Skip to main content
PeerJ logoLink to PeerJ
. 2024 Dec 18;12:e18733. doi: 10.7717/peerj.18733

The role of circRNA in breast cancer drug resistance

Shaofeng Yang 1,#, Donghai Li 1,✉,#
Editor: Vladimir Uversky
PMCID: PMC11662897  PMID: 39713143

Abstract

Among women with cancer, breast cancer has surpassed lung cancer to become the most prevalent type of cancer globally. High-throughput sequencing of breast cancer tissues from many patients has revealed significant variations in circRNA expression across different types of breast cancer. Chemotherapy is currently a very important method for treating breast cancer; however, as the number of chemotherapy sessions increases and considering factors such as the patient’s immune response, drug resistance has become a challenging issue in treating breast cancer. It is well known that drug resistance is associated with multiple factors, and different resistance mechanisms involve different roles of circRNA. This review consolidates literature from the past 5 years and addresses the shortcomings in the broad description of circRNA’s role in breast cancer drug resistance. It categorizes and describes the drug resistance and its mechanisms in different types of breast cancer, as well as the roles of circRNA and signaling pathways in drug resistance.

Keywords: BC, circRNA, Drug resistance, Antineoplastic

Introduction

Breast cancer (BC) is the most common type of cancer among everyday diseases and is also one of the leading causes of cancer-related deaths in women (Britt, Cuzick & Phillips, 2020; Harbeck et al., 2019). Due to the heterogeneity among patients and the different molecular types of tumors, breast cancer has varied treatment options and prognoses (Lüönd, Tiede & Christofori, 2021). Different subtypes of breast cancer are related to hormone receptors observed in breast cancer cells (Perou et al., 2000). CircRNA expression levels vary by breast cancer classification. With the deepening of circRNA research, it has been found to have a close relationship with breast cancer. CircRNA is a novel non-coding RNA (ncRNA) formed through the back-splicing of protein-coding genes. It plays roles in various diseases and biological processes (Zhou et al., 2023). The internal structure of circRNA is a highly stable circular molecule, which protects it from degradation by nucleases, thus providing potential for cancer prognosis or diagnosis (Wu et al., 2023). In breast cancer, circRNA can act as a tumor suppressor, influencing tumor occurrence, development, apoptosis, cell cycle, and tumor microenvironment (Ghafouri-Fard et al., 2021). Particularly, the expression of exosomal circRNA is higher in breast cancer tissues (De Palma et al., 2022). This high expression phenomenon facilitates subsequent research. Additionally, circRNA is related to breast cancer drug resistance. There are various mechanisms of drug resistance in breast cancer, such as endocrine resistance mechanisms (resistance to tamoxifen or aromatase inhibitors), chemotherapy resistance mechanisms (drug efflux), DNA damage repair (DDR) mechanisms, tumor microenvironment (TME) and tumor cell crosstalk mechanisms, targeted therapy mechanisms, etc (Zeng et al., 2018). Currently, research on circRNA in relation to breast cancer drug resistance often presents a singular description. In this review, we summarize the above mechanisms and the impact of circRNA expression on the occurrence and development of breast cancer, providing an in-depth supplement to the role of circRNA in breast cancer treatment. We also emphasize the application of circRNA as a molecular marker in breast cancer therapy.

Circrna generation and regulation

CircRNA generation: CircRNA is formed in circular transcription by reverse splicing of early messenger RNA (mRNA). During eukaryotic transcription, there is always a competition between linear transcription and reverse splicing. The presence of long introns, RBPs (RNA-binding proteins), and inverted repeat motifs facilitates reverse splicing, and the downstream splicing donor is brought closer to the upstream splice acceptor site by base-pairing of RBPs or inverted repeat motifs (Kristensen et al., 2019). Recent results from Drosophila suggest that U2snRNP depletion increases circRNA production compared with its linear counterpart (Liang et al., 2017). So when mRNA production stops, the nascent RNA can be redirected to different pathways that promote reverse splicing to produce circRNAs (Liang et al., 2017). In addition to the defective splicing mechanism described above, the flanking intron sequences on either side of the exon (i.e., splice donor site and splice acceptor site) can support efficient cyclization of different exons across eukaryotes (Kramer et al., 2015). Dimerization of RBP can bind to specific sequences of these flanking introns and facilitate the reverse splicing process (Conn et al., 2015; Errichelli et al., 2017; Verheijen & Pasterkamp, 2017). Recent studies have shown that there exists an intron-containing circRNA called ElciRNA, and the nuclear localization of ElciRNA and its association with PolII suggests that it may be involved in the regulation of transcription U1 snRNAs and EIciRNAs bind to each other through binding sites in EIciRNAs, and all three of them begin to promote gene transcription in the presence of PolII (Li et al., 2015b) (Fig. 1).

Figure 1. circRNA production and regulation.

Figure 1

Circrna expression in breast cancer

Upregulation of circRNA in different types of breast cancer

The role of circRNA in TNBC has been confirmed. On one hand, CircRNA can act on a signaling pathway to promote the initiation of bone remodeling factors and stimulate breast cancer metastasis to bone (Zhang et al., 2021). On the other hand, circBCBM1 affects the miR-125a/BRD4 axis, promoting brain metastasis of breast cancer (Huang & Zhu, 2021). Additionally, circZEB1 promotes proliferation and reduces apoptosis through the miR-448/eEF2K signaling pathway (Pei et al., 2020). An interesting study shows that circSEPT9 is overexpressed in TNBC tissues, and its high expression level is associated with advanced clinical stages and poor prognosis. Mechanistically, circSEPT9 can regulate the expression of leukemia inhibitory factor (LIF) by sponging miR-637, thereby activating the LIF/Stat3 signaling pathway involved in TNBC progression (Yi et al., 2023). Types such as circZEB1, circSKA3, circRNA-UCK2, circPDCD11, circ-UBAP2, circGFRA1, circEPSTI1, and circHIF1A are also highly expressed in TNBC, and their high expression is significantly related to the prognosis and tumor progression of TNBC (Huang & Zhu, 2021; Lyu et al., 2021; Nielsen et al., 2022; Pei et al., 2020; Xiang et al., 2019; Xing et al., 2021; Zheng et al., 2021). It is noteworthy that high expression of circRNAs can not only promote breast cancer metastasis but also have inhibitory effects. CircCDYL is also overexpressed in TNBC, promoting apoptosis and inhibiting proliferation through the miR-190a-3p/TP53INP1 axis, thereby upregulating the tumor suppressor TP53INP1 protein in TNBC (Geng et al., 2020). In HER2-positive breast cancer, ISH analysis shows that circCDYL in tumor tissues is elevated nearly 1.63 times compared to adjacent HER+BC tissues, and HER2 gene does not guide circCDYL expression in HER2 cells (Liang et al., 2021). Previous studies have shown that upregulation of circCDYL enhances proliferation and autophagy in HER2 cells (Geng et al., 2020). However, the specific biological function of circCDYL in HER2+BC cells still requires further exploration. In this study, qRT-PCR experiments validated that specific siRNA can silence circCDYL, which can consistently decrease or increase circCDYL in HER2+BC cells; overexpression of circCDYL promotes activation of the PI3K/AKT pathway in SK-RB-3 cells. CCK8 assays indicated that AKT inhibitors can partially impair the overexpression of circCDYL, suggesting that circCDYL promotes HER2+ proliferation by activating the PI3K/AKT pathway (Liang et al., 2021). Furthermore, circRNA is also related to resistance to trastuzumab; for example, circ-BGN located in the cytoplasm of trastuzumab-sensitive and resistant cells, with knockdown of circ-BGN significantly restoring sensitivity to trastuzumab (Wang et al., 2020). Additionally, circRNA not only affects the tumorigenesis of ER-positive breast cancer but can also serve as a genetic regulator influencing resistance to endocrine therapy in ER-positive breast cancer (Treeck, Haerteis & Ortmann, 2023). For instance, circRNA_0025202 interacts with miR-182-5p to upregulate FOXO3a expression, inhibiting tumor growth and tamoxifen resistance (Sang et al., 2019). Another molecule, circPVT1, is highly expressed in breast cancer cells and promotes tumorigenesis and endocrine resistance through miRNA sponging and protein scaffolding effects (Yi et al., 2023).

