- Review
- Open access
- Published:
The emerging functions and clinical implications of circRNAs in acute myeloid leukaemia
Cancer Cell International volume 25, Article number: 167 (2025)
Abstract
Acute myeloid leukaemia (AML) is a prevalent haematologic malignancy characterized by significant heterogeneity. Despite the application of aggressive therapeutic approaches, AML remains associated with poor prognosis. Circular RNAs (circRNAs) constitute a unique class of single-stranded RNAs featuring covalently closed loop structures that are ubiquitous across species. These molecules perform crucial regulatory functions in the pathogenesis of various diseases through diverse mechanisms, including acting as miRNA sponges, interacting with DNA or proteins, and encoding functional proteins/polypeptides. Recently, numerous circRNAs have been confirmed to have aberrant expression patterns in AML patients. In particular, certain circRNAs are closely associated with specific clinicopathological characteristics and thus have great potential as diagnostic/prognostic biomarkers and therapeutic targets in AML. Herein, we systematically summarize the biogenesis, degradation, and functional mechanisms of circRNAs while highlighting their clinical relevance. We also outline a series of online databases and analytical tools available to facilitate circRNA research. Finally, we discuss the current challenges and future research priorities in this evolving field.
Introduction
Acute myeloid leukaemia (AML) is a highly lethal haematologic malignancy characterized by the uncontrolled proliferation and differentiation arrest of immature myeloid cells [1]. Much progress has been made in AML management, with significant advances in molecular biology and therapeutic technologies [2] including optimized chemotherapy regimens [3], refined haematopoietic stem cell transplantation strategies [4,5,6], and novel targeted therapies such as Bcl-2 inhibitors [7], FLT3 tyrosine kinase inhibitors [7], CD33-targeting agents [8], IDH inhibitors [7], and CAR-T-cell immunotherapy [9, 10]. However, the clinical outcomes of AML remain suboptimal. Current data indicate a 5-year survival rate of barely 29.5%, and this low rate is attributed primarily to treatment failure, disease relapse, chemotherapy resistance, and limitations of the haematopoietic niche [11, 12]. These persistent challenges underscore the critical need to elucidate the mechanism underlying leukaemia initiation, maintenance and recurrence and to identify novel biomarkers for improved diagnosis, risk stratification and precision therapies [13].
Circular RNAs (circRNAs) represent a unique class of RNAs characterized by covalently closed circular structures devoid of 5’ caps and 3’ polyadenylated tails [14]. These RNAs originate predominantly through the back-splicing of linear precursor mRNAs (pre-mRNAs) [15]. They exhibit distinctive biological features, including evolutionary conservation, exceptional stability, tissue-specific expression patterns, and high abundance in mammalian cells and exosomes [14, 16, 17]. Although circRNAs were initially discovered in RNA viruses by Sanger and colleagues in 1976 [18], their significance in diseases remained poorly understood until recent breakthroughs in experimental techniques and bioinformatics analyses [19]. Recent studies have revealed that circRNAs are dysregulated in almost all cancer types [20] and can functionally modulate tumour cell proliferation, apoptosis, differentiation, invasion, metastasis, angiogenesis, autophagy, metabolism, stemness maintenance, immune escape, and drug resistance [20, 21]. Therefore, circRNAs are considered promising biomarkers and therapeutic targets for tumour treatment.
Notably, an increasing number of circRNAs have been identified as critical regulators of both the initiation and progression of AML [22, 23]. However, the precise roles and regulatory mechanisms of circRNAs in AML remain incompletely understood. Herein, we comprehensively delineate the molecular life cycle of circRNAs, including their biogenesis, degradation, regulation and function, with a particular focus on AML-associated circRNA dysregulation. We examine the expression profiles of numerous circRNAs in AML and describe their biological roles in cell proliferation, apoptosis, the cell cycle, differentiation, migration, invasion, extramedullary infiltration, ferroptosis, autophagy, stemness, drug resistance, exosomes, and the tumour microenvironment as elucidated in previous research. We also elaborate on the critical potential of these circRNAs as clinical biomarkers and innovative therapeutic targets. Additionally, we outline a series of essential online databases and tools for circRNA investigation. Finally, we discuss the current challenges and potential directions for future circRNA research. We hope that this review will help researchers achieve a better understanding of circRNAs and provide directions for further studies on circRNAs.
Biogenesis and degradation of circRNAs
Unlike the canonical linear splicing mechanism, circRNAs are typically produced through noncanonical alternative splicing, termed back-splicing, a process that joins downstream 5’ splice sites to upstream 3’ splice sites, forming covalently closed circRNAs [14]. Although their back-splicing efficiency is much lower than that of linear splicing, circRNAs exhibit greater stability than linear RNAs because of their circular conformation, which confers resistance to exonuclease-mediated degradation [14]. According to the literature, circRNAs that originate from pre-mRNAs can be classified into four broad categories: exonic circRNAs (ecircRNAs), exon‒intron circRNAs (EIciRNAs), intronic circRNAs (ciRNAs) and intergenic circRNAs [24] (Fig. 1A). Among them, ecircRNAs, composed exclusively of one or more exons, are the most common circRNA type and mainly localize in the cytoplasm [25]. In contrast, EIciRNAs retain intronic sequences between circularized exons and predominantly reside in the nucleus [26]. Notably, ecircRNAs and EIciRNAs may share common biogenesis mechanisms, including intron-pairing-driven circularization, RNA binding protein (RBP)-dependent circularization, or lariat-driven circularization [16, 24]. ciRNAs originate from intron lariats that fail to debranch during splicing and accumulate primarily in the nucleus [27]. Intergenic circRNAs originate from genomic regions between protein-coding genes, containing two intronic circRNA fragments that are spliced at flanking GT-AG splicing signals [28]. Moreover, several special types of circRNAs, such as fusion circRNAs (f-circRNAs) [29], tRNA intronic circular RNAs (tricRNAs) [30], mitochondria-encoded circRNAs (mecciRNAs) [31, 32], and read-through circRNAs (rt-circRNAs), have been identified [33] (Fig. 1A).
Biogenesis, degradation and regulatory functions of circRNAs. A Biogenesis of exonic circRNAs (ecircRNAs), exon‒intron circRNAs (EIciRNAs), intronic circRNAs (ciRNAs), intergenic circRNAs, read‒through circRNAs (rt-circRNAs), fusion circRNAs (f-circRNAs), tRNA intronic circular RNAs (tricRNAs), and mitochondria‒encoded circRNAs (mecciRNAs) and the potential mechanisms of circRNA degradation. B Potential regulatory mechanisms of circRNAs. (a) CircRNAs can serve as microRNA sponges to block microRNA-mediated target gene silencing. (b) CircRNAs can interact with DNA to form circR loops. (c) CircRNAs can regulate the functions of RBPs (I), recruit transcription activators (II), act as protein scaffolds (III), and interact with specific proteins (IV). (d) CircRNAs can be translated into proteins or peptides
Relative to linear RNAs, circRNAs exhibit superior stability across different cell and tissue types and in exosomes [17]. However, the mechanisms of circRNA degradation remain poorly understood, although recent studies have proposed several potential degradation pathways (Fig. 1A). Collectively, the available evidence suggests that circRNAs may be degraded by GW182 (a crucial component of the P-body and RNAi machine) [34], RNase L in cells upon poly(I:C) treatment or viral infection [35], or by interacting with miRNAs in an Argonaute 2 (Ago2)-dependent manner [36]. Specifically, m6A-modified circRNAs undergo endoribonucleolytic cleavage through the YTHDF2 (m6A reader protein)-HRSP12 (adaptor protein)-RNase P/MRP (endoribonuclease) pathway [37]. Furthermore, highly structured circRNAs can be degraded by G3BP1 and its associated protein UPF1 [38]. These findings indicate that circRNA degradation involves multiple coordinated mechanisms; however, comprehensive understanding requires further investigation.
Potential regulatory mechanisms of circRNAs
The regulatory mechanisms of circRNAs have been increasingly elucidated with research progress. Substantial evidence now confirms that circRNAs serve as critical regulators in diverse physiological and pathological processes. They exhibit multiple activities, including (1) sponging miRNAs, (2) interacting with DNA or proteins, and (3) encoding proteins or polypeptides (Fig. 1B). In this section, we systematically review the established circRNA regulatory paradigms, discuss controversial issues, and illustrate these topics with a few representative examples.
CircRNAs as microRNA sponges
Much of the research on circRNAs to date has focused predominantly on the activities of these molecules as miRNA sponges to regulate the expression of miRNA targets [39]. For example, circRPN2 acts as a sponge of miR-183-5p to derepress the expression of FOXO1, thereby regulating glucose metabolism and metastasis in hepatocellular carcinoma [40]. CircBCAR3 promotes oesophageal cancer tumorigenesis and metastasis by sponging miR-27a-3p and thereby upregulating TNPO1 [41]. CircMETTL3 restrains colorectal cancer development and metastasis by interacting with miR-107 to increase PER3 expression [42]. However, the biological functions of circRNA-mediated miRNA sponging requires careful validation. For example, when a circRNA harbours multiple miRNA response elements (MREs) but is expressed at low levels, its capacity to function as a miRNA sponge might be limited. Similarly, circRNAs possessing few MREs and with low cellular abundance are unlikely to exert significant sponge activity. Therefore, three critical parameters require rigorous evaluation: (1) the number of MREs per circRNA molecule, (2) the stoichiometric ratio of circRNAs to miRNAs, and (3) the expression levels of the miRNA-targeted genes. Notably, sponging activity fundamentally depends on the cytoplasmic colocalization of circRNAs and their target miRNAs.
CircRNA interact with DNA
R-loops are three-stranded nucleic acid structures comprising an RNA:DNA hybrid and single-stranded DNA that contribute significantly to DNA damage induction, genomic instability modulation, and transcriptional control [43]. Interestingly, circRNAs can form circRNA:DNA hybrids (circR loops) that critically influence malignant tumour phenotypes. A recent study demonstrated that circRNAs are enriched within leukaemia-rearranged (MLL-r) AML and can bind with DNA to form circR loops at their cognate loci [44]. In particular, circMLL(9,10) has demonstrated oncogenic potential by inducing proteasome inhibition, triggering DNA breakage, and promoting chromosomal translocation, thereby driving AML pathogenesis in vitro and in vivo [44]. Similarly, Xu et al. reported that circSMARCA5, which is expressed at reduced levels in breast cancer, forms circR loops at its parental gene locus [45]. The functional restoration of circSMARCA5 blocks SMARCA5 transcription, impairs DNA damage repair capacity, and enhances cisplatin sensitivity, suggesting that circSMARCA5 may serve as a therapeutic target in breast cancer, especially in patients with drug-resistant disease [45]. Overall, the function of circR loops in cancer is undeniably important and is usually mediated via the induction of DNA damage and genomic instability or transcription regulation. Despite these advances, the field of circR loop biology remains in its nascent exploration phase, with numerous relevant circRNAs awaiting characterization.
CircRNA interact with proteins
Regulating the functions of RBPs
Emerging evidence has revealed that circRNAs interact with RBPs to regulate gene expression. For example, circMYBL2 enhances the efficiency of FLT3 kinase translation by facilitating polypyrimidine tract-binding protein 1 (PTBP1) binding to FLT3 mRNA, thereby promoting proliferation and inhibiting differentiation in FLT3-ITD-positive AML cells [46]. In cervical cancer, circTICRR is upregulated and exerts oncogenic effects by inhibiting autophagy activation through binding to HuR and stabilizing GLUD1 mRNA [47]. Moreover, circBACH1 can directly interact with HuR and alter its translocation from the nucleus to the cytoplasm, thereby inhibiting p27 mRNA translation via recognition of its interferon-responsive sequence in the 5’-untranslated region, ultimately promoting proliferation in hepatocellular carcinoma cells [48]. Overall, these findings demonstrate that circRNAs modulate RBP subcellular localization and functional activity through specific molecular interactions.
Recruiting transcription activators
In this section, we will present three examples that illustrate the roles of circRNAs in recruiting transcriptional activators. In hepatocellular carcinoma, the highly abundant circRNA cia-MAF recruits the histone acetyltransferase complex (TIP60) complex to the MAFF promoter, initiating transcriptional activation that sustains liver tumour-initiating cell (TIC) self-renewal [49]. Similarly, circACTN4 facilitates Y-box binding protein 1 (YBX1) recruitment to activate FZD7 transcription and then activates the Wnt and Hippo signalling pathways, thereby promoting intrahepatic cholangiocarcinoma (ICC) growth and metastasis [50]. Furthermore, circ-DONSON recruits the nucleosome remodelling factor (NURF) complex to the SOX4 promoter, activating oncogenic transcription programs that increase gastric cancer cell malignancy [51]. These paradigms collectively demonstrate that circRNAs recruit specific activator complexes to target gene promoters and thereby modulate downstream oncogenic pathways.
