Back to Journals » Cancer Management and Research » Volume 10

Role of exosomal small RNA in prostate cancer metastasis

Authors Zhan F, Shen J, Wang R , Wang L , Dai Y, Zhang Y, Huang X

Received 10 April 2018

Accepted for publication 2 June 2018

Published 28 September 2018 Volume 2018:10 Pages 4029—4038

DOI https://doi.org/10.2147/CMAR.S170610

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Professor Harikrishna Nakshatri



Fei Zhan,1 Jingling Shen,2 Ruitao Wang,3 Liang Wang,4 Yao Dai,5 Yanqiao Zhang,1 Xiaoyi Huang6,7

1Department of Gastrointestinal Medical Oncology, Tumor Hospital of Harbin Medical University, Harbin 150081, China; 2Department of Histology and Embryology, Harbin Medical University, Harbin 150081, China; 3Department of Internal Medicine, Tumor Hospital of Harbin Medical University, Harbin 150081, China; 4Department of Pathology and MCW Cancer Center, Medical College of Wisconsin, Milwaukee, WI, 53226, USA; 5Department of Radiation Oncology, University of Florida, Gainesville, FL, 32610, USA; 6Biotherapy Center, Tumor Hospital of Harbin Medical University, Harbin 150081, China; 7Center of Translational Medicine, Harbin Medical University, Harbin 150086, China

Abstract: Prostate cancer (PCa) is the second most common cancer in men worldwide. When the disease becomes metastatic, limited treatment strategies exist, and metastatic disease prognoses are difficult to predict. Recently, evidence has emerged, which indicates that small RNAs are detectable in patient fluids, and exosomal small RNA ectopic expression is correlated with the development, progression, and metastasis of human PCa; however, the role of small RNAs in PCa is only partially understood. In this review, we discuss the research status regarding circulating exosomal small RNAs and applications using these small RNAs in PCa particularly looking at metastatic disease. Exosomal small RNAs could be used as potential biomarkers for the early diagnosis, micrometastasis detection, and prognosis of PCa.

Keywords: prostate cancer, metastases, ncRNA, exosome

Introduction

Prostate cancer (PCa) is the second most commonly diagnosed cancer and the fifth leading cause of cancer death in men worldwide.1 PCa is a complex disease, with multiple risk factors involving age, ethnicity, heredity, diets, and hormone levels.

PCa occurs as a localized disease in the early stages, at which time the treatment options include prostatectomy or radiotherapy. If the disease progresses as a recurrence or metastasis, androgen deprivation therapy (ADT) becomes the standard treatment. Unfortunately, patients with recurrent or metastatic PCa will inevitably develop castration-resistant PCa (CRPC), an end-stage metastatic disease, after a period of hormone responsiveness. The standard first-line CRPC treatment is docetaxel, but several newer systemic anticancer therapies that have been shown to improve overall survival are available.24 However, most cancer deaths are due to metastatic disease and are resistant to these therapies. Until now, the mechanisms that lead to metastases remain incompletely understood.

Cancer metastasis is a complex process. When tumor cells become motile and invasive during the epithelial–mesenchymal transition (EMT) period, motile cancer cells can enter the blood, where the cells become trapped in capillary beds distant from the primary tumor site. Circulating tumor cells enter secondary metastatic sites by extravasation into unfamiliar microenvironments. After converting to epithelial phenotypes, tumor cells will proliferate and form macrometastases at the second site. Behind this coherent process, the adaptation between tumor cells and the microenvironments plays a crucial role in metastasis regulation. Initially, tumor cells must overcome the extracellular matrix (ECM) barrier, which requires disruption of endothelial cell phenotypes for intravasation and extravasation. Although cancer cells survive in circulation, they cannot complete distant metastatic spread without adaptive modulation.5 The “metastatic niche model” presented below is based on the “Seed and Soil” hypothesis, which suggests that a suitable conducive microenvironment must evolve for tumor cells to engraft and proliferate at secondary sites.68 This model supports the notion that metastatic cells have preferred terminal organs, such as PCa cells that have a higher risk of bone metastasis.9,10 From the moment that metastasis occurs, cancer and stromal cells secrete molecules that adapt the environment for metastatic spread. Aside from conventional factors such as cytokines and chemokines, noncoding RNAs (ncRNAs) delivered by exosomes have been recently found to play an important role in communications between tumor cells and stromal cells in the tumor microenvironment (Figure 1).1113 In this review, we summarize the roles of exosomal ncRNAs in PCa metastases. For comprehensive information about ncRNA-mediated disease, a global network can be found at the mammalian ncRNA-disease repository (www.rna-society.org/mndr/). Besides, miR2Disease (http://www.mir2disease.org/) is a good resource for retrieving miRNA-associated diseases.

Figure 1 Schematic illustration of the cancer-derived exosome-mediated communication between cancer cell and surrounding or distal recipient.

Exosomes and exosomal ncRNAs

Exosomes are 40–100 nm vesicles that contain various molecules, including RNA, proteins, and cytokines, which are derived from several different cell types and are shed into extracellular spaces and body fluids.14 Different from microvesicles that are directly shed from cell membranes, exosomes are released when multivesicular bodies (MVBs) fuse with cell membranes. MVBs, containing intraluminal vesicles, are generated by the inward budding of clathrin-coated domains in the plasma membrane.15,16 Exosomes facilitate cell–cell communications and RNA transfer between cells and have been found to be useful in cell signaling studies and can impact biological processes in recipient cells.1720

ncRNAs represent about 98% of all transcriptional outputs in human bodies and are classified by size, small ncRNAs (<200 bp) and long ncRNAs (>200 bp).21 ncRNAs regulate gene expression using several different mechanisms such as RNA interference, cosuppression, transgene silencing, imprinting, methylation, and possibly position-effect variegation, and transvection, although no proteins are encoded.22 ncRNAs are divided into several categories: microRNAs (miRNAs), PIWI-interacting RNAs (piRNAs), small nucleolar RNAs, long ncRNAs (lncRNAs), and several other types. The most widely described ncRNAs are miRNAs and lncRNAs.23 miRNAs are single-stranded RNAs and are ~22 nucleotides in length, which play important roles in regulating gene expression. miRNAs are estimated to regulate translation in more than 60% of the protein-coding genes. As is commonly known, the way in which messenger RNAs (mRNAs) are silenced depends on the degree of complementarity between the miRNAs and the 3′-untranslated region (3′-URT) of the target mRNA. When perfect base-paring homology exists between the miRNA and the mRNA, the miRNA will cleave the mRNA with the help of Argonaute. Imperfect binding usually occurs that results in target mRNA regulation by suppressed protein translation.24

