Skip to main content

Main menu

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Advertisers
    • Editorial Board
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
  • About Us
    • General Policy
    • Contact
  • Other Publications
    • Cancer Genomics & Proteomics
    • Anticancer Research
    • In Vivo

User menu

  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
Cancer Genomics & Proteomics
  • Other Publications
    • Cancer Genomics & Proteomics
    • Anticancer Research
    • In Vivo
  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart
Cancer Genomics & Proteomics

Advanced Search

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Advertisers
    • Editorial Board
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
  • About Us
    • General Policy
    • Contact
  • Visit iiar on Facebook
  • Follow us on Linkedin
Review ArticleReview
Open Access

Breast Cancer: Circular RNAs Mediating Efficacy in Preclinical In Vivo Models

ULRICH H. WEIDLE, HUNG-EN HSIA and ULRICH BRINKMANN
Cancer Genomics & Proteomics May 2023, 20 (3) 222-238; DOI: https://doi.org/10.21873/cgp.20377
ULRICH H. WEIDLE
Roche Pharma Research and Early Development (pRED), Large Molecule Research, Roche Innovation Center Munich, Penzberg, Germany
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: weidle49@t-online.de
HUNG-EN HSIA
Roche Pharma Research and Early Development (pRED), Large Molecule Research, Roche Innovation Center Munich, Penzberg, Germany
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
ULRICH BRINKMANN
Roche Pharma Research and Early Development (pRED), Large Molecule Research, Roche Innovation Center Munich, Penzberg, Germany
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: ulrich.brinkmann@roche.com
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

In order to identify new targets and treatment modalities for breast cancer, we searched the literature for circular RNAs (circRNAs) with efficacy in preclinical breast cancer-related in vivo models. From our search, we identified 26 up-regulated and six down-regulated circRNAs which mediate efficacy in breast cancer-related preclinical in vivo models. We discuss reconstitution and inhibition of the identified circRNAs, as well as druggability and validation of the targets identified in the context of chemoresistance, inhibition of proliferation and metastasis. Pathways driven by suppressors of cytokines and high-mobility group proteins, nuclear factor Embedded ImageB and Hippo signaling emerged as important drivers of tumor growth and metastasis. The role of trefoil factor-1 with respect to metastasis of estrogen receptor-positive breast cancer also merits further investigation. In addition, mucin 19 has emerged as an unexplored target for treatment of breast cancer.

Key Words
  • Bone metastasis
  • chemoresistance
  • signaling
  • circRNA
  • si-RNA
  • sh-RNA
  • review

Breast cancer (BC) is the most common malignancy in women worldwide and occurs as ductal and lobular carcinomas (1). According to the expression of hormone receptors (HR) such as estrogen-receptor (ER), progesterone receptor and human epidermal growth factor receptor 2 (HER2), the following subtypes have been defined: Luminal A (HR+, HER2−), luminal B (HR+, HER2+), triple-negative BC (HR−, HER2−) and HER2 enriched (2). The treatment of BC is dependent on the molecular subtype and includes surgery, radiotherapy, chemotherapy and targeted therapy with agents such as tamoxifen, aromatase inhibitors, trastuzumab, pertuzumab, Kadcyla (trastuzumab-emtansine), lapatinib, poly-(ADP)-ribose polymerase inhibitors for BC with BRCA DNA repair-associated mutations, cyclin-dependent kinase (CDK) 4/6 inhibitors, tumor spread-inhibiting bisphosphonates and recently also immunotherapy with pembrolizumab (Keytruda) (3-6). BC is curable in 70-80% of patients with early-stage, non-metastatic disease (3). BC metastasizes to the bones, lungs, regional lymph nodes, liver and brain and metastases only poorly respond to therapies (7, 8). Another problem is the development of resistance to chemotherapy and targeted therapies (9, 10). In order to identify new targets and treatment modalities for BC, we searched the literature for circular RNAs (circRNAs) with efficacy in preclinical BC-related in vivo models. We excluded triple-negative BC because relevant circRNAs in this subtype will be summarized in a separate review.

Circular RNA

CircRNAs are generated by back-splicing of polymerase II transcripts, thus creating new junctions (11). Their size ranges from a hundred to several thousands of nucleotides (11). In humans, common size of a circRNA is a few hundred nucleotides comprising two to three exons (12). In mammalian cells, at least 30,000 different circRNAs have been identified (13). They affect processes such as transcription, splicing, protein scaffolding, and can act as micro-RNA (miR) sponges and decoys; in rare cases they can even function as translational templates (11-13). In BC they can act as tumor suppressors, as well as oncogenes affecting processes such as tumor initiation, progression, proliferation, cell-cycle, cell death, migration, invasion, metastasis, angiogenesis, modulation of the tumor microenvironment and chemoresistance (14-16). CircRNAs can be detected in peripheral blood encapsulated in exosomes and their roles as biomarkers, predictors of survival, metastasis and drug resistance are under active investigation (17, 18). The physiological relevance of circRNAs in cancer has recently been supported by the fact that inhibition of individual circRNAs can inhibit tumor growth in patient-derived xenografts of lung adenocarcinoma and gastric cancer (19, 20).

CircRNAs Down-regulated in BC

circRNAs affecting chemoresistance

Circ-lysine-specific demethylase 4C (circKDM4C) targets phenazine biosynthesis-like domain-containing protein (PBLD). CircKDM4C (Figure 1) was down-regulated in BC and its down-regulation correlated with poor prognosis (21). CircKDM4C repressed BC cell proliferation, metastasis and adriamycin resistance in vitro and in vivo. Mechanistically, it sponged miR-548p, which led to up-regulation of PBLD. In hepatocellular carcinoma (HCC), reduced expression of PBLD correlated with poor prognosis and forced expression of PBLD inhibited HCC growth in vitro and in vivo by interfering with mitogen-activated protein kinase (MAPK), nuclear factor Embedded ImageB (NFEmbedded ImageB), epithelial–mesenchymal transition (EMT) and angiogenesis signaling pathways (22).

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

Circular (circ) RNAs down-regulated in breast cancer with efficacy in preclinical in vivo models. CircRNAs involved in adriamycin (ADM) and tamoxifen (TAM) resistance, as well as signaling-related circRNAs are shown. Up- and down-regulation are indicated by up and down arrows, respectively. ERK1/2: Extracellular signal-regulated kinases 1,2; FOXO3A: forkhead-box protein O3A; HIPK3: homeodomain-interacting protein kinase 3; JAK: Janus kinase; KDM4C: lysine-specific demethylase 4C; MEK1: mitogen-activated protein kinase 1; MET: metastasis; miR: microRNA; NOL10: nucleolar protein 10; PBLD: phenazine biosynthesis-like domain-containing protein; SOCS 2,3: suppressor of cytokine signaling 2,3; STAT: signal transducer and activator of transcription; WNK2: WNK lysine-deficient protein kinase 2.

Circ0025202 targets forkhead-box-protein O3A (FOXO3A). Low expression of circ0025202 (Figure 1) was found in BC tissues (23). Overexpression of circ0025202 reversed tamoxifen resistance of MCF-7/TR cells (23). In vivo, it suppressed tumor growth and increased tamoxifen sensitivity of MCF-7/TR cells in a xenograft model. Mechanistically, circ0025202 acted as a sponge for miR-182-5p, and resulting in up-regulation of FOXO3A. The latter is a transcription factor which is involved in apoptosis, proliferation, cell-cycle progression, DNA damage and tumorigenesis. FOXO3A is frequently inactivated by mutation in tumors or sequestrated in the cytoplasm and overexpression of FOXO3 inhibits proliferation, tumorigenic potential and invasiveness, EMT and metastasis (24-26). FOXO3A-driven miR signatures modulate vascular endothelial growth factor/neuropilin 1 signaling and BC metastasis (27).

Circ0025202 targets homeodomain-interacting protein kinase 3 (HIPK3). Circ0025202 (Figure 1) was reduced in BC tissues and tamoxifen-resistant BC cells (MCF-7 and T47D) (28). Knockdown of circ0025202 elevated the half-maximal inhibitory concentration for tamoxifen, promoted cell proliferation, invasion and migration, and mediated cell-cycle progression and inhibition of apoptosis in vitro (28). Up-regulation of circ0025202 hindered xenograft growth of MCF-7/TR and promoted tamoxifen sensitivity in nude mice. Circ0025202 targeted miR-197-3p and thus led to up-regulation of HIPK3 (28). The latter is a member of the ser/thr kinase family with three members (HIK1, −2 and −3) and interacts with homeobox proteins and other transcription factors as transcriptional co-activators or co-repressors (29). HIPK2 acts as a suppressor of development and metastasis of many types of tumors, down-regulates vimentin and inhibits BC cell invasion (30). HIPK3 overexpression can inhibit growth of non-small-cell lung carcinoma (31) and drives p53 activation to limit colorectal cancer growth (32). HIPK1 is involved in DNA repair through its interaction with p53 (33).

circRNAs affecting suppressor of cytokine 2,3 (SOCS2,3) signaling

Circ-nucleolar protein 10 (circNOL10) targets suppressor of cytokine signaling 2 (SOCS2). CircNOL10 (Figure 1) was down-regulated in BC tissues and cell lines (34). In BT-549 and MDA-MB-231 BC cells, circNOL10 suppressed proliferation, migration, invasion, EMT by sponging miR-767 which resulted in activation of SOCS2 and inhibition of Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling. In nude mice, circNOL10 suppressed the growth of BT-549 BC xenografts.

Circ0001785 targets SOCS3. Circ0001785 (Figure 1) was reduced in T47D, MCF-7, MDA-MB-453, MDA-MB-231 and BT-549 BC cells in comparison to MCF-10A normal breast cells. It inhibited proliferation, invasion and migration of BC cells, as well as tumor growth in nude mice by sponging miR-942 through up-regulation of SOCS3 (35).

SOCS family members inhibit JAK, signal transducers and signal transducer and activator of transcription (STAT) are down-regulated in BC (36, 37). In BC, loss of SOCS2 is related to cell proliferation and tumor growth (38). In patients with BC with lymph node metastasis, reduced expression of SOCS3 was found (39).

Circ001666 targets WNK lysine-deficient protein kinase 2 (WNK2). Circ001666 (Figure 1) was down-regulated in BC tissues and cell lines (40). It inhibited proliferation, migration, invasion and promoted apoptosis of BC cells in vitro and tumor growth of BC xenografts in nude mice. Circ001666 sponged miR-620, resulting in up-regulation of WNK2 tumor suppressors. WNK2 is a ser/thr kinase, with four paralogs, which inhibits cell proliferation by modulating the activation of MAPK/extracellular signal regulated kinase 1/2 (ERK1/2) (MEK1/ERK1/2) and negatively regulates sodium transport (41). WNK2 is down-regulated in BC and inhibits BC cell proliferation (42). During pancreatic ductal adenocarcinoma development, WNK2 was also found to be down-regulated (43). However, its function seems to be context-dependent, because in HCC, WNK2 acts as a driver of carcinogenesis and is a risk factor of early recurrence (44).

CircRNAs Up-regulated in BC

circRNAs mediating chemoresistance

Circ-ATP-binding cassette subfamily B member 10 (circABCD10) targets dual-specificity phosphatase 7 (DUSP7). An abundance of circABCD10 (Figure 2A) was associated with reduced sensitivity to paclitaxel in BC tissues and cells (45). In paclitaxel-resistant BC cell lines MCF-7/PTX and MCF-MB-231/PTX, circABCD10 sponged let-7a-5p and led to up-regulation of DUSP7. In nude mice, knockdown of circABCD10 suppressed growth of MCF-7/PTX xenografts (45). The DUSP family exert their functions through dephosphorylation of MAPK (ERK1/2, JUN kinase p38) (46). DUSP7 acts as an oncogene in BC (47). DUSPs play an emerging role in human cancer (48). Paclitaxel is widely used for treatment of early and advanced BC (49).

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

Chemoresistance-related circular (circ) RNAs up-regulated in breast cancer with efficacy in preclinical in vivo models. A: CircRNAs conferring paclitaxel (PTX) resistance. B: CircRNAs conferring adriamycin (ADM) resistance. C: CircRNAs conferring tamoxifen (TAM) or lapatinib resistance (LR). Up- and down-regulation are indicated by up and down arrows, respectively. ABCD10: ATP-binding cassette subfamily B member 10; ANLN: anillin; CDK8: cyclin-dependent kinase 8; CHD4: chromodomain helicase DNA-binding protein; DUSP7: dual specificity phosphatase 7; E2F3: transcription factor E2F3; ERα: estrogen receptor α; HIPK3: homeodomain-interacting protein kinase 3; HK2: hexokinase 2; INTβ1: integrin β1; LR: lapatinib resistant; MMP11: matrix metalloprotease 11; NCOA3: nuclear receptor co-activator A3; RNF111: ring finger protein 111; UBE2D2: ubiquitin-conjugating enzyme 2 D2.

Circ-ring finger protein 111 (circRNF111) targets transcription factor E2F3. High expression of circRNF111 (Figure 2A) was observed in paclitaxel-resistant BC tissues and cell lines (50). CircRNF111 knockdown suppressed paclitaxel resistance, cell viability, colony formation, invasion and glycolysis in paclitaxel-resistant BC cells MCF-7/PTX and MDA-MB-231/PTX. In nude mice, knockdown of circRNF111 in MCF-7/PTX xenografts suppressed paclitaxel-resistance. CircRNF111 sponged miR-140-5p resulting in up-regulation of E2F3 (50). The latter is a member of the E2F family of transcription factors which are deregulated in BC (51). E2F3 is a driver of EMT, cell invasion and metastasis in BC (52, 53).

CircHIPK3 targets HK2. CircHIPK3 (Figure 2A) was upregulated in paclitaxel-resistant BC tissues and cell lines such as MCF-7/PTX and MDA-MB-231/PTX (54). Silencing of circHIPK3 enhanced drug sensitivity in vitro due to sponging of miR-1286 and up-regulation of HK2. In nude mice, silencing of circHIPK3 diminished tumor growth and promoted paclitaxel sensitivity of MDA-MB-231/PTX xenografts (54). HK2 is a tumor driver in BC and is involved in chemoresistance of BC (55, 56). It was shown that Pim2 proto-oncogene serine protein kinase (PIM2)-mediated phosphorylation of HIPK3 is critical for tumor growth and paclitaxel-resistance in BC (57).

Circ0006528 targets CDK8. Circ0006528 (Figure 2A) was up-regulated in BC tissues and paclitaxel-resistant BC cell lines BT549/PTX and ZR-75-30-PTX (58). In vitro silencing of circ0006528 repressed proliferation, migration and invasion, as well as autophagy, and induced apoptosis in vitro (58). Circ0006528 sponged miR-1299 and up-regulated CDK8. The latter promoted proliferation, migration and autophagy in paclitaxel-resistant BC cells (58). Circ0006528 increased xenograft growth of ZR-75-30/PTX in nude mice (58). CDK8 is associated with paclitaxel resistance and activation of the WNT/ß-catenin signaling pathway (59, 60). CDK8 acts both as an activator and repressor of transcription, invasiveness and EMT (61). Inhibition of CDK8 inhibits growth and proliferation of BC cells (62, 63).

