Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Oncology Letters
      • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Biomedical Reports
      • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • Information for Authors
    • Information for Reviewers
    • Information for Librarians
    • Information for Advertisers
    • Conferences
  • Language Editing
Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • For Authors
    • For Reviewers
    • For Librarians
    • For Advertisers
    • Conferences
  • Language Editing
Login Register Submit
  • This site uses cookies
  • You can change your cookie settings at any time by following the instructions in our Cookie Policy. To find out more, you may read our Privacy Policy.

    I agree
Search articles by DOI, keyword, author or affiliation
Search
Advanced Search
presentation
International Journal of Molecular Medicine
Join Editorial Board Propose a Special Issue
Print ISSN: 1107-3756 Online ISSN: 1791-244X
Journal Cover
October-2024 Volume 54 Issue 4

Full Size Image

Sign up for eToc alerts
Recommend to Library

Journals

International Journal of Molecular Medicine

International Journal of Molecular Medicine

International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.

International Journal of Oncology

International Journal of Oncology

International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.

Molecular Medicine Reports

Molecular Medicine Reports

Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.

Oncology Reports

Oncology Reports

Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.

Oncology Letters

Oncology Letters

Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.

Biomedical Reports

Biomedical Reports

Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.

Molecular and Clinical Oncology

Molecular and Clinical Oncology

International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.

World Academy of Sciences Journal

World Academy of Sciences Journal

Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.

International Journal of Functional Nutrition

International Journal of Functional Nutrition

Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.

International Journal of Epigenetics

International Journal of Epigenetics

Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.

Medicine International

Medicine International

An International Open Access Journal Devoted to General Medicine.

Journal Cover
October-2024 Volume 54 Issue 4

Full Size Image

Sign up for eToc alerts
Recommend to Library

  • Article
  • Citations
    • Cite This Article
    • Download Citation
    • Create Citation Alert
    • Remove Citation Alert
    • Cited By
  • Similar Articles
    • Related Articles (in Spandidos Publications)
    • Similar Articles (Google Scholar)
    • Similar Articles (PubMed)
  • Download PDF
  • Download XML
  • View XML
Review

Role of exosomal non‑coding RNAs in ovarian cancer (Review)

  • Authors:
    • Xinchen Wang
    • Miao Yang
    • Jiamei Zhu
    • Yu Zhou
    • Gencui Li
  • View Affiliations / Copyright

    Affiliations: Department of Obstetrics and Gynecology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310000, P.R. China, Department of Life Sciences and Technology, China Pharmaceutical University, Nanjing, Jiangsu 210009, P.R. China, Department of Obstetrics and Gynecology, Jingjiang People's Hospital, Taizhou, Jiangsu 214500, P.R. China, Oriental Fortune Capital Post‑Doctoral Innovation Center, Shenzhen, Guangdong 518040, P.R. China
  • Article Number: 87
    |
    Published online on: August 7, 2024
       https://doi.org/10.3892/ijmm.2024.5411
  • Expand metrics +
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Metrics: Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )
Cited By (CrossRef): 0 citations Loading Articles...

This article is mentioned in:



Abstract

Ovarian cancer (OC) is a common gynecological disease with a high mortality rate worldwide due to its insidious nature and undetectability at an early stage. The standard treatment, combining platinum‑based chemotherapy with cytoreductive surgery, has suboptimal results. Therefore, early diagnosis of OC is crucial. All cell types secrete extracellular vesicles, particularly exosomes. Exosomes, which contain lipids, proteins, DNA and non‑coding RNAs (ncRNAs), are novel methods of intercellular communication that participate in tumor development and progression. ncRNAs are categorized by size into long ncRNAs (lncRNAs) and small ncRNAs (sncRNAs). sncRNAs further include transfer RNAs, small nucleolar RNAs, PIWI‑interacting RNAs and microRNAs (miRNAs). miRNAs inhibit protein translation and promote messenger RNA (mRNA) cleavage to suppress gene expression. By sponging downstream miRNAs, lncRNAs and circular RNAs can regulate target gene expression, thereby weakening the interactions between miRNAs and mRNAs. Exosomes and exosomal ncRNAs, commonly present in human biological fluids, are promising biomarkers for OC. The present article aimed to review the potential role of exosomal ncRNAs in the diagnosis and prognosis of OC by summarizing the characteristics, processes, roles and isolation methods of exosomes and exosomal ncRNAs.

Introduction

Gynecological cancers are common, and ovarian cancer (OC) is the deadliest due to difficulties diagnosing this disease in the early stages. Specifically, 70–90% of patients with OC are diagnosed at an advanced stage, indicating that the cancer has already spread (1). In 2018, there were ~300,000 reported cases of OC worldwide, with ~18,000 fatalities (2). Epithelial ovarian tumors have the highest incidence, whereas germ cell and sex cord-stromal OC account for ~5% of cases (3). At present, ovarian epithelial tumors are classified into four subtypes with identical pathologies: Serous, endometrioid, mucinous and clear cell (3). These tumors can be classified as either type I or II based on the malignancy. Type II tumors are more aggressive and have poorer survival outcomes compared with type I (4). High-grade serous cancers, a subtype of type II, are the most frequent and account for 80% of OC-related deaths (5,6). Platinum-based chemotherapy and cytoreductive surgery are the primary clinical treatments for OC. However, this treatment strategy is less effective for advanced OC as the lesions are difficult to remove completely, and advanced OC tends to be drug-resistant. Therefore, early diagnosis and targeted medicine are crucial for the effective treatment of OC.

At present, tissue biopsy is the most common method for cancer diagnosis, but it is invasive and time-consuming. Additionally, biopsies carry risks such as tumor implantation and metastasis, making them unsuitable for long-term monitoring (7,8). Thus, novel and sensitive biomarkers are needed for the early and quick diagnosis of cancer. All cell types secrete extracellular vesicles (EVs), particularly exosomes, which transport various cargoes such as lipids, proteins, DNA and most non-coding RNAs (ncRNAs), including circular RNA (circRNA), long ncRNA (lncRNA) and microRNA (miRNA). Exosomes can exchange signals and materials between different cells by carrying and transporting these cargoes (9). Given that exosomes collect, transport and release cargo, they can be considered new targets in OC diagnosis and treatment.