lncRNAs are RNA molecules more than 200 nucleotides in length. With the development of RNA-sequencing techniques, the discovery of novel RNA species including lncRNAs has rapidly expanded. Although many fewer studies have been performed using lncRNAs compared with that of miRNAs, the significance of lncRNA performance in cancer therapy has gained more attention recently. lncRNAs control transcription, translation, and protein function at multiple levels. Possible lncRNA mechanisms are 1) influencing transcription of protein-coding gene upstream promoter regions to interfere with downstream gene expression, 2) inhibiting RNA polymerase II or mediating chromatin remodeling and histone modifications to affect downstream gene expression, 3) disturbing mRNA splicing by complementary binding with protein-coding pre-mRNAs, 4) producing endogenous siRNAs under control of Dicer, 5) combining with specific proteins to modulate corresponding protein activities, 6) being components of nucleic acid–protein complexes, 7) binding to the specific proteins and changing cellular localization of the proteins, and 8) serving as precursors of small RNAs.2527 Because of their functional diversity, ncRNAs are involved in the regulation of a variety of biological processes, especially those of cancer progression and metastasis.

Recently, the discovery of stable miRNAs in bodily fluids introduced new insights into ncRNAs and miRNAs that can lead to new diagnostic approaches using less invasive assays.28 In addition to being packed into exosomes or microvesicles, circulating miRNAs are also combined delivered with high-density lipoproteins29 or AGO2 proteins30 to maintain stability. Gallo et al reported that most detectable circulating miRNAs in serum and saliva were concentrated within exosomes,31 which suggests that exosomes and exosomal ncRNAs are the primary forms of cellular RNA-based communication. Huang et al performed RNA sequencing to annotate the exosomal RNA species from the plasma of 23 CRPC patients following the guideline previously reported.32 Among the mapped reads, mature miRNAs were the most common and represented 41.72% of reads, followed by piRNAs at 20.92%, lncRNAs at 20.19%, and mRNAs at 6.52%. Collectively, six other RNA species constituted 10.64% of the mappable sequences.33 During cancer metastases, exosomes can act as delivery vehicles of circulating ncRNAs and transport them from primary to metastatic cancer sites11,34,35 and mediate adaptations between cancer and stromal cells. This mechanism is similar to that of primary cancer cell motility, which is modulated by EMT processes or establishment of favorable environments at possible metastatic sites to aid in neoplastic cell survival.36

Differently expressed circulating miRNAs in PCa patients

Four miRNAs altered in the serum of transgenic adenocarcinoma of mouse prostate mice – miR-141, miR-298, miR-346, and miR-375 – also showed concordant changes in men with metastatic castration-resistant PCa (mCRPC).37 Yaman Agaoglu et al found that the differences in miRNA plasma levels between the healthy controls and the patients were highly significant for the miR-21 (P<0.001) and -221 (P<0.001) but not for the miR-141(P=0.23).38 An analysis of 742 miRNAs using plasma-derived circulating microvesicles (cMVs) from 78 PCa patients and 28 healthy individuals identified differentially expressed miRNAs that included upregulation of miR-107, -130b, -141, -2110, -301a, -326, -331-3p, -432, -484, -574-3p, and -625 and downregulation of miR-181a-2. In urine samples, miR-107 and miR-574-3p concentrations were also higher in patients compared with controls.39 These clinical results suggested that circulating miRNAs can assist in the detection of PCa. Moreover, some miRNAs have been identified that are associated with PCa progression, staging, and outcomes, and these miRNAs could be more useful in clinical practice.

Metastasis-associated circulating miRNAs in PCa patients

The first report to evaluate the utility of circulating miRNAs to distinguish PCa patients with metastasis from those with advanced, but localized disease revealed that miRNA-21, -221, and -141 plasma levels were significantly higher in the metastatic cohort.38 Circulating miRNAs were also reported to be significantly upregulated in the serum of metastatic PCa patients, and miR-375 was the most prominent and the only miRNA that showed greater serum concentrations in PCa patients with distant metastases compared with that of PCa patients with localized PCa. This study also confirmed that circulating miR-375 and miR-141 correlated with the risk of disease progression.40 An analysis using plasma samples from 16 PCa patients with metastatic disease and 55 PCa patients with localized disease found 16 miRNAs that were differently expressed in cMVs. Among these 16 miRNAs, 15 had significantly greater concentrations and one (miR-572) had significantly lower concentrations in PCa patients with metastatic disease compared with those of PCa patients with localized disease. In a subsequent study, cMVs and exosomes were separated in the serum of 47 PCa patients with metastatic disease and 72 PCa patients in remission. This study showed that miR-375 and miR-141 were significantly increased in both the cMVs and the exosomes.39 Another study demonstrated that miR-375, -141, and -378 expression was significantly upregulated and miR-409-3p expression was significantly downregulated in the serum of CRPC patients compared with patients who had a low risk of developing CRPC.41 Overall, over 30 differentially expressed circulating miRNAs have been identified. A summary of differentially circulating ncRNAs found in PCa patients with metastatic disease compared with those with localized disease is presented in Table 1. Only miR-375 and miR-141 have higher repetition rates. Some deviations in the compiled data could be due to the different study designs, cohort selections, treatment strategies, methods of sample collection, and the sensitivity and specificity of platforms used.42

Table 1 ncRNAs proven to be differently expressed in the circulation of prostate cancer patients with metastatic disease compared with those with localized disease

Abbreviations: ncRNA, noncoding RNA; PCAT18, prostate cancer-associated noncoding RNA transcript 18.