Downregulation of circRNA expression in different types of breast cancer

CircRNA not only plays a role through upregulation in TNBC but also affects proliferation, migration, and apoptosis through downregulation. An earlier analysis indicated that circAHNAK1 is downregulated in TNBC and acts by sequestering miR-421. Since miR-421 can inhibit the expression of the tumor suppressor gene RASA1, reducing miR-421 serves to suppress TNBC proliferation and metastasis (Xiao et al., 2019). According to results shared by Wang et al. (2021), downregulation of circWAC can increase the sensitivity of TNBC to chemotherapeutic drugs. This mechanism involves the circWAC/miR-142/WWP1 regulatory loop affecting drug sensitivity (Dragomir & Calin, 2018). Additionally, there is a tumor suppressor-associated circRNA, circNR3C2, whose low expression in TNBC is associated with metastasis and mortality (Fan et al., 2021). It is well known that both estrogen and circRNA play roles in breast cancer development. However, it is still undetermined whether estrogen induces circRNA expression. To investigate this, Gao, Zhang & Zhao (2018) treated cultured MCF7 cells with ribonuclease digestion and RNase R treatment and performed RNA sequencing, also sequencing a control group. The results showed that the estrogen-induced group produced a large number of coding genes. Subsequent CIRI2 analysis of circRNA-seq (Gao, Zhang & Zhao, 2018) revealed 23,830 and 25,525 circRNAs in the control and estrogen-treated groups, respectively. Experiments confirmed that estrogen induces circRNA expression. Notably, circPGR was identified. Experimental findings indicated that knockout of circPGR can inhibit, while overexpression promotes cell cycle progression, colony formation, and cell migration. Furthermore, reduction of circPGR significantly diminishes estrogen-induced tumorigenesis. Thus, circPGR is essential for the growth and tumor development of ER-positive breast cancer cells (Wang et al., 2021).

New advances in circrna in breast cancer drug resistance

The issue of drug resistance in breast cancer treatment, including drugs such as doxorubicin (Al-Malky, Al Harthi & Osman, 2020), tamoxifen (Yao et al., 2020), paclitaxel (Gao et al., 2021), and even targeted therapies, is one of the main obstacles in breast cancer prevention and treatment, leading to higher mortality rates and poor prognosis (Lee, Tan & Oon, 2018; Vasan, Baselga & Hyman, 2019). Although multidrug resistance (MDR) is currently a major barrier in treating breast cancer, significant progress has been made in recent years regarding its molecular pathways and regulatory mechanisms. Among them, non-coding RNAs are one of the most important regulatory pathways in cells. circRNA, being one of the most important members of non-coding RNAs due to its unique structure of an endless circular sequence without a 3′ cap and 5′ poly(A) tail, is especially notable. This structure makes it more stable than other non-coding RNAs (Dragomir & Calin, 2018). Furthermore, circRNA participates in various signal pathway cross-talk (such as MAPK/ERK and PTEN/PIK3/AKT pathways), protein binding, as well as sponging microRNA and interfering with the splicing of other RNAs to alter multidrug resistance (Huang et al., 2020). This intertwines different types of breast cancer resistance, sharing a common research basis.

Drug efflux

Drug efflux is related to multidrug resistance and is a mechanism of cancer cell resistance (Chen et al., 2016). Breast cancer resistance protein (BCRP/ABCG) is a transmembrane transport protein and belongs to the ATP-binding cassette (ABC) transporter family. Because the substrates of this protein include anticancer agents, chemotherapy drugs and targeted therapies are often countered by this non-selective transporter protein (Hegedus et al., 2009; Hoque et al., 2020). Membrane-localized pumps play a functional role in the drug efflux mechanism, and BCRP is involved in the functioning of (Al-Malky, Al Harthi & Osman, 2020) these membrane-localized pumps (Emran et al., 2022). In cancer tissues, the gene encoding BCRP is upregulated, indicating that the expression of BCRP in cancer tissues increases, which limits the effective delivery of chemotherapy drugs. In this mechanism, BCRP actively transports intracellular drugs across the membrane in the opposite direction, pumping the drugs out of the tumor cells and reducing drug accumulation within the body (Gillet, Efferth & Remacle, 2007; Pesic et al., 2006). Moreover, BCRP also regulates and alters drug concentrations and distribution inside and outside the cells, mediating resistance through glutathione-dependent drug efflux and preventing the drugs from reaching their intended targets (Domenichini, Adamska & Falasca, 2019). Studies have found that certain circRNAs play significant roles in cancer resistance related to BCRP efflux transporters (Liu et al., 2021). In osteosarcoma, knocking out circ-CHI3L1.2 can downregulate the expression levels of enzymes and genes, thereby reducing drug resistance (Zhang et al., 2021). The ATP-binding cassette sub-family B member 1 (ABCB1) can reverse the effect of circRNA_103615 on drug resistance (Liang et al., 2021). This suggests that circRNA may also affect certain transmembrane transport proteins in breast cancer, and discovering such RNA’s role could be pivotal in overcoming breast cancer resistance in the future.