Cancer is a genetic disease primarily caused by mutations in the non-coding genome (10). Previous genomic studies have indicated that the non-coding regions of RNA are extensively transcribed (11). The Human Genome Project has demonstrated that 1.5% of the human genome consists of genes that encode proteins; the rest are transcribed without translation into proteins (12,13). The two subtypes of ncRNAs are distinguished based on their size: lncRNA and small ncRNA (sncRNA). sncRNAs can be further classified into miRNA, transfer RNA, PIWI-interacting RNA and small nucleolar RNA (14). Particularly, miRNAs inhibit protein translation and promote messenger RNA (mRNA) cleavage to suppress gene expression (15). Furthermore, miRNAs can migrate into biological fluids (16) and ~10% of circulating miRNAs are secreted via exosomes (17,18). lncRNAs, which are transcripts of >200 nucleotides with potential non-coding functions, exhibit tissue- or cancer-type specificity (19). Serving as readouts of running cellular programs or signals of certain cellular states, lncRNAs can be used to distinguish various cellular pathologies, provide a predictive value or even used to choose appropriate therapeutics for patients with cancer (20). For instance, the circulating lncRNA-GC1, derived from EVs, serves as an early indicator of neoadjuvant chemotherapy (neoCT) effectiveness and is correlated with improved survival outcomes in patients with gastric cancer undergoing neoCT treatment (21). Serum exosomal lncRNA-UCA1, initially discovered in human bladder cancer, can serve as a potential non-invasive biomarker for the diagnosis of bladder cancer. circRNAs are closed RNAs without 5′ or 3′ ends and are classified into natural or synthetic circRNAs (22). In a number of tissues, endogenous circRNAs are involved in various biological functions such as gene transcription, protein translation, cell division, immune system dysregulation and tumorigenesis (23).

In summary, ncRNAs in exosomes are involved in numerous biological functions, including gene expression and disease development, making them valuable diagnostic tools in clinical settings. The present review summarizes the features, isolation techniques, functions and mechanisms of exosomes and exosomal ncRNAs and predicts their potential in diagnosing and prognosing OC.

Exosomes

Characteristics and main isolation techniques of exosomes

Exosomes can be isolated using several methods, such as ultrafiltration, size exclusion chromatography and density gradient ultracentrifugation. Most cells secrete EVs, first reported in 1985 (24). Exosomes are nanoparticles that facilitate cellular waste disposal into the extracellular environment. Increasing evidence suggests that exosomes participate in various physiological and disease-related processes, serving as biomarkers for several diseases (25–27). Extracting exosomes is crucial for understanding their characteristics and functions. Numerous techniques have been developed to separate exosomes based on their main features, such as surface proteins, size, shape and density (Fig. 1 and Table I).

Figure 1.

Most cells can secrete extracellular vesicles, which are widely distributed in the human body. Common methods for isolating exosomes include ultracentrifugation, ultrafiltration, magnetic bead-based exosome isolation. The figure was adapted from ‘Main isolation techniques of exosomes’, by BioRender.com (January 5th, 2024). Retrieved from https://app.biorender.com/biorend plates. Tsg101, tumor susceptibility 101.

Table I.

Different exosome isolation methods.

Table I.

Different exosome isolation methods.

MethodAdvantagesDisadvantagesPrinciplePuritySample volume(Refs.)
DUCi) Low cost; ii) for a large volume; andi) Expensive equipment; andVaried sedimentation coefficientsLow-(31,32)
iii) no other markers will be inducedii) exosomes can be destructedof particles
Density gradienti) Higher purity than DUC; andi) Low productivity; andLocalization of different particles-Medium(142)
ultracentrifugationii) classification of exosomal subpopulationsii) complex operationat a specific gradient medium
UFi) Low cost and ii) simple operationi) Exosome destruction; andUtilization of membranes with aMediumMedium(143)
ii) membrane blockagespecific pore diameter or MWCO to separate sample particles
SECi) Time efficiency; and ii) protects thei) Possibility of pollution; andExosome classification according-Small(144)
integrity and biology of the exosomeii) expensive equipmentto hydrodynamic radii
Immunoaffinity capturei) Simple operation; and ii) selectivityi) High cost; and ii) exosome destructionAntigen-antibody recognition and bindingHighSmall(145)
Polymer precipitationi) Protection of the integrity and biology of exosomes; ii) simple processing; andPossibility of pollutionExosomes surrounded and precipitated by polymersLowSmall(146)
iii) time efficiency

[i] DUC, differential ultracentrifugation; MWCO, molecular weight cut-off; SEC, size-exclusion chromatography; UF, ultrafiltration.

Exosomes in liquid biopsies for cancer diagnosis

Tissue biopsy is currently the gold standard for cancer diagnosis, but it has limitations. First, tissue biopsy only provides a snapshot of the tumor tissue, possibly neglecting tumor heterogeneity and the different processes and changes that occur during interactions with the immune system and other systems. Additionally, tissue biopsy requires time to detect cancer, thus delaying further therapeutic decisions. Tissue biopsy also cannot show whether a patient responds favorably to current treatment and is impractical for frequent sample collection (28). With the development of science and technology, liquid biopsy is regarded as a non-invasive, real-time, tumor-specific method that can track the advancement and recurrence of cancer and the response to treatment interventions (29). Assessing serum from patients for human epididymis protein 4, carbohydrate antigen-125 (CA-125) and p53 levels is one way to detect and monitor OC progression or the response to treatment (30). However, these methods lacked specificity and sensitivity. Therefore, a novel approach is required to accurately predict, diagnose and determine the prognosis of OC (14). Exosomes are among the most important biological components of liquid biopsy and possess a number of advantages over tumor-educated platelets (TEPs), circulating tumor DNA (ctDNA) and circulating tumor cells (CTCs). For instance, exosomes display exceptional stability within biological fluids, including plasma and urine (31). The immune conditions of the patient and different treatments can affect the RNA content of TEPs. Additionally, both CTCs and ctDNA have brief half-lives and are unstable, making sample collection challenging (32,33).

