IMR Press / FBL / Volume 28 / Issue 11 / DOI: 10.31083/j.fbl2811302
Open Access Review
The Function, Underlying Mechanism and Clinical Potential of Exosomes in Colorectal Cancer
Show Less
1 Department of Human Anatomy and Histology, School of Basic Medical Sciences, Xinxiang Medical University, 453000 Xinxiang, Henan, China
2 Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, 453000 Xinxiang, Henan, China
3 Department of Pathology, The Third Affiliated Hospital of Xinxiang Medical University, 453000 Xinxiang, Henan, China
4 Micromorphology Laboratory, School of Basic Medical Sciences, Xinxiang Medical University, 453000 Xinxiang, Henan, China
*Correspondence: 052106@xxmu.edu.cn (Yuhan Hu)
These authors contributed equally.
Front. Biosci. (Landmark Ed) 2023, 28(11), 302; https://doi.org/10.31083/j.fbl2811302
Submitted: 15 March 2023 | Revised: 2 June 2023 | Accepted: 8 June 2023 | Published: 24 November 2023
Copyright: © 2023 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Colorectal cancer (CRC) is a lethal malignancy worldwide. Exosomes are extracellular vesicles derived from the endosomal pathway of nearly all cells and can be found in body fluids. They can be considered an intercellular system in the human body that can mediate near- and long-distance intercellular communication due to their features and functions. Investigations have revealed that exosomes are participated in different processes, physiologically and pathologically, especially in cancer. However, the clinical value of exosomes and their mechanisms of action in CRC are unclear and have not been systematically assessed. The purpose of this review is to discuss how exosomes play a role in the occurrence and development of CRC, with a particular focus on the functions and underlying mechanisms of tumor-derived exosomes as well as non-tumor-derived exosomes. We also describe the evidence that exosomes can be used as diagnostic and prognostic markers for CRC. In addition, the possibilities of exosomes in CRC clinical transformation are also discussed.

Keywords
colorectal cancer
exosome
biomarker
diagnosis
clinical transformation
1. Introduction

Worldwide, colorectal cancer (CRC) is a common malignant tumor [1]. According to the Global Cancer Statistics 2020, CRC is the third most commonly diagnosed cancer, the second leading cause of cancer death in women, and the third leading cause of incidence and mortality in men [2]. It is a malignancy originating from the mucosal epithelium and glands of the large intestine. Histologically, there are several subtypes of CRC, and approximately 90% of CRCs are adenocarcinomas [3]. Patients with CRC often present with anemia, blood in the stools, aberrant bowel movement, and weight loss [4]. It has been demonstrated that the occurrence of CRC is related to dietary patterns, metabolism, and inflammation [5]. The initiation and progression of CRC may also be affected by environmental and genetic factors [6]. Thus, CRC is the result of a complex multi-step process involving numerous factors and genes. For patients with CRC, the systemic treatment regimens utilized for clinical management include tumor resection, 5-Fluorouracil-based chemotherapy, radiotherapy, anti-angiogenic treatment, targeted therapy, and immunotherapy [7, 8, 9]. Although the death rate of CRC has decreased due to earlier screenings and improved treatments, more than one-third of patients die within 5 years of initial diagnosis, with liver metastases being the most fatal cause of death [10, 11, 12].

Exosomes are extracellular vesicles (EVs) with diameters ranging from 40 to 160 nm. They are derived from the endosomal pathway of nearly all cells, and they can be found in cell culture fluids, breast milk, blood, urine, pleural effusion, saliva, and other body fluids [13, 14, 15]. Exosomes carry specific cargo consisting of DNA, RNA, cytosolic and cell-surface proteins, metabolites, and lipids. They can be thought of as an intercellular system in the human body that mediate communication between cells and influence a variety of cell biological behaviors [13, 16]. During the generation of exosomes, the plasma membrane is double invaginated, and intracellular multivesicular bodies (MVBs) containing intraluminal vesicles (ILVs) are formed. MVBs fuse to the plasma membrane, and ILVs are ultimately secreted as exosomes through exocytosis. The uptake of exosomes is not random but depends on the surface proteins on the membrane of exosomes and recipient cells [17]. When exosomes encounter suitable recipient cells, exosomes can adhere to the surface of recipient cells through the interaction of ligands and receptors. They can directly fuse with cell membranes or be endocytosed by recipient cells and release the cargo into target cells (Fig. 1) [13, 14]. Numerous studies have shown that exosomes are involved in different physiological and pathological processes, such as inflammatory responses, cancer development, metastasis, and immunity [17, 18].

Fig. 1.

Biogenesis and intercellular communications of exosomes. The plasma membrane is double invaginated, forming the intracellular multivesicular bodies (MVBs) containing intraluminal vesicles (ILVs). MVBs fuse to the plasma membrane, and ILVs are ultimately secreted as exosomes through exocytosis. The intercellular communications of exosomes depend on the interactions between ligands and receptors. When the exosomes meet the proper recipient cells, they can directly fuse with cell membranes or be endocytosed by recipient cells and release the cargo into target cells.

Recently, it has been demonstrated that exosomes play a role in the occurrence, development, invasion, metastasis, tumor microenvironment (TME) remodeling, chemoresistance, and other processes in CRC [19]. This paper reviews the functions and underlying mechanisms of action of tumor-derived and non-tumor-derived exosomes in CRC, intending to summarize the previous studies on potential biomarkers and effective targets for the treatment of CRC.

2. Physiological and Pathological Functions of Exosomes

Exosomes were first reported as a “type of small vesicles” in 1983, and the name “exosome” was first used in 1989 [20, 21]. They were initially believed to be superfluous membrane vesicles during cell maturation, with the effect of regulating membrane function, removing cellular debris, and eliminating surface molecules [14]. However, exosomes have successfully attracted attention among researchers because of their special roles in multiple facets of cell activity.

Exosomes can be secreted by the donor cells through exocytosis and accepted by the recipient cells through endocytosis or directly fusing to cell membranes. Thus, exosomes act as a medium in intercellular communications and transmit information to a large number of cells and locations, physiologically or pathologically (Fig. 2).

Fig. 2.

Exosomes play a role in physiological and pathological processes. Exosomes are involved in basic physiological processes such as immune response, neural communication, gamete maturation, fertilization, embryo implantation, organ development, cell differentiation and matrix synthesis, melanin synthesis, cell proliferation, homeostasis, and maturation. They are also involved in some pathological processes, such as immune disturbance, tumorigenesis, cancer cell migration and metastasis, angiogenesis, metabolic reprogramming, tumor microenvironment (TME) formation, pathogen infection, neurodegenerative diseases, and cardiovascular disease.

Exosomes are involved in the signal transduction of the nervous system due to reciprocal signal transfer among sensory and motor neurons, interneurons, and glial cells [22]. Moreover, they play an important role in the aging process [23]. Exosomes also play an important role in reproductive development. They are involved in gamete maturation, fertilization, embryo implantation, immunological communication between the mother and the fetus, and fetal protection by regulating local and/or systemic immune responses, organ development, and melanin synthesis [24, 25, 26, 27, 28]. Additionally, exosomes are related to cell proliferation, homeostasis, and maturation, such as in hepatocytes and reticulocytes [20, 29].

Exosomes are involved in immune response and infection [30, 31]. They participate in antigen presentation and induce the activation of T and/or B cells [32]. The contents of exosomes are involved in regulating innate and adaptive immune responses. It has been demonstrated that the DNA of some intracellular bacteria sorted into exosomes is able to activate innate immune responses or lower antibacterial defenses [33].

Numerous studies have demonstrated that exosomes are widely involved in the occurrence and development of cancers. They are a double-edged sword in tumor growth and progression. For example, exosomes derived from M2 macrophages can inhibit the migration and invasion of glioma cells [34]. Alternatively, the impact of tumor exosomal DNA on inflammatory responses can indirectly worsen cancer [35]. In addition, tumor-derived exosomes can promote immune evasion by cancer cells and generate an immunosuppressive microenvironment [17, 36]. Exosomes derived from tumor cells may be absorbed by surrounding cells and transform the microenvironment into one that is prone to tumor development [37]. They also participate in tumor growth, metastasis, apoptosis, metabolic reprogramming, extracellular matrix degradation, stromal reprogramming, immune surveillance escape, and drug resistance [23, 38, 39]. In addition, exosomes are involved in neurodegenerative diseases, cardiovascular disease, and pathogenic infections [14, 40].

3. The Underlying Mechanisms of Exosome Functions in CRC

Numerous studies have explored the relationship between exosomes and the pathogenesis of different cancers. The data demonstrated that exosomes have a relationship with the hallmarks of cancer, such as sustaining proliferation signaling, activating invasion and metastasis, inducing angiogenesis, metabolic reprogramming, and immune evasion [41]. We reviewed the functions and mechanisms of exosomes in CRC, including CRC cell-generated exosomes and non-tumor cell-produced exosomes.

3.1 CRC Tumor-Derived Exosomes

Tumor-derived exosomes (TDEs) were first named in 1981, and it has attracted much attention in the past years [42]. Exosomes act as signal messengers or transducers for communications between cells [43]. Once the recipient cells take up TDEs, the exosomes are internalized, and exosomal contents (such as microRNA, lncRNA, and circRNA) are released. The recipient cells respond to these exosomal contents by changing their phenotypes. This process is finely regulated and specifically determined by complex surface molecules on the extracellular vesicles and the recipient cell membrane [44]. This paper will summarize the functions and mechanisms of tumor-derived exosomes in CRC (Fig. 3; Table 1, Ref. [45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92]).

Fig. 3.

Mechanism of tumor-derived exosomes in colorectal cancer (CRC). Exosomes derived from CRC cells can transport their cargo to other cells and play an important role in the occurrence and progression of CRC, including tumor growth, invasion and metastasis, angiogenesis, immune evasion, TME remodeling, metabolic reprogramming, and therapy resistance.

Table 1.The function and mechanism of tumor-derived exosomes in CRC.
Type Molecule Function Target Ref.
miRNA miR-934 Promote colorectal cancer liver metastasis PTEN [51]
miR-106b Promote migration, invasion, and metastasis PDCD4 [53]
miR-135a-5p Promote CRC liver metastasis kinase 2-yes-associated protein-MMP7 axis [54]
miR-25-3p Facilitate vascular permeability and angiogenesis KLF2, KLF4 [68]
miR-25-3p, miR-130b-3p, miR-425-5p Enhance M2 polarization of macrophages and induce liver metastasis of CRC [95]
miR-208b Treg expansion, oxaliplatin resistance PDCD4 [72]
miR-146a-5p, miR-155-5p Activate CAFs and enhance the invasive capacity of CRC cells SOCS1, ZBTB2 [78]
miR-1246/92b-3p/27a-3p Promote metastasis [57]
miR-21-5p Induce angiogenesis and vascular permeability KRIT1 [69]
miR‐221/222 Promote metastasis SPINT1 [58]
miRNA-335-5p Promote migration, invasion, and metastasis RASA1 [59]
miR-200c-3p Inhibit migration and invasion, and promote apoptosis after LPS stimulation ZEB-1 [60]
miR-46146 OX resistance PDCD10 [89]
miR-106b-3p Promote metastasis DLC-1 [61]
miR-19b Enhance radioresistance and stemness of CRC cell FBXW7 [90]
microRNA-21-5p Induce an inflammatory premetastatic niche TLR7 [63]
miR-1229 Promote angiogenesis HIPK2 [70]
miR-17-5p Promote CRC cell growth and inhibit anti-tumor immunity SPOP [76]
miR‑10a Promote lung metastasis [66]
miR-10b Activate fibroblasts to become CAFs PIK3CA [82]
miR-1255b-5p Suppress EMT and liver metastasis hTERT [67]
miR-224-5p Promote tumor growth CMTM4 [49]
miR-203 Promote the differentiation of monocytes to M2-TAMs [83]
miR-101-3p Be related to metabolic reprogramming, promote tumor growth, migration, and 5-FU resistance HIPK3 [84]
lncRNA RPPH1 Promote metastasis and proliferation TUBB3 [55]
SNHG10 Contribute to immune escape INHBC [73]
lnc-HOXB8-1:2 Lead to TAM infiltration and M2 polarization hsa-miR-6825-5p [74]
CRNDE-h Promote Th17 cell differentiation RORγt [75]
MALAT1 Promote the invasion and metastasis miR-26a/26b [62]
PCAT1 Promote EMT and liver metastasis miR-329-3p [65]
HOTTIP Increase resistance of CRC cells to mitomycin miR-214 [92]
91H Enhance CRC metastasis HNRNPK [49]
HOTAIR Impede anti-tumor immunity PKM2 [77]
circRNA CircLPAR1 Suppress tumor growth eIF3h [45]
CircPACRGL Promote proliferation, invasion, migration, and differentiation of N1 to N2 neutrophils miR-142-3p/miR-506-3p [52]
Circ-133 Promote cell migration GEF-H1 [56]
Circ_0005963 Promote chemoresistance miR-122 [86]
circ_0000338 Improve the chemoresistance miR-217 and miR-485-3p [88]
circ_0094343 Inhibit proliferation, clone formation, and glycolysis miR-766-5p [46]
circ_PTPRA Induce CRC cell cycle arrest and inhibited cell proliferation miR-671-5p [47]
circ-FBXW7 Ameliorate chemoresistance to oxaliplatin in CRC miR-128-3p [91]
circEPB41L2 Suppress CRC cell proliferation, migration, and invasion and promote apoptosis miR-21-5p, miR-942-5p [48]
Others HSPC111 Altered lipid metabolism of CAFs and promote liver metastasis ACLY [79]
p-STAT3 Promote 5-FU resistance [87]
ANGPTL1 Attenuate CRC liver metastasis MMP9 [80]
CXCL16 Promote metastasis [57]
Wnt4 Enhance migration and invasion [64]
IRF-2 Remodel the lymphatic network and promote metastasis VEGFC [70]
CXCL1, CXCL2 Attract CRCSC-primed neutrophils to promote tumorigenesis of CRC cells IL-1β [81]
Wnt4 Promote Angiogenesis β-Catenin [71]
β-catenin promote cancer progression Wnt signaling pathway [50]
KRAS Alter the metabolic state of recipient colonic epithelial cells [85]
3.1.1 Tumor Growth

