Next Article in Journal
The Anticancer Efficacy of Plasma-Oxidized Saline (POS) in the Ehrlich Ascites Carcinoma Model In Vitro and In Vivo
Next Article in Special Issue
Colorectal Cancer after Kidney Transplantation: A Screening Colonoscopy Case-Control Study
Previous Article in Journal
Screening of the Drug-Induced Effects of Prostaglandin EP2 and FP Agonists on 3D Cultures of Dexamethasone-Treated Human Trabecular Meshwork Cells
Previous Article in Special Issue
Addition of V-Stage to Conventional TNM Staging to Create the TNVM Staging System for Accurate Prediction of Prognosis in Colon Cancer: A Multi-Institutional Retrospective Cohort Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Exosomal Components and Modulators in Colorectal Cancer: Novel Diagnosis and Prognosis Biomarkers

1
Department of Biomedical Imaging and Radiological Science, National Yang-Ming University, Taipei 112, Taiwan
2
Department of Biomedical Imaging and Radiological Science, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
3
Genomics Research Center, Academia Sinica, Taipei 115, Taiwan
4
National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 350, Taiwan
5
Department of Biochemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan
6
Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
7
Department of Pathology, Taipei Medical University Hospital, Taipei 110, Taiwan
*
Authors to whom correspondence should be addressed.
Two authors contribute equally to the work.
Biomedicines 2021, 9(8), 931; https://doi.org/10.3390/biomedicines9080931
Submission received: 3 July 2021 / Revised: 23 July 2021 / Accepted: 29 July 2021 / Published: 31 July 2021
(This article belongs to the Special Issue Colorectal Cancer: New Diagnostic and Therapeutic Approaches)

Abstract

:
The relatively high incidence and mortality rates for colorectal carcinoma (CRC) make it a formidable malignant tumor. Comprehensive strategies have been applied to predict patient survival and diagnosis. Various clinical regimens have also been developed to improve the therapeutic outcome. Extracellular vesicles (EVs) are recently proposed cellular structures that can be produced by natural or artificial methods and have been extensively studied. In addition to their innate functions, EVs can be manipulated to be drug carriers and exert many biological functions. The composition of EVs, their intravesicular components, and the surrounding tumor microenvironment are closely related to the development of colorectal cancer. Determining the expression profiles of exocytosis samples and using them as indicators for selecting effective combination therapy is an indispensable direction for EV study and should be regarded as a novel prediction platform in addition to cancer stage, prognosis, and other clinical assessments. In this review, we summarize the function, regulation, and application of EVs in the colon cancer research field. We provide an update on and discuss potential values for clinical applications of EVs. Moreover, we illustrate the specific markers, mediators, and genetic alterations of EVs in colorectal carcinogenesis. Furthermore, we outline the vital markers present in the EVs and discuss their plausible uses in colon cancer patient therapy in combination with the currently used clinical strategies. The development and application of these EVs will significantly improve the accuracy of diagnosis, lead to more precise prognoses, and may lead to the improved treatment of colorectal cancer.

1. Background of CRC

The high incidence of CRC makes it one of the malignances with the highest mortalities in the world [1]. In recent decades, many Asian countries have reported an increase in the incidence of CRC [2]. The low overall survival rate of patients diagnosed with advanced stages still cannot be improved despite recent advances in CRC screening and treatment. The morbidity and mortality of CRC after initial treatment are mainly caused by tumor recurrence and distant metastasis. Notwithstanding anti-vascular endothelial growth factor (VEGF) [3,4] and anti-epidermal growth factor receptor (EGFR) therapies [5,6], two targeted treatments currently available for CRC, relatively few means of improving survival have been reported. There is accordingly an urgent need to clarify the relevant mechanisms of tumor progression and find novel targets for their application in prognosis or treatment evaluation.
The primary lesions of colorectal cancer include bowel cancer, colon cancer, and rectal cancer. Most patients have symptoms such as abdominal pain, anemia, and bleeding. The development and derivation of aberrant polyps can eventually become CRC [7]. In addition, colonic epithelial cells are also prone to epithelial–mesenchymal transition (EMT). The EMT process is accompanied by drug resistance, cell morphology changes, metastasis, and the production/release of extracellular vesicles (EVs). There are many strategies to treat colorectal cancer (e.g., surgery, chemotherapy, radiotherapy, targeted therapy, and immunotherapy) and there are also complex combination treatments, such as folinic acid, fluorouracil, oxaliplatin (FOLFOX) and folinic acid, 5-fluorouracil (5-FU), and irinotecan (FOLFIRI) regimens [8,9,10]. Even with these approaches, colorectal cancer has not been significantly addressed. The inferior accuracy of early diagnosis and high metastatic instinct of CRC lead to a dismal survival rate among advanced patients.
In recent years, colorectal immunotherapy has become an additional standard regimen for patients with advanced diseases when the abovementioned chemotherapy/radiotherapy reaches its limit. Previous studies have reported the occurrence of mismatch repair defects (dMMR) in CRCs that resulted in hypermethylation in the promoter region of MLH1 gene [11]. This event led to an increase in high tumor mutational burden (TMB) as well as altered microsatellite sequences, leaving these tumors in a state of high microsatellite instability status (MSI-H) [12]. Therefore, TMB and MSI status have been considered as items to evaluate and select the appropriate population for immunotherapy [13,14,15]. More importantly, scientists are exploring how EVs derived from immune cells after colorectal immunotherapy can affect the immune response and promote immune system reprogramming. All in all, EVs contribute and connect the tumor microenvironment (TME) and local immune responses and play important roles in the development of CRC.

2. General Definition, Classification, and Application of EVs

Extracellular vesicles (EVs) are composed of lipid bilayers, which are mainly formed by cell secretion via exocytosis. These EVs can be classified into three subtypes, namely macrovesicles, exosomes, and apoptotic bodies, based on their release pathways, size, and content [16]. In this review, we mainly focus on the role of exosomes and cellular exocytosis in colorectal cancer. Exosomes carry mRNA, microRNA (miRNA), long non-coding RNA (lncRNA), protein, DNA fragments, and debris and transport them to the extracellular space [17,18,19]. Therefore, various studies have revealed significant exosome production in the TME, and the contents of these EVs are diverse. Research in recent years has indicated that they can be found in many body fluids. EVs have been identified and purified from blood, saliva, urine, and cerebrospinal fluid (CSF) [20,21,22,23]. There is no doubt that these EV contents can be used for diagnosis: (1) By merging them with clinicopathological factors, TNM staging, survival rate, and health/tumor definition can be easily classified [24]. (2) The properties of EVs have been used to control the rate and yield of exocytosis through gene transfection/infection, chemical compounds, growth factors, and genetic alteration events [25]. (3) EVs have targeting and homing abilities, can be modulated for their release area and target the tumor microenvironment or immune system, and have been regarded as a branch of biomedical imaging [26]. (4) Although EVs do not have the ability to replicate, they can be used as drug carriers for mass production and collection, creating more options for combination therapy [27].
The artificial manufacture of EVs has become a powerful technique to produce EVs on a large scale. Unlike traditional collection procedures, artificial methods can increase yield and reduce the scale of cell preparation and reuse. The methods of artificially promoting EV production are generally divided into physical and biochemical treatments. The physical methods can be used to produce EVs on a large scale through, for instance, an external electric field or ultrasonic vibration. The external electric field is used to stimulate the cells to release EVs. The application is based on the theory of electroporation and was developed as a new artificial manufacturing method called nano-electroporation (NEP) [28]. The NEP system can be applied to a monolayer of donor cells culturing on the surface of a chip, with the surface of the chip containing a series of nanochannels. The negative electrode is in contact with the bottom reservoir containing the cargo solution, which means that this method can also package the target cargo into EVs. After applying transient electrical pulses to cells, this NEP technique can produce large amounts of EVs. In addition, other studies have shown that cells treated with ultrasound stimulation can produce a larger quantity of EVs. The cells are repeatedly exposed to ultrasonic vibration for 10 min and then incubated for 30 min after excitation, and EV production can be expanded by 8–10 times. Through massive parallelization, Ambattu et al. used low-cost surface-reflected bulk waves to mass-produce EVs [29]. The biochemical treatments for EV production include changing the content of the cell culture medium, adding additional proteins that stimulate secretion of EVs, and genetic engineering of the gene structure of donor cells to secrete EVs on a large scale.

3. Modulation and Regulation Mechanisms for EVs and Their Components

3.1. ESCRT Complex

The formation of EVs involves a complicated mechanism and requires many steps. The core subunit is the endosomal sorting complex (ESCRT), which is composed of four main complexes (ESCRT-0, -I, -II, and -III) (Table 1). Several specific molecules are transported into the intraluminal region of the multivesicular bodies (MVBs) [30,31]. These subunits of ESCRT have unique roles in the exosome biogenesis. ESCRT-0 is a multivesicular reaction that can bind to the cell surface and accumulate receptors and/or ubiquitinated proteins. ESCRT-0 needs phosphoatidylinositol 3-phosphate to activate, directly interacts with the prosaposin domain, and then recruits the ESCRT-I subunit tumor susceptibility 101 (TSG101) to form the ESCRT-I complex [32]. In addition, ubiquitination assists ESCRT-I protein to activate and convert the bridge to connect between ESCRT-0 and ESCRT-II complexes. The ESCRT-II complex can deliver ubiquitinated proteins to endosomal membranes and plays a role in the biogenesis of multivesicular bodies. The targets are transferred from ESCRT-0 to ESCRT-I and then to ESCRT-II [33]. ESCRT-II and ESCRT-III form a cascade reaction in which the cargo containing vesicles is clamped. Therefore, ESCRT-III is responsible for cargo sorting, concentration, vesicle lysis, and material recycling [34]. ESCRT machinery also relies on several accessory proteins, including Vps4-Vta-1 complex and Bro1/ALIX proteins [34]. Once exocytosis is complete, the Vps4-Vta-1 protein strips other ESCRT components from the membrane, while the Bro1/ALIX protein helps to recruit deubiquitinases to the ESCRT-III [35]. This modification can remove the established ubiquitin tag and put an end to the exocytosis process.

