Published online Oct 24, 2025. doi: 10.5306/wjco.v16.i10.111115
Revised: July 24, 2025
Accepted: September 11, 2025
Published online: October 24, 2025
Processing time: 122 Days and 24 Hours
Non-coding RNAs, which do not encode proteins, significantly influence signal regulation. Circular RNAs (circRNAs), produced through a post-splicing mec
Core Tip: Circular RNAs (circRNAs) represents a distinct class of non-coding RNAs characterized by a closed circular conformation, wherein the terminal ends are covalently bonded, resulting in a unique structure devoid of 5' cap and 3' poly(A) tail. This structural feature imparts enhanced stability to circRNAs compared to linear RNAs. CircRNAs are implicated in a myriad of physiological processes, including cellular proliferation, differentiation, apoptosis, and immune responses. This review delineates the recent advancements in circRNAs research concerning digestive system tumors, emphasizing their pronounced influence not only on the regulation of gene expression but also on the mechanisms of chemoresistance, immune modulation and treatment.
- Citation: Xie LF, Liu SY, Dang XW. Recent advances of circular RNAs in gastrointestinal cancer. World J Clin Oncol 2025; 16(10): 111115
- URL: https://www.wjgnet.com/2218-4333/full/v16/i10/111115.htm
- DOI: https://dx.doi.org/10.5306/wjco.v16.i10.111115
Tumors of the digestive system rank among the main causes of cancer-related mortality globally, primarily encompassing esophageal, gastric, liver, pancreatic, and colorectal cancers. These tumors develop through various molecular me
Furthermore, circRNAs are also closely related to drug resistance in digestive system tumors. Researchers believe that circRNAs affect tumor cells' sensitivity and resistance to chemotherapy by regulating drug targets and related signaling pathways[7]. Liu et al[8] prove that in pancreatic cancer, circRNA hsa_circ_0014784 promotes tumor cell proliferation and metastasis by regulating the miR-214-3p/YAP1 signaling pathway. Although research on circRNAs in digestive system tumors has made some progress, further exploration of their specific biological functions and mechanisms is still needed. Future research should prioritize elucidating the role of circRNAs in the tumor microenvironment, their mutual effect with other non-coding RNAs, and clinical applications[9,10]. A rounded exploration of the roles and underlying mechanisms of circRNAs may yield novel strategies and insights for therapeutic interventions and prognostic evaluation of tumors within the digestive system. In summary, circRNAs, recognized as a promising therapeutic target, hold significant potential in the research about digestive system tumors and warrant increased focus in forthcoming investigations.
CircRNAs represent a distinctive category of non-coding RNAs, identifiable by their closed circular configuration formed via back-splicing. Compared to linear RNAs, circRNAs have significant advantages in terms of stability, expression specificity, and evolutionary conservation. The structure of circRNAs makes them resistant to degradation by nucleases, thereby positioning them as a viable marker and therapeutic target.
The biosynthesis of circRNAs is mainly generated through the back-splicing process, involving specific splicing factors and regulatory elements. Its formation mechanisms include exon circularization and intron circularization. The classification of circRNAs according to their origin can be divided into three distinct categories: Exon circRNAs (ecircRNAs), intron circRNAs (ciRNAs), and exon-intron circRNAs (EIciRNAs)[11]. Of these, ciRNAs and EIciRNAs are predominantly nuclear in their function, and further research is required to elucidate their mechanisms. In contrast, ecircRNAs primarily exert regulatory effects in the cytoplasm and can be secreted as exosomes into bodily fluids, thus making them the current focus of research. The degradation of circRNAs in cells is relatively slow and mainly relies on specific RNA-binding proteins (RBPs) and endogenous nucleases. Studies have shown that the stability and expression of circRNAs are influenced by various factors, including the regulation of transcription factors and the selection of splicing factors[4]. Typically, circRNAs modulate the downstream genes by functioning as a sponge for miRNA, whereas long non-coding RNAs may influence gene transcription via interaction with transcription factors[12,13]. The functional mechanisms of circRNAs are diverse, mainly including acting as miRNA sponges, regulating the activity of RNA-binding proteins, participating in transcriptional regulation, and encoding short peptides. Through the binding of miRNAs, circRNAs can mitigate the inhibitory influence of miRNA on target genes.
CircRNAs operate as regulators of cellular activity, principally through four discussed mechanisms (Figure 1). Firstly, they function as miRNA sponges. An increasing body of evidence substantiates that circRNAs possess miRNA binding sites, enabling them to obstruct the binding of miRNA to target mRNAs[10,14]. Secondly, circRNAs interact with RBPs. Certain circRNAs can serve as decoys, influencing gene expression by binding to or dissociating proteins, while others can act as protein scaffolds to facilitate protein-protein interactions[5,15]. Thirdly, circRNAs are implicated in the regulation of gene transcription. A majority of studies indicate that circRNAs may participate in the transcriptional regulation of their parental genes, primarily through their accumulation in the transcriptional regions of these genes, thus enhancing the activity of polymerase II (Pol II) to modulate transcription[16]. Fourthly, accumulating data suggest that circRNAs can be translated into peptides or proteins[17].
Increasing research manifests that circRNAs exhibit aberrant expression profiles in gastrointestinal cancer (Table 1).
| Type of cancer | circRNAs | Expression | miRNAs, RBPs, peptides, pathways | Function | Ref. |
| Esophageal cancer | circTMEM45A | Up | U2AF2, NLRP3/caspase 1/IL-1β | Promotes malignant progression and inflammatory progression | [52] |
| Esophageal cancer | circPRKCA | Up | YBX1/CSF2 | Promotes migration, invasion, and angiogenesis | [53] |
| Esophageal cancer | circNF1 | Up | ANXA1, JAK/STAT3 | Increases proliferation, metastasis, and tumor evasion | [54] |
| Esophageal cancer | circMMP11 | Up | miR-671-5p | Increases proliferation, migration, and invasion | [55] |
| Esophageal cancer | hsa_circ_0087104 | Down | miR-542-3p/PIK3R1 | Inhibits metastasis | [56] |
| Esophageal cancer | hsa_circ_0001165 | Up | EIF4A3, miR-381-3p/TNS3 | Promotes proliferation, invasion, and migration | [57] |
| Esophageal cancer | circJPH1 | Up | NF-κB/HERC5, XRCC6 | Promotes proliferation, invasion, and migration | [58] |
| Esophageal cancer | hsa_circ_0001615 | Down | miR-142-5p/β-catenin | Inhibits proliferation, migration, and invasion, promotes apoptosis | [59] |
| Esophageal cancer | circPDE5A | Down | PI3K/AKT | Inhibits proliferation and metastasis | [60] |
| Esophageal cancer | circSSPO | Up | miR-6820-5p/KLK8/PKD1 | Promotes tumorigenesis and metastasis | [61] |
| Esophageal cancer | circUBE4B | Up | MAPK/ERK | Promotes proliferation | [62] |
| Esophageal cancer | circ-TNRC6B | Down | miR-452-5p/DAG1 | Inhibits proliferation and invasion | [63] |
| Gastric cancer | circSCAF8 | Up | miR-1293/TIMP1 | Promotes proliferation, invasion, and migration | [64] |
| Gastric cancer | circPFKP | Down | miR-346/CAMD3 | Inhibits proliferation, invasion, and migration | [65] |
| Gastric cancer | hsa_circ_0079226 | Up | miR-155-5p/FOXK1/AKT | Promotes proliferation, invasion, and migration | [66] |
| Gastric cancer | hsa_circ_0008126 | Down | EIF4A3, APC/β-Catenin | Inhibits progression and metastasis | [67] |
| Gastric cancer | circATP8A1 | Up | miR-1-3p/STAT6 | Promotes proliferation and invasion | [68] |
