Minireviews Open Access
Copyright ©The Author(s) 2021. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Oncol. Aug 15, 2021; 13(8): 867-878
Published online Aug 15, 2021. doi: 10.4251/wjgo.v13.i8.867
Role of exosomal long non-coding RNAs in colorectal cancer
Ru Sun, Department of Blood Transfusion, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
Xiao-Yun He, Chun-Lin Ou, Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
Cheng Mei, Department of Blood Transfusion, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
ORCID number: Ru Sun (0000-0001-6483-2779); Xiao-Yun He (0000-0003-0620-420X); Cheng Mei (0000-0002-7660-7306); Chun-Lin Ou (0000-0003-2313-4186).
Author contributions: Ou CL designed the structure of the manuscript; Sun R, He XY, Mei C, and Ou CL drafted the manuscript; Sun R and Ou CL reviewed the literature; Sun R, Mei C, and Ou CL critically revised the manuscript.
Supported by National Natural Science Foundation of China, No. 81903032; China Postdoctoral Science Foundation, No. 2020M672520; Research Program of Hunan Health Commission, China, No. 202103030659; and Youth Fund of Xiangya Hospital, No. 2018Q011.
Conflict-of-interest statement: The authors declare no conflicts of interest for this manuscript.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Chun-Lin Ou, PhD, Academic Research, Department of Pathology, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha 410008, Hunan Province, China. ouchunlin@csu.edu.cn
Received: March 2, 2021
Peer-review started: March 2, 2021
First decision: May 3, 2021
Revised: June 11, 2021
Accepted: July 6, 2021
Article in press: July 6, 2021
Published online: August 15, 2021
Processing time: 164 Days and 19.2 Hours

Abstract

Exosomes are a class of small extracellular vesicles, 30-150 nm in diameter, that transfer biological information (e.g., DNA, RNA, and protein) via cell-to-cell communication. Exosomes play critical roles in the occurrence and development of human cancers, including colorectal cancer (CRC). Recent studies have shown that long non-coding RNAs (lncRNAs) can be encapsulated in exosomes, which transfer lncRNAs from secretory cells into recipient cells. This process affects the progression of CRC, since exosomal lncRNAs display special and extensive functions in CRC tumorigenesis, including malignant proliferation, metastasis, chemoresistance, and inflammatory response. Moreover, due to their specificity and sensitivity, exosomal lncRNAs are released into body fluids (e.g., urine, sputum, and plasma), which have the potential to be biomarkers of CRC tumorigenesis within screening efforts and medical and epidemiologic research. In this review, we aim to clarify the function and mechanism of exosomal lncRNAs in CRC tumorigenesis and provide a strategy for early diagnosis and medical treatment of this malignancy.

Key Words: Exosomes, Long non-coding RNAs, Colorectal cancer, Chemoresistance, Inflammatory response, Therapy

Core Tip: Recent studies have shown that exosomal long non-coding RNAs (lncRNAs) play critical roles in the occurrence and development of colorectal cancer (CRC). Exosomal lncRNAs display special and extensive functions in CRC tumorigenesis, including malignant proliferation, metastasis, chemoresistance, and inflammatory response. Moreover, due to their specificity and sensitivity, exosomal lncRNAs are released into body fluids, which have the potential to be biomarkers of CRC tumorigenesis within screening efforts and medical and epidemiologic research. In this review, we aim to clarify the function and mechanism of exosomal lncRNAs in CRC tumorigenesis and provide a strategy for early diagnosis and treatment of this malignancy.



INTRODUCTION

Colorectal cancer (CRC) is one of the most common gastrointestinal cancers and is currently the fourth most common malignant cancer worldwide in terms of incidence. CRC has an extremely high morbidity and mortality, with almost 1.8 million newly diagnosed cases and nearly 900000 deaths worldwide each year[1-3]. With the rapid progress of cancer therapeutics, the survival time of patients with early-stage CRC has been significantly prolonged. However, the 5-year survival rate for patients with advanced CRC remains low[4-6]. Therefore, exploring the molecular mechanisms of CRC tumorigenesis and identifying early and intermediate stage molecular markers are key processes in the screening and treatment of CRC.

In 1983, Harding et al[7] and Pan and Johnstone[8] were the first to identify and define exosomes, which were considered metabolic waste of cells. With the popularization of electron microscopy, researchers discovered that exosomes, which are 40-160 nm in diameter, are produced by almost all cell types and are released freely in all body fluids (e.g., urine, saliva, sputum, and plasma). Exosomes can transfer RNA, DNA, proteins, lipids, and metabolites via cell-to-cell communication, transferring these molecules from secretory cells to recipient cells and thereby inducing tumors and a range of other human diseases. In recent years, exosomes have become a potentially effective method for disease diagnosis and treatment because of their extensive presence in the body and convenience of acquisition, which may provide a promising prospect in the development of precision medicine[9-11].

With the completion of the Human Genome Project and the arrival of the post-genome era, noncoding RNAs (ncRNAs) have become a hot “star” in the era of life science[12-15]. Long ncRNAs (lncRNAs) are a subset of ncRNAs, which have a transcribing length of 200-100000 nt, lack a complete functional open reading frame, and rarely encode a functional short peptide[16-19]. Recent studies have shown that lncRNAs play an important role in human pathophysiological processes, including tissue differentiation, immunity, and reproduction, and thereby induce a range of human diseases, including diabetes mellitus (DM), Parkinson’s disease, and tumors[20-23].

An increasing number of studies have shown that lncRNAs can be encapsulated into exosomes, which transfer the lncRNAs from secretory cells into recipient cells, thereby affecting the progression of CRC[24-27]. These exosomal lncRNAs can enter receptor cells through humoral circulation and participate in multiple phenotypes of tumor progression, including malignant proliferation, metastasis, chemoresistance, and inflammatory response. In addition, due to their specificity and sensitivity, exosomal-derived lncRNAs released into the tumor microenvironment have the potential to become biomarkers of CRC. Therefore, this review aims to provide a new direction for finding new CRC biomarkers and therapeutic targets by pooling and summarizing related studies of the role of exosomal lncRNAs in CRC tumorigenesis.