One of the typical hallmarks of cancer is sustaining proliferation signaling, ultimately causing the rapid growth of the tumor. CRC tumor-derived exosomes (CTDEs) can regulate the growth of CRC cells through a variety of pathways, such as involvement in cell proliferation, cell cycle, and apoptosis. For example, circLPAR1 is encapsulated by CRC cell exosomes, internalized by CRC cells, and inhibits tumor growth. The investigation of underlying mechanisms showed that exosome circLPAR1 might bind to eIF3h directly and inhibit METTL3-eIF3h interaction, ultimately reducing the translation of BRD4 [45]. Exosome-carried circ_0094343 derived from CRC cells plays an inhibitory role in the proliferation and clone formation of HCT116 cells via the miR-766-5p/TRIM67 axis [46]. Exosomal circ_PTPRA can function as a competing endogenous RNA (ceRNA) and increase the expression of SMAD4 by binding to miR-671-5p, ultimately inducing CRC cell cycle arrest and inhibiting cell proliferation [47]. It has also been demonstrated that a hypoxic microenvironment in CRC may promote tumor cells to release exosomes rich in miR-410-3p. These exosomes may transfer to normoxic cells and enhance the proliferation of normoxic CRC cells [93]. Exosomal circEPB41L2 derived from CRC cells could suppress CRC cell proliferation and promote apoptosis by serving as a sponge for miR-21-5p and miR-942-5p and regulating the PTEN/AKT signaling pathway [48]. CRC cell exosome miR-224-5p promotes cell proliferation by downregulating CMTM4, thus promoting the malignant transformation of human normal colonic epithelial cells [49]. Oncogenic mutant β-catenin contained in the extracellular vesicles can activate the Wnt signaling pathway in the recipient cells and increase tumor burden in vivo [50].

3.1.2 Invasion and Metastasis

Invasion and metastasis are the leading causes of cancer-associated death in patients with CRC; however, the molecular mechanisms underlying tumor invasion and metastasis are complex and elusive [94]. Numerous studies have shown that CTDEs are associated with the invasion and metastasis of CRC. For example, tumor-derived exosomes miR-934 can target PTEN, resulting in downregulation of the PTEN expression, which in turn activates the PI3K/AKT signaling pathway to induce M2 macrophage polarization, ultimately promoting CRC liver metastasis [51]. Cancer-derived exosomal circPACRGL can absorb miR-142-3p/miR-506-3p as a sponge and facilitate the TGF-β1 expression, thus promoting CRC cell migration and invasion [52]. The exosomes of CRC cells which have undergone EMT contain miR-106b, and miR-106b in exosomes can directly inhibit PDCD4 post-transcriptionally, activate the PI3Kγ/AKT/mTOR signaling pathway, and promote the M2 polarization of macrophages. Activated M2 macrophages promote EMT-mediated CRC cell migration, invasion, and metastasis in a positive feedback manner [53]. The hypoxic microenvironment in the CRC primary lesion promotes exosome release, selectively initiating the formation of a favorable premetastatic niche in the liver. Molecular mechanism exploration has found that Kupffer cells (KCs) can engulf exosomes containing miR-135a-5p, which enter the liver from the blood circulation. Exosomal miR-135a-5p initiates the large tumor suppressor kinase 2-yes-associated protein-MMP7 axis and promotes CRC liver metastasis [54]. Exosome-encapsulated miRNAs, which are from CRC cells, can enhance the M2 polarization of macrophages and contribute to CXCL12/CXCR4-induced liver metastasis of CRC [95]. LncRNA RPPH1is significantly upregulated in CRC tissues, and it could be encapsulated in the exosomes of CRC cells. Then, exosomes rich in RPPH1 are transported into macrophages and mediate macrophage M2 polarization, thereby promoting metastasis and proliferation of CRC cells [55]. Hypoxia can induce the secretion of circ-133-rich exosomes, which are transported into normoxic cancer cells. Circ-133 contained in the exosomes promotes cell migration via the miR-133a/GEF-H1/RhoA axis [56]. Exosomal miR-27b-3p secreted after epithelial-mesenchymal transformation of CRC cells increases vascular permeability and promotes the production of circulating tumor cells (CTCs), thus promoting the metastasis of CRC [96]. HuR, which is an RNA-binding protein contained in CRC-derived exosomes, can stabilize c-Myc mRNA and promote lung cell proliferation [97]. Fusobacterium nucleatum infection may stimulate tumor cells to generate exosomes rich in miR-1246/92b-3p/27a-3p and CXCL16/RhoA/IL-8 that are delivered to uninfected cells to promote metastatic behaviors in CRC [57]. Exosomal miR-221/222 targets SPINT1 and plays a key role in forming a favorable premetastatic niche (PMN), thus leading to the metastasis of CRC [58]. Exosome-transmitted miRNA-335-5p derived from metastatic CRC cells promotes CRC migration, invasion, and metastasis which may be due to EMT caused by RASA1 [59]. MiR-200c-3p in exosomes derived from CRC cells influences the exosomal expression of ZEB-1 mRNA, and further alters ZEB-1 protein expression in CRC cells, ultimately inhibiting CRC migration and invasion after LPS stimulation [60]. CRC-derived exosomal miR-106b-3p promotes metastasis by down-regulating DLC-1 expression. Serum exosome miR-106b-3p could be a potential molecular biomarker for prognosis and may be a target for the treatment of CRC [61]. Exosomal MALAT1 sponges miR-26a/26b, enhances FUT4 fucosylation, and activates the PI3K/Akt pathway, ultimately promoting the invasion and metastasis of CRC [62]. MicroRNA-21-5p is highly enriched in CRC-derived exosomes and is essential for creating a liver proinflammatory phenotype and liver metastasis of CRC by inducing an inflammatory premetastatic niche through the miR-21-TLR7-IL-6 axis, therefore promoting liver metastasis [63]. Exosomal miR-193a and let-7g facilitate cancer primary CRC progression and peritoneal metastasis by targeting MMP16 and CDKN1A [98]. It is also demonstrated that hypoxia may promote the release of Wnt4-rich exosomes which are derived from tumor cells. The Wnt4-rich exosomes are delivered to normoxic cells and facilitate the migration and invasion of normoxic CRC cells [64]. Exosomal IRF-2 remodels the lymphatic network in a sentinel lymph node (SLN) and may predict the development of CRC lymph node (LN) metastases [99]. Exosomes rich in circPABPC1 are transported from CRC cells to CRC cells and are involved in the progression of CRC. In the nucleus, circPABPC1 initiates the transcription of HMGA2 by recruiting KDM4C to its promoter and reducing the H3K9me3 modification. In the cytoplasm, it inhibits the degradation of ADAM19 and BMP4, which is mediated by miR-874 and miR-1292. Thus, circPABPC1 facilitates liver metastasis in CRC via upregulating the expression of HMGA2, BMP4, and ADAM19 [100]. Exosomes carrying circ-ABCC1 from CD133+ cells have the ability to mediate cell stemness and metastasis in CRC [101]. LncRNA PCAT1, which is from CRC exosomes, can promote the EMT and liver metastasis of CRC. Because exosome lncRNA PCAT1 can influence miR-329-3p and the activity of the Netrin-1-CD146 complex in circulating tumor cells (CTCs) [65]. Exosomal‑miR‑10a derived from CRC cells reduced the expression of IL‑6, IL‑8, and IL‑1β in normal human lung fibroblasts (NHLFs), thereby reducing the proliferative and migratory activities of primary NHLFs. This will help us understand the mechanism underlying the process of CRC lung metastasis [66]. Exosomal miR-1255b-5p targets human telomerase reverse transcriptase and inhibits its expression in CRC cells, ultimately suppressing tumor progression and liver metastasis of CRC [67]. Exosomal lncRNA 91H enhances CRC metastasis by modifying HNRNPK expression in CRC [49].

3.1.3 Angiogenesis

Angiogenesis is critical for tumor growth, survival, and progression. The process is very complex, including the degradation of the vessel’s basement membrane, the activation, proliferation, and migration of vascular endothelial cells, and reconstruction to form new blood vessels and networks [102]. Tumor-derived exosomes are largely involved in this process [103]. This has been demonstrated in the angiogenesis of CRC. For example, exosomal miR-25-3p, which is CRC cell-derived, directly targets KLF2 and KLF4 and regulates VEGFR2, ZO-1, claudin5, and occludin expression in endothelial cells, consequently promoting vascular permeability and angiogenesis [68]. Exosomal miR-21-5p, which is secreted by CRC cells, induces angiogenesis and vascular permeability by targeting KRIT1 [69]. MiR-1229, which is derived from the CRC cell exosomes, inhibits the expression of HIPK2 protein, thus activating the VEGF pathway and promoting angiogenesis [70]. Exosomal Wnt4 derived from CRC cells increases β-catenin nuclear translocation in endothelial cells and promotes angiogenesis in CRC [71].

3.1.4 Immune Evasion

Exosomes are important in modulating tumor immune response and have a dual role. They may activate immune responses or exhibit strong pro-tumor immune reactions [17]. Numerous studies have revealed CRC tumor-derived exosomes (CTDEs) can function as mediators of host anti-tumor immune responses and tumor cell immune evasion. For example, miR-208b is secreted by colon cancer cells and sufficiently transported to recipient T cells. In T cells, miR-208b, which is delivered by exosomes, directly targets PDCD4 and induces immune evasion [72]. The CRC cell-derived exosomal lncRNA SNHG10s can be taken up by NK cells and suppress the function of NK cells by upregulating INHBC expression. It has been suggested that exosomal lncRNA SNHG10 could lead to the inhibition of NK cells, ultimately contributing to immune escape [73]. Exosomes containing Lnc-HOXB8-1:2 are secreted by neuroendocrine differentiated CRC cells, and Lnc-HOXB8-1:2 from exosomes competitively binds hsa-miR-6825-5p as ceRNA relieves the inhibitory effect of hsa-miR-6825-5p on CXCR3 expression, upregulates its expression, and then leads to TAM infiltration and M2 polarization, and promotes immune evasion of CRC [74]. CRNDE-h, which is derived from tumor cells, could inhibit ubiquitination and degradation of RORγt and promote Th17 cell differentiation in CRC [75]. CRC stem cell-derived exosomes (CRCSC-exos) deliver miR-17-5p to CRC cells. The high expression of miR-17-5p in CRCSC exosomes promotes tumor cell growth by promoting PD-L1 inhibition of SPOP and inhibits the anti-tumor immunity of CRC [76]. Tumor-derived HOTAIR could bind to PKM2 and inhibit its ubiquitination degradation, leading to the activation of STAT3 and expression of PDL1, thereby polarizing B cells toward a regulatory feature and suppressing CD8+ T cell activity [77].

