Skip to main content

The application of extracellular vesicles in colorectal cancer metastasis and drug resistance: recent advances and trends

Abstract

Colorectal cancer (CRC) has high incidence and mortality rates and is one of the most common cancers of the digestive tract worldwide. Metastasis and drug resistance are the main causes of cancer treatment failure. Studies have recently suggested extracellular vesicles (EVs) as a novel mechanism for intercellular communication. They are vesicular particles, which are secreted and released into biological fluids, such as blood, urine, milk, etc., by a variety of cells and carry numerous biologically active molecules, including proteins, nucleic acids, lipids, metabolites, etc. EVs play a crucial part in the metastasis and drug resistance of CRC by delivering cargo to recipient cells and modulating their behavior. An in-depth exploration of EVs might facilitate a comprehensive understanding of the biological behavior of CRC metastasis and drug resistance, which might provide a basis for developing therapeutic strategies. Therefore, considering the specific biological properties of EVs, researchers have attempted to explore their potential as next-generation delivery systems. On the other hand, EVs have also been demonstrated as biomarkers for the prediction, diagnosis, and presumed prognosis of CRC. This review focuses on the role of EVs in regulating the metastasis and chemoresistance of CRC. Moreover, the clinical applications of EVs are also discussed.

Graphical Abstract

Introduction

Colorectal cancer (CRC) is the third most deadly cancer worldwide, with high incidence and mortality [1]. Globally, approximately 10.0% of cancer incidence and 9.4% of cancer-related mortality are caused by CRC, and nearly 900,000 people die from CRC each year [1, 2]. Smoking, alcohol intake, low intake of vegetables and fruits, and excessive obesity are high-risk factors for the development of CRC [2]. Metastasis and drug resistance are major challenges for CRC treatment.

Recent studies have explored the regulatory pathways associated with extracellular vesicles (EVs) in CRC metastasis and drug resistance. EVs are a collective term, covering a variety of cell-released subtypes of membrane structures [3]. Since their contents, membrane composition, and size depend on their cell of origin and mode of biogenesis, EVs are a highly heterogeneous assemblage of subpopulations [3]. Despite their high heterogeneity, EVs can be classified based on the biogenesis patterns and characteristics into four subtypes, including exosomes, microvesicles, midbody remnants, and apoptotic vesicles (Fig. 1). For a clearer discussion, this review has focused on the EVs subtypes formed by interaction with the plasma membrane: exosomes, and microvesicles.

Fig. 1
figure 1

EVs biogenesis and structure. a the shed midbody remnants are released during cytoplasmic division and have a diameter, ranging from 200 to 600 nm; b exosomes are the smallest EV subpopulation, produced by the fusion of MVBs with the cell surface and the release of intraluminal vesicles (ILVs), with a size of 50–200 nm; c microvesicles are larger than exosomes, formed by the direct outward budding of the plasma membrane, with a diameter range of 100–1000 nm. d apoptotic vesicles are found during apoptosis with sizes, ranging from 100 to 5000 nm [4, 5]

EVs are phospholipid bilayer-based substances, which are found in a variety of bodily fluids, including blood, urine, and milk. They contain a large number of proteins, nucleic acids, lipids, and metabolites (Fig. 1) [6, 7]. And they can be obtained by differential centrifugation, density-gradient centrifugation, sucrose buffered centrifugation, affinity capture, gel-permeation chromatography, precipitation, microfluidic devices, and membrane filtration separations [8]. In terms of structure and composition (Fig. 1), the protein components in EVs include (a) membrane transport and fusion-related proteins, such as annexin, Rab-GTPase, and heat shock proteins (HSP); (b) tetraspanins, such as CD9, CD63, CD81, CD82, and intercellular cell adhesion molecule (ICAM) -1; (c) multivesicular bodies (MVBs) -related proteins, including ALIX and TSG101; and (d) other proteins, such as integrins [9]. All these proteins are essential for the EVs function. The lipid components in EVs include ceramide, cholesterol, phosphatidylserine, and sphingomyelin [10]. Notably, the CD9, CD63, CD81, CD82, endosomal sorting complex required for transport (ESCRT) proteins (TSG101 and ALIX), flotillin, ceramide, major histocompatibility complex (MHC), and HSP70/90 have been identified as potential markers for the subgroups of EVs [11]. Functionally, EVs are involved in many physiological and pathological processes. Their biological functions mainly depend on the transfer of active substances, such as non-coding RNAs, proteins, and nucleic acids, to nearby or distant receptor cells, thereby altering the behavior of receptor cells. In CRC, EVs can regulate the function and phenotype of recipient cells by transferring the cancer-associated cargoes, thereby contributing to the alteration of the microenvironment surrounding the CRC cells and enhancing their viability (Fig. 2) [12]. In addition, due to their specific biological properties, EVs are considered a biomarker for the prediction, diagnosis, and presumed prognosis of CRC. Furthermore, researchers have invested great efforts to explore their potential as a next-generation delivery system. This review focuses on the role of EVs in regulating the metastasis and chemoresistance of CRC and discusses the strategies for the clinical applications of EVs.

Fig. 2
figure 2

Diagram of EVs transport. EVs secreted by tumor and stromal cells: a captured in CRC tissues; b metastasized to metastatic sites via the blood circulation; c captured by vascular endothelial cells. EVs undergo (i) endocytosis or (ii) fusion with the recipient cells and release bioactive substances to activate signaling, ultimately leading to CRC metastasis and drug resistance

Extracellular vesicles in colorectal cancer

EVs are a significant signaling tool in the tumor microenvironment (TME). They induce the TME around CRC cells to be reprogrammed, thereby conferring immunity to CRC cells against eradication and promoting CRC progression (Fig. 3). Fibroblasts can take up the CRC cell-derived exosomes (CEXs) and transform into cancer-associated fibroblasts (CAFs). The activated CAFs can still take up the EVs from CRC cells and undergo lipid metabolic reprogramming, which promotes the secretion of EVs through the CXCL5-CXCR2 axis and forms a positive-feedback loop, thereby aiding in the CRC growth and metastasis [13].

Fig. 3
figure 3

The role of EVs in CRC. Donor cells secrete EVs into TME: a EVs act on immune cells to regulate immunosuppression; b EVs are taken up by recipient cells to promote angiogenesis and EMT; c EVs induce macrophages and fibroblasts transformation to favor CRC growth; d EVs are taken up by CRC cells to promote proliferation and chemoresistance; e EVs are also taken up by donor cells to exert some effect; f the microacidic and hypoxic environment in TME could accelerate the release of EVs from donor cells

The CRC cells are often in a hypoxic and slightly acidic microenvironment, which might be due to increased oxygen consumption, reduced oxygen entry, and difficulty in eliminating metabolic wastes from cancer cells [14]. This host microenvironment helps CRC cells in evading immune surveillance and resisting the cytotoxicity of chemotherapeutic agents. Under these hypoxic and slightly acidic microenvironments, the CRC cells accelerate the release of EVs [15]. Moreover, in order to take nutrients from the host, the CRC tissues form more ducts to transport these “spoils”. Consequently, these abundantly secreted EVs are taken up by endothelial cells, promoting vascular sprouting to meet the growing needs of the CRC tissues. Studies showed that the CRC cell-derived exosomal miR-1229 could promote angiogenesis by targeting HIPK2 and inhibiting its protein expression, thereby activating the VEGF signaling pathway [16]. In addition, a study demonstrated that the horizontal transfer of B cell-derived CD19+ EVs to the chemotherapy-treated cancer cells, followed by ATP hydrolysis to adenosine via the CD39 and CD73 vesicle-binding proteins could inhibit the CD8+ T cell responses and promote immunosuppression [17].

These findings provided a novel insight that the EVs in the TME could carry active molecules, which directly regulated receptor cells; moreover, this effect occurred without direct contact between cells. This might greatly improve the efficiency of communication because a large number of EVs derived from a single cell might be taken up by multiple cells, leading to the activation of multiple pathways and alteration of regulated receptor cells; moreover, this also illustrated the important regulatory role of EVs in the CRC microenvironment. The role of EVs in CRC metastasis and drug resistance has been discussed in detail in the following sections.

Extracellular vesicles regulate metastasis in colorectal cancer

Distant metastasis is a major cause of cancer-related deaths in CRC patients. Approximately, 15–25% of CRC patients have liver metastases at the time of initial diagnosis [18]. This section summarizes the role of EVs in the invasion and metastasis of CRC. Table 1 summarizes some EV cargos related to CRC metastasis. The molecular mechanisms by which different EVs regulate CRC metastasis are shown in Fig. 4.

Table 1 Role of cargoes in EVs in regulating metastasis in CRC
Fig. 4
figure 4

EVs regulate metastasis and drug resistance in CRC. Molecular signaling pathways of metastasis and drug resistance, including PI3K, Akt, phosphatase and tensin homolog (PTEN), STAT3, programmed cell death 4 (PDCD4), Wnt, β-catenin, and suppressor of cytokine signaling 3 (SOCS3), are regulated by EVs. Induction of metastasis and drug resistance promotes CRC progression, and targeting EVs may impair CRC growth

Extracellular vesicles in epithelial-mesenchymal transition

Epithelial-to-mesenchymal transition (EMT) is the process, by which, the cancer cells lose their epithelial cell-like characteristics and gain mesenchymal cell-like characteristics. This process is associated with enhanced CRC cell mobility, invasiveness, and resistance to apoptosis [19]. Recently, Bai's team showed that the CRC cell HCT-116-derived CEXs miR-128-3p could promote EMT by downregulating the FOXO4 expression and activating the transforming growth factor-β (TGF-β)/SMAD and JAK/STAT3 signaling pathways [20]. Furthermore, the telomerase contents and stability might affect the process of hypoxia regulation in EMT. A study showed that the decreased secretion of tumor suppressor exosomal miR-1255b-5p in the serum samples obtained from CRC patients could lead to the increased expression of human telomerase reverse transcriptase (hTERT), which might further enhance EMT and telomerase activity by activating the Wnt/β-catenin signaling pathway [21].