| Gastric cancer | circBIRC6 | Up | miR-488/GRIN2D | Promotes malignant progression | [69] |
| Gastric cancer | circPAK2 | Up | IGF2BPs/VEGFA | Promotes lymph node metastasis | [70] |
| Gastric cancer | hsa_circ_0136666 | Up | miR-375/PRKDC, PD-L1 | Promotes progression and tumor immune escape | [71] |
| Gastric cancer | circSTAU2 | Down | miR-589/CAPZA1 | Inhibits proliferation, invasion, and migration | [72] |
| Gastric cancer | circGAPVD1 | Down | miR-4424/STK4, GAPVD1-137aa | Inhibits tumor progression, encodes GAPVD1-137aa | [73] |
| Gastric cancer | circZNF131 | Down | ZNF131-354aa | Encodes ZNF131-354aa, inhibits tumor progression | [24] |
| Gastric cancer | hsa_circ_0002301 | Down | HECTD1-463aa | Encodes HECTD1-463aa, inhibits ferroptosis | [74] |
| Gastric cancer | circ0003692 | Down | FNDC3B-267aa | Encodes FNDC3B-267aa, inhibits metastasis | [75] |
| Gastric cancer | circPGD | Up | miR-16-5p/ABL2, PGD-219aa | Promotes tumor progression, encodes PGD-219aa | [25] |
| Liver cancer | circFADS1 | Up | EIF4A3, Wnt/β-Catenin | Promotes proliferation and inhibits apoptosis | [76] |
| Liver cancer | circDCUN1D4 | Down | miR-590-5p/ TIMP3 | Suppresses proliferation, migration, and invasion | [77] |
| Liver cancer | circTTC13 | Up | miR-513a-5p/SLC7A11 | Promotes sorafenib resistance | [78] |
| Liver cancer | circPIAS1 | Up | miR-455-3p/NUPR1/FTH1 | Promotes tumor progression | [79] |
| Liver cancer | circPTPN12 | Down | ESRP1, PDLIM2/ NF-κB | Suppresses proliferation, poor prognosis | [80] |
| Liver cancer | hsa_circ_0044539 | Up | miR-29a-3p/VEGFA | Promotes lymph node metastasis | [81] |
| Liver cancer | circPHKB | Up | miR-1234-3p/CYP2W1 | Promotes sorafenib resistance | [82] |
| Liver cancer | circUSP10 | Up | miR-211-5p/TCF12/EMT | Promotes tumor progression | [83] |
| Liver cancer | circMRCKα | Up | circMRCKα-227aa | Encodes circMRCKα-227aa, promotes glycolysis and progression | [84] |
| Liver cancer | circSTX6 | Up | HNRNPD/ATF3, circSTX6-144aa | Encodes circSTX6-144aa, promotes tumor progression | [85] |
| Liver cancer | circMAP3K4 | Up | circMAP3K4-455aa | Encodes circMAP3K4-455aa, inhibits apoptosis | [28] |
| Pancreatic cancer | circPHF14 | Up | EIF4E, Wnt/β-catenin | Promotes growth and metastasis | [86] |
| Pancreatic cancer | circCEACAM5 | Up | METTL3, DKC1 | Promotes proliferation, invasion, and migration, inhibits apoptosis | [87] |
| Pancreatic cancer | hsa_circ_0004781 | Up | miR-9-5p/KLF5, miR-338-3p/ADAM17 | Promotes proliferation and migration | [88] |
| Pancreatic cancer | circCGNL1 | Down | NUDT4/HDAC4/RUNX2/GAMT | Inhibits proliferation, promoting apoptosis | [89] |
| Pancreatic cancer | circRPS29 | Up | miR-770-5p/TRIM29, MEK/ERK | Promotes gemcitabine resistance | [90] |
| Pancreatic cancer | circDUSP22 | Down | miR-1178-3p/BNIP3 | Restrains cancer development | [91] |
| Pancreatic cancer | circSTK39 | Up | miR-140-3p/TRAM2 | Promotes proliferation and migration | [92] |
| Colorectal cancer | hsa_circ_0009022 | Down | miR-576-5p, FMRP | Inhibits proliferation and migration | [93] |
| Colorectal cancer | circMVP | Up | METTL3/CTNNB1/β-catenin | Increases proliferation, invasion, and tumorigenesis | [94] |
| Colorectal cancer | circEIF3I | Up | miR-328-3p/NCAPH | Promotes metastasis | [95] |
| Colorectal cancer | circMYBL2 | Down | p185 protein | Suppresses cancer progression, encodes p185 protein | [96] |
| Colorectal cancer | hsa_circ_0000467 | Up | eIF4A3/c-Myc | Promotes growth and metastasis | [97] |
| Colorectal cancer | hsa_circ_0017065 | Up | miR-3174/RBFOX2 | Promotes proliferation and metastasis | [98] |
| Colorectal cancer | circSTIL | Up | miR-431/SLC7A11 | Inhibits ferroptosis | [99] |
| Colorectal cancer | circCAPRIN1 | Up | STAT2/ACC1 | Promotes proliferation, migration, and EMT | [100] |
| Colorectal cancer | circINSIG1 | Up | circINSIG1-121aa | Encodes circINSIG1-121aa, promotes proliferation and metastasis | [101] |
| Colorectal cancer | circATG4B | Up | circATG4B-222aa | Encodes circATG4B-222aa, promotes autophagy and oxaliplatin resistance | [32] |
Esophageal cancer is a highly malignant and invasive digestive tract tumor with high incidence and mortality rates worldwide. Increasing research has demonstrated that circRNAs play a pivotal role in the pathology of this condition. Research by Lin et al[18] has indicated that circRNA TCFL5 is markedly elevated in esophageal cancer, facilitating tumor cell growth and migration through the regulation of M2 macrophage polarization. Furthermore, circRNA-0008717 has been shown to interact with miR-203, influencing the expression of Slug and thereby impacting the multiplication and migratory capabilities of esophageal cancer cells[19].
Recent years have witnessed a surge in research focusing on circRNAs in gastric cancer, revealing that numerous circRNAs are intricately related to the onset and progression of this disease. For instance, circRNA hsa_circ_0008035 enhances the growth and invasive properties of gastric cancer cells by sponging miR-375[20]. Moreover, circRNA circSMC3 exhibits upregulation in gastric cancer tissues, with its heightened expression correlating with unfavorable prognostic outcomes for patients[21]. Studies have also found that circRNA DLG1 is upregulated in anti-programmed cell death protein-1 treatment, possibly related to the immune evasion mechanism of gastric cancer[22]. Furthermore, proteins encoded by circRNAs also play an important role in tumorigenesis and progression, particularly in the regulation of key processes such as cell proliferation, migration, and apoptosis[23]. The research by Wu et al[24] indicates that circZNF131 encodes the peptide ZNF131-aa, which has been shown to suppress the progression of gastric cancer. In contrast, the research by Liu et al[25] found that the peptide PGD-219aa encoded by circPGD promotes tumor progression. These insights offer novel avenues for early diagnostic strategies and targeted therapeutic interventions in gastric cancer.
The role of circRNAs in liver cancer has gradually gained attention. Research indicates that circRNA_10156 is upregulated in hepatitis B virus-associated liver cancer, influencing the proliferation and migratory behavior of liver cancer cells through the modulation of miR-149-3p expression[26]. Additionally, circRNA KIF5B exerts its regulatory effects on miR-192 via a sponge mechanism, thereby impacting the growth and metastatic potential of liver cancer cells[27]. Duan et al’s research indicates that circMAP3K4 can encode the small peptide circMAP3K4-455aa[28], which may promote tumor progression by inhibiting apoptosis in liver cancer cells. These studies reveal the biological functions of circRNAs in liver cancer.
Pancreatic cancer is characterized by a notably poor prognosis, and the role of circRNA in its pathogenesis has increasingly come into focus. Investigations have revealed that circRNA circHIPK3 is highly expressed in pancreatic cancer cells, promoting tumor cell proliferation and migration by regulating miR-637 and FASN[29]. Additionally, circRNA-0087502 enhances the resistance of pancreatic cancer cells to chemotherapy drugs by regulating the miR-1179/TGFBR2 pathway[27].
Colorectal cancer is one of the most common malignant tumors in the digestive system, and the role of circRNAs in its development is increasingly emphasized. Research findings indicate that hsa_circ_0000190 inhibits the proliferation and migration of colorectal cancer cells by modulating the expression levels of miR-1252 and PAK3[30]. Additionally, circDENND4C promotes glycolysis and supports the proliferation of colorectal cancer cells through the regulation of GLUT1[31]. Pan et al[32] consider that circATG4B mediates autophagy and oxaliplatin resistance in colorectal cancer through the encoded protein circATG4B-222aa, thereby influencing tumor progression. These studies provide a novel idea and direction for its targeted therapy of colorectal cancer.
