v
Search
Advanced Search

Publications > Journals > Exploratory Research and Hypothesis in Medicine > Article Full Text

  • OPEN ACCESS

Emerging Roles of CircRNAs in Tumor Microenvironment

  • Liang Yu1,2 and
  • Yi Xu1,2,* 
 Author information
Exploratory Research and Hypothesis in Medicine   2023;8(2):171-176

doi: 10.14218/ERHM.2022.00047

Abstract

The tumor microenvironment (TME) is an integral part of cancer that serves as a harbor where tumor cells communicate with neighboring cells and non-cancerous components to determine the progression of the tumor. Researchers are increasingly turning their focus away from tumor cells alone and toward the dynamic and intricate tumor microenvironment to acquire a stronger insight into malignancies. The active crosstalk on TME and its heterogeneity make it challenging to reveal its characteristics, while details of the regulatory mechanisms remain unknown. A deep understanding of TME remodeling may provide potential biomarkers and treatment targets to enhance tumor therapy. Circular RNAs (circRNAs) are subpopulations of noncoding RNAs with unique characteristics and a wide range of biological properties involved in tumorigenesis and metastasis. Accumulating evidence has shown that circRNAs have abnormal expression and mediate signaling pathways. They play various roles in human malignancy events, such as angiogenesis, immune escape, and others. The role of circRNAs in the TME cannot be ignored, which may provide a novel path to elucidate the underlying regulatory mechanisms of TME remodeling. This review summarizes the history of TME and circRNAs and their roles and activities in the TME.

Keywords

Tumor microenvironment, CircRNA, CeRNA, Angiogenesis

Introduction

Cancer is the most severe social and economic burden. It will overtake heart disease as the leading cause of death by 2060, with a complex mechanism that has been poorly elucidated so far.1 Circular RNAs (circRNAs) are a distinct class of endogenous noncoding RNAs characterized by their covalently closed loop structure devoid of a 5′ cap or 3′ Poly A tail.2 They have been implicated in various pathological and physiological processes and the occurrence and progression of malignancies. The tumor microenvironment (TME) is crucial for tumor initiation, development, metastasis, and chemotherapy resistance because it provides the “soil” for tumor cells to thrive. A multimodal communication network forms between cancer cells and the TME. This crosstalk continuously remodels the TME and promotes tumor growth via various previously identified behaviors, including angiogenesis, immune escape, and extracellular matrix remodeling (ECM). Remarkably, because of their distinctive architecture and broad effects, circRNAs are extensively involved in these events to impact tumor status profoundly.

The TME refers to the space where tumor cells can survive. The tumor cells are in touch with their adjacent cells or acellular components, making the ‘space’ an ecosystem. The TME encompasses immune cells, vascular endothelial cells, CAFs, and others. The “seed and soil theory” proposed by Stephen Paget described the association between cancer cells and TME for the first time.3 Due to the general rapid growth of tumor tissue and the imperfect vascular inside, insufficient oxygen supply in the tumor tissue occurs, and the TME exhibits general hypoxia-like characteristics.4 As the result of cell death, debris and chemokines are released, allowing inflammatory cells to infiltrate and secrete inflammatory chemicals, resulting in immunosuppression.5 Additionally, stromal cells in the TME promote malignancy at the same time. Cancer-associated fibroblasts release growth factors and inflammatory ligands to stimulate cancer cell proliferation and angiogenesis,6 whereas vascular endothelial cells predominantly mediate angiogenesis by secreting high levels of proangiogenic factors,7 all of which contribute to tumor formation and metastasis. TME is essential for tumor genesis, development, invasion, metastasis, and therapeutic response.

CircRNA was initially observed in RNA viruses in 19768 and was thought to be the result of erroneously alternative splicing and ignored by researchers for almost half of a century.9 Research on circRNAs has been gradually emerging since T.B. Hansen et al. discovered that they act as microRNA sponges in 2013.10 In recent years, high-throughput technologies11 and biometrics toolbox12 have been utilized to explore the molecular characteristics and biofunctions of circRNAs. According to current research progress, the circRNA is mainly generated by a back-splicing event that joins the 3′ end of an exon to an upstream 5′ end to construct a circular molecular structure.13 Via sponging miRNAs or interacting with RNA-binding proteins (RBPs), circRNAs can regulate transcription. For instance, the circRNA MBL/MBNL1 has conserved binding sites that allow them to interact with muscleblind (MBL) proteins.14 Besides, circRNAs have been proven to be translated into the ribosome and encoded as polypeptides,15 apparently being inconsistent with the labels of noncoding RNAs. When circRNAs react with proteins, they can also serve as their molecular scaffolds. In addition, circRNAs are significantly more stable owing to their resistance to ribonuclease (RNase) and exhibit a greater abundance than corresponding linear mRNAs.16 Numerous studies have shown that circRNAs involve various biological functions, including tumorigenesis and metastasis.17 This review aims to summarize the novel progression of circRNAs, notably the events implicated in TME.