3.2. RAB Family

Among the superfamily of Ras-like small GTPase, Rab GTPase is the largest family and has an essential function in the regulation of membrane identity and vesicle formation in the entire process of transportation in human physiology [51]. Dysregulation of Rab GTPases is associated with diverse inherited disorders. Among these disorders, neuron-associated diseases, including Parkinson’s disease and Huntington’s disease, are related to the imbalance of membrane trafficking regulated by Rab, which affects the neuron connections in the synapses [52]. Rabs can also regulate the translocation of Glut4 (a glucose transporter), which is associated with the pathogenesis of type 2 diabetes [53]. The key mediators necessary for innate immunity, such as the phagocytosis of intracellular pathogens, also require Rab members [54]. In cancer, the role of the Ras proto-oncogene in tumorigenesis has been widely discussed. However, how the Rab GTPases contribute to cancer progression remains largely unknown and is worth further investigation. To date, the most common endocytic-related Rabs are reported to be Rab5, Rab21, and Rab25 [55]. Among them, Rab25, which functions as a director in modulating integrin-recycling vesicle movement, has been well characterized. It is worth noting that Rab25 is also involved in the cell movement of epithelial cells, such as transformation and motility, which are correlated to tumor progression. The aggressiveness activity of female-related cancer types, such as breast and ovarian cancer, are all related to Rab25 overexpression. Interestingly, as a controversial factor, Rab25 serves as a tumor suppressor in colon cancer [56]. Another member, Rab5, was found to participate in the fusion of the early endosome process and regulate cell survival and migration by integrating with caspase 8, and the differential expression of Rab5 in cancers has been reported [57].
Only a few RABs are involved in exocytosis (RAB3/8/26/27); other family members participate in early endosomes (RAB4/5/10/17/21/23/35), late endosomes (RAB7/9), autophagosomes (RAB2/7/24/27/33), recycling endosomes (RAB11/13/17/25/35), and in anchoring to the ER–Golgi intermediated compartment (RAB2/18) [58] (Figure 1).

3.3. Genetic Alterations

Many tumor cells are prone to gene mutations throughout their development. Upon drug treatment, the selective pressure eventually sieves out cells with mutations showing drug-resistance capability. There are various genetic alterations reported in CRC. Some of these hotspots and events are highly correlated with the formation of EVs and exocytosis, thereby leading to increased malignancies and reducing the survival of patients. According to previous profiles, KRAS, PIK3CA, BRAF, EGFR, and ERBB2 are the most frequent gene alterations in CRC patients [59]. Isogenic colorectal cancer cell lines have been demonstrated to regulate EV cargo content (miRNAs, circRNAs, mRNAs, and proteins) in a KRAS-dependent manner [60,61,62,63]. Interestingly, RAB13 is not one of the RAB members that controls exosomes, but it has been proven to promote EV production in the KRAS mutation model [64]. PIK3CA mutant status is one of the hallmarks of CRC. Through next-generation sequencing analysis, approximately 30% of PIK3CA mutants were found in CRC patients [65]. PIK3CA affects the PI3K/Akt/mTOR pathway, and there is currently no effective targeting drug [66]. This pathway increases the secretion of EVs, and EVs containing PIK3CA can transfer from malignant cells to other cells for proliferation. There are also EVs that promote PIK3/Akt signaling and the exchange of substances (proteins and lipids) [67]. In addition, although there are targeted therapeutic compounds for EGFR (HER1) and ERBB2 (HER2) (cetuximab/panitumumab and trastuzumab/pertuzumab, respectively), they are not efficient in cases of mutations and continuously activate the downstream signals, including the RAS/RAF/MEK and PI3K/Akt/mTOR axes [68]. Moreover, in colon cancer, the V600E mutant of BRAF subsequently activates the RAS/RAF/MEK pathway, leading to a poor prognosis and strong exocytosis. Most importantly, the high CpG island methylator phenotype (CIMP-H) occurs simultaneously with MSI-H, KRASmut, TP53mut, BRAFmut, and various genetic alterations. The result is the overexpression and continuous production of exocytosis, which are beneficial to cancer progression.

4. Role of EVs in Colorectal Tumorigenesis

According to previous studies, the subunits of the ESCRT complexes and members of the RAB family that are involved in exocytosis have been identified (Table 1). These components have been investigated in colon cancer research. It has been reported that the expression levels of some candidates are varied during colon tumorigenesis. According to these reports, some genes are important for their prognostic value and can increase the hazard ratio of patients. Most roles of ESCRTs and RABs in cancer progression are found to be overexpressed to confer cancer cells with diverse functions related to malignancy. Environmental stress revealing enhanced proliferation and metabolic reprogramming has been found in cancer cells showing resistance to chemotherapy/radiotherapy, and overexpression of exocytotic and EV-related genes/proteins has been observed in auxiliary cancer cells. Additionally, these altered expression profiles also persist in the metastatic cancer cells. In contrast, Vps4-Vta1 and ALIX act as gatekeepers to terminate exocytosis and, therefore, their function is often suppressed in malignant colorectal tumors [45,46]. Most RABs show excessive expression, coordinating the production and transportation of EVs, but there are also some redundant trends (Table 1). Several RABs show inconsistent indications in colon cancer studies [49,50] and need to be studied and classified in detail in patient cohorts.
The large amount of biological information carried by EV particles has been shown to be related to tumorigenesis and endows cancer cells with different phenotypes (Table 2). The most common ones contain many non-coding RNAs and proteins. It has been confirmed that abundant miRNAs are related to colon cancer development, most of which are highly expressed to regulate a variety of cancer functions (chemoresistance, radioresistance, immune response, metastasis, and proliferation). In contrast, some tumor suppressor miRNAs have also been discovered [27,69,70,71]. lncRNAs and circRNAs directly control the expression and processing of miRNAs and affect tumor progression. As a well-known long non-coding RNA, lncCRNDE regulates multiple signaling pathways and functions [72]. It is considered as a diagnostic/prognostic predictor of colon cancer. The direct targets of these lncRNAs have been explored to elucidate the pathogenic mechanisms. For example, Liu et al. discovered that lncCRNDE has a positive correlation with IRX5 RNA [73]. Cheng and his group also reported that lncRNA GAS5 can inhibit colorectal cancer cell proliferation via the miR-182-5p/FOXO3a axis [74]. Similarly, both CCAT2-miR-145 and CCAL-β-catenin regulation are presented in colon cancer proliferation and progression [75,76]. Moreover, circHIPK3 interferes with miR-7 to promote the growth and metastasis of colorectal cancer and acts as an alternative to regulate miR-1207-5p/FMNL2 signaling [77,78]. Yang et al. claimed that circ-133 acts on the miR-133a/GEF-H1/RhoA axis to promote colon cancer metastasis [79]. Hon et al. stated that circ_0000338 plays a dual regulatory role in chemoresistance CRC [80]. Consistent with this, circRNAs also reflect aberrant exocytosis caused by genetic changes and participate in its regulation [62]. Some exosomal proteins can be used for blood screening, EV collection, and analysis of patient clinical indicators [81,82,83,84,85,86,87,88,89,90,91,92,93]. These proteins can also regulate single or multiple aspects of colon cancer. Other related proteins have been observed in these studies but are not involved in exocytosis, and thus we exclude them from discussion in this article (Figure 1).
On the other hand, compared with the exosome derived from cancer cells, exosomes derived from the immune system have different contributions. They can meditate crosstalk between immunity and regulate cancer progression. First, B cell-derived exosomes were identified, and then it was found that lymphocytes, dendritic cells (DCs), natural killer cells, mast cells, macrophages, and thymocytes can all produce exosomes [123]. Yan et al. has demonstrated that the exosomes of various immune cells contain their origins, target cells, consequences, and involved molecules [124]. According to these previous studies from the literature, exosomes derived from immune cells can effectively regulate the immune response (innate/adaptive) and provide cytotoxic effects against cancer cells. Moreover, EVs derived from immune cells are known to exert similar functions as those of their parent immune cells, and the development of activated immune-derived EVs can also be used for immunotherapy applications [125]. Conversely, if EVs are derived from immune cells that are known to promote tumor development (such as TAMs), they can instead increase the malignancy of cancer phenotype in many aspects [126,127].

5. Available Omics Datasets of EVs for CRC

Previous studies have stated that EVs package a variety of fragments containing biological information, including DNA, RNA, and proteins [128]. These substances rearrange the tumor microenvironment and regulate the immune system and metabolic events, thereby promoting tumor colonization, proliferation, and metastasis [129]. The extraction of EVs, determination of their contents, and classification could be utilized to evaluate whether they can be applied as clinical parameters for prognosis and to investigate the underlying molecular mechanisms.
In the multi-omics category, many profiles of EVs and exosomes have been established (Table 3). These datasets provide potential candidates through hierarchical analysis and statistical calculations, which can accurately determine the corresponding clinical events of colorectal cancer. The core data or clustering that meets the cut-off/fold change can also predict potential upstream/downstream pathways, as well as the transcription factors involved and the consequences of gene/drug regulation. Ohshima et al. observed and verified the quality of exosomes through transmission electron microscopy (TEM) and Western blotting (CD29, Tsg101, Alip1, and Bip) [130]. Compared with the cultured cell lines and their media, they determined that in the exosomes several members of the let-7s family of miRNAs undergo significant changes. Although they illustrated the proposed model with the metastatic gastric cancer cell AZ-P7a, they also established profiles in colorectal cancer cells (SW480 and SW620) [130]. In the study by Otaga-Kawata et al., serum was collected to extract exosome-enriched fractions from CRC clinical patients. In addition to let-7, they found six more candidates (miR-1229, miR-1246, miR-150, miR-21, miR-223, and miR-23a) that were upregulated compared to healthy controls [94]. These candidates have been compared with current biomarkers of colorectal cancer (e.g., carbohydrate antigen (CA19-9) and carcinoembryonic antigen (CEA)). Their results show that these candidates are of good specificity and sensitivity for use as additional biomarkers for CRC.
Genetic alterations have always been considered an important issue in tumorigenesis and affect the secretion and exocytosis of EVs. KRAS is a typical mutation event in both CRC cell lines and clinical specimens. Cha and her team revealed that small RNA composition is correlated with KRAS status by using the wild-type KRAS cells DKs-8, mutant KRAS cells DKO-1, and their counterpart cells DLD-1 as models [60]. They listed the increase or decrease in gene expression in a single cell model and showed the common signatures in their study. The Dou and Hinger groups also chose similar models for comparison [62,63]. They detected some circular RNAs and long non-coding RNAs. These results are also consistent with the proposed model according to which EVs and exosomes contain various biomessenger fragments. This evidence also suggests that EVs are suitable for development as prediction markers. Furthermore, Yoshii et al. explored the differences in exosomal functions and miRNA expression levels in the absence of TP53 (GSE120013).
Other EV studies in CRC used various colorectal cancer cells as models. Chiba recruited exosomes from SW480 cells (GSE68979), and Ji et al. distinguished three subtypes of EVs from the human LIM1863 cancer cell line [131]. Moreover, Sun and his group reported their results in HT-29 and SW-948 cells [134,135]. Some stem cell markers (CD44v6, Tspan8, CD151, and Claudin7) have been manipulated in exosome research. In the CRC cell models, Tubita et al. also created a series of multi-omics datasets and mentioned that several miRNAs (miR-6127, miR-6746-5p, and miR-6787-5p) can be regulated in the pre-metastatic niche of colon cancer [136]. RNA-binding proteins and other cellular proteins have also been observed, and their changes have been confirmed in exosomes [137]. The EVs’ core omics profiles are not limited to the collected species and sources; transcriptomics and RNA-seq data can also be completed with patient specimens, xenografted tissues, and animal models/cell lines. These libraries include comparisons of before and after treatments, single target modulation, and normal distribution between clinical cohorts [132,133]. In addition, the multiple omics established in the Mus musculus also provide a better biological reservation and efficacy evaluation perspective for exosomal research (GSE101950, GSE101951, and GSE173202).