CEXs can also induce the activation of non-malignant cells, thereby accelerating the phenotypic changes in CRC cells. For example, CEXs could transfer miR-106b into macrophages and activate PI3Kγ/AKT/mTOR signaling pathway by directly inhibiting the PDCD4 protein, leading to the M2 polarization of macrophages. In turn, the activated M2-type macrophages mediate the EMT and metastasis of CRC cells through a positive feedback approach [12]. Similarly, the CEXs could also transfer miR-146a-5p and miR-155-5p into CAFs by targeting the suppressor of cytokine signaling 1 (SOCS1) and zinc finger and BTB domain-containing 2 (ZBTB2) and activating the JAK2-STAT3/NF-κB signaling pathway, which promoted the activation of CAFs. The activated CAFs showed a significant increase in the expression levels of IL-6, TNF-α, TGF-β, and CXCL12 while promoting EMT in the CRC cells [22].

The E-cadherin knockdown is a critical step in the initiation of EMT. ZEB can inhibit the expression of E-cadherin by binding to its promoter, while Twist can indirectly inhibit E-cadherin activity, ultimately leading to the occurrence of EMT [23]. ZEB1 was identified as a direct target of miR-150-5p. The CRC patients showed a significant decrease in the serum exosomal miR-150-5p levels and upregulation of ZEB1 expression as compared to the control group; this might lead to poor prognosis of CRC patients [24].

Extracellular vesicles in matrix activation

Multiple cancer signals delivered through EVs might contribute to the formation of an activated stromal and inflammatory microenvironment. The ITGBL (integrin β-like) 1-enriched CEXs can enter the blood circulation and are taken up by resident fibroblasts, inducing the high secretion of pro-inflammatory cytokines, such as IL-6 and IL-8. Further studies revealed that these CEXs could promote the formation of an inflammatory microenvironment by stimulating the TNFAIP3-mediated NF-κB signaling pathway, which ultimately accelerates the metastasis of CRC cells [25]. On the other hand, Hyaluronan and proteoglycan link protein-1 (HAPLN1), an extracellular matrix (ECM) protein, is involved in maintaining a normal colon microenvironment. The CRC patients demonstrated reduced mRNA levels of HAPLN1, which could induce the development of CRC [26]. Microfibril-associated glycoprotein (MAGP)-1 is another key protein in the ECM. However, its expression was also found to be down-regulated in CRC patients [27].

Extracellular vesicles in angiogenesis and vascular permeability

Angiogenesis and altered vascular permeability are the key processes in CRC growth and metastasis, especially in the initial stages of pre-metastatic niches (PMNs) formation. In fact, the serum expression levels of miR-19a-3p, miR-203-3p, miR-221-3p, and let-7f-5p were significantly higher in CRC patients as compared to those in the healthy subjects. Among them, miR-221-3p was transferred by CEXs and regulated the STAT3/VEGFR-2 axis by targeting SOCS3 in endothelial cells to promote the proliferation and migration of endothelial cells and the formation of vascular-like structures [28].

EVs also increase vascular permeability. A study showed that CEXs miR-25-3p could increase angiogenesis and vascular permeability, thereby enhancing the liver and lung metastases of CRC in mice [29]. Specifically, the exosomal miR-25-3p targets KLF2 and KLF4 to regulate the expression levels of endothelial VEGFR2, ZO-1, occludin, and claudin 5, thereby accelerating the CRC progression [29]. Similarly, the EMT-CEXs miR-27b-3p was upregulated in the CRC tissues. Further studies revealed that it could promote the expression of vascular endothelial adhesion protein (VE-Cad) and p120 at the post-transcriptional level by directly binding to the 3'-untranslated region of VE-Cad and p120 to increase the vascular permeability, which accelerated the production and metastasis of circulating CRC cells [30].

As a “double-edged sword”, not all the EVs are involved in the angiogenesis and disruption of vascular permeability. Knocking down the lncRNA-APC1 in exosomes led to the activation of the MAPK pathway in endothelial cells to promote angiogenesis. In contrast, APC inhibited the enrichment of PPARα on the lncRNA-APC1 promoter, leading to the expression of lncRNA-APC1 and inhibition of angiogenesis [31].

Extracellular vesicles in metastatic niche formation

The resident cells at metastatic sites uptake the EVs released from CRC cells, which results in promoting the formation of PMNs and providing suitable soil for CRC metastasis. Liver is a frequent target for the invasion and metastasis of CRC and might be related to its abundant blood flow and high uptake of EVs in the blood. Zhao and his team found that the miR-181a-5p-enriched CEXs could induce the activation of the hepatic stellate cells (HSCs) by targeting SOCS3 while simultaneously activating the IL6/STAT3 signaling pathway [32]. The activated HSCs secrete the chemokine CCL20, thereby promoting the remodeling of TME and the formation of PMNs [32]. Kupffer cells (KCs) are the main members of the defense system. It was suggested that the KCs could engulf the exosomal miR-135a-5p from the blood into the liver. When these CEXs are trapped, they trigger the formation of PMNs in the liver through the large tumor suppressor kinase 2/yes-associated protein/matrix metalloproteinase (MMP) 7 axis [18]. However, studies have also shown that the EVs might be involved in inhibiting EMT in CRC cells, reprogramming KCs, and reducing the expression of MMP9, thereby preventing vascular leakage and formation of PMNs in the liver [33, 34].

Extracellular vesicles in immune evasion

The TME of CRC is infiltrated by various innate or adaptive immune cells, including macrophages, neutrophils, dendritic cells (DCs), T lymphocytes, B lymphocytes, and natural killer (NK) cells [14]. In CRC, lymphangiogenesis is regulated by the VEGFC/VEGFR3 signaling pathway. The VEGFR3 signaling pathway in tumor-associated macrophages (TAMs) is activated by the abundant VEGFC in CRC, resulting in the induction of CRC immune escape and acceleration of tumor growth [35]. A study reported that the M2 macrophage-derived exosome miR-155-5p could promote the proliferation and anti-apoptotic activities of SW48 and HT29 cells; it was also confirmed in xenograft tumor models that exosomal miR-155-5p decreased the expression levels of ZC3H12B and upregulated IL-6 levels, thereby accelerating the induction of immune escape and CRC occurrence [36]. In addition, the M2 macrophage-derived EVs miR-186-5p promoted CRC growth and motility by targeting and inhibiting DLC1 expression [37]. Macrophages can take up EVs secreted by CRC cells and undergo M2 polarization and PD-L1 expression, thereby increasing the abundance of PD-L1 CD206 macrophages in the CRC microenvironment and suppressing T cell activity, ultimately leading to immunosuppression [38].

The cancer cells secrete EVs to transfer immunosuppressive molecules, which mediate the metabolic reprogramming of T cells and drive their functional depletion [39]. Meanwhile, EVs could inhibit the clonal expansion capacity of T cells by mediating adenosine production, inhibiting creatine import, and reducing ATP production, which lead to immunosuppression [40, 41]. This might be one of the reasons for the failure of adoptive CAR-T immunotherapy.

Myeloid-derived suppressor cells (MDSCs) are a heterogeneous group of immature myeloid cells with immunosuppressive activities and are induced by a variety of molecules present in the TME, such as VEGF, IFN-γ, IL-1β, IL-4, IL-6, IL-13, and TNF [15]. It was found that the plasma of CRC patients exhibited higher levels of MDSC-derived exosomal S100A9 and enhanced CRC cells stemness and growth [42].

Extracellular vesicles regulate drug resistance in colorectal cancer

The effective treatment of metastatic cancer often requires the use of drugs. Nevertheless, the development of drug resistance has been a major issue in successful CRC treatment, especially in the late stages of disease progression. This section summarizes the role of EVs in modulating drug resistance in CRC (Fig. 4, Table 2).

Table 2 Role of cargoes in EVs in regulating drug resistance in CRC

Extracellular vesicles in immune resistance

Immunotherapy has become a powerful strategy in cancer treatment. Immune checkpoint inhibitors (ICIs) and adoptive cellular immunotherapy have been clinically used to manipulate the body's immune system and kill cancer cells [43]. However, the robust adaptability of CRC enables them to escape immunotherapy. The analysis of immune profiles and tumor-immune cell interactions in human CRC showed that the miR-424-enriched CEXs were taken up by infiltrating T cells and DCs, leading to the failure of ICI immunotherapy by inhibiting the CD28-CD80/86 co-stimulatory signaling pathway [44]. Ploeg and his team attempted to bypass this EVs-mediated immune resistance to CRC. They developed a novel bispecific antibody CD73xEpCAM, which could bind to the surface marker EpCAM of EVs and inhibit CD73 (GPI-anchored ecto 5'-nucleotidase), thereby effectively enhancing the immune activity [45].