Acting as protein scaffolds
Emerging evidence demonstrates the capacity of circRNAs to orchestrate protein complex assembly through ternary interactions. One pioneering study reported that circACC1 combines with the AMPK β/γ subunits to form a ternary complex, enhancing the stability and catalytic activity of the AMPK holoenzyme [52]. Another study indicated that circ-LRIG3 assembles a circ-LRIG3–EZH2–STAT3 ternary complex that facilitates EZH2-mediated STAT3 methylation and phosphorylation, eventually activating STAT3 signalling [53]. Additionally, Du et al. revealed that in noncancerous cells, circ-Foxo3 forms a ternary complex with CDK2 and p21 that blocks the function of CDK2, ultimately inducing cell cycle arrest and suppressing proliferation [54]. The above examples indicate that circRNAs function as molecular scaffolds to perform three critical functions: (1) enzymatic activity modulation, (2) protein complex stabilization, and (3) spatial coordination of protein‒protein interactions.
Interacting with specific proteins
Nuclear circPDIA4 competitively binds the RNA helicase DHX9, disrupting its interaction with target RNAs and thereby enhancing DHX9-dependent circRNA biogenesis and accelerating gastric cancer progression [55]. Moreover, circMTCL1 interacts with C1QBP protein and augments C1QBP translational output by inhibiting ubiquitin-proteasomal degradation [56]. In cervical cancer, circVPRBP overexpression strongly represses lymph node metastasis by interacting with RACK1 and shielding its S122 O-GlcNAcylation site to accelerate RACK1 degradation [57]. Taken together, these findings indicate that circRNAs can interact with specific proteins to change their routine biological functions or influence downstream biological processes.
Several controversies
With regard to the effect of circRNA-protein binding, the abundance of circRNAs should be considered, as was argued above for miRNA sponging mechanisms. In addition, RNA immunoprecipitation (RIP), RNA pull-down, and colocalization analyses to validate these interactions are essential. Notably, the secondary and tertiary structures of proteins and circRNAs may also affect their affinities, although this aspect has not been thoroughly examined in current research.
Translating proteins or peptides
A pivotal breakthrough in 2017 emerged from three independent studies demonstrating m6A-dependent and IRES-mediated translation mechanisms in circRNAs [58,59,60]. A growing number of tumour-related circRNAs have subsequently been identified to possess protein-coding potential. For example, circZKSCAN1 encodes a 206-amino-acid polypeptide through an IRES-driven open reading frame (ORF) to promote the ubiquitination of mTOR, thereby inhibiting the PI3 K/AKT/mTOR pathway in hepatocellular carcinoma [61]. In addition, circARHGAP35 contains an ORF with an m6A-modified initiation codon that encodes a truncated protein that contributes to cancer progression [62]. Moreover, m6A modifications of circ-ZNF609, which was previously proven to have protein-coding ability owing to its IRES activity, can accelerate its IRES-mediated translation [63]. These discoveries establish two non-mutually exclusive translation initiation mechanisms, namely, m6A-dependent translation and IRES-mediated translation. These findings fundamentally expand our understanding of the mechanisms governing circRNA-derived proteins and polypeptides.
CircRNA profiles in AML
The continuous evolution of RNA sequencing and microarray technologies has revolutionized transcriptome-level gene expression analysis in AML, enabling the systematic identification of functional circRNAs. Microarray assays and next-generation sequencing (NGS) are the most widely applied methods for circRNA research.
Notably, several novel technologies, such as single-cell RNA sequencing [64], Nanopore sequencing [65,66,67], and electrochemical detection [68, 69], have been applied to increase sequencing throughput, circRNA capture efficiency, and detection specificity.
To determine whether circRNAs are involved in AML progression, extensive studies have been conducted to compare circRNA expression between AML patients and healthy controls [70,71,72,73,74,75,76,77]. These studies consistently demonstrate widespread dysregulation of circRNAs in AML [70,71,72,73,74,75,76,77]. Notably, Ding et al. reported that differentially expressed circRNAs in AML are enriched in biological processes including cell proliferation, migration, and response to drugs and are closely associated with protein binding, ATP binding and RNA binding functions [74]. KEGG pathway analysis further revealed that these circRNAs are involved mainly in ErbB signalling, EGFR tyrosine kinase inhibitor resistance and mTOR signalling pathways, all of which are related to the development of AML [74]. Moreover, three other studies reported that AML-associated circRNAs are primarily exon-derived and show chromosomal distribution biases, with high frequencies on chromosomes 1, 2, 6, and 16 and minimal representation on chromosomes 13 and 21, and in the mitochondrial DNA [71, 72, 76]. These unbalanced distribution patterns suggest that circRNAs from chromosomes 1, 2, 6, and 16 may preferentially regulate AML progression. Growing evidence has revealed that circRNAs are strongly implicated in malignancy-related behaviours and treatment response [78,79,80,81,82]. Lv et al. identified 512 differentially expressed circRNAs (253 upregulated, 259 downregulated) between samples from AML with and without extramedullary infiltration (EMI) [78]. Through network analysis, these authors mapped a circRNA‒miRNA‒gene interaction atlas and pinpointed 17 circRNAs associated with migration, adhesion, signal transduction and cell‒cell communication, suggesting that they are likely responsible for EMI [78]. In addition, Li et al. discovered 1824 dysregulated circRNAs in adriamycin-resistant AML cells that were predominantly linked to B/T-cell receptor signalling, MAPK signalling, and mTOR signalling [81]. These results suggest that circRNAs play pivotal roles in the malignant progression of AML.
Gene mutations, including Nucleophosmin (NPM1), FMS-like tyrosine kinase 3 (FLT3), and splicing factor mutations (e.g., SF3B1, SRSF2, U2 AF1), are recognized as key drivers of AML pathogenesis [83, 84]. Notably, circRNA expression profiles are obviously influenced by genetic alterations [46, 84, 85]. For example, comparative analysis of haematopoietic stem/progenitor cells (HSPCs) from AML patients and healthy controls revealed 124 dysregulated circRNAs in patients with NPM1 mutations and 42 dysregulated circRNAs in patients with splicing factor mutations [84]. Two other studies have corroborated these findings [46, 85].
Emerging studies have demonstrated that m6A modifications are prevalent in numerous circRNAs and regulate 5’-cap-independent translation and selective degradation [37, 58,59,60]. However, the biological function of m6A-modified circRNAs remains poorly understood. To address this, Issah et al. conducted a circRNA epitranscriptomic microarray assay in AML, identifying 1136 differentially expressed m6A-modified circRNAs between AML patients and healthy controls [86]. Among these genes, 1057 were upregulated, and 79 were downregulated [86]. Subsequent GO annotation and pathway analysis implicated these circRNAs in AML tumorigenesis [86].
In this section, we summarize the circRNA profiling studies that have been performed in AML to date (Table 1). While circRNAs are broadly dysregulated in AML, only a limited fraction of them have been explored. We expect that our summary will serve as a valuable resource for further investigations.
Online databases and tools for circRNA exploration
Research on circRNAs is still at a preliminary stage, with their functions and regulatory networks remaining incompletely understood. To bridge this knowledge gap, various specialized databases and analytical tools that provide circRNA profiles and annotations, assess their protein-coding potential and predict their potential functions have been developed. In this section, we summarize open-access platforms for circRNA exploration (Tables 2, 3, 4), highlighting their unique capabilities and applications.
There are already several databases containing circRNA profiles associated with various diseases, such as GEO [87], circRNADisease v2.0 [88], deepBase v3.0 [89], CircAtlas 3.0 [90], CircSC, CSCD 2.0 [91], CircNet 2.0 [92], exoRBase v2.0 [93], MiOncoCirc [94], Circ2Disease [95], TSCD [96], and CircRic [97]. These databases help users browse, search, and download information related to circRNAs. Notably, CircAtlas also provides an ID conversion service that can convert IDs from different circRNA databases [90]. In addition, circSC integrates a substantial number of full-length single-cell RNA-sequencing datasets, including a total of 196,491 human and 310,969 mouse circRNAs, and provides information on the specific expression patterns of circRNAs in different cells and samples. CircRic provides circRNA expression profiles in 935 cancer cell lines across 22 cancer lineages from the Cancer Cell Line Encyclopedia (CCLE) [97]. CIRI [28], CIRCexplorer2 [98], circRNA_finder [99], find_circ [100], DCC [101], and NCLscan [102] are commonly used tools for identifying circRNAs in large-scale RNA sequencing data. Additionally, circBase [103], CircBank [104], circVAR [105], and circRNADb [106] contain detailed annotations of circRNAs. Notably, the circVAR database provides resources for identifying circRNA-related genetic variants in healthy and diseased populations and allows users to quickly search for genetic variants in circRNAs and download all annotated variants [105].
CircRNAs can serve as miRNA sponges, interact with DNA or proteins, and encode proteins. The most likely mode of action can be predicted via bioinformatics tools. For example, CircAtlas 3.0 [90], CSCD 2.0 [91], CircNet 2.0 [92], circRNADb [106], ORF-FINDER [107], CircPro [108], IRESfinder [109], and IRESbase [110] can predict the protein-coding potential of circRNAs on the basis of the ORFs and/or IRESs present. CircCode, a Python 3-based framework for recognizing translatable circRNAs in ribo-seq reads, is also a powerful and convenient research tool [111]. Recent studies have demonstrated that m6A modification of circRNAs plays important roles in driving translation and mediating degradation [37, 58,59,60]. Therefore, predicting m6A modification sites in circRNAs is also essential. CircBank [104], TransCirc [112], and CircPrimer 2.0 [113] provide evidence of circRNA features related to translation, including ORFs, IRESs, and m6A modifications. Notably, in addition to the above services, TransCirc can perform integrative analysis to predict the coding potential of a circRNA and provides ribosome/polysome profiling, translation initiation site, sequence composition, and proteomics evidence from mass spectrometry while allowing users to search by circRNA sequence [112]. CircPrimer 2.0 can help researchers design primers for circRNAs and determine their specificity [113]. In addition, SRAMP3 serves as a useful tool to predict m6A modification sites in RNA sequences of interest [114]. Regarding other mechanisms of circRNA functions, some databases provide guidance for future research. For example, CSCD 2.0 [91], CircNet 2.0 [92], Circ2Disease [95], CircRic [97], CircBank [104], CircInteractome [115], CircFunBase [116], ENCORI [117], and TRCirc [118] provide interactions between circRNAs and miRNAs or RBPs. Interestingly, CircRic also provides information related to the regulators of circRNA biogenesis, the effect of circRNAs on drug response, and the associations between circRNAs and mutations [97]. CircBank also provides circRNA mutation information [104]. In addition, CircInteractome can be used to design divergent primers and siRNAs for circRNAs [115]. Notably, TRCirc not only contains information related to the transcriptional regulation of circRNAs but also provides related information such as data on enhancers, methylation, H3 K27ac modification and circRNA expression [118].
In conclusion, these databases and tools provide convenient platforms that allow researchers to explore the expression atlases, basic information and potential functions of circRNAs. However, predictions derived from databases require experimental validation. In addition, the lack of standardized nomenclature for circRNAs complicates database searches. Moreover, in contrast to the case for mRNAs, miRNAs, and lncRNAs, clinical databases specifically dedicated to circRNAs remain underdeveloped. Furthermore, current databases lack information on circR loops.
Roles of circRNAs in AML
Recent studies have demonstrated that circRNAs play pivotal roles in the initiation and progression of haematological malignancies, especially AML. In the following section, we describe the biological roles of circRNAs in cell proliferation, apoptosis, the cell cycle, differentiation, migration, invasion, extramedullary infiltration, ferroptosis, autophagy, stemness, drug resistance, exosomes and the tumour microenvironment in AML (Table 5, Fig. 2).
CircRNAs affect AML cell proliferation and apoptosis
Cancer cells, including AML, need to evade apoptosis in order to continue proliferating. Accumulating evidence indicates that circRNAs are highly involved in regulating cell proliferation and apoptosis. Specifically, some circRNAs, such as hsa_circ_0079480, f-circPR, and f-circM9, promote AML cell proliferation and suppress apoptosis [46, 72,73,74,75,76,77, 84, 119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150], whereas others, such as hsa_circ_0121582, hsa_circ_0001947, and hsa_circ_0003420, play diametrically opposite roles [151,152,153,154,155,156,157]. A detailed description of each circRNA is shown in Table 5. Mechanistically, circRNAs mainly exert their effects via circRNA–miRNA–mRNA networks, and few act by binding with proteins. For example, hsa_circ_0079480 can modulate AML proliferation and apoptosis through the miR-654-3p/HDGF axis [119]. Nuclear hsa_circ_0121582 can bind to the GSK3β promoter and recruit the DNA demethylase TET1 to GSK3β, thus potentiating the transcription of GSK3β and eventually suppressing cell proliferation [151]. Moreover, f-circM9 and f-circPR, which are derived from the MLL/AF9 and PML/RARα fusion genes, respectively, can also regulate AML cell proliferation and apoptosis, but the underlying mechanism remains unclear [120]. In addition, Conn et al. demonstrated that circR loops could promote AML initiation and progression both in vitro and in vivo through transcriptional pausing, proteasome inhibition, chromatin reorganization, and DNA breakage [44]. Thus, the functions of circRNAs in AML cell proliferation and apoptosis are becoming increasingly clear in the modern era of molecular biology.