Table 2 illustrates miRNA and lncRNA targets that regulate the development of PCa metastases and that are involved in signaling pathways. Overall, the mechanisms behind how miRNAs function in PCa metastases have not been substantially studied; however, a few preclinical studies have been reported. miR-375 has been shown to have multiple functions in several different types of cancer. In most cancer cases, miR-375 plays a role in tumor suppression of several cancer types, such as head and neck squamous cell carcinoma,43,44 esophageal cancer,45 gastric cancer,46 pancreatic cancer,47 and hepatocellular carcinoma.48,49 However, some studies implicate miR-375 upregulation in the propagation of PCa.50,51 miR-375 might play a dual role in prostate carcinogenesis.52 The reason for the antagonistic roles of miR-375 in different study toward PCa could be attributed to PCa heterogeneity. Circulating miR-375 expression is considered a prognostic biomarker for PCa.33,53 Higher miRNA-375 expression in circulation is the most confirmed miRNA that is related to metastatic PCa.37,40,41,50,54,55 However, scant information regarding the mechanism of circulating miR-375 in regulating metastatic spread of PCa has been discovered. A couple of studies showed that miR-375 promotes cell growth by targeting Sec23A.56,57 This was validated by a recent study, which demonstrated that miR-375 exerts its oncogenic effects by targeting CBX7 and thus regulates critical cancer pathways such as the EMT and Wnt/β-catenin signaling pathways.58 More details on these processes remain to be determined.

Table 2 Proven targets and signal pathways of ncRNAs in prostate cancer patients with metastatic disease

Abbreviations: AR, androgen receptor; EMT, epithelial–mesenchymal transition; MMP, matrix metalloproteinase; ncRNA, noncoding RNA; PCAT18, prostate cancer-associated noncoding RNA transcript 18.

miR-200 family including miR-200a, -200b, miR-200c, -141, and -429 was found to be associated with several cancer types. Members of this family directly target the metastasis-promoting protein, WAVE3, to inhibit PCa cell invasion.59 These miRNAs also target the E-cadherin transcriptional repressors, ZEB1 and ZEB2, and were identified as determining factors in the development of cancer cell epithelial phenotypes.6064 Moreover, miR-200b negatively regulates vascular endothelial growth factor (VEGF) signaling by targeting the VEGF and the VEGF receptor and could have therapeutic potential as an angiogenic inhibitor.65 However, the miR-200 family has been associated with Sec23-mediated inhibition of metastasis-suppressing proteins66 and adhesion improvements at distant sites to promote EMT and increased colonization.67 Therefore, the miR-200 family was found to have different roles in various stages of metastasis.36

miR-141, a special member of the miR-200 family, has been reported to be a tumor suppressor in several malignancies, such as gastric cancer,68,69 pancreatic cancer,70 breast cancer,71,72 renal cell carcinoma,73,74 and hepatocellular carcinoma.75 Recently, circulating miR-141 was positively upregulated in the blood of metastatic PCa patients.3739,41,76,77 miR-141 targeted the small heterodimer partner protein to modulate androgen receptor (AR)-regulated transcriptional activity in AR-responsive LNCaP cells,78 and AR signaling was closely related to PCa formation,79 progression,80 and metastasis.81 miR-141 inhibits cell migration and invasion by targeting transforming growth factor (TGF)-β282 and insulin receptor substrate 2.83 miR-141 was also reported to regulate the transcriptional coactivator, PDZ-binding motif (TAZ), a transcription cofactor that plays pivotal roles in the EMT.84

miR-21 is one of the most widely studied metastasis-related miRNAs, which have been shown to be at significantly higher levels in the blood of PCa patients with metastases compared with those who had localized or advanced disease.38 This molecule was also shown to promote cancer metastasis by regulating several aspects of the metastatic cascade. miR-21 promotes tumor cell invasion by targeting the tumor suppressor PDCD4, an inhibitor of the urokinase receptor, prometastatic factor.85 miR-21 also modulates the expression of matrix metalloproteinases (MMPs) by directly targeting the MMP inhibitors, such as PTEN,86 RECK,87 and TIMP388 to degrade the ECM and promote cancer metastasis. Exosomal miR-21 can also regulate tumor microenvironments by binding to Toll-like receptors (TLRs) in immune cells, which triggers a TLR-mediated prometastatic inflammatory response that can ultimately lead to tumor growth and metastasis.89

miR-126 was also found to be upregulated in the plasma of metastatic PCa patients,55 and miR-126* regulates protein translation and invasion of PCa LNCaP cells.90 Moreover, miR-126 has a suppressor role in cancer cell invasion through direct repression of a disintegrin and metalloprotease 9 (ADAM9).91 Another study identified that when miR-126 partnered with another miR-126, the combination repressed mesenchymal stem cell and monocyte recruitment into the tumor stroma, which effectively inhibited tumor cell invasion and metastasis.92

miR-152 upregulation in plasma samples from mCRPC patients was confirmed compared with those that had localized PCa.55 miR-152 could also control PCa cell migration and invasive potential by directly targeting TGFα.93 In other types of cancer, miR-152 was shown to inhibit non-small-cell lung cancer cell metastasis by targeting neuropilin-1.94 Elevated levels of circulating miR-221 were detected in PCa patients with metastases.38 miR-221 promotes the EMT by targeting PTEN in extrahepatic cholangiocarcinoma, which forms a positive feedback loop with the β-catenin/c-Jun signaling pathway95 and whose inhibition led to reduced cell migration in PCa cells that was partly mediated by Sirtuin 1 (SIRT1) activation.96 miR-221 also promoted cancer invasion and angiogenesis by targeting TIMP2,97 TIMP3, and PTEN.98 miR-205 and miR-409-3p were downregulated in the circulation of metastatic PCa patients, and it was confirmed that miR-409-3p regulated the EMT process.61,99,100 In summary, circulating miRNAs are present and have diverse roles throughout the entire metastatic process of PCa. In one stage of metastasis, multiple miRNAs have roles in regulating the process.