6. Current Combinations and Clinical Trials of EV-Based Carriers

Trials and combination therapies related to EVs are ongoing. The main reason is that, as we have mentioned, EVs have the characteristics of cargo loading and delivery. Through announced clinical trials (http://clinicaltrials.gov, accessed on 30 June 2021), various EV applications and combination therapies are being considered for colon cancer research (Table 4). Most of these clinical EV studies involve the use of patients’ blood samples for biomarker evaluation. These studies divide patients by healthy and tumor samples, primary/metastasis status, and other cancer subtypes to investigate the differences. In addition, they further identified components with the available parameters, including macromolecules, integrins, and metalloproteases. Furthermore, several trials have begun to use EVs as drug carriers. These drugs include curcumin (NCT01294072), Toripalimab (NCT03927898), and AL3810 (NCT03260179). In addition, in 1989, Sidney Altman and Thomas Cech discovered that small RNA and DNA have functions such as ligand binding and activation and gene regulation. They could screen for secondary structure RNA or single-stranded DNA that would specifically bind to the target protein. These specialized small molecule nucleic acids are called aptamers. Aptamers can also be coupled to EVs to promote specific targeting, such as the dart method for EVs that compete with CD63-specific aptamers [138]. This modification can direct EVs to their targeting cells or tissues and deliver the drugs only to specific tissue or cells. Moreover, recent research has made progress in the development of nucleic acid drugs (such as mRNA). Several nucleic acid drugs have been approved for marketing. The clinical data of these potential blockbuster drugs have recently been released. There are also endless mergers, acquisitions, and product introduction transactions in the field of nucleic acid drugs. Due to the recent COVID pandemic, the development of mRNA vaccines has also received more attention. These nucleic acid drugs can also be embedded in EVs and delivered to target cells or tissues. Using EVs as carriers can provide the following improvements for the delivery of nucleic acid drugs: First, EVs can avoid rapid elimination of nucleic acid drugs and extend their half-life. Second, EVs can target specific sites or home in on primary colorectal tumors through small peptides, aptamers, and surface antibodies. Finally, EVs can reduce the drug escape efficiency of endosomes to make nucleic acid drugs more likely to stay in target cells [139,140,141] (Figure 2).
At present, data on the clinical use of EVs in cancer treatment are quite scarce. In addition to various patents, there are currently many restrictions, including purification, formulation, dosage, delivery, biodistribution, etc. Xu et al. have summarized the purification methods used in EVs. Except for synthetic polymer-based precipitation approaches that can achieve higher EV yields, most other methods can only get low to medium yields and quantities (μL–mL) [142]. In addition, many purification methods must deal with particles with sizes more than 70 nm, and this relatively large particle size will seriously affect its production. In a study on colon cancer, Dai et al. tried to collect ascites-derived exosomes (Aex) from 800 mL ascites and then, after combining them with granulocyte-macrophage colony-stimulating factor (GM-CSF), compared them with the control (Aex alone, 100~500 μg) [143]. They claimed that this strategy is feasible and safe and that future research should refer to and use it as a template for improvement. Moreover, the yield and composition of EVs also have a lot to do with the source. As Allelein et al. confirmed, heterogenous populations vary greatly. This phenomenon must exist in clinical samples [144]. In order to overcome these bottlenecks, we suggest that more studies need to sort out detailed information, including source cell type, extraction method, purity, modified vesicle type, etc. [145].

7. Future Prospects

In view of their role in known mechanisms of colon cancer, EVs can be used as therapeutic targets by inhibiting the formation, release, and uptake of EVs or by targeting a biologically active substance to reverse colorectal tumor phenotypes. EVs can also be applied as therapeutic agents. Unlike common drug delivery vehicles such as liposomes or nanoparticles, EVs have minimal immunogenicity and toxicity. In addition, they are produced by endogenous cells, have fewer rejection reactions, and can carry a variety of combinations (small molecule compounds, modified peptides, monoclonal antibodies, and antagomiRs). These advantages enrich the application of EVs as a breakthrough point in combination therapy.
EVs and cellular exocytosis are still difficult to modulate, and the complete mechanism remains to be investigated. Although many candidates are related to exosomal particles, omics profiles cannot reveal the degree of consistency/dependence between them. Which is the most critical role/event in exosomes has yet to be verified. At present, most EVs are limited to clinical prognosis and diagnosis. In future basic research on EVs, it is necessary to determine the exact target genes and downstream axis regulated by miRNAs. At the same time, the identified circRNA/lncRNA should also help to elucidate the relationship with miRNA. Finally, EV-based strategies also require a comprehensive biomedical imaging platform. Through real-time feedback to patients with colorectal cancer, the potential therapeutic value can be monitored and evaluated.