In the treatment of digestive system tumors, chemotherapy remains one of the main treatment methods. However, the occurrence of chemotherapy resistance often leads to treatment failure, becoming a major obstacle to patient survival. Recent investigations have increasingly highlighted the pivotal role of circRNAs in mediating resistance to chemotherapy in tumors. CircRNAs modulate the sensitivity and resistance of tumor cells to chemotherapeutic agents through multiple mechanisms. Primarily, circRNAs sponge with miRNAs, inhibiting the latter’s regulation of target genes through miRNA binding. An illustrative example is circRNA_0014784, which enhances the growth and metastasis of pancreatic cancer by combining with miR-214-3p and modulating the expression of YAP1[8]. This regulation of miRNAs not only impacts tumor growth but may also significantly contribute to the development of resistance to chemotherapy. Moreover, circRNAs can affect the efficacy of chemotherapy by influencing drug metabolism and transport mechanisms. For instance, circRNAs may influence the concentration of drugs within tumor cells, thereby altering the cells' sensitivity to chemotherapy drugs. Evidence suggests that circRNAs can bolster the resistance of tumor cells to chemotherapeutic agents by regulating the expression of drug efflux pumps[33]. Additionally, circRNAs may further promote the deve
Research focusing on circRNAs in gastrointestinal tumors reveals their potential as both a biomarker and a therapeutic target. For instance, circRNA_0001649 has been linked to chemotherapy resistance in colorectal cancer, with its elevated expression correlating with a poor prognosis in affected patients[34]. Furthermore, circRNAs may influence tumor chemotherapy resistance through the regulation of immune responses within the tumor microenvironment. By modu
CircRNAs have a stable structure and specific tissue expression, capable of influencing tumor development through various mechanisms. Emerging evidence indicates that circRNAs play a vital regulatory role in the immune microenvironment of digestive system tumors, affecting the functions of immune cells and tumor immune evasion.
CircRNAs has the capacity to diminish the extent to which specific miRNAs suppress their target genes through direct binding, subsequently facilitating the growth and metastasis of tumor cells[2]. In digestive system tumors, various circRNAs have emerged as significant contributors to immune evasion by tumors, as they can modulate tumor cell interactions with the immune system via the regulation of immune checkpoint gene expression. A notable example is circCPA4, which has been identified as a promoter of programmed death ligand 1 (PD-L1) expression, thereby aiding the immune evasion capabilities of tumor cells[36]. Furthermore, circRNAs also influence the tumor microenvironment by modulating immune cell functions. CircRNAs can influence the anti-cancer immune response by adjusting the exhaustion state of T cells[37]. In digestive system tumors, circRNAs correlate with the degree of immune infiltration, suggesting that they may serve as important regulatory factors in the tumor immune microenvironment. Moreover, the application prospects of circRNAs in tumor immunotherapy have also garnered attention. Due to the stability and specificity of circRNAs, they may become new immunotherapy targets. For instance, the development of circRNA vaccines provides new ideas for tumor immunotherapy. Researchers are exploring how to utilize circRNAs as vaccine components to activate specific immune responses, thereby enhancing the efficacy of immunotherapy[4].
However, the specific mechanisms of circRNAs in immune regulation in digestive system tumors still require further research. We should focus on the localization, transport, and degradation mechanisms of circRNAs in the tumor microenvironment, as well as the specific pathways of their interactions with immune cells. Accomplishing this will provide fresh perspectives and strategies for the diagnosis and therapeutic management of digestive system tumors. In summary, circRNAs are an important area of research for understanding immune regulation in digestive system tumors, and future studies are expected to reveal their potential applications in tumor immunotherapy.
CircRNAs not only play a significant role in tumor occurrence and development but are also considered a potential biomarker for early diagnosis and prognostic assessment. The expression levels of circRNAs are also significantly correlated with clinical pathological features of tumors. For instance, elevated levels of hsa_circ_0001649 and hsa_circ_0001178 in gastric cancer and colorectal cancer have been correlated with unfavorable prognoses[1]. The abnormal expression not only reflects the biological characteristics of tumors but may also become an important indicator for assessing patient prognosis in clinical settings. The secretion of circRNAs into body fluids occurs in the form of exosomes, a characteristic that suggests the potential of exosomal circRNAs as an effective biomarker for tumor diagnosis and prognosis. For instance, in the context of esophageal cancer, studies have demonstrated that exosomal circ_0000337 fosters resistance to cisplatin by modulating the miR-377-3p and JAK2 signaling pathways[38]. Due to its stability in plasma and tissues, circRNAs can serve as a biomarker for non-invasive detection. More research indicates that the expression levels of specific circRNAs in plasma can effectively distinguish healthy individuals from patients with digestive system tumors, providing new ideas for early diagnosis[4]. Additionally, combining the expression profiles of multiple circRNAs can further enhance diagnostic accuracy; using multiple circRNAs for gastric cancer diagnosis can demonstrate high sensitivity and specificity[39].
In terms of prognostic assessment, the expression quantity of circRNAs are closely related to patient survival rates. Research indicates that specific oncogenic circRNAs, such as hsa_circ_0002917, correlate with unfavorable survival rates in individuals diagnosed with gastrointestinal malignancies, whereas elevated levels of tumor-suppressive circRNAs may be associated with improved prognoses[2,3]. This differential expression pattern provides clinicians with new prognostic assessment tools, aiding in the formulation of individualized treatment plans. In summary, circRNAs as a biomarker for digestive system tumors have broad application prospects. They can be used not only for early diagnosis and prognostic assessment of tumors but may also become new therapeutic targets.
Multiple studies have shown that circRNAs can regulate tumor phenotype through various mechanisms. For instance, circRNAs can sponge with miRNA, inhibiting miRNA's suppressive effects on target genes[2]. Treatment strategies targeting circRNAs mainly include using small molecule drugs, antibodies, or RNA interference techniques to inhibit the expression of specific circRNAs, thereby restoring the function of tumor suppressor genes or enhancing circRNA expression to suppress tumor progression[3]. Research has found that circRNA_0000074 acts as a tumor suppressor in gastric cancer, targeting and inhibiting the proliferation and invasion abilities of tumor cells[4]. Thus, circRNA-targeted treatment strategies present novel perspectives for managing gastrointestinal tumors.
Due to their stability and biocompatibility, circRNAs can serve as ideal carriers for drug delivery. Relevant studies have found that circRNAs can effectively encapsulate drug molecules and enter target cells through cell membranes, thereby improving the bioavailability and targeting of drugs[1]. For instance, using lipid nanoparticles to combine circRNAs with anti-cancer drugs can significantly enhance drug accumulation in tumor cells and improve anti-tumor effects[9]. Add
The tumor microenvironment is pivotal in the initiation and progression of tumors. CircRNAs influence tumor adv
CircRNA vaccines, as an emerging immunotherapy, show promising application prospects. CircRNA vaccines can effectively activate the immune system, inducing specific T cell responses, thereby enhancing immune surveillance against tumors[41]. For instance, circRNA vaccines targeting gastric cancer have shown good anti-tumor effects in mouse models, significantly inhibiting tumor growth and metastasis[42]. Moreover, the stability and long-lasting effects of circRNA vaccines make them powerful tools for cancer immunotherapy, providing new insights for the treatment of digestive system tumors.
Chimeric antigen receptor (CAR)-T cell therapy has become an effective means for cancer treatment, and the application of circRNAs in this field is gradually receiving attention. Experiments have shown that circRNAs can enhance the anti-tumor activity of CAR-T cells and improve their persistence and specificity[43]. For example, circRNAs can alleviate T cell exhaustion by modulating T cell metabolism and function, thereby enhancing their ability to kill tumor cells[44]. Therefore, incorporating circRNAs into CAR-T cell therapy holds the promise of improving therapeutic outcomes and prognoses for patients with digestive system tumors.