Autophagy, apoptosis, and cancer development

The growth of tumors essentially represents an imbalance between beneficial cells and tumor cells, with the proliferation rate of tumor cells exceeding their death rate forming the basis for cancer development (Morana, Wood & Gregory, 2022). Chemotherapy is a major current method for cancer treatment, and its effectiveness depends on the extent of cell damage and the ability to activate cell apoptosis (Sánchez-Carvajal et al., 2021). When tumor cell damage decreases and apoptosis ability is insufficient, cancer cell resistance occurs. Alternative splicing (AS) is an important mechanism in apoptosis. AS consists of four components: the spliceosome, splice sites, cis-acting elements, and trans-acting factors. The spliceosome recognizes the sites and catalyzes the removal of introns, while cis-acting elements and trans-acting factors bind to gene sites (Kumagai-Takei et al., 2018; Tilgner et al., 2012; Yang et al., 2014). Defects in AS frequently occur in cancer and may be related to gene mutations and AS mechanism disorders. In this process, circRNA inhibits AS by binding to miRNA, with circRNA 100/146 binding and collecting multiple miRNAs, promoting cancer invasion (Chen et al., 2019). In various cancers related to humans, tumor heterogeneity and treatment-induced resistance are attributed to different tumor stem cells, which can self-renew, evade cytotoxic cells, and increase the expression of resistance-related genes (Meacham & Morrison, 2013). Current research has found that stem cell autophagy has become a necessary condition for maintaining normal tissue stem cells, and like normal tissue stem cells, tumor stem cells also express autophagy. For example, in human ductal carcinoma in situ, autophagy becomes associated with invasive breast spheres (Espina et al., 2010), and can also sensitize estrogen-positive breast cancer to tamoxifen-induced cytotoxicity (Qadir et al., 2008). Autophagy has been shown to be involved in tumor cell survival and treatment resistance (Levy, Towers & Thorburn, 2017). In breast cancer, circDNMT1 is detected at high expression levels, where it binds with tp53 protein in the cytoplasm and enters the nucleus (Du et al., 2018). Excessive accumulation of tp53 in the nucleus promotes the expression of autophagy-related genes, increasing autophagy in breast cancer. Additionally, circDNMT1 also binds with HNRNPD/AUF1 and promotes its entry into the nucleus, upregulating DNMT1 expression. High DNMT1 expression then inhibits tp53 gene transcription (Du et al., 2018). Moreover, circCDYL can also promote autophagy. CircCDYL acts as a molecular sponge to adsorb MIR1275, leading to upregulation of ATG7 and ULK1 expression in cells, promoting autophagy and resulting in increased breast cancer cell proliferation (Ma et al., 2019).

circRNA and chemotherapy drug resistance

In breast cancer, the resistance to paclitaxel is closely related to circRNA. Research has found a high correlation between paclitaxel resistance in breast cancer and a substance called circular RNA angiogenic protein-like 1 (circAMOTL1). This substance participates in the regulation of the AKT pathway and promotes the expression of anti-apoptotic proteins, leading to paclitaxel resistance in breast cancer patients (Yang et al., 2020). Additionally, reports indicate that circ-ABCB10 binds with let-7a-5p, increasing paclitaxel sensitivity and related cell apoptosis, while also inhibiting the invasion and autophagy of paclitaxel-resistant breast cancer cells (Zang et al., 2020). Other studies have shown that the expression of circ-RNF111 is increased in paclitaxel-resistant tissues and cells, and knocking out circ-RNF111 reduces paclitaxel’s effectiveness on breast cancer cells (Liang et al., 2019). Further research could reveal that circRNA acts as a sponge for miRNA, and the enrichment of miRNA may affect paclitaxel resistance, suggesting that joint studies of circRNA and microRNA might yield significant findings regarding paclitaxel resistance. Tamoxifen, used as an endocrine therapy for advanced breast cancer, still faces the challenge of tamoxifen resistance. Studies have found that knocking out circBMPR2 promotes apoptosis and increases tamoxifen resistance in breast cancer cells, whereas overexpression of circBMPR2 reduces tamoxifen resistance in breast cancer (Hu et al., 2020). The mechanism involves circBMPR2 sponging miR-553, which upregulates the expression of ubiquitin-specific protease 4 (USP4), thus inhibiting breast cancer development and resistance. Additionally, a novel hsa_circ_0025202 has been discovered, which can inhibit tumors and sensitize tamoxifen both in vitro and in vivo through the miR-182-5p/FOXO3a axis. Furthermore, upregulation of circ_UBE2D2 also increases tamoxifen resistance in breast cancer cells, while downregulation has the opposite effect (Johnson-Arbor & Dubey, 2024). In ER-positive breast cancer cells with tumor-associated macrophages (TAM), circRNA UBE2D2 (circ-UBE2D2) is upregulated, and TAM-resistant cells can release exosomes containing circRNA UBE2D2 to induce drug-sensitive BC cells to develop TAM resistance. Mechanistically, circRNA UBE2D2 interacts with miR-200a-3p, subsequently reducing functional promotion of metastasis and increasing TAM resistance. Doxorubicin, one of the most effective chemotherapy drugs, still faces issues of survival and mortality due to breast cancer resistance. Doxorubicin resistance may be related to the state of molecular pathways in the apoptosis pathway and DNA double strands in target cells (Xie & Zheng, 2022). Most components of these molecular pathways are regulated by circRNA. Upregulation of Circ_0085495 leads to doxorubicin resistance in breast cancer. This mechanism sponges miR-873-5p, thereby enhancing doxorubicin resistance in breast cancer cells. Conversely, knocking out Circ_0085495 can inhibit tumor growth and increase the sensitivity of tumor cells to doxorubicin in vivo (Xie & Zheng, 2022). Besides Circ_0085495, circ_0006528 and Circ_0001667 also affect doxorubicin resistance (Cui et al., 2022; Zhou et al., 2018).

circRNA and breast cancer prognosis

According to current data, circRNA plays a very important role in chemoresistance of breast cancer. With advancements in medical research, circRNA has become a therapeutic target for overcoming breast cancer resistance (Gao et al., 2020). The dysregulation of circRNA is closely related to breast cancer resistance. CircRNA is involved in endocrine resistance of breast cancer; first, circRNA sequesters miRNA and forms a ceRNA network to exert its effects, and second, the upregulation of circ_UBE2D2 in tamoxifen-resistant BC cells suggests its potential role (Hu et al., 2020; Liang et al., 2019). Additionally, downregulation of circBMPR2 suppresses tamoxifen-induced apoptosis, and restoration can reverse resistance (Nedeljković & Damjanović, 2019). Therefore, targeting circRNA-miRNA networks might be an effective approach for treating endocrine resistance in BC patients. Another factor is the impact of signaling pathways. Increasing evidence points to the relationship between breast cancer resistance and signaling pathways, including the loss of PTEN (phosphatase and tensin homolog) and the activation of PAM (phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signaling pathways), which significantly affect breast cancer resistance (Ali et al., 2017). Targeting signaling pathways to regulate downstream gene expression might be a method to eliminate BC resistance. Third, there is evidence that DNA damage repair (DDR) mechanisms are also involved in the development of breast cancer resistance. DDR mechanisms participate in repairing DNA damage and maintaining genome integrity and stability. CircRNA acts through these pathways in breast cancer prognosis; for example, elevated levels of circCDYL in breast cancer serum and tissues are associated with shortened survival and circCDYL can be used as a prognostic marker for breast cancer (Li et al., 2023). Furthermore, Hsa_circ_0067842 is elevated in breast cancer and is associated with worse outcomes (Smid et al., 2019). Plasma levels of hsa_circ_0001785 are closely related to breast cancer histological grading, TNM staging, and distant metastasis, indicating that hsa_circ_0001785 in plasma has potential diagnostic and predictive capabilities (Liu et al., 2018).

circRNA and breast cancer diagnosis

The uniqueness and biological functions of circRNA indicate its role in tumor growth, replication, metastasis, invasion, and drug resistance, suggesting that circRNA can be used as a biomarker for tumor diagnosis (Alhasan et al., 2016). Because circRNA can evade degradation induced by nucleases and, due to its circular structure, is more soluble in blood and plasma compared to linear RNA (Li et al., 2015a). Due to its stability and tissue specificity, circRNA has already been used as a biomarker for gastric cancer and liver cancer (Kulcheski, Christoff & Margis, 2016). With advancements in high-throughput sequencing and bioinformatics technologies, there is evidence suggesting that circRNA can contribute to tumorigenesis, providing new methods for identifying diagnostic biomarkers (Jiang et al., 2023).