Circ0006528 targets chromodomain helicase DNA-binding protein 4 (CHD4). Higher levels of circ0006528 (Figure 2B) have been found in adriamycin-resistant BC tissues and cells (64). Overexpression of circ0006528 mediated proliferation, migration, invasion and adriamycin resistance in BC cells. Circ0006528 sponged miR-1236-3p and led to up-regulation of CHD4 (64). It represents the main component of the nucleosome remodeling and deacetylase complex and plays an important role in epigenetic transcriptional regulation (65). CHD4 promotes BC progression as a coactivator of hypoxia-inducible factors (66). In addition, CHD4 regulates the HER2 signaling pathway and autophagy in HER2+ BC cells (67) and is an essential gene for BC growth (68, 69). Independently, it has been shown that CHD4 is involved in DNA damage response and chemotherapy resistance (70).

Circ0001667 targets nuclear receptor co-activator A3 (NCOA3). Knockdown of circ0001667 (Figure 2B) in MCF-7/ADM and MDA-MB-231/ADM inhibited proliferation, migration, invasion and adriamycin resistance in vitro (71). Knockdown of circ0001667 repressed tumor growth and adriamycin resistance of BC cells in immunodeficient mice (71). miR-4458 was sponged by circ0001667 and led to upregulation of NCOA3, also known as amplified in BC1 (AIB1) or steroid receptor co-activator 3 (SRC3). NCOA3 is a transcriptional co-activator with several nuclear receptor-interacting domains and intrinsic histone acetyltransferase activity, resulting in acetylation of histones and assisting nuclear receptors in up-regulation of genes (72). NCOA3 is amplified in BC and acts as an oncogene (73).

Circ0085484 targets integrin β1 (INTB1). Depletion of circ0085484 (Figure 2B) repressed adriamycin resistance, proliferation, and metastasis of adriamycin-resistant BC cells (74). The phenomenon is due to sponging of miR-873-5p and subsequent up-regulation of INTB1 (74). It was shown that INTβ1 can bind to collagen type 1 and activate adriamycin efflux transporters (75).

Circ-ubiquitin-conjugating enzyme 2D2 (circUBE2D2) targets ERα. Tamoxifen is a selective ER modulator used for prevention and treatment of BC. Treatment resistance to tamoxifen is frequently observed (76, 77). CircUBE2D2 (Figure 2C) was up-regulated in tamoxifen-resistant BC tissues and cell lines such as MCF-7/TMX and T47/TMX (78). Deletion of circUBE2D2 (Figure 2C) inhibited tamoxifen resistance in MCF-7/TMX and T47/TMX cells. UBE2D2 was also found in exosomes of these cell lines. Intercellular transfer of circUBE2D2 enhanced tamoxifen resistance in vitro and in vivo. circUBE2D2 sponged miR-200a-3p and increased viability, metastasis and the level of ERα (78).

Circ-matrix metalloprotease11 (circMMP11) targets anillin (ANLN). Lapatinib, an inhibitor of HER2 and epidermal growth factor receptor is used for the treatment of HER2-overexpressing advanced and metastatic BC (79). CircMMP11 (Figure 2C) was up-regulated in lapatinib-resistant BC tissues and cell lines (MDA-MB-231/LR and MCF-7/LR) (80). CircMMP11 was transported by exosomes and enhanced lapatinib resistance in BC cell lines. CircMMP11 knockdown impeded tumor growth of MDA-MB-231/LR in nude mice and increased lapatinib resistance (80). CircMMP11 sponged miR-153-3p, resulting in upregulation of ANLN. The latter is a cytoskeletal protein, containing an actin/myosin-binding domain and a pleckstrin-homology domain, which is critical for cell division and EMT, and is frequently overexpressed in cancer (81, 82). Knockdown of ANLN inhibits growth of BC cells (82). ANLN is also involved in poor prognosis in patients treated with anthracycline-based chemotherapy (83).

CircRNAs up-regulating high-mobility group (HMG) proteins Circ0069094, circ-F-box and leucine-rich repeat protein 5 (circFBXL5), circ-homeodomain-interacting protein kinase 3 (circHIPK3) and circ0003645 target HMGA1, -A2 and - B1. Circ0069094 (Figure 3A) was up-regulated in BC samples in comparison to adjacent normal tissues and non-cancerous MC7-10A cells (84). Knockdown of circ0069094 inhibited cell glycolysis, glucose uptake, lactate production, HK2, proliferation, migration, invasion and increased apoptosis in BC cells in vitro and tumor formation in vivo in nude mice (84). From a mechanistic point of view, circ0069094 sponged miR-661, resulting in up-regulation of HMGA1.

Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.

Circular (circ) RNAs up-regulated in breast cancer with efficacy in preclinical in vivo models affecting high-mobility group (HMG) proteins, secreted factors and transmembrane proteins. A: CircRNAs up-regulating HMG proteins. B: CircRNAs up-regulating secreted and transmembrane proteins. Up- and down-regulation are indicated by up and down arrows, respectively. CXCL10: C-X-C Chemokine ligand 10; FBXL5: F-box/LRR repeat protein 5; HIPK3: homeodomain-interacting protein kinase 3 exon 2 splicing; IGF1: insulin-like growth factor 1; miR: microRNA; MUC19: mucin 19; PI3K: phosphatidylinositol-4,5-bisphosphate 3-kinase; PLK1: POLO-like kinase 1; TFF1: trefoil factor 1.

CircFBXL5 (Figure 3A) was up-regulated in BC samples, promoted migration and invasion, and inhibited apoptosis in MBA-MB-231 and MDA-MB-453 BC cells in vitro and its silencing reduced 5-fluorouracil-resistant BC growth in vivo in nude mice (85). CircFBXL5 sponged miR-216b, resulting in up-regulation of HMGA2 (85).

CircHIPK3 up-regulates HMGB1 (Figure 3A), which was increased in BC tissues and high expression predicted poor prognosis. Small interfering RNA (siRNA)- mediated knockdown of circHIPK3 reduced viability, migration and invasion of MDA-MB-231 BC cells in vitro and inhibited tumor growth in vivo in nude mice (86). CircHIPK3 sponged miR-193a, resulting in expression of HMGB1 and activation of phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/AKT serine/threonine kinase 1 (AKT) signaling in MDA-MB-231 cells (86).

Circ0003645 (Figure 3A) also up-regulated HMGB1 by sponging miR-139-3p (87). Circ0003645 was overexpressed in BC tissues, promoted proliferation of MCF-7 and MDA-MB-231 BC cells in vitro and tumor growth of MDA-MB-231 BC cells in vivo in nude mice after subcutaneous implantation (87).

HMG proteins are chromosomal DNA-binding proteins which are involved in the regulation of DNA-dependent processes such as transcription, replication, recombination and DNA repair (88). HMGA contains an AT-hook domain, HMGB contains an HMG-box domain and HMGN exhibits a nucleosomal-binding domain (89). They can directly interact with transcription factors and alter chromatin structure (89). Expression of HMGA1 and HMGA2 is correlated with malignant status and prognosis of patients with BC (90. 91). Knockdown of HMGA1 inhibited BC growth and metastasis in immunodeficient mice and attenuated BC angiogenesis (91, 92). HMGA2 is involved in proliferation, migration, invasion, acquisition of stem cell features, EMT and telomere restoration (93, 94). HMGA1 also occurs extracellularly and is involved in an autocrine loop by binding to the receptor for advanced glycation end-products, resulting in migration, invasion and metastasis, and can be targeted with monoclonal antibodies (95). HMGA2 promotes BC metastasis by modulating Hippo-YAP signaling (96). The druggability of HMGs deserves further attention.

CircRNAs up-regulating secreted and transmembrane proteins Circ-trefoil factor 1 (circTFF1) targets trefoil factor 1. CircTFF1 (Figure 3B) was found to be highly expressed in BC and its depletion restrained cell migration, invasion and EMT in BT-549 and MBA-MB-231 BC cells (97). CircTFF1 sponged miR-326, resulting in up-regulation of TFF1. CircTFF1 accelerated growth of BT-549 cells in nude mice (97). Three TFFs are known to contain at least one copy of a 40 amino acid domain with three conserved sulfide bonds (98). TFF1 is regulated by ER and can act as a tumor suppressor in mice (99). On the other hand, TFF1 stimulated migration of BC cells (100). TFF1 represents a potential prognostic biomarker with functional significance in patients with BC (101). TFF1 is expressed at higher levels in blood from patients with metastatic BC than in those without metastatic disease (102). Independently, it has been shown that TFF1 is up-regulated in ER+ BC and is correlated with increased bone metastasis (103).

Circ0000515 targets C-X-C chemokine ligand 10 (CXCL10). Circ0000515 (Figure 3B) was up-regulated in BC and its expression level correlated with poor prognosis (104). It promoted cell-cycle progression, proliferation, invasion, inflammatory response and pro-angiogenic potential of BC cells (104). Circ0000515 was shown to bind to miR-296-5p, preventing it from repressing CXCL10. CXCL10 binds to CXC motif chemokine receptor 3 (CXCR3) together with CXCL9 and CXCL11 (105). Loss of CXCL10 reduced growth of MCF-7 BC cells in nude mice (104). CXCL10 mediates BC resistance to tamoxifen and promotes estrogen-dependent and - independent proliferation (106). In BC, RAS induced CXCL10 signals through serine-threonine kinase RAF and PI3K signaling pathways. However, CXCL10 can have dual effects on cell growth (107, 108). CXCL10 plays a role in migration of tumor cells (109). CXCL10 and its receptor CXCR3 play a key role in BC metastasis to bone and osteoclast activation through NFEmbedded ImageB activation (110). However, a major function of CXCL10 is enhancement of T-cell-dependent anticancer immunity, a property which cannot be assessed in the immunodeficient in vivo model as described above (105). Therefore, the role of CXCL10 as a cancer-promoting agent has to be scrutinized in more detail.

Circ-POLO-like kinase 1 (circPLK1) targets insulin-like growth factor 1 (IGF1). CircPLK1 (Figure 3B) was upregulated in BC tissues and cells (111). In BT549 and HCC38 BC cells, circPLK1 regulated cell proliferation, cell-cycle transition from G1 to S phase, and migration and invasion in vitro. In vivo, interference with circPLK1 restrained tumor growth in nude mice (111). CircPLK1 binds to miR-4500, resulting in up-regulation of IGF1. The latter functions as a potent mitogen in the mammary gland via IGF receptor 1 signaling (112). This signaling system also includes six IGF-binding proteins, which modulate the bioavailability of IGF1 and triggers PI3K/AKT and RAF/MAPK pathways (112). In addition, IGF1 also binds to the insulin receptor (113). In preclinical models of BC, IGF1/2 antibodies inhibited bone metastasis without affecting the growth of the primary tumor (114, 115). However, clinical trials with IGF receptor 1 antibodies, IGF receptor 1 tyrosine kinase inhibitors and IGF1/2 antibodies in patients with cancer have generated negative results.

Circ0001982 targets mucin 19 (MUC19). Circ0001982 (Figure 3B) was increased and its knockdown inhibited cell glycolysis, viability of and migration and invasion by MDA-MB-231 and MDA-MB-468 BC cells (116). Circ0001082 sponged miR-1297-5p, resulting in up-regulation of MUC19 and mediated xenograft growth of MDA-MB-231 cells in nude mice (116). Mucins are large O-glycoproteins which occur as transmembrane or secreted molecules to activate signaling pathways such as NFEmbedded ImageB, WNT, SRC, p53, MAPK, hypoxia-inducible factor and JAK-STAT, affecting stemness, metabolism and chemoresistance (117). MUC1, −4 and −16 are also involved in the pathogenesis of BC (118). MUC16, which was discovered as cancer antigen 125 and functions as a ligand for mesothelin, is aberrantly expressed in many types of cancer and numerous clinical studies with corresponding monoclonal antibodies and their conjugates are ongoing. Cleavage of its extracellular domain represents a major problem for therapy (119, 120). Other approaches focus on the use of MUC1 as a cancer vaccine (121). For MUC19, more preclinical validation studies are needed to rank its significance for treatment of BC.

CircRNAs involved in signaling

Circ-inhibitor of NFEmbedded ImageB kinase subunit beta (circIKBKB) targets NFEmbedded ImageB. Overexpression of circIKBKB (Figure 4A) in MCF-7 and MDA-MB-231 BC cells induced osteoclastogenesis, formation of a bone pre-metastatic niche and bone metastasis after intracardiac injection in nude mice (122). Overexpression of circIKBKB activated NFEmbedded ImageB signaling due to IEmbedded ImageB kinase β-mediated phosphorylation of NFEmbedded ImageB inhibitor α (IEmbedded ImageBα) and its subsequent degradation. CircIKBKB recruited NFEmbedded ImageB to the promoters of several bone-remodeling factors such as receptor activator of NFEmbedded ImageB ligand, macrophage colony-stimulating factor and granulocyte-macrophage colony-stimulating factor. Eukaryotic translation initiation factor 4A3 (EIF4A3) directly bound to circIKBKB pre-mRNA and induced its cyclization. Blocking of EIF4A3 by RNA interference or a specific, highly selective non-competitive inhibitor blocked NFEmbedded ImageB signaling, osteoclastogenesis and bone metastasis (122). EIF4A3 is a nuclear matrix protein and a core component of the exon junction complex (123, 124). Bone metastasis occurs in 70% of patients with metastatic BC (125). Bisphosphonates and receptor activator of NFEmbedded ImageB ligand monoclonal antibody denosomab are approved for treatment of metastatic bone disease but they do not have an impact on survival and exhibit severe side-effects (126). It remains to be studied as to whether the inhibition of hyperactivated NFEmbedded ImageB signaling will result in improved agents.

Figure 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 4.

Circular (circ) RNAs up-regulated in breast cancer with efficacy in preclinical in vivo models affecting signaling and induction of additional targets. A: CircRNAs involved in signaling. B: CircRNAs up-regulating additional targets. Up-regulated and downregulated are indicated by up and down arrows, respectively. AKT: serine-threonine kinase AKT; CBX4: chromobox protein 4; CCND1: cyclin D1; CFL2: cofilin 2; COL1A1: collagen 1A1; DENN4C: DENN domain-containing 4C; EGFR: epidermal growth factor receptor; GRB2: growth factor receptor-bound protein 2; HK2: hexokinase 2; IKBKB: inhibitor of NFEmbedded ImageB kinase; IEmbedded ImageBα-P: phosphorylated inhibitor IEmbedded ImageBα; MAPK1: mitogen-activated protein kinase 1; MET: metastasis; miR: microRNA; NFEmbedded ImageB: nuclear factor Embedded ImageB; PAK4: serine-threonine protein kinase PAK4; RPPH1: ribonuclease P component H1; TAZ: transcriptional co-activator with a PDZ-binding motif; UBR1: ubiquitin-protein ligase 1; WWC3: WW and C3 domain-containing; YAP1: YES-associated protein 1.