Extracellular vesicles and signaling pathway activation

The Wnt/β-catenin signaling pathway plays an important role in maintaining intestinal homeostasis. However, sustained activation of the Wnt/β-catenin signaling pathway might lead to the accumulation of β-catenin, thereby promoting CRC [46]. Studies have shown a significant increase in the expression of miR-92a-3p in CRC patients. The internalization of CAF-derived exosomal miR-92a-3p by CRC cells could increase the contents of miR-92a-3p in CRC cells, resulting in the activation of the Wnt/β-catenin signaling pathway. Meanwhile, the miR-92a-3p could also inhibit mitochondrial apoptosis by directly targeting FBXW7 and MOAP1. These processes contribute to enhancing the stemness of CRC cells and promote EMT as well as chemoresistance [47]. In addition, long non-coding RNAs are involved in the development of drug resistance. lncRNA CCAL, acting on the mRNA-stabilizing protein HuR (human antigen R), could increase the mRNA and protein levels of β-catenin, which contributed to promoting oxaliplatin resistance in CRC cells [48].

The PI3K/AKT/mTOR signaling pathway is involved in regulating the proliferation, metabolism, autophagy, and protein and lipid synthesis in the cells. The aberrant activation of the PI3K/AKT/mTOR signaling pathway is frequently observed in cancers, including CRC [46]. Recently, a study reported that the CAF-derived exosomal cricN4BP2L2 promoted the resistance of CRC LoVo cells to oxaliplatin by upregulating EIF4A3, leading to the activation of PI3K/AKT/mTOR signaling pathway, while inhibiting the development of apoptosis [49].

Nowadays, the phenomenon of CRC drug resistance is becoming more prominent. The measures, required to overcome the development of drug resistance, should be explored. Finding molecular targets for CRC therapy based on signaling pathways might be an effective way to discover novel drugs due to their close correlations with CRC function, such as ICIs, which have been used in clinical practice. Analysis of the study suggested that the LncRNA HOTTIP downregulated the miR-214 to increase KPNA3 expression, enhancing CRC resistance to mitomycin [50]. The HSP 40 family protein DNAJB8 could upregulate MDR1 to promote CRC resistance to oxaliplatin by inhibiting the ubiquitinated degradation of TP53 [51]. These studies suggested numerous promising molecular targets. Taking EVs or molecular targets as a starting point, it is a very good prospect to overcome drug resistance by interfering with the secretion and uptake of these EVs or blocking their interaction with the target.

Extracellular vesicles in drug resistance transfer

EVs can enter recipient cells by horizontally transferring the specific biologically active substances, which might alter the gene expression and transcriptional regulatory programs of recipient cells as well as promote the expression of drug-resistant active molecules. A recent study demonstrated that the addition of CEXs isolated from resistant LS174T cells to the medium of sensitive LS174T cells increased the expression levels of nuclear factor erythroid 2-related factor 2 (Nrf2) and P-glycoprotein (P-gp), leading to the development of drug resistance in the sensitive LS174T cells [52]. Similarly, Zhang and his team isolated CEXs from 5-Fluorouracil (5-FU) -resistant and sensitive cell lines (RKO), which were then co-cultured with 5-FU-sensitive cells. These CEXs obtained from resistant CRC cell lines could significantly enhance the survival of CRC cells in a 5-FU-containing medium. Proteomics and Western blot analyses further confirmed that GSTP1 and p-STAT3 (Tyr705) were highly enriched in CEXs [53]. Moreover, the adriamycin-resistant breast cancer cells secreted exosomes, which caused drug resistance by directly transferring the resistance-associated protein P-gp to sensitive cells [54]. However, the studies, reporting the direct transfer of drug-resistant proteins from EVs to CRC cells, are limited.

Extracellular vesicles and colorectal cancer microenvironment

The development of CRC resistance is correlated with CRC cells and is supported by their surrounding microenvironment. The activation of CAFs is a hallmark in TME, modulates the basic characteristics of CRC cells, and produces stromal structures, which facilitate CRC growth [55]. A study, investigating the methotrexate (MTX) resistance in CRC cells, identified the differential expression of HEPH, caudal‐related homeobox (CDX) 2, and miR-24-3p in CRC using bioinformatics analysis. The miR-24-3p was carried by CAF-derived exosomes and taken up by CRC cells, resulting in the upregulation of HEPH by targeting CDX2 and accelerating the resistance of CRC cells to MTX [56].

The division between aerobic and hypoxic zones is another characteristic of TME. In general, CRC growth is often disproportionate to the rate of angiogenesis, which leaves CRC cells distant from blood vessels in a state of hypoxia. During the doxorubicin (DOX) treatment in the aerobic/hypoxic TME model, CEXs secreted by aerobic C26 cells developed resistance in the naive hypoxic C26 and RAW 264.7 cells to the cytotoxic effects of DOX. These results were related to the high levels of HIF-1α and the B-cell lymphoma-extra-large anti-apoptotic protein (Bcl-xL), mediating the anti-apoptotic response of recipient cells [57]. Furthermore, EVs can modulate the metabolic reprogramming of CRC cells, creating an acidic microenvironment, which reduces the absorption and efficacy of weakly basic chemotherapeutic drugs, such as anthracyclines, anthraquinones, and vinca alkaloids [58].

In conclusion, the mechanisms of CRC chemoresistance are complex, involving multiple dimensions. EVs can transfer a variety of drug resistance-related active substances to regulate chemoresistance in CRC cells through multiple signaling pathways. How will future studies improve drug sensitivity in drug-resistant CRC? EVs, as an entry point, might be a good choice. Further studies should elucidate the biological functions of EVs and mechanisms mediating drug resistance in CRC. This might facilitate the development of novel target drugs and the transformation of combination drugs. Moreover, EVs might also be a potential next-generation drug delivery system.

Extracellular vesicles: a novel drug delivery platform

Considering the effects of complex TME around CRC, the safe and effective delivery of drugs into CRC cells is one of the biggest challenges at present. In the past few decades, synthetic nanomaterials-based drug delivery systems have made great progress in targeted CRC therapy. However, these nanocarriers have still certain limitations and do not show satisfactory results.

Recently, in terms of drug delivery efficiency and enhanced therapeutic efficacy, natural carriers have shown surprising results. As compared to the traditional nanoparticles (NPs), EVs have great advantages in biocompatibility, evasion of phagocytic clearance, and intrinsic homing ability and might serve as a new generation of drug delivery systems. However, prior to using EVs against CRC, they should be processed first.

The preparation and bioactivity evaluation of EVs currently rely heavily on effective isolation and purification methods. Depending on the size and properties of EVs, various separation methods can be used to separate EVs from biological fluids. The commonly used methods include centrifugation techniques (differential centrifugation, density-gradient centrifugation, sucrose buffered centrifugation), affinity capture, gel-permeation chromatography, membrane filtration, precipitation and microfluidic devices [8]. Centrifugation techniques are commonly used to separate EVs using centrifugal force. They have the advantages of easy operation and high purity of EVs, and are considered the “gold standard” for EVs separation [59]. The affinity capture method uses the immunoaffinity interactions of EV surface proteins to isolate and purify them. This immunoaffinity interaction-based isolation method allows the isolation of different EV subpopulations secreted by the specified cell type [59]. However, centrifugation techniques and affinity capture are unlikely to be used for large-scale applications due to their time-consuming and costly drawbacks [60]. Gel-permeation chromatography and membrane filtration methods are used for the separation and purification of EVs based on differences in their sizes. Membrane filtration is often used as the first step in the separation of EVs and has relatively rapid separation efficiency; however, the filters are prone to clogging. Gel-permeation chromatography can isolate EVs with high purity; moreover, automated collection methods have been developed with potential translational applications. On the other hand, using this method, it is difficult to isolate fractions of sizes similar to EVs, such as lipoproteins [8, 61]. The precipitation technique relies mainly on polymers to precipitate large amounts of EVs; however, the obtained EVs are of low purity and can be used as a means of EV concentration. Microfluidic devices are based on the full integration of EVs size separation, immunoaffinity separation and dynamic separation, which have the advantages of cost-effectiveness, speed and high purity, but they require complex separation devices [59, 60]. Although these separation methods can obtain a certain quality of EVs, unfortunately, there is no method that can completely separate EVs from biofluids. It is only possible to improve the purity of EVs and reduce the proportion of impurities as much as possible. For this reason, based on the heterogeneity of EVs, it is necessary to combine multiple separation methods to isolate the target EVs.

This section summarizes the developments and applications of EVs obtained from different sources as drug delivery systems (Fig. 5).

Fig. 5
figure 5

Novel CRC treatment strategy based on EVs

Native extracellular vesicles

Currently, the EVs obtained from a variety of cells have been used as drug delivery systems in CRC studies. The lipid and protein composition of EVs depends on the parental cells, and the EVs obtained from different sources might have different activities in terms of homing properties and cancer progression. Therefore, it is necessary to carefully explore the biological properties of each EV and evaluate their pros and cons as drug carriers.

Colorectal cancer cell-derived extracellular vesicles

The components of CEXs are similar to those in the CRC cell membranes. Therefore, using CEX as a "porter" for transporting therapeutic drugs might improve their CRC specificity. Recently, Phuong and his team investigated that the exosomes derived from breast and CRC cells enhanced the uptake of nano-amorphous aspirin through clathrin-dependent and independent endocytosis pathways and efficiently delivered aspirin to the tumor sites in vivo [62]. Van et al. isolated EVs from CT26 colon cancer cells and 4T1 murine breast cancer cells and loaded them with DOX by electroporation. The results showed excellent biocompatibility and the ability to target CRC cells [63]. Interestingly, the CT26-derived EVs were preferentially taken up by parental cells and showed robust penetrating properties as compared to 4T1 cell-derived EVs [63]. This just hints at the targeting potential of EVs as drug delivery systems.