Exosomal ncRNAs

The term ‘liquid biopsy’ was first proposed in 2010 when researchers used CTCs to enhance breast cancer treatment and prognosis (34). Initially, the use of liquid biopsies focused on identifying CTCs and ctDNA in patients with cancer. However, components of CTCs and ctDNA are scarce in circulating biofluids, making them inappropriate for use as clinical diagnostic biomarkers. Conversely, other molecules secreted from cancer cells (such as ncRNAs) are more abundant than CTCs or ctDNA and are relatively stable in circulating biofluids (35). In recent decades, exosomes have been used to develop novel biomarkers, a primary goal of translational research (26,27). Exosomes are abundant and widely distributed in human bodily fluids, such as serum, saliva and urine. Exosomes carry various biomolecules, including proteins, lipids and nucleic acids, from their parent cells, making them accessible and potential biomarkers for tumor diagnosis, prognosis and monitoring (36). Additionally, exosomes are crucial for tumor development, spread and therapy resistance, particularly chemotherapy, by transferring their contents (37). ncRNAs are pivotal exosome components; exosomal ncRNAs, including miRNAs, lncRNAs and circRNAs are specific biomarkers for cancer diagnosis, prognosis, prediction and monitoring (38). With the advancements in transcriptomic analysis, ncRNAs have been shown to affect and participate in various aspects of OC (39), including cancer progression, metastasis and drug resistance (40).

miRNAs
Characteristics and biological functions of miRNAs

In 1993, Lee et al (41) first reported on miRNAs while investigating the developmental timing of Caenorhabditis elegans. miRNAs are stable RNA forms found in a number of body fluids, such as blood, urine and saliva (42). There are two types of extracellular miRNAs in biological fluids: One that departs through vesicles, such as apoptotic bodies, microvesicles and exosomes, while the other is attached to proteins, particularly argonaute 2 (AGO2) (43). At present, miRNAs are the most researched ncRNA, which may inform liquid biopsies one day.

The length of a single-stranded miRNA is 18–25 nucleotides (44), and some miRNAs might be protected against degradation by ribonucleases via the microRNA-induced silencing complex (miRISC) (45). There are two RNase III proteins, Drosha and Dicer, which produce miRNAs. The biogenesis of miRNAs is regulated at various levels, including transcription, Drosha and Dicer processing in the nucleus and cytoplasm, RNA editing, methylation, uridylation, adenylation, AGO loading and RNA decay (46). miRNA biogenesis occurs via two pathways: The canonical pathway, which is dominant, and the non-canonical pathway (43). Synthesis of the first miRNA (pri-miRNA) transcript initiates the canonical biogenesis pathway. Drosha and DiGeorge syndrome critical region 8 are components of the microprocessor complex that processes primary miRNA transcripts (47). The cleavage of pri-miRNA by the microprocessor complex generates the precursor-miRNA (pre-miRNA). Subsequently, pre-miRNAs are transferred to the cytoplasm by the exportin 5/RanGTP complex, where they are converted into mature miRNA duplexes (48). To create a miRISC, the mature miRNA duplex, with either a 5p or 3p strand, is combined with a member of the AGO family (49). miRNAs typically bind to a complementary sequence in the 3′-untranslated region (3′-UTR) of an mRNA. The primary biological role of miRNAs is to regulate the expression of target mRNA, by inhibiting mRNA expression or via mRNA degradation (50). Furthermore, miRNAs interact with additional areas, including gene promoters, the coding sequence and the 5′-UTR (51). Further investigations are necessary to fully comprehend the functional relationship between miRNAs and their target genes. Such strategies that could be used include high-throughput sequencing, bioinformatics and computational tools, RNA immunoprecipitation sequencing, cross-linking and immunoprecipitation sequencing and chromatin isolation by RNA purification. Based on these techniques, researchers will be able to identify the precise binding sites on their targets and determine how miRNAs impact cellular processes and disease states.

In general, miRISCs induce translational inhibition by binding to target mRNA. Then, P-body (GW182) family proteins binding to AGO are recruited, providing a scaffold to recruit downstream effector proteins such as poly(A)-deadenylases, PAN2/3, and the glucose-repressible alcohol dehydrogenase transcriptional effector, carbon catabolite repression 4-negative on TATA-less (52). Ultimately, the chromatin-binding exonuclease, 5′-3′ exoribonuclease 1, degrades the decapped mRNA (53,54). While most research has focused on miRNAs that prevent target gene expression, other studies have revealed that certain miRNAs can increase gene expression. For instance, AGO2 and FMR1 autosomal homolog 1 are linked to certain miRNAs, such as let-7, which stimulate translation during cell cycle arrest (55).

miRNAs in OC

miRNAs are divided into two main categories based on their target genes: Tumor suppressor miRNAs and oncogenic miRNAs (56). Tumor suppressor miRNAs silence uncontrolled genetic proliferation, thereby preventing cancer progression; tumorigenesis may occur when these miRNAs are downregulated in tumor cells (57). Oncogenic miRNAs increase oncogene expression and promote tumor development; tumorigenesis may occur when these miRNAs are upregulated in tumor cells (58).

Various miRNAs are associated with OC progression, diagnosis, prognosis and monitoring, which is summarized in Table II. miR-34a, which inhibits nearly 700 target genes, is a classical tumor suppressor (58). miR-34a can also encode a microprotein, termed miRPEP133, through an open reading frame (ORF) in the pri-miRNA-34a. A study overexpressed miRPEP133 in the SKOV3 OC cell line, finding that it may increase apoptosis and limit cell survival (59). Numerous studies have assessed the expression of miR-1246 in various cancer cell lines using various miRNA profiling techniques. The upregulation of miR-1246 has been noted in lung and liver tissues. Compared with normal colonic mucosa, the marked upregulation of miR-1246 in colorectal cancer tissues bolsters its status as a promising biomarker and suggests a role in the pathogenic processes of colorectal cancer (60–62). OC exosomes show significant levels of oncogenic miR-1246 expression (63). Based on data from The Cancer Genome Atlas, OC cells can be sensitized to paclitaxel in patients with OC receiving anti-miR-1246 therapy and exhibiting caveolin 1 (Cav1) upregulation. Moreover, miR-1246 inhibits Cav1 by acting on platelet derived growth factor receptor β receptor, preventing cell proliferation (64). miR-221 plays both oncogenic and tumor-suppressive roles in human cancer. miR-221 is capable of suppressing the proliferation of lung cancer cells, potentially through S phase arrest (65). miR-221 is upregulated in OC tissues compared with matched normal tissues. Furthermore, patients with high miR-221 expression have shorter overall survival (OS) and disease-free survival times than those with low expression. miR-221 targets protease activating factor 1 (APAF1), which induces apoptosis via gene inhibitors (66). Expressing higher levels of APAF1 by transfecting OC cell lines with miR-221 inhibitors prevents the proliferation, migration and invasion of cancer cells in vitro (66). According to a previous study, miR-27b-5p expression is lower in OC cell lines and tissues than in surrounding tissues. Furthermore, the OS rate was lower in patients with low miRNA-27b-5p expression. This study also verified that C-X-C motif chemokine ligand 1 (CXCL1), which is involved in OC malignancy, is a target gene of miR-27b-5p. Additionally, reverse transcription-quantitative PCR and immunoblotting analyses showed that the CXCL1 mRNA and protein levels were decreased in A2780 and OVCAR3 cells following miR-27b-5p overexpression (67).

Table II.

Summary of the miRNAs in ovarian cancer.