Circ-WW and C3 domain-containing (circWWC3) targets RAS pathway genes. Zinc finger E-box binding homeobox 1 (ZEB1) up-regulates circWWC3, which correlated with BC progression (127). CircWWC3 (Figure 4A) increased invasion and migration of MCF-7 and MDA-MB-231 BC cells. CircWWC3 functioned as a sponge for miR-26b-3p and -660-3p and up-regulated RAS pathway genes epidermal growth factor receptor, growth factor receptor-bound protein 2, serine-threonine kinase PAK4, MAPK1, and AKT. In nude mice, circWWC3 promoted metastasis of MDA-MB-231 cells to the lungs and liver. The RAS/RAF/MEK/ERK MAPK pathway plays a crucial role in growth, survival and differentiation of cancer cells (128). Targeting mutant RAS has recently led to approved drugs (129-131).

Circ-ribonuclease P component H1 (circRPPH1) targets YES-associated protein 1 (YAP1). CircRPPH1 (Figure 4A) was overexpressed in BC and mediated proliferation, migration and invasion in vitro of MCF-7 and MDA-MB-231 cells as well as endothelial tube formation with supernatants from circRPPH1-transfected BC cells (132). CircRPPH1 sponged miR-556-5p, up-regulated YAP1 and promoted tumor growth in nude mice bearing MDA-MB-231 BC cells (132).

Circ0005273 targets YAP1. Circ0005273 (Figure 4A) was highly expressed in BC and correlated with TNM stage, lymph node metastasis, tumor size and distant metastasis (133). It induced proliferation, migration and cell-cycle progression in MDA-MB-231, MCF-7 and SKBR3 BC cells in vitro and promoted tumor growth in MDA-MB-231 cells in nude mice (133). This was due to sponging of miR-200a-3p, which led to up-regulation of YAP1.

Circ0000511 targets transcriptional co-activator with PDZ-binding motif (TAZ). Circ0000511 (Figure 4A) was upregulated in BC and accelerated proliferation, migration, invasion, and impeded apoptosis in MCF-7 and MDA-MB-468 BC cells in vitro (134). Circ0000511 sponged miR-326 and up-regulated TAZ. In vivo, circ0000511 promoted tumor growth in nude mice (134).

YAP and TAZ are transcriptional co-activators of the Hippo pathway, which signals through components of the extracellular matrix, cell adhesion sites, cell shape and the actomyosin cytoskeleton and determines organ size in animals (135-137). YAP/TAZ regulates BC-related metastasis targets (138, 139). Therefore, targeting of the Hippo pathway in BC is a high priority issue (140). Verteporforin has been identified as a compound which inhibits YAP-TEA domain family interaction (140) and BAY1238097 interacts with YAP/TAZ and down-regulates their transcriptional activity by inhibiting bromodomain-containing protein 4 (141).

CircRNAs up-regulating additional targets

Circ-ubiquitin protein ligase 1 (circUBR1) targets cyclin D1 (CCND1). CircUBR1 (Figure 4B) was up-regulated in BC and its silencing inhibited proliferation and metastasis, promoted apoptosis in vitro and restrained tumor growth in vivo (142). CircUBR1 sponged miR-1299 and led to overexpression of CCND1. The latter is overexpressed in more than 50% of BCs, causes mammary cancer in transgenic mice, promotes cellular proliferation and cell-cycle progression, and has a role in BC stem cell expansion (143). CCND1 has been found to be amplified in BC (144). It has been found that a CCND1 G870A polymorphism increased BC risk (145).

Circ0008673 targets cofilin 2 (CFL2). Circ0008673 (Figure 4B) was up-regulated in BC, promoted proliferation, migration and invasion, and inhibited apoptosis of BC cells in vitro. It acted as a sponge for miR-153-3p and its silencing repressed BC xenograft growth in nude mice (146). This was due to up-regulation of CFL2, which mediated growth of BC cells in vitro (146, 147). CFL2 reversibly controls actin polymerization and depolymerization in a pH-dependent manner and regulation of the actin cytoskeleton in cancer cell migration and invasion (148). CFL2 promotes cancer cell invasion through O-Glc-Nacylation (149) and BC invasion and metastasis (150).

Circ002178 targets collagen 1A1 (COL1A1). Circ002178 (Figure 4B) was overexpressed in BC tissues and correlated with poor prognosis (151). Silencing of circ002178 impaired proliferation, energy metabolism and angiogenesis. Circ002178 bound to miR-328-3p and led to up-regulation of COL1A1. In vivo, circ002178 stimulated growth of MDA-MB-231 BC cells in nude mice. COL1A1 is an extracellular matrix protein that is correlated with advanced BC, poor prognosis, invasion and metastasis (152). Development of the extracellular matrix is correlated with BC cancer progression (153, 154). Taken together, these findings show COL1A1 is a potential target for treatment of BC.

Circ0008030 targets chromobox protein 4 (CBX4). Circ0008030 (Figure 4B) was up-regulated in BC tissues and cells and expedited proliferation, invasion and migration of BC cells (155). It sponged miR-515-5p, which led to upregulation of CBX4. Knockdown of circ0008030 suppressed tumor growth of BC xenografts in nude mice. CBX family proteins are canonical components of polycomb repressive complex 1 (PRC1) which bind to DNA and transcriptionally repress target genes via chromatin modification, acting as oncogenes or tumor suppressors (156). CBX4 recognizes H3K27me3, a transcriptionally suppressive epigenetic marker (157). In BC, CBX4 exhibits oncogenic activity via NOTCH1 signaling (158). In addition miR-129-5p has been shown to suppress BC proliferation by targeting CBX4 (159).

Circ-differentially expressed in normal cells neoplasia domain-containing 4C (circDENN4C) targets HK2. CircDENN4C (Figure 4B) was up-regulated in BC cells in response to hypoxia (160). Knockdown of DENN4C in MDA-MB-453 and SKBR3 BC cells inhibited glycolysis, lactate production and HK2, as well as migration and invasion under hypoxia. It was found that the observed phenomena were due to sponging of miR-200b and -c by circDENN4C. In addition, EMT and expression of MMP2 and −9 were increased under hypoxia by circDENN4C (160). However, the underlying mechanisms have to be resolved in more detail. Knockdown of circDENN4C in MDA-MB-435 cells inhibited xenograft growth in nude mice (160). Increased glycolysis is one of the hallmarks of cancer (161, 162) and a glycolysis-related expression signature predicts recurrence of BC (163).

Approaches and Challenges of Targeting CircRNAs

Down-regulated circRNAs can be reconstituted by transfection of expression vectors for the corresponding circRNA into recipient cells (164). For up-regulated circRNAs, the new junctions generated via back-splicing events may be specifically targeted (165). Options include antisense-oligonucleotides (ASO), siRNAs (siRNA), short hairpin RNA (shRNA) and the clustered regularly interspaced short palindromic repeats (CRISPR)-Cas (CRISPR-associated proteins) method for specific cleavage of the target RNA. ASOs are chemically modified single-stranded oligonucleotides (12-24-mers) that bind to their RNA targets and, depending on the design, lead to the RNaseH-mediated degradation or inhibit the function of target RNAs by steric interference (166, 167). Medicinal chemistry-based backbone and sugar modifications give rise to optimized ASOs with improved stability, high potency, and enabled intracellular delivery without the help of transfection agents (166, 167). siRNA and shRNA are RNA interference-based agents. siRNAs are 12–24-mers double-stranded RNAs with one of the strand targets circRNAs through complementary pairing and incorporate the circRNA into the RNA-induced silencing complex (168). shRNAs, on the other hand, contain a tight hairpin loop structure and target-complementary sequences which can be delivered as plasmids or viral vectors, allowing for stable integration of shRNA and long-term knockdown of the target (18, 169). The CRISPR-CAS13-based method has recently been used for screening for functional circRNAs (170). siRNA and shRNA can be delivered as lipid-based polymers or with exosomes (171). However, low stability in cells, poor intracellular delivery, and lack of cell specificity are critical issues for RNA interference therapeutics in vivo (172). Immunogenicity is another critical issue of the agents as described above (173, 174). Progress in the design of new formulations and tissue-specific targeting methods are issues of paramount importance (175-178).

Conclusion

We have identified six down-regulated and 26 up-regulated circRNAs which mediate efficacy in preclinical BC-related in vivo models. According to their corresponding targets, they can be categorized into groups which mediate chemoresistance or signaling, or are related to HMG, transmembrane or secreted proteins, as well as other targets. Two down-regulated and eight up-regulated circRNAs mediate adriamycin, tamoxifen, paclitaxel and lapatinib resistance. Interestingly, down-regulation of circ0025202 mediates tamoxifen resistance via miR-182-5p and miR-197-3p, with FOXO3A and HIPK3 as targets (Figure 1). Circ006528 can mediate paclitaxel as well as adriamycin resistance via miR-1299 and miR-1236-3p with CDK8 and CHD4 as targets (Figure 2). SOCS2 and SOCS3 (Figure 1) are frequently down-regulated in BC, resulting in constitutive activation of JAK- and STAT-related signaling. Other identified circRNAs target tumor- and metastasis-promoting pathways driven by HMG proteins (HMGA1, -A2 and -B1) (Figure 3). There are druggability issues with small molecules, but interference with antibody-related moieties emerges as an alternative, because HMGs are also present on the cell surface (179). Further identified targets are mediators of metastasis-related pathways. NFEmbedded ImageB signaling induced by circIKBKB (Figure 4) is a mediator of bone metastasis. Hippo-related components YAP1 (circRPPH1, circ0052372) and TAZ (circ0000511) are also involved in metastasis of BC (Figure 4). It remains to be established whether a therapeutic window for corresponding inhibitors can be defined. CircTFF1 up-regulates TFF1, which is involved in metastasis of ER+ BC and should be investigated in further detail in this subgroup of patients (Figure 3). A further unexplored target identified by our search is MUC19, which merits further investigation by target validation experiments (Figure 3).

Footnotes

  • Conflicts of Interest

    UHW was and UB is an employee of Roche. H-E H is a postdoctoral fellow of Roche. Roche is interested in targeted therapies and diagnostics

  • Authors’ Contributions

    The Authors contributed equally to all aspects of the article.

  • Received March 3, 2023.
  • Revision received April 3, 2023.
  • Accepted April 4, 2023.
  • Copyright © 2023, International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved

This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY-NC-ND) 4.0 international license (https://creativecommons.org/licenses/by-nc-nd/4.0).