Author Contributions

Y.-C.C. and M.-H.C. participated in the conceptualization. C.-H.L. and C.-Y.F. contributed to critical editing of manuscript. Funding is provided by M.H. and C.-L.C.; Data acquisition was done by Y.-C.C. and M.-H.C. Supervision, M.H. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by a grant from the Ministry of Science and Technology (MOST 109-2320-B-038-033) to C.L. Chen.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Rawla, P.; Sunkara, T.; Barsouk, A. Epidemiology of colorectal cancer: Incidence, mortality, survival, and risk factors. Prz. Gastroenterol. 2019, 14, 89–103. [Google Scholar] [CrossRef]
  2. Wong, M.C.; Ding, H.; Wang, J.; Chan, P.S.; Huang, J. Prevalence and risk factors of colorectal cancer in Asia. Intest. Res. 2019, 17, 317–329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Hopirtean, C.; Nagy, V. Optimizing the use of anti VEGF targeted therapies in patients with metastatic colorectal cancer: Review of literature. Clujul Med. 2018, 91, 12–17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Lieu, C.H.; Tran, H.; Jiang, Z.Q.; Mao, M.; Overman, M.J.; Lin, E.; Eng, C.; Morris, J.; Ellis, L.; Heymach, J.V.; et al. The association of alternate VEGF ligands with resistance to anti-VEGF therapy in metastatic colorectal cancer. PLoS ONE 2013, 8, e77117. [Google Scholar] [CrossRef] [PubMed]
  5. Dercle, L.; Lu, L.; Schwartz, L.H.; Qian, M.; Tejpar, S.; Eggleton, P.; Zhao, B.; Piessevaux, H. Radiomics Response Signature for Identification of Metastatic Colorectal Cancer Sensitive to Therapies Targeting EGFR Pathway. J. Natl. Cancer Inst. 2020, 112, 902–912. [Google Scholar] [CrossRef]
  6. Martinelli, E.; Ciardiello, D.; Martini, G.; Troiani, T.; Cardone, C.; Vitiello, P.P.; Normanno, N.; Rachiglio, A.M.; Maiello, E.; Latiano, T.; et al. Implementing anti-epidermal growth factor receptor (EGFR) therapy in metastatic colorectal cancer: Challenges and future perspectives. Ann. Oncol. 2020, 31, 30–40. [Google Scholar] [CrossRef] [Green Version]
  7. Kuipers, E.J.; Grady, W.M.; Lieberman, D.; Seufferlein, T.; Sung, J.J.; Boelens, P.G.; van de Velde, C.J.; Watanabe, T. Colorectal cancer. Nat. Rev. Dis. Primers 2015, 1, 15065. [Google Scholar] [CrossRef] [Green Version]
  8. Bender, U.; Rho, Y.S.; Barrera, I.; Aghajanyan, S.; Acoba, J.; Kavan, P. Adjuvant therapy for stages II and III colon cancer: Risk stratification, treatment duration, and future directions. Curr. Oncol. 2019, 26, S43–S52. [Google Scholar] [CrossRef] [Green Version]
  9. Benson, A.B., 3rd; Schrag, D.; Somerfield, M.R.; Cohen, A.M.; Figueredo, A.T.; Flynn, P.J.; Krzyzanowska, M.K.; Maroun, J.; McAllister, P.; Van Cutsem, E.; et al. American Society of Clinical Oncology recommendations on adjuvant chemotherapy for stage II colon cancer. J. Clin. Oncol. 2004, 22, 3408–3419. [Google Scholar] [CrossRef]
  10. Venook, A.P.; Niedzwiecki, D.; Lenz, H.J.; Innocenti, F.; Fruth, B.; Meyerhardt, J.A.; Schrag, D.; Greene, C.; O’Neil, B.H.; Atkins, J.N.; et al. Effect of First-Line Chemotherapy Combined with Cetuximab or Bevacizumab on Overall Survival in Patients with KRAS Wild-Type Advanced or Metastatic Colorectal Cancer: A Randomized Clinical Trial. JAMA 2017, 317, 2392–2401. [Google Scholar] [CrossRef] [Green Version]
  11. Kuismanen, S.A.; Holmberg, M.T.; Salovaara, R.; de la Chapelle, A.; Peltomäki, P. Genetic and epigenetic modification of MLH1 accounts for a major share of microsatellite-unstable colorectal cancers. Am. J. Pathol. 2000, 156, 1773–1779. [Google Scholar] [CrossRef] [Green Version]
  12. Xiao, J.; Li, W.; Huang, Y.; Huang, M.; Li, S.; Zhai, X.; Zhao, J.; Gao, C.; Xie, W.; Qin, H.; et al. A next-generation sequencing-based strategy combining microsatellite instability and tumor mutation burden for comprehensive molecular diagnosis of advanced colorectal cancer. BMC Cancer 2021, 21, 282. [Google Scholar] [CrossRef] [PubMed]
  13. Goodman, A.M.; Sokol, E.S.; Frampton, G.M.; Lippman, S.M.; Kurzrock, R. Microsatellite-Stable Tumors with High Mutational Burden Benefit from Immunotherapy. Cancer Immunol. Res. 2019, 7, 1570–1573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Luchini, C.; Bibeau, F.; Ligtenberg, M.J.L.; Singh, N.; Nottegar, A.; Bosse, T.; Miller, R.; Riaz, N.; Douillard, J.Y.; Andre, F.; et al. ESMO recommendations on microsatellite instability testing for immunotherapy in cancer, and its relationship with PD-1/PD-L1 expression and tumour mutational burden: A systematic review-based approach. Ann. Oncol. 2019, 30, 1232–1243. [Google Scholar] [CrossRef] [Green Version]
  15. Schrock, A.B.; Ouyang, C.; Sandhu, J.; Sokol, E.; Jin, D.; Ross, J.S.; Miller, V.A.; Lim, D.; Amanam, I.; Chao, J.; et al. Tumor mutational burden is predictive of response to immune checkpoint inhibitors in MSI-high metastatic colorectal cancer. Ann. Oncol. 2019, 30, 1096–1103. [Google Scholar] [CrossRef] [PubMed]
  16. Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 2018, 7, 1535750. [Google Scholar] [CrossRef] [Green Version]
  17. Hewson, C.; Capraro, D.; Burdach, J.; Whitaker, N.; Morris, K.V. Extracellular vesicle associated long non-coding RNAs functionally enhance cell viability. Noncoding RNA Res. 2016, 1, 3–11. [Google Scholar] [CrossRef] [Green Version]
  18. Huang, X.; Yuan, T.; Tschannen, M.; Sun, Z.; Jacob, H.; Du, M.; Liang, M.; Dittmar, R.L.; Liu, Y.; Liang, M.; et al. Characterization of human plasma-derived exosomal RNAs by deep sequencing. BMC Genom. 2013, 14, 319. [Google Scholar] [CrossRef] [Green Version]
  19. Mittelbrunn, M.; Gutiérrez-Vázquez, C.; Villarroya-Beltri, C.; González, S.; Sánchez-Cabo, F.; González, M.; Bernad, A.; Sánchez-Madrid, F. Unidirectional transfer of microRNA-loaded exosomes from T cells to antigen-presenting cells. Nat. Commun. 2011, 2, 282. [Google Scholar] [CrossRef] [Green Version]
  20. Erozenci, L.A.; Böttger, F.; Bijnsdorp, I.V.; Jimenez, C.R. Urinary exosomal proteins as (pan-)cancer biomarkers: Insights from the proteome. FEBS Lett. 2019, 593, 1580–1597. [Google Scholar] [CrossRef] [Green Version]
  21. Guha, D.; Lorenz, D.R.; Misra, V.; Chettimada, S.; Morgello, S.; Gabuzda, D. Proteomic analysis of cerebrospinal fluid extracellular vesicles reveals synaptic injury, inflammation, and stress response markers in HIV patients with cognitive impairment. J. Neuroinflamm. 2019, 16, 254. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Xiao, Y.; Zhong, J.; Zhong, B.; Huang, J.; Jiang, L.; Jiang, Y.; Yuan, J.; Sun, J.; Dai, L.; Yang, C.; et al. Exosomes as potential sources of biomarkers in colorectal cancer. Cancer Lett. 2020, 476, 13–22. [Google Scholar] [CrossRef] [PubMed]
  23. Zlotogorski-Hurvitz, A.; Dayan, D.; Chaushu, G.; Korvala, J.; Salo, T.; Sormunen, R.; Vered, M. Human saliva-derived exosomes: Comparing methods of isolation. J. Histochem. Cytochem. 2015, 63, 181–189. [Google Scholar] [CrossRef] [Green Version]
  24. Yan, S.; Dang, G.; Zhang, X.; Jin, C.; Qin, L.; Wang, Y.; Shi, M.; Huang, H.; Duan, Q. Downregulation of circulating exosomal miR-638 predicts poor prognosis in colon cancer patients. Oncotarget 2017, 8, 72220–72226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Yáñez-Mó, M.; Siljander, P.R.; Andreu, Z.; Zavec, A.B.; Borràs, F.E.; Buzas, E.I.; Buzas, K.; Casal, E.; Cappello, F.; Carvalho, J.; et al. Biological properties of extracellular vesicles and their physiological functions. J. Extracell. Vesicles 2015, 4, 27066. [Google Scholar] [CrossRef] [Green Version]
  26. Suthar, J.; Parsons, E.S.; Hoogenboom, B.W.; Williams, G.R.; Guldin, S. Acoustic Immunosensing of Exosomes Using a Quartz Crystal Microbalance with Dissipation Monitoring. Anal. Chem. 2020, 92, 4082–4093. [Google Scholar] [CrossRef] [Green Version]
  27. Kubota, S.; Chiba, M.; Watanabe, M.; Sakamoto, M.; Watanabe, N. Secretion of small/microRNAs including miR-638 into extracellular spaces by sphingomyelin phosphodiesterase 3. Oncol. Rep. 2015, 33, 67–73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Yang, Z.; Shi, J.; Xie, J.; Wang, Y.; Sun, J.; Liu, T.; Zhao, Y.; Zhao, X.; Wang, X.; Ma, Y.; et al. Large-scale generation of functional mRNA-encapsulating exosomes via cellular nanoporation. Nat. Biomed. Eng. 2020, 4, 69–83. [Google Scholar] [CrossRef]
  29. Ambattu, L.A.; Ramesan, S.; Dekiwadia, C.; Hanssen, E.; Li, H.; Yeo, L.Y. High frequency acoustic cell stimulation promotes exosome generation regulated by a calcium-dependent mechanism. Commun. Biol. 2020, 3, 553. [Google Scholar] [CrossRef]
  30. Abels, E.R.; Breakefield, X.O. Introduction to Extracellular Vesicles: Biogenesis, RNA Cargo Selection, Content, Release, and Uptake. Cell Mol. Neurobiol. 2016, 36, 301–312. [Google Scholar] [CrossRef]
  31. Zhang, Y.; Liu, Y.; Liu, H.; Tang, W.H. Exosomes: Biogenesis, biologic function and clinical potential. Cell Biosci. 2019, 9, 19. [Google Scholar] [CrossRef] [PubMed]
  32. Migliano, S.M.; Teis, D. ESCRT and Membrane Protein Ubiquitination. Prog. Mol. Subcell. Biol. 2018, 57, 107–135. [Google Scholar] [CrossRef] [PubMed]