Due to the easy RNA degradation, fast clearance, and immune response induction, EVs might also be used in CRC gene therapy [64]. Maryam Hosseini et al. isolated CEXs from CT26 and loaded them with miR-34a mimics. The CEX-miR-34a reduced the invasion, angiogenesis, and immune evasion-related gene expressions in CRC and prolonged the survival of mice with colon cancer. Notably, the mice, receiving CEXs alone, exhibited certain oncogenic gene expressions; however, the IL-17A secretion levels in draining lymph nodes significantly decreased [65]. This suggested that the direct application of CEXs in CRC therapy might face great challenges because the EVs secreted by CRC cells often act as "boosters" and are involved in cell proliferation, induction of angiogenesis, enhancement of invasion and metastasis, immune evasion, and metabolic reprogramming processes [66]. Therefore, ensuring the safety of CEXs for CRC treatment is a current issue, which should be addressed.

Bacterial outer membrane vesicles

The bacterial outer membrane vesicles (OMVs) might be a particular choice in immunotherapy. Although the immunogenicity of OMVs might limit their role in cancer therapy as natural drug delivery systems, their small size, immunomodulatory properties, high bacterial production, and enrichment in lymph nodes might lead to their use as vaccine carriers with applications in immunotherapy [67]. Keman and his team used genetic engineering and Plug-and-Display technology to highlight the ClyA fusion protein on the surface of OMVs with different protein capture agent modifications. The results suggested that the OMVs loaded with different tumor antigens inhibited melanoma metastasis and CRC subcutaneous graft growth and might be used as an alternative strategy for cancer immunotherapy [68]. As mentioned earlier, OMVs have the characteristics of parental bacteria and might have a great potential for development. However, further studies on OMVs, obtained from different bacterial sources, are still needed to ensure their safety while being used as vectors.

Extracellular vesicles in plants and milk

As compared to synthetic NPs, plant EVs (PEVs) have low toxicity, low immunogenicity, and better cellular uptake and gastrointestinal stability [69]. In a study, a novel oral delivery system based on grapefruit-derived EVs loaded with MTX could target intestinal macrophages, significantly reduce the toxicity of MTX, and improve the therapeutic effects on the mouse colitis models [70].

The milk-derived EVs (MEVs) have similar advantages. The bilayer lipid membrane of MEVs enables them to easily cross the blood–brain barrier and cell membranes, thereby ensuring the target-specific delivery of valuable cargo [71]. A study showed that the orally administered MEVs were stable in the gastrointestinal tract and reduced primary tumor burden in mouse models of CRC and breast cancer. Interestingly, MEVs could aggravate tumor metastasis in the mouse models of breast and pancreatic cancer, while oral administration of MEVs after primary tumor resection could attenuate the metastatic effects of breast cancer cells. Further proteomic and biochemical analyses revealed that MEVs could exert their effects by inducing the cancer cells' senescence and EMT [72].

It can be found that despite the differences in their origins, all the EVs discussed in this study have achieved promising results, highlighting their potential in developing advanced drug delivery systems for CRC treatment. However, the development of native EVs still faces enormous challenges due to the difficulties in large-scale production, isolation and purification, drug-loading efficiency, storage, and uncontrollable heterogeneity among the EVs subpopulations of native EVs [73, 74]. These challenges might greatly hinder the production and clinical applications of EV-related products.

Artificial extracellular vesicles

Increasing studies have aimed to develop artificial exosomes, such as engineered and biomimetic vesicles through nanobiotechnology. As compared to natural exosomes, these artificial exosomes have higher pharmaceutical acceptability due to solving some of the shortcomings of natural exosomes and easier standardized production rules.

Engineered extracellular vesicles

The appropriate surface modification of natural EVs gives them stronger cell-targeting abilities. Genetic engineering and chemical modification are the two commonly used modification strategies. A study investigated the expression levels of miR-1915-3p in a non-tumorigenic intestinal cell line (FHC) by lentiviral transduction. The FHC-derived EVs enhanced the sensitivity of drug-resistant CRC cell lines to oxaliplatin by downregulating the pro-oncogenes PFKFB3 and USP2 [75]. Moreover, the combined Her2-mcherry plasmid was transfected into HEK293T cells. The engineered HEK293T cells secreted exosomes, expressing the Her2-LAMP2 fusion protein. The 5-FU and miR-21i were encapsulated by electroporation, forming a co-delivery system. These engineered exosomes effectively reversed drug resistance and significantly enhanced the cytotoxicity of 5-FU-resistant colon cancer cells [76].

In terms of chemical modifications, Elnaz's team covalently attached the carboxylic acid-modified MUC1 aptamer (5TR1) to amine groups on the surface of mesenchymal stem cell-derived exosomes via EDC/NHS chemistry. The modified exosomes showed a higher tumor accumulation and significantly inhibited CRC growth in the CT26 colon adenocarcinoma model [77]. EVs were modified using polyethylene glycols (PEG) to obtain PEGylated EVs, which exhibited a longer life cycle [78]. Alternatively, magnetic NPs (MNPs) and folic acid (FA) have also been prepared into exosome-based hybrid nanostructures (EHNs). EHNs are composed of CEXs, EpCAM-MNPs, and FA and are used to load the anticancer drug DOX. This EHN enhanced the tissue permeability while applying alternating magnetic fields enhanced their CRC cell-killing effects and DOX cytotoxicity [79].

Although the surface modifications of EVs have been successfully performed, the current modification strategies have certain limitations. The prerequisite for genetic engineering techniques requires specific gene fragments for plasmid construction. Due to the huge heterogeneities of EVs, the expression of target protein in all EVs as well as the amount of expression require further verifications. In chemical modifications, ensuring the structural integrity of EVs should be considered. In addition, improving the specificity of site-specific modifications on the surface of EVs without affecting the normal functioning of other EV sites is also a major challenge.

Bionic extracellular vesicles

A large proportion of both the natural and engineered EVs are based on natural secretion from donor cells. Large-scale production is difficult to achieve due to their lower yields and higher separation costs. The recently developed artificially synthesized EVs (ASEVs), nanovesicles (NVs), and hybrid vesicles (HVs) can be commercially produced and used for the efficient delivery of drugs to CRC cells.

ASEVs are NPs synthesized based on individual small molecules by linking surface proteins or chemical moieties. The composition of these ASEVs is similar to but clearer than that of natural EVs [80]. Moreover, these ASEVs, along with overcoming the challenges of natural EVs heterogeneity, also overcome the issue of large-scale production. Zhang and his team developed PEGylated liposomes (CTB-sLip) coupled with cholera toxin subunit b protein. These CTB-sLips showed excellent affinity and cellular uptake in both human-derived CRC cell lines, including HCT-116 and HT-29 cells [81]. In another study, immunoliposomes, coated with antibodies to Frizzled 10 protein, were synthesized and loaded with 5-FU. The follow-up results showed that this immunoliposome exhibited good selectivity against CRC as well as improved cytotoxicity of 5-FU [82]. In addition, a bionic magnetic liposome exhibited similar anti-CRC advantages in vitro and in vivo [83]. Despite the great advancements in ASEVs, it is still difficult to fully simulate the natural EVs due to their complex compositions.

NVs are tiny vesicles generated from the donor cells using filters of different pore sizes, microfluidic devices, nitrogen cavitation techniques, and ultrasound or cellular vesicle methods [84]. The components of these NVs are similar to those of the donor cell membrane as well as have similar size, morphology, and protein markers to natural EVs. Notably, the production of NVs increased greatly as compared to natural EVs. Fang et al. prepared “artificially assembled macrophages” by the extrusion of macrophage-derived cell membranes. These NVs showed good CRC targeting with the help of macrophage membranes. They also reversed the immunosuppressive nature of the CRC microenvironment by inhibiting Tregs and promoting the maturation of antigen-presenting cells and achieved a CRC suppression rate of 95.3 ± 2.05% in vivo, due to the combination of photo/chemo/immunotherapy [85]. However, it is noteworthy that the filters used to prepare NVs are prone to clogging.

HVs are usually formed by fusing EVs with other types of membrane structures. A study designed a hybrid therapeutic nanovacuole (hGLV), which was obtained by isolating CD47+ gene-engineered exosomes from lentiviral vector-transfected CT26 cells and combining them with drug-loaded thermosensitive liposomes. Follow-up studies showed that CD47+ hGLV improved the macrophage-mediated phagocytosis of CRC cells and prolonged circulation time by blocking the CD47 signaling. In addition, hGLV exhibited excellent targeting properties and showed excellent phototherapeutic effects on CRC after laser irradiation. It could also promote the maturation of immature DCs with the help of a co-encapsulated immune adjuvant, thereby triggering a stronger immune response [86].

In addition to the above-mentioned applications, EVs have also been used as "invisible coats" to wrap the existing synthetic NPs. A study was conducted to synthesize a photoactivatable polymer TKPEI-Ce6 using the cross-linked branched polyethyleneimine, reactive oxygen species-sensitive thiacetal, and photosensitizer chlorin e6. Further, a nanomimetic photoactivated silencing EVs (PASEVs) were successfully developed using M1 macrophage-derived EVs wrapped with TKPEI-Ce6 and a small interfering RNA targeting PAK4. PASEVs achieved effective accumulation at CRC sites, and after using laser irradiation, PASEVs promoted immune activation in the CRC microenvironment and exerted a powerful role in killing CRC [87].

In conclusion, biomimetic NPs are highly biocompatible, have limited immunogenicity, and are capable of being produced on a large scale, thereby making them a promising drug delivery vehicle in the future. However, the molecular mechanisms of EVs biogenesis have not been fully understood yet, and there is a lack of unified technical standards. Therefore, further studies are still needed to elucidate the mechanisms of EVs biogenesis, cargo packaging, cell penetration, and regulatory evasion to further amplify the potential of EV-related drug carriers.