References

  1. ↵
    1. Siegel RL,
    2. Miller KD,
    3. Fuchs HE and
    4. Jemal A
    : Cancer statistics, 2021. CA Cancer J Clin 71(1): 7-33, 2021. PMID: 33433946. DOI: 10.3322/caac.21654
    OpenUrlCrossRefPubMed
  2. ↵
    1. Fragomeni SM,
    2. Sciallis A and
    3. Jeruss JS
    : Molecular subtypes and local-regional control of breast cancer. Surg Oncol Clin N Am 27(1): 95-120, 2018. PMID: 29132568. DOI: 10.1016/j.soc.2017.08.005
    OpenUrlCrossRefPubMed
  3. ↵
    1. Harbeck N,
    2. Penault-Llorca F,
    3. Cortes J,
    4. Gnant M,
    5. Houssami N,
    6. Poortmans P,
    7. Ruddy K,
    8. Tsang J and
    9. Cardoso F
    : Breast cancer. Nat Rev Dis Primers 5(1): 66, 2019. PMID: 31548545. DOI: 10.1038/s41572-019-0111-2
    OpenUrlCrossRefPubMed
    1. Harbeck N and
    2. Gnant M
    : Breast cancer. Lancet 389(10074): 1134-1150, 2017. PMID: 27865536. DOI: 10.1016/S0140-6736(16)31891-8
    OpenUrlCrossRefPubMed
    1. Waks AG and
    2. Winer EP
    : Breast cancer treatment: a review. JAMA 321(3): 288-300, 2019. PMID: 30667505. DOI: 10.1001/jama.2018.19323
    OpenUrlCrossRefPubMed
  4. ↵
    1. Galván Morales MA,
    2. Barrera Rodríguez R,
    3. Santiago Cruz JR and
    4. Teran LM
    : Overview of new treatments with immunotherapy for breast cancer and a proposal of a combination therapy. Molecules 25(23): 5686, 2020. PMID: 33276556. DOI: 10.3390/molecules25235686
    OpenUrlCrossRefPubMed
  5. ↵
    1. Lee YT
    : Breast carcinoma: pattern of metastasis at autopsy. J Surg Oncol 23(3): 175-180, 1983. PMID: 6345937. DOI: 10.1002/jso.2930230311
    OpenUrlCrossRefPubMed
  6. ↵
    1. Steeg PS
    : Targeting metastasis. Nat Rev Cancer 16(4): 201-218, 2016. PMID: 27009393. DOI: 10.1038/nrc.2016.25
    OpenUrlCrossRefPubMed
  7. ↵
    1. Madden E,
    2. Logue SE,
    3. Healy SJ,
    4. Manie S and
    5. Samali A
    : The role of the unfolded protein response in cancer progression: From oncogenesis to chemoresistance. Biol Cell 111(1): 1-17, 2019. PMID: 30302777. DOI: 10.1111/boc.201800050
    OpenUrlCrossRefPubMed
  8. ↵
    1. Chun KH,
    2. Park JH and
    3. Fan S
    : Predicting and overcoming chemotherapeutic resistance in breast cancer. Adv Exp Med Biol 1026: 59-104, 2017. PMID: 29282680. DOI: 10.1007/978-981-10-6020-5_4
    OpenUrlCrossRefPubMed
  9. ↵
    1. Lasda E and
    2. Parker R
    : Circular RNAs: diversity of form and function. RNA 20(12): 1829-1842, 2014. PMID: 25404635. DOI: 10.1261/rna.047126.114
    OpenUrlAbstract/FREE Full Text
  10. ↵
    1. Liu J,
    2. Zhang X,
    3. Yan M and
    4. Li H
    : Emerging role of circular RNAs in cancer. Front Oncol 10: 663, 2020. PMID: 32670861. DOI: 10.3389/fonc.2020.00663
    OpenUrlCrossRefPubMed
  11. ↵
    1. Kristensen LS,
    2. Hansen TB,
    3. Venø MT and
    4. Kjems J
    : Circular RNAs in cancer: opportunities and challenges in the field. Oncogene 37(5): 555-565, 2018. PMID: 28991235. DOI: 10.1038/onc.2017.361
    OpenUrlCrossRefPubMed
  12. ↵
    1. Li Z,
    2. Chen Z,
    3. Hu G and
    4. Jiang Y
    : Roles of circular RNA in breast cancer: present and future. Am J Transl Res 11(7): 3945-3954, 2019. PMID: 31396311.
    OpenUrlPubMed
    1. Ghafouri-Fard S,
    2. Hussen BM,
    3. Taheri M and
    4. Ayatollahi SA
    : Emerging role of circular RNAs in breast cancer. Pathol Res Pract 223: 153496, 2021. PMID: 34052769. DOI: 10.1016/j.prp.2021.153496
    OpenUrlCrossRefPubMed
  13. ↵
    1. Shao Y and
    2. Lu B
    : The crosstalk between circular RNAs and the tumor microenvironment in cancer metastasis. Cancer Cell Int 20: 448, 2020. PMID: 32943996. DOI: 10.1186/s12935-020-01532-0
    OpenUrlCrossRefPubMed
  14. ↵
    1. Sarkar D and
    2. Diermeier SD
    : Circular RNAs: Potential applications as therapeutic targets and biomarkers in breast cancer. Noncoding RNA 7(1): 2, 2021. PMID: 33466455. DOI: 10.3390/ncrna7010002
    OpenUrlCrossRefPubMed
  15. ↵
    1. He AT,
    2. Liu J,
    3. Li F and
    4. Yang BB
    : Targeting circular RNAs as a therapeutic approach: current strategies and challenges. Signal Transduct Target Ther 6(1): 185, 2021. PMID: 34016945. DOI: 10.1038/s41392-021-00569-5
    OpenUrlCrossRefPubMed
  16. ↵
    1. Zhang X,
    2. Wang S,
    3. Wang H,
    4. Cao J,
    5. Huang X,
    6. Chen Z,
    7. Xu P,
    8. Sun G,
    9. Xu J,
    10. Lv J and
    11. Xu Z
    : Circular RNA circNRIP1 acts as a microRNA-149-5p sponge to promote gastric cancer progression via the AKT1/mTOR pathway. Mol Cancer 18(1): 20, 2019. PMID: 30717751. DOI: 10.1186/s12943-018-0935-5
    OpenUrlCrossRefPubMed
  17. ↵
    1. Qiu M,
    2. Xia W,
    3. Chen R,
    4. Wang S,
    5. Xu Y,
    6. Ma Z,
    7. Xu W,
    8. Zhang E,
    9. Wang J,
    10. Fang T,
    11. Hu J,
    12. Dong G,
    13. Yin R,
    14. Wang J and
    15. Xu L
    : The circular RNA circPRKCI promotes tumor growth in lung adenocarcinoma. Cancer Res 78(11): 2839-2851, 2018. PMID: 29588350. DOI: 10.1158/0008-5472.CAN-17-2808
    OpenUrlAbstract/FREE Full Text
  18. ↵
    1. Liang Y,
    2. Song X,
    3. Li Y,
    4. Su P,
    5. Han D,
    6. Ma T,
    7. Guo R,
    8. Chen B,
    9. Zhao W,
    10. Sang Y,
    11. Zhang N,
    12. Li X,
    13. Zhang H,
    14. Liu Y,
    15. Duan Y,
    16. Wang L and
    17. Yang Q
    : circKDM4C suppresses tumor progression and attenuates doxorubicin resistance by regulating miR-548p/PBLD axis in breast cancer. Oncogene 38(42): 6850-6866, 2019. PMID: 31406252. DOI: 10.1038/s41388-019-0926-z
    OpenUrlCrossRefPubMed
  19. ↵
    1. Li A,
    2. Yan Q,
    3. Zhao X,
    4. Zhong J,
    5. Yang H,
    6. Feng Z,
    7. Du Y,
    8. Wang Y,
    9. Wang Z,
    10. Wang H,
    11. Zhou Y,
    12. Liu S and
    13. Nie Y
    : Decreased expression of PBLD correlates with poor prognosis and functions as a tumor suppressor in human hepatocellular carcinoma. Oncotarget 7(1): 524-537, 2016. PMID: 26594798. DOI: 10.18632/oncotarget.6358
    OpenUrlCrossRefPubMed
  20. ↵
    1. Sang Y,
    2. Chen B,
    3. Song X,
    4. Li Y,
    5. Liang Y,
    6. Han D,
    7. Zhang N,
    8. Zhang H,
    9. Liu Y,
    10. Chen T,
    11. Li C,
    12. Wang L,
    13. Zhao W and
    14. Yang Q
    : circRNA_0025202 regulates tamoxifen sensitivity and tumor progression via regulating the miR-182-5p/FOXO3a axis in breast cancer. Mol Ther 27(9): 1638-1652, 2019. PMID: 31153828. DOI: 10.1016/j.ymthe.2019.05.011
    OpenUrlCrossRefPubMed
  21. ↵
    1. Liu Y,
    2. Ao X,
    3. Ding W,
    4. Ponnusamy M,
    5. Wu W,
    6. Hao X,
    7. Yu W,
    8. Wang Y,
    9. Li P and
    10. Wang J
    : Critical role of FOXO3a in carcinogenesis. Mol Cancer 17(1): 104, 2018. PMID: 30045773. DOI: 10.1186/s12943-018-0856-3
    OpenUrlCrossRefPubMed
    1. Zou Y,
    2. Tsai WB,
    3. Cheng CJ,
    4. Hsu C,
    5. Chung YM,
    6. Li PC,
    7. Lin SH and
    8. Hu MC
    : Forkhead box transcription factor FOXO3a suppresses estrogen-dependent breast cancer cell proliferation and tumorigenesis. Breast Cancer Res 10(1): R21, 2008. PMID: 18312651. DOI: 10.1186/bcr1872
    OpenUrlCrossRefPubMed
  22. ↵
    1. Jin L,
    2. Zhang J,
    3. Fu HQ,
    4. Zhang X and
    5. Pan YL
    : FOXO3a inhibits the EMT and metastasis of breast cancer by regulating TWIST-1 mediated miR-10b/CADM2 axis. Transl Oncol 14(7): 101096, 2021. PMID: 33882368. DOI: 10.1016/j.tranon.2021.101096
    OpenUrlCrossRefPubMed
  23. ↵
    1. Song Y,
    2. Zeng S,
    3. Zheng G,
    4. Chen D,
    5. Li P,
    6. Yang M,
    7. Luo K,
    8. Yin J,
    9. Gu Y,
    10. Zhang Z,
    11. Jia X,
    12. Qiu N,
    13. He Z,
    14. Li H and
    15. Liu H
    : FOXO3a-driven miRNA signatures suppresses VEGF-A/NRP1 signaling and breast cancer metastasis. Oncogene 40(4): 777-790, 2021. PMID: 33262463. DOI: 10.1038/s41388-020-01562-y
    OpenUrlCrossRefPubMed
  24. ↵
    1. Li H,
    2. Li Q and
    3. He S
    : Hsa_circ_0025202 suppresses cell tumorigenesis and tamoxifen resistance via miR-197-3p/HIPK3 axis in breast cancer. World J Surg Oncol 19(1): 39, 2021. PMID: 33536026. DOI: 10.1186/s12957-021-02149-x
    OpenUrlCrossRefPubMed
  25. ↵
    1. Conte A and
    2. Pierantoni GM
    : Update on the regulation of HIPK1, HIPK2 and HIPK3 protein kinases by microRNAs. Microrna 7(3): 178-186, 2018. PMID: 29793420. DOI: 10.2174/2211536607666180525102330
    OpenUrlCrossRefPubMed
  26. ↵
    1. Nodale C,
    2. Sheffer M,
    3. Jacob-Hirsch J,
    4. Folgiero V,
    5. Falcioni R,
    6. Aiello A,
    7. Garufi A,
    8. Rechavi G,
    9. Givol D and
    10. D’Orazi G
    : HIPK2 downregulates vimentin and inhibits breast cancer cell invasion. Cancer Biol Ther 13(4): 198-205, 2012. PMID: 22236966. DOI: 10.4161/cbt.13.4.18694
    OpenUrlCrossRefPubMed
  27. ↵
    1. Liu Y,
    2. Qian L,
    3. Yang J,
    4. Huang H,
    5. Feng J,
    6. Li X,
    7. Bian T,
    8. Ke H,
    9. Liu J and
    10. Zhang J
    : The expression level and prognostic value of HIPK3 among non-small-cell lung cancer patients in China. Onco Targets Ther 11: 7459-7469, 2018. PMID: 30498360. DOI: 10.2147/OTT.S166878
    OpenUrlCrossRefPubMed
  28. ↵
    1. Rey C,
    2. Soubeyran I,
    3. Mahouche I,
    4. Pedeboscq S,
    5. Bessede A,
    6. Ichas F,
    7. De Giorgi F and
    8. Lartigue L
    : HIPK1 drives p53 activation to limit colorectal cancer cell growth. Cell Cycle 12(12): 1879-1891, 2013. PMID: 23676219. DOI: 10.4161/cc.24927
    OpenUrlCrossRefPubMed
  29. ↵
    1. Kondo S,
    2. Lu Y,
    3. Debbas M,
    4. Lin AW,
    5. Sarosi I,
    6. Itie A,
    7. Wakeham A,
    8. Tuan J,
    9. Saris C,
    10. Elliott G,
    11. Ma W,
    12. Benchimol S,
    13. Lowe SW,
    14. Mak TW and
    15. Thukral SK
    : Characterization of cells and gene-targeted mice deficient for the p53-binding kinase homeodomain-interacting protein kinase 1 (HIPK1). Proc Natl Acad Sci U S A 100(9): 5431-5436, 2003. PMID: 12702766. DOI: 10.1073/pnas.0530308100
    OpenUrlAbstract/FREE Full Text
  30. ↵
    1. Wang F,
    2. Wang X,
    3. Li J,
    4. Lv P,
    5. Han M,
    6. Li L,
    7. Chen Z,
    8. Dong L,
    9. Wang N and
    10. Gu Y
    : CircNOL10 suppresses breast cancer progression by sponging miR-767-5p to regulate SOCS2/JAK/STAT signaling. J Biomed Sci 28(1): 4, 2021. PMID: 33397365. DOI: 10.1186/s12929-020-00697-0
    OpenUrlCrossRefPubMed
  31. ↵
    1. Li Z,
    2. Zheng J,
    3. Lin W,
    4. Weng J,
    5. Hong W,
    6. Zou J,
    7. Zhang T,
    8. Ye C and
    9. Chen Y
    : Circular RNA hsa_circ_0001785 inhibits the proliferation, migration and invasion of breast cancer cells in vitro and in vivo by sponging miR-942 to upregulate SOCS3. Cell Cycle 19(21): 2811-2825, 2020. PMID: 33054543. DOI: 10.1080/15384101.2020.1824717
    OpenUrlCrossRefPubMed
  32. ↵
    1. Ghafouri-Fard S,
    2. Oskooei VK,
    3. Azari I and
    4. Taheri M
    : Suppressor of cytokine signaling (SOCS) genes are downregulated in breast cancer. World J Surg Oncol 16(1): 226, 2018. PMID: 30453988. DOI: 10.1186/s12957-018-1529-9
    OpenUrlCrossRefPubMed
  33. ↵
    1. Morris R,
    2. Kershaw NJ and
    3. Babon JJ
    : The molecular details of cytokine signaling via the JAK/STAT pathway. Protein Sci 27(12): 1984-2009, 2018. PMID: 30267440. DOI: 10.1002/pro.3519
    OpenUrlCrossRefPubMed
  34. ↵
    1. Farabegoli F,
    2. Ceccarelli C,
    3. Santini D and
    4. Taffurelli M
    : Suppressor of cytokine signalling 2 (SOCS-2) expression in breast carcinoma. J Clin Pathol 58(10): 1046-1050, 2005. PMID: 16189149. DOI: 10.1136/jcp.2004.024919
    OpenUrlAbstract/FREE Full Text
  35. ↵
    1. Nakagawa T,
    2. Iida S,
    3. Osanai T,
    4. Uetake H,
    5. Aruga T,
    6. Toriya Y,
    7. Takagi Y,
    8. Kawachi H and
    9. Sugihara K
    : Decreased expression of SOCS-3 mRNA in breast cancer with lymph node metastasis. Oncol Rep 19(1): 33-39, 2008. PMID: 18097573.
    OpenUrlPubMed
  36. ↵
    1. Su N,
    2. Liu L,
    3. He S and
    4. Zeng L
    : Circ_0001666 affects miR-620/WNK2 axis to inhibit breast cancer progression. Genes Genomics 43(8): 947-959, 2021. PMID: 34061329. DOI: 10.1007/s13258-021-01114-y
    OpenUrlCrossRefPubMed
  37. ↵
    1. Moniz S,
    2. Veríssimo F,
    3. Matos P,
    4. Brazão R,
    5. Silva E,
    6. Kotelevets L,
    7. Chastre E,
    8. Gespach C and
    9. Jordan P
    : Protein kinase WNK2 inhibits cell proliferation by negatively modulating the activation of MEK1/ERK1/2. Oncogene 26(41): 6071-6081, 2007. PMID: 17667937. DOI: 10.1038/sj.onc.1210706
    OpenUrlCrossRefPubMed
  38. ↵
    1. Huang L and
    2. Liu X
    : microRNA-370 promotes cell growth by targeting WNK2 in breast cancer. DNA Cell Biol 38(6): 501-509, 2019. PMID: 31009242. DOI: 10.1089/dna.2018.4602
    OpenUrlCrossRefPubMed
  39. ↵
    1. Dutruel C,
    2. Bergmann F,
    3. Rooman I,
    4. Zucknick M,
    5. Weichenhan D,
    6. Geiselhart L,
    7. Kaffenberger T,
    8. Rachakonda PS,
    9. Bauer A,
    10. Giese N,
    11. Hong C,
    12. Xie H,
    13. Costello JF,
    14. Hoheisel J,
    15. Kumar R,
    16. Rehli M,
    17. Schirmacher P,
    18. Werner J,
    19. Plass C,
    20. Popanda O and
    21. Schmezer P
    : Early epigenetic downregulation of WNK2 kinase during pancreatic ductal adenocarcinoma development. Oncogene 33(26): 3401-3410, 2014. PMID: 23912455. DOI: 10.1038/onc.2013.312
    OpenUrlCrossRefPubMed
  40. ↵
    1. Zhou SL,
    2. Zhou ZJ,
    3. Hu ZQ,
    4. Song CL,
    5. Luo YJ,
    6. Luo CB,
    7. Xin HY,
    8. Yang XR,
    9. Shi YH,
    10. Wang Z,
    11. Huang XW,
    12. Cao Y,
    13. Fan J and
    14. Zhou J
    : Genomic sequencing identifies WNK2 as a driver in hepatocellular carcinoma and a risk factor for early recurrence. J Hepatol 71(6): 1152-1163, 2019. PMID: 31349001. DOI: 10.1016/j.jhep.2019.07.014
    OpenUrlCrossRefPubMed
  41. ↵
    1. Yang W,
    2. Gong P,
    3. Yang Y,
    4. Yang C,
    5. Yang B and