CircRNAs are involved in the regulation of tumor microenvironment

The above is just the tip of the iceberg. Hanahan and Robert A summarized the hallmarks of tumors, including sustained angiogenesis, avoiding immune destruction, deregulating cellular energetics, and others.18 These incidents serve as breakthrough points for elucidating the mechanisms of oncogenesis and uncovering anti-tumor strategies. For instance, sorafenib, approved in 2005,19 is a targeted therapy drug that inhibits various kinases. Via suppressing tumor cell proliferation and specifically inhibiting vascular targets CRAF, VEGFR-2, etc., it is still the only systemic drug approved for treating hepatocellular carcinoma.20 Moreover, since the FDA approved anti-cytotoxic T lymphocyte-associated protein 4 (anti-CTLA-4) in 2011 for advanced melanoma,21 as well as the previously popularly adopted PD-1 inhibitors and (PD-L1), tumor immunotherapy has been revolutionized by improving T cell killing capability and avoiding immune escape.22

CircRNAs and the activation of fibroblast

Cancer-associated fibroblasts (CAFs) are fibroblasts that have been activated by cancer cells and exhibit the characteristics of myofibroblasts. The malignant alteration of CAFs phenotype is closely associated with tumor growth, and CAFs are the most crucial tissue components of TME.23 A study on HCC has suggested that CXCL11 released by CAFs in HCC tissues is significantly elevated, especially in metastatic tissues, and has a cancer-promoting function. Furthermore, CXCL11 has been shown to upregulate CircUBAP2, sponging miR-4756 to increase the expression of IL-17 and IL-1, which can contribute to tumor metastasis.24 Yanxia Zhan et al. utilized circRNA array analysis to identify that circHIF1A was selectively upregulated in hypoxic breast cancer exosomes specifically.25 Additionally, circHIF1A counteracting miR-580-5p-mediated inhibition on CD44 via sponging miR-580-5p was found to be the mechanism via which CAFs modulated the stemness of cancer cells. The discovery may establish circHIF1A as a therapeutic target in breast cancer treatment. Kaihua Yang et al. revealed that circEIF3K/miR-214/PD-L1 signaling axis was involved in the progression of colorectal cancer.26 Similarly, circEIF3K was confirmed to be produced by CFAs and delivered to tumor cells via exosomes, indicating a novel immunotherapy strategy. Through the LIF/STAT3 axis, circFARP1 promotes CAFs to induce gemcitabine resistance in pancreatic cancer.27 Chen et al. discovered that CAFs-derived circSLC7A6 modulates CXCR5, thereby promoting colon cancer cell proliferation and invasion.28 In addition, matrine was proved to inhibit the production of circSLC7A6 from CAFs, hence inhibiting tumorigenesis, paving a new path for targeted therapy against CAFs in the TME.

CircRNAs and immune escape

Tumor immune escape describes a phenomenon in which tumor progression occurs while avoiding identification and action by the immune system. In recent years, programmed Cell Death Protein 1 (PD-1) has been found to be critical for suppressing tumor immunity and increasing self-tolerance through its modulation of T-cell activity.29 Meanwhile, Immune checkpoint inhibitors directed against PD-1 or PD-L1 have demonstrated a considerable improvement in the prognosis of patients with a range of malignancies.30 Latest studies have noted that circRNAs are involved in this event. Cao Gao et al. revealed that circ-VIM binds with miR-124 competitively to promote PD-L1 expression in esophageal cancer.31 Besides, silencing circ-VIM inhibited immune escape, providing a new strategy to suppress tumor progression. Yan-Jie Xu et al. discovered the hsa_circ_0136666/miR-497/PD-L1 axis in Treg-mediated immune escape in colorectal cancer.32 Similarly, in recent research, circ 0001598 sponged miR-1184 to regulate PD-L1 expression in breast cancer, which results in CD8 T cell killing and trastuzumab resistance, exhibiting HER2-positive BC patients as a potential therapeutic target.33 However, the mode of immunological escape mediated by circRNAs is not confined to the ceRNA mechanism.

Additionally, CTLA-4, a well-known critical immune checkpoint,34 has been proven to be regulated by circRNAs. In Zikun Huang et al. research, circRNA 0003528 might function as a competing endogenous RNA to regulate the expression of CTLA-4, resulting in macrophage polarization.35 Through analysis of several GEO databases, Rongjie Zhao et al. revealed that the circUBAP2-mediated ceRNA network might regulate PAAD by altering the expression of CTLA-4 and PD-1.36 On the other hand, experiments are required to verify the initial conclusion. Taken together, circRNAs are predicted to become therapeutic targets for interfering with tumor immunity in the TME as research progresses.

