EVs were considered to be “garbage bags” that could not affect other cells (Kalluri, 2016). However, it was found that EVs could be absorbed by target cells and their cargos could be released to affect cell signaling transduction, therefore determining the fate of the recipient cells (Pan et al., 1985). Additional evidence suggested that tumor cells released EVs that promoted tumor growth and invasion in vivo (Ramírez-Ricardo et al., 2020). EVs that carried tumor suppressors, such as let-7a, could inhibited tumor growth (Melo et al., 2014).

The Function of EVs in Cell Proliferation

Indefinite proliferation is a key feature of tumor cells. The abnormal cell cycle of tumor cells is associated with un-controlled cell growth. Previous reports confirmed that miRNA-122 was involved in the cell cycle as well as the proliferation of hepatocellular carcinoma (HCC) cells (Fernández et al., 2002; Xu et al., 2011). A recent report showed that the EVs carrying circRNA plays a role in the proliferation of HCC cells (Xue et al., 2017). In addition, arsenite could increase the expression of circRNA_100284 carried by EVs, altering the cell cycle and their proliferation by acting on miR-217 (Lu et al., 2015). The expression of the cell proliferation biomarkers E2H2 and cyclin D1 were regulated by the circRNA_100284 contained within EVs, and the expression of circRASSF2 was increased in laryngeal squamous cell carcinoma (LSCC) tissue compared to paracancerous tissue. The circRASSF2 carried by EVs promoted LSCC cell growth via the miR-302B-3p/IGF-1R axis (Tian et al., 2019). Thus, EVs have the ability to regulate cell proliferation through their cargos.

The Function of EVs in Epithelial-Mesenchymal Transition

The cell-to-cell communication in tumors might promote EMT of cancers. Previous data has shown that the EV-released circRNA PED8A was associated with increased lymphatic invasion, TNM staging, and low survival rate of patients. Furthermore, the circRNA PED8A from EVs promoted tumor cell growth by activating MET, which is a tyrosine kinase receptor (Luna et al., 2019). In addition, the release of circRNA PED8A contained within EVs into the blood circulation promotes invasion and metastasis through the MACC-MET-ERK or AKT pathway. More evidence indicated that EV-released circRNA NRIP1 promoted proliferation, migration, and metastasis through AKT1/mTOR signaling pathway in gastric cancer. The involvement of this pathway has also been confirmed in breast cancer cells in patients (Wang et al., 2019b; Zhang et al., 2019). The circPTGR1 carried in EVs was found to contribute to the metastasis of hepatocellular carcinoma (Wang et al., 2019c). Interestingly, knock out of circPTGR1 in the source cells, their EVs inhibited invasion and migration of cancer cells. The increased expression of EV-released circ-IARS is related to the EMT of pancreatic cancer (Li et al., 2018). Therefore, EVs can act as messenger vehicles for cell-to-cell communication, releasing ncRNAs that contribute to the EMT in cancers.

The Function of EVs in Apoptosis and Autophagy

Cell apoptosis and autophagy are programmed cell death, both of them are abnormal in cancers. Previous reports have indicated that EVs containing anti-tumor drugs can induce cell apoptosis in HCCs (Slomka et al., 2020). Furthermore, EVs containing miRNA mimics such as let-7a have been found to induce cell apoptosis in breast cancer (Ahmed et al., 2021). In addition, EVs have the ability to regulate autophagy. There is evidence that EVs can enhance autophagy in glioblastoma (GBM) (Pavlyukov et al., 2018). These findings suggest that EVs play a role in cell apoptosis and autophagy.

EVs Stimulate Oxidative Stress

Studies have shown that low levels of reactive oxygen species (ROS) were observed in the stem cells of liver cancer and breast cancer (Shi et al., 2012). The EVs of SV-HUC-1 cells were found to mediate the P38/NF-kB signaling pathway, enhancing the levels of OS (Xi et al., 2020). This suggests that EVs were involved in OS, that may contribute to the development of cancers (Figure 4).

FIGURE 4
www.frontiersin.org

FIGURE 4. The function of EVs.

EVs Regulate the Expression of lncRNA

LncRNA usually acts as a regulator of nuclear transcription factors (Wu et al., 2021). An increasing amount of data has shown that long non-coding RNAs (lncRNAs) are associated with the development of cancers (Huang et al., 2021a). EVs containing lncRNA-APC1 inhibited tumor growth in colorectal cancer (CRC). lncRNA-APC1 is an important mediator of APC development through the APC1/RAB5B axis (Wang et al., 2021). The increased expression of lncRNA H19, which is normally regulated by DNA methylation, was observed in numerous cancers (Yang et al., 2021). Previous studies have suggested that EV-contained H19 promotes cell migration and invasion in CRC (Ren et al., 2018). The abnormal expression of XIST, a key factor in the X chromosome inactive (XCI) process, was observed in gastric cancer (Chen et al., 2016; Huang et al., 2021a; Huang et al., 2021b). EV-contained XIST was found to stimulate cell growth in breast cancer (Xing et al., 2018).

To investigate the role of EVs that contained lncRNAs in cancers, appropriate EVs were collected. The EVs were mostly obtained from the cells that were enriched in expressed lncRNA, such as the A549 cell line which exhibited increased H19 expression (Hao et al., 2017). In addition, the EVs were cultured in an environment that encouraged the increased expression of lncRNAs (Born et al., 2020).