Table II.

Summary of the miRNAs in ovarian cancer.

First author/s, yearmiRNAExpressionPathway(Refs.)
Mo et al, 2023miR-141 (an exosomal miRNA)UpYAP1/GROα/CXCRs signaling cascade(123)
Knarr et al, 2020miR-181aUpRB1(147)
Wu et al, 2022miR-96-5pUpMTSS1/Wnt signaling(148)
Luo et al, 2018miR-10a-αUp-(149)
Ying et al, 2016miR-222-3p (an exosomal miRNA)UpSOCS3/STAT3(150)
Cappellesso et al, 2014miR-21 (an exosomal miRNA)UpPDCD4(151)
Meng et al, 2016miR-373, miR-200a, miR-200b and miR-200c (an exosomal miRNA)Up-(152)
Zavesky et al, 2016miR-92a (an exosomal miRNA)Up-(153)
Zhou et al, 2015miRNA-30a-5pUp-(154)
Hong et al, 2023miR-21-5p (an exosomal miRNA)Up-(155)
Zhang et al, 2019miR-9Up BRCA1/DNAJB6/KLF4/AKT1(156)
Li et al, 2023miR-1246, miR-141-3p, miR-203a-3p and miR-429Up-(157)
Yu et al, 2021miR-29DownlncRNA H19/miR-29b/(158)
COL1A1 axis
Liang et al, 2018miR-101-3pDownZEB1(84)
Li et al, 2024miR-136DownPIK3R1(115)
Záveský et al, 2015miR-106bDown-(159)
Zhou et al, 2015miR-636, miR-3170, miR-671-5p, miR-583, miR-6076, miR-3682-3p, miR-4688, miR-3646, miR-3621, miR-4497, miR-5195-3p, miR-4656, miR-5001-5p, miR-3679-5p, miR-5006-5p, miR-6087, miR-4532, miR-6510-5p, miR-4655-5p, miR-4459, miR-328, miR-4463, miR-3935, miR-3940-5p, miR-4745-5p, miR-3911, miR-4466, miR-4739, miR-638, miR-1224-5p, miR-4672, miR-6125, miR-4644, miR-197-3p, miR-6090, miR-2861 and miR-3188Down-(154)

[i] COL1A1, collagen type I α1 chain; CXCRs, CXC chemokine receptors; DNAJB6, DnaJ heat shock protein family (Hsp40) member B6; lncRNA, long non-coding RNA; miR/miRNA, microRNA; MTSS1, MTSS I-BAR domain containing 1; PDCD4, programmed cell death 4; RB1, RB transcriptional corepressor 1; SOCS3, suppressor of cytokine signaling 3; YAP1, yes-associated protein 1; ZEB1, zinc finger E-box binding homeobox 1.

In total, four molecular subtypes of OC, characterized by low overall survival rates, have been identified through extensive transcriptional profiling of cancer tissues: Immunoreactive, differentiated, proliferative and mesenchymal (68,69). To further investigate the different patterns of immune cell infiltration in OC, Liu et al (70) utilized the expression deconvolution algorithm, CIBERSORT, to confirm the distinct infiltration of immune cells in the four subtypes. Concentrated in plasma cells, the mesenchymal subtype of high-grade serous ovarian cancer (HGSOC) can induce the mesenchymal features of OC by releasing exosomes. Exosomal miRNA profiling revealed that plasma cell-derived miR-330-3p is an essential modulator of mesenchymal identity in OC. miR-330-3p increases the transcription of junctional adhesion molecule 2 through enhancer-induced gene activation pathways (71).

lncRNAs
Characteristics and biological functions of lncRNAs

lncRNAs were first described in 1990 (72). One of the earliest known lncRNA genes, the imprinting H19 gene, encodes a 2.3-kb ncRNA molecule, which aids in the development of a number of tumor types, including OC (72,73). Previously, lncRNAs were acknowledged as non-coding molecules. However, advances in proteomics and genetic technologies have revealed that a growing proportion of lncRNAs have small ORFs (sORFs). Certain sORFs have the ability to encode micropeptides or proteins that are essential for basic biological functions (74). Similar to miRNAs, RNA polymerase II is responsible for the conventional biosynthesis of lncRNAs. After transcription, the 5′ and 3′ ends of lncRNAs are independently capped and polyadenylated (75). Compared with protein coding genes, the overall sequence of lncRNAs frequently exhibits substantially lower evolutionary conservation (76).

The promoter regions of lncRNA genes are conserved, similar to those of protein-coding genes, and the production of lncRNAs controlled by these genes is tissue-specific (77). At present, different methods are used to classify lncRNAs, mainly depending on their function, interactions with protein-coding genes, structure, sequence and metabolism (78). lncRNAs can be categorized into four functional groups: Skeleton, guide, signal and bait molecules. In terms of interactions with protein-coding genes, lncRNAs fall into four categories: Overlapping, bidirectional, intron and cis-antisense (79). lncRNAs are crucial for various biological processes, including: i) Engaging with chromatin complexes to support the regulation of epigenetic genes; ii) acting as proteins or multiprotein compound modulators; iii) influencing transcriptional expression by binding proteins linked to DNA/RNA; iv) controlling the stability of DNA by forming triple helices and R-loops; and v) assisting in the creation of higher-order chromatin structures (80).

At present, RNAs are the main targets for therapeutic and diagnostic strategies. ncRNAs, for instance, show notable variety in nucleotide length and can form different structures when interacting with proteins, DNA and other substances. Notably, lncRNAs are more likely to form intricate secondary or tertiary structures due to their long nucleotide sequences. These structures, similar to enzymes, fulfill biological functions through their unique domains that show reproducibility and conservatism (81). The function of a number of proteins is determined by their distinct structural domains. However, current understanding of the structural domains that determine their functionality and define their interactome is lacking. Recently, various methods have been developed to assess the secondary structure of lncRNAs (82). These techniques are roughly divided into two types: Experimental and computational methods. Experimental techniques include enzymatic footprinting, chemical probing, nuclear magnetic resonance, small-angle scattering, atomic force microscopy and cryo-electron microscopy (81).