Cell cycle-associated circRNAs in AML
Cell cycle acceleration is a common feature of AML, leading to uncontrolled cell division and rapid proliferation [158]. Emerging evidence suggests that circRNAs are deeply involved in regulating the cell cycle progression of AML cells, leading to cell cycle acceleration or arrest [46, 73, 75,76,77, 122, 123, 125,126,127,128,129,130,131, 133,134,135,136,137,138,139, 141, 142, 147,148,149, 151, 153, 154]. Usually, circRNAs affect cell cycle progression by controlling cell cycle regulators such as cyclins, cyclin-dependent kinases (CDKs), and proliferating cell nuclear antigen (PCNA). For example, circ_POLA2 silencing causes the downregulation of CDK4 and CDK6, leading to G1/G0 cell cycle arrest in AML cells [122]. Circ_0094100 deficiency suppresses the protein levels of cyclin D1 and PCNA in rapamycin-treated AML cells and restrains the cell cycle [125]. Circ_0040823 sponges miR-516b to relieve the inhibitory effects of PTEN in AML, thereby inhibiting the expression of cell cycle-related proteins such as cyclin D1 and cyclin E1 and increasing the percentage of cells in the G0/G1 phase [154]. These results indicate that circRNAs play prominent roles in cell cycle regulation and that targeting circRNAs that function as negative regulators of the cell cycle may be a useful therapeutic strategy for AML.
Cellular differentiation-related circRNAs in AML
Terminal differentiation block is one of the hallmarks of AML and results in the production of immature cells, ultimately leading to severe anaemia, infection, and bleeding [159]. Studies have shown that circRNAs participate in regulating AML differentiation. For example, circMYBL2, circSPI1, circSH3BGRL3 and circRNF220 are considered suppressors of differentiation in AML [46, 76, 132, 133]. In contrast, circ_0001187 and circ-HIPK2 promote AML cell differentiation [79, 156]. Mechanistically, most circRNAs function by sponging miRNAs. Due to the nonselective cytotoxicity of chemical agents towards both malignant and normal cells, differentiation-inducing therapy has emerged as a novel approach for treating AML with improved safety and efficacy. Therefore, circRNAs can serve as new targets for anticancer therapy in AML.
CircRNAs modulate invasion, migration, and extramedullary infiltration in AML
Invasion, migration, and extramedullary infiltration (EMI) are malignant behaviours that often result in a high mortality rate and poor prognosis in AML patients. CircRNAs play key regulatory roles in cell invasion and migration, and the majority of related circRNAs, such as circRNF220, circRNF13, and CircRAD18, act by sponging miRNAs to interfere with important regulators of the abovementioned processes [135,136,137,138, 141,142,143,144,145,146,147, 155, 157]. EMI is associated with poor prognosis in AML patients owing to the associated destruction of vital organs. To further understand the expression profiles of circRNAs in AML with EMI, Lv et al. performed circRNA microarray analysis, including the construction of a circRNA‒miRNA‒mRNA regulatory network [78]. Seventeen circRNAs were identified as closely associated with EMI, but the exact mechanisms of the functions of those circRNAs remain unknown and require further investigation [78].
CircRNAs regulate ferroptosis in AML
Ferroptosis is a recently identified type of cell death caused by iron-dependent lipid peroxidation, and targeting ferroptosis provides a new and promising approach for antitumour therapies [160]. In AML, Long et al. reported that circZBTB46 enhances the expression of stearoyl-CoA desaturase 1 (SCD), likely by acting as a miRNA sponge, thereby protecting AML cells from ferroptosis and promoting cell proliferation [77]. Moreover, circKDM4 C was reported to promote ferroptosis via the hsa-let-7b-5p/P53 axis [157]. Nevertheless, ferroptosis-related circRNAs have rarely been reported in AML and still require further characterization.
Autophagy-related circRNAs in AML
Autophagy, an intracellular lysosome-dependent catabolic pathway, promotes tumour cell survival in response to multiple antitumour drugs, while sustained activation of autophagy can cause cell death [161, 162]. Shang et al. demonstrated that circPAN3 might facilitate AML resistance to doxorubicin through activating autophagy and the AMPK/mTOR signalling pathway [80]. Another study revealed that hsa_circ_0035381 deficiency could reduce autophagy levels and inhibit AML cell proliferation by regulating the miR-582-3p/YWHAZ axis in AML [149]. Currently, autophagy is considered a double-edged sword in tumours that can either promote cell survival or enable apoptosis [163]. However, reports on autophagy-related circRNAs in AML are scarce, and additional relevant circRNAs need to be identified. Targeting autophagy-related circRNAs may provide new strategies for AML treatment, especially for patients with drug-resistant disease.
CircRNAs affect the stemness of leukaemia stem cells in AML
Cancer stem cells (CSCs) are considered the main cells responsible for tumour initiation, development, recurrence, metastasis and radiotherapy and chemotherapy failure [164]. Research by Lin et al. revealed that hsa_circ_0003420 was expressed at lower levels in non-M3 AML stem cells than in normal haematopoietic stem cells [165]. Its overexpression impaired the stemness of leukaemia stem cells and inhibited the expression of stemness‐related genes (HOXB4, MYB, ALDH1 A1, ABCB1, CD34, and MMRN1) [165]. However, there is a lack of related research in AML, and more studies are needed.
CircRNAs regulate drug resistance in AML
To date, both conventional chemotherapeutic agents (e.g., Cytarabine, Adriamycin, and Daunorubicin) and novel targeted drugs (e.g., Venetoclax, Quizartinib, and Ivosidenib) for the treatment of AML have encountered drug resistance as an obstacle. Therefore, a better understanding of drug resistance mechanisms is vital for improving patient outcomes. Interestingly, numerous studies have demonstrated that circRNAs are involved in the regulation of drug resistance in AML. Two of those studies revealed that circPAN3 contributed to resistance to Adriamycin (Doxorubicin) through the regulation of autophagy and through the miR-153-5p/miR-183-5p-XIAP axis [80, 166]. In addition, circNPM1 and circEHBP1 were reported to be dysregulated in AML and involved in Adriamycin resistance by sponging miR-345-5p and miR-129, respectively [147, 167]. In addition, circMYBL2 induces resistance to quizartinib, a potent and highly selective FLT3 inhibitor, in FLT3-ITD+ AML cells by activating FLT3 kinase-dependent signalling pathways [46]. F-circM9 confers resistance to arsenic trioxide and cytarabine in AML [120]. Circ-ANXA2 and circSH3BGRL3 increase the chemosensitivity of AML cells to cytarabine and/or daunorubicin by regulating their target miRNAs [74, 133]. These results indicate that circRNAs play essential roles in drug resistance and that targeting circRNAs may be a promising treatment strategy for preventing drug resistance.
Exosomal circRNAs and the AML microenvironment
The constant crosstalk between AML cells and their microenvironment affects tumour initiation and progression [168]. Exosomes are membranous vesicles secreted by virtually every type of living cell with an average diameter of ~ 100 nm. These vesicles are crucial executors of intercellular signalling and are also closely connected with the malignant behaviour of tumours [169, 170]. Notably, exosomal circ_001264, hsa_circ_0009910, and circ_0004136 are expressed at high levels in AML and play oncogenic roles in modulating AML cell behaviour [141, 148, 150]. Among them, exosomal circ_001264 can activate p38-STAT3 signalling to induce M2 macrophage polarization, thereby upregulating PD-L1 expression [150]. In addition, exosomal circ_001264 siRNA has been shown to inhibit AML tumorigenicity. PD-L1, a PD-1 ligand, interacts with PD-1 on the T-cell surface to attenuate T-cell activation and facilitate immune escape [171]. The coadministration of exosomal circ_001264 siRNA, anti-PD-L1 therapy, and cytarabine obviously increases antitumour activity in AML mouse models [150]. However, the mechanisms through which exosomal hsa_circ_0009910 and circ_0004136 function in the AML microenvironment still need to be explored [141, 148]. Taken together, these findings suggest that exosomal circRNAs play essential roles in regulating the malignant behaviours of tumour cells and cell-to-cell communication within the microenvironment and that interfering with circRNA expression may be an effective anticancer strategy. However, only a few exosomal circRNAs have been identified in AML, and their functions remain to be investigated further.
Clinical application of circRNAs in AML
Early diagnosis and timely treatment are highly important for improving cure rate and prognosis in patients with tumours. However, the current methods for the clinical diagnosis of tumours, such as tissue biopsy, endoscopy examination, and MRI, are often invasive, expensive, and time-consuming. The development of simple, minimally invasive, and relatively inexpensive approaches is essential to support early diagnosis. Moreover, the early identification of poor prognostic factors and timely delivery of targeted therapeutic interventions also improve clinical outcomes. Recent studies have demonstrated that certain circRNAs are closely associated with clinicopathologic features and possess great potential as effective biomarkers for diagnosis and prognosis, as well as therapeutic targets in AML (Fig. 3).
Correlations between circRNA expression and clinicopathological characteristics in AML patients
In this section, we summarize the correlations between circRNA expression and clinicopathological characteristics in patients with AML according to the findings of recent studies (Table 6). For example, circ_0001187 is significantly decreased in older AML patients (aged > 43 years) [156]. AML patients with high circSMC1 A expression are more likely to be female [172]. CircEHBP1 is closely associated with French–American–British (FAB) classification [167]. Moreover, the expression levels of hsa_circ_001264, hsa_circ_0001947, circ_0001187, and circKLHL8 are inversely related to the percentage of blasts in the bone marrow (BM) or peripheral blood (PB), whereas circ-ANAPC7 and circ-PVT1 are positively related [150, 152, 156, 172,173,174]. High expression of hsa_circ_0001947, circ_0001187, and circFCHO2 and low expression of circ-ANAPC7 and hsa_circ_0079480 are correlated with a low white blood cell (WBC) count [152, 156, 172, 173, 175]. Circ_0001187 has a significant positive association with the platelet (PLT) count [156]. The levels of circTASP1, hsa_circ_0001947, and circ_0001187 are positively related to haemoglobin (HGB) levels [75, 152, 156]. Gene mutations are crucial events in AML pathogenesis [83]. Accumulating evidence suggests that circSMC1 A, circKLHL8, circFCHO2, circCFLAR, and circ-PVT1 are closely linked to mutations in genes such as NPM1, FLT3-ITD, WT1, or CEBPA [172, 174]. Overall, circRNA expression levels are strongly associated with clinicopathological features in AML patients.
CircRNAs as diagnostic biomarkers in AML
Several circRNAs have been reported to have diagnostic value in AML (Table 7). For example, Lin et al. revealed that circPLXNB2 was obviously elevated in BM samples from patients with AML and was valuable for distinguishing AML patients from healthy individuals (AUC = 0.8525) [138]. Other circRNAs in BM can also serve as diagnostic biomarkers, such as hsa_circ_0004277 (AUC = 0.957), circ-ANXA2 (AUC = 0.832), hsa_circ_0044907 (AUC = 0.9447), circ-ANAPC7 (AUC = 0.915), and circ_0059706 [70, 74, 77, 129, 152, 173, 174, 176, 177]. Although BM aspiration and biopsy are still the gold standard for diagnosing AML, these tests are invasive and cause physical trauma to patients. Moreover, repeated sampling is necessary during the course of treatment, leading to recurrent trauma. In contrast, PB collection and analysis is simpler, more cost-effective and less invasive. Studies have shown that circZBTB46 (AUC = 0.830) and hsa_circ_0079480 (AUC = 0.9342) in PB are valuable biomarkers for AML diagnosis [77, 175]. In addition, circRNAs have been verified to be enriched in serum exosomes and have implications for early tumour diagnosis [178]. Circ_0004136 and hsa_circ_0009910 were reported to be highly expressed in exosomes secreted by AML cells, but regrettably, their diagnostic value was not specifically assessed by the study authors [141, 148]. Exosome detection has emerged as a promising method for liquid biopsy in tumour diagnosis with the advantage of minimal invasiveness [179]. However, research on exosomal circRNAs in AML is relatively scarce. In brief, we hope that circRNAs can serve as effective biomarkers for AML diagnosis in the future.