Biomarkers

Extracellular vesicles and biomarkers

Recently, the use of EVs as biomarkers for the prediction, diagnosis, and presumed prognosis of CRC has shown promising outcomes. EVs can be used as biomarkers for the development of CRC and are summarized in Table 3. A study using real-time quantitative polymerase chain reaction (RT-qPCR) showed a significant decrease in the plasma EV-miR-193a-5p levels in CRC patients. The EV-miR-193a-5p showed an area under the receiver operating characteristic (ROC) curve (AUC) of 0.740 in differentiating CRC from precancerous colorectal adenomas and 0.759 in differentiating CRC from non-cancerous controls. Further analysis showed that miR-193a-5p could target the tumor-associated genes, including CUT-like homeobox 1 and intersectin 1, and inhibit CRC progression. These findings suggested that miR-193a-5p might help in predicting the occurrence and progression of CRC [88].

Table 3 EVs as biomarkers in CRC

EVs might also provide novel perspectives for the early clinical diagnosis and disease pathogenesis of CRC. The diagnostic performance of exosomal CircLPAR1 in plasma was analyzed by combining the common clinical biomarkers, including CEA and CA19-9. The results demonstrated that the exosomal CircLPAR1 showed improved diagnostic performance (AUC of 0.875) while diagnosing CRC and was correlated with the overall survival of patients [89]. Moreover, CircLPAR1 encapsulated in exosomes exhibited high stability and detectability.

In addition, EVs might also predict the prognosis of CRC patients. A study suggested the superior performance of the serum EVs-associated miRNA-21 and miRNA-92a in diagnosing patients with metastatic CRC as compared to that of carcinoembryonic antigen. Further multivariate Cox analysis confirmed that the higher levels of miRNA-222 were correlated with lower overall survival of patients [90]. These data all implied that EVs might have a strong potential as a novel non-invasive biomarker for the prediction, diagnosis, and presumed prognosis of CRC.

Detection of biomarkers

Despite the great potential of EVs for liquid biopsy-based prediction, diagnosis, and presumed prognosis of CRC, the detection and characterization of such small molecules with high sensitivity are the major challenges since they are highly heterogeneous and microscopic in nature. Several techniques have been developed to determine the physical properties and biological characteristics of EVs, including transmission electron microscopy, nanoparticle-tracking analysis, Western blotting, ELISA, and high-sensitivity flow cytometry. Transmission electron microscopy is typically used to observe the size and surface characteristics of EVs. Nanoparticle-tracking analysis can be used to determine the size distribution and concentration of EVs; however, this method cannot detect specific subpopulations of EVs [60]. Western blotting is a quantitative or semi-quantitative method for monitoring the levels of characteristic proteins targeted by EVs with small sample sizes [91]. ELISA is used to quantify specific EV-binding proteins based on colorimetric or fluorescent methods, but it requires the separation of EVs from soluble fractions [61]. High-sensitivity flow cytometry classifies the subpopulations of EVs and determines the origin of EVs by simultaneously detecting their light scattering and fluorescence signals. However, efforts are still needed to improve its sensitivity [92]. Among these, ELISA and high-sensitivity flow cytometry are more likely to be used for the detection of EVs as CRC biomarkers in the routine clinical setting due to their reproducibility, low cost, and the advantage of distinguishing EV subgroups.

In addition to the above commonly used assays, studies are continuously being conducted on the development of more accurate, faster, and cost-effective technologies for the detection of biomarkers associated with EVs in CRC. For example, Wei and his team performed immunoassays on circulating EVs (CD9-CD63 and EpCAM-CD63) by developing a single-molecule array technique. The results demonstrated that the performances of the single-molecule array technique gave and Western blotting for EVs expressing EpCAM were similar, and the single-molecule array technique could distinguish between cancerous and non-cancerous plasma samples [93]. Francisco and his team presented a microfluidic amperometric immunosensor assay for detecting the biomarker claudin7 (CLD7). The results showed that the detection range of the detector was 2–1000 pg mL−1, and the detection limit of CLD7 could reach 0.1 pg mL−1 under optimal conditions [94].

However, it is worth noting that all these methods require the separation and purification of EVs. Moreover, the possibility of multiplex analysis of EVs is also not known yet. This undoubtedly increases the difficulty and complexity of sample processing. To address this issue, Zhang et al. used surface-enhanced Raman spectroscopy nanotags to simultaneously analyze multiple protein biomarkers expressed on the CEXs. As compared to most of the other reported EV detection techniques, such as Western blotting, ELISA, and flow cytometry, the surface-enhanced Raman spectroscopy nanotag analysis eliminated the need for complex separation steps and enabled the possibility of higher multiplex analysis with higher sensitivity. This might greatly simplify the sample preparation process and increase the efficiency of the assay, allowing for instant determination [95].

Conclusion and prospects

The current studies have demonstrated that EVs are involved in bidirectional crosstalk between CRC cells and recipient cells as well as in compiling the dynamic network of TME, conferring the CRC cells to proliferate, metastasize, and develop drug resistance. Based on abundant evidence, this review described the biological functions of EVs in promoting CRC cell dissemination and developing resistance to therapy by regulating the CRC microenvironment. This intrinsic property might provide a new starting point for the prediction, diagnosis, and treatment of CRC. EVs have been proposed as predictive and diagnostic biomarkers due to their easy isolation from the patient’s body fluids and their potential to highlight CRC. For therapy, investigating the pathways of EVs might potentially lead to identifying novel therapeutic targets. Considering their natural origin, EVs can improve biocompatibility and target penetration. This review also further discussed the therapeutic potential of EVs as delivery systems in CRC.

However, in practical work, there are certain limitations in the separation and purification of EVs, making their large-scale production difficult. EVs have huge heterogeneity, which might result in the uncontrollability of their clinical applications. The application of bionic technology might have good prospects for the development of EVs; however, a complete set of standardized rules to guide the production and evaluation of bionic EVs should be formed. Additionally, the available technologies for EVs detection are not yet mature, which might not be conducive to their clinical implementation. Therefore, the biogenesis process of EVs and mechanisms of their participation in cargo sorting to regulate CRC cells should be further explored. This might help in identifying a specific way to solve these defects. In conclusion, EVs are highly potential delivery media in TME; however, further research is needed to further explore them.

Availability of data and materials

Not applicable.

Abbreviations

CRC:

Colorectal cancer

EVs:

Extracellular vesicles

HSP:

Heat shock proteins

ICAM:

Intercellular cell adhesion molecule

MVBs:

Multivesicular bodies

ESCRT:

Endosomal sorting complex required for transport

MHC:

Major histocompatibility complex

ILVs:

Intraluminal vesicles

TME:

Tumor microenvironment

CEXs:

Colorectal cancer cell-derived exosomes

CAFs:

Cancer-associated fibroblasts

PTEN:

Phosphatase and tensin homolog

SOCS3:

Suppressor of cytokine signaling 3

EMT:

Epithelial-to-mesenchymal transition

TGF-β:

Transforming growth factor-β

hTERT:

Human telomerase reverse transcriptase

PDCD4:

Programmed cell death 4

SOCS1:

Suppressor of cytokine signaling 1

ZBTB2:

Zinc finger and BTB domain-containing 2

ITGBL1:

Integrin β-like 1

HAPLN1:

Hyaluronan and proteoglycan link protein -1

ECM:

Extracellular matrix

MAGP:

Microfibril-associated glycoprotein

PMNs:

Pre-metastatic niches

VE-Cad:

Vascular endothelial adhesion protein

HSCs:

Hepatic stellate cells

KCs:

Kupffer cells

MMP:

Matrix metalloproteinase

DCs:

Dendritic cells

NK cells:

Natural killer cells

TAMs:

Tumor-associated macrophages

MDSCs:

Myeloid-derived suppressor cells

ICIs:

Immune checkpoint inhibitors

HuR:

Human antigen R

Nrf2:

Nuclear factor erythroid 2-related factor 2

P-gp:

P-glycoprotein

5-FU:

5-Fluorouracil

MTX:

Methotrexate

CDX2:

Caudal‐related homeobox 2

DOX:

Doxorubicin

Bcl-xL:

B-cell lymphoma-extra-large anti-apoptotic protein

NPs:

Nanoparticles

OMVs:

Outer membrane vesicles

PEVs:

Plant EVs

MEVs:

Milk-derived EVs

PEG:

Polyethylene glycols

MNPs:

Magnetic NPs

FA:

Folic acid

EHNs:

Exosome-based hybrid nanostructures

ASEVs:

Artificially synthesized EVs

NVs:

Nanovesicles

HVs:

Hybrid vesicles

PASEVs:

Photoactivated silencing EVs

RT-qPCR:

Real-time quantitative polymerase chain reaction

ROC:

Receiver operating characteristic curve

CLD7:

Claudin7

References

  1. 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 Cancer J Clin. 2021;71(3):209–49.