circRNA and breast cancer treatment

Breast cancer stem cells (BCSCs) can resist DNA damage induced by chemotherapy drugs, leading to drug resistance (Mehraj et al., 2021). Interfering with DNA to induce mutations may increase the effectiveness of chemotherapy drugs. Tumor microenvironment (TME) cell crosstalk also promotes BC resistance. Various cells in the microenvironment interact, causing genetic alterations and uncontrolled growth and poor development, leading to hypoxia and acidosis in the microenvironment, which reshapes the TME. This reshaping of the TME can, in turn, affect cells by activating their resistance genes (Memczak et al., 2013). Additionally, circRNA can serve as a potential therapeutic target for breast cancer. The development of breast cancer involves multiple factors, and targeting downstream genes or proteins with circRNA is an important factor. circRNA RHOT1 promotes breast cancer progression by targeting the miR-204-5p/PRMT5 axis (Qu et al., 2019). The occurrence and progression of breast cancer are associated with dysregulated circRNA, so precisely targeting upregulated circRNA for unique back-splicing junctions (BSJ) for RNA interference could have therapeutic effects against tumors. Conversely, downregulating circRNA concentrations may also have specific anticancer effects. From a pharmacokinetic perspective, the high stability and long half-life of circRNA determine this effect (Sang et al., 2018). There have been exemplary studies on circRNA for breast cancer treatment. For instance, Sang et al. (2018) demonstrated that systemic injection of specific siRNAs targeting ciRS-7 inhibited migration and invasion of TNBC cells in a TNBC PDX mouse model, indicating that oncogenic ciRS-7 might be a potential therapeutic target for TNBC. They also reported that knockdown of ciRS-7 inhibits liver and lung metastases of TNBC cells in vivo by acting as a ceRNA in the sponge miR-1299. Thus, ciRS-7 exerts tumor-suppressive effects by regulating members of matrix metalloproteinases. Therefore, targeting the ciRS-7/miR-1299/MMP axis may be a new therapeutic strategy for TNBC (Zheng et al., 2021). Another strategy is that circRNA can sponge miRNA and RBP, becoming a potential carrier for breast cancer therapy. Zheng et al. (2021) validated that circGFRA1, as a sponge for miR-361-5p, promotes TNBC resistance to PTX through the TLR4 pathway. In vitro, silencing circGFRA1 upregulated the expression of miR-361-5p in PTX-treated TNBC PDX mouse models. Western blot results showed that after injecting lentivirus (LV-si-circGFRA1), TLR4 expression in PTX-treated mice was reduced. Furthermore, tumor size also decreased. Conversely, circGFRA1 and TLR4 were highly expressed in PDX mice treated with PTX alone, and tumor size increased (Tanaka et al., 2019). Additionally, circRNA can also regulate the expression of immune inhibitors through ceRNA networks, promoting tumor escape from immune surveillance. Tanaka et al. (2019) demonstrated that targeting CDR1-AS increases PD-L1 levels in CRC cells, induces T cell apoptosis, inhibits T cell activation and proliferation, ultimately leading to cancer immune escape (Conn et al., 2015). These methods provide new insights into breast cancer treatment, and more new mechanisms in circRNA therapy for breast cancer are yet to be discovered.

Discussion

Different subtypes of breast cancer have different circRNAs with high expression. Specifically, these high-expression circRNAs are related to two forms: one is the interaction and enrichment among miRNA-circRNA-lncRNA, which contributes to breast cancer drug resistance; the other is the formation of multiple drug resistances through different signaling pathways. While the processes may differ, the outcome converges on multiple drug resistance targets. We can step back and consider what affects the upstream expression of circRNAs to achieve multiple drug resistance in downstream target genes. For non-coding RNAs or signaling pathways, there might be some common factors stimulating circRNAs from upstream to control downstream targets. However, it is undeniable that circRNAs, as a highly conserved type of non-coding RNA, play a significant role in downstream enrichment or upstream integration. We could potentially produce point-to-point corrections targeting specific circRNAs, aiming to alter the adverse direction of breast cancer and possibly overcome current multiple drug resistance. It is worth noting that circRNAs, while regulating downstream target genes, can also serve as a cancer treatment method, participating in tumor occurrence and development. In the fight against breast cancer, circRNAs are playing an increasingly important role, and in-depth research on circRNAs could lead to essential breakthroughs in breast cancer treatment.

Funding Statement

This article was supported by the Natural Science Foundation of Inner Mongolia Autonomous Region (Grant No. 2022MS08010) and Graduate Student Excellence Program (YKDD2023ZY001). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Additional Information and Declarations

Competing Interests

The authors declare that they have no competing interests.

Author Contributions

Shaofeng Yang conceived and designed the experiments, performed the experiments, analyzed the data, prepared figures and/or tables, authored or reviewed drafts of the article, and approved the final draft.

Donghai Li conceived and designed the experiments, performed the experiments, analyzed the data, prepared figures and/or tables, authored or reviewed drafts of the article, and approved the final draft.

Data Availability

The following information was supplied regarding data availability:

This is a literature review.