3.1.5 TME Remodeling

The tumor microenvironment (TME) is a complex internal environmental system around tumor cells, which plays an essential role in tumorigenesis. CTDEs in the TME are essential in the formation and reprogramming of the TME. For example, miR-146a-5p and miR-155-5p contained in CRC cell exosomes can be secreted by CRC cells and taken up by the cancer-associated fibroblasts (CAFs). Then, they may activate CAFs through JAK2-STAT3/NF-κB signaling. Reciprocally, the activation of CAFs may further facilitate the invasion of CRC cells [78]. CRC cell-derived exosomal HSPC111 can be engulfed by CAFs. In CAFs, it may phosphorylate ACLY and promote the expression of acetyl-CoA. Acetyl-CoA accumulation further increases H3K27 acetylation and promotes CXCL5 expression in CAFs. Interestingly, CXCL5 from CAFs can reinforce exosomal HSPC111 excretion from CRC cells and promote liver metastasis via the CXCL5-CXCR2 axis [79]. Exosomal ANGPTL1 derived from CRC cells is mainly taken up by Kupffer cells (KCs). Then, ANGPTL1 can downregulate MMP9 levels by inhibiting the JAK2-STAT3 signaling pathway in KCs [80]. Tumor exosomal tri-phosphate RNAs sustain neutrophil survival by inducing the expression of IL-1β through a pattern recognition-NF-κB signaling axis. CRCSC-secreted CXCL1 and CXCL2 then attract CRCSC-primed neutrophils to promote tumorigenesis of CRC cells via IL-1β [81]. CRC cell-derived exosomes containing miR-10b can be transferred to fibroblast cells and activate fibroblasts to become CAFs via the PI3K/Akt pathway [82]. Studies also revealed that CRC tumor-derived exosomes contribute to generating phenotypically and functionally distinct subsets of CAFs by reprogramming their proteome and may facilitate tumor progression [77]. Exosomes that carry miR-203 can be secreted from CRC cells, and they can be swallowed by monocytes. In monocytes, miR-203 can promote the expression of M2 markers, suggesting that miR-203 may facilitate the differentiation of monocytes to M2- TAMs [83].

3.1.6 Metabolic Reprogramming

Metabolic reprogramming is an adaptive mechanism that enables tumor cells to regulate the flow of their energy to meet their needs for rapid growth. It is manifested by an increase in glucose uptake and an enhancement of glycolysis under aerobic conditions, as well as high production of lactate. These metabolic changes are also known as the “Warburg effect” [104]. A growing number of studies have shown that exosomes can mediate metabolic reprogramming in cancer [105]. As in CRC, hsa-miR-101-3p, which is derived from CRC cell exosomes, is associated with metabolic reprogramming in CRC by targeting HIPK3 [84]. Moreover, mutant KRAS exosomes are able to cause a Warburg-like effect on recipient colonic epithelial cells [85].

3.1.7 Therapy Resistance

It has been demonstrated that the delivery of exosomal cargos between different cancer cells is associated with tumor drug resistance [106]. Exosomes are used as carriers for cell-to-cell communication in the tumor microenvironment, however; drug-resistant tumor cells can also use this property to develop resistance to sensitive cells [107]. Therapy-resistance mechanisms mediated by CTDEs are summarized as follows: hsa_circ_0005963 is involved in chemoresistance in CRC. The exploration of underlying mechanisms showed that hsa_circ_0005963 could be a sponge for miR-122 [86]. Tumor-secreted miR-208b directly targets PDCD4 and promotes Treg expansion, and it may be associated with a decrease of oxaliplatin (OX)-based chemosensitivity in CRC [72]. It is also demonstrated that exosomes from RKO/R cells are able to promote acquired 5-FU resistance in CRC, which is mainly related to p-STAT3 contained in the exosomes [87]. Circ_0000338 is present in exosomes and improves the chemoresistance of CRC cells by targeting miR-217 and miR-485-3p [88]. Exosome miR-46146 can be an important promoter of OX resistance by targeting PDCD10 and may be a potential target for OX resensitization by CRC cells [89]. MiR-19b can be present in exosomes secreted by CRC cells. By delivering miR-19b, CRC-derived exosomes enhance the radiation resistance and stemness characteristics of CRC cells. Accordingly, miR-19b inhibition can enhance the efficacy of radiotherapy and reduce the stemness characteristics of CRC, suggesting that miR-19b inhibition may be a promising strategy for sensitization of CRC cells to radiotherapy [90]. Exosomal circ-FBXW7 can improve the chemoresistance of CRC to OX by direct binding to miR-128-3p, which provides a promising treatment strategy for patients with OX-resistant CRC [91]. Exosomal circATG4B plays an important role in chemoresistance in CRC. The underlying mechanisms show that it can competitively bind to TMED10, prevent TMED10 from binding to ATG4B, and induce increased autophagy, ultimately promoting chemotherapy resistance [108]. Exosomal long non-coding RNA HOTTIP derived from mitomycin-resistant CRC cells can be transferred into the parental cells and increase the resistance of CRC cells to mitomycin via impairing miR-214-mediated degradation of KPNA3 [92].

3.2 Non-Tumousr-Derived Exosomes in CRC

Although tumor cells are the main cells in the tumor microenvironment (TME), there are also many other cells, such as fibroblasts, immune cells, endothelial cells, etc. [109]. Exosomes secreted by non-CRC cells in the TME may also affect the fate of CRC cells. Here, we summarize the functions and mechanisms of non-tumor-derived exosomes in CRC (Fig. 4; Table 2, Ref. [110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134]).

Fig. 4.

The Mechanism of non-tumor-derived exosomes function in CRC. Exosomes derived from non-CRC cells can transport their cargo to CRC cells and affect most pathological processes in CRC, including stemness, EMT, migration, invasion, metastasis, angiogenesis, apoptosis, tumor growth, and therapy resistance.

Table 2.The function and mechanism of non-tumor-derived exosomes in CRC.
Type Molecule Source Function Target Ref.
miRNA miR-92a-3p CAFs Promote the stemness, EMT, metastasis, and chemotherapy resistance of CRC cells FBXW7, MOAP1 [110]
miR-93-5p CAFs Against radiation-induced apoptosis FOXA1 [112]
miR‑181d‑5p CAFs Inhibit 5-FU sensitivity NCALD [115]
miR-181b-3p CAFs Promote the occurrence and development of CRC SNX2 [116]
miR-100 MSCs Suppress proliferation, migration, invasion, and metastasis and induce apoptosis mTOR [126]
miR-3940-5p MSCs Inhibit the growth, metastasis, invasion, and EMT of CRC cells ITGA6 [127]
miR-590-3p CAFs Enhance radioresistance in CRC CLCA4 [118]
miR-625-3p CAFs Promote migration, invasion, EMT, and chemotherapeutic resistance in CRC cells CELF2 [119]
microRNA-135b-5p CAFs Promote CRC cell growth and angiogenesis TXNIP [120]
miR-424 BMSCs Promote progression of CRC TGFBR3 [129]
miR-200b-3p CAFs Increase sensitivity to 5-FU HMGB3 [122]
miR-4461 BMSCs Inhibit migration and invasion COPB2 [128]
microRNA-16-5p BMSCs Inhibit proliferation, migration, and invasion, while promoting apoptosis ITGA2 [130]
miR-203a-3p hepatocyte Induce EMT Src [134]
miR-17-5p CAFs Contribute to tumor progression RUNX3 [123]
miR-22-3p BMSCs Suppress CRC cell proliferation and invasion RAP2B [131]
LncRNA H19 CAFs Promote the stemness and chemoresistance of CRC miR-141 [111]
LINC00659 CAFs Promote CRC cell proliferation, invasion, and migration miR-342-3p [113]
WEE2-AS1 CAFs Facilitate tumorigenesis and progression MOB1A [121]
CCAL CAFs Promote oxaliplatin resistance of CRC cells HuR [124]
circRNA circEIF3K CAFs Promote the progression of CRC miR-214 [114]
cricN4BP2L2 CAFs Promote the CRC cell stemness and oxaliplatin resistance EIF4A3 [117]
Others MTTP Adipocyte Suppress ferroptosis, promote chemoresistance PRAP1 [133]
S100A9 G-MDSCs Promote the stemness of CRC cells [132]
Wnts Fibroblasts Induce the dedifferentiation of cancer cells to promote chemoresistance [125]

For example, CAFs are the main stromal cells in TME. CAFs can transfer exosomes that are rich in miR-92a-3p directly to CRC cells and increase the expression of miR-92a-3p in CRC cells significantly. In CRC cells, miR-92a-3p directly targets FBXW7 and MOAP1, ultimately promoting the stemness, EMT, metastasis, and chemotherapy resistance of CRC cells [110]. CAFs can deliver H19 in exosomes to CRC cells, promoting the stemness and resistance of CRCs. Mechanistically, H19 activates the β-catenin pathway by serving as a competing endogenous RNA sponge of miR-141, which could inhibit the stemness and chemoresistance of CRC cells after delivery to CRC cells [111]. CAFs-derived exosomes contain a large number of miR-93-5p, which can promote tumor growth in irradiated nude mice [112]. Exosomal LINC00659 could be secreted by CAFs and taken up by CRC cells. Then, it could target the miR-342-3p/ANXA2 axis and promote cell proliferation, invasion, and migration of CRC [113]. Exosomal circEIF3K derived from CAFs promotes the progression of CRC via miR-214/PD-L1 axis [114]. CAFs-derived exosomes contain miR‑181d‑5p, which is associated with 5-FU sensitivity. It has been demonstrated that miR‑181d‑5p inhibits 5-FU sensitivity through the METTL3/miR‑181d‑5p/NCALD axis in CRC cells [115]. CAFs-derived exosome miR-181b-3p promotes the occurrence and development of CRC by regulating SNX2 expression [116]. CAFs-secreted exosome circN4BP2L2 promotes the CRC cell stemness and oxaliplatin resistance through EIF4A3/PI3K/AKT/mTOR pathway [117]. CAFs can transmit miR-590-3p to CRC cells through exosomes. In CRC cells, miR-590-3p enhances the radioresistance of CRC by regulating the CLCA4-dependent PI3K/Akt signaling pathway [118]. CAFs exosomes are directly swallowed by CRC cells, and miR-625-3p in exosomes is released and may inhibit the CELF2/WWOX pathway after entering CRC cells, thereby promoting the migration, invasion, EMT and chemotherapy resistance of CRC cells [119]. CAFs-derived exosomes upregulate microRNA-135b-5p to promote CRC cell growth and angiogenesis by inhibiting TXNIP [120]. After entering CRC cells, WEE2-AS1 in the exosomes of CAFs can promote the degradation of MOB1A, thereby inhibiting the Hippo pathway and promoting the occurrence and progression of CRC tumors [121]. Hypoxia can lead to the loss of miR-200b-3p in the exosomes of CAFs, thereby reducing the sensitivity of CRC cells to 5-FU [122]. CAFs-exosomes show higher expression of miR-17-5p than normal fibroblasts-exosomes and can deliver exosomal miR-17-5p from parental CAFs to CRC cells. Further studies confirm that that miR-17-5p directly target RUNX3 3’-UTR to affect CRC metastasis. RUNX3 interacts with the MYC proto-oncogene, and both RUNX3 and MYC bind to the promoter of TGF-β1 on 1005-1296 base pairs, thereby activating the TGF-β signaling pathway and promoting tumor progression [123]. LncRNA CCAL is transferred from CAFs to the cancer cells via exosomes and promotes oxaliplatin resistance of CRC cells. Mechanistically, CCAL interacts directly with mRNA stabilizing protein HuR and promoting the expression of β-catenin [124]. Exosomes from the normal fibroblasts also influence CRC cells. For example, Exosomal Wnts derived from fibroblasts can induce the differentiation of cancer cells to promote chemoresistance in CRC [125].

Exosomes from mesenchymal stem cells (MSCs) are closely related to the therapeutic efficacy of MSCs. After being ingested by CRC cells, MSC exosomes can exert their effects through the miR-100/mTOR/miR-143 axis in CRC cells, inhibiting the proliferation, migration, invasion, and metastasis of CRC cells and inducing CRC cell apoptosis. It suggests that MSC-exosome treatment and miR-100 restoration might be considered potential therapeutic strategies for CRC [126]. MSC-exosomal miR-3940-5p inhibits the growth, metastasis, invasion, and EMT of CRC cells by targeting ITGA6 and the following TGF-β1 inactivation [127]. The exosomes produced by bone marrow mesenchymal stem cells (BMSCs) contain abundant miR-4461. After being taken up by CRC cells, miR-4461 can directly target COPB2, leading to COPB2 downregulation and inhibiting the migration and invasion of CRC cells [128]. Inhibited exosomal miR-424 from BMSCs inhibited malignant behaviors of CRC cells by targeting TGFBR3, thus suppressing the progression of CRC [129]. BMSCs exosomes contain microRNA-16-5p, and exosomes containing microRNA-16-5p are swallowed by CRC cells, which inhibit the proliferation, migration, and invasion of CRC cells and promote apoptosis of CRC cells [130]. Exosomes derived from MSCs are rich in miR-22-3p, which inhibits CRC cell proliferation and invasion by regulating RAP2B and PI3K/AKT pathways, suggesting the potential of hBMSCs-exo-miR-22-3p for the treatment of CRC in the future [131].