    Article  PubMed  Google Scholar 

  2. Dekker E, Tanis PJ, Vleugels JL, Kasi PM, Wallace MB. Colorectal cancer. Lancet. 2019;394:1467–80.

    Article  PubMed  Google Scholar 

  3. Théry C, Witwer KW, Aikawa E, Alcaraz MJ, Anderson JD, Andriantsitohaina R, et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles. 2018;7(1):1535750.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Claridge B, Lozano J, Poh QH, Greening DW. Development of extracellular vesicle therapeutics: challenges, considerations, and opportunities. Front Cell Dev Biol. 2021;9:734720.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Jeppesen DK, Fenix AM, Franklin JL, Higginbotham JN, Zhang Q, Zimmerman LJ, et al. Reassessment of exosome composition. Cell. 2019;177(2):428-445.e18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Fridman ES, Ginini L, Gil Z. The role of extracellular vesicles in metabolic reprogramming of the tumor microenvironment. Cells. 2022;11(9):1433.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Anand S, Samuel M, Kumar S, Mathivanan S. Ticket to a bubble ride: Cargo sorting into exosomes and extracellular vesicles. Biochim Biophys Acta Proteins Proteom. 2019;1867(12):140203.

    Article  CAS  PubMed  Google Scholar 

  8. Zhu L, Sun HT, Wang S, Huang SL, Zheng Y, Wang CQ, et al. Isolation and characterization of exosomes for cancer research. J Hematol Oncol. 2020;13(1):152.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Dai J, Su Y, Zhong S, Cong L, Liu B, Yang J, et al. Exosomes: key players in cancer and potential therapeutic strategy. Signal Transduct Target Ther. 2020;5(1):145.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Skotland T, Hessvik NP, Sandvig K, Llorente A. Exosomal lipid composition and the role of ether lipids and phosphoinositides in exosome biology. J Lipid Res. 2019;60(1):9–18.

    Article  CAS  PubMed  Google Scholar 

  11. Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science. 2020;367(6478):eaau6977.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. 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. Mol Ther. 2021;29(6):2088–107.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. 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 Dis. 2022;13(1):57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Anderson NM, Simon MC. The tumor microenvironment. Curr Biol. 2020;30(16):R921–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Tian X, Shen H, Li Z, Wang T, Wang S. Tumor-derived exosomes, myeloid-derived suppressor cells, and tumor microenvironment. J Hematol Oncol. 2019;12(1):84.

    Article  PubMed  PubMed Central  Google Scholar 

  16. 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. Int J Biol Macromol. 2019;132:470–7.

    Article  CAS  PubMed  Google Scholar 

  17. Zhang F, Li R, Yang Y, Shi C, Shen Y, Lu C, et al. Specific decrease in B-cell-derived extracellular vesicles enhances post-chemotherapeutic CD8+ T cell responses. Immunity. 2019;50(3):738-750.e7.

    Article  CAS  PubMed  Google Scholar 

  18. 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(5):2633–51.

    Article  CAS  PubMed  Google Scholar 

  19. Lu W, Kang Y. Epithelial-mesenchymal plasticity in cancer progression and metastasis. Dev Cell. 2019;49(3):361–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Bai J, Zhang X, Shi D, Xiang Z, Wang S, Yang C, et al. Exosomal miR-128-3p promotes epithelial-to-mesenchymal transition in colorectal cancer cells by targeting FOXO4 via TGF-β/SMAD and JAK/STAT3 signaling. Front Cell Dev Biol. 2021;9:568738.

    Article  PubMed  PubMed Central  Google Scholar 

  21. 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. Mol Oncol. 2020;14(10):2589–608.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. 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 Dis. 2022;13(4):380.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Gonzalez DM, Medici D. Signaling mechanisms of the epithelial-mesenchymal transition. Sci Signal. 2014;7(344):re8.

    Article  PubMed  PubMed Central  Google Scholar 

  24. 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 Biomark. 2019;26(1):69–77.

    Article  CAS  PubMed  Google Scholar 

  25. Ji Q, Zhou L, Sui H, Yang L, Wu X, Song Q, et al. Primary tumors release ITGBL1-rich extracellular vesicles to promote distal metastatic tumor growth through fibroblast-niche formation. Nat Commun. 2020;11(1):1211.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Wang Y, Xu X, Marshall JE, Gong M, Zhao Y, Dua K, et al. Loss of hyaluronan and proteoglycan link protein-1 induces tumorigenesis in colorectal cancer. Front Oncol. 2021;11:754240.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Gómez de Segura I, Ahechu P, Gómez-Ambrosi J, Rodríguez A, Ramírez B, Becerril S, et al. Decreased levels of microfibril-associated glycoprotein (MAGP)-1 in patients with colon cancer and obesity are associated with changes in extracellular matrix remodelling. Int J Mol Sci. 2021;22(16):8485.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Dokhanchi M, Pakravan K, Zareian S, Hussen BM, Farid M, Razmara E, et al. Colorectal cancer cell-derived extracellular vesicles transfer miR-221-3p to promote endothelial cell angiogenesis via targeting suppressor of cytokine signaling 3. Life Sci. 2021;285:119937.

    Article  CAS  PubMed  Google Scholar 

  29. 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. Nat Commun. 2018;9(1):5395.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. 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. Clin Transl Med. 2021;11(12):e595.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Wang FW, Cao CH, Han K, Zhao YX, Cai MY, Xiang ZC, et al. APC-activated long noncoding RNA inhibits colorectal carcinoma pathogenesis through reduction of exosome production. J Clin Invest. 2019;129(2):727–43.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Zhao S, Mi Y, Zheng B, Wei P, Gu Y, Zhang Z, et al. Highly-metastatic colorectal cancer cell released miR-181a-5p-rich extracellular vesicles promote liver metastasis by activating hepatic stellate cells and remodelling the tumour microenvironment. J Extracell Vesicles. 2022;11(1):e12186.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. 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(1):718.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. 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. J Exp Clin Cancer Res. 2021;40(1):21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Tacconi C, Ungaro F, Correale C, Arena V, Massimino L, Detmar M, et al. Activation of the VEGFC/VEGFR3 pathway induces tumor immune escape in colorectal cancer. Cancer Res. 2019;79(16):4196–210.

    Article  CAS  PubMed  Google Scholar 

  36. Ma YS, Wu TM, Ling CC, Yu F, Zhang J, Cao PS, et al. M2 macrophage-derived exosomal microRNA-155-5p promotes the immune escape of colon cancer by downregulating ZC3H12B. Mol Ther Oncolytics. 2021;20:484–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Guo J, Wang X, Guo Q, Zhu S, Li P, Zhang S, et al. M2 Macrophage derived extracellular vesicle-mediated transfer of MiR-186-5p promotes colon cancer progression by targeting DLC1. Int J Biol Sci. 2022;18(4):1663–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Yin Y, Liu B, Cao Y, Yao S, Liu Y, Jin G, et al. Colorectal cancer-derived small extracellular vesicles promote tumor immune evasion by upregulating PD-L1 expression in tumor-associated macrophages. Adv Sci (Weinh). 2022;9(9):2102620.

    Article  CAS  PubMed  Google Scholar 

  39. Ma F, Vayalil J, Lee G, Wang Y, Peng G. Emerging role of tumor-derived extracellular vesicles in T cell suppression and dysfunction in the tumor microenvironment. J Immunother Cancer. 2021;9(10):e003217.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Peng M, Ren J, Jing Y, Jiang X, Xiao Q, Huang J, et al. Tumour-derived small extracellular vesicles suppress CD8+ T cell immune function by inhibiting SLC6A8-mediated creatine import in NPM1-mutated acute myeloid leukaemia. J Extracell Vesicles. 2021;10(13):e12168.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Schneider E, Winzer R, Rissiek A, Ricklefs I, Meyer-Schwesinger C, Ricklefs FL, et al. CD73-mediated adenosine production by CD8 T cell-derived extracellular vesicles constitutes an intrinsic mechanism of immune suppression. Nat Commun. 2021;12(1):5911.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. 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. Adv Sci (Weinh). 2019;6(18):1901278.

    Article  CAS  PubMed  Google Scholar 

  43. Kennedy LB, Salama AKS. A review of cancer immunotherapy toxicity. CA Cancer J Clin. 2020;70(2):86–104.

    Article  PubMed  Google Scholar 

  44. Zhao X, Yuan C, Wangmo D, Subramanian S. Tumor-secreted extracellular vesicles regulate T-cell costimulation and can be manipulated to induce tumor-specific T-cell responses. Gastroenterology. 2021;161(2):560-574.e11.

    Article  CAS  PubMed  Google Scholar 

  45. Ploeg EM, Ke X, Britsch I, Hendriks MAJM, Van der Zant FA, Kruijff S, et al. Bispecific antibody CD73xEpCAM selectively inhibits the adenosine-mediated immunosuppressive activity of carcinoma-derived extracellular vesicles. Cancer Lett. 2021;521:109–18.

    Article  CAS  PubMed  Google Scholar 

  46. Prossomariti A, Piazzi G, Alquati C, Ricciardiello L. Are Wnt/β-Catenin and PI3K/AKT/mTORC1 distinct pathways in colorectal cancer? Cell Mol Gastroenterol Hepatol. 2020;10(3):491–506.

    Article  PubMed  PubMed Central  Google Scholar 

  47. 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. Mol Cancer. 2019;18(1):91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. 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. Int J Cancer. 2020;146(6):1700–16.

    Article  CAS  PubMed  Google Scholar 

  49. Qu Z, Yang KD, Luo BH, Zhang F. CAFs-secreted exosomal cricN4BP2L2 promoted colorectal cancer stemness and chemoresistance by interacting with EIF4A3. Exp Cell Res. 2022;418(2):113266.