EVs Regulate the Expression of miRNA

In contrast to lncRNAs, miRNAs are 20–22 nucleotides long. Both miRNAs and lncRNAs are single-stranded, endogenous RNAs, and play roles in the development of cancers. Some miRNAs, such as let-7a and the miR29 family, are involved in EMT, metastasis, migration, invasion, cell cycle, proliferation, and apoptosis of numerous cancers (Rostas et al., 2014; Song et al., 2020). A few miRNAs have been confirmed to be post-transcriptional regulators for target mRNAs. They can be used as the potential biomarkers for classification, prognosis, chemotherapy, and radiotherapy resistance in triple-negative breast cancer (TNBC) (Ding et al., 2019). Results show that miRNA of EVs have a curing effect on breast cancer (Ohno et al., 2013). MiRNAs can be coated by EVs and delivered to target cells, affect the H19/MAPK/ERK pathways (Ding et al., 2018; Wu et al., 2021).

A database indicated that EVs are enriched in miRNAs, lncRNAs, and proteins (Berardocco et al., 2017). In contrast to transfected mimics or miRNAs inhibitors, EVs that obtained from source cells can specifically and accurately deliver these miRNAs endogenously (Table 2). Considering the characteristics of EVs, therapies using EVs could be a potential approach for cancer treatment.

TABLE 2
www.frontiersin.org

TABLE 2. The miRNA of EVs in cancers.

EVs Regulate Gene Expression by siRNA

SiRNAs are produced by short, exogenous double-stranded RNAs (dsRNAs) as an RNA interference (RNAi) tool (Kim et al., 2018; Dharamdasani et al., 2020; Feng et al., 2020). SiRNA can be used to effectively silence target genes. A recent study showed that the use of siRNA, such as siRNA-027 can inhibit cell growth and induce apoptosis in numerous cancers (Chen et al., 2020). Hence, siRNA can be used to potentially analyze the development of cancers. A barrier to the RNAi-based therapy of cancers is the low specificity of siRNA delivery. EVs are nano-scale vesicles that can be used to deliver siRNAs as cargos to the target cells by cell-to-cell communication. Previous reports have suggested that the EVs of human plasma cells can deliver siRNA to monocytes and lymphocytes that can silence the expression of mitogen-activated protein kinase 1 (Wahlgren et al., 2012). This suggests that EVs can be used as gene delivery vehicles (GDV) to transport exogenous siRNA in cancer research. Consequently, EVs combined with siRNA are more effective and demonstrate higher specificities than traditionally siRNA delivery in cancer treatment.

EVs Regulate the Expression of Protein

The mitochondrial proteins contained in EVs can promote tumorigenesis by cell-to-cell communication (Al-Nedawi et al., 2008; Demory Beckler et al., 2013). The expression of MET (also known as hepatocyte growth factor receptors) associated with circulating EVs and phosphorylated MET (Tyr1349) was increased in patients with stage 3 and stage 4 melanoma compare to control (Peinado et al., 2012). This finding indicates that EVs can be used to detect the development of cancer (Costa-Silva et al., 2015). This assumption was confirmed when the expression of MIF and GPC-1 proteins in EVs was detected in cancer patients, allowing them to analyze the prognosis of cancer (Melo et al., 2015). Furthermore, phospholipid-binding proteins-carrying EVs can inhibit cell growth and induced apoptosis in numerous cancers (Dhondt et al., 2020). Thus, the proteins contained in EVs were useful for the detection and prognosis of cancers.

The Function of EVs in the Tumor Micro-environment

EVs are a key component of the tumor microenvironment. Tumor heterogeneity includes genomic heterogeneity in both tumor cells and non-cancerous microenvironments. Moreover, the tumor nanoenvironment (TNE) is a special nano-scale tumor microenvironment that possesses complex structures and unique components (Eguchi et al., 2018). The TNE includes EVs and apoptotic bodies. EVs released by tumor cells were absorbed by other cells in the tumor microenvironment, influencing the development of cancer through tumor heterogeneity (Tredan et al., 2007). EVs thus contribute to the formation of the tumor microenvironment in the form of cell-to-cell communication.

Discussion

Considering that EVs can carry any cargos, including nucleic acids and proteins, EVs can thus be used as clinical diagnostic biomarkers. For example, the detection of tumor-specific RNAs in EVs can be used as biomarkers for cancer diagnosis (Gurunathan et al., 2019). Furthermore, proteins contained within EVs such as TSG101, RAS-related protein RAB-11B (RAB11B), CD63, and CD81 can be used as biomarkers for diagnosis of HCCs and other cancers (Möbius et al., 2003; Valadi et al., 2007). In contrast to traditional diagnostic methods such as peripheral blood or histopathology, the accuracy and specificity of EVs were more closely associated with the development of cancers.

EVs can be combined with engineered materials to specifically affect cancer cells. Gold nanoparticles (AuNPs) can mediate photothermal therapy (PPT) to inhibit cell growth and induce cell death (Hu et al., 2020). However, most AuNPs have low specificity. EVs combined with AuNPs can increase their specificity and accelerate the release of their cargos, enhancing the anti-tumor effect of PTT (Nasseri et al., 2020). This could be an important form of therapy for the treatment of cancers in the future. Due to the endogenous nature of EVs, their cargos can escape the immune system and accurately and effectively target tumor cells. In addition, as nano-vesicles, EVs can bypass the blood-brain barrier (Yin et al., 2012). The EVs of immature dendritic cells have been engineered to contain proteins that can target tumors originated from the neuroendothelial and nerve cells in the brain (Federici et al., 2014). Therefore, EVs as nano-vesicles can be used to cross the blood-brain barrier in cancer treatment.