Some other cells may also affect the fate of CRC cells. For example, Granulocytic Myeloid-derived suppressor cells (G-MDSCs) promote the stemness of CRC cells through exosomal S100A9 [132]. Adipocyte-derived exosomal MTTP could suppress ferroptosis and promote chemoresistance in CRC [133]. MiR-203a-3p derived from hepatocyte exosomes increases the expression of E-cadherin in CRC cells and inhibits Src expression, which in turn leads to a decrease in the invasion rate of CRC cells [134].

4. The Potential Clinical Value of Exosomes in CRC

Due to their unique properties, exosomes contribute to many aspects of precise tumor diagnosis and treatment, including predicting prognosis and drug efficacy, dynamic monitoring, and precisely targeted drug delivery [14].

4.1 Exosomes in CRC Diagnosis and Prognosis

Because of the various cargos in the exosomes and the typical features on the surface of exosomes, they can reflect the status of their parent cells. Thus, exosomes have emerged as a platform with potentially broader and complementary applications to be used in the field of liquid biopsy for the diagnosis, prognostic analysis, and monitoring of cancer [135]. In this manuscript, we summarized the exosomes used in the diagnosis and prognosis of CRC (Table 3, Ref. [55, 58, 72, 75, 136, 137, 138, 139, 140, 141, 142, 143, 144, 145, 146, 147, 148, 149, 150, 151, 152, 153, 154, 155, 156, 157, 158, 159, 160, 161, 162, 163, 164, 165, 166]).

Table 3.The Clinical Potential of Exosomes “cargos” in CRC.
Type Exosomes Clinical Potential Ref.
miRNA miR-208b A predictive biomarker for oxaliplatin-based therapy response [72]
miR‐221/222 A prognostic marker and potential therapeutic target for CRC with liver metastasis [58]
miR-7641 A molecular biomarker for diagnosis and prognosis of CRC [167]
miR-96-5p and miR-149 A biomarker for early detection of CRC [169]
miR-377-3p and miR-381-3p Circulating biomarkers for diagnosis of CRC [136]
miR-1539 A potential biomarker for screening and a predictor of poor clinicopathological behavior in CRC [138]
miR-150-5p and miR-99b-5p Serve as diagnostic biomarkers for CRC [139]
miR-3937 A potential liquid biopsy marker for CRC [140]
miR-548c-5p A critical biomarker for CRC diagnosis and prognosis [141]
miRNA-139-3p A biomarker for early diagnosis and metastasis prediction in CRC [143]
miR-424-5p A biomarker for early diagnosis in CRC [75]
miR-320d A biomarker for metastatic CRC [144]
miR-221 An independent prognostic factor for CRC [145]
miR-874 An independent prognostic factor for overall survival of CRC patients [148]
miR-17-5p and miR-92a-3p A prognostic biomarker for primary and metastatic CRC [150]
miR-21 A predictor of recurrence and poor prognosis in CRC [152]
miR-6869-5p A circulating biomarker for the prognosis of CRC [153]
miR-122 A potential diagnostic and prognostic biomarker for CRC with liver metastasis [154]
miR-150 A potential prognostic factor and treatment target for CRC [157]
miR-150-5p A novel non-invasive biomarker for CRC diagnosis and prognosis [159]
miR-125b-5p Be potentially correlated with a more aggressive CRC phenotype [161]
miR-92b A promising biomarker for early detection of CRC [162]
miR-126, miR-1290, miR-23a, and miR-940 Potential biomarkers for early diagnosis of CRC [164]
microRNA-125b A biomarker of resistance to mFOLFOX6-based chemotherapy in CRC [165]
miR-181b, miR-193b, miR-195, and miR-411 A predicter for lymph node metastasis in T1 CRC patients [166]
LncRNA RPPH1 A potential biomarker for diagnosis and therapy in CRC [55]
GAS5 An independent prognostic factor for CRC [145]
FOXD2-AS1, NRIR, and XLOC_009459 Biomarkers for the diagnosis of CRC [151]
UCA1 A predicter for therapeutic efficacy of cetuximab in CRC patients [156]
CCAT2 A potential predictor of CRC [163]
circRNA circCOG2 A biomarker for poor prognosis and a therapeutic target for CRC [137]
circ-0004771 A novel potential diagnostic biomarker of CRC [146]
circ_0006174 A biomarker for the diagnosis of chemoresistance in CRC [147]
circ-PNN A potential biomarker for the detection of CRC [155]
Others FZD10 A biomarker for prognosis and diagnosis of CRC [168]
GPC1 A biomarker for early detection of CRC [169]
CPNE3 A diagnostic and prognostic biomarker for CRC [142]
FGB and β2-GP1 Biomarker for diagnostic efficacy of early CRC [149]
QSOX1 A marker for early diagnosis and non-invasive risk stratification in CRC [158]
KRAS A predictor for outcome in CRC patients with metastasis [160]

Exosomal LncRNA RPPH1 levels in blood plasma are higher in untreated CRC patients but lower after tumor resection. It displayed a better diagnostic value (AUC = 0.86) compared to CEA and CA199 and could serve as a potential target for therapy and diagnosis in CRC [55]. The miR-208b derived from exosomes of CRC cells can promote Treg amplification by regulating PDCD4, and it has the ability to reduce the sensitivity of oxaliplatin-based chemotherapy in CRC. These research results suggest that the exosomal miR-208b can serve as a biomarker for predicting oxaliplatin treatment response and may become a new target for immunotherapy [72]. Exosomal miR-221/222 promotes CRC progression and may serve as a novel prognostic marker and therapeutic target for CRC with liver metastasis [58]. By deep sequencing, it has been demonstrated that miR-7641 has the potential to be a candidate for the non-invasive and specific molecular markers for CRC diagnosis and prognosis [167]. Exosome-delivered FZD10 increases Ki-67 expression via Phospho-ERK1/2 and may be a promising novel prognostic and diagnostic biomarker for CRC [168]. Elevated plasma GPC1+ exosomes and decreased plasma miR-96-5p and miR-149 expression may be specific markers for the diagnosis of CRC and targets for the treatment of CRC. Compared with the healthy control group, the percentage of GPC1+ exosomes in tumor tissue and plasma and the expression of GPC1 protein in exosomes in CRC patients before surgery were significantly increased. However, miR-96-5p and miR-149 expression in tumor tissue and plasma and GPC1+ exosomes in CRC patients were significantly reduced compared to healthy controls [169]. The expression of exosomal miR-377-3p and miR-381-3p are decreased in CRC patients; they can serve as circulating biomarkers of diagnosis for CRC [136]. CircCOG2 is associated with poor prognosis and can be used as a therapeutic target for CRC [137]. Circulating or tissue-based miR-1539 derived from CRC cells may be used as a novel potential biomarker for CRC screening, as well as a predictor of poor clinicopathological behavior in tumors [138]. Circulating exosomal miR-150-5p and miR-99b-5p derived from CRC cells may be considered diagnostic biomarkers for CRC [139]. Hsa-miR-3937, contained in tumor-originated exosomes, is a potential and effective liquid biopsy marker for CRC detection and therapy [140]. The downregulation of exosomal miR-548c-5p in serum predicts poor prognosis in patients with CRC, and thus it may be a critical biomarker for CRC diagnosis and prognosis [141]. Circulating exosomal CPNE3 may be a diagnostic and prognostic biomarker for CRC [142]. Plasma exosomal miRNA-139-3p is decreased in CRC and may act as a novel biomarker for early diagnosis and metastasis monitoring in CRC [143]. CRC cell-derived exosomes are rich in miR-424-5p, and miR-424-5p is secreted into peripheral blood through exosomes. The circulating exosomes miR-424-5p can be used as a marker for early diagnosis of CRC [75]. Exosomal miR-320d could significantly distinguish metastatic from non-metastatic CRC patients and be a biomarker for metastatic CRC [144]. The expression of LncRNA GAS5 and miR-221 in CRC tissue, patient plasma, and exosomes can be independent prognostic factors for CRC [145]. Studies have revealed that the expression of hsa-circ-0004771 in circulating exosomes in CRC patients is significantly upregulated, which is expected to become a new biomarker for colorectal cancer diagnosis [146]. After the circ_0006174-rich exosomes enter CRC cells, the DOX resistance of CRCs is improved through the miR-1205/CCND2 axis. Therefore, exosome circ_0006174 is expected to become a biomarker for the diagnosis of chemotherapy resistance in CRC [147]. Serum exosomal miR-874 is significantly downregulated in CRC patients and has been negatively associated with distant metastasis, lymph node metastasis, differentiation, and advanced TNM stage. Thus, serum exosomal miR-874 may be a statistically significant independent prognostic factor for CRC patients [148]. It is also demonstrated that exosomal FGB and β2-GP1 can be used as significant diagnostic efficacy for early CRC by using comprehensive proteomics analyses [149]. Circulating exosomes miR-17-5p and miR-92a-3p may be non-invasive prognostic markers in patients with CRC, being either primary or metastatic CRC [150]. Exosome lncRNAs FOXD2-AS1, NRIR, and XLOC_009459 are promising biomarkers for diagnosing CRC, especially early-stage CRC [151]. Plasma exosomal miR-21 levels are a useful biomarker for the prediction of recurrence and poor prognosis in CRC patients with TNM stage II, III, or IV [152]. MiR-6869-5p may play a cancer-suppressing role in CRC, and serum exosome miR-6869-5p is a promising circulating biomarker for predicting the prognosis of CRC [153]. Serum exosomal miR-122 is a novel potential diagnostic and prognostic biomarker in CRC patients with liver metastasis (LM) [154]. Serum exosome circ-PNN may be a new biological marker for CRC detection, with non-invasive characteristics, and may play an important role in the tumorigenesis of CRC [155]. Exosomal UCA1 is detectable and stable in the serum of CRC patients. Moreover, circulating exosomes containing UCA1 can predict the clinical outcome of treatment with cetuximab in CRC patients [156]. Exosomal miR-150 can be used as a marker for the prognosis of CRC patients after surgical resection, and its low expression predicts poor prognosis after surgery. Therefore, exosome miR-150 may be a prognostic factor and potential therapeutic target for CRC [157]. Exosome-derived QSOX1 is a new promising marker for early diagnosis and non-invasive risk stratification of CRC [158]. Serum exosomal miR-150-5p is expressed differently between healthy people and CRC patients and is expected to be a potential molecular marker for the diagnosis and prognosis of CRC [159]. KRAS mutations are very common in CRC, KRAS mutations are manifested in exosomes, and plasma exosome KRAS mutation status predicts the prognosis of patients with metastatic CRC [160]. The higher the expression of miR-125b-5p in plasma exosomes, the stronger the CRC aggressiveness, suggesting that miR-125b-5p in plasma exosomes may be related to the phenotype of CRC aggressiveness [161]. Exosome-derived miR-92b is down-regulated in the plasma of CRC patients and can be used as a molecular marker for early diagnosis of CRC [162]. Circulating CCAT2 is present in exosomes, protected by exosomes, and can serve as a potential predictor of CRC [163]. In CRC cells, miRNAs can be encapsulated into exosomes and secreted outside the cell, regardless of intracellular miRNA expression. Among these miRNAs, serum exosomes miR-126, miR-1290, miR-23a, and miR-940 are novel potential molecular markers for early diagnosis of CRC [164]. In patients with advanced and recurrent CRC, plasma exosome microRNA-125b can be used as a molecular marker for detecting mFOLFOX6-based chemotherapy resistance [165]. A group of miRNAs which includes miR-181b, miR-193b, miR-195, and miR-411, is able to detect lymph node metastasis in the exosomal vs. cell-free component. This suggests that exosomal miRNA-based liquid biopsy features can strongly identify invasive submucosal CRC (T1 CRC) patients at risk of lymph node metastasis in the preoperative setting [166].

4.2 Exosomes in CRC Clinical Transformation

Because of the properties and functions of exosomes, they can be designed for drug or functional nucleic acid delivery [170] and have the therapeutic potential to be used in the treatment of CRC. For example, engineered exosomes have been used to simultaneously deliver the anticancer drug 5-FU and the miR-21 inhibitor oligonucleotide (miR-21i) to Her2-positive cancer cells. The results revealed that systematic administration of exosomes loaded with 5-FU and miR-21i in tumor-bearing mice showed significant anti-tumor effects in colon cancer [171]. Studies have shown that the expression of PGM5 antisense RNA 1 (PGM5-AS1) in colon cancer is induced by GFI1B. PGM5-AS1 prevents colon cancer cells from proliferating, migrating, and acquiring oxaliplatin tolerance, and engineered exosomes that co-deliver PGM5-AS1 and oxaliplatin can reverse colon cancer resistance [172]. MiR-506-3p delivered to CRC cells via exosomes reduces CRC proliferation and induces apoptosis, suggesting that delivery of miR-506-3p to CRC cells via exosomes may become a novel diagnosis and treatment method for CRC [173]. Tumor-derived exosomes (TEXs) may induce beneficial anti-tumor immune responses, and TEXs have shown certain beneficial anti-tumor properties in addition to miRNA delivery functions, suggesting that the introduction of TEX-miR-34a may be a new promising approach for combination therapy for CRC [174]. Studies have shown that dendritic cells are loaded with exosomes from cancer stem cell-rich spheroids, which may be a new potential immunotherapy approach [50]. It has been reported that the ascites-derived exosomes (Aex), in combination with the granulocyte-macrophage colony-stimulating factor (GM-CSF), could be used in the immunotherapy of CRC. The phase I clinical trial suggested that AEX combined with GM-CSF immunotherapy for advanced CRC is feasible and safe and may be an alternative to immunotherapy for advanced CRC [175].