Metastasis-related circulating lncRNA in PCa

As seen in Table 2, there are many fewer reports about circulating lncRNAs than miRNAs. For many years, studies mainly focused on differential lncRNA expression in PCa tissues. Recently, significant differential expression of circulating lncRNAs in metastatic PCa compared with localized PCa has been disclosed. Prostate cancer-associated noncoding RNA transcript 18 (PCAT18) was detected in PCa patient plasma, and its expression was shown to be increased as PCa progresses from localized to metastatic disease, which suggests that PCAT18 is a potential biomarker for metastatic PCa. PCAT18 expression was significantly associated with AR signaling, and PCAT18 silencing was shown to inhibit PCa cell proliferation, migration, and invasion: however, the molecular mechanisms behind its actions remain unclear.101 A few other lncRNAs were reported to be involved with PCa metastatic disease, but the differential expression in the blood of PCa patients with metastatic disease compared with PCa patients with localized disease remains uncovered. The prostate cancer antigen 3 lncRNA (PCA3; also referred to as DD3) was initially found to be significantly overexpressed in more than 95% of primary and metastatic PCa tissue sections.102 PCA3 can be detected in urine and used as a noninvasive method for PCa diagnoses.103,104 Moreover, higher PCA3 scores were associated with greater tumor aggressiveness.105

Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is the most studied metastatic-associated lncRNA and has been shown to be overexpressed during PCa progression.106 In early studies, MALAT1 transcript fragments were higher in the plasma of PCa patients compared with that of non-PCa patients.107 MALAT1 is associated with EMT; loss of MALAT1 results in reduced LPHN2 and ABCA1 levels, which are important factors of EMT.108 MALAT1 knockdown in PCa cell lines abrogates cell growth, migration, and invasion and induces G0/G1 cell cycle arrest.109 HOX transcript antisense RNA (HOTAIR) expression increases in PCa cells during growth and invasion.110 HOTAIR is also connected with both EMT and ECM remodeling. Therefore, the regulation of MMP expression partially mediates HOTAIR functions in cancer metastasis. Moreover, HOTAIR could affect EMT processes by modulating specific EMT-related genes.111113 A recent study demonstrated that lncRNA CCAT2 knockdown stimulated EMT by abrogating N-cadherin and vimentin expression and by intensifying E-cadherin expression in PCa.114

A multi-institutional high-throughput analysis of PCa tissue samples from 1,008 patients confirmed the prognostic value of measuring lncRNA, SChLAP1 (second chromosome locus associated with prostate-1) levels to detect metastatic progression.115 SChLAP1 impairs the SNF5-mediated regulation of gene expression and genomic binding to coordinate cancer cell invasion.116 A study recently identified the lncRNA, Linc00963, which regulates the epidermal growth factor receptor (EGFR) signaling pathway to promote PCa cell growth, migration, and invasion.117

Ongoing PCa clinical trials

With the accumulating evidence from preclinical studies, clinical trials have recently been launched. The Medical College of Wisconsin (Milwaukee, Wisconsin) sponsored a study in June 2017 that is recruiting PCa patients (estimated to 60), with aims to identify exosomal miRNAs that can predict responses to ADT. Patients who were intermediate to high risk at diagnosis, those who were treated but now have recurrent disease, and those with metastatic disease are recruited to the study. Blood samples will be collected at the time of pretreatment, 3 months’ posttreatment, and during disease progression. Exosomal RNA markers that predict responses to ADT will be validated.

Another observational study in 2016 sponsored by the Assuta Medical Center (Israel) and enrolling as many as 120 PCa patients is aiming to find correlations among circulating miRNAs associated with PCa metastases to bones and lymph nodes using PET imaging.

In another study sponsored by the Hospital of Ghent University (Belgium), blood from PCa patients with lymph node metastases will be collected to examine the role of circulating miRNAs as a biomarker to diagnose and predict relapse-free survival rates. The sample size is expected to be 330. None of the clinical trials mentioned have posted results, which we eagerly await.

Conclusion and prospects

The identification of circulating ncRNA provides novel biomarkers and therapeutic targets for mCRPC. Several molecules have been proven to serve as predictors of PCa metastases. Exosomes protect the stability of circulating ncRNAs in body fluids and can be easily quantified. Exosomal ncRNA expression is related to cancer progression and the microenvironments during cancer metastasis. It is well accepted that the communication between cancer cells and the extracellular environments begin in the early stage of metastasis before the cancer cells move to their colonized sites. Therefore, more studies have shown that circulating ncRNAs are associated with the PCa stages.101,118,119 The results of these studies suggest that circulating ncRNAs could be promising as biomarkers to predict cancer metastases.

Further understanding of ncRNAs has provided novel insights into PCa therapeutics. In the last few years, ncRNA-based cancer therapeutics inspire the interest of researchers.120122 However, the methods needed to deliver miRNA mimics or inhibitors to target cells steadily and effectively remain a crucial obstacle. Exosomes have been studied as vehicle transporting molecules that can deliver genetic materials to target sites with high efficiency.123,124 Ohno et al performed a study using exosomes to deliver let-7a to EGFR-expressing breast cancer tissues.125 Exosomal ncRNAs for clinical use require more research to resolve problems, such as enhancing exosome production and reducing immunogenicity. Emerging evidence suggests that exosomal ncRNA could eventually play an essential role in metastatic PCa treatment.

In summary, abundant research on exosomal miRNAs is present, while much less evidence exists that determines if lncRNAs may play an essential role in PCa metastases. With new technologies evolving at a rapid pace, lncRNA studies will overcome limitations and more promising lncRNAs will be discovered. However, in the field of ncRNAs, many challenges and opportunities still exist.

Acknowledgments

This study was jointly supported by the National Natural Science Foundation of China (Grant No. 81572528), the Haiyan Foundation of Harbin Medical University Cancer Hospital, Outstanding Youth Fund of the Heilongjiang Province (Grant No. JC2018024), and the Heilongjiang Innovation Ability Promoting Program for Scientific Research Institutions (Grant No. YC2016D002) to XH.

Disclosure

The authors report no conflicts of interest in this work.

References

1.

Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65(2):87–108.

2.

Hathaway AR, Baker MK, Sonpavde G. Emerging agents for the therapy of advanced prostate cancer. Future Oncol. 2015;11(20):2775–2787.

3.

Faleiro I, Leão R, Binnie A, de Mello RA, Maia AT, Castelo-Branco P. Epigenetic therapy in urologic cancers: an update on clinical trials. Oncotarget. 2017;8(7):12484–12500.

4.

Macfarlane RJ, Chi KN. Research in castration-resistant prostate cancer: what does the future hold? Curr Oncol. 2010;17(Suppl 2):S80–86.

5.

Torpy J, Lynm C, Glass RM. Non-coding RNAs in cancer brain metastasis. Front Biosci. 2016;8:187–202.

6.

Psaila B, Lyden D. The metastatic niche: adapting the foreign soil. Nat Rev Cancer. 2009;9(4):285–293.

7.

Paget S. The distribution of secondary growths in cancer of the breast. 1889. Cancer Metastasis Rev. 1989;8(2):98–101.

8.