    6. Ren L
    : Circ-ABCB10 contributes to paclitaxel resistance in breast cancer through Let-7a-5p/DUSP7 axis. Cancer Manag Res 12: 2327-2337, 2020. PMID: 32273769. DOI: 10.2147/CMAR.S238513
    OpenUrlCrossRefPubMed
  42. ↵
    1. Keyse SM
    : Dual-specificity MAP kinase phosphatases (MKPs) and cancer. Cancer Metastasis Rev 27(2): 253-261, 2008. PMID: 18330678. DOI: 10.1007/s10555-008-9123-1
    OpenUrlCrossRefPubMed
  43. ↵
    1. Luan T,
    2. Zhang X,
    3. Wang S,
    4. Song Y,
    5. Zhou S,
    6. Lin J,
    7. An W,
    8. Yuan W,
    9. Yang Y,
    10. Cai H,
    11. Zhang Q and
    12. Wang L
    : Long non-coding RNA MIAT promotes breast cancer progression and functions as ceRNA to regulate DUSP7 expression by sponging miR-155-5p. Oncotarget 8(44): 76153-76164, 2017. PMID: 29100300. DOI: 10.18632/oncotarget.19190
    OpenUrlCrossRefPubMed
  44. ↵
    1. Gao PP,
    2. Qi XW,
    3. Sun N,
    4. Sun YY,
    5. Zhang Y,
    6. Tan XN,
    7. Ding J,
    8. Han F and
    9. Zhang Y
    : The emerging roles of dual-specificity phosphatases and their specific characteristics in human cancer. Biochim Biophys Acta Rev Cancer 1876(1): 188562, 2021. PMID: 33964330. DOI: 10.1016/j.bbcan.2021.188562
    OpenUrlCrossRefPubMed
  45. ↵
    1. Abu Samaan TM,
    2. Samec M,
    3. Liskova A,
    4. Kubatka P and
    5. Büsselberg D
    : Paclitaxel’s mechanistic and clinical effects on breast cancer. Biomolecules 9(12): 789, 2019. PMID: 31783552. DOI: 10.3390/biom9120789
    OpenUrlCrossRefPubMed
  46. ↵
    1. Zang H,
    2. Li Y,
    3. Zhang X and
    4. Huang G
    : Circ-RNF111 contributes to paclitaxel resistance in breast cancer by elevating E2F3 expression via miR-140-5p. Thorac Cancer 11(7): 1891-1903, 2020. PMID: 32445273. DOI: 10.1111/1759-7714.13475
    OpenUrlCrossRefPubMed
  47. ↵
    1. Liu ZL,
    2. Bi XW,
    3. Liu PP,
    4. Lei DX,
    5. Wang Y,
    6. Li ZM,
    7. Jiang WQ and
    8. Xia Y
    : Expressions and prognostic values of the E2F transcription factors in human breast carcinoma. Cancer Manag Res 10: 3521-3532, 2018. PMID: 30271201. DOI: 10.2147/CMAR.S172332
    OpenUrlCrossRefPubMed
  48. ↵
    1. Jusino S,
    2. Rivera-Rivera Y,
    3. Chardón-Colón C,
    4. Ruiz-Justiz AJ,
    5. Vélez-Velázquez J,
    6. Isidro A,
    7. Cruz-Robles ME,
    8. Bonilla-Claudio M,
    9. Armaiz-Pena GN and
    10. Saavedra HI
    : E2F3 drives the epithelial-to-mesenchymal transition, cell invasion, and metastasis in breast cancer. Exp Biol Med (Maywood) 246(19): 2057-2071, 2021. PMID: 34365840. DOI: 10.1177/15353702211035693
    OpenUrlCrossRefPubMed
  49. ↵
    1. Lee M,
    2. Oprea-Ilies G and
    3. Saavedra HI
    : Silencing of E2F3 suppresses tumor growth of Her2+ breast cancer cells by restricting mitosis. Oncotarget 6(35): 37316-37334, 2015. PMID: 26512919. DOI: 10.18632/oncotarget.5686
    OpenUrlCrossRefPubMed
  50. ↵
    1. Ni J,
    2. Xi X,
    3. Xiao S and
    4. Xiao X
    : Silencing of circHIPK3 sensitizes paclitaxel-resistant breast cancer cells to chemotherapy by regulating HK2 through targeting miR-1286. Cancer Manag Res 13: 5573-5585, 2021. PMID: 34285578. DOI: 10.2147/CMAR.S307595
    OpenUrlCrossRefPubMed
  51. ↵
    1. Zhang T,
    2. Zhu X,
    3. Wu H,
    4. Jiang K,
    5. Zhao G,
    6. Shaukat A,
    7. Deng G and
    8. Qiu C
    : Targeting the ROS/PI3K/AKT/HIF-1α/HK2 axis of breast cancer cells: Combined administration of Polydatin and 2-Deoxy-d-glucose. J Cell Mol Med 23(5): 3711-3723, 2019. PMID: 30920152. DOI: 10.1111/jcmm.14276
    OpenUrlCrossRefPubMed
  52. ↵
    1. Garcia SN,
    2. Guedes RC and
    3. Marques MM
    : Unlocking the potential of HK2 in cancer metabolism and therapeutics. Curr Med Chem 26(41): 7285-7322, 2019. PMID: 30543165. DOI: 10.2174/0929867326666181213092652
    OpenUrlCrossRefPubMed
  53. ↵
    1. Yang T,
    2. Ren C,
    3. Qiao P,
    4. Han X,
    5. Wang L,
    6. Lv S,
    7. Sun Y,
    8. Liu Z,
    9. Du Y and
    10. Yu Z
    : PIM2-mediated phosphorylation of hexokinase 2 is critical for tumor growth and paclitaxel resistance in breast cancer. Oncogene 37(45): 5997-6009, 2018. PMID: 29985480. DOI: 10.1038/s41388-018-0386-x
    OpenUrlCrossRefPubMed
  54. ↵
    1. Liu G,
    2. Zhang Z,
    3. Song Q,
    4. Guo Y,
    5. Bao P and
    6. Shui H
    : Circ_0006528 contributes to paclitaxel resistance of breast cancer cells by regulating miR-1299/CDK8 axis. Onco Targets Ther 13: 9497-9511, 2020. PMID: 33061434. DOI: 10.2147/OTT.S252886
    OpenUrlCrossRefPubMed
  55. ↵
    1. Li Z,
    2. Zhang Y,
    3. Zhang Z,
    4. Zhao Z and
    5. Lv Q
    : A four-gene signature predicts the efficacy of paclitaxel-based neoadjuvant therapy in human epidermal growth factor receptor 2-negative breast cancer. J Cell Biochem 120(4): 6046-6056, 2019. PMID: 30520096. DOI: 10.1002/jcb.27891
    OpenUrlCrossRefPubMed
  56. ↵
    1. Wu D,
    2. Jia H,
    3. Zhang Z and
    4. Li S
    : Capsaicin suppresses breast cancer cell viability by regulating the CDK8/PI3K/Akt/Wnt/β catenin signaling pathway. Mol Med Rep 22(6): 4868-4876, 2020. PMID: 33173974. DOI: 10.3892/mmr.2020.11585
    OpenUrlCrossRefPubMed
  57. ↵
    1. Menzl I,
    2. Witalisz-Siepracka A and
    3. Sexl V
    : CDK8-novel therapeutic opportunities. Pharmaceuticals (Basel) 12(2): 92, 2019. PMID: 31248103. DOI: 10.3390/ph12020092
    OpenUrlCrossRefPubMed
  58. ↵
    1. Li XY,
    2. Luo QF,
    3. Wei CK,
    4. Li DF and
    5. Fang L
    : siRNA-mediated silencing of CDK8 inhibits proliferation and growth in breast cancer cells. Int J Clin Exp Pathol 7(1): 92-100, 2013. PMID: 24427329.
    OpenUrlPubMed
  59. ↵
    1. McDermott MS,
    2. Chumanevich AA,
    3. Lim CU,
    4. Liang J,
    5. Chen M,
    6. Altilia S,
    7. Oliver D,
    8. Rae JM,
    9. Shtutman M,
    10. Kiaris H,
    11. Győrffy B,
    12. Roninson IB and
    13. Broude EV
    : Inhibition of CDK8 mediator kinase suppresses estrogen dependent transcription and the growth of estrogen receptor positive breast cancer. Oncotarget 8(8): 12558-12575, 2017. PMID: 28147342. DOI: 10.18632/oncotarget.14894
    OpenUrlCrossRefPubMed
  60. ↵
    1. Hao J,
    2. Du X,
    3. Lv F and
    4. Shi Q
    : Knockdown of circ_0006528 suppresses cell proliferation, migration, invasion, and adriamycin chemoresistance via regulating the miR-1236-3p/CHD4 axis in breast cancer. J Surg Res 260: 104-115, 2021. PMID: 33333383. DOI: 10.1016/j.jss.2020.10.031
    OpenUrlCrossRefPubMed
  61. ↵
    1. Tong JK,
    2. Hassig CA,
    3. Schnitzler GR,
    4. Kingston RE and
    5. Schreiber SL
    : Chromatin deacetylation by an ATP-dependent nucleosome remodelling complex. Nature 395(6705): 917-921, 1998. PMID: 9804427. DOI: 10.1038/27699
    OpenUrlCrossRefPubMed
  62. ↵
    1. Wang Y,
    2. Chen Y,
    3. Bao L,
    4. Zhang B,
    5. Wang JE,
    6. Kumar A,
    7. Xing C,
    8. Wang Y and
    9. Luo W
    : CHD4 promotes breast cancer progression as a coactivator of hypoxia-inducible factors. Cancer Res 80(18): 3880-3891, 2020. PMID: 32699137. DOI: 10.1158/0008-5472.CAN-20-1049
    OpenUrlAbstract/FREE Full Text
  63. ↵
    1. D’Alesio C,
    2. Bellese G,
    3. Gagliani MC,
    4. Lechiara A,
    5. Dameri M,
    6. Grasselli E,
    7. Lanfrancone L,
    8. Cortese K and
    9. Castagnola P
    : The chromodomain helicase CHD4 regulates ERBB2 signaling pathway and autophagy in ERBB2(+) breast cancer cells. Biol Open 8(4): bio038323, 2019. PMID: 30967373. DOI: 10.1242/bio.038323
    OpenUrlAbstract/FREE Full Text
  64. ↵
    1. D’Alesio C,
    2. Punzi S,
    3. Cicalese A,
    4. Fornasari L,
    5. Furia L,
    6. Riva L,
    7. Carugo A,
    8. Curigliano G,
    9. Criscitiello C,
    10. Pruneri G,
    11. Pelicci PG,
    12. Faretta M,
    13. Bossi D and
    14. Lanfrancone L
    : RNAi screens identify CHD4 as an essential gene in breast cancer growth. Oncotarget 7(49): 80901-80915, 2016. PMID: 27779108. DOI: 10.18632/oncotarget.12646
    OpenUrlCrossRefPubMed
  65. ↵
    1. Novillo A,
    2. Fernández-Santander A,
    3. Gaibar M,
    4. Galán M,
    5. Romero-Lorca A,
    6. El Abdellaoui-Soussi F and
    7. Gómez-Del Arco P
    : Role of chromodomain-helicase-DNA-binding protein 4 (CHD4) in breast cancer. Front Oncol 11: 633233, 2021. PMID: 33981601. DOI: 10.3389/fonc.2021.633233
    OpenUrlCrossRefPubMed
  66. ↵
    1. Zhang J,
    2. Shih DJH and
    3. Lin SY
    : The tale of CHD4 in DNA damage response and chemotherapeutic response. J Cancer Res Cell Ther 3(1): 052, 2019. PMID: 32577620.
    OpenUrlPubMed
  67. ↵
    1. Cui Y,
    2. Fan J,
    3. Shi W and
    4. Zhou Z
    : Circ_0001667 knockdown blocks cancer progression and attenuates adriamycin resistance by depleting NCOA3 via releasing miR-4458 in breast cancer. Drug Dev Res 83(1): 75-87, 2022. PMID: 34227151. DOI: 10.1002/ddr.21845
    OpenUrlCrossRefPubMed
  68. ↵
    1. Li L,
    2. Deng CX and
    3. Chen Q
    : SRC-3, a steroid receptor coactivator: implication in cancer. Int J Mol Sci 22(9): 4760, 2021. PMID: 33946224. DOI: 10.3390/ijms22094760
    OpenUrlCrossRefPubMed
  69. ↵
    1. Lahusen T,
    2. Henke RT,
    3. Kagan BL,
    4. Wellstein A and
    5. Riegel AT
    : The role and regulation of the nuclear receptor co-activator AIB1 in breast cancer. Breast Cancer Res Treat 116(2): 225-237, 2009. PMID: 19418218. DOI: 10.1007/s10549-009-0405-2
    OpenUrlCrossRefPubMed
  70. ↵
    1. Xie H and
    2. Zheng R
    : Circ_0085495 knockdown reduces adriamycin resistance in breast cancer through miR-873-5p/integrin β1 axis. Anticancer Drugs 33(1): e166-e177, 2022. PMID: 34387598. DOI: 10.1097/CAD.0000000000001174
    OpenUrlCrossRefPubMed
  71. ↵
    1. Baltes F,
    2. Pfeifer V,
    3. Silbermann K,
    4. Caspers J,
    5. Wantoch von Rekowski K,
    6. Schlesinger M and
    7. Bendas G
    : β(1)-Integrin binding to collagen type 1 transmits breast cancer cells into chemoresistance by activating ABC efflux transporters. Biochim Biophys Acta Mol Cell Res 1867(5): 118663, 2020. PMID: 31987794. DOI: 10.1016/j.bbamcr.2020.118663
    OpenUrlCrossRefPubMed
  72. ↵
    1. Mills JN,
    2. Rutkovsky AC and
    3. Giordano A
    : Mechanisms of resistance in estrogen receptor positive breast cancer: overcoming resistance to tamoxifen/aromatase inhibitors. Curr Opin Pharmacol 41: 59-65, 2018. PMID: 29719270. DOI: 10.1016/j.coph.2018.04.009
    OpenUrlCrossRefPubMed
  73. ↵
    1. Clarke R,
    2. Tyson JJ and
    3. Dixon JM
    : Endocrine resistance in breast cancer – An overview and update. Mol Cell Endocrinol 418 Pt 3(0 3): 220-234, 2015. PMID: 26455641. DOI: 10.1016/j.mce.2015.09.035
    OpenUrlCrossRefPubMed
  74. ↵
    1. Hu K,
    2. Liu X,
    3. Li Y,
    4. Li Q,
    5. Xu Y,
    6. Zeng W,
    7. Zhong G and
    8. Yu C
    : Exosomes mediated transfer of Circ_UBE2D2 enhances the resistance of breast cancer to tamoxifen by binding to MiR-200a-3p. Med Sci Monit 26: e922253, 2020. PMID: 32756532. DOI: 10.12659/MSM.922253
    OpenUrlCrossRefPubMed
  75. ↵
    1. Oh DY and
    2. Bang YJ
    : HER2-targeted therapies - a role beyond breast cancer. Nat Rev Clin Oncol 17(1): 33-48, 2020. PMID: 31548601. DOI: 10.1038/s41571-019-0268-3
    OpenUrlCrossRefPubMed
  76. ↵
    1. Wu X,
    2. Ren Y,
    3. Yao R,
    4. Zhou L and
    5. Fan R
    : Circular RNA circ-MMP11 contributes to lapatinib resistance of breast cancer cells by regulating the miR-153-3p/ANLN axis. Front Oncol 11: 639961, 2021. PMID: 34295807. DOI: 10.3389/fonc.2021.639961
    OpenUrlCrossRefPubMed
  77. ↵
    1. Wang G,
    2. Shen W,
    3. Cui L,
    4. Chen W,
    5. Hu X and
    6. Fu J
    : Overexpression of Anillin (ANLN) is correlated with colorectal cancer progression and poor prognosis. Cancer Biomark 16(3): 459-465, 2016. PMID: 27062703. DOI: 10.3233/CBM-160585
    OpenUrlCrossRefPubMed
  78. ↵
    1. Zhou W,
    2. Wang Z,
    3. Shen N,
    4. Pi W,
    5. Jiang W,
    6. Huang J,
    7. Hu Y,
    8. Li X and
    9. Sun L
    : Knockdown of ANLN by lentivirus inhibits cell growth and migration in human breast cancer. Mol Cell Biochem 398(1-2): 11-19, 2015. PMID: 25223638. DOI: 10.1007/s11010-014-2200-6
    OpenUrlCrossRefPubMed
  79. ↵
    1. Wang Z,
    2. Chen J,
    3. Zhong MZ,
    4. Huang J,
    5. Hu YP,
    6. Feng DY,
    7. Zhou ZJ,
    8. Luo X,
    9. Liu ZQ,
    10. Jiang WZ and
    11. Zhou WB
    : Overexpression of ANLN contributed to poor prognosis of anthracycline-based chemotherapy in breast cancer patients. Cancer Chemother Pharmacol 79(3): 535-543, 2017. PMID: 28243684. DOI: 10.1007/s00280-017-3248-2
    OpenUrlCrossRefPubMed
  80. ↵
    1. Sui C,
    2. Qu W,
    3. Lian Y,
    4. Feng C and
    5. Zhan Y
    : Hsa_circ_0069094 knockdown inhibits cell proliferation, migration, invasion and glycolysis, while induces cell apoptosis by miR-661/HMGA1 axis in breast cancer. Anticancer Drugs 32(8): 829-841, 2021. PMID: 33929992. DOI: 10.1097/CAD.0000000000001076
    OpenUrlCrossRefPubMed
  81. ↵
    1. Zhu M,
    2. Wang Y,
    3. Wang F,
    4. Li L and
    5. Qiu X
    : CircFBXL5 promotes the 5-FU resistance of breast cancer via modulating miR-216b/HMGA2 axis. Cancer Cell Int 21(1): 384, 2021. PMID: 34281530. DOI: 10.1186/s12935-021-02088-3
    OpenUrlCrossRefPubMed
  82. ↵
    1. Chen ZG,
    2. Zhao HJ,
    3. Lin L,
    4. Liu JB,
    5. Bai JZ and
    6. Wang GS
    : Circular RNA CirCHIPK3 promotes cell proliferation and invasion of breast cancer by sponging miR-193a/HMGB1/PI3K/AKT axis. Thorac Cancer 11(9): 2660-2671, 2020. PMID: 32767499. DOI: 10.1111/1759-7714.13603
    OpenUrlCrossRefPubMed
  83. ↵
    1. Zhang J,
    2. Ke S,
    3. Zheng W,
    4. Zhu Z and
    5. Wu Y
    : Hsa_circ_0003645 promotes breast cancer progression by regulating miR-139-3p/HMGB1 axis. Onco Targets Ther 13: 10361-10372, 2020. PMID: 33116616. DOI: 10.2147/OTT.S265796
    OpenUrlCrossRefPubMed
  84. ↵
    1. Ozturk N,