EVs containing anti-cancer drugs, such as therapeutic agents, can be used in the treatment of cancers. In contrast to liposomes, EVs injected in vivo can be absorbed without the interference of the immune system (Ferguson and Nguyen, 2016; Kalluri, 2016; Barile and Vassalli, 2017; Fitts et al., 2019; Liao et al., 2019). Furthermore, EVs are safe and are tolerable in vivo. Recent studies have demonstrated that repeatedly injected mesenchymal cells (MHC) or the IPCs of EVs do not induce toxicity (Zhu et al., 2017; Mendt et al., 2018).

The EVs that carry chemotherapeutics can decide the cell fate by cell-to-cell communication. For example, αv integrin-specific EVs have been shown to have a therapeutic effect on breast cancer (Tian et al., 2014). Another report suggested that paclitaxel surrounding the EVs of macrophages inhibited lung cancer growth in mice (Kim et al., 2016). These reports indicated that chemotherapeutic agent encapsulating EVs have an anti-tumor effect. Recently, studies have shown that the bioavailability of EVs-engineered doxorubicin was improved compared to the free doxorubicin (Tian et al., 2014; Kojima et al., 2018). These studies suggested that as a vesicle, EVs can enhance the efficacy of drugs. Despite the advancements in the understanding of EVs, there are still some challenges that need to be solved (Figure 5).

FIGURE 5
www.frontiersin.org

FIGURE 5. The challenge of EVs.

Conclusion

EVs are derived from multivesicular bodies formed by intracellular lysosomal particles that are released into the extracellular matrix. The source cells determine the specificity of their EVs. EVs contained RNAs, proteins, and drugs that can play important roles in the development of cancers. EVs have the ability to decide the fate of cells by cell-to-cell communication. EVs have potential applications in anti-cancer treatments in the future.

Author Contributions

All authors listed have made a substantial, direct and intellectual contribution to the work, and approved it for publication.

Funding

This work was supported by the Jilin Health Commission Program under Grant 2020J05S, the Fundamental Research Funds for the Central Universities under Grant 2019JCKT-70, the Jilin Education Department Program under Grant JJKH20200950KJ, and the Jilin Scientific and Technological Development Program under Grant 20190103071JH, 202002006JC, 20210101010JC, and 2020041.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Ahmed, S. H., Espinoza-Sánchez, N. A., El-Damen, A., Fahim, S. A., Badawy, M. A., Greve, B., et al. (2021). Small Extracellular Vesicle-Encapsulated miR-181b-5p, miR-222-3p and Let-7a-5p: Next Generation Plasma Biopsy-Based Diagnostic Biomarkers for Inflammatory Breast Cancer. PLoS One 16, e0250642. doi:10.1371/journal.pone.0250642

PubMed Abstract | CrossRef Full Text | Google Scholar

Al-Nedawi, K., Meehan, B., Micallef, J., Lhotak, V., May, L., Guha, A., et al. (2008). Intercellular Transfer of the Oncogenic Receptor EGFRvIII by Microvesicles Derived from Tumour Cells. Nat. Cel Biol 10, 619–624. doi:10.1038/ncb1725

PubMed Abstract | CrossRef Full Text | Google Scholar

Bainton, D. F., and Farquhar, M. G. (1968). Differences in Enzyme Content of Azurophil and Specific Granules of Polymorphonuclear Leukocytes. J. Cel Biol 39, 299–317. doi:10.1083/jcb.39.2.299

CrossRef Full Text | Google Scholar

Balaj, L., Lessard, R., Dai, L., Cho, Y.-J., Pomeroy, S. L., Breakefield, X. O., et al. (2011). Tumour Microvesicles Contain Retrotransposon Elements and Amplified Oncogene Sequences. Nat. Commun. 2, 180. doi:10.1038/ncomms1180

PubMed Abstract | CrossRef Full Text | Google Scholar

Barile, L., and Vassalli, G. (2017). Exosomes: Therapy Delivery Tools and Biomarkers of Diseases. Pharmacol. Ther. 174, 63–78. doi:10.1016/j.pharmthera.2017.02.020

PubMed Abstract | CrossRef Full Text | Google Scholar

Berardocco, M., Radeghieri, A., Busatto, S., Gallorini, M., Raggi, C., Gissi, C., et al. (2017). RNA-seq Reveals Distinctive RNA Profiles of Small Extracellular Vesicles from Different Human Liver Cancer Cell Lines. Oncotarget 8, 82920–82939. doi:10.18632/oncotarget.20503

PubMed Abstract | CrossRef Full Text | Google Scholar

Born, L. J., Harmon, J. W., and Jay, S. M. (2020). Therapeutic Potential of Extracellular Vesicle-Associated Long Noncoding RNA. Bioeng. Transl Med. 5, e10172. doi:10.1002/btm2.10172

PubMed Abstract | CrossRef Full Text | Google Scholar

Chan, J. J., and Tay, Y. (2018). Noncoding RNA:RNA Regulatory Networks in Cancer. Int. J. Mol. Sci. 19. doi:10.3390/ijms19051310

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, D.-l., Ju, H.-q., Lu, Y.-x., Chen, L.-z., Zeng, Z.-l., Zhang, D.-s., et al. (2016). Long Non-coding RNA XIST Regulates Gastric Cancer Progression by Acting as a Molecular Sponge of miR-101 to Modulate EZH2 Expression. J. Exp. Clin. Cancer Res. 35, 142. doi:10.1186/s13046-016-0420-1