The metabolic reprogramming observed in tumor cells is a fundamental aspect of their proliferation and metastasis. CircRNAs influence tumor progression by regulating key metabolic enzymes and affecting metabolic pathways in tumor cells[45]. CircRNAs can enhance the invasiveness of tumor cells by influencing metabolic pathways such as glycolysis and fatty acid oxidation[46]. Therefore, studying the role of circRNAs in tumor metabolic reprogramming provides novel targets for the treatment of digestive system tumors.
Combination therapy is an important strategy to improve cancer treatment efficacy. The combined application of circRNAs with other treatment methods can produce synergistic effects, enhancing anti-tumor effects. For instance, using circRNAs in conjunction with chemotherapy drugs or immune checkpoint inhibitors can effectively overcome tumor resistance and improve treatment efficacy[47]. Additionally, combination therapy strategies involving circRNAs can also enhance immune responses by modulating the tumor microenvironment, further improving treatment outcomes for digestive system tumors. Thus, combination therapy strategies involving circRNAs provide new directions for the treatment of digestive system tumors.
As research continues to deepen on circRNAs in digestive system tumors, recent findings are showing their potential for clinical applications. Recent studies have not only elucidated the molecular mechanisms underlying tumorigenesis and progression but have also facilitated the identification of new biomarkers and therapeutic targets. Many researchers have made numerous discoveries and perspectives through in-depth studies of circRNAs, forming a series of research outcomes. For example, some studies have indicated that specific circRNAs are significantly upregulated or downregulated in digestive system tumors, and these circRNAs may have important impacts on tumor development by regulating gene expression[48]. However, research results regarding the role of circRNAs in different types of digestive system tumors are not entirely consistent, leading to varying opinions on their clinical applications[49,50]. To address this phenomenon, future research needs to be more systematic and standardized. On the one hand, large-scale clinical sample analyses can further validate the expression patterns and biological functions of circRNAs in various digestive system tumors. On the other hand, exploring the crosstalk between circRNAs and other molecules (miRNAs, long non-coding RNAs, etc.), as well as their role in the tumor microenvironment, will help construct a more comprehensive network model of the emergence and progression of digestive system tumors.
Multi-center studies and interdisciplinary collaborations, such as the integration of molecular biology, clinical medicine, and bioinformatics, can provide broader perspectives and deeper analytical tools for circRNA research. This can not only promote the progress of circRNAs research in digestive system tumors but also facilitate its application in clinical practice. Overall, research on circRNAs in digestive system tumors is at a rapidly developing stage. We should focus on revealing the functional mechanisms of circRNAs, clarifying their roles in tumor biology, and promoting their translation into clinical applications through clinical validation and multi-center research. With the deepening of research, circRNAs are expected to provide new ideas and methods for the early diagnosis and personalized treatment of digestive system tumors, ultimately improving patient prognosis and quality of life.
| 1. | 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. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 16] [Reference Citation Analysis (0)] |
| 2. | Fang N, Ding GW, Ding H, Li J, Liu C, Lv L, Shi YJ. Research Progress of Circular RNA in Gastrointestinal Tumors. Front Oncol. 2021;11:665246. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 4] [Cited by in RCA: 7] [Article Influence: 1.8] [Reference Citation Analysis (0)] |
| 3. | Xi SJ, Cai WQ, Wang QQ, Peng XC. Role of circular RNAs in gastrointestinal tumors and drug resistance. World J Clin Cases. 2021;9:10400-10417. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in CrossRef: 4] [Cited by in RCA: 6] [Article Influence: 1.5] [Reference Citation Analysis (0)] |
| 4. | Chen C, Xia C, Tang H, Jiang Y, Wang S, Zhang X, Huang T, Yuan X, Wang J, Peng L. Circular RNAs Involve in Immunity of Digestive Cancers From Bench to Bedside: A Review. Front Immunol. 2022;13:833058. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 2] [Cited by in RCA: 8] [Article Influence: 2.7] [Reference Citation Analysis (0)] |
| 5. | Wang H, Xiang Y, Hu R, Ji R, Wang Y. Research progress of circular RNA in digestive tract tumors: a narrative review. Transl Cancer Res. 2020;9:7632-7641. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 2] [Reference Citation Analysis (0)] |
| 6. | Li Z, Lu J. CircRNAs in osteoarthritis: research status and prospect. Front Genet. 2023;14:1173812. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 9] [Reference Citation Analysis (0)] |
| 7. | Samavarchi Tehrani S, Esmaeili F, Shirzad M, Goodarzi G, Yousefi T, Maniati M, Taheri-Anganeh M, Anushiravani A. The critical role of circular RNAs in drug resistance in gastrointestinal cancers. Med Oncol. 2023;40:116. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 5] [Article Influence: 2.5] [Reference Citation Analysis (0)] |
| 8. | Liu B, Gong Y, Jiang Q, Wu S, Han B, Chen F, Lin Q, Wang P, Yang D. Hsa_circ_0014784-induced YAP1 promoted the progression of pancreatic cancer by sponging miR-214-3p. Cell Cycle. 2023;22:1583-1596. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 9] [Reference Citation Analysis (0)] |
| 9. | Xu Y, Han J, Zhang X, Zhang X, Song J, Gao Z, Qian H, Jin J, Liang Z. Exosomal circRNAs in gastrointestinal cancer: Role in occurrence, development, diagnosis and clinical application (Review). Oncol Rep. 2024;51:19. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 5] [Cited by in RCA: 6] [Article Influence: 6.0] [Reference Citation Analysis (0)] |
| 10. | Chunyan-Zou, Yanpeng-He, Changshun-Xie, Fulai-Gao, Xiaoli-Xie, Xiukun-Chai, Chenxi-He, Dongqiang-Zhao. Potential Value of Circular RNA circTBC1D4 in Gastrointestinal Stromal Tumors. J Immunol Res. 2022;2022:9019097. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 4] [Reference Citation Analysis (0)] |
| 11. | Guo JU, Agarwal V, Guo H, Bartel DP. Expanded identification and characterization of mammalian circular RNAs. Genome Biol. 2014;15:409. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 1021] [Cited by in RCA: 1297] [Article Influence: 117.9] [Reference Citation Analysis (0)] |
| 12. | Suster I, Feng Y. Multifaceted Regulation of MicroRNA Biogenesis: Essential Roles and Functional Integration in Neuronal and Glial Development. Int J Mol Sci. 2021;22:6765. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 3] [Cited by in RCA: 25] [Article Influence: 6.3] [Reference Citation Analysis (0)] |
| 13. | Pourmehran Y, Sadri F, Hosseini SF, Mohammadi Y, Rezaei Z. Exploring the influence of non-coding RNAs on NF-κB signaling pathway regulation in ulcerative colitis. Biomed Pharmacother. 2024;179:117390. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 5] [Reference Citation Analysis (0)] |