CircRNAs as prognostic biomarkers in AML
Clinically, high recurrence rates and poor prognoses remain challenges in AML patients, especially high-risk AML patients. How to accurately stratify patients by risk profile and predict the probability of relapse at the initial visit has been a matter of intense discussion for decades. Here, we summarize several circRNAs that have significant prognostic value in AML (Table 8). For example, circ-ANXA2 is highly expressed in AML, and patients with higher circ-ANXA2 levels exhibit shorter overall survival (OS) and event-free survival (EFS), poorer risk profiles, and a lower probability of complete remission (CR) [74]. Circ_0012152 expression is significantly increased in AML patients compared with individuals without AML, and high expression of circ_0012152 is strongly associated with poor prognosis [134]. Interestingly, circ_0012152 levels were decreased in patients who achieved CR but increased again in patients who experienced relapse, indicating the great potential of circ_0012152 as a biomarker for dynamically monitoring relapse. Additionally, higher circ-PVT1 expression predicts poor outcome in AML patients, specifically, shorter OS, EFS and relapse-free survival (RFS) [146, 174]. Similarly, other circRNAs, such as hsa_circ_0001990, circTASP1, and circ-PTK2, have prognostic value in AML [73, 75, 121, 129, 138, 144, 150, 154, 172, 175, 177]. Detailed relevant information about these circRNAs is listed in Table 8. In general, circRNAs are helpful markers for evaluating the prognoses of AML patients.
CircRNAs as therapeutic targets or agents in AML
Drug resistance and disease recurrence remain the major obstacles in AML therapy. The identification of novel therapeutic targets and optimization of treatment strategies are urgently needed to improve the clinical outcomes of AML patients. Due to their extensive regulatory roles in various cellular processes, circRNAs are hypothesized to be valuable potential therapeutic targets, and interference with circRNA expression may be a promising avenue for treating cancer.
Considering that many circRNAs are upregulated in AML, RNA-based strategies for circRNA knockdown, such as RNA interference (RNAi), antisense oligonucleotide (ASO), and CRISPR/Cas approaches, are considered particularly suitable treatment methods because they can be delivered directly to the bloodstream [180,181,182,183]. Currently, RNAi molecules can be artificially designed and synthesized in the laboratory and delivered to cells via lipid nanoparticles, exosomes, polymers and other appropriate materials [184, 185]. In AML, several mouse models with circRNA deficiency (e.g., circPLXNB2, circ_0035381, and circ_0001187) have been established using RNAi technology to verify the functions of these circRNAs [138, 149, 156]. Nonetheless, rapid degradation, low intracellular delivery efficiency, immune responses and off-target effects remain to be overcome in practice [182]. Compared with RNAi, ASOs have the advantages of better cleavage efficiency and fewer off-target effects [181]. The CRISPR/Cas system is a powerful genome-editing tool that effectively impedes circRNAs biogenesis [186]. Zheng et al. revealed that silencing of circHIPK3 through the CRISPR/Cas9 system strongly inhibited human cell growth [187]. Gu et al. reported that circIPO11 deficiency induced using CRISPR/Cas9 technology apparently suppressed the progression of chemically induced liver carcinogenesis [188]. Notably, recent studies have demonstrated that CRISPR-Cas13 systems can effectively discriminate circRNAs from their cognate mRNAs and increase their silencing efficiency, which may serve as a useful tool for the functional study of circRNAs [180, 189].
For circRNAs that are downregulated in AML, overexpression can be achieved by cloning the circRNA into lentivirus or adeno-associated virus (AAV) vectors and conjugating the vector with nanoparticles or lipid carriers to drive cell type-specific expression [190,191,192,193]. For example, using recombinant AAV9 vectors, Zeng et al. constructed a circMap3k5-overexpressing mouse model to determine the ability of circMap3k5 to alleviate intimal hyperplasia [193]. Moreover, Meganck et al. developed recombinant AAV vectors carrying transgene cassettes with intronic sequences and verified their ability to promote circRNA expression in organs such as the heart, liver and brain in mice [190]. This study highlights the possibility of precise interventions targeting circRNAs in specific tissues to improve therapeutic outcomes. However, whether linear byproducts generated during circRNA overexpression exert detrimental effects on cells requires further investigation.
CircRNAs can interact with miRNAs or proteins and subsequently participate in regulating AML pathology [22, 23]. Taking advantage of circRNAs to target suppressive/oncogenic miRNAs or proteins may contribute to AML therapy. For protein-coding circRNAs, strategies such as antibody-mediated targeting or IRES insertion upstream of ORFs may provide novel therapeutic avenues [62, 194]. Taken together, these findings imply that altering circRNA expression levels may provide new strategies for AML treatment.
Moreover, circRNA vaccines also show great promise for AML therapy. Compared with normal mRNA vaccines, circRNA vaccines produce higher concentrations of antigens for a longer time because of their greater stability [195]. Qu et al. demonstrated that a novel circRNA vaccine encoding the antigen of SARS-CoV-2 effectively promoted immune activation in mice and rhesus macaques upon infection with SARS-CoV-2 [195]. In the area of cancer research, Li et al. encapsulated the synthetic circRNAOVA−luc, which encodes the restricting H2-Kb peptide OVA 257–264 and luciferase, into lipid nanoparticles to construct a circRNA vaccine and verified its antitumour effect in a variety of tumour-bearing mouse models, including colorectal carcinoma, orthotopic melanoma, and lung metastasis melanoma mouse models [196]. This circRNA vaccine triggered robust innate and adaptive antitumour immune activation in multiple mouse tumour models and showed superior antitumour efficacy [196]. Huang D et al. reported that vaccination with circFAM53B efficiently elicited antitumour immunity in an antigen-specific manner by encoding cryptic peptides and significantly inhibited tumour growth in a B16 F10 mouse melanoma model [197]. Although there is a lack of relevant studies on circRNA vaccines in AML, we believe that significant advances will be made in the coming years.
Clinical application prospects of circRNAs in AML
Although noncoding RNAs (ncRNAs) were previously considered noise in genomic transcription, their functions have become popular research topics in recent years, paving the way for their clinical application; more than 1000 miRNA-related clinical trials and more than 100 lncRNA-associated clinical trials have been registered in the ClinicalTrials.gov database (https://www.clinicaltrials.gov/). However, no circRNA-related clinical trials were found in this database. Several miRNA mimics or inhibitors have successfully entered clinical trials. For example, MRG-106, an inhibitor of miR-155, exhibits excellent antitumour efficacy without serious adverse reactions in diffuse large B-cell lymphoma (Registration ID: NCT02580552) [198]. Moreover, a miR-34a mimic (MRX34) [199], a miR-16 mimic (TargomiR) [200], and a miR-193-3p mimic (INT-IB3) have also been tested in clinical trials. For lncRNAs, clinical treatment approaches involving targeting lncRNAs are still lacking, although their possible use as tumour biomarkers has gained more attention. For example, Fayoum University recently completed a clinical trial that explored the clinical utility of the salivary expression of the lncRNA MALAT1 in the diagnosis of oral squamous cell carcinoma (Registration ID: NCT05708209). Assiut University conducted a clinical trial to evaluate the relationship between lncRNA CCAT1 and tumour staging in patients with colorectal cancer and its diagnostic value (Registration ID: NCT04269746). In addition, a clinical trial at Strasbourg University Hospital is recruiting volunteers to investigate the prognostic value of the lncRNA MFI2-AS1 in localized clear cell kidney cancers (Registration ID: NCT04946266).
Although this field of research is in its nascent stage, recent studies have demonstrated that circRNAs are characterized by high abundance, relative stability, and evolutionary conservation and are closely related to the development and progression of various diseases, making them ideal biomarkers for tumour diagnosis, prognostic assessment, and therapy [14, 16, 17, 20, 22, 23]. Nevertheless, no circRNAs are yet implemented in clinical practice, and we believe that major developments can be anticipated in the future. However, limited circRNA-specific target sites, low delivery efficiency, poor specificity and tolerability, toxicity and off-target effects are still major obstacles to the clinical application of circRNAs. Overall, inhibiting the activity of oncogenic circRNAs or overexpressing tumour-suppressor circRNAs can be beneficial treatment approaches, but some technical limitations and challenges still exist.
Recommendations and future perspectives
Although we have gradually elucidated the specific functions of circRNAs, our current understanding may represent just the tip of the iceberg, and numerous issues still need to be addressed to move the field forwards. First, there is still no generally established consensus for circRNA nomenclature, and there is a lack of universal and comprehensive circRNA-associated public databases. These problems make it difficult for investigators to obtain exact genomic locations and detailed information from databases using circRNA names. Second, superior detection methods, such as single-cell spatial noncoding transcriptomics, nanopore-based sequencing, and electrochemical detection techniques, are needed to identify and quantify circRNAs. Third, the off-target effects of circRNA knockdown must be considered carefully due to the high sequence similarities between circRNAs and their cognate mRNAs. Recent studies have demonstrated that CRISPR–Cas13 systems can effectively discriminate circRNAs from their cognate mRNAs and increase their silencing efficiency, making these systems useful tools for the functional study of circRNAs [180, 189]. Moreover, the inefficiency of circularization when circRNAs are overexpressed is an inevitable problem. Fourth, most circRNA research in AML has focused on the role of circRNAs as miRNA sponges, but the specific underlying mechanisms remain to be further elucidated, and other mechanisms also deserve investigation. Fifth, the functions and downstream mechanisms of circRNAs have attracted much attention, but the modes of circRNA biogenesis, spatial structure, transportation, degradation, and chemical modifications, in addition to m6A modifications, have been much less well studied. Sixth, circRNAs related to stem cell phenotype or function have received less attention. Seventh, the clinical translation of circRNA-based therapies still faces challenges. As a whole, research on circRNAs remains in its infancy, and associated limitations and challenges need to be addressed.
Conclusions
AML is a challenging and biologically complex disease that is driven, in part, by genetic mutations, heterogeneous clones and epigenetic alterations, and our current knowledge of its pathogenesis is insufficient [201, 202]. Previously, circRNAs were considered functionless byproducts of RNA mis-splicing [203, 204]. However, circRNAs are now considered emerging molecular regulators of various physiological and pathological processes. Many circRNAs with great physiological and clinical significance have been identified to be specifically expressed in AML, making them attractive candidate diagnostic, prognostic and therapeutic targets [22, 23]. Herein, we have summarized the biogenesis, categories, degradation, regulatory mechanisms and functions of circRNAs, with an emphasis on elucidating dysregulated circRNAs in AML and their clinical implications. Moreover, we outlined a series of online databases and tools for circRNA exploration, which can provide important guidance for subsequent studies. Although several key challenges remain, we believe that circRNAs may be developed as clinical diagnostic and prognostic markers, therapeutic targets, and even RNA drugs in the future.
Availability of data and materials
No datasets were generated or analysed during the current study.
Abbreviations
- AML:
-
Acute myeloid leukaemia
- circRNA:
-
Circular RNA
- Bcl-2:
-
B-cell lymphoma-2
- FLT3:
-
FMS-like tyrosine kinase-3
- IDH:
-
Isocitrate dehydrogenase
- CAR-T:
-
Chimeric antigen receptor-modified T
- pre-mRNAs:
-
Precursor messenger RNAs
- ecircRNAs:
-
Exonic circRNAs
- EIciRNAs:
-
Exon‒intron circRNAs
- ciRNAs:
-
Intronic circRNAs
- RBP:
-
RNA binding protein
- f-circRNAs:
-
Fusion circRNAs
- tricRNAs:
-
TRNA intronic circular RNAs
- mecciRNAs:
-
Mitochondria-encoded circRNAs
- rt-circRNAs:
-
Read-through circRNAs
- pretRNAs:
-
Precursor tRNAs
- Ago2:
-
Argonaute2
- miRNAs:
-
MicroRNAs
- lncRNAs:
-
Long non-coding RNAs
- MREs:
-
MiRNA response elements
- MLL-r:
-
Mixed lineage leukaemia-rearranged
- IRESs:
-
Internal ribosome entry sites
- m6 A:
-
N6-methyladenosine
- UTR:
-
Untranslated region
- ORF:
-
Open reading frame
- chr:
-
Chromosome
- EMI:
-
Extramedullary infiltration
- NPM1:
-
Nucleophosmin
- HSPC:
-
Healthy haematopoietic stem and progenitor cell
- CCLE:
-
Cancer cell line encyclopedia
- FAB:
-
French-American-British
- BM:
-
Bone marrow
- PB:
-
Peripheral blood
- WBC:
-
White blood cell
- PLT:
-
Platelet
- HGB:
-
Haemoglobin
- AUC:
-
Area under the curve
- OS:
-
Overall survival
- EFS:
-
Event-free survival
- LFS:
-
Leukaemia-free survival
- RFS:
-
Relapse-free survival
- DFS:
-
Disease-free survival
References
El Mesallamy HO, Rashed WM, Hamdy NM, Hamdy N. High-dose methotrexate in Egyptian pediatric acute lymphoblastic leukemia: the impact of ABCG2 C421A genetic polymorphism on plasma levels, what is next? J Cancer Res Clin Oncol. 2014;140(8):1359–65.