CircRNAs and ECM remodeling

The extracellular matrix provides a scaffold for tumor cells to form the acellular part of the TME. It is widely accepted that ECM remodeling is a prerequisite for solid tumor metastasis.37 In the study of the regulatory mechanism in breast cancer development, Yikun Qu et al. focused on the regulation of circRNAs on the extracellular matrix and observed the co-upregulated expression of circRNA-CER and matrix metalloproteinase 13 (MMP13) in breast cancer tissues.38 MMPs are zinc-dependent endopeptidases involved in extracellular matrix remodeling. Silencing circRNACER via siRNA inhibited the migration of MCF7 breast cancer cells. Furthermore, the miR-136/MMP13 axis was confirmed in ECM remodeling as a cancer driver. In another research, the dysregulation of hsa_circ_0000096 was linked to gastric cancer progression. The authors confirmed that hsa_circ_0000096 might lead to the overexpression of MMP-2 and MMP-9 in vitro and in vivo. However, the enhanced expression of E-cadherin after hsa_circ_0000096 knockdown demonstrates that ECM remodeling is complicated.39 Similarly, Li et al. discovered that circ-ERBB2/miR-637/MMP-19 is involved in ECM remodification in gastric cancer, accelerating tumor development.40 Therefore, the interaction between circRNAs and the ECM demands further exploration to identify the TME remodeling procession.

It is now well established that collagen is the primary structural protein in the EMC.41 Jinghui Yang et al. demonstrated that hsa_circRNA_0007334 sponged to both hsa-miR-144-3p and hsa-miR-577 to evaluate MMP7 and Collagen Type I Alpha 1 Chain (COL1A1) expression, resulting in pancreatic ductal adenocarcinoma advancement.42 Similarly, the hsa_circRNA_002178/miR-328-3p/COL1A1 axis was implicated in the development of breast cancer. Besides, hsa_circRNA_002178 silencing inhibits tumor development in vivo, providing us with a potential target valuable target.43 Recently, Xiaobo Chen et al. confirmed that circ_0004370 works as a molecular sponge, interacting with miR-1301-3p to regulate COL1A1 expression. Silencing circ 0004370 inhibits the EMT process in esophageal cancer.44 Therefore, the interaction between circRNAs and the ECM makes it a more suitable environment for tumor progression.

CircRNAs and angiogenesis

Angiogenesis, a biological process that is strictly controlled, is critical in tumor progression because blood vessels are nearly the only way energy enters the tumor microenvironment.44 We recently discovered that circ-CCAC1 was differently expressed in cholangiocarcinoma bile exosomes and investigated its involvement in the tumor microenvironment. Circ-CCAC1 was delivered to endothelial cells through exosomes, weakening the endothelial barrier as evaluated by rhodamine leakage assays and promoting angiogenesis as indicated by tubes and vascular sprout formation compared to the control group.45 In research by Guo Y et al., circ3823 was shown to increase the expression of MYC and CCND1 via the miR-30c-5p/TCF7 pathway, which stimulated colorectal cancer angiogenesis. Additionally, the expression of circ3823 in serum has high sensitivity and specificity (AUC was 0.8074) for the detection of CRC, according to receiver operating characteristic curves, indicating the possibility of circ3823 being a diagnosis biomarker.46 Similarly, a recent study found that the Circ 0030998/miR-567/VEGFA axis is involved in the proliferation of colorectal cancer cells and angiogenesis. The authors utilized rescue assays to demonstrate that Circ 0030998 has these effects via VEGFA.47 Yang Jiang et al. discovered that the U2AF2/circRNA ARF1/miR-342-3p/ISL2 feedback loop is essential to promote glioma angiogenesis through RNA pull-down and dual-luciferase reporter.48

In addition to the above-mentioned angiogenesis functions, circRNAs play a crucial antiangiogenic role. For instance, Yawei Li utilized RNA-seq to identify and name it bladder cancer-related circular RNA-2 (BCRC-2), which is down-regulated considerably in bladder cancer tissues and cell lines.49 Mechanistically, circHIPK3 includes two key miR-558 binding sites and may effectively decrease heparanase production to suppress angiogenesis, inhibiting its therapeutic potential. This indicates that much remains unknown about the role of circRNAs in angiogenesis.