CrossRef Full Text | Google Scholar

Chen, Z., Krishnamachary, B., Pachecho-Torres, J., Penet, M. F., and Bhujwalla, Z. M. (2020). Theranostic Small Interfering RNA Nanoparticles in Cancer Precision Nanomedicine. Wiley Interdiscip. Rev. Nanomed Nanobiotechnol 12, e1595. doi:10.1002/wnan.1595

PubMed Abstract | CrossRef Full Text | Google Scholar

Costa-Silva, B., Aiello, N. M., Ocean, A. J., Singh, S., Zhang, H., Thakur, B. K., et al. (2015). Pancreatic Cancer Exosomes Initiate Pre-metastatic Niche Formation in the Liver. Nat. Cel Biol 17, 816–826. doi:10.1038/ncb3169

PubMed Abstract | CrossRef Full Text | Google Scholar

Demory Beckler, M., Higginbotham, J. N., Franklin, J. L., Ham, A.-J., Halvey, P. J., Imasuen, I. E., et al. (2013). Proteomic Analysis of Exosomes from Mutant KRAS colon Cancer Cells Identifies Intercellular Transfer of Mutant KRAS. Mol. Cell Proteomics 12, 343–355. doi:10.1074/mcp.m112.022806

PubMed Abstract | CrossRef Full Text | Google Scholar

Dharamdasani, V., Mandal, A., Qi, Q. M., Suzuki, I., Bentley, M. V. L. B., and Mitragotri, S. (2020). Topical Delivery of siRNA into Skin Using Ionic Liquids. J. Controlled Release 323, 475–482. doi:10.1016/j.jconrel.2020.04.038

CrossRef Full Text | Google Scholar

Dhondt, B., Geeurickx, E., Tulkens, J., Van Deun, J., Vergauwen, G., Lippens, L., et al. (2020). Unravelling the Proteomic Landscape of Extracellular Vesicles in Prostate Cancer by Density‐based Fractionation of Urine. J. Extracellular Vesicles 9, 1736935. doi:10.1080/20013078.2020.1736935

CrossRef Full Text | Google Scholar

Ding, K., Liao, Y., Gong, D., Zhao, X., and Ji, W. (2018). Effect of Long Non-coding RNA H19 on Oxidative Stress and Chemotherapy Resistance of CD133+ Cancer Stem Cells via the MAPK/ERK Signaling Pathway in Hepatocellular Carcinoma. Biochem. Biophysical Res. Commun. 502, 194–201. doi:10.1016/j.bbrc.2018.05.143

CrossRef Full Text | Google Scholar

Ding, L., Gu, H., Xiong, X., Ao, H., Cao, J., Lin, W., et al. (2019). MicroRNAs Involved in Carcinogenesis, Prognosis, Therapeutic Resistance and Applications in Human Triple-Negative Breast Cancer. Cells 8, 492. doi:10.3390/cells8121492

PubMed Abstract | CrossRef Full Text | Google Scholar

Draganov, D. D., Santidrian, A. F., Minev, I., Nguyen, D., Kilinc, M. O., Petrov, I., et al. (2019). Delivery of Oncolytic Vaccinia Virus by Matched Allogeneic Stem Cells Overcomes Critical Innate and Adaptive Immune Barriers. J. Transl Med. 17, 100. doi:10.1186/s12967-019-1829-z

CrossRef Full Text | Google Scholar

Dzobo, K., Senthebane, D. A., Ganz, C., Thomford, N. E., Wonkam, A., and Dandara, C. (2020). Advances in Therapeutic Targeting of Cancer Stem Cells within the Tumor Microenvironment: An Updated Review. Cells 9, 896. doi:10.3390/cells9081896

PubMed Abstract | CrossRef Full Text | Google Scholar

Eguchi, T., Sogawa, C., Okusha, Y., Uchibe, K., Iinuma, R., Ono, K., et al. (2018). Organoids with Cancer Stem Cell-like Properties Secrete Exosomes and HSP90 in a 3D Nanoenvironment. PLoS One 13, e0191109. doi:10.1371/journal.pone.0191109

PubMed Abstract | CrossRef Full Text | Google Scholar

Federici, C., Petrucci, F., Caimi, S., Cesolini, A., Logozzi, M., Borghi, M., et al. (2014). Exosome Release and Low pH Belong to a Framework of Resistance of Human Melanoma Cells to Cisplatin. PLoS One 9, e88193. doi:10.1371/journal.pone.0088193

PubMed Abstract | CrossRef Full Text | Google Scholar

Feng, J., Yu, W., Xu, Z., Hu, J., Liu, J., and Wang, F. (2020). Multifunctional siRNA-Laden Hybrid Nanoplatform for Noninvasive PA/IR Dual-Modal Imaging-Guided Enhanced Photogenetherapy. ACS Appl. Mater. Inter. 12, 22613–22623. doi:10.1021/acsami.0c04533

PubMed Abstract | CrossRef Full Text | Google Scholar

Ferguson, S. W., and Nguyen, J. (2016). Exosomes as Therapeutics: The Implications of Molecular Composition and Exosomal Heterogeneity. J. Controlled Release 228, 179–190. doi:10.1016/j.jconrel.2016.02.037

CrossRef Full Text | Google Scholar

Fernández, P. L., Hernández, L., Farré, X., Campo, E., and Cardesa, A. (2002). Alterations of Cell Cycle-Regulatory Genes in Prostate Cancer. Pathobiology 70, 1–10. doi:10.1159/000065998

PubMed Abstract | CrossRef Full Text | Google Scholar

Fitts, C. A., Ji, N., Li, Y., and Tan, C. (2019). Exploiting Exosomes in Cancer Liquid Biopsies and Drug Delivery. Adv. Healthc. Mater. 8, e1801268. doi:10.1002/adhm.201801268