Costa-Silva B, Aiello NM, Ocean AJ, et al. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat Cell Biol. 2015;17(6):816–826.

9.

Bubendorf L, Schöpfer A, Wagner U, et al. Metastatic patterns of prostate cancer: an autopsy study of 1,589 patients. Hum Pathol. 2000;31(5):578–583.

10.

Sun YX, Schneider A, Jung Y, et al. Skeletal localization and neutralization of the SDF-1(CXCL12)/CXCR4 axis blocks prostate cancer metastasis and growth in osseous sites in vivo. J Bone Miner Res. 2005;20(2):318–329.

11.

Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ, Lötvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007;9(6):654–659.

12.

Jiang C, Li X, Zhao H, Liu H. Long non-coding RNAs: potential new biomarkers for predicting tumor invasion and metastasis. Mol Cancer. 2016;15(1):62.

13.

Ahadi A, Brennan S, Kennedy PJ, Hutvagner G, Tran N. Long non-coding RNAs harboring miRNA seed regions are enriched in prostate cancer exosomes. Sci Rep. 2016;6:24922.

14.

Zhang J, Li S, Li L, et al. Exosome and exosomal microRNA: trafficking, sorting, and function. Genomics Proteomics Bioinformatics. 2015;13(1):17–24.

15.

Sato-Kuwabara Y, Melo SA, Soares FA, Calin GA. The fusion of two worlds: non-coding RNAs and extracellular vesicles--diagnostic and therapeutic implications (Review). Int J Oncol. 2015;46(1):17–27.

16.

Heijnen HF, Schiel AE, Fijnheer R, Geuze HJ, Sixma JJ. Activated platelets release two types of membrane vesicles: microvesicles by surface shedding and exosomes derived from exocytosis of multivesicular bodies and alpha-granules. Blood. 1999;94(11):3791–3799.

17.

Ostenfeld MS, Jeppesen DK, Laurberg JR, et al. Cellular disposal of miR23b by RAB27-dependent exosome release is linked to acquisition of metastatic properties. Cancer Res. 2014;74(20):5758–5771.

18.

Ohshima K, Inoue K, Fujiwara A, et al. Let-7 microRNA family is selectively secreted into the extracellular environment via exosomes in a metastatic gastric cancer cell line. PLoS One. 2010;5(10): e13247.

19.

Montecalvo A, Larregina AT, Shufesky WJ, et al. Mechanism of transfer of functional microRNAs between mouse dendritic cells via exosomes. Blood. 2012;119(3):756–766.

20.

Liu Y, Xiang X, Zhuang X, et al. Contribution of MyD88 to the tumor exosome-mediated induction of myeloid derived suppressor cells. Am J Pathol. 2010;176(5):2490–2499.

21.

Gibb EA, Brown CJ, Lam WL. The functional role of long non-coding RNA in human carcinomas. Mol Cancer. 2011;10:38.

22.

Mattick JS. Non-coding RNAs: the architects of eukaryotic complexity. EMBO Rep. 2001;2(11):986–991.

23.

Esteller M. Non-coding RNAs in human disease. Nat Rev Genet. 2011;12(12):861–874.

24.

Doench JG, Sharp PA. Specificity of microRNA target selection in translational repression. Genes Dev. 2004;18(5):504–511.

25.

Batista PJ, Chang HY. Long noncoding RNAs: cellular address codes in development and disease. Cell. 2013;152(6):1298–1307.

26.

Wilusz JE, Sunwoo H, Spector DL. Long noncoding RNAs: functional surprises from the RNA world. Genes Dev. 2009;23(13):1494–1504.

27.

Ponting CP, Oliver PL, Reik W. Evolution and functions of long noncoding RNAs. Cell. 2009;136(4):629–641.

28.

Chen X, Ba Y, Ma L, et al. Characterization of microRNAs in serum: a novel class of biomarkers for diagnosis of cancer and other diseases. Cell Res. 2008;18(10):997–1006.

29.

Vickers KC, Palmisano BT, Shoucri BM, Shamburek RD, Remaley AT. MicroRNAs are transported in plasma and delivered to recipient cells by high-density lipoproteins. Nat Cell Biol. 2011;13(4):423–433.

30.

Arroyo JD, Chevillet JR, Kroh EM, et al. Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma. Proc Natl Acad Sci U S A. 2011;108(12):5003–5008.

31.

Gallo A, Tandon M, Alevizos I, Illei GG. The majority of microRNAs detectable in serum and saliva is concentrated in exosomes. PLoS One. 2012;7(3):e30679.

32.

Huang X, Yuan T, Tschannen M, et al. Characterization of human plasma-derived exosomal RNAs by deep sequencing. BMC Genomics. 2013;14:319.

33.

Huang X, Yuan T, Liang M, et al. Exosomal miR-1290 and miR-375 as prognostic markers in castration-resistant prostate cancer. Eur Urol. 2015;67(1):33–41.

34.

Alhasan AH, Patel PC, Choi CH, Mirkin CA. Exosome encased spherical nucleic acid gold nanoparticle conjugates as potent microRNA regulation agents. Small. 2014;10(1):186–192.

35.

Minh TN, Ph L, Guo C, et al. miR-200–containing extracellular vesicles promote breast cancer cell metastasis. J Clin Invest. 2014;124(12):5109–5128.

36.

Alečković M, Kang Y. Regulation of cancer metastasis by cell-free miRNAs. Biochim Biophys Acta. 2015;1855(1):24–42.

37.

Selth LA, Townley S, Gillis JL, et al. Discovery of circulating microRNAs associated with human prostate cancer using a mouse model of disease. Int J Cancer. 2012;131(3):652–661.

38.

Yaman Agaoglu F, Kovancilar M, Dizdar Y, et al. Investigation of miR-21, miR-141, and miR-221 in blood circulation of patients with prostate cancer. Tumour Biol. 2011;32(3):583–588.

39.

Bryant RJ, Pawlowski T, Catto JW, et al. Changes in circulating microRNA levels associated with prostate cancer. Br J Cancer. 2012;106(4):768–774.

40.

Brase JC, Johannes M, Schlomm T, et al. Circulating miRNAs are correlated with tumor progression in prostate cancer. Int J Cancer. 2011;128(3):608–616.

41.

Nguyen HC, Xie W, Yang M, et al. Expression differences of circulating microRNAs in metastatic castration resistant prostate cancer and low-risk, localized prostate cancer. Prostate. 2013;73(4):346–354.