Although exosomes have been considered promising emerging therapeutic options and tumor neo-antigen drug delivery tools and carriers in tumor precision therapy, there are still many issues waiting to be resolved. Nowadays, exosomes can be isolated and purified by ultracentrifugation, density gradient centrifugation, polymer precipitation, ultrafiltration, and size exclusion chromatography; however, flexible and fast detection platforms are still lacking. Moreover, how to load exosomes with specific cargo still need to be explored. Because few studies have focused on the fusion-based loading methods present, their detrimental effects on recipient cells remain unclear [14].

5. Conclusions and Prospects

Exosomes are derived from the endosomal pathway of nearly all cells and may be considered a cell-to-cell system in the human body that can mediate near- and long-distance intercellular communication and affect various aspects of cell biology. They are involved in different physiological and pathological processes, such as the communication of the nervous system, reproduction and development, inflammatory responses, cardiovascular diseases, and cancer development.

CRC is one of the most highly malignant tumors worldwide. Numerous studies have revealed that exosomes play important roles in tumor growth, invasion, metastasis, angiogenesis, immune evasion, TME remodeling, metabolic reprogramming, therapy resistance, and some other processes of CRC. Because of these features and functions of exosomes, they may be used in the fields of diagnosis, prognosis, and treatment of CRC. We have reviewed the current state of studies focused on the exosomes in CRC. We have compared the effects of tumor-derived and non-tumor-derived exosomes on the processes of CRC, summarized the clinical value of various types of exosomal cargos, and presented the potential of exosomes to be used in the clinical transformation process.

However, there are still many issues waiting to be resolved before exosome-based therapy can be used in the clinical treatment of CRCs. First, it is still a problem to obtain a large number of exosomes quickly so far. The isolation and purification of exosomes can be realized by the following methods, such as separation based on the size of density or particle, sedimentation or phase, affinity, microfluidic systems, or thermophoretic enrichment. However, the efficiency, quantity, and quality of exosomes separated by different separation methods are different, and there is still a lack of unified methods that can quickly and effectively obtain exosomes. Moreover, how to equip exosomes with specific drugs is also a problem. There are two methods for loading exosomes with drug, endogenous and exogenous. The two methods both have their own disadvantages; the endogenous method is not accurate in quantifying the specific substances in the exosomes, while the loading efficiency is not high using the exogenous methods. Thus, how to equip exosomes with specific drugs is also a question that needs to be resolved. Furthermore, the long-term efficacy and safety of using exosomes in the treatment of CRC remain unclear. Therefore, there is a need for comprehensive clinical studies or trials of exosomes used in CRC. It is possible that with technological developments and further clinical trials, exosomes will be a real emerging tool for the “cell-free” treatment of CRC.

Author Contributions

Conceptualization: JH, YH; writing - original draft preparation: JH, SM, YZ, BW, SD, YH; writing – review and editing: JH, YH; visualization: SM, YH; analyzing: YZ, BW, SD; interpreting data from the reviewed literature, JH, SM, YZ, BW, SD; supervision: YH. All authors have read and agreed to the published version of the manuscript.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

Thanks Dr. Jiateng Zhong for the contribution to review and editing.

Funding

This work was supported by the National Natural Science Foundation of China (No. 82203350), Natural Science Foundation of Henan Province (No. 212300410223), and the Xinxiang Medical University (No. XYBSKYZZ201820), Teaching and Research Cultivation Project of School of Basic Medical Sciences, Xinxiang Medical College (No. JCYXYKY202021), the Open Project Program of the Third Affiliated Hospital of Xinxiang Medical University (No. KFKTYB202125).

Conflict of Interest

The authors declare no conflict of interest.