Radwan SM, Hamdy NM, Hegab HM, El-Mesallamy HO. Beclin-1 and hypoxia-inducible factor-1α genes expression: Potential biomarkers in acute leukemia patients. Cancer Biomark. 2016;16(4):619–26.
Gao L, Zhang Y, Wang S, Kong P, Su Y, Hu J, Jiang M, Bai H, Lang T, Wang J, et al. Effect of rhG-CSF combined with decitabine prophylaxis on relapse of patients with high-risk MRD-negative AML after HSCT: an open-label, multicenter, randomized controlled trial. J Clin Oncol. 2020;38(36):4249–59.
Gao L, Zhang Y, Hu B, Liu J, Kong P, Lou S, Su Y, Yang T, Li H, Liu Y, et al. Phase II multicenter, randomized, double-blind controlled study of efficacy and safety of umbilical cord-derived mesenchymal stromal cells in the prophylaxis of chronic graft-versus-host disease after HLA-haploidentical stem-cell transplantation. J Clin Oncol. 2016;34(24):2843–50.
Yang G, Wang X, Huang S, Huang R, Wei J, Wang X, Zhang X. Generalist in allogeneic hematopoietic stem cell transplantation for MDS or AML: epigenetic therapy. Front Immunol. 2022;13:1034438.
Wang X, Huang R, Zhang X, Zhang X. Current status and prospects of hematopoietic stem cell transplantation in China. Chin Med J (Engl). 2022;135(12):1394–403.
Pommert L, Tarlock K. The evolution of targeted therapy in pediatric AML: gemtuzumab ozogamicin, FLT3/IDH/BCL2 inhibitors, and other therapies. Hematology Am Soc Hematol Educ Program. 2022;2022(1):603–10.
Kovtun Y, Noordhuis P, Whiteman KR, Watkins K, Jones GE, Harvey L, Lai KC, Portwood S, Adams S, Sloss CM, et al. IMGN779, a novel CD33-targeting antibody-drug conjugate with DNA-alkylating activity, exhibits potent antitumor activity in models of AML. Mol Cancer Ther. 2018;17(6):1271–9.
Huang R, Li X, He Y, Zhu W, Gao L, Liu Y, Gao L, Wen Q, Zhong JF, Zhang C, et al. Recent advances in CAR-T cell engineering. J Hematol Oncol. 2020;13(1):86.
Elanany MM, Mostafa D, Hamdy NM. Remodeled tumor immune microenvironment (TIME) parade via natural killer cells reprogramming in breast cancer. Life Sci. 2023;330:121997.
Bewersdorf JP, Abdel-Wahab O. Translating recent advances in the pathogenesis of acute myeloid leukemia to the clinic. Genes Dev. 2022;36(5–6):259–77.
Stubbins RJ, Francis A, Kuchenbauer F, Sanford D. Management of acute myeloid leukemia: a review for general practitioners in oncology. Curr Oncol. 2022;29(9):6245–59.
Kayser S, Levis MJ. The clinical impact of the molecular landscape of acute myeloid leukemia. Haematologica. 2023;108(2):308–20.
Liu CX, Chen LL. Circular RNAs: Characterization, cellular roles, and applications. Cell. 2022;185(12):2016–34.
Abaza T, El-Aziz MKA, Daniel KA, Karousi P, Papatsirou M, Fahmy SA, Hamdy NM, Kontos CK, Youness RA. Emerging role of circular RNAs in hepatocellular carcinoma immunotherapy. Int J Mol Sci. 2023;24(22):16484.
Geng Y, Jiang J, Wu C. Function and clinical significance of circRNAs in solid tumors. J Hematol Oncol. 2018;11(1):98.
Li Y, Zheng Q, Bao C, Li S, Guo W, Zhao J, Chen D, Gu J, He X, Huang S. Circular RNA is enriched and stable in exosomes: a promising biomarker for cancer diagnosis. Cell Res. 2015;25(8):981–4.
Sanger HL, Klotz G, Riesner D, Gross HJ, Kleinschmidt AK. Viroids are single-stranded covalently closed circular RNA molecules existing as highly base-paired rod-like structures. Proc Natl Acad Sci U S A. 1976;73(11):3852–6.
Hamdy NM, El-Sisi MG, Ibrahim SM, ElNokoudy H, Hady AA, Abd-Ellatef GEF, Sallam AM, Barakat BM. In silico analysis and comprehensive review of circular-RNA regulatory roles in breast diseases; a step-toward non-coding RNA precision. Pathol Res Pract. 2024;263:155651.
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.
Youness RA, Hassan HA, Abaza T, Hady AA, El Magdoub HM, Ali M, Vogel J, Thiersch M, Gassmann M, Hamdy NM, et al. A comprehensive insight and in silico analysis of CircRNAs in hepatocellular carcinoma: a step toward ncRNA-based precision medicine. Cells. 2024;13(15):1245.
Liu Y, Cheng Z, Pang Y, Cui L, Qian T, Quan L, Zhao H, Shi J, Ke X, Fu L. Role of microRNAs, circRNAs and long noncoding RNAs in acute myeloid leukemia. J Hematol Oncol. 2019;12(1):51.
Singh V, Uddin MH, Zonder JA, Azmi AS, Balasubramanian SK. Circular RNAs in acute myeloid leukemia. Mol Cancer. 2021;20(1):149.
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.
Chen I, Chen CY, Chuang TJ. Biogenesis, identification, and function of exonic circular RNAs. Wiley Interdiscip Rev RNA. 2015;6(5):563–79.
Li Z, Huang C, Bao C, Chen L, Lin M, Wang X, Zhong G, Yu B, Hu W, Dai L, et al. Exon-intron circular RNAs regulate transcription in the nucleus. Nat Struct Mol Biol. 2015;22(3):256–64.
Zhang Y, Zhang XO, Chen T, Xiang JF, Yin QF, Xing YH, Zhu S, Yang L, Chen LL. Circular intronic long noncoding RNAs. Mol Cell. 2013;51(6):792–806.
Gao Y, Wang J, Zhao F. CIRI: an efficient and unbiased algorithm for de novo circular RNA identification. Genome Biol. 2015;16(1):4.
Guarnerio J, Bezzi M, Jeong JC, Paffenholz SV, Berry K, Naldini MM, Lo-Coco F, Tay Y, Beck AH, Pandolfi PP. Oncogenic role of fusion-circRNAs derived from cancer-associated chromosomal translocations. Cell. 2016;166(4):1055–6.
Schmidt CA, Giusto JD, Bao A, Hopper AK, Matera AG. Molecular determinants of metazoan tricRNA biogenesis. Nucleic Acids Res. 2019;47(12):6452–65.
Liu X, Wang X, Li J, Hu S, Deng Y, Yin H, Bao X, Zhang QC, Wang G, Wang B, et al. Identification of mecciRNAs and their roles in the mitochondrial entry of proteins. Sci China Life Sci. 2020;63(10):1429–49.
Zhao Q, Liu J, Deng H, Ma R, Liao JY, Liang H, Hu J, Li J, Guo Z, Cai J, et al. Targeting mitochondria-located circRNA SCAR alleviates NASH via reducing mROS output. Cell. 2020;183(1):76–93.
Vidal AF. Read-through circular RNAs reveal the plasticity of RNA processing mechanisms in human cells. RNA Biol. 2020;17(12):1823–6.
Jia R, Xiao MS, Li Z, Shan G, Huang C. Defining an evolutionarily conserved role of GW182 in circular RNA degradation. Cell Discov. 2019;5:45.
Liu CX, Li X, Nan F, Jiang S, Gao X, Guo SK, Xue W, Cui Y, Dong K, Ding H, et al. Structure and degradation of circular RNAs regulate PKR activation in innate immunity. Cell. 2019;177(4):865–80.
Hansen TB, Wiklund ED, Bramsen JB, Villadsen SB, Statham AL, Clark SJ, Kjems J. miRNA-dependent gene silencing involving Ago2-mediated cleavage of a circular antisense RNA. EMBO J. 2011;30(21):4414–22.
Park OH, Ha H, Lee Y, Boo SH, Kwon DH, Song HK, Kim YK. Endoribonucleolytic cleavage of m(6)A-containing RNAs by RNase P/MRP complex. Mol Cell. 2019;74(3):494–507.
Fischer JW, Busa VF, Shao Y, Leung AKL. Structure-mediated RNA decay by UPF1 and G3BP1. Mol Cell. 2020;78(1):70–84.
Alkan AH, Akgul B. Endogenous miRNA sponges. Methods Mol Biol. 2022;2257:91–104.
Li J, Hu ZQ, Yu SY, Mao L, Zhou ZJ, Wang PC, Gong Y, Su S, Zhou J, Fan J, et al. CircRPN2 inhibits aerobic glycolysis and metastasis in hepatocellular carcinoma. Cancer Res. 2022;82(6):1055–69.
Xi Y, Shen Y, Wu D, Zhang J, Lin C, Wang L, Yu C, Yu B, Shen W. CircBCAR3 accelerates esophageal cancer tumorigenesis and metastasis via sponging miR-27a-3p. Mol Cancer. 2022;21(1):145.
Zhang F, Su T, Xiao M. RUNX3-regulated circRNA METTL3 inhibits colorectal cancer proliferation and metastasis via miR-107/PER3 axis. Cell Death Dis. 2022;13(6):550.
Tang J, Wang X, Xiao D, Liu S, Tao Y. The chromatin-associated RNAs in gene regulation and cancer. Mol Cancer. 2023;22(1):27.
Conn VM, Gabryelska M, Toubia J, Kirk K, Gantley L, Powell JA, Cildir G, Marri S, Liu R, Stringer BW, et al. Circular RNAs drive oncogenic chromosomal translocations within the MLL recombinome in leukemia. Cancer Cell. 2023;41(7):1309–1326.e10.
Xu X, Zhang J, Tian Y, Gao Y, Dong X, Chen W, Yuan X, Yin W, Xu J, Chen K, et al. CircRNA inhibits DNA damage repair by interacting with host gene. Mol Cancer. 2020;19(1):128.
Sun YM, Wang WT, Zeng ZC, Chen TQ, Han C, Pan Q, Huang W, Fang K, Sun LY, Zhou YF, et al. circMYBL2, a circRNA from MYBL2, regulates FLT3 translation by recruiting PTBP1 to promote FLT3-ITD AML progression. Blood. 2019;134(18):1533–46.
Zhu T, Cen Y, Chen Z, Zhang Y, Zhao L, Wang J, Lu W, Xie X, Wang X. Oncogenic circTICRR suppresses autophagy via binding to HuR protein and stabilizing GLUD1 mRNA in cervical cancer. Cell Death Dis. 2022;13(5):479.
Liu B, Yang G, Wang X, Liu J, Lu Z, Wang Q, Xu B, Liu Z, Li J. CircBACH1 (hsa_circ_0061395) promotes hepatocellular carcinoma growth by regulating p27 repression via HuR. J Cell Physiol. 2020;235(10):6929–41.
Chen Z, Lu T, Huang L, Wang Z, Yan Z, Guan Y, Hu W, Fan Z, Zhu P. Circular RNA cia-MAF drives self-renewal and metastasis of liver tumor-initiating cells via transcription factor MAFF. J Clin Invest. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.1172/JCI148020.
Chen Q, Wang H, Li Z, Li F, Liang L, Zou Y, Shen H, Li J, Xia Y, Cheng Z, et al. Circular RNA ACTN4 promotes intrahepatic cholangiocarcinoma progression by recruiting YBX1 to initiate FZD7 transcription. J Hepatol. 2022;76(1):135–47.
Ding L, Zhao Y, Dang S, Wang Y, Li X, Yu X, Li Z, Wei J, Liu M, Li G. Circular RNA circ-DONSON facilitates gastric cancer growth and invasion via NURF complex dependent activation of transcription factor SOX4. Mol Cancer. 2019;18(1):45.
Li Q, Wang Y, Wu S, Zhou Z, Ding X, Shi R, Thorne RF, Zhang XD, Hu W, Wu M. CircACC1 regulates assembly and activation of AMPK complex under metabolic stress. Cell Metab. 2019;30(1):157–73.