| 14. | Meng E, Deng J, Jiang R, Wu H. CircRNA-Encoded Peptides or Proteins as New Players in Digestive System Neoplasms. Front Oncol. 2022;12:944159. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 11] [Reference Citation Analysis (0)] |
| 15. | Zhao S, Ly A, Mudd JL, Rozycki EB, Webster J, Coonrod E, Othoum G, Luo J, Dang HX, Fields RC, Maher CA. Characterization of cell-type specific circular RNAs associated with colorectal cancer metastasis. NAR Cancer. 2023;5:zcad021. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 5] [Article Influence: 2.5] [Reference Citation Analysis (0)] |
| 16. | Chen W, Gu X, Lv X, Cao X, Yuan Z, Wang S, Sun W. Non-coding transcriptomic profiles in the sheep mammary gland during different lactation periods. Front Vet Sci. 2022;9:983562. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 1] [Cited by in RCA: 8] [Article Influence: 2.7] [Reference Citation Analysis (0)] |
| 17. | Hussen BM, Honarmand Tamizkar K, Hidayat HJ, Taheri M, Ghafouri-Fard S. The role of circular RNAs in the development of hepatocellular carcinoma. Pathol Res Pract. 2021;223:153495. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 6] [Cited by in RCA: 11] [Article Influence: 2.8] [Reference Citation Analysis (0)] |
| 18. | Lin C, Xi Y, Yu H, Wang Z, Chen X, Shen W. circRNA TCFL5 Promote Esophageal Cancer Progression by Modulating M2 Macrophage Polarization via the miR-543-FMNL2 Axis. J Oncol. 2022;2022:5075615. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 8] [Reference Citation Analysis (0)] |
| 19. | Wang T, Wang J, Ren W, Chen S, Cheng YF, Zhang XM. CircRNA-0008717 promotes cell proliferation, migration, and invasion by regulating miR-203/Slug in esophageal cancer cells. Ann Transl Med. 2020;8:999. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 6] [Cited by in RCA: 12] [Article Influence: 2.4] [Reference Citation Analysis (0)] |
| 20. | Huang S, Zhang X, Guan B, Sun P, Hong CT, Peng J, Tang S, Yang J. A novel circular RNA hsa_circ_0008035 contributes to gastric cancer tumorigenesis through targeting the miR-375/YBX1 axis. Am J Transl Res. 2019;11:2455-2462. [PubMed] |
| 21. | Ding P, Liu P, Wu H, Yang P, Tian Y, Zhao Q. Functional properties of circular RNAs and research progress in gastric cancer. Front Oncol. 2022;12:954637. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 3] [Cited by in RCA: 4] [Article Influence: 1.3] [Reference Citation Analysis (0)] |
| 22. | Qi L, Pan Y, Tang M, Chen X. Circulating cell-free circRNA panel predicted tumorigenesis and development of colorectal cancer. J Clin Lab Anal. 2022;36:e24431. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 6] [Reference Citation Analysis (0)] |
| 23. | Chen CK, Cheng R, Demeter J, Chen J, Weingarten-Gabbay S, Jiang L, Snyder MP, Weissman JS, Segal E, Jackson PK, Chang HY. Structured elements drive extensive circular RNA translation. Mol Cell. 2021;81:4300-4318.e13. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 176] [Cited by in RCA: 188] [Article Influence: 47.0] [Reference Citation Analysis (0)] |
| 24. | Wu X, Li Z, Cao C, Ge J, Shen X, Sun W, Guo J, Guo J. A novel DNA binding protein encoded by circZNF131 inhibits the growth of gastric cancer by suppressing CTBP2 transcription. Int J Biol Macromol. 2025;314:144236. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 2] [Reference Citation Analysis (0)] |
| 25. | Liu Y, Cao J, Zhu L, Zhao W, Zhou Y, Shao C, Shao S. Circular RNA circPGD contributes to gastric cancer progression via the sponging miR-16-5p/ABL2 axis and encodes a novel PGD-219aa protein. Cell Death Discov. 2022;8:384. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 13] [Reference Citation Analysis (0)] |
| 26. | Wang M, Gu B, Yao G, Li P, Wang K. Circular RNA Expression Profiles and the Pro-tumorigenic Function of CircRNA_10156 in Hepatitis B Virus-Related Liver Cancer. Int J Med Sci. 2020;17:1351-1365. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 16] [Cited by in RCA: 39] [Article Influence: 7.8] [Reference Citation Analysis (0)] |
| 27. | Chen M, Liu X, Lu J, Teng H, Yu C, Liu Y, Zheng Y. Dysregulation of the circ_0087502/miR-1179/TGFBR2 pathway supports gemcitabine resistance in pancreatic cancer. Cancer Biol Ther. 2023;24:2258566. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 9] [Reference Citation Analysis (0)] |
| 28. | Duan JL, Chen W, Xie JJ, Zhang ML, Nie RC, Liang H, Mei J, Han K, Xiang ZC, Wang FW, Teng K, Chen RX, Deng MH, Yin YX, Zhang N, Xie D, Cai MY. A novel peptide encoded by N6-methyladenosine modified circMAP3K4 prevents apoptosis in hepatocellular carcinoma. Mol Cancer. 2022;21:93. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 75] [Cited by in RCA: 103] [Article Influence: 34.3] [Reference Citation Analysis (0)] |
| 29. | Cao SQ, Xue ST, Li WJ, Hu GS, Wu ZG, Zheng JC, Zhang SL, Lin X, Chen C, Liu W, Zheng B. CircHIPK3 regulates fatty acid metabolism through miR-637/FASN axis to promote esophageal squamous cell carcinoma. Cell Death Discov. 2024;10:110. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 10] [Reference Citation Analysis (0)] |
| 30. | Wang GJ, Yu TY, Li YR, Liu YJ, Deng BB. Circ_0000190 suppresses gastric cancer progression potentially via inhibiting miR-1252/PAK3 pathway. Cancer Cell Int. 2020;20:351. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 5] [Cited by in RCA: 17] [Article Influence: 3.4] [Reference Citation Analysis (0)] |
| 31. | Ding N, You AB, Yang H, Hu GS, Lai CP, Liu W, Ye F. A Tumor-suppressive Molecular Axis EP300/circRERE/miR-6837-3p/MAVS Activates Type I IFN Pathway and Antitumor Immunity to Suppress Colorectal Cancer. Clin Cancer Res. 2023;29:2095-2109. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 32] [Reference Citation Analysis (0)] |
| 32. | Pan Z, Zheng J, Zhang J, Lin J, Lai J, Lyu Z, Feng H, Wang J, Wu D, Li Y. A Novel Protein Encoded by Exosomal CircATG4B Induces Oxaliplatin Resistance in Colorectal Cancer by Promoting Autophagy. Adv Sci (Weinh). 2022;9:e2204513. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 46] [Cited by in RCA: 69] [Article Influence: 23.0] [Reference Citation Analysis (0)] |
| 33. | Li Q, Zhang Y, Jin P, Chen Y, Zhang C, Geng X, Mun KS, Phang KC. New insights into the potential of exosomal circular RNAs in mediating cancer chemotherapy resistance and their clinical applications. Biomed Pharmacother. 2024;177:117027. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 9] [Reference Citation Analysis (0)] |
| 34. | Lakiotaki E, Kanakoglou DS, Pampalou A, Karatrasoglou EA, Piperi C, Korkolopoulou P. Dissecting the Role of Circular RNAs in Sarcomas with Emphasis on Osteosarcomas. Biomedicines. 2021;9:1642. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 3] [Cited by in RCA: 5] [Article Influence: 1.3] [Reference Citation Analysis (0)] |
| 35. | Zhang Y, Chao F, Lv L, Li M, Shen Z. Hsa_circ_0041150 serves as a novel biomarker for monitoring chemotherapy resistance in small cell lung cancer patients treated with a first-line chemotherapy regimen. J Cancer Res Clin Oncol. 2023;149:15365-15382. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 3] [Cited by in RCA: 6] [Article Influence: 3.0] [Reference Citation Analysis (0)] |