lncRNAs in OC

Various mechanisms have demonstrated that lncRNAs are involved in cancer progression. In the clinic, most OC cases have abdominal cavity metastases. Thus, the epithelial-to-mesenchymal transition (EMT) pathway serves a crucial role in tumor cell dissemination. The mechanisms by which lncRNAs mediate EMT in OC are diverse (83). For instance, through bioinformatics analysis, a previous study identified lncRNA pro-transition associated RNA as a lncRNA-mediated competing endogenous RNA (ceRNA) that competitively inhibits miR-101 to modulate the expression of zinc finger E-box binding homeobox 1 (84). A number of patients with OC develop carboplatin resistance. Chemotherapy-resistant strategies often involve multipotent mesenchymal stem cells or an increased EMT phenotype in cancer (85,86). In a clinical study, which included 134 primary OC cases (63 treated with carboplatin, 55 treated with cisplatin and 16 without therapy), expression of the lncRNA HOX transcript antisense RNA (HOTAIR) and the corresponding DNA methylation were detected. The research showed that patients with high HOTAIR expression receiving carboplatin had significantly lower survival rates, whereas this effect was not observed in patients who did not receive carboplatin (87). To reduce transcription, HOTAIR attracts polycomb repressive complex 2 (PRC2) to certain polycomb group target (PCGT) genes, particularly in embryonic fibroblasts (88). During differentiation, PCGTs essential for the identity of specialized cells are derepressed (89,90). However, promoters of these stem cell PCGTs are methylated and repressed in cancer (91). In several cancer types, high HOTAIR expression levels are strongly associated with metastasis, cancer invasiveness and poor prognosis (88,92).

The OC process is aided by ferroptosis, an iron-dependent type of cell death. One of the predominant methods used in cancer therapy involves triggering apoptosis to eradicate malignant cells. lncRNAs can regulate ferroptosis, a process that can trigger apoptosis and may therefore be beneficial in cancer treatment (93). A recent study found that lncRNA CACNA1G antisense RNA 1 promotes ferritin heavy chain 1 synthesis by reducing ferroptosis through insulin like growth factor 2 mRNA binding protein 1-mediated N6-methyladenosine (m6A) modification, which enhances the migration and division of OC cells (94). Additionally, another study showed that lncRNA ADAMTS9 antisense RNA 1 sponges miR-587 in OC, preventing ferroptosis and enhancing the expression of solute carrier family 7 member 11 (95).

Numerous studies have shown that lncRNAs can control the expression of tumor suppressor genes to either promote or prevent new tumor growth. Leukemia inhibitory factor receptor antisense RNA1 (LIFR-AS1), a tumor suppressor gene in colorectal cancer, was found to be downregulated in serous ovarian carcinoma (SOC) in a previous study. Generally, patients with SOC and low LIFR-AS1 expression had a poor prognosis. Low LIFR-AS1 expression was also associated with tumor size, clinical stage, lymph node metastasis and distant metastasis (96). LIFR-AS1 upregulation enhances the production of cleaved caspase-3 and E-cadherin, suppressing the malignant activities of SOC cells, including their migration, invasion and proliferation (96). lncRNAs can also repress tumor progression by regulating oncogene expression. For instance, long intergenic non-protein-coding RNA 857 (LINC00857), which inactivates the Hippo signaling pathway, can accelerate OC progression by regulating yes-associated protein 1 (YAP1), which acts as an oncogene in OC. LINC00857 competitively binds to miR-486-5p to regulate YAP1 expression, which elevates OC progression (97). A summary of the oncogenic lncRNAs in critical signaling pathways in OC is detailed in Table III.

Table III.

Oncogenic lncRNAs in critical signaling pathways in ovarian cancer.

Table III.

Oncogenic lncRNAs in critical signaling pathways in ovarian cancer.

lncRNAsPathway(Refs.)
DLEU1, HOTAIR, HOXD-AS1, LINC00511, NEAT1p53(160–164)
CCAT1, HOTAIRRAS(165,166)
CCAT1, DQ78624, EMX2OS, HAGLROS, HOTAIR, HOXD-AS1,PI3K/AKT/(165–169,161,170,163,171–173)
LINC00511, MALAT1+, NEAT1, PCAT-1mTOR
HOTAIR, NEAT1, SCAMP1, TDRG1NF-κB(174,164,175,176)
CCAT1, H19, HOTAIR, LINC00161, MIR4435-2HG, NEAT1TGF-β(177–180,172)
CCAT1, H19, HOTAIR, LINC00161, MALAT1+MAPK/ERK(177,178,166,179,181)
CCAT1, HOTAIR, LINC00511, MALAT1+, NEAT1, PCAT-1, TDRG1JAK/STAT(165,182,163,183,164,173,176)

[i] JAK, Janus kinase; lncRNA, long non-coding RNA; PI3K, phosphoinositide-3-kinase.

circRNAs
Characteristics and biological functions of circRNAs

When a tumor suppressor gene exon is spliced, circRNAs join in a different order from their genomic sequences. circRNAs were first described in 1976 (98). In 2015, high-throughput technologies were used to ascertain that circRNAs were two times more abundant in EVs than in parental cells (99). Premature mRNAs, which often do not encode proteins, can be alternatively spliced to become circRNAs, which are ncRNAs. circRNAs are more stable than other linear RNAs as they form covalently closed loops without 5′- or 3′-end features (100). This structure makes circRNAs resistant to degradation by classical RNA pathways, leading to their exocytosis from cells (101–103). circRNAs were initially thought to be non-functional byproducts of splicing mistakes. However, as high-throughput technologies and bioinformatics research progress, there is a growing body of evidence indicating that they exist widely in eukaryotic cells, where they regulate miRNAs and proteins to participate in various biological processes (104,105). Moreover, these circRNAs have been found to be stable and extremely conservative (99,106). circRNA expression in tumor tissues differs from that in normal tissues, indicating their involvement in tumorigenesis (107). circRNAs originate from various genomic regions, including intergenic, intronic, antisense and untranslated regions, and are categorized into three groups based on the region from which they are derived: Intronic, exon-intron and exonic (108). circRNAs have three major biological functions (109), including: i) Controlling the expression of target genes by competitively binding to miRNAs, similar to lncRNAs, acting as sponges for miRNAs or ceRNAs (110); ii) controlling the transcription, splicing, translation and interaction of genes with RNA-binding proteins to control the expression of genes (111); and iii) serving a role in polysomes, initiating putative ORFs and the AUG codon, and encoding regulatory peptides (112).

circRNAs in OC

circRNAs are involved in the development, metastasis and carcinogenesis of numerous human disorders (113,114). For instance, in platinum-resistant OC, the expression of circPLPP4 is notably elevated. Functionally, circPLPP4 operates as a molecular sponge for miR-136, effectively sequestering it. By doing so, circPLPP4 competitively increases the expression of PIK3R1, thereby contributing to resistance against cisplatin (115). Hsa_circ_0004712 exhibits abundant regulation in both OC tissues and cellular models, and functions by specifically targeting and reducing the expression of miR-331-3p. FZD4 was identified as a downstream target of miR-331-3p, experiencing repression when miR-331-3p levels were high, yet its expression was notably enhanced in the presence of hsa_circ_0004712, illustrating a regulatory axis between the circular RNA, microRNA and target gene (116). A summary of the oncogenic circRNAs in critical signaling pathways in OC is detailed in Table IV.