BIOLOGICAL CHARACTERISTICS AND MECHANISMS OF EXOSOMAL LNCRNAS

According to their size, extracellular vesicles (EVs) can be divided into small EVs (sEVs; with a diameter less than 200 nm) and large EVs (lEVs; with a diameter more than 200 nm). Exosomes are a class of sEVs, with a diameter of approximately 40-160 nm, derived from almost all types of human cells. After being released by secretory cells, exosomes are widely distributed to all types of human body fluids, including saliva, breast milk, cerebrospinal fluid, ascites, urine, and semen[28-30]. Exosomes enter target cells through humoral circulation and cell receptors in three ways: Direct fusion, endocytosis, and receptor ligand binding. Exosomes carry proteins, nucleic acids, lipids, and other important signal carriers, forming a cell-cell information transmission system that participates in cell communication, cell migration, angiogenesis, and tumor cell growth[31-33].

The biological function of lncRNAs depends on their subcellular localizations. When lncRNAs are mainly localized in the nucleus, they play a role in regulating epigenetic modification, which regulates the expression of downstream target genes through histone modification by binding the enhancer region of the gene. Cytoplasmic lncRNAs, in contrast, not only sponge microRNAs (miRNAs) to form competing endogenous RNAs (ceRNAs) to regulate the expression of miRNA target genes, but can also block the phosphorylation level of targeted proteins, thereby affecting the protein expression of molecules and downstream signaling pathways[34-36]. Therefore, when lncRNAs are encapsulated into exosomes, they transfer from secretory cells into recipient cells to exert their biological function, thereby affecting the progression of tumors. After entering the recipient cells, lncRNAs regulate the target gene expression of miRNAs through sponge miRNAs and affect the phosphorylation level or transcription expression of proteins by binding to proteins after entry into the target cells. Exosomal lncRNAs further regulate core molecules and signaling pathways, leading to tumor progression (Figure 1). In CRC, numerous studies have shown that exosomal lncRNAs, after entry into the target cells, not only regulate the target gene expression of miRNAs through sponge miRNAs[37-43] (Table 1), but also affect the phosphorylation level or transcription expression of proteins by binding to proteins[44-49] (Table 2). Therefore, a systematic understanding of the role of exosomal lncRNAs in cancer metastasis may provide improved diagnostic and prognostic biomarkers and therapeutic targets for malignant tumors.

Figure 1
Figure 1 The process of exosomal long non-coding RNAs secretion, transportation, and ingestion. A: Exosomal long non-coding RNAs (lncRNAs) are secreted by colorectal cancer (CRC) cells, carcinoma-associated fibroblasts, and chemoresistant CRC cells; B: Exosomal lncRNAs are released into circulatory system; C: Exosomal lncRNAs are transferred from the secretory cell into recipient cell to exert their biological function, thereby affecting the progression of tumors. CRC: Colorectal cancer; CAF: Carcinoma-associated fibroblast.
Table 1 Relationship between exosomal long non-coding RNAs and microRNAs in colorectal cancer.
Exosomal lncRNA
Expression
miRNA
Target gene
Functions
Ref.
H19miR-141β-cateninPromotes the stemness and chemoresistance of CRC cellsRen et al[37]
UCA1miR-143MYO6Promotes CRC cell proliferation and migrationLuan et al[38]
LINC00659miR-342-3pANXA2Promotes CRC cell proliferation, invasion, and migrationZhou et al[39]
MALAT1miR-26a/26bFUT4Promotes the metastasis of CRCXu et al[40]
HOTTIPmiR-214KPNA3Increases resistance of CRC cells to mitomycinChen et al[41]
LINC02418miR-1273g-3pMELKPromotes the viability of CRC cellsZhao et al[42]
GAS5miR-221-Promotes CRC cell proliferationLiu et al[43]
Table 2 Relationship between exosomal long non-coding RNAs and their binding molecules in colorectal cancer.
Exosomal lncRNA
Expression
Interaction gene/protein
Signaling pathway
Functions
Ref.
RPPH1TUBB3-Promotes metastasis and proliferation of CRC cellsLiang et al[44]
APC1 Rab5bMAPK pathwayInhibits metastasis, and angiogenesis of CRCWang et al[45]
CCALHuRβ-catenin pathwaySuppresses cell apoptosis and promotes chemoresistance of CRC cellsDeng et al[46]
ADAMTS9-AS1β-cateninWnt signaling pathwaySuppresses the CRC tumorigenesisLi et al[47]
91HHNRNPK-Promotes migration and invasion of CRC cellsGao et al[48]
CRNDE-hRORγt-Promotes Th17 cell differentiationSun et al[49]
EXOSOMAL LNCRNAS AND CRC

Hanahan and Weinberg[50] published a landmark review entitled “Hallmarks of cancer: the next generation” in the journal Cell, which summarized and clarified the ten defining characteristics of tumors, including self-sufficiency in growth signals, sustained angiogenesis, insensitivity to anti-growth signals, resisting cell death, limitless replicative potential, tissue invasion and metastasis, avoiding immune destruction, tumor promotion through inflammation, genome instability and mutation, and deregulation of cellular energetics. Recent studies have shown that exosomal lncRNA levels are closely related to the occurrence and development of tumors and that exosomal lncRNAs are involved in malignant proliferation, invasion and metastasis, chemoresistance, and inflammatory response (Figure 2).

Figure 2
Figure 2 Summary chart of exosomal long non-coding RNAs in colorectal cancer. Exosomal long non-coding RNAs are closely related to the occurrence and development of tumors, involved in malignant proliferation, invasion and metastasis, chemoresistance, and the inflammatory response. Orange arrows: High expression in colorectal cancer; green arrows: Low expression in colorectal cancer. LncRNA: Long non-coding RNA; CRC: Colorectal cancer; HuR: Human antigen R.
Exosomal lncRNAs and malignant proliferation of CRC cells

Malignant proliferation of tumor cells is the most common malignant phenotype in the development of tumors. Tumor cells usually lack contact inhibition and show enhanced proliferation. Thus, tumors are classified as a type of progressive hyperplastic disease[51,52]. Although starting from the direction of cutting off the malignant proliferation of tumors is a good choice for cancer treatment, the current basic research and clinical results are still unsatisfactory. Identifying key effective and broad-spectrum targets for inhibiting malignant tumor proliferation remains a difficult problem for scientists[53-55].