PubMed Abstract | CrossRef Full Text | Google Scholar

Gurunathan, S., Kang, M. H., Jeyaraj, M., Qasim, M., and Kim, J. H. (2019). Review of the Isolation, Characterization, Biological Function, and Multifarious Therapeutic Approaches of Exosomes. Cells 8, 307. doi:10.3390/cells8040307

PubMed Abstract | CrossRef Full Text | Google Scholar

Gusachenko, O. N., Zenkova, M. A., and Vlassov, V. V. (2013). Nucleic Acids in Exosomes: Disease Markers and Intercellular Communication Molecules. Biochem. Mosc. 78, 1–7. doi:10.1134/s000629791301001x

CrossRef Full Text | Google Scholar

Hao, Y., Wang, G., Lin, C., Li, D., Ji, Z., Gao, F., et al. (2017). Valproic Acid Induces Decreased Expression of H19 Promoting Cell Apoptosis in A549 Cells. DNA Cel Biol. 36, 428–435. doi:10.1089/dna.2016.3542

PubMed Abstract | CrossRef Full Text | Google Scholar

Harding, C., Heuser, J., and Stahl, P. (1983). Receptor-mediated Endocytosis of Transferrin and Recycling of the Transferrin Receptor in Rat Reticulocytes. J. Cel Biol 97, 329–339. doi:10.1083/jcb.97.2.329

CrossRef Full Text | Google Scholar

Hu, X., Zhang, Y., Ding, T., Liu, J., and Zhao, H. (2020). Multifunctional Gold Nanoparticles: A Novel Nanomaterial for Various Medical Applications and Biological Activities. Front. Bioeng. Biotechnol. 8, 990. doi:10.3389/fbioe.2020.00990

PubMed Abstract | CrossRef Full Text | Google Scholar

Huang, W., Yan, Y., Liu, Y., Lin, M., Ma, J., Zhang, W., et al. (2020). Exosomes with Low miR-34c-3p Expression Promote Invasion and Migration of Non-small Cell Lung Cancer by Upregulating Integrin α2β1. Signal. Transduct Target. Ther. 5, 39. doi:10.1038/s41392-020-0133-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Huang, Y., Yuan, K., Tang, M., Yue, J., Bao, L., Wu, S., et al. (2021). Melatonin Inhibiting the Survival of Human Gastric Cancer Cells under ER Stress Involving Autophagy and Ras‐Raf‐MAPK Signalling. J. Cel Mol Med 25, 1480–1492. doi:10.1111/jcmm.16237

CrossRef Full Text | Google Scholar

Huang, Y., Zhou, Z., Zhang, J., Hao, Z., He, Y., Wu, Z., et al. (2021). lncRNA MALAT1 Participates in Metformin Inhibiting the Proliferation of Breast Cancer Cell. J. Cel Mol Med 25, 7135–7145. doi:10.1111/jcmm.16742

CrossRef Full Text | Google Scholar

Kahlert, C., and Kalluri, R. (2013). Exosomes in Tumor Microenvironment Influence Cancer Progression and Metastasis. J. Mol. Med. 91, 431–437. doi:10.1007/s00109-013-1020-6

CrossRef Full Text | Google Scholar

Kahlert, C., Melo, S. A., Protopopov, A., Tang, J., Seth, S., Koch, M., et al. (2014). Identification of Double-Stranded Genomic DNA Spanning All Chromosomes with Mutated KRAS and P53 DNA in the Serum Exosomes of Patients with Pancreatic Cancer. J. Biol. Chem. 289, 3869–3875. doi:10.1074/jbc.c113.532267

CrossRef Full Text | Google Scholar

Kalluri, R. (2016). The Biology and Function of Exosomes in Cancer. J. Clin. Invest. 126, 1208–1215. doi:10.1172/jci81135

CrossRef Full Text | Google Scholar

Kim, H. J., Yi, Y., Kim, A., and Miyata, K. (2018). Small Delivery Vehicles of siRNA for Enhanced Cancer Targeting. Biomacromolecules 19, 2377–2390. doi:10.1021/acs.biomac.8b00546

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, M. S., Haney, M. J., Zhao, Y., Mahajan, V., Deygen, I., Klyachko, N. L., et al. (2016). Development of Exosome-Encapsulated Paclitaxel to Overcome MDR in Cancer Cells. Nanomedicine: Nanotechnology, Biol. Med. 12, 655–664. doi:10.1016/j.nano.2015.10.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Kojima, R., Bojar, D., Rizzi, G., Hamri, G. C.-E., El-Baba, M. D., Saxena, P., et al. (2018). Designer Exosomes Produced by Implanted Cells Intracerebrally Deliver Therapeutic Cargo for Parkinson's Disease Treatment. Nat. Commun. 9, 1305. doi:10.1038/s41467-018-03733-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Kowal, J., Arras, G., Colombo, M., Jouve, M., Morath, J. P., Primdal-Bengtson, B., et al. (2016). Proteomic Comparison Defines Novel Markers to Characterize Heterogeneous Populations of Extracellular Vesicle Subtypes. Proc. Natl. Acad. Sci. USA 113, E968–E977. doi:10.1073/pnas.1521230113

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, J., Li, Z., Jiang, P., Peng, M., Zhang, X., Chen, K., et al. (2018). 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, 177. doi:10.1186/s13046-018-0822-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Liao, W., Du, Y., Zhang, C., Pan, F., Yao, Y., Zhang, T., et al. (2019). Exosomes: The Next Generation of Endogenous Nanomaterials for Advanced Drug Delivery and Therapy. Acta Biomater. 86, 1–14. doi:10.1016/j.actbio.2018.12.045

