Skip to main content

Emerging roles of exosomal circRNAs in non-small cell lung cancer

Abstract

Despite the prevalence of non-small cell lung cancer (NSCLC) is high, the limited early detection and management of these tumors are restricted since there is an absence of reliable and precise diagnostic biomarkers and therapeutic targets. Exosomes transport functional molecules for facilitating intercellular communication, especially in the tumor microenvironment, indicating their potential as cancer biomarkers and therapeutic targets. Circular RNA (circRNA), a type of non-coding RNA possessing a covalently closed loop structure, substantial abundance, and tissue-specific expression patterns, is stably enriched in exosomes. In recent years, significant breakthroughs have been made in research on exosomal circRNA in NSCLC. This review briefly introduces the biogenesis, characterizations, and functions of circRNAs and exosomes, and systematically describes the biological functions and mechanisms of exosomal circRNAs in NSCLC. In addition, this study summarizes their role in the progression of NSCLC and discusses their clinical significance as biomarkers and therapeutic targets for NSCLC.

Background

Lung cancer, one of the most lethal malignancies globally, can be categorized into two primary forms: non-small-cell lung cancer (NSCLC) and small-cell lung cancer. NSCLC accounts for around 85% of all occurrences of lung cancer [1]. NSCLC exhibits a comparatively slower pace of growth and a prolonged disease course, and it typically does not manifest notable symptoms in its infancy, unlike small cell lung cancer [2]. The suboptimal therapeutic efficacy in NSCLC can be attributed to the paucity of reliable tumor markers and the emergence of drug resistance [3, 4]. Therefore, it is crucial to investigate NSCLC-related biomarkers to improve the efficiency of NSCLC diagnosis and treatment [5].

Exosomes, which have sizes ranging between 40 and 160 nm, are extracellular vesicles secreted by multiple kinds of active cells [6,7,8]. They form a highly heterogeneous population, and this heterogeneity manifests in four main aspects: size, content, function, and origin [9,10,11,12]. The content of exosomes influences their size, while the microenvironment of the cell and biological processes occurring within the cell can impact their content. Exosomes transport a substantial quantity of miRNAs, circRNAs, and other nucleic acids, membrane proteins, and small molecules [13, 14]. Upon being released from the cell surface, exosomes merge with the plasmalemma of recipient cells, promoting the transportation of their content into the cytoplasm [15]. Notably, cancer cells can produce a higher number of exosomes in comparison to non-malignant cells [16,17,18]. Exosomes can facilitate the occurrence and progression of NSCLC by enhancing cell migration, invasion, proliferation, and immune evasion. The study of exosomes contributes to the understanding of cell communication mechanisms and offers novel insights into the diagnosis and management of malignancies [19, 20]. Furthermore, exosomes can be modified through various methods of engineering and materials science to achieve the targeted delivery of specific drugs, which endows them with significant clinical research value and broad application prospects.

Circular RNAs (circRNAs) are plentiful endogenous RNAs characterized by circular structures created through the covalent closure of the 5’ and 3’ ends of precursor RNA [21,22,23,24]. CircRNAs are a class of highly expressed, stable, diversified and conserved RNA molecules, and the expression of circRNA has tissue specificity and disease specificity, which makes circRNA a potentially perfect biomarker [21, 25, 26]. Research has demonstrated that circRNAs have significant impacts on promoting the advancement of cancer by influencing the expression of critical genes that participate in epithelial-to-mesenchymal transition (EMT), apoptosis, proliferation, migration, and metastasis [27,28,29]. In addition, circRNAs possess the capacity to be enclosed within exosomes for intercellular transport, which are known as exosomal circRNAs [30,31,32]. In recent years, research on exosomal circRNA in cancer, such as urinary tumors, gastrointestinal cancer, etc., has received widespread attention [33,34,35,36,37,38,39,40,41,42,43,44,45]. However, the latest research progress on exosomal circRNA in NSCLC has not been reviewed yet.

In this review, we offer an in-depth exploration of the latest research progress pertaining to the biogenesis, distinctive features, and functional roles of circRNAs. In addition, we provide a comprehensive summary of the current research progress on the mechanism of circRNA entering exosomes. Finally, we explore the role of exosomal circRNAs in NSCLC and the potential of employing exosomal circRNAs as biomarkers for NSCLC.

Exosomes, circrnas and exosomal circRNAs

Characteristics and biological roles of exosomes

Exosomes, typically measuring 40–160 nm in diameter, are extracellular vesicles derived from endosomes formed through ongoing inward folding of cytomembrane, resulting in the generation of multivesicular bodies. They are able to fuse with another intracellular vesicle or organelle, resulting in the variety and heterogeneity of exosomal components [46,47,48,49].

Exosomes contain multifarious substances, including transmembrane proteins, cell adhesion molecules, scaffold proteins, RNA-binding proteins, RNA, DNA, and complex polysaccharides [50]. Exosomal proteins are specifically modificated by a significant biological process named ubiquitination. Several exosomal proteins are nonspecific molecules and can serve as a type of effective biomarkers [51,52,53,54], which can be broadly categorized into two types. According to the biological composition, the first kind of biomarker is protein biomarkers, such as tetraspanins (CD9, CD63, CD81), followed by cytoplasmic proteins including Actin and calcium phospholipid binding protein (Annexins), in addition to CEA (carcinoembryonic antigen) apoptosis through gene interactions protein X2 (ALIX), Her2 (human epidermal growth factor receptor-2), tumor susceptibility gene 101 (TSG101) protein, heat shock protein (HSP70 and HSP90), and EpCAM (epithelial cell adhesion molecule). The second type of exosomal biomarker are certain kinds of nucleic acid, such as miR-141, miR-15a-5p, miR-10b, and mutated KRAS DNA [55, 56]. Specific exosomal miRNAs have been confirmed to serve as non-invasive biomarkers for colorectal cancer, indicating information about pathological changes sensitively and accurately [57]. The potential of exosomes to serve as biomarkers has garnered extensive attention and investigation across a spectrum of diseases, including cardiovascular disorders, liver diseases, infectious diseases and cancer. Notably, a recent study elucidated the crucial role of exosomes in osteoporotic effects, identifying that exosomes originated from the plasma of murine models exposed to cold temperature (CT-EXO) sponged miR-25-3p to impact targeted gene SATB2 which ultimately attenuating autophagy in the advancement of osteoporosis [58]. The differences in exosome composition, especially on cell surface proteins impact recipient cells. Distinct cell surface indicators enable the purification of specific subpopulations of exosomes [49, 59] and also endow exosomes with the capacity to play a pivotal role in intercellular communication.

Upon the secretion into the extracellular environment, exosomes may interact with recipient cells through three mechanisms [60,61,62,63]. The first is the ligand-receptor interaction [64], which refers to the communication between molecules on the outer layer of exosomes and molecules on the target cytoplasmic membrane. The second is endocytosis [65], where recipient cells can absorb exosomes through grid protein-dependent and grid protein-independent endocytic processes, such as phagocytosis, macropinocytosis, and engulfment. The small membrane-bound structures are enriched with proteins and genetic information originating from the cells from which they are derived. Specifically, exosomes that originate from tumors contain oncogenic proteins and genes [65, 66]. The third is direct membrane fusion [67], where exosomes fuse directly with plasmalemma, secreting their contents into the cytoplasm of the recipient cell [68, 69]. In the process of direct fusion, these vesicles merge instantaneously with the plasma membrane of recipient cells through membrane fusion, which is facilitated by the interaction with ubiquitous fusion-associated molecules, including SNARE proteins [69].

Moreover, originating from a host-cell lipid membrane, exosomes exhibit organotrophic and tissue tropism modulated by surface molecules, granting them stability and targeted delivery capabilities in intercellular communication. These attributes pave the way for exosomes to play a pivotal role in transmitting cancer cell signals and influencing cancer progression. Exosomes are more abundantly secreted by cancer cells than by their normal counterparts and are capable of exchanging materials with diverse cellular populations. Numerous studies have demonstrated that exosomes either promote or inhibit the activation and metastasis of cancer cells and cause drug resistance of anti-cancer drugs in multiple aspects. Xia et al. found that exosomes derived from HCC cells rich in Smoothened (SMO) can be absorbed by hepatic stellate cells and activate cancer-associated fibroblasts (CAFs) through the Gli1-MIRLET7BHG signaling pathway, thereby influencing the tumor microenvironment [70]. A large number of studies have shown that tumor cell-derived exosomes can accelerate cancer progression by secreting a variety of secretagogues [71]. In the case of cervical squamous cell carcinoma, exosomes express elevated high levels of miR-221-3p, and they transport it to lympho-endothelial cells (LECs). This promotes migration and lymphangiogenesis in vitro by down-regulating vascular hibin-1, a well-established blocker of lymphangiogenesis, and it promotes lymph node (LN) metastasis in mice [72, 73]. Exosomal circGSE1 facilitates immune escape from HCC by promoting the growth of regulatory T cells [74]. An immunosuppressive microenvironment in exosomes is created by circTMEM181 to imbue HCC with anti-PD1 resistance by increasing CD39 expression within macrophages [75, 76]. Intercellular transportation of wild-type EGFR proteins encapsulated in exosomes activates PI3K/AKT and MAPK pathways, triggering osimertinib resistance [77]. Gastric carcinoma cell-derived exosomes induce autophagy in neutrophils and promote cancer activation through HMGB1/TLR4/NF-κB pathway [78]. HucMSC-derived exosome delivers BECN1 to regulate the xCT/GPX4 axis to induce hepatic stele iron death, and it also carries miR-378a-5p to inhibit the NLRP3 inflammasome to alleviate colitis [79, 80].

In addition to being a medium of intercellular communication, exosomes also exert a significant part in regulating immune responses. By modulating the activity of immune cells via diverse mechanisms, including the release of cytokines and bioactive molecules and the expression of immune-inhibitory cell surface markers, exosomes contribute to alleviate inflammation and immune responses [78, 81, 82]. For instance, exosomal lnc-AFTR inhibits staphylococcus aureus-induced cell apoptosis and inflammatory responses by suppressing the activation of the TNF pathway and MAPK pathway [83]. TGF-β1 enriched in MDSCs-Exos weakens NK cells’ cytotoxicity and promotes the immunosuppressive properties of MDSCs Exos by inducing Tregs or Th17 cells [84]. Tumour-derived exosomes (TEX) have potential immunogenicity due to their ability to carry tumour antigens. For example, when combined with cytokine IL-23, extracellular vesicles collected from tumor cells overexpressing Rab27a can produce strong and long-lasting anti-tumor immune effects [85]. Platelet-rich plasma-derived exosomes promote nerve repair and regeneration in patients with peripheral nerve damage by stimulating the PI3K/Akt signaling pathway in bone marrow mesenchymal stem cells [86]. In conclusion, exosomes, as crucial messengers of intercellular communication, not only participate in the exchange of substances and information between cells, but also play an important role in immune regulation.

Furthermore, exosomes possess significant characteristics including remarkable stability, high biocompatibility, minimal toxicity, low immunogenicity, the ability to permeate biological barriers and extended circulation in vitro environment, making them ideal vehicles for drug delivery [87,88,89,90]. For example, hnRNPK mediates the sorting of miR-4732-3p into exosomes, and exosomes help to form immune escape and promote the progression of NSCLC [91]. They are able to transport multifarious categories of drugs including small molecule chemical drugs, proteins, and peptide drugs, as well as large biomolecule drugs, such as antibodies and enzymes. Exosomes have the ability to actively migrate to diseased tissues and release their cargo into target cells, exerting precise targeted therapeutic effects. Benzo[a]pyrene-induced tumor exosomes can deliver circ_0011496 to activate lung fibroblasts by miR-486-5p/TWF1/MMP9 cascade, promoting HCC metastasis [92]. Additionally, exosomes mediate the link among diverse environmental stressors and negative influences and LC, particularly implicated in processes such as triggering pulmonary fibrosis, altering the tumor microenvironment, promoting apoptosis resistance, and suppressing immune reactions. For example, lung cancer cell-derived exosomes (LCCDEs) can trigger a pro-inflammatory state in mesenchymal stem cells through the NFκB-TLR signaling pathway [93]. Drug formulations based on exosomes can be applied to multiple kinds of diseases including cardiovascular diseases, infectious diseases, neurodegenerative diseases and cancer [94]. Drug delivery systems mediated by extracellular vesicles can inhibit tumor proliferation and metastasis in gastrointestinal cancer and overcome drug resistance [95].