Sun S, Gao J, Zhou S, Li Y, Wang Y, Jin L, Li J, Liu B, Zhang B, Han S, et al. A novel circular RNA circ-LRIG3 facilitates the malignant progression of hepatocellular carcinoma by modulating the EZH2/STAT3 signaling. J Exp Clin Cancer Res. 2020;39(1):252.
Du WW, Yang W, Liu E, Yang Z, Dhaliwal P, Yang BB. Foxo3 circular RNA retards cell cycle progression via forming ternary complexes with p21 and CDK2. Nucleic Acids Res. 2016;44(6):2846–58.
Shen Y, Zhang N, Chai J, Wang T, Ma C, Han L, Yang M. CircPDIA4 induces gastric cancer progression by promoting ERK1/2 activation and enhancing biogenesis of oncogenic circRNAs. Cancer Res. 2023;83(4):538–52.
Wang Z, Sun A, Yan A, Yao J, Huang H, Gao Z, Han T, Gu J, Li N, Wu H, et al. Circular RNA MTCL1 promotes advanced laryngeal squamous cell carcinoma progression by inhibiting C1QBP ubiquitin degradation and mediating beta-catenin activation. Mol Cancer. 2022;21(1):92.
Zhang C, Jiang H, Yuan L, Liao Y, Liu P, Du Q, Pan C, Liu T, Li J, Chen Y, et al. CircVPRBP inhibits nodal metastasis of cervical cancer by impeding RACK1 O-GlcNAcylation and stability. Oncogene. 2023;42(11):793–807.
Yang Y, Fan X, Mao M, Song X, Wu P, Zhang Y, Jin Y, Yang Y, Chen LL, Wang Y, et al. Extensive translation of circular RNAs driven by N(6)-methyladenosine. Cell Res. 2017;27(5):626–41.
Pamudurti NR, Bartok O, Jens M, Ashwal-Fluss R, Stottmeister C, Ruhe L, Hanan M, Wyler E, Perez-Hernandez D, Ramberger E, et al. Translation of CircRNAs. Mol Cell. 2017;66(1):9–21.
Legnini I, Di Timoteo G, Rossi F, Morlando M, Briganti F, Sthandier O, Fatica A, Santini T, Andronache A, Wade M, et al. Circ-ZNF609 is a circular RNA that can be translated and functions in myogenesis. Mol Cell. 2017;66(1):22–37.
Song R, Ma S, Xu J, Ren X, Guo P, Liu H, Li P, Yin F, Liu M, Wang Q, et al. A novel polypeptide encoded by the circular RNA ZKSCAN1 suppresses HCC via degradation of mTOR. Mol Cancer. 2023;22(1):16.
Li Y, Chen B, Zhao J, Li Q, Chen S, Guo T, Li Y, Lai H, Chen Z, Meng Z, et al. HNRNPL circularizes ARHGAP35 to produce an oncogenic protein. Adv Sci (Weinh). 2021;8(13):2001701.
Di Timoteo G, Dattilo D, Centron-Broco A, Colantoni A, Guarnacci M, Rossi F, Incarnato D, Oliviero S, Fatica A, Morlando M, et al. Modulation of circRNA metabolism by m(6)A modification. Cell Rep. 2020;31(6):107641.
Wu W, Zhang J, Cao X, Cai Z, Zhao F. Exploring the cellular landscape of circular RNAs using full-length single-cell RNA sequencing. Nat Commun. 2022;13(1):3242.
Zhang J, Hou L, Zuo Z, Ji P, Zhang X, Xue Y, Zhao F. Comprehensive profiling of circular RNAs with nanopore sequencing and CIRI-long. Nat Biotechnol. 2021;39(7):836–45.
Zhang J, Hou L, Zuo Z, Ji P, Zhang X, Xue Y, Zhao F. Author Correction: Comprehensive profiling of circular RNAs with nanopore sequencing and CIRI-long. Nat Biotechnol. 2021;39(7):893.
Rahimi K, Veno MT, Dupont DM, Kjems J. Nanopore sequencing of brain-derived full-length circRNAs reveals circRNA-specific exon usage, intron retention and microexons. Nat Commun. 2021;12(1):4825.
Zhang B, Chen M, Cao J, Liang Y, Tu T, Hu J, Li T, Cai Y, Li S, Liu B, et al. An integrated electrochemical POCT platform for ultrasensitive circRNA detection towards hepatocellular carcinoma diagnosis. Biosens Bioelectron. 2021;192:113500.
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.
Li W, Zhong C, Jiao J, Li P, Cui B, Ji C, Ma D. Characterization of hsa_circ_0004277 as a new biomarker for acute myeloid leukemia via circular RNA profile and bioinformatics analysis. Int J Mol Sci. 2017;18(3):597.
Chen H, Liu T, Liu J, Feng Y, Wang B, Wang J, Bai J, Zhao W, Shen Y, Wang X, et al. Circ-ANAPC7 is upregulated in acute myeloid leukemia and appears to target the MiR-181 family. Cell Physiol Biochem. 2018;47(5):1998–2007.
Yuan DM, Ma J, Fang WB. Identification of non-coding RNA regulatory networks in pediatric acute myeloid leukemia reveals circ-0004136 could promote cell proliferation by sponging miR-142. Eur Rev Med Pharmacol Sci. 2019;23(21):9251–8.
Ping L, Jian-Jun C, Chu-Shu L, Guang-Hua L, Ming Z. Silencing of circ_0009910 inhibits acute myeloid leukemia cell growth through increasing miR-20a-5p. Blood Cells Mol Dis. 2019;75:41–7.
Ding Y, Dong Y, Lu H, Luo X, Fu J, Xiu B, Liang A, Zhang W. Circular RNA profile of acute myeloid leukaemia indicates circular RNA annexin A2 as a potential biomarker and therapeutic target for acute myeloid leukaemia. Am J Transl Res. 2020;12(5):1683–99.
Lin Y, Huang Y, Liang C, Xie S, Xie A. Silencing of circTASP1 inhibits proliferation and induces apoptosis of acute myeloid leukaemia cells through modulating miR-515-5p/HMGA2 axis. J Cell Mol Med. 2021;25(15):7367–80.
Liu X, Liu X, Cai M, Luo A, He Y, Liu S, Zhang X, Yang X, Xu L, Jiang H. CircRNF220, not its linear cognate gene RNF220, regulates cell growth and is associated with relapse in pediatric acute myeloid leukemia. Mol Cancer. 2021;20(1):139.
Long F, Lin Z, Long Q, Lu Z, Zhu K, Zhao M, Yang M. CircZBTB46 protects acute myeloid leukemia cells from ferroptotic cell death by upregulating SCD. Cancers (Basel). 2023;15(2):459.
Lv C, Sun L, Guo Z, Li H, Kong D, Xu B, Lin L, Liu T, Guo D, Zhou J, et al. Circular RNA regulatory network reveals cell-cell crosstalk in acute myeloid leukemia extramedullary infiltration. J Transl Med. 2018;16(1):361.
Li S, Ma Y, Tan Y, Ma X, Zhao M, Chen B, Zhang R, Chen Z, Wang K. Profiling and functional analysis of circular RNAs in acute promyelocytic leukemia and their dynamic regulation during all-trans retinoic acid treatment. Cell Death Dis. 2018;9(6):651.
Shang J, Chen WM, Liu S, Wang ZH, Wei TN, Chen ZZ, Wu WB. CircPAN3 contributes to drug resistance in acute myeloid leukemia through regulation of autophagy. Leuk Res. 2019;85:106198.
Li M, Meng F, Lu Q. Expression profile screening and bioinformatics analysis of circRNA, LncRNA, and mRNA in acute myeloid leukemia drug-resistant cells. Turk J Haematol. 2020;37(2):104–10.
Ye F, Fan C, Peng M, Liu S, Dong J, Yang L, Zhang H. Screening and validating circular RNAs that estimate disease risk and treatment response of pediatric acute myeloid leukemia: a microarray-based analyses and RT-qPCR validation. J Cancer Res Clin Oncol. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00432-023-04879-9.
Bullinger L, Dohner K, Dohner H. Genomics of acute myeloid leukemia diagnosis and pathways. J Clin Oncol. 2017;35(9):934–46.
Lux S, Blatte TJ, Gillissen B, Richter A, Cocciardi S, Skambraks S, Schwarz K, Schrezenmeier H, Dohner H, Dohner K, et al. Deregulated expression of circular RNAs in acute myeloid leukemia. Blood Adv. 2021;5(5):1490–503.
Hirsch S, Blatte TJ, Grasedieck S, Cocciardi S, Rouhi A, Jongen-Lavrencic M, Paschka P, Kronke J, Gaidzik VI, Dohner H, et al. Circular RNAs of the nucleophosmin (NPM1) gene in acute myeloid leukemia. Haematologica. 2017;102(12):2039–47.
Issah MA, Wu D, Zhang F, Zheng W, Liu Y, Chen R, Lai G, Shen J. Expression profiling of N(6)-methyladenosine modified circRNAs in acute myeloid leukemia. Biochem Biophys Res Commun. 2022;601:137–45.
Barrett T, Wilhite SE, Ledoux P, Evangelista C, Kim IF, Tomashevsky M, Marshall KA, Phillippy KH, Sherman PM, Holko M, et al. NCBI GEO: archive for functional genomics data sets–update. Nucleic Acids Res. 2013;41:D991-995.
Zhao Z, Wang K, Wu F, Wang W, Zhang K, Hu H, Liu Y, Jiang T. circRNA disease: a manually curated database of experimentally supported circRNA-disease associations. Cell Death Dis. 2018;9(5):475.
Xie F, Liu S, Wang J, Xuan J, Zhang X, Qu L, Zheng L, Yang J. deepBase v3.0: expression atlas and interactive analysis of ncRNAs from thousands of deep-sequencing data. Nucleic Acids Res. 2021;49:D877–83.
Wu W, Ji P, Zhao F. CircAtlas: an integrated resource of one million highly accurate circular RNAs from 1070 vertebrate transcriptomes. Genome Biol. 2020;21(1):101.
Feng J, Chen W, Dong X, Wang J, Mei X, Deng J, Yang S, Zhuo C, Huang X, Shao L, et al. CSCD2: an integrated interactional database of cancer-specific circular RNAs. Nucleic Acids Res. 2022;50(D1):D1179–83.
Chen Y, Yao L, Tang Y, Jhong JH, Wan J, Chang J, Cui S, Luo Y, Cai X, Li W, et al. CircNet 2.0: an updated database for exploring circular RNA regulatory networks in cancers. Nucleic Acids Res. 2022;50(D1):D93–101.
Li S, Li Y, Chen B, Zhao J, Yu S, Tang Y, Zheng Q, Li Y, Wang P, He X, et al. exoRBase: a database of circRNA, lncRNA and mRNA in human blood exosomes. Nucleic Acids Res. 2018;46(D1):D106–12.
Vo JN, Cieslik M, Zhang Y, Shukla S, Xiao L, Zhang Y, Wu YM, Dhanasekaran SM, Engelke CG, Cao X, et al. The landscape of circular RNA in cancer. Cell. 2019;176(4):869–81.
Yao D, Zhang L, Zheng M, Sun X, Lu Y, Liu P. Circ2Disease: a manually curated database of experimentally validated circRNAs in human disease. Sci Rep. 2018;8(1):11018.
Xia S, Feng J, Lei L, Hu J, Xia L, Wang J, Xiang Y, Liu L, Zhong S, Han L, et al. Comprehensive characterization of tissue-specific circular RNAs in the human and mouse genomes. Brief Bioinform. 2017;18(6):984–92.
Ruan H, Xiang Y, Ko J, Li S, Jing Y, Zhu X, Ye Y, Zhang Z, Mills T, Feng J, et al. Comprehensive characterization of circular RNAs in ~ 1000 human cancer cell lines. Genome Med. 2019;11(1):55.
Ma XK, Xue W, Chen LL, Yang L. CIRCexplorer pipelines for circRNA annotation and quantification from non-polyadenylated RNA-seq datasets. Methods. 2021;196:3–10.
Westholm JO, Miura P, Olson S, Shenker S, Joseph B, Sanfilippo P, Celniker SE, Graveley BR, Lai EC. Genome-wide analysis of drosophila circular RNAs reveals their structural and sequence properties and age-dependent neural accumulation. Cell Rep. 2014;9(5):1966–80.
Memczak S, Jens M, Elefsinioti A, Torti F, Krueger J, Rybak A, Maier L, Mackowiak SD, Gregersen LH, Munschauer M, et al. Circular RNAs are a large class of animal RNAs with regulatory potency. Nature. 2013;495(7441):333–8.
Cheng J, Metge F, Dieterich C. Specific identification and quantification of circular RNAs from sequencing data. Bioinformatics. 2016;32(7):1094–6.