References

  • Al-Malky, Al Harthi & Osman (2020).Al-Malky HS, Al Harthi SE, Osman AM. Major obstacles to doxorubicin therapy: cardiotoxicity and drug resistance. Journal of Oncology Pharmacy Practice. 2020;26(2):434–444. doi: 10.1177/1078155219877931. [DOI] [PubMed] [Google Scholar]
  • Alhasan et al. (2016).Alhasan AA, Izuogu OG, Al-Balool HH, Steyn JS, Evans A, Colzani M, Ghevaert C, Mountford JC, Marenah L, Elliott DJ, Santibanez-Koref M, Jackson MS. Circular RNA enrichment in platelets is a signature of transcriptome degradation. Blood. 2016;127(9):e1–e11. doi: 10.1182/blood-2015-06-649434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Ali et al. (2017).Ali R, Rakha EA, Madhusudan S, Bryant HE. DNA damage repair in breast cancer and its therapeutic implications. Pathology. 2017;49(2):156–165. doi: 10.1016/j.pathol.2016.11.002. [DOI] [PubMed] [Google Scholar]
  • Britt, Cuzick & Phillips (2020).Britt KL, Cuzick J, Phillips KA. Key steps for effective breast cancer prevention. Nature Reviews Cancer. 2020;20(8):417–436. doi: 10.1038/s41568-020-0266-x. [DOI] [PubMed] [Google Scholar]
  • Chen et al. (2019).Chen L, Nan A, Zhang N, Jia Y, Li X, Ling Y, Dai J, Zhang S, Yang Q, Yi Y, Jiang Y. Circular RNA 100146 functions as an oncogene through direct binding to miR-361-3p and miR-615-5p in non-small cell lung cancer. Molecular Cancer. 2019;18(1):13. doi: 10.1186/s12943-019-0943-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Chen et al. (2016).Chen Z, Shi T, Zhang L, Zhu P, Deng M, Huang C, Hu T, Jiang L, Li J. Mammalian drug efflux transporters of the ATP binding cassette (ABC) family in multidrug resistance: a review of the past decade. Cancer Letters. 2016;370(1):153–164. doi: 10.1016/j.canlet.2015.10.010. [DOI] [PubMed] [Google Scholar]
  • Conn et al. (2015).Conn SJ, Pillman KA, Toubia J, Conn VM, Salmanidis M, Phillips CA, Roslan S, Schreiber AW, Gregory PA, Goodall GJ. The RNA binding protein quaking regulates formation of circRNAs. Cell. 2015;160(6):1125–1134. doi: 10.1016/j.cell.2015.02.014. [DOI] [PubMed] [Google Scholar]
  • Cui et al. (2022).Cui Y, Fan J, Shi W, Zhou Z. Circ_0001667 knockdown blocks cancer progression and attenuates adriamycin resistance by depleting NCOA3 via releasing miR-4458 in breast cancer. Drug Development Research. 2022;83(1):75–87. doi: 10.1002/ddr.21845. [DOI] [PubMed] [Google Scholar]
  • De Palma et al. (2022).De Palma FDE, Salvatore F, Pol JG, Kroemer G, Maiuri MC. Circular RNAs as potential biomarkers in breast cancer. Biomedicines. 2022;10(3):725. doi: 10.3390/biomedicines10030725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Domenichini, Adamska & Falasca (2019).Domenichini A, Adamska A, Falasca M. ABC transporters as cancer drivers: potential functions in cancer development. Biochimica et Biophysica Acta (BBA)—General Subjects. 2019;1863(1):52–60. doi: 10.1016/j.bbagen.2018.09.019. [DOI] [PubMed] [Google Scholar]
  • Dragomir & Calin (2018).Dragomir M, Calin GA. Circular RNAs in cancer—lessons learned from microRNAs. Frontiers in Oncology. 2018;8:179. doi: 10.3389/fonc.2018.00179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Du et al. (2018).Du WW, Yang W, Li X, Awan FM, Yang Z, Fang L, Lyu J, Li F, Peng C, Krylov SN, Xie Y, Zhang Y, He C, Wu N, Zhang C, Sdiri M, Dong J, Ma J, Gao C, Hibberd S, Yang BB. A circular RNA circ-DNMT1 enhances breast cancer progression by activating autophagy. Oncogene. 2018;37(44):5829–5842. doi: 10.1038/s41388-018-0369-y. [DOI] [PubMed] [Google Scholar]
  • Emran et al. (2022).Emran TB, Shahriar A, Mahmud AR, Rahman T, Abir MH, Siddiquee MF, Ahmed H, Rahman N, Nainu F, Wahyudin E, Mitra S, Dhama K, Habiballah MM, Haque S, Islam A, Hassan MM. Multidrug resistance in cancer: understanding molecular mechanisms, immunoprevention and therapeutic approaches. Frontiers in Oncology. 2022;12:891652. doi: 10.3389/fonc.2022.891652. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Errichelli et al. (2017).Errichelli L, Dini Modigliani S, Laneve P, Colantoni A, Legnini I, Capauto D, Rosa A, De Santis R, Scarfò R, Peruzzi G, Lu L, Caffarelli E, Shneider NA, Morlando M, Bozzoni I. FUS affects circular RNA expression in murine embryonic stem cell-derived motor neurons. Nature Communications. 2017;8(1):14741. doi: 10.1038/ncomms14741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Espina et al. (2010).Espina V, Mariani BD, Gallagher RI, Tran K, Banks S, Wiedemann J, Huryk H, Mueller C, Adamo L, Deng J, Petricoin EF, Pastore L, Zaman S, Menezes G, Mize J, Johal J, Edmiston K, Liotta LA. Malignant precursor cells pre-exist in human breast DCIS and require autophagy for survival. PLOS ONE. 2010;5(4):e10240. doi: 10.1371/journal.pone.0010240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Fan et al. (2021).Fan Y, Wang J, Jin W, Sun Y, Xu Y, Wang Y, Liang X, Su D. CircNR3C2 promotes HRD1-mediated tumor-suppressive effect via sponging miR-513a-3p in triple-negative breast cancer. Molecular Cancer. 2021;20(1):25. doi: 10.1186/s12943-021-01321-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Gao et al. (2020).Gao L, Shen K, Yin N, Jiang M. Comprehensive transcriptomic analysis reveals dysregulated competing endogenous RNA network in endocrine resistant breast cancer cells. Frontiers in Oncology. 2020;10:600487. doi: 10.3389/fonc.2020.600487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Gao et al. (2021).Gao L, Wu ZX, Assaraf YG, Chen ZS, Wang L. Overcoming anti-cancer drug resistance via restoration of tumor suppressor gene function. Drug Resistance Updates. 2021;57(761):100770. doi: 10.1016/j.drup.2021.100770. [DOI] [PubMed] [Google Scholar]
  • Gao, Zhang & Zhao (2018).Gao Y, Zhang J, Zhao F. Circular RNA identification based on multiple seed matching. Briefings in Bioinformatics. 2018;19(5):803–810. doi: 10.1093/bib/bbx014. [DOI] [PubMed] [Google Scholar]
  • Geng et al. (2020).Geng X, Jia Y, Zhang Y, Shi L, Li Q, Zang A, Wang H. Circular RNA: biogenesis, degradation, functions and potential roles in mediating resistance to anticarcinogens. Epigenomics. 2020;12(3):267–283. doi: 10.2217/epi-2019-0295. [DOI] [PubMed] [Google Scholar]
  • Ghafouri-Fard et al. (2021).Ghafouri-Fard S, Hussen BM, Taheri M, Ayatollahi SA. Emerging role of circular RNAs in breast cancer. Pathology—Research and Practice. 2021;223(11):153496. doi: 10.1016/j.prp.2021.153496. [DOI] [PubMed] [Google Scholar]