  33. Boura, E.; Hurley, J.H. Structural basis for membrane targeting by the MVB12-associated β-prism domain of the human ESCRT-I MVB12 subunit. Proc. Natl. Acad. Sci. USA 2012, 109, 1901–1906. [Google Scholar] [CrossRef] [Green Version]
  34. Tang, S.; Buchkovich, N.J.; Henne, W.M.; Banjade, S.; Kim, Y.J.; Emr, S.D. ESCRT-III activation by parallel action of ESCRT-I/II and ESCRT-0/Bro1 during MVB biogenesis. eLife 2016, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Piper, R.C.; Lehner, P.J. Endosomal transport via ubiquitination. Trends Cell Biol. 2011, 21, 647–655. [Google Scholar] [CrossRef] [Green Version]
  36. Sun, Y.; Zheng, W.; Guo, Z.; Ju, Q.; Zhu, L.; Gao, J.; Zhou, L.; Liu, F.; Xu, Y.; Zhan, Q.; et al. A novel TP53 pathway influences the HGS-mediated exosome formation in colorectal cancer. Sci. Rep. 2016, 6, 28083. [Google Scholar] [CrossRef] [Green Version]
  37. Džombeta, T.; Kapuralin, K.; Ulamec, M.; Tomas, D.; Gajović, S.; Krušlin, B. Immunohistochemical expression of STAM2 in gastrointestinal stromal tumors. Anticancer Res. 2014, 34, 2291–2296. [Google Scholar] [PubMed]
  38. Gheytanchi, E.; Zanjani, L.S.; Ghods, R.; Abolhasani, M.; Shahin, M.; Vafaei, S.; Naseri, M.; Fattahi, F.; Madjd, Z. High expression of tumor susceptibility gene 101 (TSG101) is associated with more aggressive behavior in colorectal carcinoma. J. Cancer Res. Clin. Oncol. 2021, 147, 1631–1646. [Google Scholar] [CrossRef]
  39. Kolmus, K.; Erdenebat, P.; Szymańska, E.; Stewig, B.; Goryca, K.; Derezińska-Wołek, E.; Szumera-Ciećkiewicz, A.; Brewińska-Olchowik, M.; Piwocka, K.; Prochorec-Sobieszek, M.; et al. Concurrent depletion of Vps37 proteins evokes ESCRT-I destabilization and profound cellular stress responses. J. Cell Sci. 2021, 134. [Google Scholar] [CrossRef]
  40. Qina, J.; Ke, T.; Li, C.; Wei-fang, L.; Rong, W.; Gui-Yuan, L. In silico expression analysis of human novel gene UBAP1 in multiple cancers. Chin. J. Cancer Res. 2002, 14, 157–160. [Google Scholar] [CrossRef]
  41. Mastrogamvraki, N.; Zaravinos, A. Signatures of co-deregulated genes and their transcriptional regulators in colorectal cancer. NPJ Syst. Biol. Appl. 2020, 6, 23. [Google Scholar] [CrossRef]
  42. Al-Temaimi, R.A.; Tan, T.Z.; Marafie, M.J.; Thiery, J.P.; Quirke, P.; Al-Mulla, F. Identification of 42 Genes Linked to Stage II Colorectal Cancer Metastatic Relapse. Int. J. Mol. Sci. 2016, 17, 598. [Google Scholar] [CrossRef] [Green Version]
  43. Mo, J.S.; Han, S.H.; Yun, K.J.; Chae, S.C. MicroRNA 429 regulates the expression of CHMP5 in the inflammatory colitis and colorectal cancer cells. Inflamm. Res. 2018, 67, 985–996. [Google Scholar] [CrossRef]
  44. Liu, J.; Song, H.; Yao, L.; Liu, Y.; Zhang, Y.; Zhao, H.; Ji, H.; Wang, Y. Over-expression of the overexpressed in lung cancer-1 is associated with poor prognosis in colorectal cancer. Anticancer Res. 2014, 34, 367–372. [Google Scholar]
  45. Szymańska, E.; Nowak, P.; Kolmus, K.; Cybulska, M.; Goryca, K.; Derezińska-Wołek, E.; Szumera-Ciećkiewicz, A.; Brewińska-Olchowik, M.; Grochowska, A.; Piwocka, K.; et al. Synthetic lethality between VPS4A and VPS4B triggers an inflammatory response in colorectal cancer. EMBO Mol. Med. 2020, 12, e10812. [Google Scholar] [CrossRef] [PubMed]
  46. Valcz, G.; Galamb, O.; Krenács, T.; Spisák, S.; Kalmár, A.; Patai, Á.V.; Wichmann, B.; Dede, K.; Tulassay, Z.; Molnár, B. Exosomes in colorectal carcinoma formation: ALIX under the magnifying glass. Mod. Pathol. 2016, 29, 928–938. [Google Scholar] [CrossRef] [Green Version]
  47. Chang, Y.C.; Su, C.Y.; Chen, M.H.; Chen, W.S.; Chen, C.L.; Hsiao, M. Secretory RAB GTPase 3C modulates IL6-STAT3 pathway to promote colon cancer metastasis and is associated with poor prognosis. Mol. Cancer 2017, 16, 135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Chen, B.; Huang, S.; Pisanic Ii, T.R.; Stark, A.; Tao, Y.; Cheng, B.; Li, Y.; Wei, Y.; Zhao, W.; Wang, T.H.; et al. Rab8 GTPase regulates Klotho-mediated inhibition of cell growth and progression by directly modulating its surface expression in human non-small cell lung cancer. EBioMedicine 2019, 49, 118–132. [Google Scholar] [CrossRef] [PubMed]
  49. Feng, F.; Jiang, Y.; Lu, H.; Lu, X.; Wang, S.; Wang, L.; Wei, M.; Lu, W.; Du, Z.; Ye, Z.; et al. Rab27A mediated by NF-κB promotes the stemness of colon cancer cells via up-regulation of cytokine secretion. Oncotarget 2016, 7, 63342–63351. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Dong, W.; Cui, J.; Yang, J.; Li, W.; Wang, S.; Wang, X.; Li, X.; Lu, Y.; Xiao, W. Decreased expression of Rab27A and Rab27B correlates with metastasis and poor prognosis in colorectal cancer. Discov. Med. 2015, 20, 357–367. [Google Scholar] [PubMed]
  51. Hutagalung, A.H.; Novick, P.J. Role of Rab GTPases in membrane traffic and cell physiology. Physiol. Rev. 2011, 91, 119–149. [Google Scholar] [CrossRef] [Green Version]
  52. Kiral, F.R.; Kohrs, F.E.; Jin, E.J.; Hiesinger, P.R. Rab GTPases and Membrane Trafficking in Neurodegeneration. Curr. Biol. 2018, 28, R471–R486. [Google Scholar] [CrossRef] [Green Version]
  53. Olson, A.L. Regulation of GLUT4 and Insulin-Dependent Glucose Flux. ISRN Mol. Biol. 2012, 2012, 856987. [Google Scholar] [CrossRef] [Green Version]
  54. Prashar, A.; Schnettger, L.; Bernard, E.M.; Gutierrez, M.G. Rab GTPases in Immunity and Inflammation. Front. Cell Infect. Microbiol. 2017, 7, 435. [Google Scholar] [CrossRef] [Green Version]
  55. Subramani, D.; Alahari, S.K. Integrin-mediated function of Rab GTPases in cancer progression. Mol. Cancer 2010, 9, 312. [Google Scholar] [CrossRef] [Green Version]
  56. Mitra, S.; Federico, L.; Zhao, W.; Dennison, J.; Sarkar, T.R.; Zhang, F.; Takiar, V.; Cheng, K.W.; Mani, S.; Lee, J.S.; et al. Rab25 acts as an oncogene in luminal B breast cancer and is causally associated with Snail driven EMT. Oncotarget 2016, 7, 40252–40265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Díaz, J.; Mendoza, P.; Ortiz, R.; Díaz, N.; Leyton, L.; Stupack, D.; Quest, A.F.; Torres, V.A. Rab5 is required in metastatic cancer cells for Caveolin-1-enhanced Rac1 activation, migration and invasion. J. Cell Sci. 2014, 127, 2401–2406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Wandinger-Ness, A.; Zerial, M. Rab proteins and the compartmentalization of the endosomal system. Cold Spring Harb. Perspect. Biol. 2014, 6, a022616. [Google Scholar] [CrossRef]
  59. Fox, E.J.; Prindle, M.J.; Loeb, L.A. Do mutator mutations fuel tumorigenesis? Cancer Metastasis Rev. 2013, 32, 353–361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Cha, D.J.; Franklin, J.L.; Dou, Y.; Liu, Q.; Higginbotham, J.N.; Beckler, M.D.; Weaver, A.M.; Vickers, K.; Prasad, N.; Levy, S.; et al. KRAS-dependent sorting of miRNA to exosomes. eLife 2015, 4, e07197. [Google Scholar] [CrossRef]
  61. Beckler, M.D.; Higginbotham, J.N.; Franklin, J.L.; Ham, A.J.; Halvey, P.J.; Imasuen, I.E.; Whitwell, C.; Li, M.; Liebler, D.C.; Coffey, R.J. Proteomic analysis of exosomes from mutant KRAS colon cancer cells identifies intercellular transfer of mutant KRAS. Mol. Cell Proteom. 2013, 12, 343–355. [Google Scholar] [CrossRef] [Green Version]
  62. Dou, Y.; Cha, D.J.; Franklin, J.L.; Higginbotham, J.N.; Jeppesen, D.K.; Weaver, A.M.; Prasad, N.; Levy, S.; Coffey, R.J.; Patton, J.G.; et al. Circular RNAs are down-regulated in KRAS mutant colon cancer cells and can be transferred to exosomes. Sci. Rep. 2016, 6, 37982. [Google Scholar] [CrossRef] [PubMed]
  63. Hinger, S.A.; Cha, D.J.; Franklin, J.L.; Higginbotham, J.N.; Dou, Y.; Ping, J.; Shu, L.; Prasad, N.; Levy, S.; Zhang, B.; et al. Diverse Long RNAs Are Differentially Sorted into Extracellular Vesicles Secreted by Colorectal Cancer Cells. Cell Rep. 2018, 25, 715–725.e4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Hinger, S.A.; Abner, J.J.; Franklin, J.L.; Jeppesen, D.K.; Coffey, R.J.; Patton, J.G. Rab13 regulates sEV secretion in mutant KRAS colorectal cancer cells. Sci. Rep. 2020, 10, 15804. [Google Scholar] [CrossRef]
  65. Zhu, Y.F.; Yu, B.H.; Li, D.L.; Ke, H.L.; Guo, X.Z.; Xiao, X.Y. PI3K expression and PIK3CA mutations are related to colorectal cancer metastases. World J. Gastroenterol. 2012, 18, 3745–3751. [Google Scholar] [CrossRef]
  66. Chen, Z.; Wang, C.; Dong, H.; Wang, X.; Gao, F.; Zhang, S.; Zhang, X. Aspirin has a better effect on PIK3CA mutant colorectal cancer cells by PI3K/Akt/Raptor pathway. Mol. Med. 2020, 26, 14. [Google Scholar] [CrossRef] [Green Version]
  67. Théry, C.; Zitvogel, L.; Amigorena, S. Exosomes: Composition, biogenesis and function. Nat. Rev. Immunol. 2002, 2, 569–579. [Google Scholar] [CrossRef]
  68. Xie, Y.H.; Chen, Y.X.; Fang, J.Y. Comprehensive review of targeted therapy for colorectal cancer. Signal. Transduct. Target. Ther. 2020, 5, 22. [Google Scholar] [CrossRef] [PubMed]
  69. Peng, Z.Y.; Gu, R.H.; Yan, B. Downregulation of exosome-encapsulated miR-548c-5p is associated with poor prognosis in colorectal cancer. J. Cell Biochem. 2018. [Google Scholar] [CrossRef]