PubMed Abstract | CrossRef Full Text | Google Scholar

Lu, L., Luo, F., Liu, Y., Liu, X., Shi, L., Lu, X., et al. (2015). Posttranscriptional Silencing of the lncRNA MALAT1 by miR-217 Inhibits the Epithelial-Mesenchymal Transition via Enhancer of Zeste Homolog 2 in the Malignant Transformation of HBE Cells Induced by Cigarette Smoke Extract. Toxicol. Appl. Pharmacol. 289, 276–285. doi:10.1016/j.taap.2015.09.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Luna, J., Boni, J., Cuatrecasas, M., Bofill-De Ros, X., Núñez-Manchón, E., Gironella, M., et al. (2019). DYRK1A Modulates C-MET in Pancreatic Ductal Adenocarcinoma to Drive Tumour Growth. Gut 68, 1465–1476. doi:10.1136/gutjnl-2018-316128

PubMed Abstract | CrossRef Full Text | Google Scholar

Melo, S. A., Luecke, L. B., Kahlert, C., Fernandez, A. F., Gammon, S. T., Kaye, J., et al. (2015). Glypican-1 Identifies Cancer Exosomes and Detects Early Pancreatic Cancer. Nature 523, 177–182. doi:10.1038/nature14581

PubMed Abstract | CrossRef Full Text | Google Scholar

Melo, S. A., Sugimoto, H., O’Connell, J. T., Kato, N., Villanueva, A., Vidal, A., et al. (2014). Cancer Exosomes Perform Cell-independent microRNA Biogenesis and Promote Tumorigenesis. Cancer Cell 26, 707–721. doi:10.1016/j.ccell.2014.09.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Mendt, M., Kamerkar, S., Sugimoto, H., McAndrews, K. M., Wu, C. C., Gagea, M., et al. (2018). Generation and Testing of Clinical-Grade Exosomes for Pancreatic Cancer. JCI Insight 3, e99263. doi:10.1172/jci.insight.99263

PubMed Abstract | CrossRef Full Text | Google Scholar

Möbius, W., van Donselaar, E., Ohno-Iwashita, Y., Shimada, Y., Heijnen, H. F. G., Slot, J. W., et al. (2003). Recycling Compartments and the Internal Vesicles of Multivesicular Bodies Harbor Most of the Cholesterol Found in the Endocytic Pathway. Traffic 4, 222–231. doi:10.1034/j.1600-0854.2003.00072.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Nasseri, B., Turk, M., Kosemehmetoglu, K., Kaya, M., Pişkin, E., Rabiee, N., et al. (2020). The Pimpled Gold Nanosphere: A Superior Candidate for Plasmonic Photothermal Therapy. Ijn Vol. 15, 2903–2920. doi:10.2147/ijn.s248327

PubMed Abstract | CrossRef Full Text | Google Scholar

Ohno, S.-i., Takanashi, M., Sudo, K., Ueda, S., Ishikawa, A., Matsuyama, N., et al. (2013). Systemically Injected Exosomes Targeted to EGFR Deliver Antitumor MicroRNA to Breast Cancer Cells. Mol. Ther. 21, 185–191. doi:10.1038/mt.2012.180

PubMed Abstract | CrossRef Full Text | Google Scholar

Pan, B. T., Teng, K., Wu, C., Adam, M., and Johnstone, R. M. (1985). Electron Microscopic Evidence for Externalization of the Transferrin Receptor in Vesicular Form in Sheep Reticulocytes. J. Cel Biol 101, 942–948. doi:10.1083/jcb.101.3.942

CrossRef Full Text | Google Scholar

Pavlyukov, M. S., Yu, H., Bastola, S., Minata, M., Shender, V. O., Lee, Y., et al. (2018). Apoptotic Cell-Derived Extracellular Vesicles Promote Malignancy of Glioblastoma via Intercellular Transfer of Splicing Factors. Cancer Cell 34, 119–135. doi:10.1016/j.ccell.2018.05.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Peinado, H., Alečković, M., Lavotshkin, S., Matei, I., Costa-Silva, B., Moreno-Bueno, G., et al. (2012). Melanoma Exosomes Educate Bone Marrow Progenitor Cells toward a Pro-metastatic Phenotype through MET. Nat. Med. 18, 883–891. doi:10.1038/nm.2753

PubMed Abstract | CrossRef Full Text | Google Scholar

Perin, E. C., Silva, G. V., Henry, T. D., Cabreira-Hansen, M. G., Moore, W. H., Coulter, S. A., et al. (2011). A Randomized Study of Transendocardial Injection of Autologous Bone Marrow Mononuclear Cells and Cell Function Analysis in Ischemic Heart Failure (FOCUS-HF). Am. Heart J. 161, 1078–1087. doi:10.1016/j.ahj.2011.01.028

PubMed Abstract | CrossRef Full Text | Google Scholar

Ramírez-Ricardo, J., Leal-Orta, E., Martínez-Baeza, E., Ortiz-Mendoza, C., Breton-Mora, F., Herrera-Torres, A., et al. (2020). Circulating Extracellular Vesicles from Patients with Breast Cancer Enhance Migration and Invasion via a Src-dependent P-athway in MDA-MB-231 B-reast C-ancer C-ells. Mol. Med. Rep. 22, 1932–1948. doi:10.3892/mmr.2020.11259