References
[1]
Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA: A Cancer Journal for Clinicians. 2022; 72: 7–33.
[2]
Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA: A Cancer Journal for Clinicians. 2021; 71: 209–249.
[3]
Munro MJ, Wickremesekera SK, Peng L, Tan ST, Itinteang T. Cancer stem cells in colorectal cancer: a review. Journal of Clinical Pathology. 2018; 71: 110–116.
[4]
Brenner H, Kloor M, Pox CP. Colorectal cancer. Lancet. 2014; 383: 1490–1502.
[5]
Moug SJ, Bryce A, Mutrie N, Anderson AS. Lifestyle interventions are feasible in patients with colorectal cancer with potential short-term health benefits: a systematic review. International Journal of Colorectal Disease. 2017; 32: 765–775.
[6]
Huang B, Feng Y, Zhu L, Xu T, Huang L, Cai G. Smaller tumor size is associated with poor survival in stage II colon cancer: An analysis of 7719 patients in the SEER database. International Journal of Surgery (London, England). 2016; 33: 157–163.
[7]
Ngune I, Jiwa M, McManus A, Parsons R, Pagey G, Hodder R. Do Patients Treated for Colorectal Cancer Benefit from General Practitioner Support? A Video Vignette Study. Journal of Medical Internet Research. 2015; 17: e249.
[8]
Sharen G, Li X, Sun J, Zhang L, Xi W, Zhao X, et al. Silencing eL31 suppresses the progression of colorectal cancer via targeting DEPDC1. Journal of Translational Medicine. 2022; 20: 493.
[9]
Al-Busaidi IS, Bailey T, Dobbs B, Eglinton TW, Wakeman CJ, Frizelle FA. Complete resection of colorectal cancer with ovarian metastases combined with chemotherapy is associated with improved survival. ANZ Journal of Surgery. 2019; 89: 1091–1096.
[10]
Rudmik LR, Magliocco AM. Molecular mechanisms of hepatic metastasis in colorectal cancer. Journal of Surgical Oncology. 2005; 92: 347–359.
[11]
Trédan O, Galmarini CM, Patel K, Tannock IF. Drug resistance and the solid tumor microenvironment. Journal of the National Cancer Institute. 2007; 99: 1441–1454.
[12]
Pancione M, Giordano G, Remo A, Febbraro A, Sabatino L, Manfrin E, et al. Immune escape mechanisms in colorectal cancer pathogenesis and liver metastasis. Journal of Immunology Research. 2014; 2014: 686879.
[13]
Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science (New York, N.Y.). 2020; 367: eaau6977.
[14]
Zhao L, Yu L, Wang X, He J, Zhu X, Zhang R, et al. Mechanisms of function and clinical potential of exosomes in esophageal squamous cell carcinoma. Cancer Letters. 2023; 553: 215993.
[15]
Zhang Y, Li J, Gao W, Xie N. Exosomes as Anticancer Drug Delivery Vehicles: Prospects and Challenges. Frontiers in Bioscience-Landmark. 2022; 27: 293.
[16]
Jia Z, Lv Y, Zhang W, Zhang X, Li F, Lu X, et al. Mesenchymal stem cell derived exosomes-based immunological signature in a rat model of corneal allograft rejection therapy. Frontiers in Bioscience-Landmark. 2022; 27: 86.
[17]
Zhang L, Yu D. Exosomes in cancer development, metastasis, and immunity. Biochimica et Biophysica Acta. Reviews on Cancer. 2019; 1871: 455–468.
[18]
Noonin C, Thongboonkerd V. Exosome-inflammasome crosstalk and their roles in inflammatory responses. Theranostics. 2021; 11: 4436–4451.
[19]
Amelimojarad M, AmeliMojarad M, Nazemalhosseini-Mojarad E. Exosomal noncoding RNAs in colorectal cancer: An overview of functions, challenges, opportunities, and clinical applications. Pathology, Research and Practice. 2022; 238: 154133.
[20]
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.
[21]
Johnstone RM, Bianchini A, Teng K. Reticulocyte maturation and exosome release: transferrin receptor containing exosomes shows multiple plasma membrane functions. Blood. 1989; 74: 1844–1851.
[22]
Budnik V, Ruiz-Cañada C, Wendler F. Extracellular vesicles round off communication in the nervous system. Nature Reviews. Neuroscience. 2016; 17: 160–172.
[23]
He C, Zheng S, Luo Y, Wang B. Exosome Theranostics: Biology and Translational Medicine. Theranostics. 2018; 8: 237–255.
[24]
Machtinger R, Laurent LC, Baccarelli AA. Extracellular vesicles: roles in gamete maturation, fertilization and embryo implantation. Human Reproduction Update. 2016; 22: 182–193.
[25]
Yáñez-Mó M, Siljander PRM, Andreu Z, Zavec AB, Borràs FE, Buzas EI, et al. Biological properties of extracellular vesicles and their physiological functions. Journal of Extracellular Vesicles. 2015; 4: 27066.
[26]
Arck PC, Hecher K. Fetomaternal immune cross-talk and its consequences for maternal and offspring’s health. Nature Medicine. 2013; 19: 548–556.
[27]
Jiang N, Xiang L, He L, Yang G, Zheng J, Wang C, et al. Exosomes Mediate Epithelium-Mesenchyme Crosstalk in Organ Development. ACS Nano. 2017; 11: 7736–7746.
[28]
Lo Cicero A, Delevoye C, Gilles-Marsens F, Loew D, Dingli F, Guéré C, et al. Exosomes released by keratinocytes modulate melanocyte pigmentation. Nature Communications. 2015; 6: 7506.
[29]
Nojima H, Freeman CM, Schuster RM, Japtok L, Kleuser B, Edwards MJ, et al. Hepatocyte exosomes mediate liver repair and regeneration via sphingosine-1-phosphate. Journal of Hepatology. 2016; 64: 60–68.
[30]
Raposo G, Nijman HW, Stoorvogel W, Liejendekker R, Harding CV, Melief CJ, et al. B lymphocytes secrete antigen-presenting vesicles. The Journal of Experimental Medicine. 1996; 183: 1161–1172.
[31]
Zitvogel L, Regnault A, Lozier A, Wolfers J, Flament C, Tenza D, et al. Eradication of established murine tumors using a novel cell-free vaccine: dendritic cell-derived exosomes. Nature Medicine. 1998; 4: 594–600.
[32]
Montecalvo A, Shufesky WJ, Stolz DB, Sullivan MG, Wang Z, Divito SJ, et al. Exosomes as a short-range mechanism to spread alloantigen between dendritic cells during T cell allorecognition. Journal of Immunology. 2008; 180: 3081–3090.
[33]
Nandakumar R, Tschismarov R, Meissner F, Prabakaran T, Krissanaprasit A, Farahani E, et al. Intracellular bacteria engage a STING-TBK1-MVB12b pathway to enable paracrine cGAS-STING signalling. Nature Microbiology. 2019; 4: 701–713.
[34]
Yao J, Wang Z, Cheng Y, Ma C, Zhong Y, Xiao Y, et al. M2 macrophage-derived exosomal microRNAs inhibit cell migration and invasion in gliomas through PI3K/AKT/mTOR signaling pathway. Journal of Translational Medicine. 2021; 19: 99.
[35]
Chennakrishnaiah S, Meehan B, D’Asti E, Montermini L, Lee TH, Karatzas N, et al. Leukocytes as a reservoir of circulating oncogenic DNA and regulatory targets of tumor-derived extracellular vesicles. Journal of Thrombosis and Haemostasis: JTH. 2018; 16: 1800–1813.
[36]
Ying X, Wu Q, Wu X, Zhu Q, Wang X, Jiang L, et al. Epithelial ovarian cancer-secreted exosomal miR-222-3p induces polarization of tumor-associated macrophages. Oncotarget. 2016; 7: 43076–43087.
[37]
Costa-Silva B, Aiello NM, Ocean AJ, Singh S, Zhang H, Thakur BK, et al. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nature Cell Biology. 2015; 17: 816–826.
[38]
Fong MY, Zhou W, Liu L, Alontaga AY, Chandra M, Ashby J, et al. Breast-cancer-secreted miR-122 reprograms glucose metabolism in premetastatic niche to promote metastasis. Nature Cell Biology. 2015; 17: 183–194.
[39]
Melo SA, Sugimoto H, O’Connell JT, Kato N, Villanueva A, Vidal A, et al. Cancer exosomes perform cell-independent microRNA biogenesis and promote tumorigenesis. Cancer Cell. 2014; 26: 707–721.
[40]
Choi SH, Kim YH, Hebisch M, Sliwinski C, Lee S, D’Avanzo C, et al. A three-dimensional human neural cell culture model of Alzheimer’s disease. Nature. 2014; 515: 274–278.
[41]
Soltész B, Buglyó G, Németh N, Szilágyi M, Pös O, Szemes T, et al. The Role of Exosomes in Cancer Progression. International Journal of Molecular Sciences. 2021; 23: 8.
[42]
Trams EG, Lauter CJ, Salem N, Jr, Heine U. Exfoliation of membrane ecto-enzymes in the form of micro-vesicles. Biochimica et Biophysica Acta. 1981; 645: 63–70.
[43]
Dioufa N, Clark AM, Ma B, Beckwitt CH, Wells A. Bi-directional exosome-driven intercommunication between the hepatic niche and cancer cells. Molecular Cancer. 2017; 16: 172.
[44]
Kok VC, Yu CC. Cancer-Derived Exosomes: Their Role in Cancer Biology and Biomarker Development. International Journal of Nanomedicine. 2020; 15: 8019–8036.
[45]
Zheng R, Zhang K, Tan S, Gao F, Zhang Y, Xu W, et al. Exosomal circLPAR1 functions in colorectal cancer diagnosis and tumorigenesis through suppressing BRD4 via METTL3-eIF3h interaction. Molecular Cancer. 2022; 21: 49.
[46]
Li C, Li X. Exosome-Derived Circ_0094343 Promotes Chemosensitivity of Colorectal Cancer Cells by Regulating Glycolysis via the miR-766-5p/TRIM67 Axis. Contrast Media & Molecular Imaging. 2022; 2022: 2878557.
[47]
Yang Y, Yang N, Jiang J. Exosomal circ_PTPRA inhibits tumorigenesis and promotes radiosensitivity in colorectal cancer by enriching the level of SMAD4 via competitively binding to miR-671-5p. Cytotechnology. 2022; 74: 51–64.
[48]
Jiang Z, Hou Z, Li L, Liu W, Yu Z, Chen S. Exosomal circEPB41L2 serves as a sponge for miR-21-5p and miR-942-5p to suppress colorectal cancer progression by regulating the PTEN/AKT signalling pathway. European Journal of Clinical Investigation. 2021; 51: e13581.
[49]
Wu F, Yang J, Shang G, Zhang Z, Niu S, Liu Y, et al. Exosomal miR-224-5p from Colorectal Cancer Cells Promotes Malignant Transformation of Human Normal Colon Epithelial Cells by Promoting Cell Proliferation through Downregulation of CMTM4. Oxidative Medicine and Cellular Longevity. 2022; 2022: 5983629.
[50]
Naseri M, Zöller M, Hadjati J, Ghods R, Ranaei Pirmardan E, Kiani J, et al. Dendritic cells loaded with exosomes derived from cancer stem cell-enriched spheroids as a potential immunotherapeutic option. Journal of Cellular and Molecular Medicine. 2021; 25: 3312–3326.
[51]
Zhao S, Mi Y, Guan B, Zheng B, Wei P, Gu Y, et al. Tumor-derived exosomal miR-934 induces macrophage M2 polarization to promote liver metastasis of colorectal cancer. Journal of Hematology & Oncology. 2020; 13: 156.
[52]
Shang A, Gu C, Wang W, Wang X, Sun J, Zeng B, et al. Exosomal circPACRGL promotes progression of colorectal cancer via the miR-142-3p/miR-506-3p- TGF-β1 axis. Molecular Cancer. 2020; 19: 117.
[53]
Yang C, Dou R, Wei C, Liu K, Shi D, Zhang C, et al. Tumor-derived exosomal microRNA-106b-5p activates EMT-cancer cell and M2-subtype TAM interaction to facilitate CRC metastasis. Molecular Therapy: the Journal of the American Society of Gene Therapy. 2021; 29: 2088–2107.
[54]
Sun H, Meng Q, Shi C, Yang H, Li X, Wu S, et al. Hypoxia-Inducible Exosomes Facilitate Liver-Tropic Premetastatic Niche in Colorectal Cancer. Hepatology. 2021; 74: 2633–2651.
[55]
Liang ZX, Liu HS, Wang FW, Xiong L, Zhou C, Hu T, et al. LncRNA RPPH1 promotes colorectal cancer metastasis by interacting with TUBB3 and by promoting exosomes-mediated macrophage M2 polarization. Cell Death & Disease. 2019; 10: 829.
[56]
Yang H, Zhang H, Yang Y, Wang X, Deng T, Liu R, et al. Hypoxia induced exosomal circRNA promotes metastasis of Colorectal Cancer via targeting GEF-H1/RhoA axis. Theranostics. 2020; 10: 8211–8226.
[57]
Guo S, Chen J, Chen F, Zeng Q, Liu WL, Zhang G. Exosomes derived from Fusobacterium nucleatum-infected colorectal cancer cells facilitate tumour metastasis by selectively carrying miR-1246/92b-3p/27a-3p and CXCL16. Gut. 2020. (Online ahead of print)
[58]
Tian F, Wang P, Lin D, Dai J, Liu Q, Guan Y, et al. Exosome-delivered miR-221/222 exacerbates tumor liver metastasis by targeting SPINT1 in colorectal cancer. Cancer Science. 2021; 112: 3744–3755.
[59]
Sun X, Lin F, Sun W, Zhu W, Fang D, Luo L, et al. Exosome-transmitted miRNA-335-5p promotes colorectal cancer invasion and metastasis by facilitating EMT via targeting RASA1. Molecular Therapy. Nucleic Acids. 2021; 24: 164–174.
[60]
Jiang Y, Ji X, Liu K, Shi Y, Wang C, Li Y, et al. Exosomal miR-200c-3p negatively regulates the migraion and invasion of lipopolysaccharide (LPS)-stimulated colorectal cancer (CRC). BMC Molecular and Cell Biology. 2020; 21: 48.
[61]
Liu H, Liu Y, Sun P, Leng K, Xu Y, Mei L, et al. Colorectal cancer-derived exosomal miR-106b-3p promotes metastasis by down-regulating DLC-1 expression. Clinical Science. 2020; 134: 419–434.
[62]
Xu J, Xiao Y, Liu B, Pan S, Liu Q, Shan Y, et al. Exosomal MALAT1 sponges miR-26a/26b to promote the invasion and metastasis of colorectal cancer via FUT4 enhanced fucosylation and PI3K/Akt pathway. Journal of Experimental & Clinical Cancer Research: CR. 2020; 39: 54.
[63]
Shao Y, Chen T, Zheng X, Yang S, Xu K, Chen X, et al. Colorectal cancer-derived small extracellular vesicles establish an inflammatory premetastatic niche in liver metastasis. Carcinogenesis. 2018; 39: 1368–1379.
[64]