Biogenesis, characterization, and functions of circRNAs

CircRNA is generated through a reverse splicing mechanism in which the 5’ splicing site is linked to the 3’ splicing site, resulting in the formation of a closed-loop structure [96,97,98]. CircRNAs are categorized into three groups based on their composition: Exon-intron circRNAs (EIciRNAs), Exonic circRNAs (EcircRNAs), Circular intronic RNAs (CiRNAs). (1) The majority of ElciRNAs, when located in the nucleus, interact with U1 small nuclear ribonucleoproteins (snRNPs) and positively regulate the transcription of their parent genes in a cis approach [99]. (2) EcircRNAs are identified with the cytoplasm and contain one or more exons, accounting for 80% of the identified circRNAs [100]. (3) CiRNAs are predominantly localized in the nucleus, contain only introns, and possess the ability to transcriptionally promote Pol II within the nucleus [101].

To date, there are four proposed models of circRNA biogenesis: intron pairing-driven circulation, RNA-driven circularization, lariat-driven circularization and circularization intronic RNA, as depicted in Fig. 1. In intron pairing-driven circulation, splicing sites are approached through the complementary base pairing of reverse repeat sequences in introns on both sides of the circular exon. Reverse splicing involves the upstream branching point (BP) attacking, in which BP in turn attacks the upstream SA site, forming EIciRNAs or EcircRNAs [21]. Additionally, RNA binding proteins (RBPs) can bind to specific motifs in flanking introns, promoting RNA-driven circularization [102, 103]. CircRNAs can also be formed through the splicing of intermediates called lariat precursors, which are generated when an exon is skipped during linear splicing or when intronic lariat precursors manage to avoid the debranching process of canonical linear splicing [104]. In circularization intronic RNA, CiRNAs containing both a 7 nt GU-rich element around the 5’ splice site and an 11 nt C-rich element close to the branch-point site are spared, then CiRNAs appear.

Fig. 1
figure 1

Biogenesis and functions of circRNAs. CircRNAs form EIciRNAs, EcircRNAs, and CiRNAs through four hypothesized mechanisms: intron pairing-driven circulation, RNA-driven circularization, lariat-driven circularization and circularization intronic RNA. Reverse direct splicing or RNA-binding proteins (RBPs) bind to specific motifs in flanking introns, promoting RNA-driven circularization. Splicing intermediates known as lariat precursors, which result from intronic lariat precursors that elude the debranching stage of canonical linear splicing or from an exon-skipping event during linear splicing, can also produce circRNAs. CiRNAs are generated by the retention of a 7 nt GU-rich element and an 11 nt C-rich element. CircRNAs can be sponges, interact with proteins, regulate gene expression, and translate proteins. They serve as biomarkers, therapeutic targets, and therapeutic agents. Detailed information on circRNAs was sourced from Lingling Chen

The molecular mechanisms underlying the formation of circRNA are still under continuous exploration, and the latest research has revealed some new molecular mechanisms underlying the generation of circRNAs. For example, ESRP1 (epithelial splicing regulatory protein 1) controls the biosynthesis of circDOCK1 by binding to the repeating GGU motifs in intron 1 and preventing its splicing until Pol II completes its 157 kb journey to exon 27 [105]. Tang et al. found that back-splicing predominantly occurs at m6A-enriched locations, which are often found near the start and stop codons in linear mRNAs, for a subset of circRNAs [106]. Another study demonstrates a widespread role for NOVA2 in enhancing circRNA biogenesis during neuronal differentiation and YCAY clusters located in flanking introns were found to be responsible for the impairment of NOVA2-mediated back-splicing of circEfnb2 [107]. Liang et al. showed that the steady-state levels of circRNAs are elevated when canonical pre-mRNA processing activities are inhibited or slowed down. This is partially due to the fact that developing RNAs are diverted into other pathways that result in the synthesis of circRNAs [24, 108]. Additionally, studies on viral circRNA biogenesis revealed that they primarily use the alternative 5’ splicing site (A5SS) for variable splicing and exhibit a non-standard reverse splicing signal that is significantly higher than that of mammals and plants [109]. In summary, the molecular mechanism of circRNA generation is complex and further research is needed.

CircRNAs are abundant in eukaryotic cells, and they display characteristics as follows [110]. (1) High stability: CircRNAs are more steady than linear RNAs. CircRNAs are rarely degraded by exonuclease R and niacin phosphatase 5′-terminal exonuclease and have a longer half-life [111]. (2) Evolutionary conservation: CircRNA sequences exhibit evolutionary conservation across various species. One example of conserved circRNAs originates from the human PHF21A locus and the mouse Phf21a locus [111]. (3) High abundance: CircRNAs have higher expression than that of corresponding linear RNAs [112]. In the environment of HEK293 and whole blood specimens, the abundance of circRNA can exceed that of the corresponding linear RNA by at least 30 times [113]. (4) Tissue specificity: CircRNAs also exhibit tissue, disease, and developmental stage-specific expression, endowing them with significant potential as biomarkers. Notably, there is abundant circRNA derived from normal cells and cancer cells in human body fluids, which provides potential for circRNA to be used as a biomarker for forensic body fluid identification [114,115,116,117] and clinical liquid biopsy diagnosis of various types of cancers [28, 118, 119]. Importantly, mounting evidence has demonstrated that numerous circRNAs participate in stem cell differentiation, highlighting the role of circRNAs in a wide range of physiopathological processes and the potential as therapeutic targets for stem cell-based therapy [120].

Functionally, circRNAs can be sponges, interact with proteins, regulate gene expression, and protein translation [121,122,123,124] (Fig. 1). Circ6834 functions as a molecular sponge for miR-873-5p and increased the expression of the TXNIP gene, leading to the inactivation of the TGF-β/Smad signaling pathway in NSCLC cells [125]. CircRNAs are also capable of regulating tumor cell proliferation, migration, and apoptosis, inducing multidrug resistance, modulating the tumor microenvironment, and facilitating immune evasion through diverse signaling pathways. For instance, lung cancer progression is impeded by o8G-modified circPLCE1 through the mechanism of chaperone-mediated autophagy [126]. Besides, recent studies have revealed that circRNAs can function through novel regulatory mechanisms, including the formation of circR-loops and the interaction with protein kinase R (PKR) to impact innate immune responses. CircRNAs combine with DNA to form an R-loop structure [127], influencing chromatin structure, interacting with other ncRNAs [128, 129], and transcriptionally impacting gene expression [130]. Moreover, for immune regulation, circRNAs form a small stem-loop structure that binds to PKR in normal circumstances. In a Lewis lung carcinoma model, circRNA encoding interleukin-12 (IL-12)) induces a robust immune response to regress lung tumors [131]. Upon viral stimulation, RNase L degrades the circRNAs to release PKR and initiate immune responses [132]. CircRNAs can also influence intracellular activities through phosphorylation processes. The circLIFRSA/miR-1305/PTEN pathway mitigates oncogenic processes in NSCLC through the modulation of AKT phosphorylation [133]. Research on the functions of circRNA is continuously expanding as an emerging field. Recent studies have focused on the impact of circRNA folding structures on their biological functions [134], hinting at future discoveries of additional functions and mechanisms with improved research techniques.

Biogenesis of exosomal circRNAs

Due to the multiple biological functions of exosomal circRNA that are currently being discovered and confirmed, the mechanism of circRNA entering exosomes is of significant research value. Researchers have observed that circRNAs are stably and abundantly expressed in exosomes [135]. In recent years, the process by which circRNAs are transported into exosomes has been extensively studied. Although the specific mechanisms are still being explored, there have been some important discoveries and hypotheses (Fig. 2).

Fig. 2
figure 2

Bioactive molecules on exosomes, the mechanisms of circRNAs’ entry into exosomes, and the therapy strategies of exosomes. Various bioactive molecules on exosomes can be used as biomarkers for disease. They can be roughly divided into two categories: protein biomarkers such as CD9, CD63, CD81, CEA (carcinoembryonic antigen), Her2 (human epidermal growth factor receptor-2), and EpCAM (epithelial cell adhesion molecule); nucleic acid biomarkers. Exosomes play a role in gene therapy, targeted therapy, drug delivery, immune regulation, etc. The mechanisms by which circRNAs enter exosomes include (A) RNA-binding protein mediated mechanism, (B) miRNA-mediated mechanism, (C) mechanisms mediated by RNA modification, and (D) mechanisms under cellular stress. In RNA-binding protein-mediated mechanism, circRHOBTB3 can be specifically sorted into exosomes by interacting with SNF8, a member of the ESCRT-II complexes, via its distinctive components. CircCCAR1, circ-CDYL and circNEIL3 can be packaged into exosomes by hnRNPA2B. In miRNA-mediated mechanism, exosomes with miR-7 mimics can effectively decrease the levels of competing endogenous spongy circCDR1as, and miR-671-AGO2 mediates the degradation of circCDR1as in source cells. In mechanism mediated by RNA modification, the structure and function of circRNAs may be affected by RNA modifications. M6A modification of circ-CDYL can promote the sorting of circ-CDYL into exosomes. In addition, when cells are exposed to external stimuli or stress conditions, the integration of circRNAs into exosomes is also affected. Exosome therapy strategies primarily include drug delivery, targeted therapy, gene therapy, and immune regulation

RNA-binding protein mediated mechanism

Studies have revealed that RNA-binding proteins (RBPs) extensively participate in RNA transport and regulation within cells and circRNAs can form complexes with RBPs and enter exosomes through interaction [136, 137]. Protein heterogeneous nuclear ribonucleoprotein A2B1 (hnRNPA2B1) is an RBP exerting significant regulatory function in controlling the transport and subcellular localization of specific mRNAs in neurons, thereby influencing neuronal function and development [138]. Moreover, hnRNPA2B1 is essential for the selective packaging of miRNAs and lncARSR into exosomes [139, 140]. CircRNAs are preferentially incorporated into exosomes over linear RNAs, yet the specific sorting mechanism for circRNAs in exosomes is not well understood [141]. For circRNAs, it has been reported that hnRNPA2B1 can facilitate the packaging of circCCAR1, circ-CDYL and circNEIL3 into exosomes [75, 142, 143]. Another study suggested that KHDRBS3 mediates the entry of circ_0088300 into exosomes [144]. Additionally, the mechanism of circRNAs sorted into exosomes may also be influenced by protein complexes in exosomes. For example, circRHOBTB3 has been shown to be specifically sorted into exosomes through the interaction with SNF8, a constituent part of ESCRT-II complex, with its own unique components [141]. CircFAT1 may be transported into exosomes by some specific RNA-binding proteins [145]. Another study found that circRNAs presumably utilize similar transportation procedure as that of linear RNAs, where RBPs specifically selectively identify RNAs that possess matching binding motifs. A study showed that exosomes have the capability to specifically encapsulate circRNAs compromising 5’-GMWGVWGRAG-3’ motifs [146]. However, the molecular mechanism that contributes to the selective entry of circRNA into exosomes still needs further investigation.

MiRNA-mediated mechanism

Some studies suggest that circRNAs can connect to miRNAs, perform as miRNA baits, and inhibit their activities [147]. Studies have shown that the miR-7 simulation of cells may trigger a notable decrease of competitive endogenous spongiform circCDR1as within exosomes, suggesting that the process of selecting of circRNAs into exosomes is partially controlled by alterations in the amounts of associated miRNAs [135, 148, 149]. Furthermore, miRNAs can regulate the intracellular breakdown of circRNAs, leading to decreased expression of circRNAs in exosomes. For instance, miR-671-AGO2 mediates the degradation of circCDR1as in source cells, which may result in decreased circCDR1as expression level in exosomes [150].

Mechanism mediated by RNA modification

RNA modifications play an important regulatory role in many aspects of the RNA post-transcriptional process, including splicing, processing, export, stability, degradation, and translation [151, 152]. In eukaryotic cells, the predominant form of RNA modification is regulated by N6-methyladenosine (m6A) [153]. M6A modification exerts a crucial role in enriching and regulating the physiological properties of circRNAs [154]. Research has shown that m6A modification of circNSUN2 promotes its translocation to the cytoplasm [155]. In addition, RNA modifications may affect the process of circRNAs’ entry into exosomes [156]. Of note, it has been reported that m6A modification of circ-CDYL promoted the exosomal sorting of circ-CDYL [143]. However, the role of RNA modification in the process of circRNA entering exosomes still needs further investigation.