Chuang TJ, Wu CS, Chen CY, Hung LY, Chiang TW, Yang MY. NCLscan: accurate identification of non-co-linear transcripts (fusion, trans-splicing and circular RNA) with a good balance between sensitivity and precision. Nucleic Acids Res. 2016;44(3):e29.
Glazar P, Papavasileiou P, Rajewsky N. circBase: a database for circular RNAs. RNA. 2014;20(11):1666–70.
Liu M, Wang Q, Shen J, Yang BB, Ding X. Circbank: a comprehensive database for circRNA with standard nomenclature. RNA Biol. 2019;16(7):899–905.
Zhao M, Qu H. circVAR database: genome-wide archive of genetic variants for human circular RNAs. BMC Genomics. 2020;21(1):750.
Chen X, Han P, Zhou T, Guo X, Song X, Li Y. circRNADb: a comprehensive database for human circular RNAs with protein-coding annotations. Sci Rep. 2016;6:34985.
Rombel IT, Sykes KF, Rayner S, Johnston SA. ORF-FINDER: a vector for high-throughput gene identification. Gene. 2002;282(1–2):33–41.
Meng X, Chen Q, Zhang P, Chen M. CircPro: an integrated tool for the identification of circRNAs with protein-coding potential. Bioinformatics. 2017;33(20):3314–6.
Zhao J, Wu J, Xu T, Yang Q, He J, Song X. IRESfinder: identifying RNA internal ribosome entry site in eukaryotic cell using framed k-mer features. J Genet Genomics. 2018;45(7):403–6.
Zhao J, Li Y, Wang C, Zhang H, Zhang H, Jiang B, Guo X, Song X. IRESbase: a comprehensive database of experimentally validated internal ribosome entry sites. Genomics Proteomics Bioinform. 2020;18(2):129–39.
Sun P, Li G. CircCode: a powerful tool for identifying circRNA coding ability. Front Genet. 2019;10:981.
Huang W, Ling Y, Zhang S, Xia Q, Cao R, Fan X, Fang Z, Wang Z, Zhang G. TransCirc: an interactive database for translatable circular RNAs based on multi-omics evidence. Nucleic Acids Res. 2021;49(D1):D236–42.
Zhong S, Feng J. CircPrimer 2.0: a software for annotating circRNAs and predicting translation potential of circRNAs. BMC Bioinform. 2022;23(1):215.
Zhou Y, Zeng P, Li YH, Zhang Z, Cui Q. SRAMP: prediction of mammalian N6-methyladenosine (m6A) sites based on sequence-derived features. Nucleic Acids Res. 2016;44(10):e91.
Dudekula DB, Panda AC, Grammatikakis I, De S, Abdelmohsen K, Gorospe M. CircInteractome: a web tool for exploring circular RNAs and their interacting proteins and microRNAs. RNA Biol. 2016;13(1):34–42.
Meng X, Hu D, Zhang P, Chen Q, Chen M. CircFunBase: a database for functional circular RNAs. Database. 2019. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/database/baz003.
Li JH, Liu S, Zhou H, Qu LH, Yang JH. starBase v2.0: decoding miRNA-ceRNA, miRNA-ncRNA and protein-RNA interaction networks from large-scale CLIP-Seq data. Nucleic Acids Res. 2014;42:D92-97.
Tang Z, Li X, Zhao J, Qian F, Feng C, Li Y, Zhang J, Jiang Y, Yang Y, Wang Q, et al. TRCirc: a resource for transcriptional regulation information of circRNAs. Brief Bioinform. 2019;20(6):2327–33.
Hu Q, Gu Y, Chen S, Tian Y, Yang S. Hsa_circ_0079480 promotes tumor progression in acute myeloid leukemia via miR-654-3p/HDGF axis. Aging (Albany NY). 2020;13(1):1120–31.
Guarnerio J, Bezzi M, Jeong JC, Paffenholz SV, Berry K, Naldini MM, Lo-Coco F, Tay Y, Beck AH, Pandolfi PP. Oncogenic role of fusion-circRNAs derived from cancer-associated chromosomal translocations. Cell. 2016;165(2):289–302.
Yi L, Zhou L, Luo J, Yang Q. Circ-PTK2 promotes the proliferation and suppressed the apoptosis of acute myeloid leukemia cells through targeting miR-330-5p/FOXM1 axis. Blood Cells Mol Dis. 2021;86:102506.
Li H, Bi K, Feng S, Wang Y, Zhu C. CircRNA circ_POLA2 is upregulated in acute myeloid leukemia (AML) and promotes cell proliferation by suppressing the production of mature miR-34a. Cancer Manag Res. 2021;13:3629–37.
Dong S, Zhong H, Li L. Circ_DLEU2 knockdown represses cell proliferation, migration and invasion, and induces cell apoptosis through the miR-582-5p/COX2 pathway in acute myeloid leukemia. Histol Histopathol. 2023;38(2):171–83.
Wu DM, Wen X, Han XR, Wang S, Wang YJ, Shen M, Fan SH, Zhang ZF, Shan Q, Li MQ et al: Role of Circular RNA DLEU2 in Human Acute MyeloidLeukemia. Mol Cell Biol 2018, 38(20).
Cao J, Huang S, Li X. Rapamycin inhibits the progression of human acute myeloid leukemia by regulating the circ_0094100/miR-217/ATP1B1 axis. Exp Hematol. 2022;112:60–9.
Huang L, Huang L, Ming X, Wu J, Liu W, Xiao Y. CircNFIX knockdown inhibited AML tumorigenicity by the miR-876-3p/TRIM31 axis. Hematology. 2022;27(1):1046–55.
Zhang L, Bu Z, Shen J, Shang L, Chen Y, Wang Y. A novel circular RNA (hsa_circ_0000370) increases cell viability and inhibits apoptosis of FLT3-ITD-positive acute myeloid leukemia cells by regulating miR-1299 and S100A7A. Biomed Pharmacother. 2020;122:109619.
Chen K, Ning X, Yan X, Song L. Circ_0104700 contributes to acute myeloid leukemia progression by enhancing MCM2 expression through targeting miR-665. Hematology. 2023;28(1):2227489.
Liu L, Qiang X. Hsa_circ_0044907 promotes acute myeloid leukemia progression through upregulating oncogene KIT via sequestering miR-186-5p. Hematology. 2022;27(1):960–70.
Xiao Y, Ming X, Wu J. Hsa_circ_0002483 regulates miR-758-3p/MYC axis to promote acute myeloid leukemia progression. Hematol Oncol. 2021;39(2):243–53.
Li Q, Luan Q, Zhu H, Zhao Y, Ji J, Wu F, Yan J. Circular RNA circ_0005774 contributes to proliferation and suppresses apoptosis of acute myeloid leukemia cells via circ_0005774/miR-192-5p/ULK1 ceRNA pathway. Biochem Biophys Res Commun. 2021;551:78–85.
Wang X, Jin P, Zhang Y, Wang K. CircSPI1 acts as an oncogene in acute myeloid leukemia through antagonizing SPI1 and interacting with microRNAs. Cell Death Dis. 2021;12(4):297.
Yang X, Wang Y, Rong S, An J, Lan X, Yin B, Sun Y, Wang P, Tan B, Xuan Y, et al. Gene SH3BGRL3 regulates acute myeloid leukemia progression through circRNA_0010984 based on competitive endogenous RNA mechanism. Front Cell Dev Biol. 2023;11:1173491.
Shang Z, Ming X, Wu J, Xiao Y. Downregulation of circ_0012152 inhibits proliferation and induces apoptosis in acute myeloid leukemia cells through the miR-625-5p/SOX12 axis. Hematol Oncol. 2021;39(4):539–48.
Zhang Z, Lin S, Yin J, Yu W, Xu C. CircRNF220 plays a pathogenic role to facilitate cell progression of AML in vitro via sponging miR-330-5p to induce upregulation of SOX4. Histol Histopathol. 2022;37(10):1019–30.
Zhang R, Li Y, Wang H, Zhu K, Zhang G. The regulation of circRNA RNF13/miRNA-1224-5p axis promotes the malignant evolution in acute myeloid leukemia. Biomed Res Int. 2020;2020:5654380.
Wang Y, Guo T, Liu Q, Xie X. CircRAD18 accelerates the progression of acute myeloid leukemia by modulation of miR-206/PRKACB Axis. Cancer Manag Res. 2020;12:10887–96.
Lin L, Wang Y, Bian S, Sun L, Guo Z, Kong D, Zhao L, Guo D, Li Q, Wu M, et al. A circular RNA derived from PLXNB2 as a valuable predictor of the prognosis of patients with acute myeloid leukaemia. J Transl Med. 2021;19(1):123.
Wang J, Wu C, Zhou W. CircPLXNB2 regulates acute myeloid leukemia progression through miR-654-3p/CCND1 axis. Hematology. 2023;28(1):2220522.
Hu X, Yin J, He R, Chao R, Zhu S. Circ_KCNQ5 participates in the progression of childhood acute myeloid leukemia by enhancing the expression of RAB10 via binding to miR-622. Hematology. 2022;27(1):431–40.
Bi J, Pu Y, Yu X. Exosomal circ_0004136 enhances the progression of pediatric acute myeloid leukemia depending on the regulation of miR-570-3p/TSPAN3 axis. Anticancer Drugs. 2021;32(8):802–11.
Zhang H, Tao Y, Ding X, Wang Y, Wang X. Roles of the hsa_circ_0013880/USP32/Rap1b axis in the proliferation and apoptosis of acute myeloid leukemia cells. Acta Biochim Biophys Sin (Shanghai). 2023;55(3):382–93.
Chang W, Shang Z, Ming X, Wu J, Xiao Y. Circ-SFMBT2 facilitates the malignant growth of acute myeloid leukemia cells by modulating miR-582-3p/ZBTB20 pathway. Histol Histopathol. 2022;37(2):137–49.
Ye Q, Li N, Zhou K, Liao C. Homo sapiens circular RNA 0003602 (Hsa_circ_0003602) accelerates the tumorigenicity of acute myeloid leukemia by modulating miR-502-5p/IGF1R axis. Mol Cell Biochem. 2022;477(2):635–44.
Zhang T, Zhou Y, Guan J, Cheng H. Circ_0058058 knockdown inhibits acute myeloid leukemia progression by sponging miR-4319 to regulate EIF5A2 expression. Cancer Biother Radiopharm. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.1089/cbr.2020.4170.
Sheng XF, Hong LL, Fan L, Zhang Y, Chen KL, Mu J, Shen SY, Zhuang HF. Circular RNA PVT1 regulates cell proliferation, migration, and apoptosis by stabilizing c-Myc and downstream target CXCR4 expression in acute myeloid leukemia. Turk J Haematol. 2023;40(2):82–91.
Ding J, Zhang X, Xue J, Fang L, Ban C, Song B, Wu L. CircNPM1 strengthens Adriamycin resistance in acute myeloid leukemia by mediating the miR-345-5p/FZD5 pathway. Cent Eur J Immunol. 2021;46(2):162–82.
Wang D, Ming X, Xu J, Xiao Y. Circ_0009910 shuttled by exosomes regulates proliferation, cell cycle and apoptosis of acute myeloid leukemia cells by regulating miR-5195-3p/GRB10 axis. Hematol Oncol. 2021;39(3):390–400.
Xue F, Li M, Liu Y, Xu C, Li H, Liu H. Circ_0035381 regulates acute myeloid leukemia development by modulating YWHAZ expression via adsorbing miR-582-3p. Biochem Genet. 2023;61(1):354–71.
Du A, Yang Q, Sun X, Zhao Q. Exosomal circRNA-001264 promotes AML immunosuppression through induction of M2-like macrophages and PD-L1 overexpression. Int Immunopharmacol. 2023;124:110868.
Chen JJ, Lei P, Zhou M. hsa_circ_0121582 inhibits leukemia growth by dampening Wnt/beta-catenin signaling. Clin Transl Oncol. 2020;22(12):2293–302.
Han F, Zhong C, Li W, Wang R, Zhang C, Yang X, Ji C, Ma D. hsa_circ_0001947 suppresses acute myeloid leukemia progression via targeting hsa-miR-329-5p/CREBRF axis. Epigenomics. 2020;12(11):935–53.
Liu W, Cheng F. Circular RNA circCRKL inhibits the proliferation of acute myeloid leukemia cells via the miR-196a-5p/miR-196b-5p/p27 axis. Bioengineered. 2021;12(1):7704–13.
Wang N, Yang B, Jin J, He Y, Wu X, Yang Y, Zhou W, He Z. Circular RNA circ_0040823 inhibits the proliferation of acute myeloid leukemia cells and induces apoptosis by regulating miR-516b/PTEN. J Gene Med. 2022;24(3):e3404.