In recent years, exosomal lncRNAs have attracted wide attention as targets for the malignant proliferation of CRC. Exosomal lncRNA-UCA1 can be transmitted into CRC cells, thereby increasing the expression of myosin VI (MYO6) by sponging with miR-143, thus promoting CRC cell proliferation[38]. Moreover, Zhao et al[42] demonstrated that circulating lncRNA-LINC02418 can be transmitted into CRC cells via exosomes, and lncRNA-LINC02418 can function as a ceRNA to regulate the expression of maternal embryonic leucine zipper kinase (MELK) by competing for miR-1273g-3p binding. Li et al[47] revealed that exosomal lncRNA-ADAMTS9-AS1 can promote the malignant proliferation of CRC cells by increasing the expression of β-catenin and activating the Wnt signaling pathway. Interestingly, exosomal lncRNA-APC1 was also found to be able to inhibit CRC cell proliferation by activating the MAPK pathway through directly binding to Ras-related protein rab-5b (Rab5b) mRNA and hence reducing its stability[45]. These findings show that exosomal lncRNAs play a key regulatory role in CRC cells; hence, lncRNAs may provide a new target for CRC therapy.

Exosomal lncRNAs and CRC metastasis

The term tumor metastasis usually refers to the process by which malignant tumor cells detach from the primary tumor site and are transferred through the circulatory system to secondary tissues or organs, where they colonize and form secondary tumors. Tumor metastasis is a complex process that involves cytoskeletal reconstruction, decreased cell adhesion, extracellular matrix degradation, and so on, thereby inducing angiogenesis and the epithelial-mesenchymal transition (EMT) of tumors[56]. Tumor metastasis is a major problem with regard to cancer therapy because it affects the prognosis and survivorship of tumor patients and represents the leading cause of tumor-related deaths[57]. Therefore, it is of great significance to study the mechanisms of tumor metastasis for the sake of improved treatment and prevention of tumors.

Exosomal lncRNAs are a class of molecules that, in recent years, have been shown to play an important role in the CRC metastasis. A study[40] demonstrated that exosomal lncRNA-MALAT1 derived from highly metastatic CRC cells can enhance the metastatic abilities of primary CRC cells. The underlying mechanism is that exosomal lncRNA-MALAT absorbs miR-26a/26b, hence increasing the expression of fucosyltransferase 4 (FUT4) and activating the PI3K/AKT/mTOR pathway. Meanwhile, exosomal lncRNA 91H was also found to be able to directly interact with heterogeneous ribosomal protein K (HNRNPK) in CRC cells, thereby positively regulating the expression of HNRNPK to enhance CRC metastasis[48]. Furthermore, exosomal lncRNAs are closely related to EMT in CRC cells. Zhou et al[39] found that cancer-associated fibroblast-derived exosomal LINC00659 can promote the progression of EMT in CRC cells via the miR-342-3p/ANXA2 axis. These studies indicate that exosomal lncRNAs might be potential biomarkers and therapeutic targets for the prediction, screening, and treatment of CRC.

Exosomal lncRNAs and CRC chemoresistance

The chemoresistance of tumors is caused by interaction between internal and external factors. Internal factors include protective autophagy, EMT, oxidative stress, and metabolic reprogramming. External factors include hypoxia and the tumor microenvironment. Corresponding molecules and the signaling pathways of cancer cells are changed through both internal and external factors, reducing the drug sensitivity of cancer cells and ultimately inducing the chemoresistance of cancer cells[58-60]. The mechanism of chemoresistance has not been fully elucidated. Thus, chemoresistance is still one of the key reasons for the failure of tumor treatment.

Recent studies have demonstrated the important role of exosomal lncRNAs in CRC chemoresistance. The chemotherapy drugs for CRC contain cetuximab, mitomycin, and oxaliplatin. Yang et al[61] reported that cetuximab-resistant CRC cells secrete the exosomal lncRNA-UCA1, which can transmit cetuximab resistance to sensitive cells, and that the expression of exosomal lncRNA-UCA1 is closely related to the clinical outcome of cetuximab therapy in CRC patients. Interestingly, Chen et al[41] revealed that lncRNA-HOTTIP is highly expressed in mitomycin-resistant CRC cells and can be encapsulated into exosomes, transferring lncRNAs from mitomycin-resistant cells to sensitive cells; after entering the sensitive cells, lncRNA-HOTTIP can upregulate the expression of karyopherin subunit alpha 3 (KPNA3) by binding to miR-214, thereby promoting drug resistance in sensitive cells. Moreover, Deng et al[46] found that carcinoma-associated fibroblasts (CAFs) can secrete exosomal lncRNA-CCAL to promote oxaliplatin resistance in CRC cells. Functional studies revealed that lncRNA-CCAL can interact directly with the mRNA stabilizing protein HuR (human antigen R) to increase the expression of β-catenin, thereby inducing chemoresistance in CRC. Targeting exosomal lncRNAs might thus be a promising strategy for overcoming drug resistance in CRC.

Exosomal lncRNAs and inflammatory response in CRC

The connection of inflammation with tumorigenesis and tumor promotion, progression, and metastasis has been a major concern in recent years. Studies have shown that inflammation can accelerate the inflammation-tumor transformational reaction chain, thereby increasing the risk of the occurrence of tumors. Therefore, this type of tumor is referred to as an inflammation-associated tumor. Inflammation is an important biological risk factor for malignancy, and is considered the seventh defining characteristic of tumors[62-64]. Emerging evidence suggests that exosomal lncRNAs participate in inflammatory tumor response, which is closely related to the tumor microenvironment, comprised of immune cells, inflammatory factors, chemokines, etc.[65,66].

The association between inflammatory bowel disease (IBD) and CRC has long been recognized[64]. Colitis-associated cancer (CAC) is a CRC subtype that is associated with IBD[67]. By constructing an AOM/DSS-induced CAC mouse model, Ren et al[37] found that CAF-derived exosomal H19 can act as a ceRNA to increase the expression of β-catenin via sponging with miR-141, thereby contributing to the stemness and development of CRC.

Furthermore, proinflammatory conditions can be reflected in the tumor microenvironment, which contains a series of immune cells, such as dendritic cells, natural killer cells, macrophages, and T helper (Th) cells[68,69]. Recent studies have shown that exosomal lncRNAs play a crucial role in the differentiation of CRC immune cells. For example, Sun et al[49] demonstrated that serum exosomal lncRNA-CRNDE-h can be transmitted into CD4+ T cells to increase the Th17 cell proportion and promote Th17 cell differentiation by inhibiting the E3 ubiquitin ligase Itch-mediated ubiquitination and degradation of RAR-related orphan receptor γt (RORγt) in CRC. Moreover, Liang et al[44] found that CRC cells-derived exosomal lncRNA-RPPH1 can be transmitted into tumor-associated macrophages to mediate macrophage M2 polarization, which is associated with CRC inflammatory response. Taken together, exosomal lncRNAs may serve as a promising target for tumor immune therapy.