Mechanism under cellular stress

When cells are exposed to external stimuli or stress conditions, the composition and release of exosomes may change, which in turn affects the types and amounts of circRNAs contained in them. This change may be related to the biological response of cells under stress [157]. For example, the characteristics of exosomal circRNAs observed in tumor development, immune response, and metabolic diseases are different from those under normal conditions, so exosomal cirRNAs can also be employed as diagnostic markers for multiple kinds of diseases [158, 159].

These studies provide some clues for understanding the mechanism of circRNAs’ entry into exosomes, but further experimental validation and in-depth research are still needed. With the continuous advancement of technology and a deeper understanding of exosome biology, more mechanisms regarding the entry of circRNAs into exosomes will be revealed.

Exosomal circRNAs in NSCLC

In recent years, the research interest in the exosomal circRNAs in NSCLC has rapidly increased. We conducted a search on the PubMed website for all articles on the research of exosomal circRNA in NSCLC published before August 12, 2024. After manual verification and exclusion of reviews and retracted articles, a total of 30 articles have been identified as target articles to be included in our review.

Exosomal circRNAs and angiogenesis

Angiogenesis is one of the pivotal areas of investigation in oncology due to its crucial role in tumor progression. In particular, angiogenesis plays an integral part in NSCLC, facilitating the growth and metastasis of tumor cells by establishing a robust vascular network that supplies essential nutrients and oxygen. This network supports the sustained proliferation of cancer cells and enables their dissemination throughout the body via the bloodstream. Research has uncovered novel mechanisms underlying NSCLC tumorigenicity, with one study highlighting that cancer-released exosomal circ0008717 promotes angiogenesis in NSCLC progression. The number of formed branches of human umbilical vein endothelial cells (HUVECs) was reduced in the si-circ_0008717 group. Mechanism studies showed that exosomal circ0008717 specifically targeted the miRNA-1287-5p/P21-activated kinase 2 (PAK2) axis to speed up NSCLC advancement [160].

Exosomal circRNAs and lung cancer proliferation, migration, and invasion

Exosomal circRNAs exhibit a substantial impact on proliferation, metastasis, angiogenesis, and apoptosis of NSCLC. Chen et al. reported that the upregulation of NSCLC cell-derived exosomal circFARSA leads to increased metastasis of NSCLC cells, which is prompted by the induction of M2 polarization via the ubiquitylation and degradation of PTEN, ultimately activating the PI3K/AKT pathway [161]. CircSHKBP1 was upregulated in NSCLC tissues and cell lines and was enriched in their exosomes. It was proved that circSHKBP1 within exosomes promotes NSCLC proliferation, migration, invasion, and stemness [162]. Fang et al. proved that serum-derived exosome-mediated circARHGAP10 drives NSCLC cell growth via the miR-638/FAM83F pathway [163]. Additionally, high expression of circPLK1 in serum exosomes enhances HMGA1 levels by sponging miR-1294 and promotes the growth of NSCLC [164]. It has shown that the up-regulated expression of has-circ-0002130 in serum exosomes of patients with ocitinib-resistant NSCLC promotes NSCLC progression [165]. Moreover, Ning et al. found that exosomal circ0007385 promotes the NSCLC process by targeting miR-1253 and FAM83A [166]. The abundance of circCD226 was elevated in serum exosomes of NSCLC. Exosomal circCD226 can promote NSCLC advancement via the miR-1224-3p/HMGA2 axis [167]. Furthermore, exosomal circPIP5K1A relies on the miR101/ABCC1 axis, enhancing the proliferation and migration of NSCLC cells [168]. It was found that circCCDC134 highly expressed in the serum exosomes of NSCLC patients. Exosomal circCCDC134 regulates NFAT5 by sponging miR-625-5p and promotes the NSCLC process [169]. Circ_0002476 is transported between cells by exosomes, and it is found that circ-0002476 promotes NSCLC progression and mtDNA damage via the miR-1182/TFAM axis [170]. Zhang et al. observed that circSATB2 was highly upregulated in NSCLC cells and tissues. Databases for circRNA-miRNA binding prediction and FISH (fluorescence in situ hybridization) to detect the sublocalization of circRNAs were utilized. Mechanism studies revealed that circSATB2 positively regulated fascin homolog 1, actin-bundling protein 1 (FSCN1) expression by sponging miR-326, and transits through exosomes to promote NSCLC progression, NSCLC progression, and may stimulate anomalous growth in healthy bronchial epithelial cells [32]. Exosomal ERBB2IP is pivotal in promoting tumor growth in NSCLC by upregulating PSAT1 expression [171]. Additionally, exosomal circ-0008928 inhibits NSCLC development through the miR-488/HK2 axis [172]. Wang et al. proved that a hypoxia-induced exosomal circPLEKHM1 facilitates PABPC1-eIF4G interaction to promote NSCLC cell metastasis by increasing OSMR expression and M2 polarization of macrophages [173]. In the experiment of Liu et al., circPLK1 highly expressed in serum exosomes from NSCLC patients, and it was demonstrated that glycolysis and NSCLC progression can be accelerated through the circFTO/miR-148a-3p/PDK4 axis [174]. These studies demonstrate that exosomal circRNA is involved in multiple aspects of NSCLC progression (Fig. 3; Table 1), suggesting it may be a potential therapeutic target that requires further investigation.

Fig. 3
figure 3

Diagram of the functions of exosomal circRNAs in various aspects of NSCLC. The functions of exosomal circRNAs in NSCLC include lung cancer proliferation, metastasis, angiogenesis and apoptosis, in addition to playing roles in metabolism, tumor immune microenvironment, and chemoresistance, making exosomal circRNAs potential biomarkers and therapeutic targets

Table 1 Exosomal circRNAs in non-small cell lung cancer

Exosomal circRNAs and metabolism

Altered energy metabolism is a hallmark of malignancies as a result of the necessity to provide the essential nutrients for tumorigenesis and advancement, and the swift growth of cancer cells is dependent on glycolysis [175, 176]. Chen et al. have found that exosomal circSHKBP1 modulates glycolysis with the aid of PKM2 in a HIF-1α-dependent manner, promoting NSCLC progression [162]. Ding et al. have investigated that knocking down serum exosomal circ-MEMO1 inhibits glucose absorption and attenuates lactate synthesis, indicating its role in promoting glycolysis in them [177]. Elevating serum-derived exosome-mediated circARHGAP10 leads to a significant increase in glucose consumption and lactic acid production, contributing to glycolysis in NSCLC cells [163]. Furthermore, exosomal hsa-circ0002130 influences glycolysis in osimertinib-resistant NSCLC cells by conducting as miR-498 sponge and facilitating the modulation of GLUT1, HK2, and LDHA, which are key factors involved in glycolysis [165]. Exosomal circCCDC134 contributes to NSCLC development by suppressing the glucose utilization, lactate generation, and ATP levels of PC9 and A549 cells to hinder glycolysis [169]. It is shown that exosomal circ-0008928 inhibits the glycolytic metabolism of NSCLC via the miR-488/HK2 axis [172] (Fig. 3; Table 1).

Exosomal circRNAs and tumor immune microenvironment

The tumor immune microenvironment is a dynamic ecosystem in which cancer cells interact with stromal components, immune cells, and various soluble factors in complex ways. In this microenvironment, exosomes have gained attention due to their potential dual role as drivers of tumor development and regulators of immune responses [8]. Research has revealed that exosomal circRNAs function as innovative genetic information molecules which can facilitate the interaction between malignant cells and microenvironmental cells [35, 178]. Chen and colleagues have demonstrated that exosomal circUSP7 suppresses the activities of CD8 + T cells by increasing the levels of Src homology region 2 (SH2)-containing protein tyrosine phosphatase 2 (SHP2) through sequestering miR-934 [179] (Fig. 3; Table 1). These studies indicate that exosomal circRNAs affect NSCLC by regulating specific components in the tumor immune microenvironment. However, as a complex and dynamically changing microenvironment, the exact regulatory role of exosomal circRNA in the tightly interconnected microenvironment of various components may not yet be fully explored when based on one or several cell lines cultured in vitro. Therefore, further research is required in this regard. (Fig. 3; Table 1).

Exosomal circRNAs and chemoresistance

More and more evidence indicates that exosomal circRNAs play an essential role in tumor chemotherapy resistance [39, 180]. Chen et al. elucidated the role of exosomal circUSP7 in anti-PD1 therapy resistance in NSCLC. ExoQuick Exosome Precipitation Solution kit was utilized to isolate exosomes from patients’ plasma. Transmission electronic microscopy (TEM), NanoSight, and western blotting were employed to characterize exosomes. CircRNA precipitation (circRIP) and luciferase reporter revealed that exosomal circUSP7 blocked CD8 + T cell function by enhancing the expression of Src homology region 2 (SH2)-containing protein tyrosine phosphatase 2 (SHP2) through sponging miR-934, inducing CD8 + T cell dysfunction and diminishing the efficacy of anti-PD1 therapy in NSCLC [179]. Exosomal circ_PIP5K1A decreases the sensitivity of NSCLC to cisplatin therapy by sponging miR-101 [168], and overexpression of circ_0014235 in NSCLC serum-derived exosomes increases the resistance of A549 and H1299 cells to cisplatin (DDP) [181]. In addition, expression of circ_0008928 increased in serum exosomes of CDDP-resistant NSCLC patients promotes CDDP resistance in NSCLC [172], and serum exosomal circVMP1 knockdown exhibits inhibitory effects on resistance to cisplatin (DDP) [182]. Furthermore, circ_0076305 increased in NSCLC cell-derived exosomes can promote DDP resistance within NSCLC cells by modulating ABCC1 via miR-186-5p [183]. Increasing exosome-derived circKIF20B restores gefitinib sensitivity by modulating the cell cycle, accelerating apoptosis, and reducing mitochondrial oxidative phosphorylation [184]. A recent study showed that exosomal circRNA_102481 derived from tumor cells exerts a significant function in the advancement of resistance to EGFR-TKIs (epidermal growth factor receptor tyrosine kinase inhibitors) in NSCLC mediated through the miR-30a-5p/ROR1 axis [185]. (Fig. 3; Table 1).

Exosomal circRNAs and apoptosis

Apoptosis is a fundamental mechanism in tumor biology that ensures cellular homeostasis and eliminates potentially oncogenic cells. In NSCLC, apoptosis functions as a crucial checkpoint, curbing the uncontrolled proliferation of cancer cells and thereby influencing disease progression and therapeutic responsiveness. Ding et al. have shown that the silencing of exosomal circMEMO1 potently induces apoptosis in NSCLC cells, whereas its overexpression significantly dampens the apoptotic rate. Further mechanism exploration revealed that the downregulation of exosomal circ-MEMO1 inhibits NSCLC tumor growth by decreasing KRAS protein expression and elevating miR-101-3p levels [177].

Exosomal circRNAs and epithelial mesenchymal transition

Epithelial-mesenchymal transition (EMT) plays a crucial role in the progression of non-small cell lung cancer (NSCLC) by enhancing cancer cell mobility and invasiveness, which is characterized by the downregulated expression of the epithelial cadherin (E-cadherin) and upregulated expression of the mesenchymal cadherin (N‐cadherin and Vimentin). Zhang et al. demonstrated that overexpression of exosomal circ_100395 hampered the EMT of H1650 cells and the Hippo/YAP pathway, thereby inhibiting NSCLC advancement [186]. Liu et al. observed that downregulated expression of exosomal circ_0000735 enhanced E‐cadherin expression and decreased the expression of N‐cadherin and Vimentin, indicating the potential of exosomal circ_0000735 to induce EMT in NSCLC [187]. These findings highlight the potential of exosomal circ_100395, and circ_0000735 in regulating NSCLC progression through its influence on EMT.

Therapy potential of Exosomal circRNAs in NSCLC

Exosomal circRNAs are widely involved in various physiological and pathological processes, and their roles as biomarkers, therapeutic targets, and therapeutic agents are being extensively investigated. In non-small cell lung cancer, current research primarily focuses on the exploration and validation of diagnostic biomarkers and therapeutic targets.