    Article  CAS  PubMed  Google Scholar 

  50. 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. Front Cell Dev Biol. 2021;8:582723.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Wang Z, Li Y, Mao R, Zhang Y, Wen J, Liu Q, et al. DNAJB8 in small extracellular vesicles promotes Oxaliplatin resistance through TP53/MDR1 pathway in colon cancer. Cell Death Dis. 2022;13(2):151.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Mostafazadeh M, Kahroba H, Haiaty S, TazeKand AP, Samadi N, Rahbarghazi R, et al. In vitro exosomal transfer of Nrf2 led to the oxaliplatin resistance in human colorectal cancer LS174T cells. Cell Biochem Funct. 2022;40(4):391–402.

    Article  CAS  PubMed  Google Scholar 

  53. 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. J Exp Clin Cancer Res. 2019;38(1):320.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Ning K, Wang T, Sun X, Zhang P, Chen Y, Jin J, et al. UCH-L1-containing exosomes mediate chemotherapeutic resistance transfer in breast cancer. J Surg Oncol. 2017;115(8):932–40.

    Article  CAS  PubMed  Google Scholar 

  55. Chen Y, McAndrews KM, Kalluri R. Clinical and therapeutic relevance of cancer-associated fibroblasts. Nat Rev Clin Oncol. 2021;18(12):792–804.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Zhang HW, Shi Y, Liu JB, Wang HM, Wang PY, Wu ZJ, et al. Cancer-associated fibroblast-derived exosomal microRNA-24-3p enhances colon cancer cell resistance to MTX by down-regulating CDX2/HEPH axis. J Cell Mol Med. 2021;25(8):3699–713.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Patras L, Fens MHAM, Vader P, Barendrecht A, Sesarman A, Banciu M, et al. Normoxic tumour extracellular vesicles modulate the response of hypoxic cancer and stromal cells to doxorubicin in vitro. Int J Mol Sci. 2020;21(17):5951.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Wojtkowiak JW, Verduzco D, Schramm KJ, Gillies RJ. Drug resistance and cellular adaptation to tumor acidic pH microenvironment. Mol Pharm. 2011;8(6):2032–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Yang D, Zhang W, Zhang H, Zhang F, Chen L, Ma L, et al. Progress, opportunity, and perspective on exosome isolation - efforts for efficient exosome-based theranostics. Theranostics. 2020;10(8):3684–707.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Gebeyehu A, Kommineni N, Meckes DG Jr, Sachdeva MS. Role of exosomes for delivery of chemotherapeutic drugs. Crit Rev Ther Drug Carrier Syst. 2021;38(5):53–97.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Thietart S, Rautou PE. Extracellular vesicles as biomarkers in liver diseases: a clinician’s point of view. J Hepatol. 2020;73(6):1507–25.

    Article  CAS  PubMed  Google Scholar 

  62. Tran PHL, Wang T, Yin W, Tran TTD, Nguyen TNG, Lee BJ, et al. Aspirin-loaded nanoexosomes as cancer therapeutics. Int J Pharm. 2019;572:118786.

    Article  CAS  PubMed  Google Scholar 

  63. Nguyen VD, Kim HY, Choi YH, Park JO, Choi E. Tumor-derived extracellular vesicles for the active targeting and effective treatment of colorectal tumors in vivo. Drug Deliv. 2022;29(1):2621–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Burnett JC, Rossi JJ. RNA-based therapeutics: current progress and future prospects. Chem Biol. 2012;19(1):60–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. 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 Sci. 2022;290:120234.

    Article  CAS  PubMed  Google Scholar 

  66. Meng W, Hao Y, He C, Li L, Zhu G. Exosome-orchestrated hypoxic tumor microenvironment. Mol Cancer. 2019;18(1):57.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Sartorio MG, Pardue EJ, Feldman MF, Haurat MF. Bacterial outer membrane vesicles: from discovery to applications. Annu Rev Microbiol. 2021;75:609–30.

    Article  PubMed  PubMed Central  Google Scholar 

  68. Cheng K, Zhao R, Li Y, Qi Y, Wang Y, Zhang Y, et al. Bioengineered bacteria-derived outer membrane vesicles as a versatile antigen display platform for tumor vaccination via Plug-and-Display technology. Nat Commun. 2021;12(1):2041.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Fujita D, Arai T, Komori H, Shirasaki Y, Wakayama T, Nakanishi T, et al. Apple-derived nanoparticles modulate expression of organic-anion-transporting polypeptide (OATP) 2B1 in Caco-2 cells. Mol Pharm. 2018;15(12):5772–80.

    Article  CAS  PubMed  Google Scholar 

  70. Wang B, Zhuang X, Deng ZB, Jiang H, Mu J, Wang Q, et al. Targeted drug delivery to intestinal macrophages by bioactive nanovesicles released from grapefruit. Mol Ther. 2014;22(3):522–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Babaker MA, Aljoud FA, Alkhilaiwi F, Algarni A, Ahmed A, Khan MI, et al. The therapeutic potential of milk extracellular vesicles on colorectal cancer. Int J Mol Sci. 2022;23(12):6812.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Samuel M, Fonseka P, Sanwlani R, Gangoda L, Chee SH, Keerthikumar S, et al. Oral administration of bovine milk-derived extracellular vesicles induces senescence in the primary tumor but accelerates cancer metastasis. Nat Commun. 2021;12(1):3950.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Meng W, He C, Hao Y, Wang L, Li L, Zhu G. Prospects and challenges of extracellular vesicle-based drug delivery system: considering cell source. Drug Deliv. 2020;27(1):585–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Cheng Y, Zeng Q, Han Q, Xia W. Effect of pH, temperature and freezing-thawing on quantity changes and cellular uptake of exosomes. Protein Cell. 2019;10(4):295–9.

    Article  CAS  PubMed  Google Scholar 

  75. Xiao Z, Liu Y, Li Q, Liu Q, Liu Y, Luo Y, et al. EVs delivery of miR-1915-3p improves the chemotherapeutic efficacy of oxaliplatin in colorectal cancer. Cancer Chemother Pharmacol. 2021;88(6):1021–31.

    Article  CAS  PubMed  Google Scholar 

  76. 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. J Nanobiotechnology. 2020;18(1):10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Bagheri E, Abnous K, Farzad SA, Taghdisi SM, Ramezani M, Alibolandi M. Targeted doxorubicin-loaded mesenchymal stem cells-derived exosomes as a versatile platform for fighting against colorectal cancer. Life Sci. 2020;261:118369.

    Article  CAS  PubMed  Google Scholar 

  78. Emam SE, Abu Lila AS, Elsadek NE, Ando H, Shimizu T, Okuhira K, et al. Cancer cell-type tropism is one of crucial determinants for the efficient systemic delivery of cancer cell-derived exosomes to tumor tissues. Eur J Pharm Biopharm. 2019;145:27–34.

    Article  CAS  PubMed  Google Scholar 

  79. Kwon SH, Faruque HA, Kee H, Kim E, Park S. Exosome-based hybrid nanostructures for enhanced tumor targeting and hyperthermia therapy. Colloids Surf B Biointerfaces. 2021;205:111915.

    Article  CAS  PubMed  Google Scholar 

  80. Molinaro R, Martinez JO, Zinger A, De Vita A, Storci G, Arrighetti N, et al. Leukocyte-mimicking nanovesicles for effective doxorubicin delivery to treat breast cancer and melanoma. Biomater Sci. 2020;8(1):333–41.

    Article  CAS  PubMed  Google Scholar 

  81. Zhang X, Tang W, Wen H, Wu E, Ding T, Gu J, et al. Evaluation of CTB-sLip for targeting lung metastasis of colorectal cancer. Pharmaceutics. 2022;14(4):868.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Scavo MP, Cutrignelli A, Depalo N, Fanizza E, Laquintana V, Gasparini G, et al. Effectiveness of a controlled 5-FU delivery based on FZD10 antibody-conjugated liposomes in colorectal cancer in vitro models. Pharmaceutics. 2020;12(7):650.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Garcia-Pinel B, Jabalera Y, Ortiz R, Cabeza L, Jimenez-Lopez C, Melguizo C, et al. Biomimetic magnetoliposomes as oxaliplatin nanocarriers: in vitro study for potential application in colon cancer. Pharmaceutics. 2020;12(6):589.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Li YJ, Wu JY, Liu J, Xu W, Qiu X, Huang S, et al. Artificial exosomes for translational nanomedicine. J Nanobiotechnol. 2021;19(1):242.

    Article  Google Scholar 

  85. Fang Y, Zhang Z, Liu Y, Gao T, Liang S, Chu Q, et al. Artificial assembled macrophage co-deliver black phosphorus quantum dot and CDK4/6 inhibitor for colorectal cancer triple-therapy. ACS Appl Mater Interfaces. 2022;14(18):20628–40.

    Article  CAS  PubMed  Google Scholar 

  86. Cheng L, Zhang X, Tang J, Lv Q, Liu J. Gene-engineered exosomes-thermosensitive liposomes hybrid nanovesicles by the blockade of CD47 signal for combined photothermal therapy and cancer immunotherapy. Biomaterials. 2021;275:120964.

    Article  CAS  PubMed  Google Scholar 

  87. Lu M, Xing H, Shao W, Zhang T, Zhang M, Wang Y, et al. Photoactivatable silencing extracellular vesicle (PASEV) sensitizes cancer immunotherapy. Adv Mater. 2022;34(35):e2204765.

    Article  PubMed  Google Scholar 

  88. Wei R, Chen L, Qin D, Guo Q, Zhu S, Li P, et al. Liquid biopsy of extracellular vesicle-derived miR-193a-5p in colorectal cancer and discovery of its tumor-suppressor functions. Front Oncol. 2020;10:1372.