CircRNAs and hypoxia

Hypoxia is a common microenvironmental feature of solid tumors due to uncontrolled proliferation.50 Numerous recent studies have established a link between circRNAs and tumor microenvironment hypoxia. Rui Yang et al. revealed that circWSB1 was elevated in hypoxic circumstances by HIF1α and could compete with deubiquitinase USP10 in breast cancer cells, which is essential for maintaining the stability of P53, resulting in p53 degradation and tumorigenesis.51 However, Jun Liu et al. confirmed the high expression of circ 0004543 in cervical cancer tissues and cell lines and its upstream regulation mechanism of HIF1α, i.e., circ 0004543 binds to hsa-miR-217 to increase the expression of HIF-1a, triggering tumorgenesis, providing us with a novel possible therapeutic target.52 Xiwu Ouyang et al. focused on the phenomenon of androgen receptor (AR) restraining HCC progression in hypoxia and demonstrated that AR/circ-LNPEP/miR-532-3p/RAB9A axis is involved; the following rescue assay also demonstrated the function and the core role in the regulation.53 Metabolic reprogramming is a hallmark of malignancy with the deepening understanding of tumor biology and the complexity of tumor metabolism.54 In TME, hypoxia of tumor cells also results in metabolic reprogramming, leading to angiogenesis and metastasis.55 The role of circRNAs in this process has also been gradually revealed. Yiming Jiang et al. investigated the regulation of HIF-1α by circRBM33 in breast cancer and evaluated glycolysis by glucose consumption and lactic acid production.56 The authors also discovered that silencing circRBM33 inhibits the production of HIF-1α, targeted by miR-542-3p, which results in glycolysis inhibition and tumor cell death. CircRNAs are found in exosomes and act as critical regulators. Recent years have seen an increase in relevant reports, but hypoxia is rare. Li Xu et al. discovered that exosomes released by hypoxic lung adenocarcinoma cells could induce malignant phenotypes in normoxic ones.57 Following that, exosomes were isolated from hypoxic and normal cells and analyzed using microarrays; circSETDB1 was initially identified and named. Additionally, they established that circSETDB1 interacts with miR-7, as a competing endogenous RNA, to regulate the expression of specificity protein1, thereby promoting tumor invasion and metastasis.

Clinical application of circRNAs

The diverse functions and characteristics of circRNAs in TME provide us with novel methods to monitor the presence of cancers or their progression. For instance, circRNA_0000392 was found to be abundant in colorectal cancer tissues, associated with the distant metastasis and lymph node metastasis of CRC positively,58 and may serve as a biomarker. Stella M et al. considered that serum exosomes make circRNA an ideal marker for improving stability and enrichment.59 They discovered that the expression of circSMARCA5 and circHIPK3 were increased in exosomes isolated from glioblastoma multiforme patients’ serum. Both circRNAs can differentiate between GBM and a healthy group. A diagnostic model which involves the two circRNAs and some known GBM markers, including NLR, PLR, and LMR, was established to achieve a more precise prediction (AUC 0.901 (0.7912 to 1.000), 95% CI). In addition to the function of biomarkers, circRNAs are also performing as potential therapeutic targets due to their diverse roles in TME. For example, Ke Hu et al. confirmed that CircUBE2D2 capsuled by exosomes enhances the level of ER alpha by binding to miR-200a-3p to increase tamoxifen resistance, offering novel insights on the enhancement of tamoxifen’s performance in breast cancer patients.60 This shows that the application of the exosome makes it a more effective therapy method.

Future direction

Complex interactions in the TME provide more realistic cancer incidences and allow for more in-depth research into cancer progression mechanisms. The stable characteristics and regulator roles of circRNAs become active molecules in the TME, as we know from considerable progress. Future research may make significant advances in the following directions: a. The analysis of circRNAs in TME is still in its early stages. Exosomes have increasingly gained interest because they are carriers in the tumor microenvironment. CircRNAs are encapsulated in exosomes, which provide them more stability and specificity, taking this as an opportunity to make breakthrough progress; b. The tumor microenvironment is heterogeneous, and the function of circRNAs in specific conditions, such as acidic environments or changes in interstitial pressure, has yet to be determined. These works will give a more accurate and comprehensive view of tumor progression. c. There have been numerous advances in tumor immunotherapy. The expression level or functions of several essential receptor molecules regulated by circRNAs may disclose their fundamental roles in tumor immunity. With the accumulation of basic research, circRNAs are continuously transformed into clinical applications to improve the treatment of cancers. How to improve the safety of a treatment or precise drug delivery determines the application prospects of these star circRNAs.

Summary and outlook

CircRNAs, a novel family of noncoding RNAs, perform critical roles in the TME (Fig. 1). In this review, we summarized recent advances in the regulatory functions of circRNAs in many events, including angiogenesis, ECM remodeling, and immune escape. However, the physiological and pathological activities of circRNAs in the TME remain largely unclear. In general, circRNAs work primarily via the ceRNA mechanism; nevertheless, most circRNAs cannot do so because they lack miRNA binding sites. Therefore, research on circRNAs translation or interaction with RNA-binding proteins is expected. As summarized in our study, exosomes, as stable carriers of cellular communication in TME, may amplify the structural and functional benefits of circRNAs and deserve concern. Last but not least, more clinical trials are to be conducted to ensure the safety of circRNAs research results to clinical translation. We are convinced that the roles of circRNAs in critical events in the TME will be gradually uncovered to serve as tumor markers or therapeutic targets.