Regarding clinical biomarkers, exosomal circRNAs present potential as diagnostic biomarkers for NSCLC. It has been shown that exosomal circFARSA can facilitate communication between macrophages and NSCLC cells via the PTEN/PI3K/AKT pathway, highlighting its potential as a promising biomarker for the diagnosis of NSCLC [161]. Serum exosomal hsa_circ_0069313 can differentiate benign lung tumors from NSCLC with AUC values of 0.803 and 0.749, respectively [188]. Moreover, serum exo-circ-MEMO1 [177], exo-circ_0048856 [189], and exosomal circCCDC134 [169] also show diagnostic potential for NSCLC. Xian et al. reported that in serum exosomes, circ0047921 exhibits the ability to distinguish between NSCLC cases and COPD controls, and the combination of circ0056285 and circ-0007761 shows potential in distinguishing NSCLC cases [190]. In addition, Chen et al. demonstrated that encapsulated within exosomes, circ_0061407 and circ_0008103 are transported to recipient cells, having good diagnostic value for NSCLC [191]. Also, the expression of serum exosomal circVMP1 is significantly upregulated in DDP-resistant NSCLC patients, which shows its diagnostic value [182] (Fig. 3; Table 1).

In recent years, targeted therapy has become a therapeutic tool in oncological therapy, replacing chemotherapy, which is prone to side effects and drug resistance [166, 192]. CircRNAs with anticancer properties, which are facilitated by extracellular vesicles, have the ability to enter tumor cells or hinder their production, so they hold promise as therapeutic targets and the basis for developing circRNA-based therapies [175]. Has-circ-0002130 was upregulated in serum exosomes from osimertinib-resistant NSCLC patients. Hsa-circ-0002130 knockdown suppresses cancer development in vivo, making it a potential therapeutic target for osimertinib-resistant NSCLC. It also significantly promotes the advancement of oxitinib-resistant NSCLC [165]. Similarly, the suppression of exosomal circ-0007385 inhibited the advancement of NSCLC by modulating the expression of miR-1253 and FAM83A [166]. Additionally, the inhibition of exosomal circCCDC134 effectively suppresses the growth, metastasis, and glycolysis of NSCLC [169]. Exosomes secreted by Tanreqing-treated NSCLC cells deliver circ-WDR78 to untreated NSCLC cells, thereby curbing the malignancy of the recipient tumor cells [193]. These all indicate the great potential of exosomal circRNAs as potential therapeutic targets for NSCLC treatment (Fig. 3; Table 1). However, further investigation is required to determine their specific physiopathology activities and underlying mechanisms in NSCLC.

However, regarding therapeutic agents based on exosomal circRNAs, relevant research in NSCLC is still in its infancy. Despite the promising results obtained from studies on diagnostic biomarkers and therapeutic targets, the development of circRNA-based therapeutic agents requires further investigation. Future efforts should be directed towards exploring the specific physiopathology activities and underlying mechanisms of exosomal circRNAs in NSCLC, as well as optimizing their therapeutic potential.

Conclusions and perspectives

Extracellular vesicle-derived circRNA is closely associated with the occurrence and progression of NSCLC. It participates in various mechanisms involved in NSCLC development, including promoting tumor proliferation, metastasis, angiogenesis, and apoptosis. It can also suppress anti-tumor immune response, regulate NSCLC immune evasion, and induce chemotherapy resistance. Furthermore, an increasing number of extracellular vesicle-derived circRNAs have been identified as potential early diagnostic markers and therapeutic targets for NSCLC, such as circ0047921, circFARSA, circ0069313, and circCCDC134.

In recent years, some clinical trials and applications on exosomes, exosomal RNA and NSCLC have begun to be implemented. Clinical trials have been registered to predict the immunotherapeutic effects, efficacy, and adverse reactions of anlotinib in the treatment of advanced NSCLC using exosome detection (NCT04427475, NCT05218759) [194, 195]. However, exosomal circRNAs have not yet been applied in clinical settings. This may be due to technical challenges in detecting specific circRNAs in exosomes, as circRNAs have common exonic sequences with their corresponding linear counterparts. Furthermore, the obstacles of exosome purification and circRNA extraction also limit the clinical validation of the reliability of exosomal circRNAs as diagnostic markers and therapeutic targets [36]. However, the rapidly developed sequencing technology and droplet digital PCR (ddPCR) with high specificity and sensitivity are increasingly being used in liquid biopsies, which are more suitable for testing with a limited size of patient cohorts. Moreover, machine learning combined with ddPCR is being utilized to upgrade the accuracy of clinical applications [196]. Moreover, novel modalities for exosomal circRNA detection depended on electrochemistry, rolling circle amplification and biosensors are gradually emerging [197, 198]. Excitingly, various engineering materials science methods are widely used to study non-coding RNAs and exosomes, which significantly expands their application scenarios [64, 199, 200]. In recent years, engineered exosomes designed with additional surface features and medicinal compounds within their structure, such as antitumor drugs and siRNA, have attracted increasing attention, particularly in the field of cancer, due to their temporal and spatial targeting capability [66, 94, 201, 202]. However, despite the increasing reports on exosome engineering strategies, there is still a lack of ideal targeting molecules that can guide exosomes to tumor sites to prevent them from being absorbed by normal cells. Notably, the anticancer potential of circRNAs is currently under extensive investigation. Consequently, engineered exosomes carrying these circRNAs may exhibit heightened anticancer efficacy and reduced side effects. Moreover, significant progress has been made in the synthesis of circRNA with biological functions in vitro, which increases the clinical value of circRNA with low expression levels but high anti-cancer activity. However, artificially synthesizing circRNAs still faces the problems of standardization and immunogenicity [203]. In addition, the size of circRNA molecules may affect their efficiency in loading into exosome. More and more evidence show that the nanoparticle-based circRNA delivery system could also be a highly valuable therapeutic strategy. Research shows that a lipid nanoparticle system for delivering circRNA can activate the adaptive immune response and demonstrate excellent anti-tumor effects in various murine maligancy models, providing novel prospects for the advancement of tumor circRNA vaccines.

Excitingly, in addition to the mentioned techniques of exosome engineering and artificial synthesis of circRNA, 3D bioprinting has gained extensive attention in recent years and demonstrated tremendous potential in the field of tumor research due to its significant advantages in closely simulating in vivo conditions. Research has shown that 3D bioprinting can simulate various tumor microenvironments [204,205,206,207], thereby providing strong support for studying the cancer biology and enhancing medical intervention of tumors. As an important component of the tumor microenvironment, exosomes play a crucial role in the intercellular communication between various cells within the tumor microenvironment. Clearly elucidating the role and precise molecular mechanisms by which exosomes mediate intercellular communication in tumor microenvironment holds great potential for both treating cancer and achieving personalized therapy. It is worth noting that exosomes will receive significant technical support in exploring their biological role in tumor progression through 3D bioprinting. Most essentially, 3D bioprinting provides high submicron scale resolution and precise spatiotemporal control, which can be utilized to incorporate and direct the gradient release of exosomes, including engineered and natural exosomes. Furthermore, screening for synthetic circRNA with ideal biological functions is crucial for its clinical translation. Compared to traditional 2D cell culture-based screening, 3D bioprinting will provide a more physiologically relevant environment for circRNA screening and overcome many limitations of in vivo experiments. Therefore, 3D bioprinting will inevitably promote the clinical translation of exosome and circRNA. In the future, people will witness the clinical applications of 3D bioprinting in facilitating the targeted gradient release of engineered exosomes carrying artificially synthesized circRNAs to exert biological functions in specific diseases. Moreover, hydrogels are confirmed to possess substantial adjustability, biocompatibility, degradability, and physical characteristics akin to tissues. The synergistic combination of exosomes and hydrogels promotes the efficiency of exosomes’ activities, which holds enormous potential for future application.

Intriguingly, the research on plant or food exosomes in drug delivery has gained widespread attention due to their advantages of a lower cost, broader availability, and easy accessibility [208, 209]. Exosome-like nanovesicles generated from Yam possess a capability to promote the biogenesis of osteoblasts and protect against osteoporosis [210]. Bovine milk-derived exosomes show cross-species tolerance in drug delivery without adverse immune and inflammatory responses [208]. Nevertheless, natural exosomes exhibit certain limitations, such as their heterogeneity, which may diminish their anti-cancer efficacy and even promote tumor progression.

Future studies delving into exosomal circRNAs across various biological settings and diseases such as cancer will continue to unravel the enigma surrounding exosomal circRNAs and benefit humankind.

Data availability

All data generated or analyzed during this study are included in this article.

Abbreviations

NSCLC:

Non-small cell lung cancer

circRNA:

Circular RNA

EMT:

Epithelial-to-mesenchymal transition

LC:

Lung cancer

LCCDEs:

Lung cancer cell-derived exosomes

EIciRNAs:

Exon-intron circRNAs

EcircRNAs:

Exonic circRNAs

CiRNAs:

Circular intronic RNAs

BP:

Branching point

RBPs:

RNA binding proteins

ESRP1:

Epithelial splicing regulatory protein1

A5SS:

Alternative 5’ splicing site

m6A:

N6-methyladenosine

PAK2:

P21-activated kinase 2

EGFR-TKIs:

Epidermal growth factor receptor tyrosine kinase inhibitors

ddPCR:

Droplet digital PCR

References

  1. Herbst RS, Morgensztern D, Boshoff C. The biology and management of non-small cell lung cancer. Nature. 2018;553(7689):446–54.

    Article  CAS  PubMed  Google Scholar 

  2. Remon J, Soria JC, Peters S. Early and locally advanced non-small-cell lung cancer: an update of the ESMO clinical practice guidelines focusing on diagnosis, staging, systemic and local therapy. Ann Oncol. 2021;32(12):1637–42.

    Article  CAS  PubMed  Google Scholar 

  3. di Noia V, D’Argento E, Pilotto S, Grizzi G, Caccese M, Iacovelli R, et al. Necitumumab in the treatment of non-small-cell lung cancer: clinical controversies. Expert Opin Biol Ther. 2018;18(9):937–45.

    Article  PubMed  Google Scholar 

  4. Skribek M, Rounis K, Tsakonas G, Ekman S. Complications following novel therapies for non-small cell lung cancer. J Intern Med. 2022;291(6):732–54.

    Article  PubMed  Google Scholar 

  5. Zhao L, Wang H, Fu J, Wu X, Liang XY, Liu XY, et al. Microfluidic-based exosome isolation and highly sensitive aptamer exosome membrane protein detection for lung cancer diagnosis. Biosens Bioelectron. 2022;214:114487.

    Article  CAS  PubMed  Google Scholar 

  6. Kalluri R. The biology and function of exosomes in cancer. J Clin Invest. 2016;126(4):1208–15.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Meldolesi J. Exosomes and ectosomes in intercellular communication. Curr Biol. 2018;28(8):R435–44.

    Article  CAS  PubMed  Google Scholar 

  8. Paskeh MDA, Entezari M, Mirzaei S, Zabolian A, Saleki H, Naghdi MJ, et al. Emerging role of exosomes in cancer progression and tumor microenvironment remodeling. J Hematol Oncol. 2022;15(1):83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Kimiz-Gebologlu I, Oncel SS, Exosomes. Large-scale production, isolation, drug loading efficiency, and biodistribution and uptake. J Control Release. 2022;347:533–43.

    Article  CAS  PubMed  Google Scholar 

  10. Mashouri L, Yousefi H, Aref AR, Ahadi AM, Molaei F, Alahari SK. Exosomes: composition, biogenesis, and mechanisms in cancer metastasis and drug resistance. Mol Cancer. 2019;18(1):75.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Zhou Z, Wang R, Wang J, Hao Y, Xie Q, Wang L, et al. Melatonin pretreatment on exosomes: heterogeneity, therapeutic effects, and usage. Front Immunol. 2022;13:933736.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Wang H, Zhu Y, Tang C, Zhou Z, Wang Z, Li Z, et al. Reassessment of the proteomic composition and function of extracellular vesicles in the seminal plasma. Endocrinology. 2022;163(1):bqab214.

    Article  PubMed  Google Scholar 

  13. van Niel G, D’Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol. 2018;19(4):213–28.

    Article  PubMed  Google Scholar 

  14. Lin Z, Ji Y, Zhou J, Li G, Wu Y, Liu W, et al. Exosomal circRNAs in cancer: implications for therapy resistance and biomarkers. Cancer Lett. 2023;566:216245.

    Article  CAS  PubMed  Google Scholar 

  15. Yang B, Chen Y, Shi J. Exosome biochemistry and advanced nanotechnology for Next-Generation theranostic platforms. Adv Mater. 2019;31(2):e1802896.