Table IV.

Summary of the circRNAs in ovarian cancer.

Table IV.

Summary of the circRNAs in ovarian cancer.

First author/s, yearcircRNAExpressionPathway(Refs.)
Li et al, 2024circPLPP4UpmiR-136/PIK3R1(115)
An et al, 2020circKRT7Up miR-29a-3p/COL1A1(184)
Wang et al, 2023circATP2B4Up miR-532-3p/SREBF1/PI3Kα/AK(185)
Wu et al, 2023circ_0010467Up miR-637/LIF/STAT3(186)
Li et al, 2022circITGB6Up GF2BP2/FGF9/RNA-PROTEIN(187)
Wu et al, 2022circFBXO7Down miR-96-5p/MTSS1(148)
Luo et al, 2018circ-ITCHDownmiR-10a-α(149)
Zou et al, 2018circLARP4Down-(188)
Zhang et al, 2019circPLEKHM3Down miR-9/BRCA1/DNAJB6/KLF4/AKT1(156)

[i] circRNA, circular RNA.

Relationship between the tumor microenvironment (TME) and exosomes

The majority of the tumor mass consists of the TME, which is mostly composed of tumor stroma (117). Stem cells, fibroblasts, endothelial cells, immune cells and numerous other cells that produce growth factors and cytokines are found in the TME (118). EVs are essential for communication between stromal and tumor cells in both nearby and distant microenvironments. EVs support tumor growth and establish a complex microenvironment called the pre-metastatic niche (PMN). The PMN has been primed by the primary tumor in distant organs or regions free of tumors, anticipating metastasis spread (119). The characteristic features of the PMN include extracellular matrix remodeling and deposition, lymphangiogenesis, vascular permeability, angiogenesis and immunological suppression (120).

Among the deadliest gynecological cancers, OC frequently results in peritoneal metastases, leading to higher recurrence rates and resistance to standard platinum-based treatments, which can result in a poor prognosis (121,122). Metastatic cancer cells spread to secondary sites, where they find a supportive TME owing to the biological components of the host. For instance, OC-generated EVs target YAP1, a key effector of the Hippo pathway. This interaction reduces the nuclear YAP1/transcriptional co-activator with PDZ-binding motif protein ratio and increases CXCL1 production by stromal fibroblasts. Pro-inflammatory cytokines, such as CXCL1, play a crucial role in promoting tumor development and metastasis (123). This finding indicates that pro-inflammatory CXCL1 is extensively expressed in the TME of ovarian malignancies and has oncogenic-promoting properties (123). Exosomes derived from OC that contain activating transcription factor 2 and metastasis-associated protein 1 may enhance angiogenesis (124). Tumor-secreted EVs (TEVs) are taken up by macrophages and lymphocytes in animal models, significantly decreasing the cross-presentation of tumor antigens from dendritic cells, which in turn affects CD8+ T cell activity (125). Another study demonstrated that TEVs expressing programmed cell death ligand 1 reduced the number of CD8+ T cells that invaded the lymph nodes (126). Phosphatidylserine, which impedes T-cell activation by obstructing intracellular signaling cascades, is present in some exosomes from OC (127). To counteract the immunosuppressive effects, normal cells, particularly immune cells, also produce EVs. These findings highlight the critical role of EVs in controlling the immune system. For instance, exosomes release cytokines, respond to specific antigens and mitogens, stimulate or inhibit B cell antibody production and directly kill target cells (128). Cytotoxic EVs are also released by natural killer (NK) cells. Fais et al (129) provided the first evidence that NK-derived EVs are lethal to cancer cells. It was shown that NK cells obtained from healthy donors, both in the resting and active states, were capable of killing a range of cancer cell lines and extending their cytotoxicity to resting immune cells but not to activated cells. Another study has reported that NK-derived EVs exhibit characteristic protein markers that are typical of NK cells, showing a propensity to be selectively internalized by SKOV3 cells, and demonstrating cytotoxic effects against OC cells (130). FasL and perforin are expressed in NK-derived EVs. These proteins can induce target cell demise via two pathways: i) A conventional ligand/receptor engagement between the membrane-bound FasL of NK-derived EVs and the Fas receptor on the surface of the target cell and; ii) an atypical cytotoxic mechanism initiated upon the uptake of exosomes by the target cells, potentially involving the intracellular release of effector molecules, such as perforin (129).

EVs have become important mediators of immunological evasion and repression of complex interactions between tumors and normal cells. TEVs disrupt immunological functions and diminish the effectiveness of immunotherapy. Normal cells, particularly immune cells, release EVs to counteract these effects.

Discussion

Current clinical screening methods for OC include imaging and blood testing. Imaging techniques comprise positron emission tomography, magnetic resonance imaging, computed tomography and ultrasound screening. Serum tests primarily measure CA-125 and cancer antigen 19-9 (131). Common diagnostic methods for OC, such as pelvic examination, ultrasound screening and CA-125 measurement, lack sensitivity and specificity (132), and only 50–60% of individuals diagnosed with stage I or II OC exhibit elevated CA-125 levels (133). Therefore, the discovery of new biomarkers is urgently needed to improve detection rates. Patients with OC show specific miRNA expression patterns in exosomes (134). Thus, these patterns may serve as potential biomarkers for various cancer types, including OC. For instance, ~95 exosomal miRNAs are differentially expressed in patients with epithelial ovarian cancer (EOC) and healthy donors (135). Individuals with EOC exhibit significantly lower levels of miR-320d, miR-4479 and miR-6763-5p than healthy donors. According to a study that used receiver operating characteristic curves to assess the diagnostic efficiency of exosomal miRNAs, these three exosomal miRNAs have diagnostic potential (135). A study also reported that exosomal miR-200b and CA-125, which are used for EOC screening, are associated with OS. According to this study, miR-200b influences cell proliferation and apoptosis. Furthermore, exosomal miR-200b can distinguish patients with EOC from healthy individuals with a 64% sensitivity and 86% specificity (136). Additionally, the level of serum exosomal miR-34a can be used to differentiate patients with early-stage and advanced-stage OC, suggesting that miR-34 is a potential biomarker for OC (137).