  70. Yan, S.; Liu, G.; Jin, C.; Wang, Z.; Duan, Q.; Xu, J.; Xu, D. MicroRNA-6869-5p acts as a tumor suppressor via targeting TLR4/NF-κB signaling pathway in colorectal cancer. J. Cell Physiol. 2018, 233, 6660–6668. [Google Scholar] [CrossRef]
  71. Zou, S.L.; Chen, Y.L.; Ge, Z.Z.; Qu, Y.Y.; Cao, Y.; Kang, Z.X. Downregulation of serum exosomal miR-150-5p is associated with poor prognosis in patients with colorectal cancer. Cancer Biomark. 2019, 26, 69–77. [Google Scholar] [CrossRef]
  72. Lu, Y.; Sha, H.; Sun, X.; Zhang, Y.; Wu, Y.; Zhang, J.; Zhang, H.; Wu, J.; Feng, J. CRNDE: An oncogenic long non-coding RNA in cancers. Cancer Cell Int. 2020, 20, 162. [Google Scholar] [CrossRef]
  73. Liu, T.; Zhang, X.; Yang, Y.M.; Du, L.T.; Wang, C.X. Increased expression of the long noncoding RNA CRNDE-h indicates a poor prognosis in colorectal cancer, and is positively correlated with IRX5 mRNA expression. Onco Targets Ther. 2016, 9, 1437–1448. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Cheng, K.; Zhao, Z.; Wang, G.; Wang, J.; Zhu, W. lncRNA GAS5 inhibits colorectal cancer cell proliferation via the miR-182-5p/FOXO3a axis. Oncol. Rep. 2018, 40, 2371–2380. [Google Scholar] [CrossRef] [PubMed]
  75. Deng, X.; Ruan, H.; Zhang, X.; Xu, X.; Zhu, Y.; Peng, H.; Zhang, X.; 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. [Google Scholar] [CrossRef]
  76. Yu, Y.; Nangia-Makker, P.; Farhana, L.; Majumdar, A.P.N. A novel mechanism of lncRNA and miRNA interaction: CCAT2 regulates miR-145 expression by suppressing its maturation process in colon cancer cells. Mol. Cancer 2017, 16, 155. [Google Scholar] [CrossRef]
  77. Yan, Y.; Su, M.; Qin, B. CircHIPK3 promotes colorectal cancer cells proliferation and metastasis via modulating of miR-1207-5p/FMNL2 signal. Biochem. Biophys. Res. Commun. 2020, 524, 839–846. [Google Scholar] [CrossRef] [PubMed]
  78. Zeng, K.; Chen, X.; Xu, M.; Liu, X.; Hu, X.; Xu, T.; Sun, H.; Pan, Y.; He, B.; Wang, S. CircHIPK3 promotes colorectal cancer growth and metastasis by sponging miR-7. Cell Death Dis. 2018, 9, 417. [Google Scholar] [CrossRef] [PubMed]
  79. Yang, H.; Zhang, H.; Yang, Y.; Wang, X.; Deng, T.; Liu, R.; Ning, T.; Bai, M.; Li, H.; Zhu, K.; et al. Hypoxia induced exosomal circRNA promotes metastasis of Colorectal Cancer via targeting GEF-H1/RhoA axis. Theranostics 2020, 10, 8211–8226. [Google Scholar] [CrossRef]
  80. Hon, K.W.; Ab-Mutalib, N.S.; Abdullah, N.M.A.; Jamal, R.; Abu, N. Extracellular Vesicle-derived circular RNAs confers chemoresistance in Colorectal cancer. Sci. Rep. 2019, 9, 16497. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Campanella, C.; Rappa, F.; Sciumè, C.; Gammazza, A.M.; Barone, R.; Bucchieri, F.; David, S.; Curcurù, G.; Bavisotto, C.C.; Pitruzzella, A.; et al. Heat shock protein 60 levels in tissue and circulating exosomes in human large bowel cancer before and after ablative surgery. Cancer 2015, 121, 3230–3239. [Google Scholar] [CrossRef]
  82. Dong, L.; Lin, W.; Qi, P.; Xu, M.D.; Wu, X.; Ni, S.; Huang, D.; Weng, W.W.; Tan, C.; Sheng, W.; et al. Circulating Long RNAs in Serum Extracellular Vesicles: Their Characterization and Potential Application as Biomarkers for Diagnosis of Colorectal Cancer. Cancer Epidemiol. Biomark. Prev. 2016, 25, 1158–1166. [Google Scholar] [CrossRef] [Green Version]
  83. Hu, Y.B.; Yan, C.; Mu, L.; Mi, Y.L.; Zhao, H.; Hu, H.; Li, X.L.; Tao, D.D.; Wu, Y.Q.; Gong, J.P.; et al. Exosomal Wnt-induced dedifferentiation of colorectal cancer cells contributes to chemotherapy resistance. Oncogene 2019, 38, 1951–1965. [Google Scholar] [CrossRef] [Green Version]
  84. Li, J.; Chen, Y.; Guo, X.; Zhou, L.; Jia, Z.; Peng, Z.; Tang, Y.; Liu, W.; Zhu, B.; Wang, L.; et al. GPC1 exosome and its regulatory miRNAs are specific markers for the detection and target therapy of colorectal cancer. J. Cell Mol. Med. 2017, 21, 838–847. [Google Scholar] [CrossRef]
  85. Kumara, H.M.S.; Grieco, M.J.; Caballero, O.L.; Su, T.; Ahmed, A.; Ritter, E.; Gnjatic, S.; Cekic, V.; Old, L.J.; Simpson, A.J.; et al. MAGE-A3 is highly expressed in a subset of colorectal cancer patients. Cancer Immun. 2012, 12, 16. [Google Scholar]
  86. Sun, B.; Li, Y.; Zhou, Y.; Ng, T.K.; Zhao, C.; Gan, Q.; Gu, X.; Xiang, J. Circulating exosomal CPNE3 as a diagnostic and prognostic biomarker for colorectal cancer. J. Cell Physiol. 2019, 234, 1416–1425. [Google Scholar] [CrossRef] [PubMed]
  87. Sun, B.; Zhou, Y.; Fang, Y.; Li, Z.; Gu, X.; Xiang, J. Colorectal cancer exosomes induce lymphatic network remodeling in lymph nodes. Int. J. Cancer 2019, 145, 1648–1659. [Google Scholar] [CrossRef] [PubMed]
  88. Tian, Y.; Ma, L.; Gong, M.; Su, G.; Zhu, S.; Zhang, W.; Wang, S.; Li, Z.; Chen, C.; Li, L.; et al. Protein Profiling and Sizing of Extracellular Vesicles from Colorectal Cancer Patients via Flow Cytometry. ACS Nano 2018, 12, 671–680. [Google Scholar] [CrossRef]
  89. Wang, Y.; Yin, K.; Tian, J.; Xia, X.; Ma, J.; Tang, X.; Xu, H.; Wang, S. Granulocytic Myeloid-Derived Suppressor Cells Promote the Stemness of Colorectal Cancer Cells through Exosomal S100A9. Adv. Sci. 2019, 6, 1901278. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Wu, B.; Sun, D.; Ma, L.; Deng, Y.; Zhang, S.; Dong, L.; Chen, S. Exosomes isolated from CAPS1-overexpressing colorectal cancer cells promote cell migration. Oncol. Rep. 2019, 42, 2528–2536. [Google Scholar] [CrossRef]
  91. Xiao, Y.; Li, Y.; Yuan, Y.; Liu, B.; Pan, S.; Liu, Q.; Qi, X.; Zhou, H.; Dong, W.; Jia, L. The potential of exosomes derived from colorectal cancer as a biomarker. Clin. Chim. Acta 2019, 490, 186–193. [Google Scholar] [CrossRef] [PubMed]
  92. Yokoyama, S.; Takeuchi, A.; Yamaguchi, S.; Mitani, Y.; Watanabe, T.; Matsuda, K.; Hotta, T.; Shively, J.E.; Yamaue, H. Clinical implications of carcinoembryonic antigen distribution in serum exosomal fraction-Measurement by ELISA. PLoS ONE 2017, 12, e0183337. [Google Scholar] [CrossRef]
  93. Zhong, M.E.; Chen, Y.; Xiao, Y.; Xu, L.; Zhang, G.; Lu, J.; Qiu, H.; Ge, W.; Wu, B. Serum extracellular vesicles contain SPARC and LRG1 as biomarkers of colon cancer and differ by tumour primary location. EBioMedicine 2019, 50, 211–223. [Google Scholar] [CrossRef] [Green Version]
  94. Ogata-Kawata, H.; Izumiya, M.; Kurioka, D.; Honma, Y.; Yamada, Y.; Furuta, K.; Gunji, T.; Ohta, H.; Okamoto, H.; Sonoda, H.; et al. Circulating exosomal microRNAs as biomarkers of colon cancer. PLoS ONE 2014, 9, e92921. [Google Scholar] [CrossRef]
  95. Wang, J.; Yan, F.; Zhao, Q.; Zhan, F.; Wang, R.; Wang, L.; Zhang, Y.; Huang, X. Circulating exosomal miR-125a-3p as a novel biomarker for early-stage colon cancer. Sci. Rep. 2017, 7, 4150. [Google Scholar] [CrossRef] [Green Version]
  96. Fu, F.; Jiang, W.; Zhou, L.; Chen, Z. Circulating Exosomal miR-17-5p and miR-92a-3p Predict Pathologic Stage and Grade of Colorectal Cancer. Transl. Oncol. 2018, 11, 221–232. [Google Scholar] [CrossRef] [PubMed]
  97. Bjørnetrø, T.; Redalen, K.R.; Meltzer, S.; Thusyanthan, N.S.; Samiappan, R.; Jegerschöld, C.; Handeland, K.R.; Ree, A.H. An experimental strategy unveiling exosomal microRNAs 486-5p, 181a-5p and 30d-5p from hypoxic tumour cells as circulating indicators of high-risk rectal cancer. J. Extracell. Vesicles 2019, 8, 1567219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Matsumura, T.; Sugimachi, K.; Iinuma, H.; Takahashi, Y.; Kurashige, J.; Sawada, G.; Ueda, M.; Uchi, R.; Ueo, H.; Takano, Y.; et al. Exosomal microRNA in serum is a novel biomarker of recurrence in human colorectal cancer. Br. J. Cancer 2015, 113, 275–281. [Google Scholar] [CrossRef] [PubMed]
  99. Teng, Y.; Ren, Y.; Hu, X.; Mu, J.; Samykutty, A.; Zhuang, X.; Deng, Z.; Kumar, A.; Zhang, L.; Merchant, M.L.; et al. MVP-mediated exosomal sorting of miR-193a promotes colon cancer progression. Nat. Commun. 2017, 8, 14448. [Google Scholar] [CrossRef]
  100. Ren, D.; Lin, B.; Zhang, X.; Peng, Y.; Ye, Z.; Ma, Y.; Liang, Y.; Cao, L.; Li, X.; Li, R.; et al. Maintenance of cancer stemness by miR-196b-5p contributes to chemoresistance of colorectal cancer cells via activating STAT3 signaling pathway. Oncotarget 2017, 8, 49807–49823. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Takano, Y.; Masuda, T.; Iinuma, H.; Yamaguchi, R.; Sato, K.; Tobo, T.; Hirata, H.; Kuroda, Y.; Nambara, S.; Hayashi, N.; et al. Circulating exosomal microRNA-203 is associated with metastasis possibly via inducing tumor-associated macrophages in colorectal cancer. Oncotarget 2017, 8, 78598–78613. [Google Scholar] [CrossRef] [Green Version]
  102. Bigagli, E.; Luceri, C.; Guasti, D.; Cinci, L. Exosomes secreted from human colon cancer cells influence the adhesion of neighboring metastatic cells: Role of microRNA-210. Cancer Biol. Ther. 2016, 17, 1062–1069. [Google Scholar] [CrossRef] [Green Version]
  103. Jin, G.; Liu, Y.; Zhang, J.; Bian, Z.; Yao, S.; Fei, B.; Zhou, L.; Yin, Y.; Huang, Z. A panel of serum exosomal microRNAs as predictive markers for chemoresistance in advanced colorectal cancer. Cancer Chemother. Pharmacol. 2019, 84, 315–325. [Google Scholar] [CrossRef]