EXOSOMAL LNCRNAS AS NOVEL CRC BIOMARKERS AND TARGETS

Exosomal miRNAs have a higher specificity compared to proteins and are easier to extract and detect. Thus, testing exosomal miRNAs by quantitative reverse transcription-polymerase chain reaction and in situ hybridization assays is more specific and sensitive than detecting proteins by an antigen-antibody reaction[70,71]. Exosomal lncRNAs can be detected in body fluids, and the content of lncRNAs can provide significant information about physiological and/or pathological changes in tumor patients[72,73]. Therefore, exosomal lncRNAs have been widely studied as CRC markers in recent years[37,38,40,42,44,45,47,48,74-78] (Table 3).

Table 3 Exosomal long non-coding RNAs as novel biomarkers and therapeutic targets for colorectal cancer.
Exosomal lncRNA
Expression
Relationship with clinicopathologic features
AUC
Ref.
APC1Lymph node and distant metastasis-Wang et al[45]
GAS5TNM stage, Dukes stage, lymph node metastasis, local recurrence, and distant metastasis0.791Xu et al[40]
LINC02418-0.898Zhao et al[42]
SPINT1-AS1Lymph node metastasis, distant metastasis, and poor RFS0.865Li et al[74]
ADAMTS9-AS1-0.835Li et al[47]
CCAT2TNM stages and lymph node metastasis-Wang et al[75]
UCA1Tumor size, tumor stage, and metastasis status-Luan et al[38]
CRNDE-hLymph node metastasis and distant metastasis0.892Liu et al[76]
CRNDE-pAdvanced clinical stages, tumor classification, and lymph node or distant metastasis0.854Yu et al[77]
HOTTIPLymph node metastasis and better OS0.750Oehme et al[78]
91HTNM stage-Gao et al[48]
RPPH1TNM stage, metastasis, and poor OS and DFS0.856Liang et al[44]
H19TNM stage-Ren et al[37]

Many clinical studies have indicated a close association between exosomal lncRNAs and various clinical symptoms. Li et al[74] found that elevated expression of exosomal lncRNA-SPINT1-AS1 was associated with regional lymph node metastasis, distant metastasis, and short recurrence-free survival of CRC patients. Liu et al[43] showed that elevated expression of exosomal lncRNA-GAS5 was correlated with TNM stage, Dukes stage, local recurrence rate, and distant metastasis in CRC. In addition, elevated expression of CCAT2 was associated with local invasion and lymph node metastasis in CRC[79].

Exosomal lncRNAs serve as novel potential diagnostic and prognostic biomarkers of CRC. Using exosomal lncRNA-CRNDE-h as a diagnostic biomarker[76], receiver operating characteristic (ROC) curve analysis showed that lncRNA-CRNDE-h expression was a good candidate for distinguishing CRC patients from healthy control participants providing serum samples (sensitivity 70.3%, specificity 94.4%). The area under the ROC curve was 0.892 [95% confidence interval (CI): 0.860-0.918, P < 0.05]. Moreover, exosomal miRNAs act as prognostic biomarkers, and Oehme et al[78] found that low expression of exosomal lncRNA-HOTTIP was positively correlated with a poor overall survival (OS) in CRC patients (P = 0.0009), and further found that low expression of lncRNA-HOTTIP was an independent prognostic marker for OS (hazard ratio: 4.5, 95%CI: 1.69-11.98, P = 0.0027) within a multivariate analysis. With the widespread use and development of new technologies, the detection of exosomal lncRNAs may provide a novel strategy for the screening, early diagnosis, and therapy of CRC.

FUTURE PERSPECTIVES

To summarize, an increasing number of studies have shown that exosomal lncRNAs are closely associated with the progression of CRC. Exosomal lncRNAs have been recognized as ‘new stars’ in the research field of tumors, and, as a class of novel regulatory molecules, they participate in the progression of CRC. Therefore, we can silence or activate exosomal lncRNAs in CRC patients via an exogenous means. For example, we can wrap the silenced or active lentiviral vector of lncRNAs into exosomes in vitro, then perform a targeted injection into the corresponding organs of humans through fluid circulation. This can be developed as a therapy for CRC. With the development of high-throughput sequencing and related technologies in recent decades[80], an increasing number of exosomal lncRNAs have been discovered and identified in human diseases. Because the extraction and detection of exosomal lncRNAs present a higher specificity and sensitivity compared to exosomal proteins, lncRNAs have great potential as biological tools for the diagnosis or treatment of tumors[81]. However, there is a long road from scientific research on exosomal lncRNAs to clinical applications. Recently, research on exosomal lncRNAs has faced a series of challenges and limitations: (1) Although high-throughput sequencing has shown that many exosomal lncRNAs are abnormally expressed in tumor body fluids (e.g., urine, sputum, and plasma), the specific mechanisms and functions of exosomal lncRNAs have not been fully understood; (2) The technology and methods for isolation and purification of exosomes, including ultra-high-speed centrifugation, filtration, precipitation, and immunoconcentration, are not mature[82,83]. Existing purification methods can hardly distinguish exosomes from non-vesicle components, which may affect the subsequent functional experiments of exosomal lncRNAs in vivo and in vitro; (3) Although exosomal lncRNAs have been shown to play a role in tumor treatment in many animal models, there is still a lack of clinical trials to confirm the accuracy and safety of these findings; and (4) There are still some difficulties in developing exosome-based drug delivery systems and functionally introducing them to specific cells.

Although our existing understanding of exosomal lncRNAs is just the tip of the iceberg, novel approaches and techniques will ultimately shed light on these processes. It is likely that in the near future, detection kits and therapeutic drugs targeting exosomal lncRNAs will be used in clinical research for CRC screening, diagnosis, and treatment.