Exosomal ncRNAs show promise as potential novel diagnostic biomarkers for OC and are currently in the preclinical stage of development. Numerous prior studies have demonstrated that exosomal ncRNAs can function as prognostic biomarkers for OC. For instance, exosomal metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) has been linked to more malignant tendencies and a late International Federation of Gynecology and Obstetrics stage of EOC. Based on nomogram modeling and multivariate survival analysis, serum exosomal MALAT1 may be a potential predictive biomarker for EOC (138). Zhang et al (139) demonstrated that OS was associated with high levels of exosomal miR-200b. Additionally, circFoxp1 is identified as an independent factor that can predict survival and disease recurrence in individuals with EOC, according to the International Union of Obstetrics and Gynecology (140). Unlike other human cancer types, OC preferably invades the peritoneal cavity, with a particular potency at interacting with different viscera inside the compartment (121). The exosomes of the primary ovarian tumor could prime the distant TME for an expedited metastatic invasion. Within the TME, exosomes control intercellular communication between tumor cells and the normal stroma, cancer-associated fibroblasts and local immune cells.

Conclusion

Globally, epithelial ovarian cancer leads to >185,000 fatalities annually. HGSOC, the predominant variant, contributes to ~60% of these deaths. Despite advancements in surgical techniques and chemotherapy protocols, the mortality rate for HGSOC has remained high over the past several decades (141). The primary cause is the lack of reliable biomarkers for early diagnosis. Exosomes, which contain numerous ncRNAs, have increasingly been recognized as key players in intercellular communication. ncRNAs, including circRNAs, lncRNAs and miRNAs, are crucial for tumor development. Exosomal ncRNAs can be packaged, secreted and transported by tumor and normal cells to influence each other and complete the establishment of OC. Exosomes from OC could stimulate the establishment of PMNs by immunosuppression, angiogenesis, stromal cell modification and oncogenic reprogramming to promote tumor metastasis and growth. Although exosomes and exosomal ncRNAs are innovative indicators for the prognosis and diagnosis of OC, several issues hinder their use in clinical settings. First, only a limited quantity of exosomes is present in biological fluids. Second, there is limited knowledge about the biological functions of ncRNAs. Despite these challenges, exosomes and ncRNAs are promising biomarkers for the identification and management of OC.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

XW was responsible for writing and revising the manuscript. MY participated in the literature review and provided feedback. JZ and YZ provided substantial contributions to conception and design. GL was responsible for revising the manuscript and providing financial support. Data authentication is not applicable. All the authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Authors' information

Dr Gencui Li ORCID: 0009-0004-1397-3204.

References

1 

Yang C, Kim HS, Song G and Lim W: The potential role of exosomes derived from ovarian cancer cells for diagnostic and therapeutic approaches. J Cell Physiol. 234:21493–21503. 2019. View Article : Google Scholar : PubMed/NCBI

2 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Stewart C, Ralyea C and Lockwood S: Ovarian cancer: An integrated review. Semin Oncol Nurs. 35:151–156. 2019. View Article : Google Scholar : PubMed/NCBI

4 

Koshiyama M, Matsumura N and Konishi I: Subtypes of ovarian cancer and ovarian cancer screening. Diagnostics (Basel). 7:122017. View Article : Google Scholar : PubMed/NCBI

5 

Lheureux S, Braunstein M and Oza AM: Epithelial ovarian cancer: Evolution of management in the era of precision medicine. CA Cancer J Clin. 69:280–304. 2019. View Article : Google Scholar : PubMed/NCBI

6 

Lisio MA, Fu L, Goyeneche A, Gao ZH and Telleria C: High-grade serous ovarian cancer: Basic sciences, clinical and therapeutic standpoints. Int J Mol Sci. 20:9522019. View Article : Google Scholar : PubMed/NCBI

7 

Sierra J, Marrugo-Ramirez J, Rodríguez-Trujillo R, Mir M and Samitier J: Sensor-integrated microfluidic approaches for liquid biopsies applications in early detection of cancer. Sensors (Basel). 20:13172020. View Article : Google Scholar : PubMed/NCBI

8 

Bellassai N, D'Agata R, Jungbluth V and Spoto G: Surface plasmon resonance for biomarker detection: Advances in non-invasive cancer diagnosis. Front Chem. 7:5702019. View Article : Google Scholar : PubMed/NCBI

9 

Miao M, Miao Y, Zhu Y, Wang J and Zhou H: Advances in exosomes as diagnostic and therapeutic biomarkers for gynaecological malignancies. Cancers (Basel). 14:47432022. View Article : Google Scholar : PubMed/NCBI

10 

Maurano MT, Humbert R, Rynes E, Thurman RE, Haugen E, Wang H, Reynolds AP, Sandstrom R, Qu H, Brody J, et al: Systematic localization of common disease-associated variation in regulatory DNA. Science. 337:1190–1195. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Morris KV and Mattick JS: The rise of regulatory RNA. Nat Rev Genet. 15:423–437. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Hauptman N and Glavač D: Long non-coding RNA in cancer. Int J Mol Sci. 14:4655–4669. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Lander ES, Linton LM, Birren B, Nusbaum C, Zody MC, Baldwin J, Devon K, Dewar K, Doyle M, FitzHugh W, et al: Initial sequencing and analysis of the human genome. Nature. 409:860–921. 2001. View Article : Google Scholar : PubMed/NCBI

14 

Beg A, Parveen R, Fouad H, Yahia ME and Hassanein AS: Role of different non-coding RNAs as ovarian cancer biomarkers. J Ovarian Res. 15:722022. View Article : Google Scholar : PubMed/NCBI

15 

Fu G, Brkić J, Hayder H and Peng C: MicroRNAs in human placental development and pregnancy complications. Int J Mol Sci. 14:5519–5544. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Weber JA, Baxter DH, Zhang S, Huang DY, Huang KH, Lee MJ, Galas DJ and Wang K: The microRNA spectrum in 12 body fluids. Clin Chem. 56:1733–1741. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Théry C: Exosomes: Secreted vesicles and intercellular communications. F1000 Biol Rep. 3:152011. View Article : Google Scholar : PubMed/NCBI

18 

Théry C, Zitvogel L and Amigorena S: Exosomes: Composition, biogenesis and function. Nat Rev Immunol. 2:569–579. 2002. View Article : Google Scholar : PubMed/NCBI