  104. Karimi, N.; Ali Feizi, M.H.; Safaralizadeh, R.; Hashemzadeh, S.; Baradaran, B.; Shokouhi, B.; Teimourian, S. Serum overexpression of miR-301a and miR-23a in patients with colorectal cancer. J. Chin. Med. Assoc. 2019, 82, 215–220. [Google Scholar] [CrossRef] [PubMed]
  105. Zeng, Z.; Li, Y.; Pan, Y.; Lan, X.; Song, F.; Sun, J.; Zhou, K.; Liu, X.; Ren, X.; Wang, F.; et al. Cancer-derived exosomal miR-25-3p promotes pre-metastatic niche formation by inducing vascular permeability and angiogenesis. Nat. Commun. 2018, 9, 5395. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Liu, X.; Chen, X.; Zeng, K.; Xu, M.; He, B.; Pan, Y.; Sun, H.; Pan, B.; Xu, X.; Xu, T.; et al. DNA-methylation-mediated silencing of miR-486-5p promotes colorectal cancer proliferation and migration through activation of PLAGL2/IGF2/β-catenin signal pathways. Cell Death Dis. 2018, 9, 1037. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Yan, S.; Han, B.; Gao, S.; Wang, X.; Wang, Z.; Wang, F.; Zhang, J.; Xu, D.; Sun, B. Exosome-encapsulated microRNAs as circulating biomarkers for colorectal cancer. Oncotarget 2017, 8, 60149–60158. [Google Scholar] [CrossRef] [Green Version]
  108. Yan, S.; Jiang, Y.; Liang, C.; Cheng, M.; Jin, C.; Duan, Q.; Xu, D.; Yang, L.; Zhang, X.; Ren, B.; et al. Exosomal miR-6803-5p as potential diagnostic and prognostic marker in colorectal cancer. J. Cell Biochem. 2018, 119, 4113–4119. [Google Scholar] [CrossRef]
  109. Hu, J.L.; Wang, W.; Lan, X.L.; Zeng, Z.C.; Liang, Y.S.; Yan, Y.R.; Song, F.Y.; Wang, F.F.; Zhu, X.H.; Liao, W.J.; et al. CAFs secreted exosomes promote metastasis and chemotherapy resistance by enhancing cell stemness and epithelial-mesenchymal transition in colorectal cancer. Mol. Cancer 2019, 18, 91. [Google Scholar] [CrossRef] [Green Version]
  110. Min, L.; Zhu, S.; Chen, L.; Liu, X.; Wei, R.; Zhao, L.; Yang, Y.; Zhang, Z.; Kong, G.; Li, P.; et al. Evaluation of circulating small extracellular vesicles derived miRNAs as biomarkers of early colon cancer: A comparison with plasma total miRNAs. J. Extracell. Vesicles 2019, 8, 1643670. [Google Scholar] [CrossRef] [Green Version]
  111. Barbagallo, C.; Brex, D.; Caponnetto, A.; Cirnigliaro, M.; Scalia, M.; Magnano, A.; Caltabiano, R.; Barbagallo, D.; Biondi, A.; Cappellani, A.; et al. LncRNA UCA1, Upregulated in CRC Biopsies and Downregulated in Serum Exosomes, Controls mRNA Expression by RNA-RNA Interactions. Mol. Ther. Nucleic Acids 2018, 12, 229–241. [Google Scholar] [CrossRef] [Green Version]
  112. Wang, Y.; Zhang, H.; Wang, J.; Li, B.; Wang, X. Circular RNA expression profile of lung squamous cell carcinoma: Identification of potential biomarkers and therapeutic targets. Biosci. Rep. 2020, 40. [Google Scholar] [CrossRef] [Green Version]
  113. Pan, B.; Qin, J.; Liu, X.; He, B.; Wang, X.; Pan, Y.; Sun, H.; Xu, T.; Xu, M.; Chen, X.; et al. Identification of Serum Exosomal hsa-circ-0004771 as a Novel Diagnostic Biomarker of Colorectal Cancer. Front. Genet. 2019, 10, 1096. [Google Scholar] [CrossRef] [Green Version]
  114. Wang, Y.; Liu, J.; Ma, J.; Sun, T.; Zhou, Q.; Wang, W.; Wang, G.; Wu, P.; Wang, H.; Jiang, L.; et al. Exosomal circRNAs: Biogenesis, effect and application in human diseases. Mol. Cancer 2019, 18, 116. [Google Scholar] [CrossRef]
  115. 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. [Google Scholar] [CrossRef] [PubMed]
  116. Liu, L.; Meng, T.; Yang, X.H.; Sayim, P.; Lei, C.; Jin, B.; Ge, L.; Wang, H.J. 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. [Google Scholar] [CrossRef]
  117. 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. [Google Scholar] [CrossRef] [PubMed]
  118. 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. [Google Scholar] [CrossRef] [PubMed]
  119. Ling, H.; Spizzo, R.; Atlasi, Y.; Nicoloso, M.; Shimizu, M.; Redis, R.S.; Nishida, N.; Gafà, R.; Song, J.; Guo, Z.; et al. CCAT2, a novel noncoding RNA mapping to 8q24, underlies metastatic progression and chromosomal instability in colon cancer. Genome Res. 2013, 23, 1446–1461. [Google Scholar] [CrossRef] [Green Version]
  120. 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. [Google Scholar] [CrossRef] [PubMed]
  121. Ogunwobi, O.O.; Mahmood, F.; Akingboye, A. Biomarkers in Colorectal Cancer: Current Research and Future Prospects. Int. J. Mol. Sci. 2020, 21, 5311. [Google Scholar] [CrossRef]
  122. Wu, H.; Wei, M.; Jiang, X.; Tan, J.; Xu, W.; Fan, X.; Zhang, R.; Ding, C.; Zhao, F.; Shao, X.; et al. lncRNA PVT1 Promotes Tumorigenesis of Colorectal Cancer by Stabilizing miR-16-5p and Interacting with the VEGFA/VEGFR1/AKT Axis. Mol. Ther. Nucleic Acids 2020, 20, 438–450. [Google Scholar] [CrossRef]
  123. Del Vecchio, F.; Martinez-Rodriguez, V.; Schukking, M.; Cocks, A.; Broseghini, E.; Fabbri, M. Professional killers: The role of extracellular vesicles in the reciprocal interactions between natural killer, CD8+ cytotoxic T-cells and tumour cells. J. Extracell. Vesicles 2021, 10, e12075. [Google Scholar] [CrossRef] [PubMed]
  124. Yan, W.; Jiang, S. Immune Cell-Derived Exosomes in the Cancer-Immunity Cycle. Trends Cancer 2020, 6, 506–517. [Google Scholar] [CrossRef]
  125. Mittal, S.; Gupta, P.; Chaluvally-Raghavan, P.; Pradeep, S. Emerging Role of Extracellular Vesicles in Immune Regulation and Cancer Progression. Cancers 2020, 12, 3563. [Google Scholar] [CrossRef]
  126. Lan, J.; Sun, L.; Xu, F.; Liu, L.; Hu, F.; Song, D.; Hou, Z.; Wu, W.; Luo, X.; Wang, J.; et al. M2 Macrophage-Derived Exosomes Promote Cell Migration and Invasion in Colon Cancer. Cancer Res. 2019, 79, 146–158. [Google Scholar] [CrossRef] [Green Version]
  127. Mannavola, F.; Salerno, T.; Passarelli, A.; Tucci, M.; Internò, V.; Silvestris, F. Revisiting the Role of Exosomes in Colorectal Cancer: Where Are We Now? Front. Oncol. 2019, 9, 521. [Google Scholar] [CrossRef]
  128. Milane, L.; Singh, A.; Mattheolabakis, G.; Suresh, M.; Amiji, M.M. Exosome mediated communication within the tumor microenvironment. J. Control. Release 2015, 219, 278–294. [Google Scholar] [CrossRef]
  129. Sun, Z.; Yang, S.; Zhou, Q.; Wang, G.; Song, J.; Li, Z.; Zhang, Z.; Xu, J.; Xia, K.; Chang, Y.; et al. Emerging role of exosome-derived long non-coding RNAs in tumor microenvironment. Mol. Cancer 2018, 17, 82. [Google Scholar] [CrossRef] [PubMed]
  130. Ohshima, K.; Inoue, K.; Fujiwara, A.; Hatakeyama, K.; Kanto, K.; Watanabe, Y.; Muramatsu, K.; Fukuda, Y.; Ogura, S.; Yamaguchi, K.; 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, e13247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Ji, H.; Chen, M.; Greening, D.W.; He, W.; Rai, A.; Zhang, W.; Simpson, R.J. Deep sequencing of RNA from three different extracellular vesicle (EV) subtypes released from the human LIM1863 colon cancer cell line uncovers distinct miRNA-enrichment signatures. PLoS ONE 2014, 9, e110314. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Li, S.; Li, Y.; Chen, B.; Zhao, J.; Yu, S.; Tang, Y.; Zheng, Q.; Li, Y.; Wang, P.; He, X.; et al. exoRBase: A database of circRNA, lncRNA and mRNA in human blood exosomes. Nucleic Acids Res. 2018, 46, D106–D112. [Google Scholar] [CrossRef] [Green Version]
  133. Mizoguchi, A.; Takayama, A.; Arai, T.; Kawauchi, J.; Sudo, H. MicroRNA-8073: Tumor suppressor and potential therapeutic treatment. PLoS ONE 2018, 13, e0209750. [Google Scholar] [CrossRef]
  134. Kyuno, D.; Bauer, N.; Schnölzer, M.; Provaznik, J.; Ryschich, E.; Hackert, T.; Zöller, M. Distinct Origin of Claudin7 in Early Tumor Endosomes Affects Exosome Assembly. Int. J. Biol. Sci. 2019, 15, 2224–2239. [Google Scholar] [CrossRef] [PubMed]
  135. Sun, H.; Rana, S.; Wang, Z.; Zhao, K.; Schnölzer, M.; Provaznik, J.; Hackert, T.; Lv, Q.; Zöller, M. The Pancreatic Cancer-Initiating Cell Marker CD44v6 Affects Transcription, Translation, and Signaling: Consequences for Exosome Composition and Delivery. J. Oncol. 2019, 2019, 3516973. [Google Scholar] [CrossRef] [Green Version]
  136. Tubita, V.; Segui-Barber, J.; Lozano, J.J.; Banon-Maneus, E.; Rovira, J.; Cucchiari, D.; Moya-Rull, D.; Oppenheimer, F.; Del Portillo, H.; Campistol, J.M.; et al. Effect of immunosuppression in miRNAs from extracellular vesicles of colorectal cancer and their influence on the pre-metastatic niche. Sci. Rep. 2019, 9, 11177. [Google Scholar] [CrossRef] [Green Version]
  137. Jeppesen, D.K.; Fenix, A.M.; Franklin, J.L.; Higginbotham, J.N.; Zhang, Q.; Zimmerman, L.J.; Liebler, D.C.; Ping, J.; Liu, Q.; Evans, R.; et al. Reassessment of Exosome Composition. Cell 2019, 177, 428–445.e18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Yu, X.; He, L.; Pentok, M.; Yang, H.; Yang, Y.; Li, Z.; He, N.; Deng, Y.; Li, S.; Liu, T.; et al. An aptamer-based new method for competitive fluorescence detection of exosomes. Nanoscale 2019, 11, 15589–15595. [Google Scholar] [CrossRef]
  139. De Voogt, W.S.; Tanenbaum, M.E.; Vader, P. Illuminating RNA trafficking and functional delivery by extracellular vesicles. Adv. Drug Deliv. Rev. 2021, 174, 250–264. [Google Scholar] [CrossRef] [PubMed]