42.

Josson S, Chung LW, Gururajan M. microRNAs and prostate cancer. Adv Exp Med Biol. 2015;889:105–118.

43.

Kinoshita T, Nohata N, Yoshino H, et al. Tumor suppressive microRNA-375 regulates lactate dehydrogenase B in maxillary sinus squamous cell carcinoma. Int J Oncol. 2012;40(1):185–193.

44.

Harris T, Jimenez L, Kawachi N, et al. Low-level expression of miR-375 correlates with poor outcome and metastasis while altering the invasive properties of head and neck squamous cell carcinomas. Am J Pathol. 2012;180(3):917–928.

45.

Kong KL, Kwong DL, Chan TH, et al. MicroRNA-375 inhibits tumour growth and metastasis in oesophageal squamous cell carcinoma through repressing insulin-like growth factor 1 receptor. Gut. 2012;61(1):33–42.

46.

Tsukamoto Y, Nakada C, Noguchi T, et al. MicroRNA-375 is downregulated in gastric carcinomas and regulates cell survival by targeting PDK1 and 14-3-3zeta. Cancer Res. 2010;70(6):2339–2349.

47.

Bhatti I, Lee A, James V, et al. Knockdown of microRNA-21 inhibits proliferation and increases cell death by targeting programmed cell death 4 (PDCD4) in pancreatic ductal adenocarcinoma. J Gastrointest Surg. 2011;15(1):199–208.

48.

Liu AM, Poon RT, Luk JM. MicroRNA-375 targets Hippo-signaling effector YAP in liver cancer and inhibits tumor properties. Biochem Biophys Res Commun. 2010;394(3):623–627.

49.

He XX, Chang Y, Meng FY, et al. MicroRNA-375 targets AEG-1 in hepatocellular carcinoma and suppresses liver cancer cell growth in vitro and in vivo. Oncogene. 2012;31(28):3357–3369.

50.

Cheng HH, Mitchell PS, Kroh EM, et al. Circulating microRNA profiling identifies a subset of metastatic prostate cancer patients with evidence of cancer-associated hypoxia. PLoS One. 2013;8(7):e69239.

51.

Nguyen HC, Xie W, Yang M, et al. Expression differences of circulating microRNAs in metastatic castration resistant prostate cancer and low-risk, localized prostate cancer. Prostate. 2013;73(4):346–354.

52.

Costa-Pinheiro P, Ramalho-Carvalho J, Vieira FQ, et al. MicroRNA-375 plays a dual role in prostate carcinogenesis. Clin Epigenetics. 2015;7:42.

53.

Kachakova D, Mitkova A, Popov E, et al. Combinations of serum prostate-specific antigen and plasma expression levels of let-7c, miR-30c, miR-141, and miR-375 as potential better diagnostic biomarkers for prostate cancer. DNA Cell Biol. 2015;34(3):189–200.

54.

Hori N, Narita M, Yamashita A, et al. Changes in the expression of IL-6-mediated microRNAs in the dorsal root ganglion under neuropathic pain in mice. Synapse. 2016;70(8):317–324.

55.

Watahiki A, Macfarlane RJ, Gleave ME, et al. Plasma miRNAs as biomarkers to identify patients with castration-resistant metastatic prostate cancer. Int J Mol Sci. 2013;14(4):7757–7770.

56.

Szczyrba J, Nolte E, Wach S, et al. Downregulation of Sec23A protein by miRNA-375 in prostate carcinoma. Mol Cancer Res. 2011;9(6):791–800.

57.

Wang Y, Lieberman R, Pan J, et al. miR-375 induces docetaxel resistance in prostate cancer by targeting SEC23A and YAP1. Mol Cancer. 2016;15(1):70.

58.

Pickl JM, Tichy D, Kuryshev VY, et al. Ago-RIP-Seq identifies Polycomb repressive complex I member CBX7 as a major target of miR-375 in prostate cancer progression. Oncotarget. 2016;7(37):59589–59603.

59.

Sossey-Alaoui K, Bialkowska K, Plow EF. The miR200 family of microRNAs regulates WAVE3-dependent cancer cell invasion. J Biol Chem. 2009;284(48):33019–33029.

60.

Park SM, Gaur AB, Lengyel E, Peter ME. The miR-200 family determines the epithelial phenotype of cancer cells by targeting the E-cadherin repressors ZEB1 and ZEB2. Genes Dev. 2008;22(7):894–907.

61.

Gregory PA, Bert AG, Paterson EL, et al. The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat Cell Biol. 2008;10(5):593–601.

62.

Hurteau GJ, Carlson JA, Spivack SD, Brock GJ. Overexpression of the microRNA hsa-miR-200c leads to reduced expression of transcription factor 8 and increased expression of E-cadherin. Cancer Res. 2007;67(17):7972–7976.

63.

Burk U, Schubert J, Wellner U, et al. A reciprocal repression between ZEB1 and members of the miR-200 family promotes EMT and invasion in cancer cells. EMBO Rep. 2008;9(6):582–589.

64.

Korpal M, Lee ES, Hu G, Kang Y. The miR-200 family inhibits epithelial-mesenchymal transition and cancer cell migration by direct targeting of E-cadherin transcriptional repressors ZEB1 and ZEB2. J Biol Chem. 2008;283(22):14910–14914.

65.

Choi YC, Yoon S, Jeong Y, Yoon J, Baek K. Regulation of vascular endothelial growth factor signaling by miR-200b. Mol Cells. 2011;32(1):77–82.

66.

Korpal M, Ell BJ, Buffa FM, et al. Direct targeting of Sec23a by miR-200s influences cancer cell secretome and promotes metastatic colonization. Nat Med. 2011;17(9):1101–1108.

67.

Dykxhoorn DM, Wu Y, Xie H, et al. miR-200 enhances mouse breast cancer cell colonization to form distant metastases. PLoS One. 2009;4(9):e7181.

68.

Zhou X, Wang Y, Shan B, et al. The downregulation of miR-200c/141 promotes ZEB1/2 expression and gastric cancer progression. Med Oncol. 2015;32(1):428.

69.

du Y, Xu Y, Ding L, et al. Down-regulation of miR-141 in gastric cancer and its involvement in cell growth. J Gastroenterol. 2009;44(6):556–561.

70.