CONCLUSION

Recent studies have shown that exosomal lncRNAs play critical roles in the occurrence and development of CRC. Exosomal lncRNAs display special and extensive functions in CRC tumorigenesis, including malignant proliferation, metastasis, chemoresistance, and inflammatory response. Moreover, due to their specificity and sensitivity, exosomal lncRNAs are released into body fluids, which have the potential to be biomarkers of CRC tumorigenesis within screening efforts and medical and epidemiologic research. In this review, we aim to clarify the function and mechanism of exosomal lncRNAs in CRC tumorigenesis and provide a strategy for early diagnosis and treatment.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Oncology

Country/Territory of origin: China

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B, B

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: An Q, Ros J S-Editor: Gao CC L-Editor: Wang TQ P-Editor: Yuan YY

References
1.  Siegel RL, Miller KD, Fedewa SA, Ahnen DJ, Meester RGS, Barzi A, Jemal A. Colorectal cancer statistics, 2017. CA Cancer J Clin. 2017;67:177-193.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2526]  [Cited by in F6Publishing: 2784]  [Article Influence: 397.7]  [Reference Citation Analysis (2)]
2.  Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer Statistics, 2021. CA Cancer J Clin. 2021;71:7-33.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8287]  [Cited by in F6Publishing: 10426]  [Article Influence: 3475.3]  [Reference Citation Analysis (2)]
3.  Tejpar S, Shen L, Wang X, Schilsky RL. Integrating biomarkers in colorectal cancer trials in the West and China. Nat Rev Clin Oncol. 2015;12:553-560.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 10]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
4.  Walsh JM, Terdiman JP. Colorectal cancer screening: scientific review. JAMA. 2003;289:1288-1296.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 286]  [Cited by in F6Publishing: 309]  [Article Influence: 14.7]  [Reference Citation Analysis (0)]
5.  Ou C, Sun Z, Li S, Li G, Li X, Ma J. Dual roles of yes-associated protein (YAP) in colorectal cancer. Oncotarget. 2017;8:75727-75741.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 40]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
6.  Wang Y, He X, Nie H, Zhou J, Cao P, Ou C. Application of artificial intelligence to the diagnosis and therapy of colorectal cancer. Am J Cancer Res. 2020;10:3575-3598.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 46]  [Article Influence: 11.5]  [Reference Citation Analysis (1)]
7.  Harding C, Heuser J, Stahl P. Receptor-mediated endocytosis of transferrin and recycling of the transferrin receptor in rat reticulocytes. J Cell Biol. 1983;97:329-339.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1101]  [Cited by in F6Publishing: 1207]  [Article Influence: 29.4]  [Reference Citation Analysis (0)]
8.  Pan BT, Johnstone RM. Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: selective externalization of the receptor. Cell. 1983;33:967-978.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1188]  [Cited by in F6Publishing: 1330]  [Article Influence: 32.4]  [Reference Citation Analysis (0)]
9.  Tyner JW. Functional genomics for personalized cancer therapy. Sci Transl Med. 2014;6:243fs26.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 13]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
10.  Zhou R, Chen KK, Zhang J, Xiao B, Huang Z, Ju C, Sun J, Zhang F, Lv XB, Huang G. The decade of exosomal long RNA species: an emerging cancer antagonist. Mol Cancer. 2018;17:75.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 103]  [Article Influence: 17.2]  [Reference Citation Analysis (0)]
11.  Sun Z, Shi K, Yang S, Liu J, Zhou Q, Wang G, Song J, Li Z, Zhang Z, Yuan W. Effect of exosomal miRNA on cancer biology and clinical applications. Mol Cancer. 2018;17:147.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 311]  [Cited by in F6Publishing: 480]  [Article Influence: 80.0]  [Reference Citation Analysis (0)]
12.  Wang Y, Nie H, He X, Liao Z, Zhou Y, Zhou J, Ou C. The emerging role of super enhancer-derived noncoding RNAs in human cancer. Theranostics. 2020;10:11049-11062.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 46]  [Article Influence: 11.5]  [Reference Citation Analysis (1)]
13.  Tang Y, Wang J, Lian Y, Fan C, Zhang P, Wu Y, Li X, Xiong F, Li G, Xiong W, Zeng Z. Linking long non-coding RNAs and SWI/SNF complexes to chromatin remodeling in cancer. Mol Cancer. 2017;16:42.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 103]  [Article Influence: 14.7]  [Reference Citation Analysis (0)]
14.  Wang JP, Tang YY, Fan CM, Guo C, Zhou YH, Li Z, Li XL, Li Y, Li GY, Xiong W, Zeng ZY, Xiong F. The role of exosomal non-coding RNAs in cancer metastasis. Oncotarget. 2018;9:12487-12502.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 42]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
15.  Long F, Lin Z, Li L, Ma M, Lu Z, Jing L, Li X, Lin C. Comprehensive landscape and future perspectives of circular RNAs in colorectal cancer. Mol Cancer. 2021;20:26.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 84]  [Article Influence: 28.0]  [Reference Citation Analysis (0)]
16.  Dempsey JL, Cui JY. Long Non-Coding RNAs: A Novel Paradigm for Toxicology. Toxicol Sci. 2017;155:3-21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 72]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
17.  Ou C, Sun Z, He X, Li X, Fan S, Zheng X, Peng Q, Li G, Ma J. Targeting YAP1/LINC00152/FSCN1 Signaling Axis Prevents the Progression of Colorectal Cancer. Adv Sci (Weinh). 2020;7:1901380.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 103]  [Article Influence: 25.8]  [Reference Citation Analysis (0)]
18.  Tang Y, He Y, Zhang P, Wang J, Fan C, Yang L, Xiong F, Zhang S, Gong Z, Nie S, Liao Q, Li X, Li Y, Li G, Zeng Z, Xiong W, Guo C. LncRNAs regulate the cytoskeleton and related Rho/ROCK signaling in cancer metastasis. Mol Cancer. 2018;17:77.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 123]  [Article Influence: 20.5]  [Reference Citation Analysis (0)]
19.  MacDonald WA, Mann MRW. Long noncoding RNA functionality in imprinted domain regulation. PLoS Genet. 2020;16:e1008930.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 39]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
20.  Fan C, Tang Y, Wang J, Xiong F, Guo C, Wang Y, Zhang S, Gong Z, Wei F, Yang L, He Y, Zhou M, Li X, Li G, Xiong W, Zeng Z. Role of long non-coding RNAs in glucose metabolism in cancer. Mol Cancer. 2017;16:130.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 128]  [Cited by in F6Publishing: 147]  [Article Influence: 21.0]  [Reference Citation Analysis (0)]