    Article  PubMed  Google Scholar 

  16. Bastos N, Ruivo CF, da Silva S, Melo SA. Exosomes in cancer: use them or target them? Semin Cell Dev Biol. 2018;78:13–21.

    Article  CAS  PubMed  Google Scholar 

  17. Zhou Y, Zhang Y, Gong H, Luo S, Cui Y. The role of exosomes and their applications in cancer. Int J Mol Sci. 2021;22(22):12204.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Huang J, Shen M, Yan M, Cui Y, Gao Z, Meng X. Exosome-mediated transfer of miR-1290 promotes cell proliferation and invasion in gastric cancer via < italic > NKD1. ABBS. 2019;51(9):900–7.

    Article  CAS  PubMed  Google Scholar 

  19. Li Y, Feng W, Kong M, Liu R, Wu A, Shen L, et al. Exosomal circRNAs: A new star in cancer. Life Sci. 2021;269:119039.

    Article  CAS  PubMed  Google Scholar 

  20. Wang S, Dong Y, Gong A, Kong H, Gao J, Hao X, et al. Exosomal circRNAs as novel cancer biomarkers: challenges and opportunities. Int J Biol Sci. 2021;17(2):562–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Kristensen LS, Andersen MS, Stagsted LVW, Ebbesen KK, Hansen TB, Kjems J. The biogenesis, biology and characterization of circular RNAs. Nat Rev Genet. 2019;20(11):675–91.

    Article  CAS  PubMed  Google Scholar 

  22. Lei M, Zheng G, Ning Q, Zheng J, Dong D. Translation and functional roles of circular RNAs in human cancer. Mol Cancer. 2020;19(1):30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Yang L, Wilusz JE, Chen LL. Biogenesis and regulatory roles of circular RNAs. Annu Rev Cell Dev Biol. 2022;38:263–89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Chen LL. The expanding regulatory mechanisms and cellular functions of circular RNAs. Nat Rev Mol Cell Biol. 2020;21(8):475–90.

    Article  CAS  PubMed  Google Scholar 

  25. Yuan W, Zhang X, Cong H. Advances in the protein–encoding functions of circular RNAs associated with cancer (Review). Oncol Rep. 2023;50(2):160.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Memczak S, Jens M, Elefsinioti A, Torti F, Krueger J, Rybak A, et al. Circular RNAs are a large class of animal RNAs with regulatory potency. Nature. 2013;495(7441):333–8.

    Article  CAS  PubMed  Google Scholar 

  27. Li J, Sun D, Pu W, Wang J, Peng Y. Circular RNAs in cancer: biogenesis, function, and clinical significance. Trends Cancer. 2020;6(4):319–36.

    Article  CAS  PubMed  Google Scholar 

  28. Kristensen LS, Jakobsen T, Hager H, Kjems J. The emerging roles of circRNAs in cancer and oncology. Nat Rev Clin Oncol. 2022;19(3):188–206.

    Article  CAS  PubMed  Google Scholar 

  29. Yu T, Wang Y, Fan Y, Fang N, Wang T, Xu T, et al. CircRNAs in cancer metabolism: a review. J Hematol Oncol. 2019;12(1):90.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Cao YZ, Sun JY, Chen YX, Wen CC, Wei L. The roles of circRNAs in cancers: perspectives from molecular functions. Gene. 2021;767:145182.

    Article  CAS  PubMed  Google Scholar 

  31. Li S, Zhao Y, Chen X. Microarray expression profile analysis of circular RNAs and their potential regulatory role in bladder carcinoma. Oncol Rep. 2021;45(1):239–53.

    Article  PubMed  Google Scholar 

  32. Zhang N, Nan A, Chen L, Li X, Jia Y, Qiu M, et al. Circular RNA circSATB2 promotes progression of non-small cell lung cancer cells. Mol Cancer. 2020;19(1):101.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Zhou Q, Xie D, Wang R, Liu L, Yu Y, Tang X, et al. The emerging landscape of Exosomal circRNAs in solid cancers and hematological malignancies. Biomark Res. 2022;10(1):28.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Zhang Y, Liu Q, Zhang X, Huang H, Tang S, Chai Y, et al. Recent advances in exosome-mediated nucleic acid delivery for cancer therapy. J Nanobiotechnol. 2022;20(1):279.

    Article  CAS  Google Scholar 

  35. Zhang F, Jiang J, Qian H, Yan Y, Xu W. Exosomal circRNA: emerging insights into cancer progression and clinical application potential. J Hematol Oncol. 2023;16(1):67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Liu Q, Li S. Exosomal circRNAs: novel biomarkers and therapeutic targets for urinary tumors. Cancer Lett. 2024;588:216759.

    Article  CAS  PubMed  Google Scholar 

  37. Wu X, Shi M, Lian Y, Zhang H. Exosomal circRNAs as promising liquid biopsy biomarkers for glioma. Front Immunol. 2023;14:1039084.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Xu Y, Han J, Zhang X, Zhang X, Song J, Gao Z, et al. Exosomal circRNAs in Gastrointestinal cancer: role in occurrence, development, diagnosis and clinical application (Review). Oncol Rep. 2024;51(2):19.

    Article  CAS  PubMed  Google Scholar 

  39. Wang D, Li R, Jiang J, Qian H, Xu W. Exosomal circRNAs: novel biomarkers and therapeutic targets for Gastrointestinal tumors. Biomed Pharmacother. 2023;157:114053.

    Article  CAS  PubMed  Google Scholar 

  40. Marangon D, Lecca D. Exosomal non-coding RNAs in glioma progression: insights into tumor microenvironment dynamics and therapeutic implications. Front Cell Dev Biol. 2023;11:1275755.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Sun W, Jiang C, Liu Q, Wang N, Huang R, Jiang G, et al. Exosomal noncoding RNAs: decoding their role in thyroid cancer progression. Front Endocrinol (Lausanne). 2024;15:1337226.

    Article  PubMed  Google Scholar 

  42. Zeng Y, Hu S, Luo Y, He K. Exosome cargos as biomarkers for diagnosis and prognosis of hepatocellular carcinoma. Pharmaceutics. 2023;15(9):2365.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Wang R, Wang S, Jiang H, Lan Y, Yu S. Prospects for the clinical application of Exosomal circular RNA in squamous cell carcinoma. Front Oncol. 2024;14:1430684.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Han M, Zhang M, Qi M, Zhou Y, Li F, Fang S. Regulatory mechanism and promising clinical application of Exosomal circular RNA in gastric cancer. Front Oncol. 2023;13:1236679.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Gopikrishnan M, Ashour RHCRG, Pintus HM, Hammad G. Therapeutic and diagnostic applications of Exosomal circRNAs in breast cancer. Funct Integr Genomics. 2023;23(2):184.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Pegtel DM, Gould SJ, Exosomes. Annu Rev Biochem. 2019;88:487–514.

  48. Zhang L, Yu D. Exosomes in cancer development, metastasis, and immunity. Biochim Biophys Acta Rev Cancer. 2019;1871(2):455–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Jeppesen DK, Fenix AM, Franklin JL, Higginbotham JN, Zhang Q, Zimmerman LJ, et al. Reassessment Exosome Composition Cell. 2019;177(2):428–e44518.

  50. Li X, Li C, Zhang L, Wu M, Cao K, Jiang F, et al. The significance of exosomes in the development and treatment of hepatocellular carcinoma. Mol Cancer. 2020;19(1):1.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Ghafourian M, Mahdavi R, Akbari Jonoush Z, Sadeghi M, Ghadiri N, Farzaneh M, et al. The implications of exosomes in pregnancy: emerging as new diagnostic markers and therapeutics targets. Cell Commun Signal. 2022;20(1):51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Gao X, Ran N, Dong X, Zuo B, Yang R, Zhou Q, et al. Anchor peptide captures, targets, and loads exosomes of diverse origins for diagnostics and therapy. Sci Transl Med. 2018;10(444):eaat0195.

    Article  PubMed  Google Scholar 

  53. Min L, Shen J, Tu C, Hornicek F, Duan Z. The roles and implications of exosomes in sarcoma. Cancer Metastasis Rev. 2016;35(3):377–90.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Xu Z, Zeng S, Gong Z, Yan Y. Exosome-based immunotherapy: a promising approach for cancer treatment. Mol Cancer. 2020;19(1):160.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Yu D, Li Y, Wang M, Gu J, Xu W, Cai H, et al. Exosomes as a new frontier of cancer liquid biopsy. Mol Cancer. 2022;21(1):56.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Barile L, Vassalli G, Exosomes. Therapy delivery tools and biomarkers of diseases. Pharmacol Ther. 2017;174:63–78.

  57. Xiao Y, Zhong J, Zhong B, Huang J, Jiang L, Jiang Y, et al. Exosomes as potential sources of biomarkers in colorectal cancer. Cancer Lett. 2020;476:13–22.

    Article  CAS  PubMed  Google Scholar 

  58. Lei LM, Li FXZ, Lin X, Xu F, Shan SK, Guo B, et al. Cold exposure-induced plasma exosomes impair bone mass by inhibiting autophagy. J Nanobiotechnol. 2024;22(1):361.

    Article  CAS  Google Scholar 

  59. Mathieu M, Martin-Jaular L, Lavieu G, Théry C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat Cell Biol. 2019;21(1):9–17.

    Article  CAS  PubMed  Google Scholar 

  60. Isaac R, Reis FCG, Ying W, Olefsky JM. Exosomes as mediators of intercellular crosstalk in metabolism. Cell Metab. 2021;33(9):1744–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Colombo M, Raposo G, Théry C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu Rev Cell Dev Biol. 2014;30:255–89.

    Article  CAS  PubMed  Google Scholar 

  62. Wang Y, Liu J, Ma J, Sun T, Zhou Q, Wang W, et al. Exosomal circRNAs: biogenesis, effect and application in human diseases. Mol Cancer. 2019;18(1):116.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Wu Y, Li J, Zeng Y, Pu W, Mu X, Sun K, et al. Exosomes rewire the cartilage microenvironment in osteoarthritis: from intercellular communication to therapeutic strategies. Int J Oral Sci. 2022;14(1):40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Salunkhe S, Dheeraj null, Basak M, Chitkara D, Mittal A. Surface functionalization of exosomes for target-specific delivery and in vivo imaging & tracking: strategies and significance. J Control Release. 2020;326:599–614.

    Article  CAS  PubMed  Google Scholar 

  65. Gonda A, Kabagwira J, Senthil GN, Wall NR. Internalization of exosomes through Receptor-Mediated endocytosis. Mol Cancer Res. 2019;17(2):337–47.

    Article  CAS  PubMed  Google Scholar 

  66. Iyaswamy A, Thakur A, Guan XJ, Krishnamoorthi S, Fung TY, Lu K, et al. Fe65-engineered neuronal exosomes encapsulating corynoxine-B ameliorate cognition and pathology of Alzheimer’s disease. Signal Transduct Target Ther. 2023;8(1):404.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Milane L, Singh A, Mattheolabakis G, Suresh M, Amiji MM. Exosome mediated communication within the tumor microenvironment. J Control Release. 2015;219:278–94.

    Article  CAS  PubMed  Google Scholar 

  68. Xu J, Wu M, Yang J, Zhao D, He D, Liu Y et al. Multimodal smart systems reprogramme macrophages and remove urate to treat gouty arthritis. Nat Nanotechnol. 2024.

  69. Rahbarghazi R, Jabbari N, Sani NA, Asghari R, Salimi L, Kalashani SA, et al. Tumor-derived extracellular vesicles: reliable tools for cancer diagnosis and clinical applications. Cell Commun Signal. 2019;17(1):73.

    Article  PubMed  PubMed Central  Google Scholar 

  70. Xia Y, Zhen L, Li H, Wang S, Chen S, Wang C, et al. MIRLET7BHG promotes hepatocellular carcinoma progression by activating hepatic stellate cells through Exosomal SMO to trigger Hedgehog pathway. Cell Death Dis. 2021;12(4):326.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Lin Z, Wu Y, Xu Y, Li G, Li Z, Liu T. Mesenchymal stem cell-derived exosomes in cancer therapy resistance: recent advances and therapeutic potential. Mol Cancer. 2022;21(1):179.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Wortzel I, Dror S, Kenific CM, Lyden D. Exosome-Mediated metastasis: communication from a distance. Dev Cell. 2019;49(3):347–60.