Liu Y, Chen X, Liu J, Jin Y, Wang W. Circular RNA circ_0004277 inhibits acute myeloid leukemia progression through MicroRNA-134-5p / single stranded DNA binding protein 2. Bioengineered. 2022;13(4):9662–73.
Yang X, Han F, Hu X, Li G, Wu H, Can C, Wei Y, Liu J, Wang R, Jia W, et al. EIF4A3-induced Circ_0001187 facilitates AML suppression through promoting ubiquitin-proteasomal degradation of METTL3 and decreasing m6A modification level mediated by miR-499a-5p/RNF113A pathway. Biomark Res. 2023;11(1):59.
Dong LH, Huang JJ, Zu P, Liu J, Gao X, Du JW, Li YF. CircKDM4C upregulates P53 by sponging hsa-let-7b-5p to induce ferroptosis in acute myeloid leukemia. Environ Toxicol. 2021;36(7):1288–302.
Bouligny IM, Maher KR, Grant S. Mechanisms of myeloid leukemogenesis: current perspectives and therapeutic objectives. Blood Rev. 2023;57:100996.
De Kouchkovsky I, Abdul-Hay M. Acute myeloid leukemia: a comprehensive review and 2016 update. Blood Cancer J. 2016;6(7):e441.
Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, Patel DN, Bauer AJ, Cantley AM, Yang WS, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell. 2012;149(5):1060–72.
Levine B, Kroemer G. Autophagy in the pathogenesis of disease. Cell. 2008;132(1):27–42.
Levy JMM, Towers CG, Thorburn A. Targeting autophagy in cancer. Nat Rev Cancer. 2017;17(9):528–42.
Seo W, Silwal P, Song IC, Jo EK. The dual role of autophagy in acute myeloid leukemia. J Hematol Oncol. 2022;15(1):51.
Yang L, Shi P, Zhao G, Xu J, Peng W, Zhang J, Zhang G, Wang X, Dong Z, Chen F, et al. Targeting cancer stem cell pathways for cancer therapy. Signal Transduct Target Ther. 2020;5(1):8.
Lin G, Fei Y, Zhang Y. Hsa-circ_0003420 induces apoptosis in acute myeloid leukemia stem cells and impairs stem cell properties. Immunopharmacol Immunotoxicol. 2021;43(5):622–31.
Shang J, Chen WM, Wang ZH, Wei TN, Chen ZZ, Wu WB. CircPAN3 mediates drug resistance in acute myeloid leukemia through the miR-153-5p/miR-183-5p-XIAP axis. Exp Hematol. 2019;70(42–54):e43.
Li H, Bi K, Feng S, Wang Y, Zhu C. CircRNA CircEHBP1 regulates the maturation of MiR-129 to increase the chemoresistance of cancer cells to adriamycin in acute myeloid leukaemia. Mediterr J Hematol Infect Dis. 2022;14(1):e2022062.
Tettamanti S, Pievani A, Biondi A, Dotti G, Serafini M. Catch me if you can: how AML and its niche escape immunotherapy. Leukemia. 2022;36(1):13–22.
Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science. 2020;367(6478):eaau6977.
Paskeh MDA, Entezari M, Mirzaei S, Zabolian A, Saleki H, Naghdi MJ, Sabet S, Khoshbakht MA, Hashemi M, Hushmandi K, et al. Emerging role of exosomes in cancer progression and tumor microenvironment remodeling. J Hematol Oncol. 2022;15(1):83.
Qiu Y, Chen T, Hu R, Zhu R, Li C, Ruan Y, Xie X, Li Y. Next frontier in tumor immunotherapy: macrophage-mediated immune evasion. Biomark Res. 2021;9(1):72.
Papaioannou D, Volinia S, Nicolet D, Swierniak M, Petri A, Mrozek K, Bill M, Pepe F, Walker CJ, Walker AE, et al. Clinical and functional significance of circular RNAs in cytogenetically normal AML. Blood Adv. 2020;4(2):239–51.
Shen Y, Jia Y, Zhang R, Chen H, Feng Y, Li F, Wang T, Bai J, He A, Yang Y. Using Circ-ANAPC7 as a novel type of biomarker in the monitoring of acute myeloid leukemia. Acta Haematol. 2022;145(2):176–83.
Chen T, Chen F. The role of circular RNA plasmacytoma variant translocation 1 as a biomarker for prognostication of acute myeloid leukemia. Hematology. 2021;26(1):1018–24.
Guo L, Kou R, Song Y, Li G, Jia X, Li Z, Zhang Y. Serum hsa_circ_0079480 is a novel prognostic marker for acute myeloid leukemia. J Clin Lab Anal. 2022;36(4):e24337.
Zhou J, Zhou LY, Tang X, Zhang J, Zhai LL, Yi YY, Yi J, Lin J, Qian J, Deng ZQ. Circ-Foxo3 is positively associated with the Foxo3 gene and leads to better prognosis of acute myeloid leukemia patients. BMC Cancer. 2019;19(1):930.
Ma J, Wen X, Xu Z, Xia P, Jin Y, Lin J, Qian J. Predicting the influence of Circ_0059706 expression on prognosis in patients with acute myeloid leukemia using classical statistics and machine learning. Front Genet. 2022;13:961142.
Wang Y, Liu J, Ma J, Sun T, Zhou Q, Wang W, Wang G, Wu P, Wang H, Jiang L, et al. Exosomal circRNAs: biogenesis, effect and application in human diseases. Mol Cancer. 2019;18(1):116.
Yu D, Li Y, Wang M, Gu J, Xu W, Cai H, Fang X, Zhang X. Exosomes as a new frontier of cancer liquid biopsy. Mol Cancer. 2022;21(1):56.
Abudayyeh OO, Gootenberg JS, Essletzbichler P, Han S, Joung J, Belanto JJ, Verdine V, Cox DBT, Kellner MJ, Regev A, et al. RNA targeting with CRISPR-Cas13. Nature. 2017;550(7675):280–4.
Crooke ST, Baker BF, Crooke RM, Liang XH. Antisense technology: an overview and prospectus. Nat Rev Drug Discov. 2021;20(6):427–53.
Setten RL, Rossi JJ, Han SP. The current state and future directions of RNAi-based therapeutics. Nat Rev Drug Discov. 2019;18(6):421–46.
Wang SW, Gao C, Zheng YM, Yi L, Lu JC, Huang XY, Cai JB, Zhang PF, Cui YH, Ke AW. Current applications and future perspective of CRISPR/Cas9 gene editing in cancer. Mol Cancer. 2022;21(1):57.
Dong Y, Siegwart DJ, Anderson DG. Strategies, design, and chemistry in siRNA delivery systems. Adv Drug Deliv Rev. 2019;144:133–47.
Zhang Y, Liu Q, Zhang X, Huang H, Tang S, Chai Y, Xu Z, Li M, Chen X, Liu J, et al. Recent advances in exosome-mediated nucleic acid delivery for cancer therapy. J Nanobiotechnology. 2022;20(1):279.
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.
Zheng Q, Bao C, Guo W, Li S, Chen J, Chen B, Luo Y, Lyu D, Li Y, Shi G, et al. Circular RNA profiling reveals an abundant circHIPK3 that regulates cell growth by sponging multiple miRNAs. Nat Commun. 2016;7:11215.
Gu Y, Wang Y, He L, Zhang J, Zhu X, Liu N, Wang J, Lu T, He L, Tian Y, et al. Circular RNA circIPO11 drives self-renewal of liver cancer initiating cells via Hedgehog signaling. Mol Cancer. 2021;20(1):132.
Li S, Li X, Xue W, Zhang L, Yang LZ, Cao SM, Lei YN, Liu CX, Guo SK, Shan L, et al. Screening for functional circular RNAs using the CRISPR-Cas13 system. Nat Methods. 2021;18(1):51–9.
Meganck RM, Borchardt EK, Castellanos Rivera RM, Scalabrino ML, Wilusz JE, Marzluff WF, Asokan A. Tissue-dependent expression and translation of circular RNAs with recombinant AAV vectors in vivo. Mol Ther Nucleic Acids. 2018;13:89–98.
Palombarini F, Masciarelli S, Incocciati A, Liccardo F, Di Fabio E, Iazzetti A, Fabrizi G, Fazi F, Macone A, Bonamore A, et al. Self-assembling ferritin-dendrimer nanoparticles for targeted delivery of nucleic acids to myeloid leukemia cells. J Nanobiotechnology. 2021;19(1):172.
Zheng H, Huang S, Wei G, Sun Y, Li C, Si X, Chen Y, Tang Z, Li X, Chen Y, et al. CircRNA Samd4 induces cardiac repair after myocardial infarction by blocking mitochondria-derived ROS output. Mol Ther. 2022;30(11):3477–98.
Zeng Z, Xia L, Fan S, Zheng J, Qin J, Fan X, Liu Y, Tao J, Liu Y, Li K, et al. Circular RNA CircMAP3K5 acts as a MicroRNA-22-3p sponge to promote resolution of intimal hyperplasia Via TET2-mediated smooth muscle cell differentiation. Circulation. 2021;143(4):354–71.
Chen CK, Cheng R, Demeter J, Chen J, Weingarten-Gabbay S, Jiang L, Snyder MP, Weissman JS, Segal E, Jackson PK, et al. Structured elements drive extensive circular RNA translation. Mol Cell. 2021;81(20):4300–18.
Qu L, Yi Z, Shen Y, Lin L, Chen F, Xu Y, Wu Z, Tang H, Zhang X, Tian F, et al. Circular RNA vaccines against SARS-CoV-2 and emerging variants. Cell. 2022;185(10):1728–44.
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(14):6422–36.
Huang D, Zhu X, Ye S, Zhang J, Liao J, Zhang N, Zeng X, Wang J, Yang B, Zhang Y, et al. Tumour circular RNAs elicit anti-tumour immunity by encoding cryptic peptides. Nature. 2024;625(7995):593–602.
Anastasiadou E, Seto AG, Beatty X, Hermreck M, Gilles ME, Stroopinsky D, Pinter-Brown LC, Pestano L, Marchese C, Avigan D, et al. Cobomarsen, an oligonucleotide inhibitor of miR-155, slows DLBCL tumor cell growth in vitro and in vivo. Clin Cancer Res. 2021;27(4):1139–49.
Hong DS, Kang YK, Borad M, Sachdev J, Ejadi S, Lim HY, Brenner AJ, Park K, Lee JL, Kim TY, et al. Phase 1 study of MRX34, a liposomal miR-34a mimic, in patients with advanced solid tumours. Br J Cancer. 2020;122(11):1630–7.
van Zandwijk N, Pavlakis N, Kao SC, Linton A, Boyer MJ, Clarke S, Huynh Y, Chrzanowska A, Fulham MJ, Bailey DL, et al. Safety and activity of microRNA-loaded minicells in patients with recurrent malignant pleural mesothelioma: a first-in-man, phase 1, open-label, dose-escalation study. Lancet Oncol. 2017;18(10):1386–96.
DiNardo CD, Erba HP, Freeman SD, Wei AH. Acute myeloid leukaemia. Lancet. 2023;401(10393):2073–86.
Wang Y, Chang YJ, Chen J, Han M, Hu J, Hu J, Huang H, Lai Y, Liu D, Liu Q, et al. Consensus on the monitoring, treatment, and prevention of leukaemia relapse after allogeneic haematopoietic stem cell transplantation in China: 2024 update. Cancer Lett. 2024;605:217264.
Nigro JM, Cho KR, Fearon ER, Kern SE, Ruppert JM, Oliner JD, Kinzler KW, Vogelstein B. Scrambled exons. Cell. 1991;64(3):607–13.
Cocquerelle C, Mascrez B, Hetuin D, Bailleul B. Mis-splicing yields circular RNA molecules. FASEB J. 1993;7(1):155–60.
Acknowledgements
Not applicable.
Funding
This work was supported by the National Key R&D Program of China (2022YFA1103300), the Natural Science Foundation of the Chongqing Innovation Group Science Program (cstc2021jcyjcxttX0001), the Youth Program of the National Natural Science Foundation of China (82300197), the Special Project for Talent Construction in Xinqiao Hospital (2022XKRC001), and the Young Talent Project of Xinqiao Hospital (2022YQB076).
Author information
Authors and Affiliations
Contributions
SL, XW, and YG wrote the manuscript and made the figures and tables. WY, WX, and YD collected the related literature. XP conceived the structure of the paper. XW provided critical revision of the manuscript and figures. XZ designed the review, critically revised and edited the manuscript. All authors reviewed the manuscript.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare no competing interests.
Additional information
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
About this article
Cite this article
Liu, S., Wan, X., Gou, Y. et al. The emerging functions and clinical implications of circRNAs in acute myeloid leukaemia. Cancer Cell Int 25, 167 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12935-025-03772-4
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12935-025-03772-4