21.  Sun Z, Liu J, Chen C, Zhou Q, Yang S, Wang G, Song J, Li Z, Zhang Z, Xu J, Sun X, Chang Y, Yuan W. The Biological Effect and Clinical Application of Long Noncoding RNAs in Colorectal Cancer. Cell Physiol Biochem. 2018;46:431-441.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 25]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
22.  Kung JT, Colognori D, Lee JT. Long noncoding RNAs: past, present, and future. Genetics. 2013;193:651-669.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1443]  [Cited by in F6Publishing: 1400]  [Article Influence: 127.3]  [Reference Citation Analysis (0)]
23.  Cheng J, Meng J, Zhu L, Peng Y. Exosomal noncoding RNAs in Glioma: biological functions and potential clinical applications. Mol Cancer. 2020;19:66.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 164]  [Cited by in F6Publishing: 206]  [Article Influence: 51.5]  [Reference Citation Analysis (0)]
24.  Galamb O, Barták BK, Kalmár A, Nagy ZB, Szigeti KA, Tulassay Z, Igaz P, Molnár B. Diagnostic and prognostic potential of tissue and circulating long non-coding RNAs in colorectal tumors. World J Gastroenterol. 2019;25:5026-5048.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 68]  [Cited by in F6Publishing: 73]  [Article Influence: 14.6]  [Reference Citation Analysis (0)]
25.  Francavilla A, Turoczi S, Tarallo S, Vodicka P, Pardini B, Naccarati A. Exosomal microRNAs and other non-coding RNAs as colorectal cancer biomarkers: a review. Mutagenesis. 2020;35:243-260.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 25]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
26.  Camacho CV, Choudhari R, Gadad SS. Long noncoding RNAs and cancer, an overview. Steroids. 2018;133:93-95.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 61]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
27.  Cheshomi H, Matin MM. Exosomes and their importance in metastasis, diagnosis, and therapy of colorectal cancer. J Cell Biochem. 2018;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 39]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
28.  Zhang HG, Grizzle WE. Exosomes: a novel pathway of local and distant intercellular communication that facilitates the growth and metastasis of neoplastic lesions. Am J Pathol. 2014;184:28-41.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 224]  [Cited by in F6Publishing: 270]  [Article Influence: 24.5]  [Reference Citation Analysis (0)]
29.  Dai J, Su Y, Zhong S, Cong L, Liu B, Yang J, Tao Y, He Z, Chen C, Jiang Y. Exosomes: key players in cancer and potential therapeutic strategy. Signal Transduct Target Ther. 2020;5:145.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 256]  [Cited by in F6Publishing: 533]  [Article Influence: 133.3]  [Reference Citation Analysis (0)]
30.  Hashemipour M, Boroumand H, Mollazadeh S, Tajiknia V, Nourollahzadeh Z, Rohani Borj M, Pourghadamyari H, Rahimian N, Hamblin MR, Mirzaei H. Exosomal microRNAs and exosomal long non-coding RNAs in gynecologic cancers. Gynecol Oncol. 2021;161:314-327.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 52]  [Article Influence: 17.3]  [Reference Citation Analysis (0)]
31.  Vlassov AV, Magdaleno S, Setterquist R, Conrad R. Exosomes: current knowledge of their composition, biological functions, and diagnostic and therapeutic potentials. Biochim Biophys Acta. 2012;1820:940-948.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1267]  [Cited by in F6Publishing: 1398]  [Article Influence: 116.5]  [Reference Citation Analysis (0)]
32.  Xu K, Zhang C, Du T, Gabriel ANA, Wang X, Li X, Sun L, Wang N, Jiang X, Zhang Y. Progress of exosomes in the diagnosis and treatment of lung cancer. Biomed Pharmacother. 2021;134:111111.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 80]  [Article Influence: 20.0]  [Reference Citation Analysis (0)]
33.  Choi JU, Park IK, Lee YK, Hwang SR. The Biological Function and Therapeutic Potential of Exosomes in Cancer: Exosomes as Efficient Nanocommunicators for Cancer Therapy. Int J Mol Sci. 2020;21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
34.  Klinge CM. Non-Coding RNAs in Breast Cancer: Intracellular and Intercellular Communication. Noncoding RNA. 2018;4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 94]  [Article Influence: 15.7]  [Reference Citation Analysis (0)]
35.  Bhan A, Soleimani M, Mandal SS. Long Noncoding RNA and Cancer: A New Paradigm. Cancer Res. 2017;77:3965-3981.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1358]  [Cited by in F6Publishing: 1918]  [Article Influence: 274.0]  [Reference Citation Analysis (0)]
36.  He X, Yu B, Kuang G, Wu Y, Zhang M, Cao P, Ou C. Long noncoding RNA DLEU2 affects the proliferative and invasive ability of colorectal cancer cells. J Cancer. 2021;12:428-437.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 17]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
37.  Ren J, Ding L, Zhang D, Shi G, Xu Q, Shen S, Wang Y, Wang T, Hou Y. Carcinoma-associated fibroblasts promote the stemness and chemoresistance of colorectal cancer by transferring exosomal lncRNA H19. Theranostics. 2018;8:3932-3948.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 296]  [Cited by in F6Publishing: 486]  [Article Influence: 81.0]  [Reference Citation Analysis (0)]
38.  Luan Y, Li X, Luan Y, Zhao R, Li Y, Liu L, Hao Y, Oleg Vladimir B, Jia L. Circulating lncRNA UCA1 Promotes Malignancy of Colorectal Cancer via the miR-143/MYO6 Axis. Mol Ther Nucleic Acids. 2020;19:790-803.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 89]  [Article Influence: 17.8]  [Reference Citation Analysis (0)]
39.  Zhou L, Li J, Tang Y, Yang M. Exosomal LncRNA LINC00659 transferred from cancer-associated fibroblasts promotes colorectal cancer cell progression via miR-342-3p/ANXA2 axis. J Transl Med. 2021;19:8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 71]  [Article Influence: 23.7]  [Reference Citation Analysis (0)]
40.  Xu J, Xiao Y, Liu B, Pan S, Liu Q, Shan Y, Li S, Qi Y, Huang Y, Jia L. Exosomal MALAT1 sponges miR-26a/26b to promote the invasion and metastasis of colorectal cancer via FUT4 enhanced fucosylation and PI3K/Akt pathway. J Exp Clin Cancer Res. 2020;39:54.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 95]  [Article Influence: 23.8]  [Reference Citation Analysis (0)]
41.  Chen X, Liu Y, Zhang Q, Liu B, Cheng Y, Zhang Y, Sun Y, Liu J, Gen H. Exosomal Long Non-coding RNA HOTTIP Increases Resistance of Colorectal Cancer Cells to Mitomycin via Impairing MiR-214-Mediated Degradation of KPNA3. Front Cell Dev Biol. 2020;8:582723.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 16]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