Zhu ZM, Xu YF, Su QJ, Yf X, Qj S, et al. Prognostic significance of microRNA-141 expression and its tumor suppressor function in human pancreatic ductal adenocarcinoma. Mol Cell Biochem. 2014;388(1–2):39–49.

71.

Xu F, He H, Huang W, et al. Decreased expression of microRNA-200 family in human breast cancer is associated with lymph node metastasis. Clin Transl Oncol. 2016;18(3):283–288.

72.

O’Day E, Lal A. MicroRNAs and their target gene networks in breast cancer. Breast Cancer Res. 2010;12(2):201.

73.

Yu XY, Zhang Z, Liu J, Zhan B, Kong CZ. MicroRNA-141 is downregulated in human renal cell carcinoma and regulates cell survival by targeting CDC25B. Onco Targets Ther. 2013;6:349–354.

74.

Nakada C, Matsuura K, Tsukamoto Y, et al. Genome-wide microRNA expression profiling in renal cell carcinoma: significant down-regulation of miR-141 and miR-200c. J Pathol. 2008;216(4):418–427.

75.

Xue J, Niu YF, Huang J, et al. miR-141 suppresses the growth and metastasis of HCC cells by targeting E2F3. Tumour Biol. 2014;35(12):12103–12107.

76.

Li Z, Ma YY, Wang J, et al. Exosomal microRNA-141 is upregulated in the serum of prostate cancer patients. Onco Targets Ther. 2016;9:139–148.

77.

Zhang HL, Qin XJ, Cao DL, et al. An elevated serum miR-141 level in patients with bone-metastatic prostate cancer is correlated with more bone lesions. Asian J Androl. 2013;15(2):231–235.

78.

Xiao J, Gong AY, Eischeid AN, et al. miR-141 modulates androgen receptor transcriptional activity in human prostate cancer cells through targeting the small heterodimer partner protein. Prostate. 2012;72(14):1514–1522.

79.

Culig Z, Bartsch G. Androgen axis in prostate cancer. J Cell Biochem. 2006;99(2):373–381.

80.

Li H, Xie N, Chen R, et al. UGT2B17 expedites progression of castration-resistant prostate cancers by promoting ligand-independent AR signaling. Cancer Res. 2016;76(22):6701–6711.

81.

Hu S, Li L, Yeh S, et al. Infiltrating T cells promote prostate cancer metastasis via modulation of FGF11→miRNA-541→androgen receptor (AR)→MMP9 signaling. Mol Oncol. 2015;9(1):44–57.

82.

Peng T, Zhang S, Li W, Fu S, Luan Y, Zuo L. MicroRNA-141 inhibits glioma cells growth and metastasis by targeting TGF-β2. Am J Transl Res. 2016;8(8):3513–3521.

83.

Dong S, Meng X, Xue S, Yan Z, Ren P, Liu J. MicroRNA-141 inhibits thyroid cancer cell growth and metastasis by targeting insulin receptor substrate 2. Am J Transl Res. 2016;8(3):1471–1481.

84.

Zuo QF, Zhang R, Li BS, et al. MicroRNA-141 inhibits tumor growth and metastasis in gastric cancer by directly targeting transcriptional co-activator with PDZ-binding motif, TAZ. Cell Death Dis. 2015;6:e1623.

85.

Asangani IA, Rasheed SA, Nikolova DA, et al. MicroRNA-21 (miR-21) post-transcriptionally downregulates tumor suppressor Pdcd4 and stimulates invasion, intravasation and metastasis in colorectal cancer. Oncogene. 2008;27(15):2128–2136.

86.

Meng F, Henson R, Wehbe-Janek H, Ghoshal K, Jacob ST, Patel T. MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer. Gastroenterology. 2007;133(2):647–658.

87.

Reis ST, Pontes-Junior J, Antunes AA, et al. miR-21 may acts as an oncomir by targeting RECK, a matrix metalloproteinase regulator, in prostate cancer. BMC Urol. 2012;12:14.

88.

Gabriely G, Wurdinger T, Kesari S, et al. MicroRNA 21 promotes glioma invasion by targeting matrix metalloproteinase regulators. Mol Cell Biol. 2008;28(17):5369–5380.

89.

Fabbria M. MicroRNAs bind to Toll like receptors to induce prometastatic inflammatory response. 2012;109(31):E2110–2116.

90.

Musiyenko A, Bitko V, Barik S. Ectopic expression of miR-126*, an intronic product of the vascular endothelial EGF-like 7 gene, regulates prostein translation and invasiveness of prostate cancer LNCaP cells. J Mol Med. 2008;86(3):313–322.

91.

Wang CZ, Yuan P, Li Y. miR-126 regulated breast cancer cell invasion by targeting ADAM9. Int J Clin Exp Pathol. 2015;8(6):6547–6553.

92.

Zhang Y, Yang P, Sun T, et al. miR-126 and miR-126* repress recruitment of mesenchymal stem cells and inflammatory monocytes to inhibit breast cancer metastasis. Nat Cell Biol. 2013;15(3):284–294.

93.

Zhu C, Li J, Ding Q, et al. miR-152 controls migration and invasive potential by targeting TGFα in prostate cancer cell lines. Prostate. 2013;73(10):1082–1089.

94.

Zhang YJ, Liu XC, du J, Zhang YJ. miR-152 regulates metastases of non-small cell lung cancer cells by targeting neuropilin-1. Int J Clin Exp Pathol. 2015;8(11):14235–14240.

95.

Li J, Yao L, Li G, et al. miR-221 promotes epithelial-mesenchymal transition through targeting PTEN and forms a positive feedback loop with β-catenin/c-Jun signaling pathway in extra-hepatic cholangiocarcinoma. PLoS One. 2015;10(10):e0141168.

96.

Yang X, Yang Y, Gan R, et al. Down-regulation of mir-221 and mir-222 restrain prostate cancer cell proliferation and migration that is partly mediated by activation of SIRT1. PLoS One. 2014;9(6):e98833.

97.

Yang F, Wang W, Zhou C, et al. miR-221/222 promote human glioma cell invasion and angiogenesis by targeting TIMP2. Tumour Biol. 2015;36(5):3763–3773.

98.

Garofalo M, di Leva G, Romano G, et al. miR-221&222 regulate TRAIL resistance and enhance tumorigenicity through PTEN and TIMP3 downregulation. Cancer Cell. 2009;16(6):498–509.