  • Gillet, Efferth & Remacle (2007).Gillet JP, Efferth T, Remacle J. Chemotherapy-induced resistance by ATP-binding cassette transporter genes. Biochimica et Biophysica Acta (BBA)—Reviews on Cancer. 2007;1775(2):237–262. doi: 10.1016/j.bbcan.2007.05.002. [DOI] [PubMed] [Google Scholar]
  • Harbeck et al. (2019).Harbeck N, Penault-Llorca F, Cortes J, Gnant M, Houssami N, Poortmans P, Ruddy K, Tsang J, Cardoso F. Breast cancer. Nature Reviews Disease Primers. 2019;5(1):66. doi: 10.1038/s41572-019-0111-2. [DOI] [PubMed] [Google Scholar]
  • Hegedus et al. (2009).Hegedus C, Ozvegy-Laczka C, Szakács G, Sarkadi B. Interaction of ABC multidrug transporters with anticancer protein kinase inhibitors: substrates and/or inhibitors? Current Cancer Drug Targets. 2009;9(3):252–272. doi: 10.2174/156800909788166565. [DOI] [PubMed] [Google Scholar]
  • Hoque et al. (2020).Hoque KM, Dixon EE, Lewis RM, Allan J, Gamble GD, Phipps-Green AJ, Halperin Kuhns VL, Horne AM, Stamp LK, Merriman TR, Dalbeth N, Woodward OM. The ABCG2 Q141K hyperuricemia and gout associated variant illuminates the physiology of human urate excretion. Nature Communications. 2020;11(1):2767. doi: 10.1038/s41467-020-16525-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Hu et al. (2020).Hu K, Liu X, Li Y, Li Q, Xu Y, Zeng W, Zhong G, Yu C. Exosomes mediated transfer of Circ_UBE2D2 enhances the resistance of breast cancer to tamoxifen by binding to MiR-200a-3p. Medical Science Monitor. 2020;26:e922253. doi: 10.12659/MSM.922253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Huang et al. (2020).Huang A, Zheng H, Wu Z, Chen M, Huang Y. Circular RNA-protein interactions: functions, mechanisms, and identification. Theranostics. 2020;10(8):3503–3517. doi: 10.7150/thno.42174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Huang & Zhu (2021).Huang Y, Zhu Q. Mechanisms regulating abnormal circular RNA biogenesis in cancer. Cancers (Basel) 2021;13(16):4185. doi: 10.3390/cancers13164185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Jiang et al. (2023).Jiang W, Yu Y, Ou J, Li Y, Zhu N. Exosomal circRNA RHOT1 promotes breast cancer progression by targeting miR-204-5p/ PRMT5 axis. Cancer Cell International. 2023;23(1):260. doi: 10.1186/s12935-023-03111-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Johnson-Arbor & Dubey (2024).Johnson-Arbor K, Dubey R. Doxorubicin. StatPearls. Treasure Island (FL) ineligible companies. Disclosure: Ramin Dubey declares no relevant financial relationships with ineligible companies.: StatPearls Publishing, Copyright © 2024, StatPearls Publishing LLC. 2024.
  • Kramer et al. (2015).Kramer MC, Liang D, Tatomer DC, Gold B, March ZM, Cherry S, Wilusz JE. Combinatorial control of Drosophila circular RNA expression by intronic repeats, hnRNPs, and SR proteins. Genes & Development. 2015;29(20):2168–2182. doi: 10.1101/gad.270421.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Kristensen et al. (2019).Kristensen LS, Andersen MS, Stagsted LVW, Ebbesen KK, Hansen TB, Kjems J. The biogenesis, biology and characterization of circular RNAs. Nature Reviews Genetics. 2019;20(11):675–691. doi: 10.1038/s41576-019-0158-7. [DOI] [PubMed] [Google Scholar]
  • Kulcheski, Christoff & Margis (2016).Kulcheski FR, Christoff AP, Margis R. Circular RNAs are miRNA sponges and can be used as a new class of biomarker. Journal of Biotechnology. 2016;238(175):42–51. doi: 10.1016/j.jbiotec.2016.09.011. [DOI] [PubMed] [Google Scholar]
  • Kumagai-Takei et al. (2018).Kumagai-Takei N, Yamamoto S, Lee S, Maeda M, Masuzzaki H, Sada N, Yu M, Yoshitome K, Nishimura Y, Otsuki T. Inflammatory alteration of human T cells exposed continuously to asbestos. International Journal of Molecular Sciences. 2018;19(2):504. doi: 10.3390/ijms19020504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Lee, Tan & Oon (2018).Lee YT, Tan YJ, Oon CE. Molecular targeted therapy: treating cancer with specificity. European Journal of Pharmacology. 2018;834:188–196. doi: 10.1016/j.ejphar.2018.07.034. [DOI] [PubMed] [Google Scholar]
  • Levy, Towers & Thorburn (2017).Levy JMM, Towers CG, Thorburn A. Targeting autophagy in cancer. Nature Reviews Cancer. 2017;17(9):528–542. doi: 10.1038/nrc.2017.53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Li et al. (2015a).Li C, Chen J, Zhang K, Feng B, Wang R, Chen L. Progress and prospects of long noncoding RNAs (lncRNAs) in hepatocellular carcinoma. Cellular Physiology and Biochemistry. 2015a;36(2):423–434. doi: 10.1159/000430109. [DOI] [PubMed] [Google Scholar]
  • Li et al. (2023).Li J, Dong X, Kong X, Wang Y, Li Y, Tong Y, Zhao W, Duan W, Li P, Wang Y, Wang C. Circular RNA hsa_circ_0067842 facilitates tumor metastasis and immune escape in breast cancer through HuR/CMTM6/PD-L1 axis. Biology Direct. 2023;18(1):48. doi: 10.1186/s13062-023-00397-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Li et al. (2015b).Li Z, Huang C, Bao C, Chen L, Lin M, Wang X, Zhong G, Yu B, Hu W, Dai L, Zhu P, Chang Z, Wu Q, Zhao Y, Jia Y, Xu P, Liu H, Shan G. Exon-intron circular RNAs regulate transcription in the nucleus. Nature Structural & Molecular Biology. 2015b;22(3):256–264. doi: 10.1038/nsmb.2959. [DOI] [PubMed] [Google Scholar]
  • Liang et al. (2021).Liang H, Lin Z, Lin H, Zhao L, Huang W. circRNA_103615 contributes to tumor progression and cisplatin resistance in NSCLC by regulating ABCB1. Experimental and Therapeutic Medicine. 2021;22(3):934. doi: 10.3892/etm.2021.10366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Liang et al. (2019).Liang Y, Song X, Li Y, Ma T, Su P, Guo R, Chen B, Zhang H, Sang Y, Liu Y, Duan Y, Zhang N, Li X, Zhao W, Wang L, Yang Q. Targeting the circBMPR2/miR-553/USP4 axis as a potent therapeutic approach for breast cancer. Molecular Therapy—Nucleic Acids. 2019;17:347–361. doi: 10.1016/j.omtn.2019.05.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Liang et al. (2017).Liang D, Tatomer DC, Luo Z, Wu H, Yang L, Chen LL, Cherry S, Wilusz JE. The output of protein-coding genes shifts to circular RNAs when the pre-mRNA processing machinery is limiting. Molecular Cell. 2017;68(5):940–954.e943. doi: 10.1016/j.molcel.2017.10.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Liu et al. (2021).Liu XY, Zhang Q, Guo J, Zhang P, Liu H, Tian ZB, Zhang CP, Li XY. The role of circular RNAs in the drug resistance of cancers. Frontiers in Oncology. 