| 36. | Ju C, He J, Wang C, Sheng J, Jia J, Du D, Li H, Zhou M, He F. Current advances and future perspectives on the functional roles and clinical implications of circular RNAs in esophageal squamous cell carcinoma: more influential than expected. Biomark Res. 2022;10:41. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 11] [Cited by in RCA: 19] [Article Influence: 6.3] [Reference Citation Analysis (0)] |
| 37. | Li Z, Yin S, Yang K, Zhang B, Wu X, Zhang M, Gao D. CircRNA Regulation of T Cells in Cancer: Unraveling Potential Targets. Int J Mol Sci. 2024;25:6383. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 6] [Reference Citation Analysis (0)] |
| 38. | Zang R, Qiu X, Song Y, Wang Y. Exosomes Mediated Transfer of Circ_0000337 Contributes to Cisplatin (CDDP) Resistance of Esophageal Cancer by Regulating JAK2 via miR-377-3p. Front Cell Dev Biol. 2021;9:673237. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 13] [Cited by in RCA: 43] [Article Influence: 10.8] [Reference Citation Analysis (0)] |
| 39. | Li J, Li H, Lv X, Yang Z, Gao M, Bi Y, Zhang Z, Wang S, Cui Z, Zhou B, Yin Z. Diagnostic performance of circular RNAs in human cancers: A systematic review and meta-analysis. Mol Genet Genomic Med. 2019;7:e00749. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 14] [Cited by in RCA: 17] [Article Influence: 2.8] [Reference Citation Analysis (0)] |
| 40. | Yan J, Chen X, Dong Q, Lin J, Sun X. Analysis of Potential Circular RNAs in Regulating Imatinib Resistance of Gastrointestinal Stromal Tumor. Comb Chem High Throughput Screen. 2024;27:1371-1380. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 2] [Article Influence: 2.0] [Reference Citation Analysis (0)] |
| 41. | Li H, Peng K, Yang K, Ma W, Qi S, Yu X, He J, Lin X, Yu G. Circular RNA cancer vaccines drive immunity in hard-to-treat malignancies. Theranostics. 2022;12:6422-6436. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 132] [Reference Citation Analysis (0)] |
| 42. | Wei X, Shi Y, Dai Z, Wang P, Meng X, Yin B. Underlying metastasis mechanism and clinical application of exosomal circular RNA in tumors (Review). Int J Oncol. 2021;58:289-297. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 6] [Cited by in RCA: 25] [Article Influence: 6.3] [Reference Citation Analysis (0)] |
| 43. | Huang S, Xu J, Baran N, Ma W. Advancing the next generation of cancer treatment with circular RNAs in CAR-T cell therapy. Biomed Pharmacother. 2024;181:117753. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 9] [Article Influence: 9.0] [Reference Citation Analysis (0)] |
| 44. | Khan SU, Khan MU, Khan MI, Fadahunsi AA, Khan A, Gao S, Bilal M, Li F. Role of circular RNAs in disease progression and diagnosis of cancers: An overview of recent advanced insights. Int J Biol Macromol. 2022;220:973-984. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 2] [Cited by in RCA: 22] [Article Influence: 7.3] [Reference Citation Analysis (0)] |
| 45. | Alkhathami AG, Sahib AS, Al Fayi MS, Fadhil AA, Jawad MA, Shafik SA, Sultan SJ, Almulla AF, Shen M. Glycolysis in human cancers: Emphasis circRNA/glycolysis axis and nanoparticles in glycolysis regulation in cancer therapy. Environ Res. 2023;234:116007. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 2] [Cited by in RCA: 12] [Article Influence: 6.0] [Reference Citation Analysis (0)] |
| 46. | Hsu CY, Faisal A, Jumaa SS, Gilmanova NS, Ubaid M, Athab AH, Mirzaei R, Karampoor S. Exploring the impact of circRNAs on cancer glycolysis: Insights into tumor progression and therapeutic strategies. Noncoding RNA Res. 2024;9:970-994. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 7] [Cited by in RCA: 8] [Article Influence: 8.0] [Reference Citation Analysis (0)] |
| 47. | Hashemi M, Khosroshahi EM, Daneii P, Hassanpoor A, Eslami M, Koohpar ZK, Asadi S, Zabihi A, Jamali B, Ghorbani A, Nabavi N, Memarkashani MR, Salimimoghadam S, Taheriazam A, Tan SC, Entezari M, Farahani N, Hushmandi K. Emerging roles of CircRNA-miRNA networks in cancer development and therapeutic response. Noncoding RNA Res. 2025;10:98-115. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 18] [Reference Citation Analysis (0)] |
| 48. | Zhao X, Wang Y, Yu Q, Yu P, Zheng Q, Yang X, Gao D. Circular RNAs in gastrointestinal cancer: Current knowledge, biomarkers and targeted therapy (Review). Int J Mol Med. 2020;46:1611-1632. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 5] [Reference Citation Analysis (0)] |
| 49. | Sun Z, Chen C, Su Y, Wang W, Yang S, Zhou Q, Wang G, Li Z, Song J, Zhang Z, Yuan W, Liu J. Regulatory mechanisms and clinical perspectives of circRNA in digestive system neoplasms. J Cancer. 2019;10:2885-2891. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 20] [Cited by in RCA: 33] [Article Influence: 5.5] [Reference Citation Analysis (0)] |
| 50. | Chen Z, Zhang L, Han G, Zuo X, Zhang Y, Zhu Q, Wu J, Wang X. A Meta-Analysis of the Diagnostic Accuracy of Circular RNAs in Digestive System Malignancy. Cell Physiol Biochem. 2018;45:962-972. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 15] [Cited by in RCA: 20] [Article Influence: 2.9] [Reference Citation Analysis (0)] |
| 51. | Jiang S, Li H, Zhang L, Mu W, Zhang Y, Chen T, Wu J, Tang H, Zheng S, Liu Y, Wu Y, Luo X, Xie Y, Ren J. Generic Diagramming Platform (GDP): a comprehensive database of high-quality biomedical graphics. Nucleic Acids Res. 2025;53:D1670-D1676. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 4] [Cited by in RCA: 202] [Article Influence: 202.0] [Reference Citation Analysis (0)] |
| 52. | Meng L, Wu H, Wu J, Ding P, He J, Li T, Niu X, Sang M, Liu L. Aberrant CircTMEM45A Facilitates Inflammatory Progression of Esophageal Squamous Cell Carcinoma through m5C-Mediated NLRP3 Activation. Cancer Res. 2025;85:2694-2713. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 2] [Article Influence: 2.0] [Reference Citation Analysis (0)] |
| 53. | Wu L, Gu L, Zheng Y, Liu J, Wei Z, Liu F, Li J, Meng L, Sang Y, Sang M, Zhao L, Shan B. CircPRKCA facilitates esophageal squamous cell carcinoma metastasis via m5C-dependent CSF2 mRNA stabilization. J Transl Med. 2025;23:385. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 2] [Reference Citation Analysis (0)] |
| 54. | Wang C, Ju C, Du D, Zhu P, Yin J, Jia J, Wang X, Xu X, Zhao L, Wan J, Sun T, Yang L, Li H, He F, Zhou M, He J. CircNF1 modulates the progression and immune evasion of esophageal squamous cell carcinoma through dual regulation of PD-L1. Cell Mol Biol Lett. 2025;30:37. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 5] [Reference Citation Analysis (0)] |
| 55. | Yuan J, Cui Y, Zhang J, Cai Y, Xu X. CircMMP11 is a Potential Recurrence Biomarker and Facilitates Progression of Esophageal Squamous Cell Carcinoma. Biochem Genet. 2025. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 1] [Reference Citation Analysis (0)] |
| 56. | Gao S, Lou W. miR-542-3p/PIK3R1 axis is involved in hsa_circ_0087104-mediated inhibition of esophageal squamous cell carcinoma metastasis. Am J Cancer Res. 2024;14:5665-5679. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 2] [Reference Citation Analysis (0)] |
| 57. | Zhang X, Bian Y, Li Q, Yu C, Gao Y, Tian B, Xia W, Wang W, Xin L, Lin H, Wang L. EIF4A3-mediated oncogenic circRNA hsa_circ_0001165 advances esophageal squamous cell carcinoma progression through the miR-381-3p/TNS3 pathway. Cell Biol Toxicol. 2024;40:84. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 7] [Reference Citation Analysis (0)] |