PubMed Abstract | CrossRef Full Text | Google Scholar

Raposo, G., and Stoorvogel, W. (2013). Extracellular Vesicles: Exosomes, Microvesicles, and Friends. J. Cel Biol 200, 373–383. doi:10.1083/jcb.201211138

CrossRef Full Text | Google Scholar

Ren, J., Ding, L., Zhang, D., Shi, G., Xu, Q., Shen, S., et al. (2018). Carcinoma-associated Fibroblasts Promote the Stemness and Chemoresistance of Colorectal Cancer by Transferring Exosomal lncRNA H19. Theranostics 8, 3932–3948. doi:10.7150/thno.25541

PubMed Abstract | CrossRef Full Text | Google Scholar

Rostas, J. W., Pruitt, H. C., Metge, B. J., Mitra, A., Bailey, S. K., Bae, S., et al. (2014). microRNA-29 Negatively Regulates EMT Regulator N-Myc Interactor in Breast Cancer. Mol. Cancer 13, 200. doi:10.1186/1476-4598-13-200

PubMed Abstract | CrossRef Full Text | Google Scholar

Shi, X., Zhang, Y., Zheng, J., and Pan, J. (2012). Reactive Oxygen Species in Cancer Stem Cells. Antioxid. Redox Signaling 16, 1215–1228. doi:10.1089/ars.2012.4529

CrossRef Full Text | Google Scholar

Silva, A. M., Lazaro-Ibanez, E., Gunnarsson, A., Dhande, A., Daaboul, G., Peacock, B., et al. (2021). Quantification of Protein Cargo Loading into Engineered Extracellular Vesicles at Single-Vesicle and Single-Molecule Resolution. J. Extracellular Vesicles 10, e12130. doi:10.1002/jev2.12130

CrossRef Full Text | Google Scholar

Slomka, A., Mocan, T., Wang, B., Nenu, I., Urban, S. K., Gonzales-Carmona, M., et al. (2020). EVs as Potential New Therapeutic Tool/Target in Gastrointestinal Cancer and HCC. Cancers (Basel) 12, 3019. doi:10.3390/cancers12103019

CrossRef Full Text | Google Scholar

Song, X., Liang, Y., Sang, Y., Li, Y., Zhang, H., Chen, B., et al. (2020). circHMCU Promotes Proliferation and Metastasis of Breast Cancer by Sponging the Let-7 Family. Mol. Ther. - Nucleic Acids 20, 518–533. doi:10.1016/j.omtn.2020.03.014

PubMed Abstract | CrossRef Full Text | Google Scholar

Su, C. Y., Zhang, J. Y., Yarden, Y., and Fu, L. W. (2021). The Key Roles of Cancer Stem Cell-Derived Extracellular Vesicles. Signal. Transduct Tar 6, 109. doi:10.1038/s41392-021-00499-2

CrossRef Full Text | Google Scholar

Taylor, D. D., and Gercel-Taylor, C. (2008). MicroRNA Signatures of Tumor-Derived Exosomes as Diagnostic Biomarkers of Ovarian Cancer. Gynecol. Oncol. 110, 13–21. doi:10.1016/j.ygyno.2008.04.033

PubMed Abstract | CrossRef Full Text | Google Scholar

Théry, C., Zitvogel, L., and Amigorena, S. (2002). Exosomes: Composition, Biogenesis and Function. Nat. Rev. Immunol. 2, 569–579. doi:10.1038/nri855

PubMed Abstract | CrossRef Full Text | Google Scholar

Tian, L., Cao, J., Jiao, H., Zhang, J., Ren, X., Liu, X., et al. (2019). CircRASSF2 Promotes Laryngeal Squamous Cell Carcinoma Progression by Regulating the miR-302b-3p/IGF-1R axis. Clin. Sci. (Lond) 133, 1053–1066. doi:10.1042/cs20190110

PubMed Abstract | CrossRef Full Text | Google Scholar

Tian, Y., Li, S., Song, J., Ji, T., Zhu, M., Anderson, G. J., et al. (2014). A Doxorubicin Delivery Platform Using Engineered Natural Membrane Vesicle Exosomes for Targeted Tumor Therapy. Biomaterials 35, 2383–2390. doi:10.1016/j.biomaterials.2013.11.083

PubMed Abstract | CrossRef Full Text | Google Scholar

Tredan, O., Galmarini, C. M., Patel, K., and Tannock, I. F. (2007). Drug Resistance and the Solid Tumor Microenvironment. JNCI J. Natl. Cancer Inst. 99, 1441–1454. doi:10.1093/jnci/djm135

CrossRef Full Text | Google Scholar

Valadi, H., Ekström, K., Bossios, A., Sjöstrand, M., Lee, J. J., and Lötvall, J. O. (2007). Exosome-mediated Transfer of mRNAs and microRNAs Is a Novel Mechanism of Genetic Exchange between Cells. Nat. Cel Biol 9, 654–659. doi:10.1038/ncb1596

CrossRef Full Text | Google Scholar

Wahlgren, J., Karlson, T. D. L., Brisslert, M., Vaziri Sani, F., Telemo, E., Sunnerhagen, P., et al. (2012). Plasma Exosomes Can Deliver Exogenous Short Interfering RNA to Monocytes and Lymphocytes. Nucleic Acids Res. 40, e130. doi:10.1093/nar/gks463

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, F. W., Cao, C. H., Han, K., Zhao, Y. X., Cai, M. Y., Xiang, Z. C., et al. (2021). APC-activated Long Noncoding RNA Inhibits Colorectal Carcinoma Pathogenesis through Reduction of Exosome Production. J. Clin. Invest. 131, e149666. doi:10.1172/jci149666