Overview of circRNA in tumor microenvironment.
Fig. 1  Overview of circRNA in tumor microenvironment.

(a) The primary Biogenesis model of circRNAs is alternative splicing; (b) CircRNAs perform biological functions via MicroRNA sponging, protein complex stabilization, and promoting proliferation; (c) In TME, circRNAs are involved in many core processes, including ECM remodeling, CAFs formation, immune escape, and angiogenesis. CAF, cancer-associated fibroblasts; ECM, extracellular matrix remodeling; Exon, expressed region; TME, tumor microenvironment.

Abbreviations

CAF: 

cancer-associated fibroblasts

CeRNA: 

competing endogenouse RNA

circRNAs: 

circular RNA

COL1A1: 

Collagen Type I Alpha 1 Chain

CTLA-4: 

cytotoxic T lymphocyte-associated protein 4

ECM: 

extracellular matrix remodeling

MMP13: 

matrix metalloproteinase 13

PD-1: 

programmed Cell Death Protein 1

RBP: 

RNA binding protein

TME: 

tumor microenvironment

Declarations

Acknowledgement

None.

Funding

The work was supported in part by a grant from the following projects: Hong Kong Scholars Program (Grant No. XJ2020012); National Natural Science Foundation of China (Grant No. 81902431); Excellent Young Scientists Project of Natural Science Foundation of Heilongjiang Province (Grant No. YQ2019H007); Special Project of China Postdoctoral Science Foundation (Grant No. 2019T120279); Special Project of Heilongjiang Postdoctoral Science Foundation (Grant No. LBH-TZ1016); China Postdoctoral Science Foundation (Grant No. 2018M641849); Heilongjiang Postdoctoral Science Foundation (Grant No. LBH-Z18107); The Fundamental Research Funds for the Heilongjiang Provincial Universities (Grant No. 2018-KYYWF-0511); Postgraduate Innovative Research Project of Harbin Medical University (Grant No. YJSCX2016-21HYD); Foundation of Key Laboratory of Myocardial Ischemia, Ministry of Education (Grant No. KF201810); Chen Xiaoping Foundation for the Development of Science and Technology of Hubei Province (Grant No. CXPJJH11800004-001); Medjaden Young Scientists Research Fund (Grant No. MJR20220903).

Conflict of interest

YX has been an editorial board member of Exploratory Research and Hypothesis in Medicine since March 2022. The authors have no other conflict of interest related to this publication.

Authors’ contributions

Contributed to study concept and design (YX), drafting of the manuscript (LY), critical revision of the manuscript (YX), and supervision (YX).