    Article  PubMed  PubMed Central  Google Scholar 

  89. 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. Mol Cancer. 2022;21(1):49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. de Miguel PD, Rodriguez Martínez A, Ortigosa Palomo A, Delgado Ureña M, Garcia Puche JL, Robles Remacho A, et al. Extracellular vesicle-miRNAs as liquid biopsy biomarkers for disease identification and prognosis in metastatic colorectal cancer patients. Sci Rep. 2020;10(1):3974.

    Article  Google Scholar 

  91. Moritz CP. 40 years Western blotting: a scientific birthday toast. J Proteomics. 2020;212:103575.

    Article  CAS  PubMed  Google Scholar 

  92. van der Pol E, Böing AN, Gool EL, Nieuwland R. Recent developments in the nomenclature, presence, isolation, detection and clinical impact of extracellular vesicles. J Thromb Haemost. 2016;14(1):48–56.

    Article  PubMed  Google Scholar 

  93. Wei P, Wu F, Kang B, Sun X, Heskia F, Pachot A, et al. Plasma extracellular vesicles detected by single molecule array technology as a liquid biopsy for colorectal cancer. J Extracell Vesicles. 2020;9(1):1809765.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Ortega FG, Gomez GE, Boni C, García IC, Navas CG, Dvries RF, et al. Microfluidic amperometric immunosensor based on porous nanomaterial towards claudin7 determination for colorectal cancer diagnosis. Talanta. 2023;251:123766.

    Article  CAS  PubMed  Google Scholar 

  95. Zhang W, Jiang L, Diefenbach RJ, Campbell DH, Walsh BJ, Packer NH, et al. Enabling sensitive phenotypic profiling of cancer-derived small extracellular vesicles using surface-enhanced raman spectroscopy nanotags. ACS Sens. 2020;5(3):764–71.

    Article  CAS  PubMed  Google Scholar 

  96. 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 Lett. 2020;474:36–52.

    Article  CAS  PubMed  Google Scholar 

  97. 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. J Hematol Oncol. 2020;13(1):156.

    Article  PubMed  PubMed Central  Google Scholar 

  98. 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. Clin Sci (Lond). 2020;134(4):419–34.

    Article  CAS  PubMed  Google Scholar 

  99. Wang S, Zhang Z, Gao Q. Transfer of microRNA-25 by colorectal cancer cell-derived extracellular vesicles facilitates colorectal cancer development and metastasis. Mol Ther Nucleic Acids. 2020;23:552–64.

    Article  PubMed  PubMed Central  Google Scholar 

  100. 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 Sci. 2021;112(9):3744–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. 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 Lett. 2020;491:22–35.

    Article  CAS  PubMed  Google Scholar 

  102. Lan J, Sun L, Xu F, Liu L, Hu F, Song D, et al. M2 macrophage-derived exosomes promote cell migration and invasion in colon cancer. Cancer Res. 2019;79(1):146–58.

    Article  CAS  PubMed  Google Scholar 

  103. 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. Mol Cancer. 2020;19(1):117.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Han K, Wang FW, Cao CH, Ling H, Chen JW, Chen RX, et al. CircLONP2 enhances colorectal carcinoma invasion and metastasis through modulating the maturation and exosomal dissemination of microRNA-17. Mol Cancer. 2020;19(1):60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. 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(18):8211–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Zhao H, Chen S, Fu Q. Exosomes from CD133+ cells carrying circ-ABCC1 mediate cell stemness and metastasis in colorectal cancer. J Cell Biochem. 2020;121(5–6):3286–97.

    Article  CAS  PubMed  Google Scholar 

  107. 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(1):933.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. 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 Dis. 2019;10(11):829.

    Article  PubMed  PubMed Central  Google Scholar 

  109. Zhang J, Li S, Zhang X, Li C, Zhang J, Zhou W. LncRNA HLA-F-AS1 promotes colorectal cancer metastasis by inducing PFN1 in colorectal cancer-derived extracellular vesicles and mediating macrophage polarization. Cancer Gene Ther. 2021;28(12):1269–84.

    Article  CAS  PubMed  Google Scholar 

  110. 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. J Exp Clin Cancer Res. 2020;39(1):54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Sun B, Zhou Y, Fang Y, Li Z, Gu X, Xiang J. Colorectal cancer exosomes induce lymphatic network remodeling in lymph nodes. Int J Cancer. 2019;145(6):1648–59.

    Article  CAS  PubMed  Google Scholar 

  112. Yun CW, Lee JH, Go G, Jeon J, Yoon S, Lee SH. Prion protein of extracellular vesicle regulates the progression of colorectal cancer. Cancers (Basel). 2021;13(9):2144.

    Article  CAS  PubMed  Google Scholar 

  113. Huang M, Chen M, Qi M, Ye G, Pan J, Shi C, et al. Perivascular cell-derived extracellular vesicles stimulate colorectal cancer revascularization after withdrawal of antiangiogenic drugs. J Extracell Vesicles. 2021;10(7):e12096.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Zheng X, Liu J, Li X, Tian R, Shang K, Dong X, et al. Angiogenesis is promoted by exosomal DPP4 derived from 5-fluorouracil-resistant colon cancer cells. Cancer Lett. 2021;497:190–201.

    Article  CAS  PubMed  Google Scholar 

  115. 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. Dig Dis Sci. 2021;66(6):1916–27.

    Article  CAS  PubMed  Google Scholar 

  116. Chen C, Yu H, Han F, Lai X, Ye K, Lei S, et al. Tumor-suppressive circRHOBTB3 is excreted out of cells via exosome to sustain colorectal cancer cell fitness. Mol Cancer. 2022;21(1):46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. 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. Mol Ther. 2021;29(9):2723–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Xu Y, Zhu M. Novel exosomal miR-46146 transfer oxaliplatin chemoresistance in colorectal cancer. Clin Transl Oncol. 2020;22(7):1105–16.

    Article  PubMed  Google Scholar 

  119. Sun LH, Tian D, Yang ZC, Li JL. Exosomal miR-21 promotes proliferation, invasion and therapy resistance of colon adenocarcinoma cells through its target PDCD4. Sci Rep. 2020;10(1):8271.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. 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. Mol Oncol. 2020;14(3):539–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. 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. Mol Cell Biol. 2021;41(5):e00517-e520.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Yao S, Yin Y, Jin G, Li D, Li M, Hu Y, et al. Exosome-mediated delivery of miR-204-5p inhibits tumor growth and chemoresistance. Cancer Med. 2020;9(16):5989–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. 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 Mol Imaging. 2022;2022:2878557.

    PubMed  PubMed Central  Google Scholar 

  124. Yang CK, Hsu HC, Liu YH, Tsai WS, Ma CP, Chen YT, et al. EV-miRome-wide profiling uncovers miR-320c for detecting metastatic colorectal cancer and monitoring the therapeutic response. Cell Oncol (Dordr). 2022;45(4):621–38.

    Article  CAS  PubMed  Google Scholar 

  125. 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. Mol Cancer. 2023;22(1):2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Li J, Yang P, Chen F, Tan Y, Huang C, Shen H, et al. Hypoxic colorectal cancer-derived extracellular vesicles deliver microRNA-361-3p to facilitate cell proliferation by targeting TRAF3 via the noncanonical NF-κB pathways. Clin Transl Med. 2021;11(3):e349.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Zhong ME, Chen Y, Xiao Y, Xu L, Zhang G, Lu J, et al. Serum extracellular vesicles contain SPARC and LRG1 as biomarkers of colon cancer and differ by tumour primary location. EBioMedicine. 2019;50:211–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. 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 (Basel). 2021;13(6):1351.

    Article  CAS  PubMed  Google Scholar 

  129. Dash S, Wu CC, Wu CC, Chiang SF, Lu YT, Yeh CY, et al. Extracellular vesicle membrane protein profiling and targeted mass spectrometry unveil CD59 and tetraspanin 9 as novel plasma biomarkers for detection of colorectal cancer. Cancers (Basel). 2022;15(1):177.

    Article  PubMed  PubMed Central  Google Scholar 

  130. 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. Front Oncol. 2021;11:725211.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This study was supported by the central government guides the local science and technology development special fund (No.2022ZYD0084), Key R&D Projects (No. 2022YFS0619, 2022YFS0630, 2022YFS0635, 2022YFS0627), the Youth Science and Technology Innovation Research Team (No. 2021JDTD0008) of the Science and Technology Department of Sichuan province of China, the Science and Technology Innovation Team from Jiucheng Science and Technology Talent Cultivation Plan in Luzhou (No.2019-1), Key Research and Development Projectors of Luzhou (No. 2021-SYF-26, 2022-SYF-85), the Research Fund (No. 210027-01SZ, 200017-01SZ, 230008-01SZ, 230007-01SZ) of Sichuan Credit Pharmaceutical CO., Ltd., Central Nervous System Drug Key Laboratory of Sichuan Province, Chongqing Traditional Chinese Medicine Inheritance and Innovation Team Construction Project “Traditional Chinese Medicine New Drug and Safety Research Inheritance and Innovation Team” (No.2022-8).

Author information

Authors and Affiliations

Authors

Contributions

LX, YW and QJ wrote the manuscript. FL, HY and LZ conceived the structure and revised the manuscript. JC, TC, JY, CP, HL, JT, SY, YZ, and XZ revised the manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Furong Liu, Hongru Yang or Ling Zhao.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Yes.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xiong, L., Wei, Y., Jia, Q. et al. The application of extracellular vesicles in colorectal cancer metastasis and drug resistance: recent advances and trends. J Nanobiotechnol 21, 143 (2023). https://doi.org/10.1186/s12951-023-01888-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12951-023-01888-1

Keywords