| 58. | Liu J, Wei Z, Meng L, Wu L, Liu F, Sang M, Zhao L, Gu L, Shan B. CircJPH1 regulates the NF-κB/HERC5 axis to promote the malignant progression of esophageal squamous cell carcinoma through binding to XRCC6. Cell Signal. 2024;124:111403. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 2] [Reference Citation Analysis (0)] |
| 59. | Dai Y, Xu Q, Xia M, Chen C, Xiong X, Yang X, Wang W. Hsa_circ_0001615 downregulation inhibits esophageal cancer development through miR-142-5p/β-catenin. PeerJ. 2024;12:e17089. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 2] [Reference Citation Analysis (0)] |
| 60. | Lei K, Liang R, Liang J, Lu N, Huang J, Xu K, Tan B, Wang K, Liang Y, Wang W, Lin H, Wang M. CircPDE5A-encoded novel regulator of the PI3K/AKT pathway inhibits esophageal squamous cell carcinoma progression by promoting USP14-mediated de-ubiquitination of PIK3IP1. J Exp Clin Cancer Res. 2024;43:124. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 16] [Reference Citation Analysis (0)] |
| 61. | Luo Q, Li J, Miao H, Su S, Chen Y, Xu C, Zhao C, Huang J, Ling K, Lin C, Yan H, Zhang S. circSSPO boosts growth of esophageal squamous cell carcinoma through upregulation of micrRNA-6820-5p-mediated KLK8 and PKD1 expression. Cell Biol Toxicol. 2023;39:3219-3234. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 3] [Article Influence: 1.5] [Reference Citation Analysis (0)] |
| 62. | Lyu Y, Tan B, Li L, Liang R, Lei K, Wang K, Wu D, Lin H, Wang M. A novel protein encoded by circUBE4B promotes progression of esophageal squamous cell carcinoma by augmenting MAPK/ERK signaling. Cell Death Dis. 2023;14:346. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 11] [Reference Citation Analysis (0)] |
| 63. | Xu R, Ding P, Zhao X, Li Z, Liu F, Gu L, Zheng Y, Sang M, Meng L. Circular RNA circ-TNRC6B inhibits the proliferation and invasion of esophageal squamous cell carcinoma cells by regulating the miR-452-5p/DAG1 axis. Mol Oncol. 2023;17:1437-1452. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 13] [Reference Citation Analysis (0)] |
| 64. | Mei B, Chen J, Peng Y. The circRNA circSCAF8 promotes tumor growth and metastasis of gastric cancer via miR-1293/TIMP1signaling. Gene Ther. 2025;32:142-153. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 2] [Article Influence: 2.0] [Reference Citation Analysis (0)] |
| 65. | Song X, Zhang G, Niu J, Liu H, Li C, Ning W, Zhou L. Circular RNA circPFKP suppress gastric cancer progression through targeting miR-346/CAMD3 axis. Exp Cell Res. 2025;445:114390. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 2] [Reference Citation Analysis (0)] |
| 66. | Zhang H, Huang Z, Zhong Y. Circular RNA Circ_0079226 Plays an Oncogenic Role in Gastric Cancer via the miR-155-5p/FOXK1/AKT Pathway. Anal Cell Pathol (Amst). 2025;2025:6619550. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 1] [Reference Citation Analysis (0)] |
| 67. | Wang Z, Chen W, Wang Z, Dai X. EIF4A3-Mediated circ_0008126 Inhibits the Progression and Metastasis of Gastric Cancer by Modulating the APC/β-Catenin Pathway. Cancers (Basel). 2025;17:253. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 1] [Reference Citation Analysis (0)] |
| 68. | Deng C, Huo M, Chu H, Zhuang X, Deng G, Li W, Wei H, Zeng L, He Y, Liu H, Li J, Zhang C, Chen H. Exosome circATP8A1 induces macrophage M2 polarization by regulating the miR-1-3p/STAT6 axis to promote gastric cancer progression. Mol Cancer. 2024;23:49. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 80] [Reference Citation Analysis (0)] |
| 69. | Tang Z, Li J, Lu B, Zhang X, Yang L, Qi Y, Jiang S, Wu Q, Wang Y, Cheng T, Xu M, Sun P, Wang X, Miao K, Wu H, Huang J. CircBIRC6 facilitates the malignant progression via miR-488/GRIN2D-mediated CAV1-autophagy signal axis in gastric cancer. Pharmacol Res. 2024;202:107127. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 10] [Reference Citation Analysis (0)] |
| 70. | Ding P, Wu H, Wu J, Li T, He J, Ju Y, Liu Y, Li F, Deng H, Gu R, Zhang L, Guo H, Tian Y, Yang P, Meng N, Li X, Guo Z, Meng L, Zhao Q. N6-methyladenosine modified circPAK2 promotes lymph node metastasis via targeting IGF2BPs/VEGFA signaling in gastric cancer. Oncogene. 2024;43:2548-2563. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 19] [Reference Citation Analysis (0)] |
| 71. | Miao Z, Li J, Wang Y, Shi M, Gu X, Zhang X, Wei F, Tang X, Zheng L, Xing Y. Hsa_circ_0136666 stimulates gastric cancer progression and tumor immune escape by regulating the miR-375/PRKDC Axis and PD-L1 phosphorylation. Mol Cancer. 2023;22:205. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 33] [Cited by in RCA: 51] [Article Influence: 25.5] [Reference Citation Analysis (0)] |
| 72. | Zhang C, Wei G, Zhu X, Chen X, Ma X, Hu P, Liu W, Yang W, Ruan T, Zhang W, Wu C, Tao K. Exosome-Delivered circSTAU2 Inhibits the Progression of Gastric Cancer by Targeting the miR-589/CAPZA1 Axis. Int J Nanomedicine. 2023;18:127-142. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 22] [Reference Citation Analysis (0)] |
| 73. | Zhu L, Yao Z, Liu Y, Shao S, Zhou W, Ding J, Mao B, Shao C, Cui F. circGAPVD1 inhibits the progression of gastric cancer through miR-4424/STK4 axis and encoding GAPVD1-137aa protein. Int J Biol Macromol. 2025;319:145408. [PubMed] [DOI] [Full Text] |
| 74. | Wang S, Wu C, Wang J, Yuan F, Hou Y, Cao T, Xu L, Qian L, Xia Y, Xu L, Zeng A, Wang X, Wang L, Huang X. Hsa_circ_0002301 inhibits ferroptosis in gastric cancer by encoding the de novo protein HECTD1-463aa. J Transl Med. 2025;23:250. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 3] [Reference Citation Analysis (0)] |
| 75. | Liu YY, Zhang YY, Ran LY, Huang B, Ren JW, Ma Q, Pan XJ, Yang FF, Liang C, Wang XL, Wang SM, Ran A, Ning H, Jiang Y, Qin CH, Xiao B. A novel protein FNDC3B-267aa encoded by circ0003692 inhibits gastric cancer metastasis via promoting proteasomal degradation of c-Myc. J Transl Med. 2024;22:507. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 8] [Cited by in RCA: 11] [Article Influence: 11.0] [Reference Citation Analysis (0)] |
| 76. | Tang L, Ji Y, Ni C, Xu Z, Shen Y, Lu H, Zhang C, Yang S. EIF4A3-Mediated Biogenesis of CircFADS1 Promotes the Progression of Hepatocellular Carcinoma via Wnt/β-Catenin Pathway. Adv Sci (Weinh). 2025;12:e2411869. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 6] [Reference Citation Analysis (0)] |
| 77. | Li H, Su B, Jiang Y, Zhang B, Du R, Song C, Hou B, Xu K, Wu L, Gu Y. Circular RNA circDCUN1D4 suppresses hepatocellular carcinoma development via targeting the miR-590-5p/ TIMP3 axis. Mol Cancer. 2025;24:95. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 4] [Reference Citation Analysis (0)] |
| 78. | Zhang Y, Yao R, Li M, Fang C, Feng K, Chen X, Wang J, Luo R, Shi H, Chen X, Zhao X, Huang H, Liu S, Yin B, Zhong C. CircTTC13 promotes sorafenib resistance in hepatocellular carcinoma through the inhibition of ferroptosis by targeting the miR-513a-5p/SLC7A11 axis. Mol Cancer. 2025;24:32. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 10] [Reference Citation Analysis (0)] |
| 79. | Zhang XY, Li SS, Gu YR, Xiao LX, Ma XY, Chen XR, Wang JL, Liao CH, Lin BL, Huang YH, Lian YF. CircPIAS1 promotes hepatocellular carcinoma progression by inhibiting ferroptosis via the miR-455-3p/NUPR1/FTH1 axis. Mol Cancer. 2024;23:113. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 36] [Reference Citation Analysis (0)] |