References

  1. Li R, Jiang J, Shi H, Qian H, Zhang X, Xu W. CircRNA: A rising star in gastric cancer. Cell Mol Life Sci 2020;77(9):1661-1680 View Article PubMed/NCBI
  2. Kristensen LS, Jakobsen T, Hager H, Kjems J. The emerging roles of circRNAs in cancer and oncology. Nat Rev Clin Oncol 2022;19(3):188-206 View Article PubMed/NCBI
  3. Paget S. The distribution of secondary growths in cancer of the breast. Cancer Metastasis Rev 1989;8(2):98-101 PubMed/NCBI
  4. Jing X, Yang F, Shao C, Wei K, Xie M, Shen H, et al. Role of hypoxia in cancer therapy by regulating the tumor microenvironment. Mol Cancer 2019;18(1):157 View Article PubMed/NCBI
  5. Riera-Domingo C, Audigé A, Granja S, Cheng WC, Ho PC, Baltazar F, et al. Immunity, Hypoxia, and Metabolism-the Ménage à Trois of Cancer: Implications for Immunotherapy. Physiol Rev 2020;100(1):1-102 View Article PubMed/NCBI
  6. Biffi G, Tuveson DA. Diversity and Biology of Cancer-Associated Fibroblasts. Physiol Rev 2021;101(1):147-176 View Article PubMed/NCBI
  7. Viallard C, Larrivée B. Tumor angiogenesis and vascular normalization: alternative therapeutic targets. Angiogenesis 2017;20(4):409-426 View Article PubMed/NCBI
  8. Sanger HL, Klotz G, Riesner D, Gross HJ, Kleinschmidt AK. Viroids are single-stranded covalently closed circular RNA molecules existing as highly base-paired rod-like structures. Proc Natl Acad Sci USA 1976;73(11):3852-3856 View Article PubMed/NCBI
  9. Danan M, Schwartz S, Edelheit S, Sorek R. Transcriptome-wide discovery of circular RNAs in Archaea. Nucleic Acids Res 2012;40(7):3131-3142 View Article PubMed/NCBI
  10. Hansen TB, Jensen TI, Clausen BH, Bramsen JB, Finsen B, Damgaard CK, et al. Natural RNA circles function as efficient microRNA sponges. Nature 2013;495(7441):384-388 View Article PubMed/NCBI
  11. Reon BJ, Dutta A. Biological Processes Discovered by High-Throughput Sequencing. Am J Pathol 2016;186(4):722-732 View Article PubMed/NCBI
  12. Chen L, Wang C, Sun H, Wang J, Liang Y, Wang Y, et al. The bioinformatics toolbox for circRNA discovery and analysis. Brief Bioinform 2021;22(2):1706-1728 View Article PubMed/NCBI
  13. Yin Y, Long J, He Q, Li Y, Liao Y, He P, et al. Emerging roles of circRNA in formation and progression of cancer. J Cancer 2019;10(21):5015-5021 View Article PubMed/NCBI
  14. Ashwal-Fluss R, Meyer M, Pamudurti NR, Ivanov A, Bartok O, Hanan M, et al. circRNA biogenesis competes with pre-mRNA splicing. Mol Cell 2014;56(1):55-66 View Article PubMed/NCBI
  15. Lei M, Zheng G, Ning Q, Zheng J, Dong D. Translation and functional roles of circular RNAs in human cancer. Mol Cancer 2020;19(1):30 View Article PubMed/NCBI
  16. Tang X, Ren H, Guo M, Qian J, Yang Y, Gu C. Review on circular RNAs and new insights into their roles in cancer. Comput Struct Biotechnol J 2021;19:910-928 View Article PubMed/NCBI
  17. Shao Y, Lu B. The crosstalk between circular RNAs and the tumor microenvironment in cancer metastasis. Cancer Cell Int 2020;20:448 View Article PubMed/NCBI
  18. Hanahan D, Weinberg RA. Hallmarks of cancer: The next generation. Cell 2011;144(5):646-674 View Article PubMed/NCBI
  19. Strumberg D. Sorafenib for the treatment of renal cancer. Expert Opin Pharmacother 2012;13(3):407-419 View Article PubMed/NCBI
  20. Escudier B, Worden F, Kudo M. Sorafenib: Key lessons from over 10 years of experience. Expert Rev Anticancer Ther 2019;19(2):177-189 View Article PubMed/NCBI
  21. Hosseini A, Gharibi T, Marofi F, Babaloo Z, Baradaran B. CTLA-4: From mechanism to autoimmune therapy. Int Immunopharmacol 2020;80:106221 View Article PubMed/NCBI
  22. Bagchi S, Yuan R, Engleman EG. Immune Checkpoint Inhibitors for the Treatment of Cancer: Clinical Impact and Mechanisms of Response and Resistance. Annu Rev Pathol 2021;16:223-249 View Article PubMed/NCBI
  23. Liao Z, Tan ZW, Zhu P, Tan NS. Cancer-associated fibroblasts in tumor microenvironment - Accomplices in tumor malignancy. Cell Immunol 2019;343:103729 View Article PubMed/NCBI
  24. Liu G, Sun J, Yang ZF, Zhou C, Zhou PY, Guan RY, et al. Cancer-associated fibroblast-derived CXCL11 modulates hepatocellular carcinoma cell migration and tumor metastasis through the circUBAP2/miR-4756/IFIT1/3 axis. Cell Death Dis 2021;12(3):260 View Article PubMed/NCBI