    Article  CAS  PubMed  Google Scholar 

  73. Zhou CF, Ma J, Huang L, Yi HY, Zhang YM, Wu XG, et al. Cervical squamous cell carcinoma-secreted Exosomal miR-221-3p promotes lymphangiogenesis and lymphatic metastasis by targeting VASH1. Oncogene. 2019;38(8):1256–68.

    Article  CAS  PubMed  Google Scholar 

  74. Huang M, Huang X, Huang N. Exosomal circGSE1 promotes immune escape of hepatocellular carcinoma by inducing the expansion of regulatory T cells. Cancer Sci. 2022;113(6):1968–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Hu Z, Chen G, Zhao Y, Gao H, Li L, Yin Y, et al. Exosome-derived circCCAR1 promotes CD8 + T-cell dysfunction and anti-PD1 resistance in hepatocellular carcinoma. Mol Cancer. 2023;22(1):55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Lu JC, Zhang PF, Huang XY, Guo XJ, Gao C, Zeng HY, et al. Amplification of spatially isolated adenosine pathway by tumor–macrophage interaction induces anti-PD1 resistance in hepatocellular carcinoma. J Hematol Oncol. 2021;14:200.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Wu S, Luo M, To KKW, Zhang J, Su C, Zhang H, et al. Intercellular transfer of Exosomal wild type EGFR triggers osimertinib resistance in non-small cell lung cancer. Mol Cancer. 2021;20:17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Zhang X, Shi H, Yuan X, Jiang P, Qian H, Xu W. Tumor-derived exosomes induce N2 polarization of neutrophils to promote gastric cancer cell migration. Mol Cancer. 2018;17(1):146.

    Article  PubMed  PubMed Central  Google Scholar 

  79. Tan Y, Huang Y, Mei R, Mao F, Yang D, Liu J, et al. HucMSC-derived exosomes delivered BECN1 induces ferroptosis of hepatic stellate cells via regulating the xCT/GPX4 axis. Cell Death Dis. 2022;13(4):319.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Cai X, Zhang ZY, Yuan JT, Ocansey DKW, Tu Q, Zhang X, et al. hucMSC-derived exosomes attenuate colitis by regulating macrophage pyroptosis via the miR-378a-5p/NLRP3 axis. Stem Cell Res Ther. 2021;12(1):416.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Li C, Teixeira AF, Zhu HJ, Ten Dijke P. Cancer associated-fibroblast-derived exosomes in cancer progression. Mol Cancer. 2021;20(1):154.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Fanini F, Fabbri M. Cancer-derived exosomic MicroRNAs shape the immune system within the tumor microenvironment: state of the Art. Semin Cell Dev Biol. 2017;67:23–8.

    Article  CAS  PubMed  Google Scholar 

  83. Chen Y, Zhang X, Yang J, Feng W, Deng G, Xu S, et al. Extracellular vesicles derived from Selenium-Deficient MAC-T cells aggravated inflammation and apoptosis by triggering the Endoplasmic reticulum (ER) Stress/PI3K-AKT-mTOR pathway in bovine mammary epithelial cells. Antioxid (Basel). 2023;12(12):2077.

    Article  CAS  Google Scholar 

  84. Shi M, Zhu J, Wang R, Chen X, Mi L, Walz T, et al. Latent TGF-β structure and activation. Nature. 2011;474(7351):343–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Ostrowski M, Carmo NB, Krumeich S, Fanget I, Raposo G, Savina A, et al. Rab27a and Rab27b control different steps of the exosome secretion pathway. Nat Cell Biol. 2010;12(1):19–30. sup pp 1-13.

    Article  CAS  PubMed  Google Scholar 

  86. Zhang Y, Yi D, Hong Q, Cao J, Geng X, Liu J, et al. Platelet-rich plasma-derived exosomes boost mesenchymal stem cells to promote peripheral nerve regeneration. J Controlled Release. 2024;367:265–82.

    Article  CAS  Google Scholar 

  87. Batrakova EV, Kim MS. Using exosomes, naturally-equipped nanocarriers, for drug delivery. J Controlled Release. 2015;219:396–405.

    Article  CAS  Google Scholar 

  88. Dad HA, Gu TW, Zhu AQ, Huang LQ, Peng LH. Plant Exosome-like nanovesicles: emerging therapeutics and drug delivery nanoplatforms. Mol Ther. 2021;29(1):13–31.

    Article  CAS  PubMed  Google Scholar 

  89. Donoso-Quezada J, Ayala-Mar S, González-Valdez J. State-of-the-art exosome loading and functionalization techniques for enhanced therapeutics: a review. Crit Rev Biotechnol. 2020;40(6):804–20.

    Article  CAS  PubMed  Google Scholar 

  90. Hade MD, Suire CN, Suo Z. Mesenchymal stem Cell-Derived exosomes: applications in regenerative medicine. Cells. 2021;10(8):1959.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Zhuang W, Liu C, Hong Y, Zheng Y, Huang M, Tang H, et al. Tumor-suppressive miR-4732-3p is sorted into fucosylated exosome by HnRNPK to avoid the Inhibition of lung cancer progression. J Exp Clin Cancer Res. 2024;43(1):123.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Mu W, Gu P, Li H, Zhou J, Jian Y, Jia W, et al. Exposure of benzo[a]pyrene induces HCC exosome-circular RNA to activate lung fibroblasts and trigger organotropic metastasis. Cancer Commun (Lond). 2024;44(7):718–38.

    Article  PubMed  Google Scholar 

  93. Li MY, Liu LZ, Dong M. Progress on pivotal role and application of exosome in lung cancer carcinogenesis, diagnosis, therapy and prognosis. Mol Cancer. 2021;20(1):22.

    Article  PubMed  PubMed Central  Google Scholar 

  94. Liang Y, Duan L, Lu J, Xia J. Engineering exosomes for targeted drug delivery. Theranostics. 2021;11(7):3183–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. 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 Nanobiotechnol. 2020;18(1):10.

    Article  CAS  Google Scholar 

  96. Hatibaruah A, Rahman M, Agarwala S, Singh SA, Shi J, Gupta S, et al. Circular RNAs in cancer and diabetes. J Genet. 2021;100:21.

    Article  CAS  PubMed  Google Scholar 

  97. Zhang S, Long F, Lin H, Wang X, Jiang G, Wang T. Regulatory roles of phytochemicals on circular RNAs in cancer and other chronic diseases. Pharmacol Res. 2021;174:105936.

    Article  CAS  PubMed  Google Scholar 

  98. Almouh M, Razmara E, Bitaraf A, Ghazimoradi MH, Hassan ZM, Babashah S. Circular RNAs play roles in regulatory networks of cell signaling pathways in human cancers. Life Sci. 2022;309:120975.

    Article  CAS  PubMed  Google Scholar 

  99. Li Z, Huang C, Bao C, Chen L, Lin M, Wang X, et al. Exon-intron circular RNAs regulate transcription in the nucleus. Nat Struct Mol Biol. 2015;22(3):256–64.

    Article  PubMed  Google Scholar 

  100. Patop IL, Wüst S, Kadener S. Past, present, and future of circRNAs. EMBO J. 2019;38(16):e100836.

    Article  PubMed  PubMed Central  Google Scholar 

  101. Zhang Y, Zhang XO, Chen T, Xiang JF, Yin QF, Xing YH, et al. Circular intronic long noncoding RNAs. Mol Cell. 2013;51(6):792–806.

    Article  CAS  PubMed  Google Scholar 

  102. Montañés-Agudo P, van der Made I, Aufiero S, Tijsen AJ, Pinto YM, Creemers EE. Quaking regulates circular RNA production in cardiomyocytes. J Cell Sci. 2023;136(13):jcs261120.

    Article  PubMed  PubMed Central  Google Scholar 

  103. Hentze MW, Castello A, Schwarzl T, Preiss T. A brave new world of RNA-binding proteins. Nat Rev Mol Cell Biol. 2018;19(5):327–41.

    Article  CAS  PubMed  Google Scholar 

  104. He AT, Liu J, Li F, Yang BB. Targeting circular RNAs as a therapeutic approach: current strategies and challenges. Signal Transduct Target Ther. 2021;6(1):185.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Liu D, Dredge BK, Bert AG, Pillman KA, Toubia J, Guo W, et al. ESRP1 controls biogenesis and function of a large abundant multiexon circRNA. Nucleic Acids Res. 2024;52(3):1387–403.

    Article  CAS  PubMed  Google Scholar 

  106. Tang C, Xie Y, Yu T, Liu N, Wang Z, Woolsey RJ, et al. m6A-dependent biogenesis of circular RNAs in male germ cells. Cell Res. 2020;30(3):211–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Knupp D, Cooper DA, Saito Y, Darnell RB, Miura P. NOVA2 regulates neural circRNA biogenesis. Nucleic Acids Res. 2021;49(12):6849–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Liang D, Tatomer DC, Luo Z, Wu H, Yang L, Chen LL, et al. The output of Protein-Coding genes shifts to circular RNAs when the Pre-mRNA processing machinery is limiting. Mol Cell. 2017;68(5):940–e9543.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Fu P, Cai Z, Zhang Z, Meng X, Peng Y. An updated database of virus circular RNAs provides new insights into the biogenesis mechanism of the molecule. Emerg Microbes Infect. 2023;12(2):2261558.

    Article  PubMed  PubMed Central  Google Scholar 

  110. Chen L, Shan G. CircRNA in cancer: fundamental mechanism and clinical potential. Cancer Lett. 2021;505:49–57.

    Article  CAS  PubMed  Google Scholar 

  111. Guo JU, Agarwal V, Guo H, Bartel DP. Expanded identification and characterization of mammalian circular RNAs. Genome Biol. 2014;15(7):409.

    Article  PubMed  PubMed Central  Google Scholar 

  112. Piccinni MP, Raghupathy R, Saito S, Szekeres-Bartho J. Cytokines, hormones and cellular regulatory mechanisms favoring successful reproduction. Front Immunol. 2021;12:717808.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Heo JI, Ryu J. Exosomal noncoding RNA: A potential therapy for retinal vascular diseases. Mol Ther Nucleic Acids. 2024;35(1):102128.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Liu B, Song F, Yang Q, Zhou Y, Shao C, Shen Y, et al. Characterization of tissue-specific biomarkers with the expression of circRNAs in forensically relevant body fluids. Int J Legal Med. 2019;133(5):1321–31.

    Article  PubMed  Google Scholar 

  115. Liu B, Yang Q, Meng H, Shao C, Jiang J, Xu H, et al. Development of a multiplex system for the identification of forensically relevant body fluids. Forensic Sci Int Genet. 2020;47:102312.

    Article  CAS  PubMed  Google Scholar 

  116. Zhang Y, Liu B, Shao C, Xu H, Xue A, Zhao Z, et al. Evaluation of the inclusion of circular RNAs in mRNA profiling in forensic body fluid identification. Int J Legal Med. 2018;132(1):43–52.

    Article  PubMed  Google Scholar 

  117. Yang Q, Liu B, Zhou Y, Yao Y, Zhou Z, Li H, et al. Evaluation of one-step RT-PCR multiplex assay for body fluid identification. Int J Legal Med. 2021;135(5):1727–35.

    Article  PubMed  Google Scholar 

  118. Li Y, Ye J, Xu S, Wang J. Circulating noncoding RNAs: promising biomarkers in liquid biopsy for the diagnosis, prognosis, and therapy of NSCLC. Discov Oncol. 2023;14(1):142.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Roy S, Kanda M, Nomura S, Zhu Z, Toiyama Y, Taketomi A, et al. Diagnostic efficacy of circular RNAs as noninvasive, liquid biopsy biomarkers for early detection of gastric cancer. Mol Cancer. 2022;21(1):42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Lin Z, Tang X, Wan J, Zhang X, Liu C, Liu T. Functions and mechanisms of circular RNAs in regulating stem cell differentiation. RNA Biol. 2021;18(12):2136–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Liu CX, Chen LL. Circular RNAs: characterization, cellular roles, and applications. Cell. 2022;185(13):2390.

    Article  CAS  PubMed  Google Scholar 

  122. Zhou WY, Cai ZR, Liu J, Wang DS, Ju HQ, Xu RH. Circular RNA: metabolism, functions and interactions with proteins. Mol Cancer. 2020;19(1):172.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Chen R, Wang SK, Belk JA, Amaya L, Li Z, Cardenas A, et al. Engineering circular RNA for enhanced protein production. Nat Biotechnol. 2023;41(2):262–72.