2021;11:790589. doi: 10.3389/fonc.2021.790589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Liu et al. (2018).Liu F, Zhang J, Qin L, Yang Z, Xiong J, Zhang Y, Li R, Li S, Wang H, Yu B, Zhao W, Wang W, Li Z, Liu J. Circular RNA EIF6 (Hsa_circ_0060060) sponges miR-144-3p to promote the cisplatin-resistance of human thyroid carcinoma cells by autophagy regulation. Aging (Albany NY) 2018;10(12):3806–3820. doi: 10.18632/aging.101674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Lüönd, Tiede & Christofori (2021).Lüönd F, Tiede S, Christofori G. Breast cancer as an example of tumour heterogeneity and tumour cell plasticity during malignant progression. British Journal of Cancer. 2021;125(2):164–175. doi: 10.1038/s41416-021-01328-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Lyu et al. (2021).Lyu L, Zhang S, Deng Y, Wang M, Deng X, Yang S, Wu Y, Dai Z. Regulatory mechanisms, functions, and clinical significance of CircRNAs in triple-negative breast cancer. Journal of Hematology & Oncology. 2021;14(1):41. doi: 10.1186/s13045-021-01052-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Ma et al. (2019).Ma J, Fang L, Yang Q, Hibberd S, Du WW, Wu N, Yang BB. Posttranscriptional regulation of AKT by circular RNA angiomotin-like 1 mediates chemoresistance against paclitaxel in breast cancer cells. Aging (Albany NY) 2019;11(23):11369–11381. doi: 10.18632/aging.102535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Meacham & Morrison (2013).Meacham CE, Morrison SJ. Tumour heterogeneity and cancer cell plasticity. Nature. 2013;501(7467):328–337. doi: 10.1038/nature12624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Mehraj et al. (2021).Mehraj U, Dar AH, Wani NA, Mir MA. Tumor microenvironment promotes breast cancer chemoresistance. Cancer Chemotherapy and Pharmacology. 2021;87(2):147–158. doi: 10.1007/s00280-020-04222-w. [DOI] [PubMed] [Google Scholar]
  • Memczak et al. (2013).Memczak S, Jens M, Elefsinioti A, Torti F, Krueger J, Rybak A, Maier L, Mackowiak SD, Gregersen LH, Munschauer M, Loewer A, Ziebold U, Landthaler M, Kocks C, le Noble F, Rajewsky N. Circular RNAs are a large class of animal RNAs with regulatory potency. Nature. 2013;495(7441):333–338. doi: 10.1038/nature11928. [DOI] [PubMed] [Google Scholar]
  • Morana, Wood & Gregory (2022).Morana O, Wood W, Gregory CD. The apoptosis paradox in cancer. International Journal of Molecular Sciences. 2022;23(3):1328. doi: 10.3390/ijms23031328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Nedeljković & Damjanović (2019).Nedeljković M, Damjanović A. Mechanisms of chemotherapy resistance in triple-negative breast cancer—how we can rise to the challenge. Cells. 2019;8(9):957. doi: 10.3390/cells8090957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Nielsen et al. (2022).Nielsen AF, Bindereif A, Bozzoni I, Hanan M, Hansen TB, Irimia M, Kadener S, Kristensen LS, Legnini I, Morlando M, Jarlstad Olesen MT, Pasterkamp RJ, Preibisch S, Rajewsky N, Suenkel C, Kjems J. Best practice standards for circular RNA research. Nature Methods. 2022;19(10):1208–1220. doi: 10.1038/s41592-022-01487-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Pei et al. (2020).Pei X, Zhang Y, Wang X, Xue B, Sun M, Li H. Circular RNA circ-ZEB1 acts as an oncogene in triple negative breast cancer via sponging miR-448. The International Journal of Biochemistry & Cell Biology. 2020;126(2019):105798. doi: 10.1016/j.biocel.2020.105798. [DOI] [PubMed] [Google Scholar]
  • Perou et al. (2000).Perou CM, Sørlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, Pollack JR, Ross DT, Johnsen H, Akslen LA, Fluge O, Pergamenschikov A, Williams C, Zhu SX, Lønning PE, Børresen-Dale AL, Brown PO, Botstein D. Molecular portraits of human breast tumours. Nature. 2000;406(6797):747–752. doi: 10.1038/35021093. [DOI] [PubMed] [Google Scholar]
  • Pesic et al. (2006).Pesic M, Markovic JZ, Jankovic D, Kanazir S, Markovic ID, Rakic L, Ruzdijic S. Induced resistance in the human non small cell lung carcinoma (NCI-H460) cell line in vitro by anticancer drugs. Journal of Chemotherapy. 2006;18(1):66–73. doi: 10.1179/joc.2006.18.1.66. [DOI] [PubMed] [Google Scholar]
  • Qadir et al. (2008).Qadir MA, Kwok B, Dragowska WH, To KH, Le D, Bally MB, Gorski SM. Macroautophagy inhibition sensitizes tamoxifen-resistant breast cancer cells and enhances mitochondrial depolarization. Breast Cancer Research and Treatment. 2008;112(3):389–403. doi: 10.1007/s10549-007-9873-4. [DOI] [PubMed] [Google Scholar]
  • Qu et al. (2019).Qu Y, Dou P, Hu M, Xu J, Xia W, Sun H. circRNA‐CER mediates malignant progression of breast cancer through targeting the miR‐136/MMP13 axis. Molecular Medicine Reports. 2019;19:3314–3320. doi: 10.3892/mmr.2019.9965. [DOI] [PubMed] [Google Scholar]
  • Sánchez-Carvajal et al. (2021).Sánchez-Carvajal JM, Ruedas-Torres I, Carrasco L, Pallarés FJ, Mateu E, Rodríguez-Gómez IM, Gómez-Laguna J. Activation of regulated cell death in the lung of piglets infected with virulent PRRSV-1 Lena strain occurs earlier and mediated by cleaved Caspase-8. Veterinary Research. 2021;52(1):12. doi: 10.1186/s13567-020-00882-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Sang et al. (2019).Sang Y, Chen B, Song X, Li Y, Liang Y, Han D, Zhang N, Zhang H, Liu Y, Chen T, Li C, Wang L, Zhao W, Yang Q. circRNA_0025202 regulates tamoxifen sensitivity and tumor progression via regulating the miR-182-5p/FOXO3a axis in breast cancer. Molecular Therapy. 2019;27(9):1638–1652. doi: 10.1016/j.ymthe.2019.05.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Sang et al. (2018).Sang M, Meng L, Liu S, Ding P, Chang S, Ju Y, Liu F, Gu L, Lian Y, Geng C. Circular RNA ciRS-7 maintains metastatic phenotypes as a ceRNA of miR-1299 to target MMPs. Molecular Cancer Research. 2018;16(11):1665–1675. doi: 10.1158/1541-7786.MCR-18-0284. [DOI] [PubMed] [Google Scholar]
  • Smid et al. (2019).Smid M, Wilting SM, Uhr K, Rodríguez-González FG, de Weerd V, Prager-Van der Smissen WJC, van der Vlugt-Daane M, van Galen A, Nik-Zainal S, Butler A, Martin S, Davies HR, Staaf J, van de Vijver MJ, Richardson AL, MacGrogan G, Salgado R, van den Eynden G, Purdie CA, Thompson AM, Caldas C, Span PN, Sweep F, Simpson PT, Lakhani SR, Van Laere S, Desmedt C, Paradiso A, Eyfjord J, Broeks A, Vincent-Salomon A, Futreal AP, Knappskog S, King T, Viari A, Børresen-Dale AL, Stunnenberg HG, Stratton M, Foekens JA, Sieuwerts AM, Martens JWM. The circular RNome of primary breast cancer. Genome Research. 2019;29(3):356–366. doi: 10.1101/gr.238121.118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Tanaka et al. (2019).Tanaka E, Miyakawa Y, Kishikawa T, Seimiya T, Iwata T, Funato K, Odawara N, Sekiba K, Yamagami M, Suzuki T, Ishibashi R, Otsuka M, Koike K. Expression of circular RNA CDR1‐AS in colon cancer cells increases cell surface PD‐L1 protein levels. Oncology Reports. 2019;42:1459–1466. doi: 10.3892/or.2019.7244. [DOI] [PubMed] [Google Scholar]