    2. Singh I,
    3. Mehta A,
    4. Braun T and
    5. Barreto G
    : HMGA proteins as modulators of chromatin structure during transcriptional activation. Front Cell Dev Biol 2: 5, 2014. PMID: 25364713. DOI: 10.3389/fcell.2014.00005
    OpenUrlCrossRefPubMed
  85. ↵
    1. Huang R,
    2. Huang D,
    3. Dai W and
    4. Yang F
    : Overexpression of HMGA1 correlates with the malignant status and prognosis of breast cancer. Mol Cell Biochem 404(1-2): 251-257, 2015. PMID: 25772486. DOI: 10.1007/s11010-015-2384-4
    OpenUrlCrossRefPubMed
  86. ↵
    1. Wu J,
    2. Zhang S,
    3. Shan J,
    4. Hu Z,
    5. Liu X,
    6. Chen L,
    7. Ren X,
    8. Yao L,
    9. Sheng H,
    10. Li L,
    11. Ann D,
    12. Yen Y,
    13. Wang J and
    14. Wang X
    : Elevated HMGA2 expression is associated with cancer aggressiveness and predicts poor outcome in breast cancer. Cancer Lett 376(2): 284-292, 2016. PMID: 27063096. DOI: 10.1016/j.canlet.2016.04.005
    OpenUrlCrossRefPubMed
  87. ↵
    1. Di Cello F,
    2. Shin J,
    3. Harbom K and
    4. Brayton C
    : Knockdown of HMGA1 inhibits human breast cancer cell growth and metastasis in immunodeficient mice. Biochem Biophys Res Commun 434(1): 70-74, 2013. PMID: 23545254. DOI: 10.1016/j.bbrc.2013.03.064
    OpenUrlCrossRefPubMed
  88. ↵
    1. Zanin R,
    2. Pegoraro S,
    3. Ros G,
    4. Ciani Y,
    5. Piazza S,
    6. Bossi F,
    7. Bulla R,
    8. Zennaro C,
    9. Tonon F,
    10. Lazarevic D,
    11. Stupka E,
    12. Sgarra R and
    13. Manfioletti G
    : HMGA1 promotes breast cancer angiogenesis supporting the stability, nuclear localization and transcriptional activity of FOXM1. J Exp Clin Cancer Res 38(1): 313, 2019. PMID: 31311575. DOI: 10.1186/s13046-019-1307-8
    OpenUrlCrossRefPubMed
  89. ↵
    1. Mansoori B,
    2. Duijf PHG,
    3. Mohammadi A,
    4. Najafi S,
    5. Roshani E,
    6. Shanehbandi D,
    7. Hajiasgharzadeh K,
    8. Shirjang S,
    9. Ditzel HJ,
    10. Kazemi T,
    11. Mokhtarzadeh A,
    12. Gjerstorff MF and
    13. Baradaran B
    : Overexpression of HMGA2 in breast cancer promotes cell proliferation, migration, invasion and stemness. Expert Opin Ther Targets: 1-11, 2020. PMID: 32172636. DOI: 10.1080/14728222.2020.1736559
    OpenUrlCrossRefPubMed
  90. ↵
    1. Zhang S,
    2. Mo Q and
    3. Wang X
    : Oncological role of HMGA2 (Review). Int J Oncol 55(4): 775-788, 2019. PMID: 31432151. DOI: 10.3892/ijo.2019.4856
    OpenUrlCrossRefPubMed
  91. ↵
    1. Méndez O,
    2. Pérez J,
    3. Soberino J,
    4. Racca F,
    5. Cortés J and
    6. Villanueva J
    : Clinical implications of extracellular HMGA1 in breast cancer. Int J Mol Sci 20(23): 5950, 2019. PMID: 31779212. DOI: 10.3390/ijms20235950
    OpenUrlCrossRefPubMed
  92. ↵
    1. Xu J,
    2. Fang X,
    3. Long L,
    4. Wang S,
    5. Qian S and
    6. Lyu J
    : HMGA2 promotes breast cancer metastasis by modulating Hippo-YAP signaling pathway. Cancer Biol Ther 22(1): 5-11, 2021. PMID: 33307962. DOI: 10.1080/15384047.2020.1832429
    OpenUrlCrossRefPubMed
  93. ↵
    1. Pan G,
    2. Mao A,
    3. Liu J,
    4. Lu J,
    5. Ding J and
    6. Liu W
    : Circular RNA hsa_circ_0061825 (circ-TFF1) contributes to breast cancer progression through targeting miR-326/TFF1 signalling. Cell Prolif 53(2): e12720, 2020. PMID: 31961997. DOI: 10.1111/cpr.12720
    OpenUrlCrossRefPubMed
  94. ↵
    1. Thim L
    : A new family of growth factor-like peptides. ‘Trefoil’ disulphide loop structures as a common feature in breast cancer associated peptide (pS2), pancreatic spasmolytic polypeptide (PSP), and frog skin peptides (spasmolysins). FEBS Lett 250(1): 85-90, 1989. PMID: 2737304. DOI: 10.1016/0014-5793(89)80690-8
    OpenUrlCrossRefPubMed
  95. ↵
    1. Buache E,
    2. Etique N,
    3. Alpy F,
    4. Stoll I,
    5. Muckensturm M,
    6. Reina-San-Martin B,
    7. Chenard MP,
    8. Tomasetto C and
    9. Rio MC
    : Deficiency in trefoil factor 1 (TFF1) increases tumorigenicity of human breast cancer cells and mammary tumor development in TFF1-knockout mice. Oncogene 30(29): 3261-3273, 2011. PMID: 21358676. DOI: 10.1038/onc.2011.41
    OpenUrlCrossRefPubMed
  96. ↵
    1. Prest SJ,
    2. May FE and
    3. Westley BR
    : The estrogen-regulated protein, TFF1, stimulates migration of human breast cancer cells. FASEB J 16(6): 592-594, 2002. PMID: 11919164. DOI: 10.1096/fj.01-0498fje
    OpenUrlCrossRefPubMed
  97. ↵
    1. Yi J,
    2. Ren L,
    3. Li D,
    4. Wu J,
    5. Li W,
    6. Du G and
    7. Wang J
    : Trefoil factor 1 (TFF1) is a potential prognostic biomarker with functional significance in breast cancers. Biomed Pharmacother 124: 109827, 2020. PMID: 31986408. DOI: 10.1016/j.biopha.2020.109827
    OpenUrlCrossRefPubMed
  98. ↵
    1. Elnagdy MH,
    2. Farouk O,
    3. Seleem AK and
    4. Nada HA
    : TFF1 and TFF3 mRNAs are higher in blood from breast cancer patients with metastatic disease than those without. J Oncol 2018: 4793498, 2018. PMID: 29977293. DOI: 10.1155/2018/4793498
    OpenUrlCrossRefPubMed
  99. ↵
    1. Spadazzi C,
    2. Mercatali L,
    3. Esposito M,
    4. Wei Y,
    5. Liverani C,
    6. De Vita A,
    7. Miserocchi G,
    8. Carretta E,
    9. Zanoni M,
    10. Cocchi C,
    11. Bongiovanni A,
    12. Recine F,
    13. Kang Y and
    14. Ibrahim T
    : Trefoil factor-1 upregulation in estrogen-receptor positive breast cancer correlates with an increased risk of bone metastasis. Bone 144: 115775, 2021. PMID: 33249323. DOI: 10.1016/j.bone.2020.115775
    OpenUrlCrossRefPubMed
  100. ↵
    1. Cai F,
    2. Fu W,
    3. Tang L,
    4. Tang J,
    5. Sun J,
    6. Fu G and
    7. Ye G
    : Hsa_circ_0000515 is a novel circular RNA implicated in the development of breast cancer through its regulation of the microRNA-296-5p/CXCL10 axis. FEBS J 288(3): 861-883, 2021. PMID: 32446265. DOI: 10.1111/febs.15373
    OpenUrlCrossRefPubMed
  101. ↵
    1. Liu M,
    2. Guo S and
    3. Stiles JK
    : The emerging role of CXCL10 in cancer (Review). Oncol Lett 2(4): 583-589, 2011. PMID: 22848232. DOI: 10.3892/ol.2011.300
    OpenUrlCrossRefPubMed
  102. ↵
    1. Wu X,
    2. Sun A,
    3. Yu W,
    4. Hong C and
    5. Liu Z
    : CXCL10 mediates breast cancer tamoxifen resistance and promotes estrogen-dependent and independent proliferation. Mol Cell Endocrinol 512: 110866, 2020. PMID: 32417506. DOI: 10.1016/j.mce.2020.110866
    OpenUrlCrossRefPubMed
  103. ↵
    1. Datta D,
    2. Flaxenburg JA,
    3. Laxmanan S,
    4. Geehan C,
    5. Grimm M,
    6. Waaga-Gasser AM,
    7. Briscoe DM and
    8. Pal S
    : Ras-induced modulation of CXCL10 and its receptor splice variant CXCR3-B in MDA-MB-435 and MCF-7 cells: relevance for the development of human breast cancer. Cancer Res 66(19): 9509-9518, 2006. PMID: 17018607. DOI: 10.1158/0008-5472.CAN-05-4345
    OpenUrlAbstract/FREE Full Text
  104. ↵
    1. Aksoy MO,
    2. Yang Y,
    3. Ji R,
    4. Reddy PJ,
    5. Shahabuddin S,
    6. Litvin J,
    7. Rogers TJ and
    8. Kelsen SG
    : CXCR3 surface expression in human airway epithelial cells: cell cycle dependence and effect on cell proliferation. Am J Physiol Lung Cell Mol Physiol 290(5): L909-L918, 2006. PMID: 16339779. DOI: 10.1152/ajplung.00430.2005
    OpenUrlCrossRefPubMed
  105. ↵
    1. Alassaf E and
    2. Mueller A
    : The role of PKC in CXCL8 and CXCL10 directed prostate, breast and leukemic cancer cell migration. Eur J Pharmacol 886: 173453, 2020. PMID: 32777211. DOI: 10.1016/j.ejphar.2020.173453
    OpenUrlCrossRefPubMed
  106. ↵
    1. Balkwill F
    : Cancer and the chemokine network. Nat Rev Cancer 4(7): 540-550, 2004. PMID: 15229479. DOI: 10.1038/nrc1388
    OpenUrlCrossRefPubMed
  107. ↵
    1. Lin G,
    2. Wang S,
    3. Zhang X and
    4. Wang D
    : Circular RNA circPLK1 promotes breast cancer cell proliferation, migration and invasion by regulating miR-4500/IGF1 axis. Cancer Cell Int 20(1): 593, 2020. PMID: 33298061. DOI: 10.1186/s12935-020-01694-x
    OpenUrlCrossRefPubMed
  108. ↵
    1. Christopoulos PF,
    2. Msaouel P and
    3. Koutsilieris M
    : The role of the insulin-like growth factor-1 system in breast cancer. Mol Cancer 14: 43, 2015. PMID: 25743390. DOI: 10.1186/s12943-015-0291-7
    OpenUrlCrossRefPubMed
  109. ↵
    1. Malaguarnera R and
    2. Belfiore A
    : The insulin receptor: a new target for cancer therapy. Front Endocrinol (Lausanne) 2: 93, 2011. PMID: 22654833. DOI: 10.3389/fendo.2011.00093
    OpenUrlCrossRefPubMed
  110. ↵
    1. Rieunier G,
    2. Wu X,
    3. Macaulay VM,
    4. Lee AV,
    5. Weyer-Czernilofsky U and
    6. Bogenrieder T
    : Bad to the bone: the role of the insulin-like growth factor axis in osseous metastasis. Clin Cancer Res 25(12): 3479-3485, 2019. PMID: 30745299. DOI: 10.1158/1078-0432.CCR-18-2697
    OpenUrlAbstract/FREE Full Text
  111. ↵
    1. Kang Y,
    2. Siegel PM,
    3. Shu W,
    4. Drobnjak M,
    5. Kakonen SM,
    6. Cordón-Cardo C,
    7. Guise TA and
    8. Massagué J
    : A multigenic program mediating breast cancer metastasis to bone. Cancer Cell 3(6): 537-549, 2003. PMID: 12842083. DOI: 10.1016/s1535-6108(03)00132-6
    OpenUrlCrossRefPubMed
  112. ↵
    1. Qiu Z,
    2. Wang L and
    3. Liu H
    : Hsa_circ_0001982 promotes the progression of breast cancer through miR-1287-5p/MUC19 axis under hypoxia. World J Surg Oncol 19(1): 161, 2021. PMID: 34082777. DOI: 10.1186/s12957-021-02273-8
    OpenUrlCrossRefPubMed
  113. ↵
    1. Ratan C,
    2. Cicily K D D,
    3. Nair B and
    4. Nath LR
    : MUC glycoproteins: potential biomarkers and molecular targets for cancer therapy. Curr Cancer Drug Targets 21(2): 132-152, 2021. PMID: 33200711. DOI: 10.2174/1568009620666201116113334
    OpenUrlCrossRefPubMed
  114. ↵
    1. Mukhopadhyay P,
    2. Chakraborty S,
    3. Ponnusamy MP,
    4. Lakshmanan I,
    5. Jain M and
    6. Batra SK
    : Mucins in the pathogenesis of breast cancer: implications in diagnosis, prognosis and therapy. Biochim Biophys Acta 1815(2): 224-240, 2011. PMID: 21277939. DOI: 10.1016/j.bbcan.2011.01.001
    OpenUrlCrossRefPubMed
  115. ↵
    1. Aithal A,
    2. Rauth S,
    3. Kshirsagar P,
    4. Shah A,
    5. Lakshmanan I,
    6. Junker WM,
    7. Jain M,
    8. Ponnusamy MP and
    9. Batra SK
    : MUC16 as a novel target for cancer therapy. Expert Opin Ther Targets 22(8): 675-686, 2018. PMID: 29999426. DOI: 10.1080/14728222.2018.1498845
    OpenUrlCrossRefPubMed
  116. ↵
    1. Stewart D and
    2. Cristea M
    : Antibody-drug conjugates for ovarian cancer: current clinical development. Curr Opin Obstet Gynecol 31(1): 18-23, 2019. PMID: 30531606. DOI: 10.1097/GCO.0000000000000515
    OpenUrlCrossRefPubMed
  117. ↵
    1. Gao T,
    2. Cen Q and
    3. Lei H
    : A review on development of MUC1-based cancer vaccine. Biomed Pharmacother 132: 110888, 2020. PMID: 33113416. DOI: 10.1016/j.biopha.2020.110888
    OpenUrlCrossRefPubMed
  118. ↵
    1. Xu Y,
    2. Zhang S,
    3. Liao X,
    4. Li M,
    5. Chen S,
    6. Li X,
    7. Wu X,
    8. Yang M,
    9. Tang M,
    10. Hu Y,
    11. Li Z,
    12. Yu R,
    13. Huang M,
    14. Song L and
    15. Li J
    : Circular RNA circIKBKB promotes breast cancer bone metastasis through sustaining NF-κB/bone remodeling factors signaling. Mol Cancer 20(1): 98, 2021. PMID: 34325714. DOI: 10.1186/s12943-021-01394-8
    OpenUrlCrossRefPubMed
  119. ↵
    1. Ye J,
    2. She X,
    3. Liu Z,
    4. He Z,
    5. Gao X,
    6. Lu L,
    7. Liang R and
    8. Lin Y
    : Eukaryotic initiation factor 4A-3: a review of its physiological role and involvement in oncogenesis. Front Oncol 11: 712045, 2021. PMID: 34458150. DOI: 10.3389/fonc.2021.712045
    OpenUrlCrossRefPubMed
  120. ↵
    1. Zhu Y,
    2. Ren C and
    3. Yang L
    : Effect of eukaryotic translation initiation factor 4A3 in malignant tumors. Oncol Lett 21(5): 358, 2021. PMID: 33747215. DOI: 10.3892/ol.2021.12619
    OpenUrlCrossRefPubMed
  121. ↵
    1. Weidle UH,
    2. Birzele F,
    3. Kollmorgen G and
    4. Rüger R
    : Molecular mechanisms of bone metastasis. Cancer Genomics Proteomics 13(1): 1-12, 2016. PMID: 26708594.
    OpenUrlAbstract/FREE Full Text
  122. ↵
    1. Fornetti J,
    2. Welm AL and
    3. Stewart SA
    : Understanding the bone in cancer metastasis. J Bone Miner Res 33(12): 2099-2113, 2018. PMID: 30476357. DOI: 10.1002/jbmr.3618
    OpenUrlCrossRefPubMed
  123. ↵
    1. Meng L,
    2. Liu S,
    3. Liu F,
    4. Sang M,
    5. Ju Y,
    6. Fan X,
    7. Gu L,
    8. Li Z,
    9. Geng C and
    10. Sang M
    : ZEB1-mediated transcriptional upregulation of circWWC3 promotes breast cancer progression through activating Ras signaling pathway. Mol Ther Nucleic Acids 22: 124-137, 2020. PMID: 32916598. DOI: 10.1016/j.omtn.2020.08.015
    OpenUrlCrossRefPubMed
  124. ↵
    1. Degirmenci U,
    2. Wang M and
    3. Hu J
    : Targeting aberrant RAS/RAF/MEK/ERK signaling for cancer therapy. Cells 9(1): 198, 2020. PMID: 31941155. DOI: 10.3390/cells9010198
    OpenUrlCrossRefPubMed
  125. ↵
    1. Thein KZ,
    2. Biter AB and
    3. Hong DS
    : Therapeutics targeting mutant KRAS. Annu Rev Med 72: 349-364, 2021. PMID: 33138715. DOI: 10.1146/annurev-med-080819-033145
    OpenUrlCrossRefPubMed
    1. Uprety D and
    2. Adjei AA
    : KRAS: From undruggable to a druggable cancer target. Cancer Treat Rev 89: 102070, 2020. PMID: 32711246. DOI: 10.1016/j.ctrv.2020.102070
    OpenUrlCrossRefPubMed
  126. ↵
    1. Ghimessy A,
    2. Radeczky P,
    3. Laszlo V,
    4. Hegedus B,
    5. Renyi-Vamos F,
    6. Fillinger J,
    7. Klepetko W,
    8. Lang C,
    9. Dome B and
    10. Megyesfalvi Z
    : Current therapy of KRAS-mutant lung cancer. Cancer Metastasis Rev 39(4): 1159-1177, 2020. PMID: 32548736. DOI: 10.1007/s10555-020-09903-9
    OpenUrlCrossRefPubMed
  127. ↵
    1. Zhou Y,
    2. Liu X,
    3. Lan J,
    4. Wan Y and
    5. Zhu X
    : Circular RNA circRPPH1 promotes triple-negative breast cancer progression via the miR-556-5p/YAP1 axis. Am J Transl Res 12(10): 6220-6234, 2020. PMID: 33194025.
    OpenUrlPubMed
  128. ↵
    1. Wang X,
    2. Ji C,
    3. Hu J,
    4. Deng X,
    5. Zheng W,
    6. Yu Y,
    7. Hua K,
    8. Zhou X and
    9. Fang L
    : Hsa_circ_0005273 facilitates breast cancer tumorigenesis by regulating YAP1-hippo signaling pathway. J Exp Clin Cancer Res 40(1): 29, 2021. PMID: 33436041. DOI: 10.1186/s13046-021-01830-z
    OpenUrlCrossRefPubMed
  129. ↵
    1. Wu D,
    2. Jia H,
    3. Zhang Z and
    4. Li S
    : Circ_0000511 accelerates the proliferation, migration and invasion, and restrains the apoptosis of breast cancer cells through the miR-326/TAZ axis. Int J Oncol 58(4): 1, 2021. PMID: 33649821. DOI: 10.3892/ijo.2021.5181
    OpenUrlCrossRefPubMed
  130. ↵
    1. Zanconato F,
    2. Cordenonsi M and
    3. Piccolo S
    : YAP and TAZ: a signalling hub of the tumour microenvironment. Nat Rev Cancer 19(8): 454-464, 2019. PMID: 31270418. DOI: 10.1038/s41568-019-0168-y
    OpenUrlCrossRefPubMed
    1. Halder G,
    2. Dupont S and
    3. Piccolo S
    : Transduction of mechanical and cytoskeletal cues by YAP and TAZ. Nat Rev Mol Cell Biol 13(9): 591-600, 2012. PMID: 22895435. DOI: 10.1038/nrm3416
    OpenUrlCrossRefPubMed
  131. ↵
    1. Zanconato F,
    2. Cordenonsi M and
    3. Piccolo S
    : YAP/TAZ at the roots of cancer. Cancer Cell 29(6): 783-803, 2016. PMID: 27300434. DOI: 10.1016/j.ccell.2016.05.005
    OpenUrlCrossRefPubMed
  132. ↵
    1. Zhao W,
    2. Wang M,
    3. Cai M,
    4. Zhang C,
    5. Qiu Y,
    6. Wang X,
    7. Zhang T,
    8. Zhou H,
    9. Wang J,
    10. Zhao W and
    11. Shao R
    : Transcriptional co-activators YAP/TAZ: Potential therapeutic targets for metastatic breast cancer. Biomed Pharmacother 133: 110956, 2021. PMID: 33189066. DOI: 10.1016/j.biopha.2020.110956
    OpenUrlCrossRefPubMed
  133. ↵
    1. Thompson BJ
    : YAP/TAZ: Drivers of tumor growth, metastasis, and resistance to therapy. Bioessays 42(5): e1900162, 2020. PMID: 32128850. DOI: 10.1002/bies.201900162
    OpenUrlCrossRefPubMed
  134. ↵
    1. Wu L and
    2. Yang X
    : Targeting the Hippo pathway for breast cancer therapy. Cancers (Basel) 10(11): 422, 2018. PMID: 30400599. DOI: 10.3390/cancers10110422
    OpenUrlCrossRefPubMed
  135. ↵
    1. Zanconato F,
    2. Battilana G,
    3. Forcato M,
    4. Filippi L,
    5. Azzolin L,
    6. Manfrin A,
    7. Quaranta E,
    8. Di Biagio D,
    9. Sigismondo G,
    10. Guzzardo V,
    11. Lejeune P,
    12. Haendler B,
    13. Krijgsveld J,
    14. Fassan M,
    15. Bicciato S,
    16. Cordenonsi M and
    17. Piccolo S
    : Transcriptional addiction in cancer cells is mediated by YAP/TAZ through BRD4. Nat Med 24(10): 1599-1610, 2018. PMID: 30224758. DOI: 10.1038/s41591-018-0158-8
    OpenUrlCrossRefPubMed
  136. ↵
    1. Zhang L,
    2. Sun D,
    3. Zhang J and
    4. Tian Y
    : Circ-UBR1 facilitates proliferation, metastasis, and inhibits apoptosis in breast cancer by regulating the miR-1299/CCND1 axis. Life Sci 266: 118829, 2021. PMID: 33259864. DOI: 10.1016/j.lfs.2020.118829
    OpenUrlCrossRefPubMed
  137. ↵
    1. Casimiro MC,
    2. Crosariol M,
    3. Loro E,
    4. Li Z and
    5. Pestell RG
    : Cyclins and cell cycle control in cancer and disease. Genes Cancer 3(11-12): 649-657, 2012. PMID: 23634253. DOI: 10.1177/1947601913479022
    OpenUrlCrossRefPubMed
  138. ↵
    1. Roy PG and
    2. Thompson AM
    : Cyclin D1 and breast cancer. Breast 15(6): 718-727, 2006. PMID: 16675218. DOI: 10.1016/j.breast.2006.02.005
    OpenUrlCrossRefPubMed
  139. ↵
    1. Akhter N,
    2. Alzahrani FA,
    3. Dar SA,
    4. Wahid M,
    5. Sattar RSA,
    6. Hussain S,
    7. Haque S,
    8. Ansari SA,
    9. Jawed A,
    10. Mandal RK,
    11. Almalki S,
    12. Alharbi RA and
    13. Husain SA
    : AA genotype of cyclin D1 G870A polymorphism increases breast cancer risk: Findings of a case-control study and meta-analysis. J Cell Biochem 120(10): 16452-16466, 2019. PMID: 31243808. DOI: 10.1002/jcb.28800
    OpenUrlCrossRefPubMed
  140. ↵
    1. Zhang L,
    2. Zhang W,
    3. Zuo Z,
    4. Tang J,
    5. Song Y,
    6. Cao F,
    7. Yu X,
    8. Liu S and
    9. Cai X
    : Circ_0008673 regulates breast cancer malignancy by miR-153-3p/CFL2 axis. Arch Gynecol Obstet 305(1): 223-232, 2022. PMID: 34324029. DOI: 10.1007/s00404-021-06149-w
    OpenUrlCrossRefPubMed
  141. ↵
    1. Liu C,
    2. Xing H,
    3. Luo X and
    4. Wang Y
    : MicroRNA-342 targets Cofilin 1 to suppress the growth, migration and invasion of human breast cancer cells. Arch Biochem Biophys 687: 108385, 2020. PMID: 32335050. DOI: 10.1016/j.abb.2020.108385
    OpenUrlCrossRefPubMed
  142. ↵
    1. Yamaguchi H and
    2. Condeelis J
    : Regulation of the actin cytoskeleton in cancer cell migration and invasion. Biochim Biophys Acta 1773(5): 642-652, 2007. PMID: 16926057. DOI: 10.1016/j.bbamcr.2006.07.001
    OpenUrlCrossRefPubMed
  143. ↵
    1. Huang X,
    2. Pan Q,
    3. Sun D,
    4. Chen W,
    5. Shen A,
    6. Huang M,
    7. Ding J and
    8. Geng M
    : O-GlcNAcylation of cofilin promotes breast cancer cell invasion. J Biol Chem 288(51): 36418-36425, 2013. PMID: 24214978. DOI: 10.1074/jbc.M113.495713
    OpenUrlAbstract/FREE Full Text
  144. ↵
    1. Wang W,
    2. Eddy R and
    3. Condeelis J
    : The cofilin pathway in breast cancer invasion and metastasis. Nat Rev Cancer 7(6): 429-440, 2007. PMID: 17522712. DOI: 10.1038/nrc2148
    OpenUrlCrossRefPubMed
  145. ↵
    1. Liu T,
    2. Ye P,
    3. Ye Y,
    4. Lu S and
    5. Han B
    : Circular RNA hsa_circRNA_002178 silencing retards breast cancer progression via microRNA-328-3p-mediated inhibition of COL1A1. J Cell Mol Med 24(3): 2189-2201, 2020. PMID: 31957232. DOI: 10.1111/jcmm.14875
    OpenUrlCrossRefPubMed
  146. ↵
    1. Wang Y,
    2. Xu H,
    3. Zhu B,
    4. Qiu Z and
    5. Lin Z
    : Systematic identification of the key candidate genes in breast cancer stroma. Cell Mol Biol Lett 23: 44, 2018. PMID: 30237810. DOI: 10.1186/s11658-018-0110-4
    OpenUrlCrossRefPubMed
  147. ↵
    1. Insua-Rodríguez J and
    2. Oskarsson T
    : The extracellular matrix in breast cancer. Adv Drug Deliv Rev 97: 41-55, 2016. PMID: 26743193. DOI: 10.1016/j.addr.2015.12.017
    OpenUrlCrossRefPubMed
  148. ↵
    1. Liu J,
    2. Shen JX,
    3. Wu HT,
    4. Li XL,
    5. Wen XF,
    6. Du CW and
    7. Zhang GJ
    : Collagen 1A1 (COL1A1) promotes metastasis of breast cancer and is a potential therapeutic target. Discov Med 25(139): 211-223, 2018. PMID: 29906404.
    OpenUrlPubMed
  149. ↵
    1. Huang FJ,
    2. Dang JQ,
    3. Zhang S and
    4. Cheng ZY
    : Circular RNA hsa_circ_0008039 promotes proliferation, migration and invasion of breast cancer cells through upregulating CBX4 via sponging miR-515-5p. Eur Rev Med Pharmacol Sci 24(4): 1887-1898, 2020. PMID: 32141558. DOI: 10.26355/eurrev_202002_20367
    OpenUrlCrossRefPubMed
  150. ↵
    1. Liang YK,
    2. Lin HY,
    3. Chen CF and
    4. Zeng D
    : Prognostic values of distinct CBX family members in breast cancer. Oncotarget 8(54): 92375-92387, 2017. PMID: 29190923. DOI: 10.18632/oncotarget.21325
    OpenUrlCrossRefPubMed
  151. ↵
    1. van Wijnen AJ,
    2. Bagheri L,
    3. Badreldin AA,
    4. Larson AN,
    5. Dudakovic A,
    6. Thaler R,
    7. Paradise CR and
    8. Wu Z
    : Biological functions of chromobox (CBX) proteins in stem cell self-renewal, lineage-commitment, cancer and development. Bone 143: 115659, 2021. PMID: 32979540. DOI: 10.1016/j.bone.2020.115659
    OpenUrlCrossRefPubMed
  152. ↵
    1. Zeng JS,
    2. Zhang ZD,
    3. Pei L,
    4. Bai ZZ,
    5. Yang Y,
    6. Yang H and
    7. Tian QH
    : CBX4 exhibits oncogenic activities in breast cancer via Notch1 signaling. Int J Biochem Cell Biol 95: 1-8, 2018. PMID: 29229426. DOI: 10.1016/j.biocel.2017.12.006
    OpenUrlCrossRefPubMed
  153. ↵
    1. Meng R,
    2. Fang J,
    3. Yu Y,
    4. Hou LK,
    5. Chi JR,
    6. Chen AX,
    7. Zhao Y and
    8. Cao XC
    : miR-129-5p suppresses breast cancer proliferation by targeting CBX4. Neoplasma 65(4): 572-578, 2018. PMID: 29940764. DOI: 10.4149/neo_2018_170814N530
    OpenUrlCrossRefPubMed
  154. ↵
    1. Ren S,
    2. Liu J,
    3. Feng Y,
    4. Li Z,
    5. He L,
    6. Li L,
    7. Cao X,
    8. Wang Z and
    9. Zhang Y
    : Knockdown of circDENND4C inhibits glycolysis, migration and invasion by up-regulating miR-200b/c in breast cancer under hypoxia. J Exp Clin Cancer Res 38(1): 388, 2019. PMID: 31488193. DOI: 10.1186/s13046-019-1398-2
    OpenUrlCrossRefPubMed
  155. ↵
    1. Pavlova NN and
    2. Thompson CB
    : The emerging hallmarks of cancer metabolism. Cell Metab 23(1): 27-47, 2016. PMID: 26771115. DOI: 10.1016/j.cmet.2015.12.006
    OpenUrlCrossRefPubMed
  156. ↵
    1. Abdel-Wahab AF,
    2. Mahmoud W and
    3. Al-Harizy RM
    : Targeting glucose metabolism to suppress cancer progression: prospective of anti-glycolytic cancer therapy. Pharmacol Res 150: 104511, 2019. PMID: 31678210. DOI: 10.1016/j.phrs.2019.104511
    OpenUrlCrossRefPubMed
  157. ↵
    1. Tang J,
    2. Luo Y and
    3. Wu G
    : A glycolysis-related gene expression signature in predicting recurrence of breast cancer. Aging (Albany NY) 12(24): 24983-24994, 2020. PMID: 33201835. DOI: 10.18632/aging.103806
    OpenUrlCrossRefPubMed
  158. ↵
    1. Meganck RM,
    2. Borchardt EK,
    3. Castellanos Rivera RM,
    4. Scalabrino ML,
    5. Wilusz JE,
    6. Marzluff WF and
    7. Asokan A
    : Tissue-dependent expression and translation of circular RNAs with recombinant AAV vectors in vivo. Mol Ther Nucleic Acids 13: 89-98, 2018. PMID: 30245471. DOI: 10.1016/j.omtn.2018.08.008
    OpenUrlCrossRefPubMed
  159. ↵
    1. Lei B,
    2. Tian Z,
    3. Fan W and
    4. Ni B
    : Circular RNA: a novel biomarker and therapeutic target for human cancers. Int J Med Sci 16(2): 292-301, 2019. PMID: 30745810. DOI: 10.7150/ijms.28047
    OpenUrlCrossRefPubMed
  160. ↵
    1. Crooke ST,
    2. Baker BF,
    3. Crooke RM and
    4. Liang XH
    : Antisense technology: an overview and prospectus. Nat Rev Drug Discov 20(6): 427-453, 2021. PMID: 33762737. DOI: 10.1038/s41573-021-00162-z
    OpenUrlCrossRefPubMed
  161. ↵
    1. Frieden M and
    2. Ørum H
    : Locked nucleic acid holds promise in the treatment of cancer. Curr Pharm Des 14(11): 1138-1142, 2008. PMID: 18473860. DOI: 10.2174/138161208784246234
    OpenUrlCrossRefPubMed
  162. ↵
    1. Rao DD,
    2. Vorhies JS,
    3. Senzer N and
    4. Nemunaitis J
    : siRNA vs. shRNA: similarities and differences. Adv Drug Deliv Rev 61(9): 746-759, 2009. PMID: 19389436. DOI: 10.1016/j.addr.2009.04.004
    OpenUrlCrossRefPubMed
  163. ↵
    1. Rossi JJ
    : Expression strategies for short hairpin RNA interference triggers. Hum Gene Ther 19(4): 313-317, 2008. PMID: 18363506. DOI: 10.1089/hum.2008.026
    OpenUrlCrossRefPubMed
  164. ↵
    1. Li S,
    2. Li X,
    3. Xue W,
    4. Zhang L,
    5. Yang LZ,
    6. Cao SM,
    7. Lei YN,
    8. Liu CX,
    9. Guo SK,
    10. Shan L,
    11. Wu M,
    12. Tao X,
    13. Zhang JL,
    14. Gao X,
    15. Zhang J,
    16. Wei J,
    17. Li J,
    18. Yang L and
    19. Chen LL
    : Screening for functional circular RNAs using the CRISPR-Cas13 system. Nat Methods 18(1): 51-59, 2021. PMID: 33288960. DOI: 10.1038/s41592-020-01011-4
    OpenUrlCrossRefPubMed
  165. ↵
    1. Nikam RR and
    2. Gore KR
    : Journey of siRNA: Clinical developments and targeted delivery. Nucleic Acid Ther 28(4): 209-224, 2018. PMID: 29584585. DOI: 10.1089/nat.2017.0715
    OpenUrlCrossRefPubMed
  166. ↵
    1. Rivera S and
    2. Yuan F
    : Critical issues in delivery of RNAi therapeutics in vivo. Curr Pharm Biotechnol 13(7): 1279-1291, 2012. PMID: 22201583. DOI: 10.2174/138920112800624300
    OpenUrlCrossRefPubMed
  167. ↵
    1. Gavrilov K and
    2. Saltzman WM
    : Therapeutic siRNA: principles, challenges, and strategies. Yale J Biol Med 85(2): 187-200, 2012. PMID: 22737048.
    OpenUrlPubMed
  168. ↵
    1. Subhan MA and
    2. Torchilin VP
    : siRNA based drug design, quality, delivery and clinical translation. Nanomedicine 29: 102239, 2020. PMID: 32544449. DOI: 10.1016/j.nano.2020.102239
    OpenUrlCrossRefPubMed
  169. ↵
    1. Blanco E and
    2. Ferrari M
    : Emerging nanotherapeutic strategies in breast cancer. Breast 23(1): 10-18, 2014. PMID: 24215984. DOI: 10.1016/j.breast.2013.10.006
    OpenUrlCrossRefPubMed
    1. Charbe NB,
    2. Amnerkar ND,
    3. Ramesh B,
    4. Tambuwala MM,
    5. Bakshi HA,
    6. Aljabali AAA,
    7. Khadse SC,
    8. Satheeshkumar R,
    9. Satija S,
    10. Metha M,
    11. Chellappan DK,
    12. Shrivastava G,
    13. Gupta G,
    14. Negi P,
    15. Dua K and
    16. Zacconi FC
    : Small interfering RNA for cancer treatment: overcoming hurdles in delivery. Acta Pharm Sin B 10(11): 2075-2109, 2020. PMID: 33304780. DOI: 10.1016/j.apsb.2020.10.005
    OpenUrlCrossRefPubMed
    1. Mainini F and
    2. Eccles MR
    : Lipid and polymer-based nanoparticle siRNA delivery systems for cancer therapy. Molecules 25(11): 2692, 2020. PMID: 32532030. DOI: 10.3390/molecules25112692
    OpenUrlCrossRefPubMed
  170. ↵
    1. Fucikova J,
    2. Kepp O,
    3. Kasikova L,
    4. Petroni G,
    5. Yamazaki T,
    6. Liu P,
    7. Zhao L,
    8. Spisek R,
    9. Kroemer G and
    10. Galluzzi L
    : Detection of immunogenic cell death and its relevance for cancer therapy. Cell Death Dis 11(11): 1013, 2020. PMID: 33243969. DOI: 10.1038/s41419-020-03221-2
    OpenUrlCrossRefPubMed
  171. ↵
    1. Musumeci D,
    2. Roviello GN and
    3. Montesarchio D
    : An overview on HMGB1 inhibitors as potential therapeutic agents in HMGB1-related pathologies. Pharmacol Ther 141(3): 347-357, 2014. PMID: 24220159. DOI: 10.1016/j.pharmthera.2013.11.001
    OpenUrlCrossRefPubMed
PreviousNext
Back to top

In this issue

Cancer Genomics - Proteomics: 20 (3)
Cancer Genomics & Proteomics
Vol. 20, Issue 3
May-June 2023
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
  • Back Matter (PDF)
  • Ed Board (PDF)
  • Front Matter (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word on Cancer Genomics & Proteomics.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Breast Cancer: Circular RNAs Mediating Efficacy in Preclinical In Vivo Models
(Your Name) has sent you a message from Cancer Genomics & Proteomics
(Your Name) thought you would like to see the Cancer Genomics & Proteomics web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
11 + 0 =
Solve this simple math problem and enter the result. E.g. for 1+3, enter 4.
Citation Tools
Breast Cancer: Circular RNAs Mediating Efficacy in Preclinical In Vivo Models
ULRICH H. WEIDLE, HUNG-EN HSIA, ULRICH BRINKMANN
Cancer Genomics & Proteomics May 2023, 20 (3) 222-238; DOI: 10.21873/cgp.20377

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Reprints and Permissions
Share
Breast Cancer: Circular RNAs Mediating Efficacy in Preclinical In Vivo Models
ULRICH H. WEIDLE, HUNG-EN HSIA, ULRICH BRINKMANN
Cancer Genomics & Proteomics May 2023, 20 (3) 222-238; DOI: 10.21873/cgp.20377
Reddit logo Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Circular RNA
    • CircRNAs Down-regulated in BC
    • CircRNAs Up-regulated in BC
    • Approaches and Challenges of Targeting CircRNAs
    • Conclusion
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Cited By...

  • No citing articles found.
  • Google Scholar

More in this TOC Section

  • Biospecimen Digital Twins: Moving from a “High Quality” to a “Fit-for-Purpose” Concept in the Era of Omics Sciences
  • Triple-negative Breast Cancer: Identification of circRNAs With Efficacy in Preclinical In Vivo Models
Show more Review

Similar Articles

Keywords

  • Bone metastasis
  • chemoresistance
  • signaling
  • circRNA
  • si-RNA
  • sh-RNA
  • review
Cancer & Genome Proteomics

© 2023 Cancer Genomics & Proteomics

Powered by HighWire