99.

Tucci P, Agostini M, Grespi F, et al. Loss of p63 and its microRNA-205 target results in enhanced cell migration and metastasis in prostate cancer. Proc Natl Acad Sci U S A. 2012;109(38):15312–15317.

100.

Josson S, Gururajan M, Hu P, et al. miR-409-3p/-5p promotes tumorigenesis, epithelial-to-mesenchymal transition, and bone metastasis of human prostate cancer. Clin Cancer Res. 2014;20(17):4636–4646.

101.

Crea F, Watahiki A, Quagliata L, et al. Identification of a long non-coding RNA as a novel biomarker and potential therapeutic target for metastatic prostate cancer. Oncotarget. 2014;5(3):764–774.

102.

Hessels D, Klein Gunnewiek JMT, van Oort I, van OI, et al. DD3PCA3-based molecular urine analysis for the diagnosis of prostate cancer. Eur Urol. 2003;44(1):8–16.

103.

Xu Y-H, Xue W-J, Ying X-L, Jiang J-H, Yh X. Prostate cancer antigen 3 as a biomarker in the urine for prostate cancer diagnosis: a meta-analysis. J Cancer Res Ther. 2014;10(7):218–221.

104.

Hu B, Yang H, Yang H. Diagnostic value of urine prostate cancer antigen 3 test using a cutoff value of 35 μg/L in patients with prostate cancer. Tumor Biol. 2014;35(9):8573–8580.

105.

Merola R, Tomao L, Antenucci A, et al. PCA3 in prostate cancer and tumor aggressiveness detection on 407 high-risk patients: a National Cancer Institute experience. J Exp Clin Cancer Res. 2015;34:15.

106.

Wang D, Ding L, Wang L, et al. lncRNA MALAT1 enhances oncogenic activities of EZH2 in castration-resistant prostate cancer. Oncotarget. 2015;6(38):41045–41055.

107.

Ren S, Wang F, Shen J, et al. Long non-coding RNA metastasis associated in lung adenocarcinoma transcript 1 derived miniRNA as a novel plasma-based biomarker for diagnosing prostate cancer. Eur J Cancer. 2013;49(13):2949–2959.

108.

Gutschner T, Hämmerle M, Eissmann M, et al. The noncoding RNA MALAT1 is a critical regulator of the metastasis phenotype of lung cancer cells. Cancer Res. 2013;73(3):1180–1189.

109.

Ren S, Liu Y, Xu W, et al. Long noncoding RNA MALAT-1 is a new potential therapeutic target for castration resistant prostate cancer. J Urol. 2013;190(6):2278–2287.

110.

Zhang A, Zhao JC, Kim J, et al. LncRNA HOTAIR enhances the androgen-receptor-mediated transcriptional program and drives castration-resistant prostate cancer. Cell Rep. 2015;13(1):209–221.

111.

Xu ZY, Yu QM, Du YA, et al. Knockdown of long non-coding RNA HOTAIR suppresses tumor invasion and reverses epithelial-mesenchymal transition in gastric cancer. Int J Biol Sci. 2013;9(6):587–597.

112.

Wu S, Zheng C, Chen S, et al. Overexpression of long non-coding RNA HOTAIR predicts a poor prognosis in patients with acute myeloid leukemia. Oncol Lett. 2015;10(4):2410–2414.

113.

Wu Y, Zhang L, Zhang L, et al. Long non-coding RNA HOTAIR promotes tumor cell invasion and metastasis by recruiting EZH2 and repressing E-cadherin in oral squamous cell carcinoma. Int J Oncol. 2015;46(6):2586–2594.

114.

Zheng J, Zhao S, He X, et al. The up-regulation of long non-coding RNA CCAT2 indicates a poor prognosis for prostate cancer and promotes metastasis by affecting epithelial-mesenchymal transition. Biochem Biophys Res Commun. 2016;480(4):508–514.

115.

Prensner JR, Zhao S, Erho N, et al. RNA biomarkers associated with metastatic progression in prostate cancer: a multi-institutional high-throughput analysis of SChLAP1. Lancet Oncol. 2014;15(13):1469–1480.

116.

Mehra R, Shi Y, Udager AM, et al. A novel RNA in situ hybridization assay for the long noncoding RNA SChLAP1 predicts poor clinical outcome after radical prostatectomy in clinically localized prostate cancer. Neoplasia. 2014;16(12):1121–1127.

117.

Wang L, Han S, Jin G, et al. Linc00963: a novel, long non-coding RNA involved in the transition of prostate cancer from androgen-dependence to androgen-independence. Int J Oncol. 2014;44(6):2041–2049.

118.

Alhasan AH, Scott AW, Wu JJ, Jj W, et al. Circulating microRNA signature for the diagnosis of very high-risk prostate cancer. Proc Natl Acad Sci U S A. 2016;113(38):10655–10660.

119.

Kelly BD, Miller N, Sweeney KJ, et al. A circulating microRNA signature as a biomarker for prostate cancer in a high risk group. J Clin Med. 2015;4(7):1369–1379.

120.

van Rooij E, Kauppinen S. Development of microRNA therapeutics is coming of age. EMBO Mol Med. 2014;6(7):851–864.

121.

Wen D, Danquah M, Chaudhary AK, Mahato RI. Small molecules targeting microRNA for cancer therapy: promises and obstacles. J Control Release. 2015;219:237–247.

122.

Mouraviev V, Lee B, Patel V, et al. Clinical prospects of long noncoding RNAs as novel biomarkers and therapeutic targets in prostate cancer. Prostate Cancer Prostatic Dis. 2016;19(1):14–20.

123.

Alvarez-Erviti L, Seow Y, Yin H, Betts C, Lakhal S, Wood MJ. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat Biotechnol. 2011;29(4):341–345.

124.

Zhang D, Lee H, Zhu Z, Minhas JK, Jin Y. Enrichment of selective miRNAs in exosomes and delivery of exosomal miRNAs in vitro and in vivo. Am J Physiol Lung Cell Mol Physiol. 2017;312(1):L110–L121.

125.

Ohno S, Takanashi M, Sudo K, et al. Systemically injected exosomes targeted to EGFR deliver antitumor microRNA to breast cancer cells. Mol Ther. 2013;21(1):185–191.

Creative Commons License © 2018 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.