Huang Z, Yang M, Li Y, Yang F, Feng Y. Exosomes Derived from Hypoxic Colorectal Cancer Cells Transfer Wnt4 to Normoxic Cells to Elicit a Prometastatic Phenotype. International Journal of Biological Sciences. 2018; 14: 2094–2102.
[65]
Fang X, Xu Y, Li K, Liu P, Zhang H, Jiang Y, et al. Exosomal lncRNA PCAT1 Promotes Tumor Circulating Cell-Mediated Colorectal Cancer Liver Metastasis by Regulating the Activity of the miR-329-3p/Netrin-1-CD146 Complex. Journal of Immunology Research. 2022; 2022: 9916228.
[66]
Wang J, Liu Y, Li Y, Zheng X, Gan J, Wan Z, et al. Exosomal miR 10a derived from colorectal cancer cells suppresses migration of human lung fibroblasts, and expression of IL 6, IL 8 and IL 1β. Molecular Medicine Reports. 2021; 23: 84.
[67]
Zhang X, Bai J, Yin H, Long L, Zheng Z, Wang Q, et al. Exosomal miR-1255b-5p targets human telomerase reverse transcriptase in colorectal cancer cells to suppress epithelial-to-mesenchymal transition. Molecular Oncology. 2020; 14: 2589–2608.
[68]
Zeng Z, Li Y, Pan Y, Lan X, Song F, Sun J, et al. Cancer-derived exosomal miR-25-3p promotes pre-metastatic niche formation by inducing vascular permeability and angiogenesis. Nature Communications. 2018; 9: 5395.
[69]
He Q, Ye A, Ye W, Liao X, Qin G, Xu Y, et al. Cancer-secreted exosomal miR-21-5p induces angiogenesis and vascular permeability by targeting KRIT1. Cell Death & Disease. 2021; 12: 576.
[70]
Hu HY, Yu CH, Zhang HH, Zhang SZ, Yu WY, Yang Y, et al. Exosomal miR-1229 derived from colorectal cancer cells promotes angiogenesis by targeting HIPK2. International Journal of Biological Macromolecules. 2019; 132: 470–477.
[71]
Huang Z, Feng Y. Exosomes Derived From Hypoxic Colorectal Cancer Cells Promote Angiogenesis Through Wnt4-Induced β-Catenin Signaling in Endothelial Cells. Oncology Research. 2017; 25: 651–661.
[72]
Ning T, Li J, He Y, Zhang H, Wang X, Deng T, et al. Exosomal miR-208b related with oxaliplatin resistance promotes Treg expansion in colorectal cancer. Molecular Therapy: the Journal of the American Society of Gene Therapy. 2021; 29: 2723–2736.
[73]
Huang Y, Luo Y, Ou W, Wang Y, Dong D, Peng X, et al. Exosomal lncRNA SNHG10 derived from colorectal cancer cells suppresses natural killer cell cytotoxicity by upregulating INHBC. Cancer Cell International. 2021; 21: 528.
[74]
Li X, Lan Q, Lai W, Wu H, Xu H, Fang K, et al. Exosome-derived lnc-HOXB8-1:2 induces tumor-associated macrophage infiltration to promote neuroendocrine differentiated colorectal cancer progression by sponging hsa-miR-6825-5p. BMC Cancer. 2022; 22: 928.
[75]
Dai W, Zhou J, Wang H, Zhang M, Yang X, Song W. miR-424-5p promotes the proliferation and metastasis of colorectal cancer by directly targeting SCN4B. Pathology, Research and Practice. 2020; 216: 152731.
[76]
Sun W, Cui J, Ge Y, Wang J, Yu Y, Han B, et al. Tumor stem cell-derived exosomal microRNA-17-5p inhibits anti-tumor immunity in colorectal cancer via targeting SPOP and overexpressing PD-L1. Cell Death Discovery. 2022; 8: 223.
[77]
Xie Z, Xia J, Jiao M, Zhao P, Wang Z, Lin S, et al. Exosomal lncRNA HOTAIR induces PDL1+ B cells to impede anti-tumor immunity in colorectal cancer. Biochemical and Biophysical Research Communications. 2023; 644: 112–121.
[78]
Wang D, Wang X, Song Y, Si M, Sun Y, Liu X, et al. Exosomal miR-146a-5p and miR-155-5p promote CXCL12/CXCR7-induced metastasis of colorectal cancer by crosstalk with cancer-associated fibroblasts. Cell Death & Disease. 2022; 13: 380.
[79]
Zhang C, Wang XY, Zhang P, He TC, Han JH, Zhang R, et al. Cancer-derived exosomal HSPC111 promotes colorectal cancer liver metastasis by reprogramming lipid metabolism in cancer-associated fibroblasts. Cell Death & Disease. 2022; 13: 57.
[80]
Jiang K, Chen H, Fang Y, Chen L, Zhong C, Bu T, et al. Exosomal ANGPTL1 attenuates colorectal cancer liver metastasis by regulating Kupffer cell secretion pattern and impeding MMP9 induced vascular leakiness. Journal of Experimental & Clinical Cancer Research: CR. 2021; 40: 21.
[81]
Hwang WL, Lan HY, Cheng WC, Huang SC, Yang MH. Tumor stem-like cell-derived exosomal RNAs prime neutrophils for facilitating tumorigenesis of colon cancer. Journal of Hematology & Oncology. 2019; 12: 10.
[82]
Dai G, Yao X, Zhang Y, Gu J, Geng Y, Xue F, et al. Colorectal cancer cell-derived exosomes containing miR-10b regulate fibroblast cells via the PI3K/Akt pathway. Bulletin Du Cancer. 2018; 105: 336–349.
[83]
Takano Y, Masuda T, Iinuma H, Yamaguchi R, Sato K, Tobo T, et al. Circulating exosomal microRNA-203 is associated with metastasis possibly via inducing tumor-associated macrophages in colorectal cancer. Oncotarget. 2017; 8: 78598–78613.
[84]
Tao L, Xu C, Shen W, Tan J, Li L, Fan M, et al. HIPK3 Inhibition by Exosomal hsa-miR-101-3p Is Related to Metabolic Reprogramming in Colorectal Cancer. Frontiers in Oncology. 2022; 11: 758336.
[85]
Zhang Q, Jeppesen DK, Higginbotham JN, Demory Beckler M, Poulin EJ, Walsh AJ, et al. Mutant KRAS Exosomes Alter the Metabolic State of Recipient Colonic Epithelial Cells. Cellular and Molecular Gastroenterology and Hepatology. 2018; 5: 627–629.e6.
[86]
Wang X, Zhang H, Yang H, Bai M, Ning T, Deng T, et al. Exosome-delivered circRNA promotes glycolysis to induce chemoresistance through the miR-122-PKM2 axis in colorectal cancer. Molecular Oncology. 2020; 14: 539–555.
[87]
Zhang Q, Liu RX, Chan KW, Hu J, Zhang J, Wei L, et al. Exosomal transfer of p-STAT3 promotes acquired 5-FU resistance in colorectal cancer cells. Journal of Experimental & Clinical Cancer Research: CR. 2019; 38: 320.
[88]
Zhao K, Cheng X, Ye Z, Li Y, Peng W, Wu Y, et al. Exosome-Mediated Transfer of circ_0000338 Enhances 5-Fluorouracil Resistance in Colorectal Cancer through Regulating MicroRNA 217 (miR-217) and miR-485-3p. Molecular and Cellular Biology. 2021; 41: e00517–e00520.
[89]
Xu Y, Zhu M. Novel exosomal miR-46146 transfer oxaliplatin chemoresistance in colorectal cancer. Clinical & Translational Oncology: Official Publication of the Federation of Spanish Oncology Societies and of the National Cancer Institute of Mexico. 2020; 22: 1105–1116.
[90]
Sun T, Yin YF, Jin HG, Liu HR, Tian WC. Exosomal microRNA-19b targets FBXW7 to promote colorectal cancer stem cell stemness and induce resistance to radiotherapy. The Kaohsiung Journal of Medical Sciences. 2022; 38: 108–119.
[91]
Xu Y, Qiu A, Peng F, Tan X, Wang J, Gong X. Exosomal transfer of circular RNA FBXW7 ameliorates the chemoresistance to oxaliplatin in colorectal cancer by sponging miR-18b-5p. Neoplasma. 2021; 68: 108–118.
[92]
Chen X, Liu Y, Zhang Q, Liu B, Cheng Y, Zhang Y, et al. Exosomal Long Non-coding RNA HOTTIP Increases Resistance of Colorectal Cancer Cells to Mitomycin via Impairing MiR-214-Mediated Degradation of KPNA3. Frontiers in Cell and Developmental Biology. 2021; 8: 582723.
[93]
Hu X, Mu Y, Liu J, Mu X, Gao F, Chen L, et al. Exosomes Derived from Hypoxic Colorectal Cancer Cells Transfer miR-410-3p to Regulate Tumor Progression. Journal of Cancer. 2020; 11: 4724–4735.
[94]
Goldstein DA, Zeichner SB, Bartnik CM, Neustadter E, Flowers CR. Metastatic Colorectal Cancer: A Systematic Review of the Value of Current Therapies. Clinical Colorectal Cancer. 2016; 15: 1–6.
[95]
Wang D, Wang X, Si M, Yang J, Sun S, Wu H, et al. Exosome-encapsulated miRNAs contribute to CXCL12/CXCR4-induced liver metastasis of colorectal cancer by enhancing M2 polarization of macrophages. Cancer Letters. 2020; 474: 36–52.
[96]
Dou R, Liu K, Yang C, Zheng J, Shi D, Lin X, et al. EMT-cancer cells-derived exosomal miR-27b-3p promotes circulating tumour cells-mediated metastasis by modulating vascular permeability in colorectal cancer. Clinical and Translational Medicine. 2021; 11: e595.
[97]
Xiao H, Ye X, Vishwakarma V, Preet R, Dixon DA. CRC-derived exosomes containing the RNA binding protein HuR promote lung cell proliferation by stabilizing c-Myc mRNA. Cancer Biology & Therapy. 2022; 23: 139–149.
[98]
Cho WC, Kim M, Park JW, Jeong SY, Ku JL. Exosomal miR-193a and let-7g accelerate cancer progression on primary colorectal cancer and paired peritoneal metastatic cancer. Translational Oncology. 2021; 14: 101000.
[99]
Sun B, Zhou Y, Fang Y, Li Z, Gu X, Xiang J. Colorectal cancer exosomes induce lymphatic network remodeling in lymph nodes. International Journal of Cancer. 2019; 145: 1648–1659.
[100]
Li Y, Hu J, Wang M, Yuan Y, Zhou F, Zhao H, et al. Exosomal circPABPC1 promotes colorectal cancer liver metastases by regulating HMGA2 in the nucleus and BMP4/ADAM19 in the cytoplasm. Cell Death Discovery. 2022; 8: 335.
[101]
Zhao H, Chen S, Fu Q. Exosomes from CD133+ cells carrying circ-ABCC1 mediate cell stemness and metastasis in colorectal cancer. Journal of Cellular Biochemistry. 2020; 121: 3286–3297.
[102]
Sun Z, Yang S, Zhou Q, Wang G, Song J, Li Z, et al. Emerging role of exosome-derived long non-coding RNAs in tumor microenvironment. Molecular Cancer. 2018; 17: 82.
[103]
Yang K, Zhou Q, Qiao B, Shao B, Hu S, Wang G, et al. Exosome-derived noncoding RNAs: Function, mechanism, and application in tumor angiogenesis. Molecular Therapy. Nucleic Acids. 2022; 27: 983–997.
[104]
Yang E, Wang X, Gong Z, Yu M, Wu H, Zhang D. Exosome-mediated metabolic reprogramming: the emerging role in tumor microenvironment remodeling and its influence on cancer progression. Signal Transduction and Targeted Therapy. 2020; 5: 242.
[105]
Fridman ES, Ginini L, Gil Z. The Role of Extracellular Vesicles in Metabolic Reprogramming of the Tumor Microenvironment. Cells. 2022; 11: 1433.
[106]
Shedden K, Xie XT, Chandaroy P, Chang YT, Rosania GR. Expulsion of small molecules in vesicles shed by cancer cells: association with gene expression and chemosensitivity profiles. Cancer Research. 2003; 63: 4331–4337.
[107]
Corrado C, Raimondo S, Chiesi A, Ciccia F, De Leo G, Alessandro R. Exosomes as intercellular signaling organelles involved in health and disease: basic science and clinical applications. International Journal of Molecular Sciences. 2013; 14: 5338–5366.
[108]
Pan Z, Zheng J, Zhang J, Lin J, Lai J, Lyu Z, et al. A Novel Protein Encoded by Exosomal CircATG4B Induces Oxaliplatin Resistance in Colorectal Cancer by Promoting Autophagy. Advanced Science. 2022; 9: e2204513.
[109]
Mashouri L, Yousefi H, Aref AR, Ahadi AM, Molaei F, Alahari SK. Exosomes: composition, biogenesis, and mechanisms in cancer metastasis and drug resistance. Molecular Cancer. 2019; 18: 75.
[110]
Hu JL, Wang W, Lan XL, Zeng ZC, Liang YS, Yan YR, et al. CAFs secreted exosomes promote metastasis and chemotherapy resistance by enhancing cell stemness and epithelial-mesenchymal transition in colorectal cancer. Molecular Cancer. 2019; 18: 91.
[111]
Ren J, Ding L, Zhang D, Shi G, Xu Q, Shen S, et al. Carcinoma-associated fibroblasts promote the stemness and chemoresistance of colorectal cancer by transferring exosomal lncRNA H19. Theranostics. 2018; 8: 3932–3948.
[112]
Chen X, Liu J, Zhang Q, Liu B, Cheng Y, Zhang Y, et al. Exosome-mediated transfer of miR-93-5p from cancer-associated fibroblasts confer radioresistance in colorectal cancer cells by downregulating FOXA1 and upregulating TGFB3. Journal of Experimental & Clinical Cancer Research: CR. 2020; 39: 65.
[113]
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. Journal of Translational Medicine. 2021; 19: 8.
[114]
Yang K, Zhang J, Bao C. Exosomal circEIF3K from cancer-associated fibroblast promotes colorectal cancer (CRC) progression via miR-214/PD-L1 axis. BMC Cancer. 2021; 21: 933.
[115]
Pan S, Deng Y, Fu J, Zhang Y, Zhang Z, Qin X. N6 methyladenosine upregulates miR 181d 5p in exosomes derived from cancer associated fibroblasts to inhibit 5 FU sensitivity by targeting NCALD in colorectal cancer. International Journal of Oncology. 2022; 60: 14.
[116]
Jiang Y, Qiu Q, Jing X, Song Z, Zhang Y, Wang C, et al. Cancer-associated fibroblast-derived exosome miR-181b-3p promotes the occurrence and development of colorectal cancer by regulating SNX2 expression. Biochemical and Biophysical Research Communications. 2023; 641: 177–185.
[117]
Qu Z, Yang KD, Luo BH, Zhang F. CAFs-secreted exosomal cricN4BP2L2 promoted colorectal cancer stemness and chemoresistance by interacting with EIF4A3. Experimental Cell Research. 2022; 418: 113266.
[118]
Chen X, Liu Y, Zhang Q, Liu B, Cheng Y, Zhang Y, et al. Exosomal miR-590-3p derived from cancer-associated fibroblasts confers radioresistance in colorectal cancer. Molecular Therapy. Nucleic Acids. 2020; 24: 113–126.
[119]