| 80. | Ji Y, Ni C, Shen Y, Xu Z, Tang L, Yu F, Zhu L, Lu H, Zhang C, Yang S, Wang X. ESRP1-mediated biogenesis of circPTPN12 inhibits hepatocellular carcinoma progression by PDLIM2/ NF-κB pathway. Mol Cancer. 2024;23:143. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 15] [Cited by in RCA: 13] [Article Influence: 13.0] [Reference Citation Analysis (0)] |
| 81. | Yang Y, Chen XQ, Jia YX, Ma J, Xu D, Xiang ZL. Circ-0044539 promotes lymph node metastasis of hepatocellular carcinoma through exosomal-miR-29a-3p. Cell Death Dis. 2024;15:630. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 8] [Reference Citation Analysis (0)] |
| 82. | Chen L, Xiao H, Wu Y, Yan D, Shan M, Sun L, Yan X, Liu D, Li T, Zhang Y, Xiang L, Chen A, Li S, Xiang W, Ni Z, He F, Yang M, Lian J. CircPHKB decreases the sensitivity of liver cancer cells to sorafenib via miR-1234-3p/CYP2W1 axis. Genomics. 2024;116:110764. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 4] [Article Influence: 4.0] [Reference Citation Analysis (0)] |
| 83. | Chen X, Xu Y, Zhou Z, Zhao P, Zhou Z, Wang F, Zhong F, Du H. CircUSP10 promotes liver cancer progression by regulating miR-211-5p/TCF12/EMT signaling pathway. Heliyon. 2023;9:e20649. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 4] [Reference Citation Analysis (0)] |
| 84. | Yu S, Su S, Wang P, Li J, Chen C, Xin H, Gong Y, Wang H, Ye X, Mao L, Zhou Z, Zhou S, Hu Z, Huang X. Tumor-associated macrophage-induced circMRCKα encodes a peptide to promote glycolysis and progression in hepatocellular carcinoma. Cancer Lett. 2024;591:216872. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 9] [Reference Citation Analysis (0)] |
| 85. | Lu J, Ru J, Chen Y, Ling Z, Liu H, Ding B, Jiang Y, Ma J, Zhang D, Ge J, Li Y, Sun F, Chen D, Zheng S, Wu J. N(6) -methyladenosine-modified circSTX6 promotes hepatocellular carcinoma progression by regulating the HNRNPD/ATF3 axis and encoding a 144 amino acid polypeptide. Clin Transl Med. 2023;13:e1451. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 31] [Reference Citation Analysis (0)] |
| 86. | Fang Z, Wu Z, Yu C, Xie Q, Zeng L, Chen R. EIF4E-mediated biogenesis of circPHF14 promotes the growth and metastasis of pancreatic ductal adenocarcinoma via Wnt/β-catenin pathway. Mol Cancer. 2025;24:56. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 1] [Cited by in RCA: 6] [Article Influence: 6.0] [Reference Citation Analysis (0)] |
| 87. | Zhang J, Sun W, Wu W, Qin Z, Wei B, Li T. METTL3-dependent m6A methylation of circCEACAM5 fuels pancreatic cancer progression through DKC1 activation. Cell Mol Life Sci. 2025;82:132. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 3] [Reference Citation Analysis (0)] |
| 88. | Lee KL, Liu JJ, Huang WJ, Hung CS, Liang YC. CircRNA hsa_circ_0004781 promoted cell proliferation by acting as a sponge for miR-9-5p and miR-338-3p and upregulating KLF5 and ADAM17 expression in pancreatic ductal adenocarcinoma. Cancer Cell Int. 2025;25:56. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 1] [Reference Citation Analysis (0)] |
| 89. | Yuan H, Chen C, Li H, Qu G, Chen L, Liu Y, Zhang Y, Zhao Q, Lian C, Ji A, Hou X, Liu X, Jiang K, Zhu Y, He Y. Role of a novel circRNA-CGNL1 in regulating pancreatic cancer progression via NUDT4-HDAC4-RUNX2-GAMT-mediated apoptosis. Mol Cancer. 2024;23:27. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 5] [Cited by in RCA: 13] [Article Influence: 13.0] [Reference Citation Analysis (0)] |
| 90. | Huang W, Hu X, He X, Pan D, Huang Z, Gu Z, Huang G, Wang P, Cui C, Fan Y. TRIM29 facilitates gemcitabine resistance via MEK/ERK pathway and is modulated by circRPS29/miR-770-5p axis in PDAC. Drug Resist Updat. 2024;74:101079. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 13] [Reference Citation Analysis (0)] |
| 91. | Shi Y, Shen M, Yang Y, Qiu J. CircDUSP22 Overexpression Restrains Pancreatic Cancer Development via Modulating miR-1178-3p and Downstream BNIP3. Biochem Genet. 2023;61:651-668. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 2] [Reference Citation Analysis (0)] |
| 92. | Li C, Cai J, Liu W, Gao Z, Li G. Downregulation of circ-STK39 suppresses pancreatic cancer progression by sponging mir-140-3p and regulating TRAM2-mediated epithelial-mesenchymal transition. Apoptosis. 2023;28:1024-1034. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 4] [Cited by in RCA: 7] [Article Influence: 3.5] [Reference Citation Analysis (0)] |
| 93. | Guo J, Wang Y, Yang Y, Feng S, Cai J, Fan S, Wang J, Li N, Qian X, Zhu H. CircRNA hsa_ circ_0009022 inhibits colorectal cancer progression by sponging miR-576-5p and complexing with FMRP. Tissue Cell. 2025;95:102924. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 1] [Reference Citation Analysis (0)] |
| 94. | Wang F, Wang Q, Wu Y, Huang Z, Zhong X, Wang H, Yang C, Qin Y, Qi X, Ge X, Mao Y. CircMVP promotes METTL3 activation mediated CTNNB1 m6A modification in the inhibition of colorectal cancer in B7-H3 dependence antitumor immunity. Int J Biol Sci. 2025;21:306-327. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 5] [Cited by in RCA: 5] [Article Influence: 5.0] [Reference Citation Analysis (0)] |
| 95. | Zhao Y, He Y, Xiao Z, Xin L, Deng M, Yao M, Huang G. circEIF3I Promotes Colorectal Cancer Metastasis by Regulating the miR-328-3p/NCAPH Axis. Mol Carcinog. 2025;64:450-462. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 2] [Reference Citation Analysis (0)] |
| 96. | Zhao N, Cao Y, Tao R, Zhu X, Li R, Chen Y, Tao K, Li L, Chen H, Ma X. The circMYBL2-Encoded p185 Protein Suppresses Colorectal Cancer Progression by Inhibiting Serine Biosynthesis. Cancer Res. 2024;84:2155-2168. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 6] [Reference Citation Analysis (0)] |
| 97. | Jiang X, Peng M, Liu Q, Peng Q, Oyang L, Li S, Xu X, Shen M, Wang J, Li H, Wu N, Tan S, Lin J, Xia L, Tang Y, Luo X, Liao Q, Zhou Y. Circular RNA hsa_circ_0000467 promotes colorectal cancer progression by promoting eIF4A3-mediated c-Myc translation. Mol Cancer. 2024;23:151. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 15] [Reference Citation Analysis (0)] |
| 98. | Wang X, Sun T, Fan J, Zuo X, Mao J. Gastrin-related circRNA_0017065 promotes the proliferation and metastasis of colorectal cancer through the miR-3174/RBFOX2 axis. Biol Direct. 2024;19:75. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 3] [Reference Citation Analysis (0)] |
| 99. | Li Q, Li K, Guo Q, Yang T. CircRNA circSTIL inhibits ferroptosis in colorectal cancer via miR-431/SLC7A11 axis. Environ Toxicol. 2023;38:981-989. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 25] [Article Influence: 12.5] [Reference Citation Analysis (0)] |
| 100. | Yang Y, Luo D, Shao Y, Shan Z, Liu Q, Weng J, He W, Zhang R, Li Q, Wang Z, Li X. circCAPRIN1 interacts with STAT2 to promote tumor progression and lipid synthesis via upregulating ACC1 expression in colorectal cancer. Cancer Commun (Lond). 2023;43:100-122. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 43] [Reference Citation Analysis (0)] |
| 101. | Xiong L, Liu HS, Zhou C, Yang X, Huang L, Jie HQ, Zeng ZW, Zheng XB, Li WX, Liu ZZ, Kang L, Liang ZX. A novel protein encoded by circINSIG1 reprograms cholesterol metabolism by promoting the ubiquitin-dependent degradation of INSIG1 in colorectal cancer. Mol Cancer. 2023;22:72. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 48] [Reference Citation Analysis (0)] |