    Article  CAS  PubMed  Google Scholar 

  124. Zhang Y, Jiang J, Zhang J, Shen H, Wang M, Guo Z, et al. CircDIDO1 inhibits gastric cancer progression by encoding a novel DIDO1-529aa protein and regulating PRDX2 protein stability. Mol Cancer. 2021;20(1):101.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Wang M, Ding X, Fang X, Xu J, Chen Y, Qian Y, et al. Circ6834 suppresses non-small cell lung cancer progression by destabilizing ANHAK and regulating miR-873-5p/TXNIP axis. Mol Cancer. 2024;23(1):128.

    Article  PubMed  PubMed Central  Google Scholar 

  126. Zhao Q, Cai D, Xu H, Gao Y, Zhang R, Zhou X, et al. o8G-modified circPLCE1 inhibits lung cancer progression via chaperone-mediated autophagy. Mol Cancer. 2025;24(1):82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Chen L, Wang Y, Lin J, Song Z, Wang Q, Zhao W, et al. Exportin 4 depletion leads to nuclear accumulation of a subset of circular RNAs. Nat Commun. 2022;13(1):5769.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Liu S, Wang Z, Su Y, Qi L, Yang W, Fu M, et al. A neuroanatomical basis for electroacupuncture to drive the vagal-adrenal axis. Nature. 2021;598(7882):641–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Su X, Feng Y, Chen R, Duan S. CircR-loop: a novel RNA:DNA interaction on genome instability. Cell Mol Biol Lett. 2024;29(1):89.

    Article  PubMed  PubMed Central  Google Scholar 

  130. Wilusz JE. A 360° view of circular RNAs: from biogenesis to functions. Wiley Interdiscip Rev RNA. 2018;9(4):e1478.

    Article  PubMed  PubMed Central  Google Scholar 

  131. Xu S, Xu Y, Solek NC, Chen J, Gong F, Varley AJ, et al. Tumor-Tailored ionizable lipid nanoparticles facilitate IL-12 circular RNA delivery for enhanced lung cancer immunotherapy. Adv Mater. 2024;36(29):e2400307.

    Article  PubMed  Google Scholar 

  132. Liu CX, Li X, Nan F, Jiang S, Gao X, Guo SK, et al. Structure and degradation of circular RNAs regulate PKR activation in innate immunity. Cell. 2019;177(4):865–e88021.

    Article  CAS  PubMed  Google Scholar 

  133. Jiang M, Bai H, Fang S, Zhou C, Shen W, Gong Z. CircLIFRSA/miR-1305/PTEN axis attenuates malignant cellular processes in non-small cell lung cancer by regulating AKT phosphorylation. Mol Cancer. 2024;23(1):208.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Liu CX, Yang L, Chen LL. Dynamic conformation: marching toward circular RNA function and application. Mol Cell. 2024;84(19):3596–609.

    Article  CAS  PubMed  Google Scholar 

  135. Li Y, Zheng Q, Bao C, Li S, Guo W, Zhao J, et al. Circular RNA is enriched and stable in exosomes: a promising biomarker for cancer diagnosis. Cell Res. 2015;25(8):981–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Huang A, Zheng H, Wu Z, Chen M, Huang Y. Circular RNA-protein interactions: functions, mechanisms, and identification. Theranostics. 2020;10(8):3503–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Wang S, Sun Z, Lei Z, Zhang HT. RNA-binding proteins and cancer metastasis. Semin Cancer Biol. 2022;86(Pt 2):748–68.

    Article  CAS  PubMed  Google Scholar 

  138. Munro TP, Magee RJ, Kidd GJ, Carson JH, Barbarese E, Smith LM, et al. Mutational analysis of a heterogeneous nuclear ribonucleoprotein A2 response element for RNA trafficking. J Biol Chem. 1999;274(48):34389–95.

    Article  CAS  PubMed  Google Scholar 

  139. Villarroya-Beltri C, Gutiérrez-Vázquez C, Sánchez-Cabo F, Pérez-Hernández D, Vázquez J, Martin-Cofreces N, et al. Sumoylated hnRNPA2B1 controls the sorting of MiRNAs into exosomes through binding to specific motifs. Nat Commun. 2013;4:2980.

    Article  PubMed  Google Scholar 

  140. Qu L, Ding J, Chen C, Wu ZJ, Liu B, Gao Y, et al. Exosome-Transmitted LncARSR promotes Sunitinib resistance in renal cancer by acting as a competing endogenous RNA. Cancer Cell. 2016;29(5):653–68.

    Article  CAS  PubMed  Google Scholar 

  141. 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 

  142. Pan Z, Zhao R, Li B, Qi Y, Qiu W, Guo Q, et al. EWSR1-induced circNEIL3 promotes glioma progression and exosome-mediated macrophage immunosuppressive polarization via stabilizing IGF2BP3. Mol Cancer. 2022;21(1):16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Wei Y, Fu J, Zhang H, Ling Y, Tang X, Liu S, et al. N6-methyladenosine modification promotes hepatocarcinogenesis through circ-CDYL-enriched and EpCAM-positive liver tumor-initiating exosomes. iScience. 2023;26(10):108022.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Shi H, Huang S, Qin M, Xue X, Guo X, Jiang L, et al. Exosomal circ_0088300 derived from Cancer-Associated fibroblasts acts as a miR-1305 sponge and promotes gastric carcinoma cell tumorigenesis. Front Cell Dev Biol. 2021;9:676319.

    Article  PubMed  PubMed Central  Google Scholar 

  145. Dou Y, Cha DJ, Franklin JL, Higginbotham JN, Jeppesen DK, Weaver AM, et al. Circular RNAs are down-regulated in KRAS mutant colon cancer cells and can be transferred to exosomes. Sci Rep. 2016;6(1):37982.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Zhang J, Zhang X, Li C, Yue L, Ding N, Riordan T, et al. Circular RNA profiling provides insights into their subcellular distribution and molecular characteristics in HepG2 cells. RNA Biol. 2019;16(2):220–32.

    Article  PubMed  PubMed Central  Google Scholar 

  147. Misir S, Wu N, Yang BB. Specific expression and functions of circular RNAs. Cell Death Differ. 2022;29(3):481–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Hu X, Qin H, Yan Y, Wu W, Gong S, Wang L, et al. Exosomal circular RNAs: biogenesis, effect, and application in cardiovascular diseases. Front Cell Dev Biol. 2022;10:948256.

    Article  PubMed  PubMed Central  Google Scholar 

  149. Hansen TB, Jensen TI, Clausen BH, Bramsen JB, Finsen B, Damgaard CK, et al. Natural RNA circles function as efficient MicroRNA sponges. Nature. 2013;495(7441):384–8.

    Article  CAS  PubMed  Google Scholar 

  150. Hansen TB, Wiklund ED, Bramsen JB, Villadsen SB, Statham AL, Clark SJ, et al. miRNA-dependent gene Silencing involving Ago2-mediated cleavage of a circular antisense RNA. EMBO J. 2011;30(21):4414–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Lee Y, Choe J, Park OH, Kim YK. Molecular mechanisms driving mRNA degradation by m6A modification. Trends Genet. 2020;36(3):177–88.

    Article  CAS  PubMed  Google Scholar 

  152. Zaccara S, Ries RJ, Jaffrey SR. Reading, writing and erasing mRNA methylation. Nat Rev Mol Cell Biol. 2019;20(10):608–24.

    Article  CAS  PubMed  Google Scholar 

  153. Wang X, Wu R, Liu Y, Zhao Y, Bi Z, Yao Y, et al. m6A mRNA methylation controls autophagy and adipogenesis by targeting Atg5 and Atg7. Autophagy. 2020;16(7):1221–35.

    Article  CAS  PubMed  Google Scholar 

  154. Zhang L, Hou C, Chen C, Guo Y, Yuan W, Yin D, et al. The role of N6-methyladenosine (m6A) modification in the regulation of circRNAs. Mol Cancer. 2020;19(1):105.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Chen RX, Chen X, Xia LP, Zhang JX, Pan ZZ, Ma XD, et al. N6-methyladenosine modification of circNSUN2 facilitates cytoplasmic export and stabilizes HMGA2 to promote colorectal liver metastasis. Nat Commun. 2019;10(1):4695.

    Article  PubMed  PubMed Central  Google Scholar 

  156. Wen SY, Qadir J, Yang BB. Circular RNA translation: novel protein isoforms and clinical significance. Trends Mol Med. 2022;28(5):405–20.

    Article  CAS  PubMed  Google Scholar 

  157. Fischer JW, Leung AKL. CircRNAs: a regulator of cellular stress. Crit Rev Biochem Mol Biol. 2017;52(2):220–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Zhang W, Zhou B, Yang X, Zhao J, Hu J, Ding Y, et al. Exosomal circEZH2_005, an intestinal injury biomarker, alleviates intestinal ischemia/reperfusion injury by mediating Gprc5a signaling. Nat Commun. 2023;14(1):5437.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Wen N, Peng D, Xiong X, Liu G, Nie G, Wang Y, et al. Cholangiocarcinoma combined with biliary obstruction: an Exosomal circRNA signature for diagnosis and early recurrence monitoring. Signal Transduct Target Ther. 2024;9(1):107.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Wang H, Tang Z, Duan J, Zhou C, Xu K, Mu H. Cancer-released Exosomal circular RNA circ_0008717 promotes cell tumorigenicity through microRNA-1287-5p/P21-activated kinase 2 (PAK2) axis in non-small cell lung cancer. Bioengineered. 2022;13(4):8937–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Chen T, Liu Y, Li C, Xu C, Ding C, Chen J, et al. Tumor-derived Exosomal circfarsa mediates M2 macrophage polarization via the PTEN/PI3K/AKT pathway to promote non-small cell lung cancer metastasis. Cancer Treat Res Commun. 2021;28:100412.

    Article  PubMed  Google Scholar 

  162. Chen W, Tang D, Lin J, Huang X, Lin S, Shen G, et al. Exosomal circSHKBP1 participates in non-small cell lung cancer progression through PKM2-mediated Glycolysis. Mol Therapy - Oncolytics. 2022;24:470–85.

    Article  CAS  Google Scholar 

  163. Fang K, Chen X, Qiu F, Xu J, Xiong H, Zhang Z. Serum-Derived Exosomes-Mediated circular RNA ARHGAP10 modulates the progression of Non-Small cell lung cancer through the miR-638/FAM83F axis. Cancer Biotherapy Radiopharmaceuticals. 2022;37(2):96–110.

    Article  CAS  PubMed  Google Scholar 

  164. Li C, Wang G, Ma X, Tao T, Li Q, Yang Y, et al. Upregulation of Exosomal circPLK1 promotes the development of non-small cell lung cancer through the miR-1294/ high mobility group protein A1 axis. Bioengineered. 2022;13(2):4185–200.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Ma J, Qi G, Li L. A novel serum Exosomes-Based biomarker Hsa_circ_0002130 facilitates Osimertinib-Resistance in Non-Small cell lung cancer by sponging miR-498. OTT. 2020;13:5293–307.

    Article  CAS  Google Scholar 

  166. Ning Z, Tian Y, Li Y, Zhao X, Zhang J, Wang C, et al. Exosomal circ_0007385 enhances non-small cell lung cancer cell proliferation and stemness via regulating miR-1253/FAM83A axis. Anticancer Drugs. 2022;33(1):61–74.

    Article  CAS  PubMed  Google Scholar 

  167. Peng L, Cao Y, Hu C. Identification of Exosomal circRNA CD226 as a potent driver of nonsmall cell lung cancer through miR-1224-3p/high mobility group AT-hook 2 axis. Anticancer Drugs. 2022;33(10):1126–38.

    Article  CAS  PubMed  Google Scholar 

  168. Shao N, Song L, Sun X. Exosomal circ_PIP5K1A regulates the progression of non-small cell lung cancer and cisplatin sensitivity by miR-101/ABCC1 axis. Mol Cell Biochem. 2021;476(6):2253–67.