Zhang Y, Yin C, Wei C, Xia S, Qiao Z, Zhang XW, et al. Exosomal miR-625-3p secreted by cancer-associated fibroblasts in colorectal cancer promotes EMT and chemotherapeutic resistance by blocking the CELF2/WWOX pathway. Pharmacological Research. 2022; 186: 106534.
[120]
Yin H, Yu S, Xie Y, Dai X, Dong M, Sheng C, et al. Cancer-associated fibroblasts-derived exosomes upregulate microRNA-135b-5p to promote colorectal cancer cell growth and angiogenesis by inhibiting thioredoxin-interacting protein. Cellular Signalling. 2021; 84: 110029.
[121]
Yang P, Zhang D, Wang T, Ji J, Jin C, Peng C, et al. CAF-derived exosomal WEE2-AS1 facilitates colorectal cancer progression via promoting degradation of MOB1A to inhibit the Hippo pathway. Cell Death & Disease. 2022; 13: 796.
[122]
Yuan H, Chen B, Chai R, Gong W, Wan Z, Zheng B, et al. Loss of exosomal micro-RNA-200b-3p from hypoxia cancer-associated fibroblasts reduces sensitivity to 5-flourouracil in colorectal cancer through targeting high-mobility group box 3. Frontiers in Oncology. 2022; 12: 920131.
[123]
Zhang Y, Wang S, Lai Q, Fang Y, Wu C, Liu Y, et al. Cancer-associated fibroblasts-derived exosomal miR-17-5p promotes colorectal cancer aggressive phenotype by initiating a RUNX3/MYC/TGF-β1 positive feedback loop. Cancer Letters. 2020; 491: 22–35.
[124]
Deng X, Ruan H, Zhang X, Xu X, Zhu Y, Peng H, et al. Long noncoding RNA CCAL transferred from fibroblasts by exosomes promotes chemoresistance of colorectal cancer cells. International Journal of Cancer. 2020; 146: 1700–1716.
[125]
Hu YB, Yan C, Mu L, Mi YL, Zhao H, Hu H, et al. Exosomal Wnt-induced dedifferentiation of colorectal cancer cells contributes to chemotherapy resistance. Oncogene. 2019; 38: 1951–1965.
[126]
Jahangiri B, Khalaj-Kondori M, Asadollahi E, Purrafee Dizaj L, Sadeghizadeh M. MSC-Derived exosomes suppress colorectal cancer cell proliferation and metastasis via miR-100/mTOR/miR-143 pathway. International Journal of Pharmaceutics. 2022; 627: 122214.
[127]
Li T, Wan Y, Su Z, Li J, Han M, Zhou C. Mesenchymal Stem Cell-Derived Exosomal microRNA-3940-5p Inhibits Colorectal Cancer Metastasis by Targeting Integrin α6. Digestive Diseases and Sciences. 2021; 66: 1916–1927.
[128]
Chen HL, Li JJ, Jiang F, Shi WJ, Chang GY. MicroRNA-4461 derived from bone marrow mesenchymal stem cell exosomes inhibits tumorigenesis by downregulating COPB2 expression in colorectal cancer. Bioscience, Biotechnology, and Biochemistry. 2020; 84: 338–346.
[129]
Zhang N, Li L, Luo J, Tan J, Hu W, Li Z, et al. Inhibiting microRNA-424 in bone marrow mesenchymal stem cells-derived exosomes suppresses tumor growth in colorectal cancer by upregulating TGFBR3. Archives of Biochemistry and Biophysics. 2021; 709: 108965.
[130]
Xu Y, Shen L, Li F, Yang J, Wan X, Ouyang M. microRNA-16-5p-containing exosomes derived from bone marrow-derived mesenchymal stem cells inhibit proliferation, migration, and invasion, while promoting apoptosis of colorectal cancer cells by downregulating ITGA2. Journal of Cellular Physiology. 2019; 234: 21380–21394.
[131]
Wang Y, Lin C. Exosomes miR-22-3p Derived from Mesenchymal Stem Cells Suppress Colorectal Cancer Cell Proliferation and Invasion by Regulating RAP2B and PI3K/AKT Pathway. Journal of Oncology. 2021; 2021: 3874478.
[132]
Wang Y, Yin K, Tian J, Xia X, Ma J, Tang X, et al. Granulocytic Myeloid-Derived Suppressor Cells Promote the Stemness of Colorectal Cancer Cells through Exosomal S100A9. Advanced Science. 2019; 6: 1901278.
[133]
Zhang Q, Deng T, Zhang H, Zuo D, Zhu Q, Bai M, et al. Adipocyte-Derived Exosomal MTTP Suppresses Ferroptosis and Promotes Chemoresistance in Colorectal Cancer. Advanced Science. 2022; 9: e2203357.
[134]
Xu H, Lan Q, Huang Y, Zhang Y, Zeng Y, Su P, et al. The mechanisms of colorectal cancer cell mesenchymal-epithelial transition induced by hepatocyte exosome-derived miR-203a-3p. BMC Cancer. 2021; 21: 718.
[135]
Yu W, Hurley J, Roberts D, Chakrabortty SK, Enderle D, Noerholm M, et al. Exosome-based liquid biopsies in cancer: opportunities and challenges. Annals of Oncology: Official Journal of the European Society for Medical Oncology. 2021; 32: 466–477.
[136]
Wang L, Song X, Yu M, Niu L, Zhao Y, Tang Y, et al. Serum exosomal miR-377-3p and miR-381-3p as diagnostic biomarkers in colorectal cancer. Future Oncology. 2022; 18: 793–805.
[137]
Gao L, Tang X, He Q, Sun G, Wang C, Qu H. Exosome-transmitted circCOG2 promotes colorectal cancer progression via miR-1305/TGF-β2/SMAD3 pathway. Cell Death Discovery. 2021; 7: 281.
[138]
Cui X, Lv Z, Ding H, Xing C, Yuan Y. MiR-1539 and Its Potential Role as a Novel Biomarker for Colorectal Cancer. Frontiers in Oncology. 2021; 10: 531244.
[139]
Zhao YJ, Song X, Niu L, Tang Y, Song X, Xie L. Circulating Exosomal miR-150-5p and miR-99b-5p as Diagnostic Biomarkers for Colorectal Cancer. Frontiers in Oncology. 2019; 9: 1129.
[140]
Qiao D, Gu C, Wang W, Yan W, Jiang C, Hu J, et al. Tumor-Originated Exosomal hsa-miR-3937 as a Minimally Invasive Early Biomarker for Liquid Biopsy of Colorectal Cancer. Journal of Oncology. 2022; 2022: 6990955.
[141]
Peng ZY, Gu RH, Yan B. Downregulation of exosome-encapsulated miR-548c-5p is associated with poor prognosis in colorectal cancer. Journal of Cellular Biochemistry. 2019; 120: 1457–1463.
[142]
Sun B, Li Y, Zhou Y, Ng TK, Zhao C, Gan Q, et al. Circulating exosomal CPNE3 as a diagnostic and prognostic biomarker for colorectal cancer. Journal of Cellular Physiology. 2019; 234: 1416–1425.
[143]
Liu W, Yang D, Chen L, Liu Q, Wang W, Yang Z, et al. Plasma Exosomal miRNA-139-3p is a Novel Biomarker of Colorectal Cancer. Journal of Cancer. 2020; 11: 4899–4906.
[144]
Tang Y, Zhao Y, Song X, Song X, Niu L, Xie L. Tumor-derived exosomal miRNA-320d as a biomarker for metastatic colorectal cancer. Journal of Clinical Laboratory Analysis. 2019; 33: e23004.
[145]
Liu L, Meng T, Yang XH, Sayim P, Lei C, Jin B, et al. 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 Biomarkers: Section a of Disease Markers. 2018; 22: 283–299.
[146]
Pan B, Qin J, Liu X, He B, Wang X, Pan Y, et al. Identification of Serum Exosomal hsa-circ-0004771 as a Novel Diagnostic Biomarker of Colorectal Cancer. Frontiers in Genetics. 2019; 10: 1096.
[147]
Zhang Y, Tan X, Lu Y. Exosomal transfer of circ_0006174 contributes to the chemoresistance of doxorubicin in colorectal cancer by depending on the miR-1205/CCND2 axis. Journal of Physiology and Biochemistry. 2022; 78: 39–50.
[148]
Zhang N, Zhang PP, Huang JJ, Wang ZY, Zhang ZH, Yuan JZ, et al. Reduced serum exosomal miR-874 expression predicts poor prognosis in colorectal cancer. European Review for Medical and Pharmacological Sciences. 2020; 24: 664–672.
[149]
Sun Z, Ji S, Wu J, Tian J, Quan W, Shang A, et al. Proteomics-Based Identification of Candidate Exosomal Glycoprotein Biomarkers and Their Value for Diagnosing Colorectal Cancer. Frontiers in Oncology. 2021; 11: 725211.
[150]
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. Translational Oncology. 2018; 11: 221–232.
[151]
Yu M, Song XG, Zhao YJ, Dong XH, Niu LM, Zhang ZJ, et al. Circulating Serum Exosomal Long Non-Coding RNAs FOXD2-AS1, NRIR, and XLOC_009459 as Diagnostic Biomarkers for Colorectal Cancer. Frontiers in Oncology. 2021; 11: 618967.
[152]
Tsukamoto M, Iinuma H, Yagi T, Matsuda K, Hashiguchi Y. Circulating Exosomal MicroRNA-21 as a Biomarker in Each Tumor Stage of Colorectal Cancer. Oncology. 2017; 92: 360–370.
[153]
Yan S, Liu G, Jin C, Wang Z, Duan Q, Xu J, et al. MicroRNA-6869-5p acts as a tumor suppressor via targeting TLR4/NF-κB signaling pathway in colorectal cancer. Journal of Cellular Physiology. 2018; 233: 6660–6668.
[154]
Sun L, Liu X, Pan B, Hu X, Zhu Y, Su Y, et al. Serum exosomal miR-122 as a potential diagnostic and prognostic biomarker of colorectal cancer with liver metastasis. Journal of Cancer. 2020; 11: 630–637.
[155]
Xie Y, Li J, Li P, Li N, Zhang Y, Binang H, et al. RNA-Seq Profiling of Serum Exosomal Circular RNAs Reveals Circ-PNN as a Potential Biomarker for Human Colorectal Cancer. Frontiers in Oncology. 2020; 10: 982.
[156]
Yang YN, Zhang R, Du JW, Yuan HH, Li YJ, Wei XL, et al. Predictive role of UCA1-containing exosomes in cetuximab-resistant colorectal cancer. Cancer Cell International. 2018; 18: 164.
[157]
Zhang Y, Liu WS, Zhang XY, Tong HX, Yang H, Liu WF, et al. Low expression of exosomal miR-150 predicts poor prognosis in colorectal cancer patients after surgical resections. Carcinogenesis. 2022; 43: 930–940.
[158]
Ganig N, Baenke F, Thepkaysone ML, Lin K, Rao VS, Wong FC, et al. Proteomic Analyses of Fibroblast- and Serum-Derived Exosomes Identify QSOX1 as a Marker for Non-invasive Detection of Colorectal Cancer. Cancers. 2021; 13: 1351.
[159]
Zou SL, Chen YL, Ge ZZ, Qu YY, Cao Y, Kang ZX. Downregulation of serum exosomal miR-150-5p is associated with poor prognosis in patients with colorectal cancer. Cancer Biomarkers: Section a of Disease Markers. 2019; 26: 69–77.
[160]
Lucchetti D, Zurlo IV, Colella F, Ricciardi-Tenore C, Di Salvatore M, Tortora G, et al. Mutational status of plasma exosomal KRAS predicts outcome in patients with metastatic colorectal cancer. Scientific Reports. 2021; 11: 22686.
[161]
Sur D, Balacescu L, Cainap SS, Visan S, Pop L, Burz C, et al. Predictive Efficacy of MiR-125b-5p, MiR-17-5p, and MiR-185-5p in Liver Metastasis and Chemotherapy Response Among Advanced Stage Colorectal Cancer Patients. Frontiers in Oncology. 2021; 11: 651380.
[162]
Min L, Chen L, Liu S, Yu Y, Guo Q, Li P, et al. Loss of Circulating Exosomal miR-92b is a Novel Biomarker of Colorectal Cancer at Early Stage. International Journal of Medical Sciences. 2019; 16: 1231–1237.
[163]
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. Biomedicine & Pharmacotherapy. 2019; 113: 108758.
[164]
Shi Y, Zhuang Y, Zhang J, Chen M, Wu S. Four circulating exosomal miRNAs as novel potential biomarkers for the early diagnosis of human colorectal cancer. Tissue & Cell. 2021; 70: 101499.
[165]
Yagi T, Iinuma H, Hayama T, Matsuda K, Nozawa K, Tsukamoto M, et al. Plasma exosomal microRNA-125b as a monitoring biomarker of resistance to mFOLFOX6-based chemotherapy in advanced and recurrent colorectal cancer patients. Molecular and Clinical Oncology. 2019; 11: 416–424.
[166]
Miyazaki K, Wada Y, Okuno K, Murano T, Morine Y, Ikemoto T, et al. An exosome-based liquid biopsy signature for pre-operative identification of lymph node metastasis in patients with pathological high-risk T1 colorectal cancer. Molecular Cancer. 2023; 22: 2.
[167]
Chen M, Xu R, Rai A, Suwakulsiri W, Izumikawa K, Ishikawa H, et al. Distinct shed microvesicle and exosome microRNA signatures reveal diagnostic markers for colorectal cancer. PLoS ONE. 2019; 14: e0210003.
[168]
Scavo MP, Rizzi F, Depalo N, Armentano R, Coletta S, Serino G, et al. Exosome Released FZD10 Increases Ki-67 Expression via Phospho-ERK1/2 in Colorectal and Gastric Cancer. Frontiers in Oncology. 2021; 11: 730093.
[169]
Li J, Chen Y, Guo X, Zhou L, Jia Z, Peng Z, et al. GPC1 exosome and its regulatory miRNAs are specific markers for the detection and target therapy of colorectal cancer. Journal of Cellular and Molecular Medicine. 2017; 21: 838–847.
[170]
Heydari R, Koohi F, Rasouli M, Rezaei K, Abbasgholinejad E, Bekeschus S, et al. Exosomes as Rheumatoid Arthritis Diagnostic Biomarkers and Therapeutic Agents. Vaccines. 2023; 11: 687.
[171]
Liang G, Zhu Y, Ali DJ, Tian T, Xu H, Si K, et al. Engineered exosomes for targeted co-delivery of miR-21 inhibitor and chemotherapeutics to reverse drug resistance in colon cancer. Journal of Nanobiotechnology. 2020; 18: 10.
[172]
Hui B, Lu C, Wang J, Xu Y, Yang Y, Ji H, et al. Engineered exosomes for co-delivery of PGM5-AS1 and oxaliplatin to reverse drug resistance in colon cancer. Journal of Cellular Physiology. 2022; 237: 911–933.
[173]
Xuan B, Wang Y. Exosome-Transmitted miR-506-3p Inhibits Colorectal Cancer Cell Malignancy via Regulating GSTP1. Applied Biochemistry and Biotechnology. 2023; 195: 2015–2027.
[174]
Hosseini M, Baghaei K, Hajivalili M, Zali MR, Ebtekar M, Amani D. The anti-tumor effects of CT-26 derived exosomes enriched by MicroRNA-34a on murine model of colorectal cancer. Life Sciences. 2022; 290: 120234.
[175]
Dai S, Wei D, Wu Z, Zhou X, Wei X, Huang H, et al. Phase I clinical trial of autologous ascites-derived exosomes combined with GM-CSF for colorectal cancer. Molecular Therapy: the Journal of the American Society of Gene Therapy. 2008; 16: 782–790.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top