  • Tilgner et al. (2012).Tilgner H, Knowles DG, Johnson R, Davis CA, Chakrabortty S, Djebali S, Curado J, Snyder M, Gingeras TR, Guigó R. Deep sequencing of subcellular RNA fractions shows splicing to be predominantly co-transcriptional in the human genome but inefficient for lncRNAs. Genome Research. 2012;22(9):1616–1625. doi: 10.1101/gr.134445.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Treeck, Haerteis & Ortmann (2023).Treeck O, Haerteis S, Ortmann O. Non-coding RNAs modulating estrogen signaling and response to endocrine therapy in breast cancer. Cancers (Basel) 2023;15(6):1632. doi: 10.3390/cancers15061632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Vasan, Baselga & Hyman (2019).Vasan N, Baselga J, Hyman DM. A view on drug resistance in cancer. Nature. 2019;575(7782):299–309. doi: 10.1038/s41586-019-1730-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Verheijen & Pasterkamp (2017).Verheijen BM, Pasterkamp RJ. Commentary: FUS affects circular RNA expression in murine embryonic stem cell-derived motor neurons. Frontiers in Molecular Neuroscience. 2017;10:412. doi: 10.3389/fnmol.2017.00412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Wang et al. (2020).Wang S, Liu F, Ma H, Cui X, Yang S, Qin R. circCDYL acts as a tumor suppressor in triple negative breast cancer by sponging miR-190a-3p and upregulating TP53INP1. Clinical Breast Cancer. 2020;20(5):422–430. doi: 10.1016/j.clbc.2020.04.006. [DOI] [PubMed] [Google Scholar]
  • Wang et al. (2021).Wang L, Yi J, Lu LY, Zhang YY, Wang L, Hu GS, Liu YC, Ding JC, Shen HF, Zhao FQ, Huang HH, Liu W. Estrogen-induced circRNA, circPGR, functions as a ceRNA to promote estrogen receptor-positive breast cancer cell growth by regulating cell cycle-related genes. Theranostics. 2021;11(4):1732–1752. doi: 10.7150/thno.45302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Wu et al. (2023).Wu S, Wu Y, Deng S, Lei X, Yang X. Emerging roles of noncoding RNAs in human cancers. Discover Oncology. 2023;14(1):128. doi: 10.1007/s12672-023-00728-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Xiang et al. (2019).Xiang Z, Xu C, Wu G, Liu B, Wu D. CircRNA-UCK2 increased TET1 inhibits proliferation and invasion of prostate cancer cells via sponge MiRNA-767-5p. Open Medicine (Wars) 2019;14(1):833–842. doi: 10.1515/med-2019-0097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Xiao et al. (2019).Xiao W, Zheng S, Zou Y, Yang A, Xie X, Tang H, Xie X. CircAHNAK1 inhibits proliferation and metastasis of triple-negative breast cancer by modulating miR-421 and RASA1. Aging (Albany NY) 2019;11(24):12043–12056. doi: 10.18632/aging.102539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Xie & Zheng (2022).Xie H, Zheng R. Circ_0085495 knockdown reduces adriamycin resistance in breast cancer through miR-873-5p/integrin β1 axis. Anti-Cancer Drugs. 2022;33(1):e166–e177. doi: 10.1097/CAD.0000000000001174. [DOI] [PubMed] [Google Scholar]
  • Xing et al. (2021).Xing Z, Wang R, Wang X, Liu J, Zhang M, Feng K, Wang X. CircRNA circ-PDCD11 promotes triple-negative breast cancer progression via enhancing aerobic glycolysis. Cell Death Discovery. 2021;7(1):218. doi: 10.1038/s41420-021-00604-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Yang et al. (2020).Yang W, Gong P, Yang Y, Yang C, Yang B, Ren L. Circ-ABCB10 contributes to paclitaxel resistance in breast cancer through let-7a-5p/DUSP7 axis. Cancer Management Research. 2020;12:2327–2337. doi: 10.2147/cmar.S238513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Yang et al. (2014).Yang J, Hung LH, Licht T, Kostin S, Looso M, Khrameeva E, Bindereif A, Schneider A, Braun T. RBM24 is a major regulator of muscle-specific alternative splicing. Developmental Cell. 2014;31(1):87–99. doi: 10.1016/j.devcel.2014.08.025. [DOI] [PubMed] [Google Scholar]
  • Yao et al. (2020).Yao J, Deng K, Huang J, Zeng R, Zuo J. Progress in the understanding of the mechanism of tamoxifen resistance in breast cancer. Frontiers in Pharmacology. 2020;11:592912. doi: 10.3389/fphar.2020.592912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Yi et al. (2023).Yi J, Wang L, Hu GS, Zhang YY, Du J, Ding JC, Ji X, Shen HF, Huang HH, Ye F, Liu W. CircPVT1 promotes ER-positive breast tumorigenesis and drug resistance by targeting ESR1 and MAVS. The EMBO Journal. 2023;42(10):e112408. doi: 10.15252/embj.2022112408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Zang et al. (2020).Zang H, Li Y, Zhang X, Huang G. Circ-RNF111 contributes to paclitaxel resistance in breast cancer by elevating E2F3 expression via miR-140-5p. Thoracic Cancer. 2020;11(7):1891–1903. doi: 10.1111/1759-7714.13475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Zeng et al. (2018).Zeng K, He B, Yang BB, Xu T, Chen X, Xu M, Liu X, Sun H, Pan Y, Wang S. The pro-metastasis effect of circANKS1B in breast cancer. Molecular Cancer. 2018;17(1):160. doi: 10.1186/s12943-018-0914-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Zhang et al. (2021).Zhang Z, Zhou Q, Luo F, Zhou R, Xu J, Xiao J, Dai F, Song L. Circular RNA circ-CHI3L1.2 modulates cisplatin resistance of osteosarcoma cells via the miR-340-5p/LPAATβ axis. Human Cell. 2021;34(5):1558–1568. doi: 10.1007/s13577-021-00564-6. [DOI] [PubMed] [Google Scholar]
  • Zheng et al. (2021).Zheng SR, Huang QD, Zheng ZH, Zhang ZT, Guo GL. circGFRA1 affects the sensitivity of triple-negative breast cancer cells to paclitaxel via the miR-361-5p/TLR4 pathway. The Journal of Biochemistry. 2021;169(5):601–611. doi: 10.1093/jb/mvaa148. [DOI] [PubMed] [Google Scholar]
  • Zhou et al. (2023).Zhou Q, Boeckel JN, Yao J, Zhao J, Bai Y, Lv Y, Hu M, Meng D, Xie Y, Yu P, Xi P, Xu J, Zhang Y, Dimmeler S, Xiao J. Diagnosis of acute myocardial infarction using a combination of circulating circular RNA cZNF292 and clinical information based on machine learning. MedComm. 2023;4(3):e299. doi: 10.1002/mco2.299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • Zhou et al. (2018).Zhou R, Wu Y, Wang W, Su W, Liu Y, Wang Y, Fan C, Li X, Li G, Li Y, Xiong W, Zeng Z. Circular RNAs (circRNAs) in cancer. Cancer Letters. 2018;425(1):134–142. doi: 10.1016/j.canlet.2018.03.035. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

The following information was supplied regarding data availability:

This is a literature review.


Articles from PeerJ are provided here courtesy of PeerJ, Inc

RESOURCES