CrossRef Full Text | Google Scholar

Wang, G., Liu, W., Zou, Y., Wang, G., Deng, Y., Luo, J., et al. (2019). Three Isoforms of Exosomal circPTGR1 Promote Hepatocellular Carcinoma Metastasis via the miR449a-MET Pathway. EBioMedicine 40, 432–445. doi:10.1016/j.ebiom.2018.12.062

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, J., Zhang, Q., Zhou, S., Xu, H., Wang, D., Feng, J., et al. (2019). Circular RNA Expression in Exosomes Derived from Breast Cancer Cells and Patients. Epigenomics 11, 411–421. doi:10.2217/epi-2018-0111

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, X., Qiao, D., Chen, L., Xu, M., Chen, S., Huang, L., et al. (2019). Chemotherapeutic Drugs Stimulate the Release and Recycling of Extracellular Vesicles to Assist Cancer Cells in Developing an Urgent Chemoresistance. Mol. Cancer 18, 182. doi:10.1186/s12943-019-1114-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, S., Li, T. Y., Liu, W. W., and Huang, Y. Y. (2021). Ferroptosis and Cancer: Complex Relationship and Potential Application of Exosomes. Front Cel Dev Biol 9, 733751. doi:10.3389/fcell.2021.733751

CrossRef Full Text | Google Scholar

Xi, X. j., Zeng, J. j., Lu, Y., Chen, S. h., Jiang, Z. w., He, P. j., et al. (2020). Extracellular Vesicles Enhance Oxidative Stress through P38/NF‐kB Pathway in Ketamine‐induced Ulcerative Cystitis. J. Cel Mol Med 24, 7609–7624. doi:10.1111/jcmm.15397

PubMed Abstract | CrossRef Full Text | Google Scholar

Xing, F., Liu, Y., Wu, S.-Y., Wu, K., Sharma, S., Mo, Y.-Y., et al. (2018). Loss of XIST in Breast Cancer Activates MSN-C-Met and Reprograms Microglia via Exosomal miRNA to Promote Brain Metastasis. Cancer Res. 78, 4316–4330. doi:10.1158/0008-5472.can-18-1102

PubMed Abstract | CrossRef Full Text | Google Scholar

Xu, Y., Xia, F., Ma, L., Shan, J., Shen, J., Yang, Z., et al. (2011). MicroRNA-122 Sensitizes HCC Cancer Cells to Adriamycin and Vincristine through Modulating Expression of MDR and Inducing Cell Cycle Arrest. Cancer Lett. 310, 160–169. doi:10.1016/j.canlet.2011.06.027

PubMed Abstract | CrossRef Full Text | Google Scholar

Xue, J., Liu, Y., Luo, F., Lu, X., Xu, H., Liu, X., et al. (2017). Circ100284, via miR-217 Regulation of EZH2, Is Involved in the Arsenite-Accelerated Cell Cycle of Human Keratinocytes in Carcinogenesis. Biochim. Biophys. Acta (Bba) - Mol. Basis Dis. 1863, 753–763. doi:10.1016/j.bbadis.2016.12.018

CrossRef Full Text | Google Scholar

Yang, J., Qi, M., Fei, X., Wang, X., and Wang, K. (2021). LncRNA H19: A Novel Oncogene in Multiple Cancers. Int. J. Biol. Sci. 17, 3188–3208. doi:10.7150/ijbs.62573

CrossRef Full Text | Google Scholar

Yin, J., Yan, X., Yao, X., Zhang, Y., Shan, Y., Mao, N., et al. (2012). Secretion of Annexin A3 from Ovarian Cancer Cells and its Association with Platinum Resistance in Ovarian Cancer Patients. J. Cel Mol Med 16, 337–348. doi:10.1111/j.1582-4934.2011.01316.x

CrossRef Full Text | Google Scholar

Zhang, X., Wang, S., Wang, H., Cao, J., Huang, X., Chen, Z., et al. (2019). Circular RNA circNRIP1 Acts as a microRNA-149-5p Sponge to Promote Gastric Cancer Progression via the AKT1/mTOR Pathway. Mol. Cancer 18, 20. doi:10.1186/s12943-018-0935-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhu, X., Badawi, M., Pomeroy, S., Sutaria, D. S., Xie, Z., Baek, A., et al. (2017). Comprehensive Toxicity and Immunogenicity Studies Reveal Minimal Effects in Mice Following Sustained Dosing of Extracellular Vesicles Derived from HEK293T Cells. J. Extracellular Vesicles 6, 1324730. doi:10.1080/20013078.2017.1324730

CrossRef Full Text | Google Scholar

Keywords: EVS, Cancer, ncRNA, drug loading, target

Citation: Zhang X, Liu D, Gao Y, Lin C, An Q, Feng Y, Liu Y, Liu D, Luo H and Wang D (2021) The Biology and Function of Extracellular Vesicles in Cancer Development. Front. Cell Dev. Biol. 9:777441. doi: 10.3389/fcell.2021.777441

Received: 15 September 2021; Accepted: 22 October 2021;
Published: 05 November 2021.

Edited by:

Dong-Hua Yang, St. John’s University, United States

Reviewed by:

Wanhua Xie, Shenyang Medical College, China
Cecilia Battistelli, Sapienza University of Rome, Italy

Copyright © 2021 Zhang, Liu, Gao, Lin, An, Feng, Liu, Liu, Luo and Wang. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Haoming Luo, Luo.haoming@163.com; Dongxu Wang, wang_dong_xu@jlu.edu.cn

These authors have contributed equally to this work

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.