  140. Yao, X.; Lyu, P.; Yoo, K.; Yadav, M.K.; Singh, R.; Atala, A.; Lu, B. Engineered extracellular vesicles as versatile ribonucleoprotein delivery vehicles for efficient and safe CRISPR genome editing. J. Extracell. Vesicles 2021, 10, e12076. [Google Scholar] [CrossRef] [PubMed]
  141. Zhuang, J.; Tan, J.; Wu, C.; Zhang, J.; Liu, T.; Fan, C.; Li, J.; Zhang, Y. Extracellular vesicles engineered with valency-controlled DNA nanostructures deliver CRISPR/Cas9 system for gene therapy. Nucleic Acids Res. 2020, 48, 8870–8882. [Google Scholar] [CrossRef] [PubMed]
  142. Xu, R.; Greening, D.W.; Zhu, H.J.; Takahashi, N.; Simpson, R.J. Extracellular vesicle isolation and characterization: Toward clinical application. J. Clin. Investig. 2016, 126, 1152–1162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Dai, S.; Wei, D.; Wu, Z.; Zhou, X.; Wei, X.; Huang, H.; Li, G. Phase I clinical trial of autologous ascites-derived exosomes combined with GM-CSF for colorectal cancer. Mol. Ther. 2008, 16, 782–790. [Google Scholar] [CrossRef] [PubMed]
  144. Allelein, S.; Medina-Perez, P.; Lopes, A.L.H.; Rau, S.; Hause, G.; Kölsch, A.; Kuhlmeier, D. Potential and challenges of specifically isolating extracellular vesicles from heterogeneous populations. Sci. Rep. 2021, 11, 11585. [Google Scholar] [CrossRef] [PubMed]
  145. Lener, T.; Gimona, M.; Aigner, L.; Börger, V.; Buzas, E.; Camussi, G.; Chaput, N.; Chatterjee, D.; Court, F.A.; Del Portillo, H.A.; et al. Applying extracellular vesicles based therapeutics in clinical trials—An ISEV position paper. J. Extracell. Vesicles 2015, 4, 30087. [Google Scholar] [CrossRef]
Figure 1. The classification of extracellular vesicles (EVs), the process of exocytosis, and components of EVs. Members of the RAB family regulate different EVs (early/late endosome, recycling endosome, autophagosome, etc.). EVs embed various substances and anchor receptors and factors on the membrane.
Figure 1. The classification of extracellular vesicles (EVs), the process of exocytosis, and components of EVs. Members of the RAB family regulate different EVs (early/late endosome, recycling endosome, autophagosome, etc.). EVs embed various substances and anchor receptors and factors on the membrane.
Biomedicines 09 00931 g001
Figure 2. Current exosomal materials and application aspects. DNA, RNA, protein, and metabolites can be extracted from EVs in cancer cells, and potential clinical events (risk assessment, detection, diagnosis, and prognosis) can be analyzed. In animal models, EVs can be used to carry drugs as a strategy for biomedical imaging and therapy.
Figure 2. Current exosomal materials and application aspects. DNA, RNA, protein, and metabolites can be extracted from EVs in cancer cells, and potential clinical events (risk assessment, detection, diagnosis, and prognosis) can be analyzed. In animal models, EVs can be used to carry drugs as a strategy for biomedical imaging and therapy.
Biomedicines 09 00931 g002
Table 1. Components of ESCRT complexes and regulators in colorectal cancer exocytosis.
Table 1. Components of ESCRT complexes and regulators in colorectal cancer exocytosis.
ComplexGene SymbolHazard RatioExpressionRef.
ESCRT-0VPS27 (HRS/HGS)Uni: 2.27
Multi: 3.34
Up[36]
STAM1/2----Up[37]
ESCRT-IVPS23 (TSG101)----Up[38]
VPS28-------------
VPS37A/B/C/D----Down[39]
MVB12A/B------------
UBAP1----Down[40]
ESCRT-IIVPS22 (SNF8, EAP30)-------------
VPS25 (EAP20)-------------
VPS36 (EAP45)-------------
ESCRT-IIIVPS2A/B (CHMP2A/B)----Up[41]
VPS20 (CHMP6)-------------
VPS24 (CHMP3)-------------
SNF7A/B/C (CHMP4A/B/C)----Down[42]
VPS60 (CHMP5)----Up[43]
DID2A/B (CHMP7, CHMP1A/B)----Down[42]
IST1 (OLC1)Uni: 10.43
Multi: 7.9
Up[44]
Vps4-Vta1VPS4A/B (SKD1)----Down[45]
VTA1 (LIP5)------------
Bro1/ALIXALIX (PDCD6IP)----Down[46]
RABsRAB3Uni: 2.58
Multi: 2.39
Up[47]
RAB8----Up[48]
RAB26----Up[47]
RAB27Multi: 0.45Up
Down
[49]
[50]
Table 2. Vesicle-related substances carried by colorectal cancer.
Table 2. Vesicle-related substances carried by colorectal cancer.
CategoryNameExpressFunctionRef.
miRNAmiR-1246UpDiagnosis[94]
miR-125a-3pUpDiagnosis[95]
miR-150-5pDownDiagnosis[71]
miR-17-5p/miR-92a-3pUpMetastasis[96]
miR-181a-5pDownMetastasis[97]
miR-19aUpRecurrence[98]
miR-193aUpMetastasis[99]
miR-196b-5pUpChemoresistance[100]
miR-203UpMetastasis[101]
miR-210UpChemoresistance/metastasis[102]
miR-21UpRecurrence/chemoresistance/metastasis[94]
miR-21-5p/miR-1246/miR-96-5p/miR-1229-5pUpChemoresistance[103]
miR-23aUpDiagnosis[94,104]
miR-25-3pUpMetastasis[105]
miR-30d-5pUpMetastasis[97]
miR-301aUpDiagnosis[104]
miR-486-5pUpDiagnosis[106]
miR-548c-5pDownMetastasis[69]
miR-638DownMetastasis[107]
miR-6803-5pUpDiagnosis[108]
miR-6869-5pDownMetastasis[70]
miR-92a-3pUpChemoresistance[109]
Let-7b-3p/miR-139-3p/miR-145-3pUpDiagnosis[110]
circRNAcircHIPK3UpDiagnosis/metastasis[78,111]
circ-133UpMetastasis[79]
ciRS-122UpChemoresistance[112]
hsa-circ_0004771UpDiagnosis[113]
hsa-circ_0000338UpChemoresistance[80]
circRTN4UpChemoresistance[114]
lncRNAlncRNA CRNDE-hUpRecurrence/chemoresistance/
metastasis/diagnosis
[72,115]
lncRNA GAS5UpDiagnosis[116]
LNCV6_116109UPDiagnosis[117]
LNCV6_98390UpDiagnosis[117]
LNCV6_84003UPDiagnosis[117]
LNCV6_98602UpDiagnosis[117]
LNCV_108266UpDiagnosis[117]
LNCV6_38772UpDiagnosis[117]
lncRNA 91HUpRecurrence[118]
lncRNA CCAT2UpDiagnosis[119]
lncRNA H19UpChemoresistance[120]
lncRNA CCALUpChemoresistance[75]
lncRNA PVT1UpMetastasis[121,122]
ProteinHsp60UpDiagnosis/proliferation[81]
GPC1UpDiagnosis/metastasis[84]
CD147UpDiagnosis[88]
CPNE3UpDiagnosis[86]
TAG72UpChemoresistance[91]
S100A9UpRecurrence[89]
SPARCUpDiagnosis/angiogenesis[93]
LRG1UpDiagnosis[93]
CEAUpDiagnosis/metastasis[92]
IRF-2UpMetastasis[87]
WntUpChemoresistance[83]
CAPS1UpMetastasis[90]
MAGEA3UpDiagnosis/metastasis[82,85]
Table 3. Available microarray chips and RNA-seq datasets of exosomal research in colorectal cancer from the GEO website.
Table 3. Available microarray chips and RNA-seq datasets of exosomal research in colorectal cancer from the GEO website.
Array Chip and RNA-Seq PlatformSpeciesObjectsRef.
GSE21350Agilent-021827. Human miRNA Microarray G4470CHumanCell line
(SW480,SW620)
[130]
GSE39833Agilent-021827. Human miRNA Microarray G4470CHumanCRC patients’ serum[94]
GSE40246Agilent-021827. Human miRNA Microarray G4470CHumanCRC patients’ serum[94]
GSE67004Illumina HiSeq 2000HumanCell line
(DKO-1,DLD-1,DKs-8)
[60]
GSE68979Agilent-019052 Homo sapiens 45KHumanCell line(SW480)N/A
GSE72577Illumina HiSeq 2000HumanCell line
(DKO-1,DLD-1,DKs-8)
[62]
GSE87839Applied Biosystems Taqman Array Human Micro A+B Cards Set v3.0HumanCell line
(LIM1863)
[131]
GSE100063Illumina HiSeq 2000HumanCRC patients’ blood[132]
GSE101950
GSE101951
Illumina HiSeq 2500MusCell line
(CT26)
N/A
GSE114316
GSE114317
GSE114318
3D-Gene Human Oligo Chip 25K V2.13D-Gene Human miRNA V21_1.0.0HumanColorectal xenografts[133]
GSE115114Illumina NextSeq 500HumanCRC patientsN/A
GSE116589Illumina HiSeq 3000HumanCRC patientsN/A
GSE119031Agilent-040150 EMBL-rel18_30rep 031181HumanCell line
(HT-29,SW-948)
[134,135]
GSE119032
GSE119033
Agilent-070156 Human_miRNA_ V21.0_Microarray 046064HumanCell line
(HT-29,SW-948)
[134,135]
GSE1200133D-Gene Human miRNA V21_1.0.0HumanCell line
(HCT116)
N/A
GSE121964Illumina HiSeq 2000HumanCell line
(DKO-1,DLD-1,DKs-8)
[63]
GSE123708
GSE123709
GSE123710
Affymetrix Multispecies miRNA-4 ArrayAffymetrix Clariom S Assay HT, HumanHumanCell line
(HCT116,SW480)
[136]
GSE125905Illumina HiSeq 2000HumanCell line
(DKO-1,Gli36)
[137]
GSE173202Illumina HiSeq 2000MusCell line
(CT26,MC38)
N/A
Table 4. Current clinical trials involve EVs for colorectal cancer.
Table 4. Current clinical trials involve EVs for colorectal cancer.
NumberParticipantsPhaseStatusApplication
NCT0129407235Phase IRecruitingTreatment
NCT0343280680----RecruitingDiagnostic
NCT0439457275----RecruitingDiagnostic
NCT04523389172----RecruitingDiagnostic
NCT04298398108----Not yet recruitingDiagnostic
NCT0439457275----RecruitingDiagnostic
NCT04523389172----RecruitingDiagnostic
NCT0392789840Phase IIRecruitingTreatment
NCT0243900828----TerminatedDiagnostic
NCT0326017960Phase IUnknownTreatment
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Chang, Y.-C.; Chan, M.-H.; Li, C.-H.; Fang, C.-Y.; Hsiao, M.; Chen, C.-L. Exosomal Components and Modulators in Colorectal Cancer: Novel Diagnosis and Prognosis Biomarkers. Biomedicines 2021, 9, 931. https://doi.org/10.3390/biomedicines9080931

AMA Style

Chang Y-C, Chan M-H, Li C-H, Fang C-Y, Hsiao M, Chen C-L. Exosomal Components and Modulators in Colorectal Cancer: Novel Diagnosis and Prognosis Biomarkers. Biomedicines. 2021; 9(8):931. https://doi.org/10.3390/biomedicines9080931

Chicago/Turabian Style

Chang, Yu-Chan, Ming-Hsien Chan, Chien-Hsiu Li, Chih-Yeu Fang, Michael Hsiao, and Chi-Long Chen. 2021. "Exosomal Components and Modulators in Colorectal Cancer: Novel Diagnosis and Prognosis Biomarkers" Biomedicines 9, no. 8: 931. https://doi.org/10.3390/biomedicines9080931

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop