Skip to main content

Main menu

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • 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
    • 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
Research ArticleReview
Open Access

Circular RNAs With Efficacy in Preclinical In Vitro and In Vivo Models of Esophageal Squamous Cell Carcinoma

ULRICH H. WEIDLE, TATJANA SELA, ULRICH BRINKMANN and JENS NIEWOEHNER
Cancer Genomics & Proteomics May 2022, 19 (3) 283-298; DOI: https://doi.org/10.21873/cgp.20320
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 ulrich.brinkmann@roche.com
TATJANA SELA
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: weidle49@t-online.de ulrich.brinkmann@roche.com
JENS NIEWOEHNER
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
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Esophageal cancer is associated with a dismal prognosis. The armamentarium of approved drugs is focused on chemotherapy with modest therapeutic benefit. Recently, checkpoint inhibitory monoclonal antibody Pembrolizumab was approved. In order to identify new targets and modalities for the treatment of esophagus squamous cell carcinoma (ESCC) we searched the literature for circRNAs involved in the pathogenesis of ESCC. We identified two down-regulated and 17 up-regulated circRNAs as well as a synthetic circRNA with efficacy in preclinical in vivo systems. Down-regulated circRNAs sponge microRNAs directed against tumor suppressor genes. Up-regulated circRNAs sponge microRNAs directed against mRNAs, which encode proteins with pro-tumoral functions. We discuss issues such as reconstitution of down-regulated circRNAs and inhibition of up-regulated circRNAs with short interfering RNA (siRNA)- related entities. Also, we address druggability issues of the identified targets.

Key Words:
  • microRNA sponges
  • short interfering RNA (siRNA)
  • synthetic circ RNA
  • target validation
  • xenografts
  • review

Esophageal carcinoma is the seventh most common cancer worldwide with 570,000 cases annually (1). Two major histological sub-types are esophageal squamous cell carcinoma (ESCC) and esophageal adenocarcinoma (EAC). ESCCs arise in squamous cells that line the esophagus, whereas EACs derive from glandular cells of the esophagus (2). The ratio of incidence between ESCC and EAC depends on the geographic location with ESCC emerging as the most frequent subtype worldwide (3). Therapies such as endoscopic resection, surgery, chemo- and radio-therapy and treatment with immune-checkpoint inhibitory monoclonal antibodies (mAbs) only confer minimal survival advantages (4-7). Among the agents under development are epidermal growth factor receptor (EGFR), human epidermal growth factor receptor 2 (HER2), vascular endothelial growth factor receptor (VEGFR), hepatocyte growth factor (HGF)-tyrosine protein kinase c-MET and fibroblast growth factor receptor 2 (FGFR2) signaling inhibitors in the format of mAbs or small molecule tyrosine kinase inhibitors (4) as well as small molecule inhibitors of mechanistic target of rapamycin (mTOR), poly(ADP)ribose-polymerase 1 (PARP1) and cyclin-dependent kinases (CDKs) (8-10), most of them affecting rat sarcoma (RAS), rapidly accelerated firbrosarcoma (RAF), extracellular signal-regulated kinase (ERK), mitogen-activated protein kinase(MAPK), phosphoinosite-3-kinase (PI3K), serine-threonine kinase AKT and mTOR pathways.

Despite these ongoing efforts, identification of new targets and treatment modalities for ESCC is an issue of high priority. In this review, we focus on circRNAs with documented efficacy in preclinical efficacy in in vivo models for treatment of ESCC.

Circular RNA

Several types of non-coding RNAs such as t-RNA, r-RNA, siRNA, lncRNA and circRNA have been described (11). CircRNAs are covalently closed circles of RNA and are products of backsplicing events in which one exon is spliced to a preceding exon (12). A unique junction is generated which allows their specific detection by molecular probes (12). At least 30000 circRNAs have been identified by deep sequencing (12). circRNAs are expressed in all cells and tissues and are deregulated in cancer (13). Only a few of them are expressed at significant abundance (13). They are transcribed by RNA polymerase II (RNA pol II) and have no 5’-caps and polyA extensions at the 3’-end (14). In cancer they can be expressed in tumor cells, in cells of the tumor-microenvironment, immune-related cells such as T- and natural killer (NK) cells, fibroblasts and endothelial cells thus regulating tumor growth, angiogenesis and anti-tumoral immune responses (15) (Figure 1). Also, they can be incorporated into exosomes and transferred into tumor cells and cells of the tumor microenvironment (16) (Figure 1). Most circRNAs are located in the cytoplasm, and a minority is located in the nucleus. CircRNAs can function as sponges and decoys for microRNAs (miRs), as binders of RNA transcripts, mediators of protein scaffolding (17) and recently it was found that they can represent short open reading frames encoding small proteins (Figure 1) (17). circRNAs located in the nucleus can associate with RNA pol II and modulate transcription (Figure 1) (17). In this review we focus on their role as sponges for miRs. Depending on the type of miRs they are interacting with, and their decreased or increased status of deregulation they can exert an oncogenic or tumor-suppressive effect (Figure 2) (16, 17). Due to their stability and presence in blood and saliva, circRNAs are potential biomarkers for cancer (18).

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

Roles of circRNAs in cancer. circRNAs can be found intracellulary in the nucleus and the cytoplasm and extracellulary in exosomes. In the nucleus, they can regulate splicing and transcription, in the cytoplasm they are involved in RNA binding, sponging of miRs, protein scaffolding, synthesis of new proteins, proliferation, apoptosis, EMT, remodeling of the ECM and drug resistance. circRNAs can be localized in exosomes in blood and saliva as biomarkers for detection and progression of cancer. CircRNAs can be transferred to cells of the TME such es endothelial cells and NK cells and regulate angiogenesis and cytotoxicity. Figures were created via BioRender.com. CircRNAs are shown as coloured circles. EC: Endothelial cell; ECM: extracellular matrix; EMT: epithelial mesenchymal transition; miR: microRNA; NK: natural killer cell; TC: tumor cell.

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

circRNAs can modulate the activity of oncogenes and tumor suppressor genes. A) Up-regulated circRNAs can sponge miRs targeting oncogenic mRNAs and thus up-regulate oncogenes. B) Down-regulated miRs targeting mRNAs for tumor suppressors lead to increased miRs targeting tumors suppressors and decrease of the corresponding proteins. TS: Tumor suppressive; ONC: oncogenic. Figures were created with Biorender.com

circRNAs were first identified as parasitic viroids in plants (19) and later in additional eukaryotic cells by electron microscopy (20). It has also been shown that circRNAs have an impact on brain function in mice (21).

Functional roles of circRNA in ESCC and their emerging role as cancer biomarkers are described in several reviews (22-25). In this review we focus on circRNAs with efficacy in ESCC-related preclinical in vivo systems. We also describe a synthetic circRNA which mediates an anti-tumoral effect. Taken together, circRNAs can affect hallmarks of cancer such as proliferation, apoptosis, epithelial mesenchymal transition (EMT), invasion, migration, metastasis, angiogenesis, anti-tumoral immune response and drug resistance.

Down-regulated circRNAs

Down-regulation of circRNAs sponging miRs can result in excess of corresponding miRs which degrade tumor suppressors.

circ-FOXO3 targets phosphatase and tensin homolog on chromosome 10. Circ-FOXO3 (Figure 3) is encoded by the human FOXO3 gene (26, 27). It was found to be poorly expressed in ESCC cell lines and tissues and inhibited growth, migration and invasion of ESCC cells in vitro and in vivo (28). circ-FOXO3 acted as a sponge for miR-23a which targets phosphatase and tensin homolog on chromosome 10 (PTEN) (28), a gatekeeper of the phosphoinosite-3-kinase (PI-3K) pathway (29).

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

circRNAs sponging miRs targeting tumor suppressors and transcription factors with efficacy in preclinical in vivo models of oesophageal squamous cell carcinoma. Down- or up-ward arrows: Down- or up-regulated. A) circRNAs targeting tumor suppressors. circ FOXO3 and circ CNTAP3 are down-regulated. Sponged miRs and targeted proteins are indicated. B) circRNAs sponging microRNAs targeting mRNAs for transcription factors circ 6948, circ LRP6 and CiRS-7 are up-regulated and corresponding microRNAs and targets are shown. HMGA2: High mobility group A2; HOXB13: homeobox protein B13; MYC: transcription factor MYC; PTEN: tensin homolog on chromosome 10. Figures were created with Biorender.com

circ-CNTNAP3 targets RNA-binding motif protein 25. circ-CNTNAP3 (Figure 3) derives from exons 5 to 9 of the CNTNAP3 gene and was identified by screening for circRNA expression in ESCC (30). It was poorly expressed in ESCC cell lines and tissues (30). Knock-down promotes cell proliferation in wild-type p53 Eca-109 ESCC cells, but not in p53 mutant ESCC cells such as KYSE-450 and TE-1 cells (30). In vivo, over-expression of circ-CNTNAP3 suppressed growth of Eca-109 cells by acting as a sponge for miR-153-5p which targets p53 (30). miR-153-5p reversed the TS role of circ-CNTNAP3 in p53 wild-type ESCCs (30). RNA-binding motif protein 25 (RBM25) was found to be a direct transcriptional target of p53 (30, 31). A positive feedback expression loop was identified based on over-expression of p53 and increased expression of circ-CNTNAP3 (30). Low expression of circ-CNTNAP3 indicated poor prognosis in p53 wild-type ESCC patients (30). p53 acts as a TS and its loss is a driver of metastasis (32, 33). Gene transfer of p53 into ESCC tumors has been pursued as a modality for treatment of ESCC (34).

Up-regulated circ-RNAs. Up-regulation of circRNAs can lead to down-regulation of miRs which inhibit proteins with oncogenic functions such as transcription factors, mediators of cellular signaling, components of the extracellular matrix and further protumoral functionalities.

CircRNAs Sponging miRs Directed Against Transcription Factors

Circ-0006948 targets high mobility group A2. Circ-0006948 (Figure 3) originates from exons 2, 3 and 4 of the fibronectin type III domain containing 3B (FND3B) gene (35). Circ-0006948 was shown to be up-regulated in ESCC and was related to poor survival (35). It promoted proliferation, invasion, migration and epithelial mesenchymal transition in TE-1 and KYSE30 cells (35). In vivo, circ-0006948 mediated tumor growth of TE-1 cells after subcutaneous injection into nude mice. It functioned as a sponge for miR-490-3p and mediated up-regulation of its target high mobility group A2 (HMGA2) (35). The latter acts as a transcription factor by binding to AT-rich regions of DNA and functions as a critical regulator in cancer development (36). HMGA2 is over-expressed in several types of cancer and its over-expression can induce tumorigenesis (37-39). HMGA2 promotes cell-cycle entry, inhibits apoptosis, promotes EMT, supports cancer stem cell (CSC) phenotype and chemo-resistance and activates MAPK/ERK, transforming growth factor β (TGFβ)/SMAD), PI3K/AKT/mTOR, nuclear factor ĸB (NF-ĸB) and signal transducer and activator of transcription 3 (STAT3) pathways (37-39). Several small molecule inhibitors interfering with HMGA2/DNA interaction have been identified (40). HMGA2 over-expression plays a critical role in the progression of ESCC (41, 42).

circ-LRP6 targets the transcription factor MYC. circ-LRP6 (Figure 3) derives from the low-density lipoprotein receptor-related protein 6 (LRP6) gene (43, 44). It was found to be up-regulated in ESCC tissues and was mainly located in the cytoplasm (44). Knockdown of circ-LRP6 inhibited proliferation and invasion of TE-1 and EC109 ESCC cells in vitro and decreased TG of EC109 ESCC cells in vivo (44). Circ-LRP6 acted as a sponge for miR-182, which targets transcription factor MYC (44). MYC dimerizes with MYC-associated factor X (MAX) to exert its oncogenic functions such as proliferation, invasion, metastasis and angiogenesis (45-48). Inhibition of MYC transcription, disruption of MYC/MAX dimers, inhibition of post-translational regulation and proteolytic degradation of MYC are drug discovery approaches in preclinical and clinical stages of development (49, 50).

CiR-S7 targets homeobox protein B31. Up-regulation of CiR-S7 (Figure 3) and down-regulation of miR-7 have been shown to predict poor prognosis of ESCC patients (51). Over-expression of CiR-S7 promotes proliferation and invasion of Eca109 and KYSE150 ESCC cells by sponging miR-7 (51). Homeobox protein B31 (HOXB13) has been identified as a target of miR-7 (51). In vivo, CiR-S7 increased lung metastasis of Eca109 cells after tail vein injection (51). HOXB13 activates NFĸB signaling by phosphorylation of NFĸB/p65 (51). Forced expression of HOXB13 confers oncogenic hallmarks to esophageal keratinocytes (52), promotes cancer cell growth and predicts poor survival in ESCC patients (53, 54). HOX genes are master regulatory transcription factors during embryogenesis and HOXB13 is commonly altered in human cancers (55).

circRNAs Sponging miRs Directed Against Signaling Components

circ-0000654 targets interleukin 6. High circ-0000654 (Figure 4) is correlated with higher T stage and lymph node metastasis in ESCC patients (56). circ-0000654 enhanced proliferation, migration and invasion of KYSE450 ESCCs in vitro (56). In vivo, circ-0000654 promoted TG after subcutaneous implantation and lung metastasis after tail vein injection in nude mice (56). The effects are mediated by sponging of miR-149-5p which targets interleukin 6 (IL6) (56). IL6 is a tumor promoting cytokine, which binds to IL6 receptor (IL6R) and activates intracellular signals such as STAT3 phosphorylation via glycoprotein 130 (gp130). STAT3 is transferred to the nucleus and activates genes such as cyclin B1 and B-cell lymphoma extra-large (Bcl-xL), MYC, induced myeloid leukemia cell differentiation protein (MCL1) and vascular endothelial growth factor (VEGF) (57, 58). IL6 signaling is aberrantly hyperactivated in many types of cancer (59). IL6 is linked to the progression of ESCC and its inhibition is a potential strategy for treatment of this disease (60).

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

circRNAs sponging miRs affecting mRNAs for components of signaling pathways and the extracellular matrix with efficacy in preclinical in vivo models of esophageal squamous cell carcinoma. Down- or up-ward arrows: Down- or up-regulated. CDC25A: Cell-division cycle 25A; COL1A1: collagen1A1; FNDC3B: fibronectin type III domain containing protein 3B; IL6: interleukin 6; MET: transmembrane tyrosine kinase MET; RAB10: ras-related protein 10; RHOC: Ras homology member, member C; WNT3: proto-oncogene protein WNT3. Figures were created with Biorender.com.

circ-0072088 targets vascular endothelial growth factor A. circ-0072088 (Figure 4) is increased in ESCC tissues and cell lines (61). It derives from exons 13 to 17 of the ZFR gene, which encodes a zinc finger transcription factor (62). It promoted proliferation, migration and invasion in TE-13 and ECA109 ESCCs in vitro (61). In vivo, circ-0072088 promoted TG in nude mice after subcutaneous implantation (61). This leads to up-regulation of VEGF by sponging of miR-377 (61). Bevacizumab, a mAb directed against VEGF-A and Ramicurumab, a mAb directed against VEGFR2, are under clinical investigation in ESCC (63, 64), as well as several small molecule inhibitors of the VEGFR tyrosine kinases such as sunitinib, sorafenib, apatinib and anlotinib (65).

circ-LPAR3 targets transmembrane receptor kinase MET. circ-LPAR3 (Figure 4), which derives from the lysophophosphatic acid receptor 3 gene, was increased in human ESCC tissues and cell lines (66). It promoted migration and invasion of KYSE-450 ESCC cells, but had no impact on proliferation (66). In vivo, in TE-13 ESCC xenografted cells, circ-LPAR3 functioned as a mediator of lung metastases after tail vein injection into nude mice (66). circ-LPAR3 mediated activation of MET by sponging miR-198 which targets MET. Together with its ligand HGF, MET is an essential driver of oncogenesis and metastasis in many types of tumors (67). MET can activate many oncogenic pathways such as PI3K/AKT/mTOR, MAPK/ERK1/2 and cell division control protein 42 homolog (CDC42)/ras-related C3 botulinum toxin substrate1 (RAC1) (68). Furthermore, MET can be activated by gene amplification, mutations, gene fusion or exon 14 skipping (68, 69). MET is over-expressed preferentially in EAC, however, MET expression in ESCC has to be studied in more detail (70). Therefore, mAbs directed against HGF (Rilotuzumab, AMG102) and MET (Obinutuzumab) are in clinical development in patients with EAC or tumors of the gastroesophageal junction (71). AMG 377, a highly selective small molecule, which inhibits HGF/MET signaling, is in clinical trials in patients with several types of cancer (72).

circ-100367 targets proto-oncogene WNT3. circ-100367 (Figure 4) was found to be highly expressed in radio-resistant ESCC cell lines such as KYSE-150R and its silencing inhibited proliferation, invasion and migration of this cell line (73). Silencing of circ-100367 reduced the growth of KYSE-150R cells under radiation in nude mice (73). circ-100367 interacted with miR-217, which targets proto-oncogene protein WNT3 (73). The latter promotes stabilization of β-catenin to regulate radio-sensitivity (73, 74). WNTs are secreted lipid modified glycoproteins with 16 members in humans (75). They interact with the frizzled family of receptors and co-receptors such as lipoprotein-related protein 5 and 6 (LRP5,6) and their signaling activity is highly context-dependent (76).

circ-AKT3 targets ras homolog family, member C and transcription factor STAT3. circ-AKT3 (Figure 4) derives from the AKT3 gene and was identified in gastric cancer cells with cis-Pt resistance (77). Circ-AKT3 was found to be over-expressed in ESCC and induced proliferation, migration and invasion of TE-1 ESCCs (78). circ-AKT3 knockdown suppressed TG of TE-1 ESCCs in nude mice (78). miR-17-5p was sponged by circ-AKT3 resulting in expression of ras homolog family, member C (RHOC) and STAT3 (78). RHOC is a member of the protumoral ras homolog family and acts as a cytoskeleton organizer (79, 80). RHOC is related to lymph node metastasis and survival in ESCC patients (81). The role of transcription factor STAT3 in tumor formation, metastasis and drug resistance has been discussed previously in this review (57, 58). Recently, STAT3 has been identified as a mediator of immune suppression in tumors (82).

circ-UBAP2 targets ras-related protein RAB10. circ-UBAP2 (Figure 4) derives from ubiquitin-associated protein 2 and is up-regulated in ESCC tissues (83). Knockdown of circ-UBAP2 with lentivirus small hairpin RNA (shRNA) mediated inhibition of proliferation, invasion and migration of ESCC cells in vitro (83). Vice versa, circ-UBAP2 induced proliferation, migration and invasion of ESCCs (83). In vivo, down-regulation of circ-UBAP2 suppressed TG of ESCCs in nude mice (83). Circ-UBAP2 sponged miR-422a, which targets ras-related protein RAB10 (83). circ-UBAP2 promoted tumorigenesis in osteosarcoma, lung cancer, triple-negative breast cancer and cervical cancer (84-87). RAB10, a member of the superfamily of small GTPases, can regulate intracellular vesicle trafficking and its over-expression promotes TG of hepatocellular carcinoma (HCC) and correlates with poor prognosis (88).

Circ-0004771 targets cell-division cycle 25A. Circ-0004771 (Figure 4) was found to be up-regulated in ESCC tissues and its expression correlated with poor prognosis (89). Knockdown of circ-0004771 inhibited proliferation of ESCC cells in vitro and in vivo (89). It acted as a sponge for miR-339-5p, which targets cell-division cycle 25A (CDC25A) (89). CDC25A is a dual-specific phosphatase, which hydrolyses tyr- and ser-phosphates by dephosphorylation (90). CDC25A is over-expressed in many types of tumors and activates the cyclin-dependent kinase complex which promotes transition of G1/S and G2/M phase of the cell-cycle and plays a role in the DNA damage response (91, 92). In ESCC, targeting of CDC25A mediates radio-sensitivity (93).

CircRNAs Sponging miRs Directed Against Components of The Extracellular Matrix

Circ-0004370 targets collagen 1A1. Expression of circ-0004370 (Figure 4) was found to be increased in ESCC tissues in comparison to normal controls (94). In OE19 and EC109 ESCCs, knockdown of circ-0004370 suppressed proliferation, invasion and migration in vitro and in vivo after subcutaneous implantation into nude mice (94). Circ-0004370 sponged mir-1310-3p, which targets collagen 1A1 (Col1A1) (94). Collagen, an abundant component of the ECM, plays an important role in EMT and TG in gastric cancer (95). COL1A1 knockdown suppresses metastasis of breast cancer (96). COL1A1 was found to be up-regulated in ESCC compared to normal controls (97) and its expression is associated with poor prognosis (98).

Circ-0005948 targets fibronectin type III domain-containing protein 3B. circ-0005948 (Figure 4) was up-regulated in ESCC tissues and cells (99). Down-regulation of expression of circ-0005948 repressed growth, migration and invasion and EMT in vitro and TG in vivo of ESCCs (99). circ-0005948 sponged miR-4262, which targets fibronectin type III domain-containing protein 3B (FNDC3B) (99). FNDC3B, a protein of the extracellular matrix (ECM), also known as factor for adipocyte differentiation 104, is a member of the fibronectin family and contains nine fibronectin III domains and one transmembrane domain (100). It can be amplified in HCC, act as an oncogenic driver and promote EMT, PI3K/AKT, retinoblastoma (Rb) and TGFβ signaling (100). Over-expression of FNDC3B predicts poor prognosis in lung adeno-carcinoma (101).

Circ RNAs Sponging miRs Targeting Additional Protein Categories

Circ-0003340 targets protein for Xklp2. circ-0003340 (Figure 5) was shown to be over-expressed in ESCC in comparison to adjacent normal tissues and non-transformed human esophagus squamous cells Het-1a (102). Knockdown of circ-0003340 induced apoptosis and inhibited proliferation in EC1 and EC9706 ESCCs in vitro and TG in vivo (102). circ-0003340 has a binding site for mir-562, which inhibits targeting protein for Xklp2 (TPX2) (102). TPX2 acts as a spindle assembly factor, mediates growth during M phase as well as cell invasion and metastasis (103, 104). TPX2 recruits and activates ser-thr kinase aurora A, which is a validated, druggable target for cancer therapy and corresponding inhibitors are evaluated in several clinical studies in cancer patients (105). TPX2 has been shown to promote growth of EC9706 xenografts in vivo (106). TPX2 may be useful as a potential biomarker for tumor progression, metastasis and prognosis in patients with ESCC (107).

Figure 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 5.

circRNAs sponging miRs affecting mRNAs of diverse other components with efficacy in preclinical in vivo models of esophageal squamous cell carcinoma. Down- or up-ward arrows: Down- or up-regulated. PARP9: Poly(ADP-ribose) polymerase, family member 9; PDCD4: programmed cell death protein 4; PTEN: tensin homolog on chromosome 10; NRIP1: nuclear receptor interacting protein 1; TKTL1: transketolase-like 1; TPX2: targeting protein for Xklp2; TXNRD1: thioredoxin 1. Figures were created with Biorender.com.

Circ-PRKCI targets poly(ADP-ribose) family member 9. circ-PRKCI (Figure 5) and poly(ADP-ribose) family member 9 (PARP9) were up-regulated and miR-186polymerase-5p was down-regulated in ESCC tissues in comparison to adjacent normal tissues (108). circ-PRKCI derives from the protein kinase C iota type (108). Knockdown of circ-PRKCI decreased cell viability, colony formation and elevated radiosensitivity of ESCC in vitro and inhibited TG in vivo (108). Mechanistically, circ-PRKCI sponged miR-186-5p and up-regulated PARP9 (108, 109). PARP9 is over-expressed in breast cancer and promotes cell migration (110). In lymphoma and prostate cancer, PARP9 promotes metastasis and correlates with poor prognosis (111, 112). From a mechanistic point of view, the function of PARP9 in tumor progression is not yet resolved.

Circ-DUSP16 targets dual specificity phosphatase 16. circ-DUSP16 (Figure 5) derives from dual specificity phosphatase 16 and was found to be elevated in ESCC tissues and hypoxia stimulated ESCC cells (113). Knockdown of DUSP16 suppressed hypoxia-induced ESCC viability, colony formation, migration, invasion and glycolysis in vitro and inhibited TG of ESCC cells in vivo (113). DUSP16 positively regulated transketolase like-1 (TKTL1) by sponging miR-497 (113). TKTL1 is a thiamine diphosphate (vitamin1)-dependent enzyme, which catalyses several key reactions in the non-oxidative branch of the pentose phosphate pathway; It has been identified as a marker of the anaerobic glucose metabolism (Warburg effect) and is involved in the generation of NADPH and ribose-5-phosphate (114, 115). It is also involved in cell proliferation, lipid synthesis, ATP generation, inhibition of apoptosis, angiogenesis and lactate-based inhibition of the immune system by T- and NK cells and therapy resistance (114, 115). TKTL1 promotes proliferation and metastasis of ESCC (116, 117). The druggability of TKTL1 and its prospect for cancer treatment are still under evaluation.

circ-12086 targets thioredoxin 1. circ-12086 (Figure 5) was shown to be up-regulated in ESCC tissues, enhanced cell proliferation, cell viability and cell adhesion and inhibited apoptosis in KYSE450 and KYSE510 ESCCs in vitro (118). In vivo, its inhibition led to TG inhibition and attenuation of lung metastasis after subcutaneous implantation in KYSE450 cells (118). circ-12086 sponged miR-1305, which targets thioredoxin 1 (TXNRD1) (118). The latter is a cytosolic selenocysteine containing enzyme, which is over-expressed in many types of cancer (119). It is involved in adaption of cancer cells to stress (120, 121). Irreversible TXNRD1 inhibitors have been identified, which inhibit growth of xenografts and syngeneic mouse tumors without overt cytotoxicity (122).

circ-NTRK2 targets nuclear receptor-interacting protein 2. circ-NTRK2 (Figure 5) derives from the neurotrophin receptor tyrosine kinase 2 gene, and was found to be up-regulated in ESCC patients and its expression correlated with poor prognosis (123). In vitro, knockdown of crc-NTRK2 inhibited proliferation, invasion and EMT and accelerated apoptosis in Eca-109 and KYSE-150 ESCCs (123). In vivo, silencing of NTRK2 led to inhibition of TG of KYSE 150 cells after subcutaneous implantation in nude mice (123). circ-NTRK2 sponged miR-140-3p leading to up-regulation of nuclear receptor-interacting protein 2 (NRIP2), also known as receptor interacting protein 140 (RIP140) (123). It interacts with the hormone-dependent activation function 2 domain (AF2) of nuclear receptors and modulates transcriptional activity of several transcription factors, including estrogen receptor (124). NRIP1 regulates various oncogenic signaling pathways and participates in progression of solid tumors (125). Suppression of NRIP1 inhibits growth of breast cancer cells in vitro and in vivo (126). NRIP1 is also involved in cachexia in mice (127).

circ 21-93 targets PTEN, programmed cell death protein 4 and transforming growth factor receptor β2. Using enzymatic ligation, circ 21-93, a circular multi-miR sponge was generated which is more stable than its linear counterpart (128). It contains two binding sites for miR-21 and three binding sites for miR-93 (128). circ 21-93 decreased SKGT4 and OE33 ESCC transfectants proliferation and migration and increased apoptosis in vitro (128). In vivo, circ 21-93 suppressed TG of xenografts after subcutaneous implantation into nude mice (128). Tumor suppressors PTEN, programmed cell death protein 4 (PDCD4) and transforming growth factor receptor β2 (TGFRβ2) have been identified as targets for miR-21 and -93 (129-132).

Technical Issues

Down-regulated circRNAs. circ-FOXO3 and Circ-CNTNAP3 are down-regulated in ESCC tissues and have been identified as sponges for miRs-23a and -513a-5p, which target PTEN and p53, respectively (28, 30). Therapeutic intervention can be based on reconstitution of the corresponding TS by small molecules or gene transfer. However, both approaches have their limitations with respect to specificity and delivery issues. Proof-of-concept (POC) experiments have shown that TG can be inhibited in vitro and in vivo by transfecting ESCC with the corresponding circRNAs. However, extrapolation of these preclinical findings to clinical studies would require to solve issues such as targeting ESCC cells and optimization of transfection efficacy. The same considerations hold true for transfer of synthetic circ-RNAs acting as sponges for miRs targeting TS mRNAs (128).

Up-regulated circRNAs. POC experiments as outlined in the previous sections have indicated that up-regulated circRNAs can exhibit an oncogenic function by sponging miRs, which inactivate mRNA encoding for tumor-promoting proteins in ESCC tumor cells. circRNAs are generated by a backsplicing event generating specific new junctional sequences; therefore, circRNA can be specifically targeted by short interfering RNA (siRNA) or short hair-pin RNA (shRNA) (133-135). siRNA is chemically synthesized double-stranded RNA, 19-23 base pairs in length with 2nts unpaired at the 5’-phosphorylated and 3’-unphosphorylated ends (136). shRNA are artificial RNA molecules with tight hairpin loops, which also silence target gene expression via RNA interference (137). siRNA and shRNA represent exogenously added RNAs, whereas miRs are synthesized intracellularly. All rely on the endoribonuclease DICER for processing (136). RNAi requires stringent complementarity pairing to the corresponding target, whereas in the case miRs more relaxed specificity of base-pairing has been observed (137). As of January 2020, ten oligonucleotide drugs have received regulatory approval. These drugs are focused on local delivery such as eye, spinal cord or liver (138, 139). For example, Onpattra (Patisiran), an agent approved for treatment of transerythrin-mediated amyloidosis that silences production of an abnormal form of transerythretin, represents a chemically modified anti-transerythrin oligo-nucleotide formulated in liposomes targeted to the liver (139).

However, from a broader perspective, delivery of siRNA into tumor cells is impeded by several factors such as intravascular degradation, renal clearance, recognition by the immune system, tumor tissue penetration, uptake into TC, endosomal release and off-target-effects (140, 141). Delivery of siRNA directly is not recommended due to its short half-life ranging from a couple of minutes to half-an hour (140) and its low targeting and transfection efficacy. Its negative charge due to the anionic backbone prevents it from permeating the plasma membrane and gives rise to toxicity (141). Therefore, chemical modification and development of delivery methods and corresponding vehicles are of paramount importance. Chemical modifications of the backbone, nucleobases, ribose and alternative chemistries, have improved pharmaco-kinetic and pharmaco-dynamic properties of siRNA (142). Conjugation of siRNA with polyethylene glycole and cholesterol have improved serum stability and prevented renal clearance (143, 144). Formulation of siRNA into nanoparticle-based structures has improved half-life of siRNA and its delivery to TC due to facilitated membrane fusion. Lipid nano-particles, composed of lipids play an important role in delivery of si-RNA (145), and prevention of immunogenicity and cytokine-release syndrome. An important step in the development of siRNA delivery is the development of dynamic polyconjugates (146). This technology includes a membrane active polymer, which is masked until it reaches the acid environment of the endosomes and induces burst of their membrane. Targeting of lipid nanoparticles can be achieved by conjugation to antibodies, ligands, aptamers and RGD peptides as shown for transmembrane antigens such as integrins, prostate-specific membrane antigen and folate receptor (147-149). A breakthrough for delivery of siRNA to the liver was the identification of the asialoglycoprotein complex on hepatocytes which is targeted by entities containing N-acetylgalactosamine (GalNAc) and allow targeting of siRNA conjugated with GalNAc to hepatocytes (150, 151). For ESCC, specific ligands, antibodies or transmembrane receptors have not yet been identified. In order to improve cellular uptake of siRNA complexation with viral proteins such as trans-activator of transcriptin (TAT) (152) or penetratin (drosophila) (153), have improved delivery of siRNA through the cell membrane as well as endosomal escape.

Targets

P53, PTEN, PDCD4 and TGFβR2 are TS frequently down-regulated in ESCC (129-132). As already outlined previously, their functional reconstitution by small molecules or gene therapy is problematic from a drug discovery point of view.

Regarding oncogenic circRNAs, delivery of siRNA into TC can inhibit their growth in vitro and in vivo. It is important to mention that in lung adenocarcinoma and gastric patient derived xenografts (PDX), corresponding cholesterol-conjugated siRNAs have mediated in vivo efficacy (154, 155). For ESCC, data derived from PDX-derived models are not yet available.

circRNAs can mediate an oncogenic effect by sponging miRs which inactivate oncogenic proteins. We have identified targets which can be grouped into transcription factors, signaling related entities, components of the ECM and targets, which do not match with these major categories. Inhibitors of the identified targets VEGF and MET, such as small molecule tyrosine kinase inhibitors or antagonizing antibody moieties, are in clinical trials in esophageal cancer patients as outlined in the previous text (63-65, 71, 72). TPX2 activates Aurora kinase A, which is clinically evaluated in several types of cancer (105). WNT3 signaling is context-dependent and deserves further validation in ESCC (73). CDC25A, is difficult to target and alerts toxicity issues due to ubiquitous expression of this enzyme (91, 92). It will be of interest to evaluate the identified TXNRD1 inhibitors in several in vivo models of ESCC as a further path for their clinical evaluation (122). A couple of potential targets such as the transcription factors HMGA2 (37-39), MYC (45-48), HOXB13 (53, 54), STAT3 (57, 58), the GTPases RHOC and RAB10 (79, 80, 88) and NRIP1 are associated with druggability problems (125). However, proteolysis targeting chimeras (PROTACs) are on the horizon. PROTACs mediate degradation of the corresponding targets by the proteasome after their ubiquitinylation (156-158). At least 15 targeted degraders will be in clinical studies until the end of 2021 with targets such as androgen receptor (AR), estrogen receptor (ER), Bcl-xL, the zinc finger transcription factors Helios, Ikaros, Aiolos, GTPase1, glutathion P-transferase 1 and members of the bromodomain family, such as BRD9 (159). Another interesting target identified by our search is TKTL1, which is dependent on vitamin B1 as a cofactor (114, 115). The druggability of this enzyme is unclear, since it has not been shown yet that vitamin B1 antagonists or other inhibitory compounds give rise to in vivo inhibition of ESCC-related xenografts. The identified components of the ECM, COL1A1 and FNDC3B, might be targeted with corresponding antibodies (94, 99). With respect to targeting ESCC, identification of corresponding transmembrane antigens is an important issue. Claudin 18.2 is expressed in some esophageal cancers and members of the FGFR family deserve further investigation (160, 161). Taken together, the field of circRNAs in ESCC is still in its infancy and its translational impact deserves further investigation.

Footnotes

  • Authors’ Contributions

    The Authors contributed equally to all aspects of the article.

  • Conflicts of Interest

    UB, TS, JN are and UHW was an employee of Roche.

  • Received January 27, 2022.
  • Revision received March 2, 2022.
  • Accepted March 3, 2022.
  • Copyright© 2022, 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. Yang J,
    2. Liu X,
    3. Cao S,
    4. Dong X,
    5. Rao S and
    6. Cai K
    : Understanding esophageal cancer: the challenges and opportunities for the next decade. Front Oncol 10: 1727, 2020. PMID: 33014854. DOI: 10.3389/fonc.2020.01727
    OpenUrlCrossRefPubMed
  2. ↵
    1. Smyth EC,
    2. Lagergren J,
    3. Fitzgerald RC,
    4. Lordick F,
    5. Shah MA,
    6. Lagergren P and
    7. Cunningham D
    : Oesophageal cancer. Nat Rev Dis Primers 3: 17048, 2017. PMID: 28748917. DOI: 10.1038/nrdp.2017.48
    OpenUrlCrossRefPubMed
  3. ↵
    1. Abnet CC,
    2. Arnold M and
    3. Wei WQ
    : Epidemiology of esophageal squamous cell carcinoma. Gastroenterology 154(2): 360-373, 2018. PMID: 28823862. DOI: 10.1053/j.gastro.2017.08.023
    OpenUrlCrossRefPubMed
  4. ↵
    1. Yang YM,
    2. Hong P,
    3. Xu WW,
    4. He QY and
    5. Li B
    : Advances in targeted therapy for esophageal cancer. Signal Transduct Target Ther 5(1): 229, 2020. PMID: 33028804. DOI: 10.1038/s41392-020-00323-3
    OpenUrlCrossRefPubMed
    1. Fatehi Hassanabad A,
    2. Chehade R,
    3. Breadner D and
    4. Raphael J
    : Esophageal carcinoma: Towards targeted therapies. Cell Oncol (Dordr) 43(2): 195-209, 2020. PMID: 31848929. DOI: 10.1007/s13402-019-00488-2
    OpenUrlCrossRefPubMed
    1. Vivaldi C,
    2. Catanese S,
    3. Massa V,
    4. Pecora I,
    5. Salani F,
    6. Santi S,
    7. Lencioni M,
    8. Vasile E,
    9. Falcone A and
    10. Fornaro L
    : Immune checkpoint inhibitors in esophageal cancers: are we finally finding the right path in the mist? Int J Mol Sci 21(5): 1658, 2020. PMID: 32121290. DOI: 10.3390/ijms21051658
    OpenUrlCrossRefPubMed
  5. ↵
    1. Peng C and
    2. Cohen DJ
    : Advances in the pharmacotherapeutic management of esophageal squamous cell carcinoma. Expert Opin Pharmacother 22(1): 93-107, 2021. PMID: 33034212. DOI: 10.1080/14656566.2020.1813278
    OpenUrlCrossRefPubMed
  6. ↵
    1. Zhu DS,
    2. Dong JY,
    3. Xu YY,
    4. Zhang XT,
    5. Fu SB and
    6. Liu W
    : Omipalisib inhibits esophageal squamous cell carcinoma growth through inactivation of phosphoinositide 3-Kinase (PI3K)/AKT/Mammalian target of rapamycin (mTOR) and ERK signaling. Med Sci Monit 26: e927106, 2020. PMID: 32804918. DOI: 10.12659/MSM.927106
    OpenUrlCrossRefPubMed
    1. Miyamoto K,
    2. Minegaki T,
    3. Hirano S,
    4. Hayashi I,
    5. Tsujimoto M and
    6. Nishiguchi K
    : Olaparib potentiates anticancer drug cytotoxicity via 53BP1 in oesophageal squamous cell carcinoma cells. Anticancer Res 40(2): 813-823, 2020. PMID: 32014924. DOI: 10.21873/anticanres.14013
    OpenUrlAbstract/FREE Full Text
  7. ↵
    1. Schettini F,
    2. De Santo I,
    3. Rea CG,
    4. De Placido P,
    5. Formisano L,
    6. Giuliano M,
    7. Arpino G,
    8. De Laurentiis M,
    9. Puglisi F,
    10. De Placido S and
    11. Del Mastro L
    : CDK 4/6 inhibitors as single agent in advanced solid tumors. Front Oncol 8: 608, 2018. PMID: 30631751. DOI: 10.3389/fonc.2018.00608
    OpenUrlCrossRefPubMed
  8. ↵
    1. Goodall GJ and
    2. Wickramasinghe VO
    : RNA in cancer. Nat Rev Cancer 21(1): 22-36, 2021. PMID: 33082563. DOI: 10.1038/s41568-020-00306-0
    OpenUrlCrossRefPubMed
  9. ↵
    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
  10. ↵
    1. Tang Q and
    2. Hann SS
    : Biological roles and mechanisms of circular RNA in human cancers. Onco Targets Ther 13: 2067-2092, 2020. PMID: 32210574. DOI: 10.2147/OTT.S233672
    OpenUrlCrossRefPubMed
  11. ↵
    1. Zhu LP,
    2. He YJ,
    3. Hou JC,
    4. Chen X,
    5. Zhou SY,
    6. Yang SJ,
    7. Li J,
    8. Zhang HD,
    9. Hu JH,
    10. Zhong SL,
    11. Zhao JH and
    12. Tang JH
    : The role of circRNAs in cancers. Biosci Rep 37(5): BSR20170750, 2017. PMID: 28928231. DOI: 10.1042/BSR20170750
    OpenUrlAbstract/FREE Full Text
  12. ↵
    1. Zhang Q,
    2. Wang W,
    3. Zhou Q,
    4. Chen C,
    5. Yuan W,
    6. Liu J,
    7. Li X and
    8. Sun Z
    : Roles of circRNAs in the tumour microenvironment. Mol Cancer 19(1): 14, 2020. PMID: 31973726. DOI: 10.1186/s12943-019-1125-9
    OpenUrlCrossRefPubMed
  13. ↵
    1. Shi X,
    2. Wang B,
    3. Feng X,
    4. Xu Y,
    5. Lu K and
    6. Sun M
    : circRNAs and exosomes: a mysterious frontier for human cancer. Mol Ther Nucleic Acids 19: 384-392, 2020. PMID: 31887549. DOI: 10.1016/j.omtn.2019.11.023
    OpenUrlCrossRefPubMed
  14. ↵
    1. Lei M,
    2. Zheng G,
    3. Ning Q,
    4. Zheng J and
    5. Dong D
    : Translation and functional roles of circular RNAs in human cancer. Mol Cancer 19(1): 30, 2020. PMID: 32059672. DOI: 10.1186/s12943-020-1135-7
    OpenUrlCrossRefPubMed
  15. ↵
    1. Meng S,
    2. Zhou H,
    3. Feng Z,
    4. Xu Z,
    5. Tang Y,
    6. Li P and
    7. Wu M
    : CircRNA: functions and properties of a novel potential biomarker for cancer. Mol Cancer 16(1): 94, 2017. PMID: 28535767. DOI: 10.1186/s12943-017-0663-2
    OpenUrlCrossRefPubMed
  16. ↵
    1. Sanger HL,
    2. Klotz G,
    3. Riesner D,
    4. Gross HJ and
    5. Kleinschmidt AK
    : Viroids are single-stranded covalently closed circular RNA molecules existing as highly base-paired rod-like structures. Proc Natl Acad Sci USA 73(11): 3852-3856, 1976. PMID: 1069269. DOI: 10.1073/pnas.73.11.3852
    OpenUrlAbstract/FREE Full Text
  17. ↵
    1. Hsu MT and
    2. Coca-Prados M
    : Electron microscopic evidence for the circular form of RNA in the cytoplasm of eukaryotic cells. Nature 280(5720): 339-340, 1979. PMID: 460409. DOI: 10.1038/280339a0
    OpenUrlCrossRefPubMed
  18. ↵
    1. Piwecka M,
    2. Glažar P,
    3. Hernandez-Miranda LR,
    4. Memczak S,
    5. Wolf SA,
    6. Rybak-Wolf A,
    7. Filipchyk A,
    8. Klironomos F,
    9. Cerda Jara CA,
    10. Fenske P,
    11. Trimbuch T,
    12. Zywitza V,
    13. Plass M,
    14. Schreyer L,
    15. Ayoub S,
    16. Kocks C,
    17. Kühn R,
    18. Rosenmund C,
    19. Birchmeier C and
    20. Rajewsky N
    : Loss of a mammalian circular RNA locus causes miRNA deregulation and affects brain function. Science 357(6357): eaam8526, 2017. PMID: 28798046. DOI: 10.1126/science.aam8526
    OpenUrlAbstract/FREE Full Text
  19. ↵
    1. Feng Q,
    2. Zhang H,
    3. Yao D,
    4. Chen WD and
    5. Wang YD
    : Emerging role of non-coding RNAs in esophageal squamous cell carcinoma. Int J Mol Sci 21(1): 258, 2019. PMID: 31905958. DOI: 10.3390/ijms21010258
    OpenUrlCrossRefPubMed
    1. Niu C,
    2. Zhao L,
    3. Guo X,
    4. Shen Y,
    5. Shao Y and
    6. Liu F
    : Diagnostic accuracy of circRNAs in esophageal cancer: a meta-analysis. Dis Markers 2019: 9673129, 2019. PMID: 31534563. DOI: 10.1155/2019/9673129
    OpenUrlCrossRefPubMed
    1. Zhang H,
    2. Shen Y,
    3. Li Z,
    4. Ruan Y,
    5. Li T,
    6. Xiao B and
    7. Sun W
    : The biogenesis and biological functions of circular RNAs and their molecular diagnostic values in cancers. J Clin Lab Anal 34(1): e23049, 2020. PMID: 31556152. DOI: 10.1002/jcla.23049
    OpenUrlCrossRefPubMed
  20. ↵
    1. Zhang X,
    2. Lu N,
    3. Wang L,
    4. Wang Y,
    5. Li M,
    6. Zhou Y,
    7. Yan H,
    8. Cui M,
    9. Zhang M and
    10. Zhang L
    : Circular RNAs and esophageal cancer. Cancer Cell Int 20: 362, 2020. PMID: 32774156. DOI: 10.1186/s12935-020-01451-0
    OpenUrlCrossRefPubMed
  21. ↵
    1. Du WW,
    2. Yang W,
    3. Liu E,
    4. Yang Z,
    5. Dhaliwal P and
    6. Yang BB
    : Foxo3 circular RNA retards cell cycle progression via forming ternary complexes with p21 and CDK2. Nucleic Acids Res 44(6): 2846-2858, 2016. PMID: 26861625. DOI: 10.1093/nar/gkw027
    OpenUrlCrossRefPubMed
  22. ↵
    1. Anderson MJ,
    2. Viars CS,
    3. Czekay S,
    4. Cavenee WK and
    5. Arden KC
    : Cloning and characterization of three human forkhead genes that comprise an FKHR-like gene subfamily. Genomics 47(2): 187-199, 1998. PMID: 9479491. DOI: 10.1006/geno.1997.5122
    OpenUrlCrossRefPubMed
  23. ↵
    1. Xing Y,
    2. Zha WJ,
    3. Li XM,
    4. Li H,
    5. Gao F,
    6. Ye T,
    7. Du WQ and
    8. Liu YC
    : Circular RNA circ-Foxo3 inhibits esophageal squamous cell cancer progression via the miR-23a/PTEN axis. J Cell Biochem 121(3): 2595-2605, 2020. PMID: 31680314. DOI: 10.1002/jcb.29481
    OpenUrlCrossRefPubMed
  24. ↵
    1. Liu T,
    2. Wang Y,
    3. Wang Y and
    4. Chan AM
    : Multifaceted regulation of PTEN subcellular distributions and biological functions. Cancers (Basel) 11(9): 1247, 2019. PMID: 31454965. DOI: 10.3390/cancers11091247
    OpenUrlCrossRefPubMed
  25. ↵
    1. Wang H,
    2. Song X,
    3. Wang Y,
    4. Yin X,
    5. Liang Y,
    6. Zhang T,
    7. Xu L,
    8. Jiang F and
    9. Dong G
    : CircCNTNAP3-TP53-positive feedback loop suppresses malignant progression of esophageal squamous cell carcinoma. Cell Death Dis 11(11): 1010, 2020. PMID: 33239613. DOI: 10.1038/s41419-020-03217-y
    OpenUrlCrossRefPubMed
  26. ↵
    1. Yang Z,
    2. Qu CB,
    3. Zhang Y,
    4. Zhang WF,
    5. Wang DD,
    6. Gao CC,
    7. Ma L,
    8. Chen JS,
    9. Liu KL,
    10. Zheng B,
    11. Zhang XH,
    12. Zhang ML,
    13. Wang XL,
    14. Wen JK and
    15. Li W
    : Dysregulation of p53-RBM25-mediated circAMOTL1L biogenesis contributes to prostate cancer progression through the circAMOTL1L-miR-193a-5p-Pcdha pathway. Oncogene 38(14): 2516-2532, 2019. PMID: 30531834. DOI: 10.1038/s41388-018-0602-8
    OpenUrlCrossRefPubMed
  27. ↵
    1. Levine AJ and
    2. Oren M
    : The first 30 years of p53: growing ever more complex. Nat Rev Cancer 9(10): 749-758, 2009. PMID: 19776744. DOI: 10.1038/nrc2723
    OpenUrlCrossRefPubMed
  28. ↵
    1. Wellenstein MD,
    2. Coffelt SB,
    3. Duits DEM,
    4. van Miltenburg MH,
    5. Slagter M,
    6. de Rink I,
    7. Henneman L,
    8. Kas SM,
    9. Prekovic S,
    10. Hau CS,
    11. Vrijland K,
    12. Drenth AP,
    13. de Korte-Grimmerink R,
    14. Schut E,
    15. van der Heijden I,
    16. Zwart W,
    17. Wessels LFA,
    18. Schumacher TN,
    19. Jonkers J and
    20. de Visser KE
    : Loss of p53 triggers WNT-dependent systemic inflammation to drive breast cancer metastasis. Nature 572(7770): 538-542, 2019. PMID: 31367040. DOI: 10.1038/s41586-019-1450-6
    OpenUrlCrossRefPubMed
  29. ↵
    1. Shimada H
    : p53 molecular approach to diagnosis and treatment of esophageal squamous cell carcinoma. Ann Gastroenterol Surg 2(4): 266-273, 2018. PMID: 30003189. DOI: 10.1002/ags3.12179
    OpenUrlCrossRefPubMed
  30. ↵
    1. Pan Z,
    2. Lin J,
    3. Wu D,
    4. He X,
    5. Wang W,
    6. Hu X,
    7. Zhang L and
    8. Wang M
    : Hsa_circ_0006948 enhances cancer progression and epithelial-mesenchymal transition through the miR-490-3p/HMGA2 axis in esophageal squamous cell carcinoma. Aging (Albany NY) 11(24): 11937-11954, 2019. PMID: 31881015. DOI: 10.18632/aging.102519
    OpenUrlCrossRefPubMed
  31. ↵
    1. Mansoori B,
    2. Mohammadi A,
    3. Ditzel HJ,
    4. Duijf PHG,
    5. Khaze V,
    6. Gjerstorff MF and
    7. Baradaran B
    : HMGA2 as a critical regulator in cancer development. Genes (Basel) 12(2): 269, 2021. PMID: 33668453. DOI: 10.3390/genes12020269
    OpenUrlCrossRefPubMed
  32. ↵
    1. Young AR and
    2. Narita M
    : Oncogenic HMGA2: short or small? Genes Dev 21(9): 1005-1009, 2007. PMID: 17473167. DOI: 10.1101/gad.1554707
    OpenUrlFREE Full Text
    1. Wang Y,
    2. Hu L,
    3. Wang J,
    4. Li X,
    5. Sahengbieke S,
    6. Wu J and
    7. Lai M
    : HMGA2 promotes intestinal tumorigenesis by facilitating MDM2-mediated ubiquitination and degradation of p53. J Pathol 246(4): 508-518, 2018. PMID: 30175854. DOI: 10.1002/path.5164
    OpenUrlCrossRefPubMed
  33. ↵
    1. Fedele M,
    2. Visone R,
    3. De Martino I,
    4. Troncone G,
    5. Palmieri D,
    6. Battista S,
    7. Ciarmiello A,
    8. Pallante P,
    9. Arra C,
    10. Melillo RM,
    11. Helin K,
    12. Croce CM and
    13. Fusco A
    : HMGA2 induces pituitary tumorigenesis by enhancing E2F1 activity. Cancer Cell 9(6): 459-471, 2006. PMID: 16766265. DOI: 10.1016/j.ccr.2006.04.024
    OpenUrlCrossRefPubMed
  34. ↵
    1. Su L,
    2. Bryan N,
    3. Battista S,
    4. Freitas J,
    5. Garabedian A,
    6. D’Alessio F,
    7. Romano M,
    8. Falanga F,
    9. Fusco A,
    10. Kos L,
    11. Chambers J,
    12. Fernandez-Lima F,
    13. Chapagain PP,
    14. Vasile S,
    15. Smith L and
    16. Leng F
    : Identification of HMGA2 inhibitors by AlphaScreen-based ultra-high-throughput screening assays. Sci Rep 10(1): 18850, 2020. PMID: 33139812. DOI: 10.1038/s41598-020-75890-0
    OpenUrlCrossRefPubMed
  35. ↵
    1. Palumbo Júnior A,
    2. Da Costa NM,
    3. Esposito F,
    4. Fusco A and
    5. Pinto LF
    : High Mobility Group A proteins in esophageal carcinomas. Cell Cycle 15(18): 2410-2413, 2016. PMID: 27484584. DOI: 10.1080/15384101.2016.1215388
    OpenUrlCrossRefPubMed
  36. ↵
    1. Palumbo A Jr.,
    2. Da Costa NM,
    3. Esposito F,
    4. De Martino M,
    5. D’Angelo D,
    6. de Sousa VP,
    7. Martins I,
    8. Nasciutti LE,
    9. Fusco A and
    10. Ribeiro Pinto LF
    : HMGA2 overexpression plays a critical role in the progression of esophageal squamous carcinoma. Oncotarget 7(18): 25872-25884, 2016. PMID: 27027341. DOI: 10.18632/oncotarget.8288
    OpenUrlCrossRefPubMed
  37. ↵
    1. Hall IF,
    2. Climent M,
    3. Quintavalle M,
    4. Farina FM,
    5. Schorn T,
    6. Zani S,
    7. Carullo P,
    8. Kunderfranco P,
    9. Civilini E,
    10. Condorelli G and
    11. Elia L
    : Circ_Lrp6, a circular RNA enriched in vascular smooth muscle cells, acts as a sponge regulating miRNA-145 function. Circ Res 124(4): 498-510, 2019. PMID: 30582454. DOI: 10.1161/CIRCRESAHA.118.314240
    OpenUrlCrossRefPubMed
  38. ↵
    1. Wang J,
    2. Zhu W,
    3. Tao G and
    4. Wang W
    : Circular RNA circ-LRP6 facilitates Myc-driven tumorigenesis in esophageal squamous cell cancer. Bioengineered 11(1): 932-938, 2020. PMID: 32867570. DOI: 10.1080/21655979.2020.1809922
    OpenUrlCrossRefPubMed
  39. ↵
    1. Baluapuri A,
    2. Wolf E and
    3. Eilers M
    : Target gene-independent functions of MYC oncoproteins. Nat Rev Mol Cell Biol 21(5): 255-267, 2020. PMID: 32071436. DOI: 10.1038/s41580-020-0215-2
    OpenUrlCrossRefPubMed
    1. Schuhmacher M,
    2. Staege MS,
    3. Pajic A,
    4. Polack A,
    5. Weidle UH,
    6. Bornkamm GW,
    7. Eick D and
    8. Kohlhuber F
    : Control of cell growth by c-Myc in the absence of cell division. Curr Biol 9(21): 1255-1258, 1999. PMID: 10556095. DOI: 10.1016/s0960-9822(99)80507-7
    OpenUrlCrossRefPubMed
    1. Schuhmacher M,
    2. Kohlhuber F,
    3. Hölzel M,
    4. Kaiser C,
    5. Burtscher H,
    6. Jarsch M,
    7. Bornkamm GW,
    8. Laux G,
    9. Polack A,
    10. Weidle UH and
    11. Eick D
    : The transcriptional program of a human B cell line in response to Myc. Nucleic Acids Res 29(2): 397-406, 2001. PMID: 11139609. DOI: 10.1093/nar/29.2.397
    OpenUrlCrossRefPubMed
  40. ↵
    1. Schlosser I,
    2. Hölzel M,
    3. Hoffmann R,
    4. Burtscher H,
    5. Kohlhuber F,
    6. Schuhmacher M,
    7. Chapman R,
    8. Weidle UH and
    9. Eick D
    : Dissection of transcriptional programmes in response to serum and c-Myc in a human B-cell line. Oncogene 24(3): 520-524, 2005. PMID: 15516975. DOI: 10.1038/sj.onc.1208198
    OpenUrlCrossRefPubMed
  41. ↵
    1. Massó-Vallés D and
    2. Soucek L
    : Blocking Myc to treat cancer: reflecting on two decades of Omomyc. Cells 9(4): 883, 2020. PMID: 32260326. DOI: 10.3390/cells9040883
    OpenUrlCrossRefPubMed
  42. ↵
    1. Allen-Petersen BL and
    2. Sears RC
    : Mission possible: Advances in MYC therapeutic targeting in cancer. BioDrugs 33(5): 539-553, 2019. PMID: 31392631. DOI: 10.1007/s40259-019-00370-5
    OpenUrlCrossRefPubMed
  43. ↵
    1. Li RC,
    2. Ke S,
    3. Meng FK,
    4. Lu J,
    5. Zou XJ,
    6. He ZG,
    7. Wang WF and
    8. Fang MH
    : CiRS-7 promotes growth and metastasis of esophageal squamous cell carcinoma via regulation of miR-7/HOXB13. Cell Death Dis 9(8): 838, 2018. PMID: 30082829. DOI: 10.1038/s41419-018-0852-y
    OpenUrlCrossRefPubMed
  44. ↵
    1. Nesteruk K,
    2. Janmaat VT,
    3. Liu H,
    4. Ten Hagen TLM,
    5. Peppelenbosch MP and
    6. Fuhler GM
    : Forced expression of HOXA13 confers oncogenic hallmarks to esophageal keratinocytes. Biochim Biophys Acta Mol Basis Dis 1866(8): 165776, 2020. PMID: 32222541. DOI: 10.1016/j.bbadis.2020.165776
    OpenUrlCrossRefPubMed
  45. ↵
    1. Zhang E,
    2. Han L,
    3. Yin D,
    4. He X,
    5. Hong L,
    6. Si X,
    7. Qiu M,
    8. Xu T,
    9. De W,
    10. Xu L,
    11. Shu Y and
    12. Chen J
    : H3K27 acetylation activated-long non-coding RNA CCAT1 affects cell proliferation and migration by regulating SPRY4 and HOXB13 expression in esophageal squamous cell carcinoma. Nucleic Acids Res 45(6): 3086-3101, 2017. PMID: 27956498. DOI: 10.1093/nar/gkw1247
    OpenUrlCrossRefPubMed
  46. ↵
    1. Gu ZD,
    2. Shen LY,
    3. Wang H,
    4. Chen XM,
    5. Li Y,
    6. Ning T and
    7. Chen KN
    : HOXA13 promotes cancer cell growth and predicts poor survival of patients with esophageal squamous cell carcinoma. Cancer Res 69(12): 4969-4973, 2009. PMID: 19491265. DOI: 10.1158/0008-5472.CAN-08-4546
    OpenUrlAbstract/FREE Full Text
  47. ↵
    1. Bhatlekar S,
    2. Fields JZ and
    3. Boman BM
    : HOX genes and their role in the development of human cancers. J Mol Med (Berl) 92(8): 811-823, 2014. PMID: 24996520. DOI: 10.1007/s00109-014-1181-y
    OpenUrlCrossRefPubMed
  48. ↵
    1. Xu Z,
    2. Tie X,
    3. Li N,
    4. Yi Z,
    5. Shen F and
    6. Zhang Y
    : Circular RNA hsa_circ_0000654 promotes esophageal squamous cell carcinoma progression by regulating the miR-149-5p/IL-6/STAT3 pathway. IUBMB Life 72(3): 426-439, 2020. PMID: 31778020. DOI: 10.1002/iub.2202
    OpenUrlCrossRefPubMed
  49. ↵
    1. Weidle UH,
    2. Klostermann S,
    3. Eggle D and
    4. Krüger A
    : Interleukin 6/interleukin 6 receptor interaction and its role as a therapeutic target for treatment of cachexia and cancer. Cancer Genomics Proteomics 7(6): 287-302, 2010. PMID: 21156962.
    OpenUrlAbstract/FREE Full Text
  50. ↵
    1. Kumari N,
    2. Dwarakanath BS,
    3. Das A and
    4. Bhatt AN
    : Role of interleukin-6 in cancer progression and therapeutic resistance. Tumour Biol 37(9): 11553-11572, 2016. PMID: 27260630. DOI: 10.1007/s13277-016-5098-7
    OpenUrlCrossRefPubMed
  51. ↵
    1. Johnson DE,
    2. O’Keefe RA and
    3. Grandis JR
    : Targeting the IL-6/JAK/STAT3 signalling axis in cancer. Nat Rev Clin Oncol 15(4): 234-248, 2018. PMID: 29405201. DOI: 10.1038/nrclinonc.2018.8
    OpenUrlCrossRefPubMed
  52. ↵
    1. Groblewska M,
    2. Mroczko B,
    3. Sosnowska D and
    4. Szmitkowski M
    : Interleukin 6 and C-reactive protein in esophageal cancer. Clin Chim Acta 413(19-20): 1583-1590, 2012. PMID: 22609487. DOI: 10.1016/j.cca.2012.05.009
    OpenUrlCrossRefPubMed
  53. ↵
    1. Fang N,
    2. Shi Y,
    3. Fan Y,
    4. Long T,
    5. Shu Y and
    6. Zhou J
    : Circ_0072088 promotes proliferation, migration, and invasion of esophageal squamous cell cancer by absorbing miR-377. J Oncol 2020: 8967126, 2020. PMID: 33061973. DOI: 10.1155/2020/8967126
    OpenUrlCrossRefPubMed
  54. ↵
    1. Li Z,
    2. Huang C,
    3. Bao C,
    4. Chen L,
    5. Lin M,
    6. Wang X,
    7. Zhong G,
    8. Yu B,
    9. Hu W,
    10. Dai L,
    11. Zhu P,
    12. Chang Z,
    13. Wu Q,
    14. Zhao Y,
    15. Jia Y,
    16. Xu P,
    17. Liu H and
    18. Shan G
    : Exon-intron circular RNAs regulate transcription in the nucleus. Nat Struct Mol Biol 22(3): 256-264, 2015. PMID: 25664725. DOI: 10.1038/nsmb.2959
    OpenUrlCrossRefPubMed
  55. ↵
    1. Cunningham D,
    2. Stenning SP,
    3. Smyth EC,
    4. Okines AF,
    5. Allum WH,
    6. Rowley S,
    7. Stevenson L,
    8. Grabsch HI,
    9. Alderson D,
    10. Crosby T,
    11. Griffin SM,
    12. Mansoor W,
    13. Coxon FY,
    14. Falk SJ,
    15. Darby S,
    16. Sumpter KA,
    17. Blazeby JM and
    18. Langley RE
    : Peri-operative chemotherapy with or without bevacizumab in operable oesophagogastric adenocarcinoma (UK Medical Research Council ST03): primary analysis results of a multicentre, open-label, randomised phase 2-3 trial. Lancet Oncol 18(3): 357-370, 2017. PMID: 28163000. DOI: 10.1016/S1470-2045(17)30043-8
    OpenUrlCrossRefPubMed
  56. ↵
    1. Yoon HH,
    2. Bendell JC,
    3. Braiteh FS,
    4. Firdaus I,
    5. Philip PA,
    6. Cohn AL,
    7. Lewis N,
    8. Anderson DM,
    9. Arrowsmith E,
    10. Schwartz JD,
    11. Gao L,
    12. Hsu Y,
    13. Xu Y,
    14. Ferry D,
    15. Alberts SR and
    16. Wainberg ZA
    : Ramucirumab combined with FOLFOX as front-line therapy for advanced esophageal, gastroesophageal junction, or gastric adenocarcinoma: a randomized, double-blind, multicenter Phase II trial. Ann Oncol 27(12): 2196-2203, 2016. PMID: 27765757. DOI: 10.1093/annonc/mdw423
    OpenUrlCrossRefPubMed
  57. ↵
    1. Kashyap MK and
    2. Abdel-Rahman O
    : Expression, regulation and targeting of receptor tyrosine kinases in esophageal squamous cell carcinoma. Mol Cancer 17(1): 54, 2018. PMID: 29455652. DOI: 10.1186/s12943-018-0790-4
    OpenUrlCrossRefPubMed
  58. ↵
    1. Shi Y,
    2. Fang N,
    3. Li Y,
    4. Guo Z,
    5. Jiang W,
    6. He Y,
    7. Ma Z and
    8. Chen Y
    : Circular RNA LPAR3 sponges microRNA-198 to facilitate esophageal cancer migration, invasion, and metastasis. Cancer Sci 111(8): 2824-2836, 2020. PMID: 32495982. DOI: 10.1111/cas.14511
    OpenUrlCrossRefPubMed
  59. ↵
    1. Matsumoto K,
    2. Umitsu M,
    3. De Silva DM,
    4. Roy A and
    5. Bottaro DP
    : Hepatocyte growth factor/MET in cancer progression and biomarker discovery. Cancer Sci 108(3): 296-307, 2017. PMID: 28064454. DOI: 10.1111/cas.13156
    OpenUrlCrossRefPubMed
  60. ↵
    1. Comoglio PM,
    2. Trusolino L and
    3. Boccaccio C
    : Known and novel roles of the MET oncogene in cancer: a coherent approach to targeted therapy. Nat Rev Cancer 18(6): 341-358, 2018. PMID: 29674709. DOI: 10.1038/s41568-018-0002-y
    OpenUrlCrossRefPubMed
  61. ↵
    1. Recondo G,
    2. Che J,
    3. Jänne PA and
    4. Awad MM
    : Targeting MET dysregulation in cancer. Cancer Discov 10(7): 922-934, 2020. PMID: 32532746. DOI: 10.1158/2159-8290.CD-19-1446
    OpenUrlAbstract/FREE Full Text
  62. ↵
    1. Kashyap MK and
    2. Abdel-Rahman O
    : Expression, regulation and targeting of receptor tyrosine kinases in esophageal squamous cell carcinoma. Mol Cancer 17(1): 54, 2018. PMID: 29455652. DOI: 10.1186/s12943-018-0790-4
    OpenUrlCrossRefPubMed
  63. ↵
    1. Van Cutsem E,
    2. Karaszewska B,
    3. Kang YK,
    4. Chung HC,
    5. Shankaran V,
    6. Siena S,
    7. Go NF,
    8. Yang H,
    9. Schupp M and
    10. Cunningham D
    : A multicenter phase II study of AMG 337 in patients with MET-amplified gastric/gastroesophageal junction/esophageal adenocarcinoma and other MET-amplified solid tumors. Clin Cancer Res 25(8): 2414-2423, 2019. PMID: 30366938. DOI: 10.1158/1078-0432.CCR-18-1337
    OpenUrlAbstract/FREE Full Text
  64. ↵
    1. Hong DS,
    2. LoRusso P,
    3. Hamid O,
    4. Janku F,
    5. Kittaneh M,
    6. Catenacci DVT,
    7. Chan E,
    8. Bekaii-Saab T,
    9. Gadgeel SM,
    10. Loberg RD,
    11. Amore BM,
    12. Hwang YC,
    13. Tang R,
    14. Ngarmchamnanrith G and
    15. Kwak EL
    : Phase I study of AMG 337, a highly selective small-molecule MET inhibitor, in patients with advanced solid tumors. Clin Cancer Res 25(8): 2403-2413, 2019. PMID: 30425090. DOI: 10.1158/1078-0432.CCR-18-1341
    OpenUrlAbstract/FREE Full Text
  65. ↵
    1. Liu J,
    2. Xue N,
    3. Guo Y,
    4. Niu K,
    5. Gao L,
    6. Zhang S,
    7. Gu H,
    8. Wang X,
    9. Zhao D and
    10. Fan R
    : CircRNA_100367 regulated the radiation sensitivity of esophageal squamous cell carcinomas through miR-217/Wnt3 pathway. Aging (Albany NY) 11(24): 12412-12427, 2019. PMID: 31851619. DOI: 10.18632/aging.102580
    OpenUrlCrossRefPubMed
  66. ↵
    1. Derksen PW,
    2. Tjin E,
    3. Meijer HP,
    4. Klok MD,
    5. MacGillavry HD,
    6. van Oers MH,
    7. Lokhorst HM,
    8. Bloem AC,
    9. Clevers H,
    10. Nusse R,
    11. van der Neut R,
    12. Spaargaren M and
    13. Pals ST
    : Illegitimate WNT signaling promotes proliferation of multiple myeloma cells. Proc Natl Acad Sci USA 101(16): 6122-6127, 2004. PMID: 15067127. DOI: 10.1073/pnas.0305855101
    OpenUrlAbstract/FREE Full Text
  67. ↵
    1. Nie X,
    2. Liu H,
    3. Liu L,
    4. Wang YD and
    5. Chen WD
    : Emerging roles of Wnt ligands in human colorectal cancer. Front Oncol 10: 1341, 2020. PMID: 32923386. DOI: 10.3389/fonc.2020.01341
    OpenUrlCrossRefPubMed
  68. ↵
    1. Jung YS and
    2. Park JI
    : Wnt signaling in cancer: therapeutic targeting of Wnt signaling beyond β-catenin and the destruction complex. Exp Mol Med 52(2): 183-191, 2020. PMID: 32037398. DOI: 10.1038/s12276-020-0380-6
    OpenUrlCrossRefPubMed
  69. ↵
    1. Huang X,
    2. Li Z,
    3. Zhang Q,
    4. Wang W,
    5. Li B,
    6. Wang L,
    7. Xu Z,
    8. Zeng A,
    9. Zhang X,
    10. Zhang X,
    11. He Z,
    12. Li Q,
    13. Sun G,
    14. Wang S,
    15. Li Q,
    16. Wang L,
    17. Zhang L,
    18. Xu H and
    19. Xu Z
    : Circular RNA AKT3 upregulates PIK3R1 to enhance cisplatin resistance in gastric cancer via miR-198 suppression. Mol Cancer 18(1): 71, 2019. PMID: 30927924. DOI: 10.1186/s12943-019-0969-3
    OpenUrlCrossRefPubMed
  70. ↵
    1. Zang HL,
    2. Ji FJ,
    3. Ju HY and
    4. Tian XF
    : Circular RNA AKT3 governs malignant behaviors of esophageal cancer cells by sponging miR-17-5p. World J Gastroenterol 27(3): 240-254, 2021. PMID: 33519139. DOI: 10.3748/wjg.v27.i3.240
    OpenUrlCrossRefPubMed
  71. ↵
    1. Guan X,
    2. Chen S and
    3. Zhao Y
    : The role of RhoC in malignant tumor invasion, metastasis and targeted therapy. Histol Histopathol 33(3): 255-260, 2018. PMID: 28664531. DOI: 10.14670/HH-11-915
    OpenUrlCrossRefPubMed
  72. ↵
    1. Thomas P,
    2. Pranatharthi A,
    3. Ross C and
    4. Srivastava S
    : RhoC: a fascinating journey from a cytoskeletal organizer to a Cancer stem cell therapeutic target. J Exp Clin Cancer Res 38(1): 328, 2019. PMID: 31340863. DOI: 10.1186/s13046-019-1327-4
    OpenUrlCrossRefPubMed
  73. ↵
    1. Zhang HZ,
    2. Liu JG,
    3. Wei YP,
    4. Wu C,
    5. Cao YK and
    6. Wang M
    : Expression of G3BP and RhoC in esophageal squamous carcinoma and their effect on prognosis. World J Gastroenterol 13(30): 4126-4130, 2007. PMID: 17696235. DOI: 10.3748/wjg.v13.i30.4126
    OpenUrlCrossRefPubMed
  74. ↵
    1. Zou S,
    2. Tong Q,
    3. Liu B,
    4. Huang W,
    5. Tian Y and
    6. Fu X
    : Targeting STAT3 in cancer immunotherapy. Mol Cancer 19(1): 145, 2020. PMID: 32972405. DOI: 10.1186/s12943-020-01258-7
    OpenUrlCrossRefPubMed
  75. ↵
    1. Wu Y,
    2. Zhi L,
    3. Zhao Y,
    4. Yang L and
    5. Cai F
    : Knockdown of circular RNA UBAP2 inhibits the malignant behaviours of esophageal squamous cell carcinoma by microRNA-422a/Rab10 axis. Clin Exp Pharmacol Physiol 47(7): 1283-1290, 2020. PMID: 32012318. DOI: 10.1111/1440-1681.13269
    OpenUrlCrossRefPubMed
  76. ↵
    1. Ma W,
    2. Xue N,
    3. Zhang J,
    4. Wang D,
    5. Yao X,
    6. Lin L and
    7. Xu Q
    : circUBAP2 regulates osteosarcoma progression via the miR-204-3p/HMGA2 axis. Int J Oncol 58(3): 298-311, 2021. PMID: 33650644. DOI: 10.3892/ijo.2021.5178
    OpenUrlCrossRefPubMed
    1. Yin Y,
    2. Gao H,
    3. Guo J and
    4. Gao Y
    : Effect of circular RNA UBAP2 silencing on proliferation and invasion of human lung cancer A549 cells and its mechanism. Zhongguo Fei Ai Za Zhi 20(12): 800-807, 2017. PMID: 29277177. DOI: 10.3779/j.issn.1009-3419.2017.12.02
    OpenUrlCrossRefPubMed
    1. Wang S,
    2. Li Q,
    3. Wang Y,
    4. Li X,
    5. Wang R,
    6. Kang Y,
    7. Xue X,
    8. Meng R,
    9. Wei Q and
    10. Feng X
    : Upregulation of circ-UBAP2 predicts poor prognosis and promotes triple-negative breast cancer progression through the miR-661/MTA1 pathway. Biochem Biophys Res Commun 505(4): 996-1002, 2018. PMID: 30314706. DOI: 10.1016/j.bbrc.2018.10.026
    OpenUrlCrossRefPubMed
  77. ↵
    1. Meng L,
    2. Jia X,
    3. Yu W,
    4. Wang C,
    5. Chen J and
    6. Liu F
    : Circular RNA UBAP2 contributes to tumor growth and metastasis of cervical cancer via modulating miR-361-3p/SOX4 axis. Cancer Cell Int 20: 357, 2020. PMID: 32760224. DOI: 10.1186/s12935-020-01436-z
    OpenUrlCrossRefPubMed
  78. ↵
    1. Wang W,
    2. Jia WD,
    3. Hu B and
    4. Pan YY
    : RAB10 overexpression promotes tumor growth and indicates poor prognosis of hepatocellular carcinoma. Oncotarget 8(16): 26434-26447, 2017. PMID: 28460436. DOI: 10.18632/oncotarget.15507
    OpenUrlCrossRefPubMed
  79. ↵
    1. Huang E,
    2. Fu J,
    3. Yu Q,
    4. Xie P,
    5. Yang Z,
    6. Ji H,
    7. Wang L,
    8. Luo G,
    9. Zhang Y and
    10. Li K
    : CircRNA hsa_circ_0004771 promotes esophageal squamous cell cancer progression via miR-339-5p/CDC25A axis. Epigenomics 12(7): 587-603, 2020. PMID: 32050790. DOI: 10.2217/epi-2019-0404
    OpenUrlCrossRefPubMed
  80. ↵
    1. Shen T and
    2. Huang S
    : The role of Cdc25A in the regulation of cell proliferation and apoptosis. Anticancer Agents Med Chem 12(6): 631-639, 2012. PMID: 22263797. DOI: 10.2174/187152012800617678
    OpenUrlCrossRefPubMed
  81. ↵
    1. Boutros R,
    2. Lobjois V and
    3. Ducommun B
    : CDC25 phosphatases in cancer cells: key players? Good targets? Nat Rev Cancer 7(7): 495-507, 2007. PMID: 17568790. DOI: 10.1038/nrc2169
    OpenUrlCrossRefPubMed
  82. ↵
    1. Sur S and
    2. Agrawal DK
    : Phosphatases and kinases regulating CDC25 activity in the cell cycle: clinical implications of CDC25 overexpression and potential treatment strategies. Mol Cell Biochem 416(1-2): 33-46, 2016. PMID: 27038604. DOI: 10.1007/s11010-016-2693-2
    OpenUrlCrossRefPubMed
  83. ↵
    1. Luo A,
    2. Zhou X,
    3. Shi X,
    4. Zhao Y,
    5. Men Y,
    6. Chang X,
    7. Chen H,
    8. Ding F,
    9. Li Y,
    10. Su D,
    11. Xiao Z,
    12. Hui Z and
    13. Liu Z
    : Exosome-derived miR-339-5p mediates radiosensitivity by targeting Cdc25A in locally advanced esophageal squamous cell carcinoma. Oncogene 38(25): 4990-5006, 2019. PMID: 30858545. DOI: 10.1038/s41388-019-0771-0
    OpenUrlCrossRefPubMed
  84. ↵
    1. Chen X,
    2. Sun H,
    3. Zhao Y,
    4. Zhang J,
    5. Xiong G,
    6. Cui Y and
    7. Lei C
    : CircRNA circ_0004370 promotes cell proliferation, migration, and invasion and inhibits cell apoptosis of esophageal cancer via miR-1301-3p/COL1A1 axis. Open Med (Wars) 16(1): 104-116, 2021. PMID: 33506107. DOI: 10.1515/med-2021-0001
    OpenUrlCrossRefPubMed
  85. ↵
    1. Li J,
    2. Ding Y and
    3. Li A
    : Identification of COL1A1 and COL1A2 as candidate prognostic factors in gastric cancer. World J Surg Oncol 14(1): 297, 2016. PMID: 27894325. DOI: 10.1186/s12957-016-1056-5
    OpenUrlCrossRefPubMed
  86. ↵
    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
  87. ↵
    1. Li J,
    2. Wang X,
    3. Zheng K,
    4. Liu Y,
    5. Li J,
    6. Wang S,
    7. Liu K,
    8. Song X,
    9. Li N,
    10. Xie S and
    11. Wang S
    : The clinical significance of collagen family gene expression in esophageal squamous cell carcinoma. PeerJ 7: e7705, 2019. PMID: 31598423. DOI: 10.7717/peerj.7705
    OpenUrlCrossRefPubMed
  88. ↵
    1. Fang S,
    2. Dai Y,
    3. Mei Y,
    4. Yang M,
    5. Hu L,
    6. Yang H,
    7. Guan X and
    8. Li J
    : Clinical significance and biological role of cancer-derived Type I collagen in lung and esophageal cancers. Thorac Cancer 10(2): 277-288, 2019. PMID: 30604926. DOI: 10.1111/1759-7714.12947
    OpenUrlCrossRefPubMed
  89. ↵
    1. Yue M,
    2. Liu Y,
    3. Zuo T,
    4. Jiang Y,
    5. Pan J,
    6. Zhang S and
    7. Shen X
    : Circ_0006948 contributes to cell growth, migration, invasion and epithelial-mesenchymal transition in esophageal carcinoma. Dig Dis Sci 67(2): 492-503, 2022. PMID: 33630215. DOI: 10.1007/s10620-021-06894-7
    OpenUrlCrossRefPubMed
  90. ↵
    1. Cai C,
    2. Rajaram M,
    3. Zhou X,
    4. Liu Q,
    5. Marchica J,
    6. Li J and
    7. Powers RS
    : Activation of multiple cancer pathways and tumor maintenance function of the 3q amplified oncogene FNDC3B. Cell Cycle 11(9): 1773-1781, 2012. PMID: 22510613. DOI: 10.4161/cc.20121
    OpenUrlCrossRefPubMed
  91. ↵
    1. Bian T,
    2. Zheng L,
    3. Jiang D,
    4. Liu J,
    5. Zhang J,
    6. Feng J,
    7. Zhang Q,
    8. Qian L,
    9. Qiu H,
    10. Liu Y and
    11. Yao S
    : Overexpression of fibronectin type III domain containing 3B is correlated with epithelial-mesenchymal transition and predicts poor prognosis in lung adenocarcinoma. Exp Ther Med 17(5): 3317-3326, 2019. PMID: 30988707. DOI: 10.3892/etm.2019.7370
    OpenUrlCrossRefPubMed
  92. ↵
    1. Hou Y,
    2. Liu H and
    3. Pan W
    : Knockdown of circ_0003340 induces cell apoptosis, inhibits invasion and proliferation through miR-564/TPX2 in esophageal cancer cells. Exp Cell Res 394(2): 112142, 2020. PMID: 32535036. DOI: 10.1016/j.yexcr.2020.112142
    OpenUrlCrossRefPubMed
  93. ↵
    1. Pérez de Castro I and
    2. Malumbres M
    : Mitotic stress and chromosomal instability in cancer: the case for TPX2. Genes Cancer 3(11-12): 721-730, 2012. PMID: 23634259. DOI: 10.1177/1947601912473306
    OpenUrlCrossRefPubMed
  94. ↵
    1. Neumayer G,
    2. Belzil C,
    3. Gruss OJ and
    4. Nguyen MD
    : TPX2: of spindle assembly, DNA damage response, and cancer. Cell Mol Life Sci 71(16): 3027-3047, 2014. PMID: 24556998. DOI: 10.1007/s00018-014-1582-7
    OpenUrlCrossRefPubMed
  95. ↵
    1. Lin X,
    2. Xiang X,
    3. Hao L,
    4. Wang T,
    5. Lai Y,
    6. Abudoureyimu M,
    7. Zhou H,
    8. Feng B,
    9. Chu X and
    10. Wang R
    : The role of Aurora-A in human cancers and future therapeutics. Am J Cancer Res 10(9): 2705-2729, 2020. PMID: 33042612.
    OpenUrlPubMed
  96. ↵
    1. Liu HC,
    2. Zhang GH,
    3. Liu YH,
    4. Wang P,
    5. Ma JF,
    6. Su LS,
    7. Li SL,
    8. Zhang L and
    9. Liu JW
    : TPX2 siRNA regulates growth and invasion of esophageal cancer cells. Biomed Pharmacother 68(7): 833-839, 2014. PMID: 25239289. DOI: 10.1016/j.biopha.2014.08.008
    OpenUrlCrossRefPubMed
  97. ↵
    1. Sui C,
    2. Song Z,
    3. Yu H and
    4. Wang H
    : Prognostic significance of TPX2 and NIBP in esophageal cancer. Oncol Lett 18(4): 4221-4229, 2019. PMID: 31516617. DOI: 10.3892/ol.2019.10747
    OpenUrlCrossRefPubMed
  98. ↵
    1. Ma Y,
    2. Zhang D,
    3. Wu H,
    4. Li P,
    5. Zhao W,
    6. Yang X,
    7. Xing X,
    8. Li S and
    9. Li J
    : Circular RNA PRKCI silencing represses esophageal cancer progression and elevates cell radiosensitivity through regulating the miR-186-5p/PARP9 axis. Life Sci 259: 118168, 2020. PMID: 32739469. DOI: 10.1016/j.lfs.2020.118168
    OpenUrlCrossRefPubMed
  99. ↵
    1. Iwata H,
    2. Goettsch C,
    3. Sharma A,
    4. Ricchiuto P,
    5. Goh WW,
    6. Halu A,
    7. Yamada I,
    8. Yoshida H,
    9. Hara T,
    10. Wei M,
    11. Inoue N,
    12. Fukuda D,
    13. Mojcher A,
    14. Mattson PC,
    15. Barabási AL,
    16. Boothby M,
    17. Aikawa E,
    18. Singh SA and
    19. Aikawa M
    : PARP9 and PARP14 cross-regulate macrophage activation via STAT1 ADP-ribosylation. Nat Commun 7: 12849, 2016. PMID: 27796300. DOI: 10.1038/ncomms12849
    OpenUrlCrossRefPubMed
  100. ↵
    1. Tang X,
    2. Zhang H,
    3. Long Y,
    4. Hua H,
    5. Jiang Y and
    6. Jing J
    : PARP9 is overexpressed in human breast cancer and promotes cancer cell migration. Oncol Lett 16(3): 4073-4077, 2018. PMID: 30128030. DOI: 10.3892/ol.2018.9124
    OpenUrlCrossRefPubMed
  101. ↵
    1. Juszczynski P,
    2. Kutok JL,
    3. Li C,
    4. Mitra J,
    5. Aguiar RC and
    6. Shipp MA
    : BAL1 and BBAP are regulated by a gamma interferon-responsive bidirectional promoter and are overexpressed in diffuse large B-cell lymphomas with a prominent inflammatory infiltrate. Mol Cell Biol 26(14): 5348-5359, 2006. PMID: 16809771. DOI: 10.1128/MCB.02351-05
    OpenUrlAbstract/FREE Full Text
  102. ↵
    1. Bachmann SB,
    2. Frommel SC,
    3. Camicia R,
    4. Winkler HC,
    5. Santoro R and
    6. Hassa PO
    : DTX3L and ARTD9 inhibit IRF1 expression and mediate in cooperation with ARTD8 survival and proliferation of metastatic prostate cancer cells. Mol Cancer 13: 125, 2014. PMID: 24886089. DOI: 10.1186/1476-4598-13-125
    OpenUrlCrossRefPubMed
  103. ↵
    1. Ma L,
    2. Li H,
    3. Lin Y,
    4. Wang G,
    5. Xu Q,
    6. Chen Y,
    7. Xiao K and
    8. Rao X
    : CircDUSP16 contributes to cell development in esophageal squamous cell carcinoma by regulating miR-497-5p/TKTL1 axis. J Surg Res 260: 64-75, 2021. PMID: 33326930. DOI: 10.1016/j.jss.2020.11.052
    OpenUrlCrossRefPubMed
  104. ↵
    1. Zhao J and
    2. Zhong CJ
    : A review on research progress of transketolase. Neurosci Bull 25(2): 94-99, 2009. PMID: 19290028. DOI: 10.1007/s12264-009-1113-y
    OpenUrlCrossRefPubMed
  105. ↵
    1. Coy JF
    : EDIM-TKTL1/Apo10 blood test: an innate immune system based liquid biopsy for the early detection, characterization and targeted treatment of cancer. Int J Mol Sci 18(4): 878, 2017. PMID: 28425973. DOI: 10.3390/ijms18040878
    OpenUrlCrossRefPubMed
  106. ↵
    1. Li J,
    2. Zhu SC,
    3. Li SG,
    4. Zhao Y,
    5. Xu JR and
    6. Song CY
    : TKTL1 promotes cell proliferation and metastasis in esophageal squamous cell carcinoma. Biomed Pharmacother 74: 71-76, 2015. PMID: 26349965. DOI: 10.1016/j.biopha.2015.07.004
    OpenUrlCrossRefPubMed
  107. ↵
    1. Shi Z,
    2. Tang Y,
    3. Li K and
    4. Fan Q
    : TKTL1 expression and its downregulation is implicated in cell proliferation inhibition and cell cycle arrest in esophageal squamous cell carcinoma. Tumour Biol 36(11): 8519-8529, 2015. PMID: 26032094. DOI: 10.1007/s13277-015-3608-7
    OpenUrlCrossRefPubMed
  108. ↵
    1. Li X,
    2. Song L,
    3. Wang B,
    4. Tao C,
    5. Shi L and
    6. Xu M
    : Circ0120816 acts as an oncogene of esophageal squamous cell carcinoma by inhibiting miR-1305 and releasing TXNRD1. Cancer Cell Int 20(1): 526, 2020. PMID: 33292234. DOI: 10.1186/s12935-020-01617-w
    OpenUrlCrossRefPubMed
  109. ↵
    1. Lincoln DT,
    2. Ali Emadi EM,
    3. Tonissen KF and
    4. Clarke FM
    : The thioredoxin-thioredoxin reductase system: over-expression in human cancer. Anticancer Res 23(3B): 2425-2433, 2003. PMID: 12894524.
    OpenUrlPubMed
  110. ↵
    1. Powis G and
    2. Kirkpatrick DL
    : Thioredoxin signaling as a target for cancer therapy. Curr Opin Pharmacol 7(4): 392-397, 2007. PMID: 17611157. DOI: 10.1016/j.coph.2007.04.003
    OpenUrlCrossRefPubMed
  111. ↵
    1. Dai B,
    2. Yoo SY,
    3. Bartholomeusz G,
    4. Graham RA,
    5. Majidi M,
    6. Yan S,
    7. Meng J,
    8. Ji L,
    9. Coombes K,
    10. Minna JD,
    11. Fang B and
    12. Roth JA
    : KEAP1-dependent synthetic lethality induced by AKT and TXNRD1 inhibitors in lung cancer. Cancer Res 73(17): 5532-5543, 2013. PMID: 23824739. DOI: 10.1158/0008-5472.CAN-13-0712
    OpenUrlAbstract/FREE Full Text
  112. ↵
    1. Stafford WC,
    2. Peng X,
    3. Olofsson MH,
    4. Zhang X,
    5. Luci DK,
    6. Lu L,
    7. Cheng Q,
    8. Trésaugues L,
    9. Dexheimer TS,
    10. Coussens NP,
    11. Augsten M,
    12. Ahlzén HM,
    13. Orwar O,
    14. Östman A,
    15. Stone-Elander S,
    16. Maloney DJ,
    17. Jadhav A,
    18. Simeonov A,
    19. Linder S and
    20. Arnér ESJ
    : Irreversible inhibition of cytosolic thioredoxin reductase 1 as a mechanistic basis for anticancer therapy. Sci Transl Med 10(428): 2018. PMID: 29444979. DOI: 10.1126/scitranslmed.aaf7444
    OpenUrlCrossRefPubMed
  113. ↵
    1. Chen X,
    2. Jiang J,
    3. Zhao Y,
    4. Wang X,
    5. Zhang C,
    6. Zhuan L,
    7. Zhang D and
    8. Zheng Y
    : Circular RNA circNTRK2 facilitates the progression of esophageal squamous cell carcinoma through up-regulating NRIP1 expression via miR-140-3p. J Exp Clin Cancer Res 39(1): 133, 2020. PMID: 32653032. DOI: 10.1186/s13046-020-01640-9
    OpenUrlCrossRefPubMed
  114. ↵
    1. Cavaillès V,
    2. Dauvois S,
    3. L’Horset F,
    4. Lopez G,
    5. Hoare S,
    6. Kushner PJ and
    7. Parker MG
    : Nuclear factor RIP140 modulates transcriptional activation by the estrogen receptor. EMBO J 14(15): 3741-3751, 1995. PMID: 7641693.
    OpenUrlPubMed
  115. ↵
    1. Lapierre M,
    2. Docquier A,
    3. Castet-Nicolas A,
    4. Gitenay D,
    5. Jalaguier S,
    6. Teyssier C and
    7. Cavaillès V
    : The emerging role of the transcriptional coregulator RIP140 in solid tumors. Biochim Biophys Acta 1856(1): 144-150, 2015. PMID: 26116758. DOI: 10.1016/j.bbcan.2015.06.006
    OpenUrlCrossRefPubMed
  116. ↵
    1. Aziz MH,
    2. Chen X,
    3. Zhang Q,
    4. DeFrain C,
    5. Osland J,
    6. Luo Y,
    7. Shi X and
    8. Yuan R
    : Suppressing NRIP1 inhibits growth of breast cancer cells in vitro and in vivo. Oncotarget 6(37): 39714-39724, 2015. PMID: 26492163. DOI: 10.18632/oncotarget.5356
    OpenUrlCrossRefPubMed
  117. ↵
    1. Berriel Diaz M,
    2. Krones-Herzig A,
    3. Metzger D,
    4. Ziegler A,
    5. Vegiopoulos A,
    6. Klingenspor M,
    7. Müller-Decker K and
    8. Herzig S
    : Nuclear receptor cofactor receptor interacting protein 140 controls hepatic triglyceride metabolism during wasting in mice. Hepatology 48(3): 782-791, 2008. PMID: 18712775. DOI: 10.1002/hep.22383
    OpenUrlCrossRefPubMed
  118. ↵
    1. Wang Z,
    2. Ma K,
    3. Cheng Y,
    4. Abraham JM,
    5. Liu X,
    6. Ke X,
    7. Wang Z and
    8. Meltzer SJ
    : Synthetic circular multi-miR sponge simultaneously inhibits miR-21 and miR-93 in esophageal carcinoma. Lab Invest 99(10): 1442-1453, 2019. PMID: 31217510. DOI: 10.1038/s41374-019-0273-2
    OpenUrlCrossRefPubMed
  119. ↵
    1. Fu X,
    2. He Y,
    3. Wang X,
    4. Peng D,
    5. Chen X,
    6. Li X and
    7. Wang Q
    : Overexpression of miR-21 in stem cells improves ovarian structure and function in rats with chemotherapy-induced ovarian damage by targeting PDCD4 and PTEN to inhibit granulosa cell apoptosis. Stem Cell Res Ther 8(1): 187, 2017. PMID: 28807003. DOI: 10.1186/s13287-017-0641-z
    OpenUrlCrossRefPubMed
    1. Wen SW,
    2. Zhang YF,
    3. Li Y,
    4. Liu ZX,
    5. Lv HL,
    6. Li ZH,
    7. Xu YZ,
    8. Zhu YG and
    9. Tian ZQ
    : Characterization and effects of miR-21 expression in esophageal cancer. Genet Mol Res 14(3): 8810-8818, 2015. PMID: 26345812. DOI: 10.4238/2015.August.3.4
    OpenUrlCrossRefPubMed
    1. Liang H,
    2. Wang F,
    3. Chu D,
    4. Zhang W,
    5. Liao Z,
    6. Fu Z,
    7. Yan X,
    8. Zhu H,
    9. Guo W,
    10. Zhang Y,
    11. Guan W and
    12. Chen X
    : miR-93 functions as an oncomiR for the downregulation of PDCD4 in gastric carcinoma. Sci Rep 6: 23772, 2016. PMID: 27021515. DOI: 10.1038/srep23772
    OpenUrlCrossRefPubMed
  120. ↵
    1. David S and
    2. Meltzer SJ
    : MicroRNA involvement in esophageal carcinogenesis. Curr Opin Pharmacol 11(6): 612-616, 2011. PMID: 21992930. DOI: 10.1016/j.coph.2011.09.006
    OpenUrlCrossRefPubMed
  121. ↵
    1. Meister G and
    2. Tuschl T
    : Mechanisms of gene silencing by double-stranded RNA. Nature 431(7006): 343-349, 2004. PMID: 15372041. DOI: 10.1038/nature02873
    OpenUrlCrossRefPubMed
    1. Martinez J,
    2. Patkaniowska A,
    3. Urlaub H,
    4. Lührmann R and
    5. Tuschl T
    : Single-stranded antisense siRNAs guide target RNA cleavage in RNAi. Cell 110(5): 563-574, 2002. PMID: 12230974. DOI: 10.1016/s0092-8674(02)00908-x
    OpenUrlCrossRefPubMed
  122. ↵
    1. Soutschek J,
    2. Akinc A,
    3. Bramlage B,
    4. Charisse K,
    5. Constien R,
    6. Donoghue M,
    7. Elbashir S,
    8. Geick A,
    9. Hadwiger P,
    10. Harborth J,
    11. John M,
    12. Kesavan V,
    13. Lavine G,
    14. Pandey RK,
    15. Racie T,
    16. Rajeev KG,
    17. Röhl I,
    18. Toudjarska I,
    19. Wang G,
    20. Wuschko S,
    21. Bumcrot D,
    22. Koteliansky V,
    23. Limmer S,
    24. Manoharan M and
    25. Vornlocher HP
    : Therapeutic silencing of an endogenous gene by systemic administration of modified siRNAs. Nature 432(7014): 173-178, 2004. PMID: 15538359. DOI: 10.1038/nature03121
    OpenUrlCrossRefPubMed
  123. ↵
    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
  124. ↵
    1. Carthew RW and
    2. Sontheimer EJ
    : Origins and mechanisms of miRNAs and siRNAs. Cell 136(4): 642-655, 2009. PMID: 19239886. DOI: 10.1016/j.cell.2009.01.035
    OpenUrlCrossRefPubMed
  125. ↵
    1. Roberts TC,
    2. Langer R and
    3. Wood MJA
    : Advances in oligonucleotide drug delivery. Nat Rev Drug Discov 19(10): 673-694, 2020. PMID: 32782413. DOI: 10.1038/s41573-020-0075-7
    OpenUrlCrossRefPubMed
  126. ↵
    1. Hoy SM
    : Patisiran: first global approval. Drugs 78(15): 1625-1631, 2018. PMID: 30251172. DOI: 10.1007/s40265-018-0983-6
    OpenUrlCrossRefPubMed
  127. ↵
    1. Huang Y,
    2. Hong J,
    3. Zheng S,
    4. Ding Y,
    5. Guo S,
    6. Zhang H,
    7. Zhang X,
    8. Du Q and
    9. Liang Z
    : Elimination pathways of systemically delivered siRNA. Mol Ther 19(2): 381-385, 2011. PMID: 21119623. DOI: 10.1038/mt.2010.266
    OpenUrlCrossRefPubMed
  128. ↵
    1. Zuckerman JE and
    2. Davis ME
    : Clinical experiences with systemically administered siRNA-based therapeutics in cancer. Nat Rev Drug Discov 14(12): 843-856, 2015. PMID: 26567702. DOI: 10.1038/nrd4685
    OpenUrlCrossRefPubMed
  129. ↵
    1. Setten RL,
    2. Rossi JJ and
    3. Han SP
    : The current state and future directions of RNAi-based therapeutics. Nat Rev Drug Discov 18(6): 421-446, 2019. PMID: 30846871. DOI: 10.1038/s41573-019-0017-4
    OpenUrlCrossRefPubMed
  130. ↵
    1. Hattori Y,
    2. Tamaki K,
    3. Sakasai S,
    4. Ozaki KI and
    5. Onishi H
    : Effects of PEG anchors in PEGylated siRNA lipoplexes on in vitro gene silencing effects and siRNA biodistribution in mice. Mol Med Rep 22(5): 4183-4196, 2020. PMID: 33000194. DOI: 10.3892/mmr.2020.11525
    OpenUrlCrossRefPubMed
  131. ↵
    1. Ambardekar VV,
    2. Han HY,
    3. Varney ML,
    4. Vinogradov SV,
    5. Singh RK and
    6. Vetro JA
    : The modification of siRNA with 3’ cholesterol to increase nuclease protection and suppression of native mRNA by select siRNA polyplexes. Biomaterials 32(5): 1404-1411, 2011. PMID: 21047680. DOI: 10.1016/j.biomaterials.2010.10.019
    OpenUrlCrossRefPubMed
  132. ↵
    1. Guevara ML,
    2. Persano F and
    3. Persano S
    : Advances in lipid nanoparticles for mRNA-based cancer immunotherapy. Front Chem 8: 589959, 2020. PMID: 33195094. DOI: 10.3389/fchem.2020.589959
    OpenUrlCrossRefPubMed
  133. ↵
    1. Rozema DB,
    2. Lewis DL,
    3. Wakefield DH,
    4. Wong SC,
    5. Klein JJ,
    6. Roesch PL,
    7. Bertin SL,
    8. Reppen TW,
    9. Chu Q,
    10. Blokhin AV,
    11. Hagstrom JE and
    12. Wolff JA
    : Dynamic PolyConjugates for targeted in vivo delivery of siRNA to hepatocytes. Proc Natl Acad Sci USA 104(32): 12982-12987, 2007. PMID: 17652171. DOI: 10.1073/pnas.0703778104
    OpenUrlAbstract/FREE Full Text
  134. ↵
    1. Lo JH,
    2. Hao L,
    3. Muzumdar MD,
    4. Raghavan S,
    5. Kwon EJ,
    6. Pulver EM,
    7. Hsu F,
    8. Aguirre AJ,
    9. Wolpin BM,
    10. Fuchs CS,
    11. Hahn WC,
    12. Jacks T and
    13. Bhatia SN
    : iRGD-guided tumor-penetrating nanocomplexes for therapeutic siRNA delivery to pancreatic cancer. Mol Cancer Ther 17(11): 2377-2388, 2018. PMID: 30097486. DOI: 10.1158/1535-7163.MCT-17-1090
    OpenUrlAbstract/FREE Full Text
    1. Chen Z,
    2. Penet MF,
    3. Nimmagadda S,
    4. Li C,
    5. Banerjee SR,
    6. Winnard PT Jr.,
    7. Artemov D,
    8. Glunde K,
    9. Pomper MG and
    10. Bhujwalla ZM
    : PSMA-targeted theranostic nanoplex for prostate cancer therapy. ACS Nano 6(9): 7752-7762, 2012. PMID: 22866897. DOI: 10.1021/nn301725w
    OpenUrlCrossRefPubMed
  135. ↵
    1. Chen J,
    2. Dou Y,
    3. Tang Y and
    4. Zhang X
    : Folate receptor-targeted RNAi nanoparticles for silencing STAT3 in tumor-associated macrophages and tumor cells. Nanomedicine 25: 102173, 2020. PMID: 32084593. DOI: 10.1016/j.nano.2020.102173
    OpenUrlCrossRefPubMed
  136. ↵
    1. Springer AD and
    2. Dowdy SF
    : GalNAc-siRNA conjugates: leading the way for delivery of RNAi therapeutics. Nucleic Acid Ther 28(3): 109-118, 2018. PMID: 29792572. DOI: 10.1089/nat.2018.0736
    OpenUrlCrossRefPubMed
  137. ↵
    1. Nair JK,
    2. Attarwala H,
    3. Sehgal A,
    4. Wang Q,
    5. Aluri K,
    6. Zhang X,
    7. Gao M,
    8. Liu J,
    9. Indrakanti R,
    10. Schofield S,
    11. Kretschmer P,
    12. Brown CR,
    13. Gupta S,
    14. Willoughby JLS,
    15. Boshar JA,
    16. Jadhav V,
    17. Charisse K,
    18. Zimmermann T,
    19. Fitzgerald K,
    20. Manoharan M,
    21. Rajeev KG,
    22. Akinc A,
    23. Hutabarat R and
    24. Maier MA
    : Impact of enhanced metabolic stability on pharmacokinetics and pharmacodynamics of GalNAc-siRNA conjugates. Nucleic Acids Res 45(19): 10969-10977, 2017. PMID: 28981809. DOI: 10.1093/nar/gkx818
    OpenUrlCrossRefPubMed
  138. ↵
    1. Mailhiot SE,
    2. Thompson MA,
    3. Eguchi AE,
    4. Dinkel SE,
    5. Lotz MK,
    6. Dowdy SF and
    7. June RK
    : The TAT protein transduction domain as an intra-articular drug delivery technology. Cartilage 13(2_suppl): 1637S-1645S, 2021. PMID: 32954793. DOI: 10.1177/1947603520959392
    OpenUrlCrossRefPubMed
  139. ↵
    1. Moschos SA,
    2. Jones SW,
    3. Perry MM,
    4. Williams AE,
    5. Erjefalt JS,
    6. Turner JJ,
    7. Barnes PJ,
    8. Sproat BS,
    9. Gait MJ and
    10. Lindsay MA
    : Lung delivery studies using siRNA conjugated to TAT(48-60) and penetratin reveal peptide induced reduction in gene expression and induction of innate immunity. Bioconjug Chem 18(5): 1450-1459, 2007. PMID: 17711319. DOI: 10.1021/bc070077d
    OpenUrlCrossRefPubMed
  140. ↵
    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
  141. ↵
    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
  142. ↵
    1. Liu J,
    2. Ma J,
    3. Liu Y,
    4. Xia J,
    5. Li Y,
    6. Wang ZP and
    7. Wei W
    : PROTACs: A novel strategy for cancer therapy. Semin Cancer Biol 67(Pt 2): 171-179, 2020. PMID: 32058059. DOI: 10.1016/j.semcancer.2020.02.006
    OpenUrlCrossRefPubMed
    1. Khan S,
    2. He Y,
    3. Zhang X,
    4. Yuan Y,
    5. Pu S,
    6. Kong Q,
    7. Zheng G and
    8. Zhou D
    : PROteolysis TArgeting Chimeras (PROTACs) as emerging anticancer therapeutics. Oncogene 39(26): 4909-4924, 2020. PMID: 32475992. DOI: 10.1038/s41388-020-1336-y
    OpenUrlCrossRefPubMed
  143. ↵
    1. Deshaies RJ
    : Multispecific drugs herald a new era of biopharmaceutical innovation. Nature 580(7803): 329-338, 2020. PMID: 32296187. DOI: 10.1038/s41586-020-2168-1
    OpenUrlCrossRefPubMed
  144. ↵
    1. Mullard A
    : Targeted protein degraders crowd into the clinic. Nat Rev Drug Discov 20(4): 247-250, 2021. PMID: 33737725. DOI: 10.1038/d41573-021-00052-4
    OpenUrlCrossRefPubMed
  145. ↵
    1. Arnold A,
    2. Daum S,
    3. von Winterfeld M,
    4. Berg E,
    5. Hummel M,
    6. Rau B,
    7. Stein U and
    8. Treese C
    : Prognostic impact of Claudin 18.2 in gastric and esophageal adenocarcinomas. Clin Transl Oncol 22(12): 2357-2363, 2020. PMID: 32488802. DOI: 10.1007/s12094-020-02380-0
    OpenUrlCrossRefPubMed
  146. ↵
    1. Islam F,
    2. Gopalan V and
    3. Lam AK
    : Immunoblotting in detection of tumor-associated antigens in esophageal squamous cell carcinoma. Methods Mol Biol 2129: 269-277, 2020. PMID: 32056184. DOI: 10.1007/978-1-0716-0377-2_20
    OpenUrlCrossRefPubMed
PreviousNext
Back to top

In this issue

Cancer Genomics - Proteomics: 19 (3)
Cancer Genomics & Proteomics
Vol. 19, Issue 3
May-June 2022
  • Table of Contents
  • Table of Contents (PDF)
  • 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.
Circular RNAs With Efficacy in Preclinical In Vitro and In Vivo Models of Esophageal Squamous Cell Carcinoma
(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.
12 + 1 =
Solve this simple math problem and enter the result. E.g. for 1+3, enter 4.
Citation Tools
Circular RNAs With Efficacy in Preclinical In Vitro and In Vivo Models of Esophageal Squamous Cell Carcinoma
ULRICH H. WEIDLE, TATJANA SELA, ULRICH BRINKMANN, JENS NIEWOEHNER
Cancer Genomics & Proteomics May 2022, 19 (3) 283-298; DOI: 10.21873/cgp.20320

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Reprints and Permissions
Share
Circular RNAs With Efficacy in Preclinical In Vitro and In Vivo Models of Esophageal Squamous Cell Carcinoma
ULRICH H. WEIDLE, TATJANA SELA, ULRICH BRINKMANN, JENS NIEWOEHNER
Cancer Genomics & Proteomics May 2022, 19 (3) 283-298; DOI: 10.21873/cgp.20320
del.icio.us logo Digg logo Reddit logo Twitter logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Circular RNA
    • Down-regulated circRNAs
    • CircRNAs Sponging miRs Directed Against Transcription Factors
    • circRNAs Sponging miRs Directed Against Signaling Components
    • CircRNAs Sponging miRs Directed Against Components of The Extracellular Matrix
    • Circ RNAs Sponging miRs Targeting Additional Protein Categories
    • Technical Issues
    • Targets
    • 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

  • MicroRNAs and Corresponding Targets in Esophageal Cancer as Shown In Vitro and In Vivo in Preclinical Models
  • The Application of Bayesian Methods in Cancer Prognosis and Prediction
Show more Review

Similar Articles

Keywords

  • microRNA sponges
  • short interfering RNA (siRNA)
  • synthetic circ RNA
  • target validation
  • xenografts
  • review
Cancer & Genome Proteomics

© 2022 Cancer Genomics & Proteomics

Powered by HighWire