42.  Zhao Y, Du T, Du L, Li P, Li J, Duan W, Wang Y, Wang C. Long noncoding RNA LINC02418 regulates MELK expression by acting as a ceRNA and may serve as a diagnostic marker for colorectal cancer. Cell Death Dis. 2019;10:568.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 50]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
43.  Liu L, Meng T, Yang XH, Sayim P, Lei C, Jin B, Ge L, Wang HJ. Prognostic and predictive value of long non-coding RNA GAS5 and mircoRNA-221 in colorectal cancer and their effects on colorectal cancer cell proliferation, migration and invasion. Cancer Biomark. 2018;22:283-299.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 83]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
44.  Liang ZX, Liu HS, Wang FW, Xiong L, Zhou C, Hu T, He XW, Wu XJ, Xie D, Wu XR, Lan P. LncRNA RPPH1 promotes colorectal cancer metastasis by interacting with TUBB3 and by promoting exosomes-mediated macrophage M2 polarization. Cell Death Dis. 2019;10:829.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 221]  [Article Influence: 44.2]  [Reference Citation Analysis (0)]
45.  Wang FW, Cao CH, Han K, Zhao YX, Cai MY, Xiang ZC, Zhang JX, Chen JW, Zhong LP, Huang Y, Zhou SF, Jin XH, Guan XY, Xu RH, Xie D. APC-activated long noncoding RNA inhibits colorectal carcinoma pathogenesis through reduction of exosome production. J Clin Invest. 2021;131.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
46.  Deng X, Ruan H, Zhang X, Xu X, Zhu Y, Peng H, Kong F, Guan M. Long noncoding RNA CCAL transferred from fibroblasts by exosomes promotes chemoresistance of colorectal cancer cells. Int J Cancer. 2020;146:1700-1716.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 138]  [Article Influence: 27.6]  [Reference Citation Analysis (0)]
47.  Li N, Li J, Mi Q, Xie Y, Li P, Wang L, Binang H, Wang Q, Wang Y, Chen Y, Mao H, Du L, Wang C. Long non-coding RNA ADAMTS9-AS1 suppresses colorectal cancer by inhibiting the Wnt/β-catenin signalling pathway and is a potential diagnostic biomarker. J Cell Mol Med. 2020;24:11318-11329.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 45]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
48.  Gao T, Liu X, He B, Nie Z, Zhu C, Zhang P, Wang S. Exosomal lncRNA 91H is associated with poor development in colorectal cancer by modifying HNRNPK expression. Cancer Cell Int. 2018;18:11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 80]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
49.  Sun J, Jia H, Bao X, Wu Y, Zhu T, Li R, Zhao H. Tumor exosome promotes Th17 cell differentiation by transmitting the lncRNA CRNDE-h in colorectal cancer. Cell Death Dis. 2021;12:123.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 47]  [Article Influence: 15.7]  [Reference Citation Analysis (0)]
50.  Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646-674.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39812]  [Cited by in F6Publishing: 43707]  [Article Influence: 3362.1]  [Reference Citation Analysis (4)]
51.  Pietras K, Ostman A. Hallmarks of cancer: interactions with the tumor stroma. Exp Cell Res. 2010;316:1324-1331.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 780]  [Cited by in F6Publishing: 819]  [Article Influence: 58.5]  [Reference Citation Analysis (0)]
52.  Balkwill F, Mantovani A. Cancer and inflammation: implications for pharmacology and therapeutics. Clin Pharmacol Ther. 2010;87:401-406.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 177]  [Cited by in F6Publishing: 186]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
53.  Schwartz GK, Shah MA. Targeting the cell cycle: a new approach to cancer therapy. J Clin Oncol. 2005;23:9408-9421.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 530]  [Cited by in F6Publishing: 551]  [Article Influence: 30.6]  [Reference Citation Analysis (0)]
54.  Ou C, Sun Z, Li X, Ren W, Qin Z, Zhang X, Yuan W, Wang J, Yu W, Zhang S, Peng Q, Yan Q, Xiong W, Li G, Ma J. MiR-590-5p, a density-sensitive microRNA, inhibits tumorigenesis by targeting YAP1 in colorectal cancer. Cancer Lett. 2017;399:53-63.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 88]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
55.  Li J, Tian H, Yang J, Gong Z. Long Noncoding RNAs Regulate Cell Growth, Proliferation, and Apoptosis. DNA Cell Biol. 2016;35:459-470.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 117]  [Article Influence: 14.6]  [Reference Citation Analysis (0)]
56.  Liu C, Wu Y, Ma J. Interaction of non-coding RNAs and Hippo signaling: Implications for tumorigenesis. Cancer Lett. 2020;493:207-216.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 6]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
57.  Mantovani A. Cancer: Inflaming metastasis. Nature. 2009;457:36-37.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 451]  [Cited by in F6Publishing: 471]  [Article Influence: 31.4]  [Reference Citation Analysis (0)]
58.  Shao Y, Li H, Du R, Meng J, Yang G. Involvement of non-coding RNAs in chemotherapy resistance of ovarian cancer. J Cancer. 2018;9:1966-1972.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 7]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
59.  Karagiannis GS, Condeelis JS, Oktay MH. Chemotherapy-induced metastasis: mechanisms and translational opportunities. Clin Exp Metastasis. 2018;35:269-284.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 102]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
60.  Wambecke A, Ahmad M, Lambert B, Joly F, Poulain L, Denoyelle C, Meryet-Figuiere M. The influence of long non-coding RNAs on the response to chemotherapy in ovarian cancer. Gynecol Oncol. 2020;156:726-733.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 9]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
61.  Yang YN, Zhang R, Du JW, Yuan HH, Li YJ, Wei XL, Du XX, Jiang SL, Han Y. Predictive role of UCA1-containing exosomes in cetuximab-resistant colorectal cancer. Cancer Cell Int. 2018;18:164.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 73]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
62.  Zhang X, Ai F, Li X, She X, Li N, Tang A, Qin Z, Ye Q, Tian L, Li G, Shen S, Ma J. Inflammation-induced S100A8 activates Id3 and promotes colorectal tumorigenesis. Int J Cancer. 2015;137:2803-2814.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 39]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
63.  Zhang Q, Wang W, Zhou Q, Chen C, Yuan W, Liu J, Li X, Sun Z. Roles of circRNAs in the tumour microenvironment. Mol Cancer. 2020;19:14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 150]  [Article Influence: 37.5]  [Reference Citation Analysis (0)]