    Article  CAS  PubMed  Google Scholar 

  169. Tong Z, Wang Z, Jiang J, Tong C, Wu L. A novel molecular mechanism mediated by circ CCDC134 regulates non-small cell lung cancer progression. Thorac Cancer. 2023;14(20):1958–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Wang W, Sun H, Ma X, Zhu T, Zhang H. Circ_0002476 regulates cell growth, invasion, and MtDNA damage in non–small cell lung cancer by targeting MiR -1182/ TFAM axis. Thorac Cancer. 2022;13(20):2867–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Peng X, Zhao L, Yao L, Dong J, Wu W, Luo T. Exosomal ERBB2IP contributes to tumor growth via elevating PSAT1 expression in non-small cell lung carcinoma. Thorac Cancer. 2023;14(19):1812–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Shi Q, Ji T, Ma Z, Tan Q, Liang J. Serum Exosomes-Based biomarker circ_0008928 regulates cisplatin sensitivity, tumor progression, and Glycolysis metabolism by miR-488/ HK2 axis in cisplatin-Resistant nonsmall cell lung carcinoma. Cancer Biotherapy Radiopharmaceuticals. 2023;38(8):558–71.

    Article  CAS  PubMed  Google Scholar 

  173. Wang D, Wang S, Jin M, Zuo Y, Wang J, Niu Y, et al. Hypoxic Exosomal circPLEKHM1-Mediated crosstalk between tumor cells and macrophages drives lung cancer metastasis. Adv Sci (Weinh). 2024;11(22):e2309857.

    Article  PubMed  Google Scholar 

  174. Liu Q, Xu P, Jin M, Wang L, Hu F, Yang Q, et al. CircFTO from M2 macrophage-derived small extracellular vesicles (sEV) enhances NSCLC malignancy by regulation miR-148a-3pPDK4 axis. Cancer Immunol Immunother. 2024;73(5):91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Zhang Y, Luo J, Yang W, Ye WC. CircRNAs in colorectal cancer: potential biomarkers and therapeutic targets. Cell Death Dis. 2023;14(6):353.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Lin J, Wang X, Zhai S, Shi M, Peng C, Deng X, et al. Hypoxia-induced Exosomal circPDK1 promotes pancreatic cancer Glycolysis via c-myc activation by modulating miR-628-3p/BPTF axis and degrading BIN1. J Hematol Oncol. 2022;15(1):128.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Ding C, Xi G, Wang G, Cui D, Zhang B, Wang H, et al. Exosomal Circ-MEMO1 promotes the progression and aerobic Glycolysis of Non-small cell lung cancer through targeting MiR-101-3p/KRAS axis. Front Genet. 2020;11:962.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Zhang Q, Wang W, Zhou Q, Chen C, Yuan W, Liu J, et al. Roles of circRNAs in the tumour microenvironment. Mol Cancer. 2020;19(1):14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Chen SW, Zhu SQ, Pei X, Qiu BQ, Xiong D, Long X, et al. Cancer cell-derived Exosomal circUSP7 induces CD8 + T cell dysfunction and anti-PD1 resistance by regulating the miR-934/SHP2 axis in NSCLC. Mol Cancer. 2021;20(1):144.

    Article  PubMed  PubMed Central  Google Scholar 

  180. Guo X, Gao C, Yang DH, Li S. Exosomal circular RNAs: A chief culprit in cancer chemotherapy resistance. Drug Resist Updat. 2023;67:100937.

    Article  CAS  PubMed  Google Scholar 

  181. Xu X, Tao R, Sun L, Ji X. Exosome-transferred Hsa_circ_0014235 promotes DDP chemoresistance and deteriorates the development of non-small cell lung cancer by mediating the miR-520a-5p/CDK4 pathway. Cancer Cell Int. 2020;20(1):552.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Xie H, Yao J, Wang Y, Ni B. Exosome-transmitted circVMP1 facilitates the progression and cisplatin resistance of non-small cell lung cancer by targeting miR-524-5p-METTL3/SOX2 axis. Drug Delivery. 2022;29(1):1257–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Wang X, Wang H, Jiang H, Qiao L, Guo C. Circular RNAcirc_0076305 promotes cisplatin (DDP) resistance of Non-Small cell lung cancer cells by regulating ABCC1 through miR-186-5p. Cancer Biotherapy Radiopharmaceuticals. 2023;38(5):293–304.

    Article  CAS  PubMed  Google Scholar 

  184. Wei SL, Ye JJ, Sun L, Hu L, Wei YY, Zhang DW, et al. Exosome-derived circKIF20B suppresses gefitinib resistance and cell proliferation in non-small cell lung cancer. Cancer Cell Int. 2023;23(1):129.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Yang B, Teng F, Chang L, Wang J, Liu DL, Cui YS, et al. Tumor-derived Exosomal CircRNA_102481 contributes to EGFR-TKIs resistance via the miR-30a-5p/ROR1 axis in non-small cell lung cancer. Aging. 2021;13(9):13264–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Zhang C, Cao J, Lv W, Mou H. CircRNA_100395 carried by exosomes from Adipose-Derived mesenchymal stem cells inhibits the malignant transformation of Non-Small cell lung carcinoma through the miR-141-3p-LATS2 axis. Front Cell Dev Biol. 2021;9:663147.

    Article  PubMed  PubMed Central  Google Scholar 

  187. Liu S, Wu X, Wang Y, Chen Y. Exosomal circ_0000735 contributes to non-small lung cancer malignant progression. J Biochem Mol Tox. 2024;38(4):e23700.

    Article  CAS  Google Scholar 

  188. Chen Y, Lou C, Ma X, Zhou C, Zhao X, Li N, et al. Serum Exosomal Hsa_circ_0069313 has a potential to diagnose more aggressive non-small cell lung cancer. Clin Biochem. 2022;102:56–64.

    Article  CAS  PubMed  Google Scholar 

  189. He Y, Liu Y, Cha N, Gao Y, Li F, Zhang M, et al. Exosomal circ_0048856 derived from non-small cell lung cancer contributes to aggressive cancer progression through downregulation of miR-1287–5p. Pathol - Res Pract. 2022;232:153659.

    Article  CAS  PubMed  Google Scholar 

  190. Xian J, Su W, Liu L, Rao B, Lin M, Feng Y, et al. Identification of three circular RNA cargoes in serum exosomes as diagnostic biomarkers of Non–Small-Cell lung cancer in the Chinese population. J Mol Diagn. 2020;22(8):1096–108.

    Article  CAS  PubMed  Google Scholar 

  191. Chen Z, Ma X, Chen Z, Chen W, Li L, Lin Y, et al. Exosome-transported circ_0061407 and circ_0008103 play a tumour-repressive role and show diagnostic value in non-small-cell lung cancer. J Transl Med. 2024;22(1):427.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Xu YX, Pu SD, Li X, Yu ZW, Zhang YT, Tong XW, et al. Exosomal NcRNAs: novel therapeutic target and biomarker for diabetic complications. Pharmacol Res. 2022;178:106135.

    Article  CAS  PubMed  Google Scholar 

  193. Hong W, Du K, Zhang Q, Ren Z, Gao X. Tanreqing suppresses the proliferation and migration of non-small cell lung cancer cells by mediating the inactivation of the HIF1α signaling pathway via Exosomal circ-WDR78. J Biomol Struct Dynamics. 2024;1–12.

  194. Seo K, Yoo JH, Kim J, Min SJ, Heo DN, Kwon IK, et al. Ginseng-derived exosome-like nanovesicles extracted by sucrose gradient ultracentrifugation to inhibit osteoclast differentiation. Nanoscale. 2023;15(12):5798–808.

    Article  CAS  PubMed  Google Scholar 

  195. Chen Q, Zu M, Gong H, Ma Y, Sun J, Ran S, et al. Tea leaf-derived exosome-like nanotherapeutics retard breast tumor growth by pro-apoptosis and microbiota modulation. J Nanobiotechnol. 2023;21(1):6.

    Article  CAS  Google Scholar 

  196. Hu X, Wu D, He X, Zhao H, He Z, Lin J, et al. circGSK3β promotes metastasis in esophageal squamous cell carcinoma by augmenting β-catenin signaling. Mol Cancer. 2019;18(1):160.

    Article  PubMed  PubMed Central  Google Scholar 

  197. Cheng L, Yang F, Zhao Y, Liu Z, Yao X, Zhang J. Tetrahedron supported CRISPR/Cas13a cleavage for electrochemical detection of circular RNA in bladder cancer. Biosens Bioelectron. 2023;222:114982.

    Article  CAS  PubMed  Google Scholar 

  198. Dong J, Zeng Z, Sun R, Zhang X, Cheng Z, Chen C, et al. Specific and sensitive detection of circRNA based on netlike hybridization chain reaction. Biosens Bioelectron. 2021;192:113508.

    Article  CAS  PubMed  Google Scholar 

  199. Zhang X, Zhang H, Gu J, Zhang J, Shi H, Qian H, et al. Engineered extracellular vesicles for cancer therapy. Adv Mater. 2021;33(14):e2005709.

    Article  PubMed  Google Scholar 

  200. Zhang J, Ji C, Zhang H, Shi H, Mao F, Qian H, et al. Engineered neutrophil-derived exosome-like vesicles for targeted cancer therapy. Sci Adv. 2022;8(2):eabj8207.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Zhang M, Hu S, Liu L, Dang P, Liu Y, Sun Z, et al. Engineered exosomes from different sources for cancer-targeted therapy. Signal Transduct Target Ther. 2023;8(1):124.

    Article  PubMed  PubMed Central  Google Scholar 

  202. Xu M, Feng T, Liu B, Qiu F, Xu Y, Zhao Y, et al. Engineered exosomes: desirable target-tracking characteristics for cerebrovascular and neurodegenerative disease therapies. Theranostics. 2021;11(18):8926–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Chen YG, Chen R, Ahmad S, Verma R, Kasturi SP, Amaya L, et al. N6-Methyladenosine modification controls circular RNA immunity. Mol Cell. 2019;76(1):96–e1099.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Li Y, Liu J, Xu S, Wang J. 3D bioprinting: an important tool for tumor microenvironment research. Int J Nanomed. 2023;18:8039–57.

    Article  CAS  Google Scholar 

  205. Wang X, Luo Y, Ma Y, Wang P, Yao R. Converging Bioprinting and organoids to better recapitulate the tumor microenvironment. Trends Biotechnol. 2024;42(5):648–63.

    Article  CAS  PubMed  Google Scholar 

  206. Li J, Parra-Cantu C, Wang Z, Zhang YS. Improving bioprinted volumetric tumor microenvironments in vitro. Trends Cancer. 2020;6(9):745–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  207. Zhou Z, Pang Y, Ji J, He J, Liu T, Ouyang L, et al. Harnessing 3D in vitro systems to model immune responses to solid tumours: a step towards improving and creating personalized immunotherapies. Nat Rev Immunol. 2024;24(1):18–32.

    Article  CAS  PubMed  Google Scholar 

  208. Munagala R, Aqil F, Jeyabalan J, Gupta RC. Bovine milk-derived exosomes for drug delivery. Cancer Lett. 2016;371(1):48–61.

    Article  CAS  PubMed  Google Scholar 

  209. Cao M, Diao N, Cai X, Chen X, Xiao Y, Guo C, et al. Plant exosome nanovesicles (PENs): green delivery platforms. Mater Horiz. 2023;10(10):3879–94.

    Article  CAS  PubMed  Google Scholar 

  210. Hwang JH, Park YS, Kim HS, Kim DH, Lee SH, Lee CH, et al. Yam-derived exosome-like nanovesicles stimulate osteoblast formation and prevent osteoporosis in mice. J Control Release. 2023;355:184–98.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank Wenlong Xue for the support in reading and revising the manuscript.

Funding

This work was supported by grants from the National Natural Science Foundation of China (82272766, 82204837), the Shanghai Municipal Health Commission-Excellence Project (20234Z0001), the Shanghai Municipal Health Commission (Grant no. 20224Y0128), and the Shanghai Sailing Program (No. 22YF1448800).

Author information

Authors and Affiliations

Authors

Contributions

Hongyuan Yin: conceptualization, formal analysis, and writing the original draft. Jiayi Shi: validation, project administration, and writing the original draft. Shaoling Li: validation, project administration, and writing the original draft. Qianhui You: validation and project administration. Huici Zhu: investigation. Chinying Koo: investigation. Baonian Liu: writing - review and editing. Likun Hou: writing - review and editing. Chunyan Wu: writing - review, editing, and conceptualization. All authors contributed to the article and approved the submitted version.

Corresponding authors

Correspondence to Baonian Liu, Likun Hou or Chunyan Wu.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

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-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, 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 you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. 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-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yin, H., Shi, J., Li, S. et al. Emerging roles of exosomal circRNAs in non-small cell lung cancer. J Transl Med 23, 490 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-025-06463-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-025-06463-w

Keywords