  25. Zhan Y, Du J, Min Z, Ma L, Zhang W, Zhu W, et al. Carcinoma-associated fibroblasts derived exosomes modulate breast cancer cell stemness through exonic circHIF1A by miR-580-5p in hypoxic stress. Cell Death Discov 2021;7(1):141 View Article PubMed/NCBI
  26. Yang K, Zhang J, Bao C. Exosomal circEIF3K from cancer-associated fibroblast promotes colorectal cancer (CRC) progression via miR-214/PD-L1 axis. BMC Cancer 2021;21(1):933 View Article PubMed/NCBI
  27. Hu C, Xia R, Zhang X, Li T, Ye Y, Li G, et al. circFARP1 enables cancer-associated fibroblasts to promote gemcitabine resistance in pancreatic cancer via the LIF/STAT3 axis. Mol Cancer 2022;21(1):24 View Article PubMed/NCBI
  28. Gu C, Lu H, Qian Z. Matrine reduces the secretion of exosomal circSLC7A6 from cancer-associated fibroblast to inhibit tumorigenesis of colorectal cancer by regulating CXCR5. Biochem Biophys Res Commun 2020;527(3):638-645 View Article PubMed/NCBI
  29. Han Y, Liu D, Li L. PD-1/PD-L1 pathway: current researches in cancer. Am J Cancer Res 2020;10(3):727-742 PubMed/NCBI
  30. Doroshow DB, Bhalla S, Beasley MB, Sholl LM, Kerr KM, Gnjatic S, et al. PD-L1 as a biomarker of response to immune-checkpoint inhibitors. Nat Rev Clin Oncol 2021;18(6):345-362 View Article PubMed/NCBI
  31. Gao C, Xu YJ, Qi L, Bao YF, Zhang L, Zheng L. CircRNA VIM silence synergizes with sevoflurane to inhibit immune escape and multiple oncogenic activities of esophageal cancer by simultaneously regulating miR-124/PD-L1 axis. Cell Biol Toxicol 2021 View Article PubMed/NCBI
  32. Xu YJ, Zhao JM, Gao C, Ni XF, Wang W, Hu WW, et al. Hsa_circ_0136666 activates Treg-mediated immune escape of colorectal cancer via miR-497/PD-L1 pathway. Cell Signal 2021;86:110095 View Article PubMed/NCBI
  33. Huang L, Ma J, Cui M. Circular RNA hsa_circ_0001598 promotes programmed death-ligand-1-mediated immune escape and trastuzumab resistance via sponging miR-1184 in breast cancer cells. Immunol Res 2021;69(6):558-567 View Article PubMed/NCBI
  34. Van Coillie S, Wiernicki B, Xu J. Molecular and Cellular Functions of CTLA-4. Adv Exp Med Biol 2020;1248:7-32 View Article PubMed/NCBI
  35. Huang Z, Yao F, Liu J, Xu J, Guo Y, Su R, et al. Up-regulation of circRNA-0003528 promotes mycobacterium tuberculosis associated macrophage polarization via down-regulating miR-224-5p, miR-324-5p and miR-488-5p and up-regulating CTLA4. Aging (Albany NY) 2020;12(24):25658-25672 View Article PubMed/NCBI
  36. Zhao R, Ni J, Lu S, Jiang S, You L, Liu H, et al. CircUBAP2-mediated competing endogenous RNA network modulates tumorigenesis in pancreatic adenocarcinoma. Aging (Albany NY) 2019;11(19):8484-8501 View Article PubMed/NCBI
  37. Najafi M, Farhood B, Mortezaee K. Extracellular matrix (ECM) stiffness and degradation as cancer drivers. J Cell Biochem 2019;120(3):2782-2790 View Article PubMed/NCBI
  38. Qu Y, Dou P, Hu M, Xu J, Xia W, Sun H. circRNA-CER mediates malignant progression of breast cancer through targeting the miR-136/MMP13 axis. Mol Med Rep 2019;19(4):3314-3320 View Article PubMed/NCBI
  39. Li P, Chen H, Chen S, Mo X, Li T, Xiao B, et al. Circular RNA 0000096 affects cell growth and migration in gastric cancer. Br J Cancer 2017;116(5):626-633 View Article PubMed/NCBI
  40. Li X, He M, Guo J, Cao T. Upregulation of circular RNA circ-ERBB2 predicts unfavorable prognosis and facilitates the progression of gastric cancer via miR-503/CACUL1 and miR-637/MMP-19 signaling. Biochem Biophys Res Commun 2019;511(4):926-930 View Article PubMed/NCBI
  41. Rahman MA. Collagen of Extracellular Matrix from Marine Invertebrates and Its Medical Applications. Mar Drugs 2019;17(2):118 View Article PubMed/NCBI
  42. Yang J, Cong X, Ren M, Sun H, Liu T, Chen G, et al. Circular RNA hsa_circRNA_0007334 is Predicted to Promote MMP7 and COL1A1 Expression by Functioning as a miRNA Sponge in Pancreatic Ductal Adenocarcinoma. J Oncol 2019;2019:7630894 View Article PubMed/NCBI