64.  Zhang X, Wei L, Wang J, Qin Z, Lu Y, Zheng X, Peng Q, Ye Q, Ai F, Liu P, Wang S, Li G, Shen S, Ma J. Suppression Colitis and Colitis-Associated Colon Cancer by Anti-S100a9 Antibody in Mice. Front Immunol. 2017;8:1774.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 59]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
65.  Sun Z, Yang S, Zhou Q, Wang G, Song J, Li Z, Zhang Z, Xu J, Xia K, Chang Y, Liu J, Yuan W. Emerging role of exosome-derived long non-coding RNAs in tumor microenvironment. Mol Cancer. 2018;17:82.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 196]  [Cited by in F6Publishing: 281]  [Article Influence: 46.8]  [Reference Citation Analysis (0)]
66.  Drak Alsibai K, Meseure D. Tumor microenvironment and noncoding RNAs as co-drivers of epithelial-mesenchymal transition and cancer metastasis. Dev Dyn. 2018;247:405-431.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 33]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
67.  Lakatos PL, Lakatos L. Risk for colorectal cancer in ulcerative colitis: changes, causes and management strategies. World J Gastroenterol. 2008;14:3937-3947.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 303]  [Cited by in F6Publishing: 297]  [Article Influence: 18.6]  [Reference Citation Analysis (0)]
68.  Guo Y, Ji X, Liu J, Fan D, Zhou Q, Chen C, Wang W, Wang G, Wang H, Yuan W, Ji Z, Sun Z. Effects of exosomes on pre-metastatic niche formation in tumors. Mol Cancer. 2019;18:39.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 178]  [Cited by in F6Publishing: 260]  [Article Influence: 52.0]  [Reference Citation Analysis (0)]
69.  Que RS, Lin C, Ding GP, Wu ZR, Cao LP. Increasing the immune activity of exosomes: the effect of miRNA-depleted exosome proteins on activating dendritic cell/cytokine-induced killer cells against pancreatic cancer. J Zhejiang Univ Sci B. 2016;17:352-360.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 47]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
70.  Baassiri A, Nassar F, Mukherji D, Shamseddine A, Nasr R, Temraz S. Exosomal Non Coding RNA in LIQUID Biopsies as a Promising Biomarker for Colorectal Cancer. Int J Mol Sci. 2020;21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 64]  [Article Influence: 16.0]  [Reference Citation Analysis (0)]
71.  Jiang N, Tian H, Pan J, Gong Z. [Circulating long noncoding RNAs as biomarkers in tumor diagnosis]. Sheng Wu Gong Cheng Xue Bao. 2017;33:910-922.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
72.  Kahroba H, Hejazi MS, Samadi N. Exosomes: from carcinogenesis and metastasis to diagnosis and treatment of gastric cancer. Cell Mol Life Sci. 2019;76:1747-1758.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 89]  [Article Influence: 17.8]  [Reference Citation Analysis (0)]
73.  Zhou J, Li XL, Chen ZR, Chng WJ. Tumor-derived exosomes in colorectal cancer progression and their clinical applications. Oncotarget. 2017;8:100781-100790.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 42]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
74.  Li C, Li W, Zhang Y, Zhang X, Liu T, Yang Y, Wang L, Pan H, Ji J, Wang C. Increased expression of antisense lncRNA SPINT1-AS1 predicts a poor prognosis in colorectal cancer and is negatively correlated with its sense transcript. Onco Targets Ther. 2018;11:3969-3978.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 34]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
75.  Wang L, Duan W, Yan S, Xie Y, Wang C. Circulating long non-coding RNA colon cancer-associated transcript 2 protected by exosome as a potential biomarker for colorectal cancer. Biomed Pharmacother. 2019;113:108758.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 46]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
76.  Liu T, Zhang X, Gao S, Jing F, Yang Y, Du L, Zheng G, Li P, Li C, Wang C. Exosomal long noncoding RNA CRNDE-h as a novel serum-based biomarker for diagnosis and prognosis of colorectal cancer. Oncotarget. 2016;7:85551-85563.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 196]  [Cited by in F6Publishing: 242]  [Article Influence: 40.3]  [Reference Citation Analysis (0)]
77.  Yu B, Du Q, Li H, Liu HY, Ye X, Zhu B, Zhai Q, Li XX. Diagnostic potential of serum exosomal colorectal neoplasia differentially expressed long non-coding RNA (CRNDE-p) and microRNA-217 expression in colorectal carcinoma. Oncotarget. 2017;8:83745-83753.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 45]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
78.  Oehme F, Krahl S, Gyorffy B, Muessle B, Rao V, Greif H, Ziegler N, Lin K, Thepkaysone ML, Polster H, Tonn T, Schneider M, Weitz J, Baenke F, Kahlert C. Low level of exosomal long non-coding RNA HOTTIP is a prognostic biomarker in colorectal cancer. RNA Biol. 2019;16:1339-1345.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 61]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
79.  Hu D, Zhan Y, Zhu K, Bai M, Han J, Si Y, Zhang H, Kong D. Plasma Exosomal Long Non-Coding RNAs Serve as Biomarkers for Early Detection of Colorectal Cancer. Cell Physiol Biochem. 2018;51:2704-2715.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 62]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
80.  Huang X, Sun L, Wen S, Deng D, Wan F, He X, Tian L, Liang L, Wei C, Gao K, Fu Q, Li Y, Jiang J, Zhai R, He M. RNA sequencing of plasma exosomes revealed novel functional long noncoding RNAs in hepatocellular carcinoma. Cancer Sci. 2020;111:3338-3349.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 50]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
81.  Tan S, Xia L, Yi P, Han Y, Tang L, Pan Q, Tian Y, Rao S, Oyang L, Liang J, Lin J, Su M, Shi Y, Cao D, Zhou Y, Liao Q. Exosomal miRNAs in tumor microenvironment. J Exp Clin Cancer Res. 2020;39:67.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 108]  [Article Influence: 27.0]  [Reference Citation Analysis (0)]
82.  Kowal J, Arras G, Colombo M, Jouve M, Morath JP, Primdal-Bengtson B, Dingli F, Loew D, Tkach M, Théry C. Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes. Proc Natl Acad Sci USA. 2016;113:E968-E977.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2360]  [Cited by in F6Publishing: 2302]  [Article Influence: 287.8]  [Reference Citation Analysis (0)]
83.  van Niel G, D'Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol. 2018;19:213-228.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3060]  [Cited by in F6Publishing: 4552]  [Article Influence: 758.7]  [Reference Citation Analysis (0)]