  43. Liu T, Ye P, Ye Y, Lu S, Han B. Circular RNA hsa_circRNA_002178 silencing retards breast cancer progression via microRNA-328-3p-mediated inhibition of COL1A1. J Cell Mol Med 2020;24(3):2189-2201 View Article PubMed/NCBI
  44. Chen X, Sun H, Zhao Y, Zhang J, Xiong G, Cui Y, et al. 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) 2021;16(1):104-116 View Article PubMed/NCBI
  45. Xu Y, Leng K, Yao Y, Kang P, Liao G, Han Y, et al. A Circular RNA, Cholangiocarcinoma-Associated Circular RNA 1, Contributes to Cholangiocarcinoma Progression, Induces Angiogenesis, and Disrupts Vascular Endothelial Barriers. Hepatology 2021;73(4):1419-1435 View Article PubMed/NCBI
  46. Guo Y, Guo Y, Chen C, Fan D, Wu X, Zhao L, et al. Circ3823 contributes to growth, metastasis and angiogenesis of colorectal cancer: involvement of miR-30c-5p/TCF7 axis. Mol Cancer 2021;20(1):93 View Article PubMed/NCBI
  47. Jin L, Han C, Zhai T, Zhang X, Chen C, Lian L. Circ_0030998 promotes tumor proliferation and angiogenesis by sponging miR-567 to regulate VEGFA in colorectal cancer. Cell Death Discov 2021;7(1):160 View Article PubMed/NCBI
  48. Jiang Y, Zhou J, Zhao J, Zhang H, Li L, Li H, et al. The U2AF2 /circRNA ARF1/miR-342-3p/ISL2 feedback loop regulates angiogenesis in glioma stem cells. J Exp Clin Cancer Res 2020;39(1):182 View Article PubMed/NCBI
  49. Li Y, Zheng F, Xiao X, Xie F, Tao D, Huang C, et al. CircHIPK3 sponges miR-558 to suppress heparanase expression in bladder cancer cells. EMBO Rep 2017;18(9):1646-1659 View Article PubMed/NCBI
  50. Jing X, Yang F, Shao C, Wei K, Xie M, Shen H, et al. Role of hypoxia in cancer therapy by regulating the tumor microenvironment. Mol Cancer 2019;18(1):157 View Article PubMed/NCBI
  51. Yang R, Chen H, Xing L, Wang B, Hu M, Ou X, et al. Hypoxia-induced circWSB1 promotes breast cancer progression through destabilizing p53 by interacting with USP10. Mol Cancer 2022;21(1):88 View Article PubMed/NCBI
  52. Liu J, Liu Y, Zhang Y, Zheng J, Wang S, Cao G. Circular RNA hsa_circ_0004543 Aggravates Cervical Cancer Development by Sponging MicroRNA hsa-miR-217 to Upregulate Hypoxia-Inducible Factor. J Oncol 2022;2022:4031403 View Article PubMed/NCBI
  53. Ouyang X, Yao L, Liu G, Liu S, Gong L, Xiao Y. Loss of androgen receptor promotes HCC invasion and metastasis via activating circ-LNPEP/miR-532-3p/RAB9A signal under hypoxi-a. Biochem Biophys Res Commun 2021;557:26-32 View Article PubMed/NCBI
  54. Faubert B, Solmonson A, DeBerardinis RJ. Metabolic reprogramming and cancer progression. Science 2020;368(6487):eaaw5473 View Article PubMed/NCBI
  55. Miranda-Galvis M, Teng Y. Targeting Hypoxia-Driven Metabolic Reprogramming to Constrain Tumor Progression and Metastasis. Int J Mol Sci 2020;21(15):5487 View Article PubMed/NCBI
  56. Jiang Y, Zhang M, Yu D, Hou G, Wu J, Li F. CircRBM33 downregulation inhibits hypoxia-induced glycolysis and promotes apoptosis of breast cancer cells via a microRNA-542-3p/HIF-1α axis. Cell Death Discov 2022;8(1):126 View Article PubMed/NCBI
  57. Xu L, Liao WL, Lu QJ, Zhang P, Zhu J, Jiang GN. Hypoxic tumor-derived exosomal circular RNA SETDB1 promotes invasive growth and EMT via the miR-7/Sp1 axis in lung adenocarcinoma. Mol Ther Nucleic Acids 2021;23:1078-1092 View Article PubMed/NCBI
  58. Xu H, Liu Y, Cheng P, Wang C, Liu Y, Zhou W, et al. CircRNA_0000392 promotes colorectal cancer progression through the miR-193a-5p/PIK3R3/AKT axis. J Exp Clin Cancer Res 2020;39(1):283 View Article PubMed/NCBI
  59. Stella M, Falzone L, Caponnetto A, Gattuso G, Barbagallo C, Battaglia R, et al. Serum Extracellular Vesicle-Derived circHIPK3 and circSMARCA5 Are Two Novel Diagnostic Biomarkers for Glioblastoma Multiforme. Pharmaceuticals (Basel) 2021;14(7):618 View Article PubMed/NCBI
  60. Hu K, Liu X, Li Y, Li Q, Xu Y, Zeng W, et al. Exosomes Mediated Transfer of Circ_UBE2D2 Enhances the Resistance of Breast Cancer to Tamoxifen by Binding to MiR-200a-3p. Med Sci Monit 2020;26:e922253 View Article PubMed/NCBI
  • Exploratory Research and Hypothesis in Medicine
  • pISSN 2993-5113
  • eISSN 2472-0712
Back to Top

Emerging Roles of CircRNAs in Tumor Microenvironment

Liang Yu, Yi Xu
  • Reset Zoom
  • Download TIFF