19 

Iyer MK, Niknafs YS, Malik R, Singhal U, Sahu A, Hosono Y, Barrette TR, Prensner JR, Evans JR, Zhao S, et al: The landscape of long noncoding RNAs in the human transcriptome. Nat Genet. 47:199–208. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Wang KC and Chang HY: Molecular mechanisms of long noncoding RNAs. Mol Cell. 43:904–914. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Guo X, Gao Y, Song Q, Wei J, Wu J, Dong J, Chen L, Xu S, Wu D, Yang X, et al: Early assessment of circulating exosomal lncRNA-GC1 for monitoring neoadjuvant chemotherapy response in gastric cancer. Int J Surg. 109:1094–1104. 2023. View Article : Google Scholar : PubMed/NCBI

22 

Obi P and Chen YG: The design and synthesis of circular RNAs. Methods. 196:85–103. 2021. View Article : Google Scholar : PubMed/NCBI

23 

Chen LL: The expanding regulatory mechanisms and cellular functions of circular RNAs. Nat Rev Mol Cell Biol. 21:475–490. 2020. View Article : Google Scholar : PubMed/NCBI

24 

Pan BT, Teng K, Wu C, Adam M and Johnstone RM: Electron microscopic evidence for externalization of the transferrin receptor in vesicular form in sheep reticulocytes. J Cell Biol. 101:942–948. 1985. View Article : Google Scholar : PubMed/NCBI

25 

Isaac R, Reis FCG, Ying W and Olefsky JM: Exosomes as mediators of intercellular crosstalk in metabolism. Cell Metab. 33:1744–1762. 2021. View Article : Google Scholar : PubMed/NCBI

26 

Qi Y, Xu R, Song C, Hao M, Gao Y, Xin M, Liu Q, Chen H, Wu X, Sun R, et al: A comprehensive database of exosome molecular biomarkers and disease-gene associations. Sci Data. 11:2102024. View Article : Google Scholar : PubMed/NCBI

27 

Kalluri R and McAndrews KM: The role of extracellular vesicles in cancer. Cell. 186:1610–1626. 2023. View Article : Google Scholar : PubMed/NCBI

28 

Mannelli C: Tissue vs liquid biopsies for cancer detection: Ethical issues. J Bioeth Inq. 16:551–557. 2019. View Article : Google Scholar : PubMed/NCBI

29 

Xie H and Kim RD: The application of circulating tumor DNA in the screening, surveillance, and treatment monitoring of colorectal cancer. Ann Surg Oncol. 28:1845–1858. 2021. View Article : Google Scholar : PubMed/NCBI

30 

Yang WL, Lu Z, Guo J, Fellman BM, Ning J, Lu KH, Menon U, Kobayashi M, Hanash SM, Celestino J, et al: Human epididymis protein 4 antigen-autoantibody complexes complement cancer antigen 125 for detecting early-stage ovarian cancer. Cancer. 126:725–736. 2020. View Article : Google Scholar : PubMed/NCBI

31 

Yu W, Hurley J, Roberts D, Chakrabortty SK, Enderle D, Noerholm M, Breakefield XO and Skog JK: Exosome-based liquid biopsies in cancer: Opportunities and challenges. Ann Oncol. 32:466–477. 2021. View Article : Google Scholar : PubMed/NCBI

32 

Kalluri R: The biology and function of exosomes in cancer. J Clin Invest. 126:1208–1215. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Spina V and Rossi D: Liquid biopsy in tissue-born lymphomas. Swiss Med Wkly. 149:w147092019.PubMed/NCBI

34 

Lianidou ES, Mavroudis D, Sotiropoulou G, Agelaki S and Pantel K: What's new on circulating tumor cells? A meeting report. Breast Cancer Res. 12:3072010. View Article : Google Scholar : PubMed/NCBI

35 

Shigeyasu K, Toden S, Zumwalt TJ, Okugawa Y and Goel A: Emerging role of MicroRNAs as liquid biopsy biomarkers in gastrointestinal cancers. Clin Cancer Res. 23:2391–2399. 2017. View Article : Google Scholar : PubMed/NCBI

36 

Ebrahimi N, Faghihkhorasani F, Fakhr SS, Moghaddam PR, Yazdani E, Kheradmand Z, Rezaei-Tazangi F, Adelian S, Mobarak H, Hamblin MR and Aref AR: Tumor-derived exosomal non-coding RNAs as diagnostic biomarkers in cancer. Cell Mol Life Sci. 79:5722022. View Article : Google Scholar : PubMed/NCBI

37 

Kalluri R and LeBleu VS: The biology, function, and biomedical applications of exosomes. Science. 367:eaau69772020. View Article : Google Scholar : PubMed/NCBI

38 

Li C, Ni YQ, Xu H, Xiang QY, Zhao Y, Zhan JK, He JY, Li S and Liu YS: Roles and mechanisms of exosomal non-coding RNAs in human health and diseases. Signal Transduct Target Ther. 6:3832021. View Article : Google Scholar : PubMed/NCBI

39 

Jacobs IJ, Menon U, Ryan A, Gentry-Maharaj A, Burnell M, Kalsi JK, Amso NN, Apostolidou S, Benjamin E, Cruickshank D, et al: Ovarian cancer screening and mortality in the UK collaborative trial of ovarian cancer screening (UKCTOCS): A randomised controlled trial. Lancet. 387:945–956. 2016. View Article : Google Scholar : PubMed/NCBI

40 

Anastasiadou E, Jacob LS and Slack FJ: Non-coding RNA networks in cancer. Nat Rev Cancer. 18:5–18. 2018. View Article : Google Scholar : PubMed/NCBI

41 

Lee RC, Feinbaum RL and Ambros V: The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 75:843–854. 1993. View Article : Google Scholar : PubMed/NCBI

42 

Gareev I, Beylerli O, Yang G, Sun J, Pavlov V, Izmailov A, Shi H and Zhao S: The current state of MiRNAs as biomarkers and therapeutic tools. Clin Exp Med. 20:349–359. 2020. View Article : Google Scholar : PubMed/NCBI

43 

O'Brien J, Hayder H, Zayed Y and Peng C: Overview of MicroRNA biogenesis, mechanisms of actions, and circulation. Front Endocrinol (Lausanne). 9:4022018. View Article : Google Scholar : PubMed/NCBI

44 

Toden S and Goel A: Non-coding RNAs as liquid biopsy biomarkers in cancer. Br J Cancer. 126:351–360. 2022. View Article : Google Scholar : PubMed/NCBI

45 

Zhang Z, Qin YW, Brewer G and Jing Q: MicroRNA degradation and turnover: Regulating the regulators. Wiley Interdiscip Rev RNA. 3:593–600. 2012. View Article : Google Scholar : PubMed/NCBI

46 

Ha M and Kim VN: Regulation of microRNA biogenesis. Nat Rev Mol Cell Biol. 15:509–524. 2014. View Article : Google Scholar : PubMed/NCBI

47 

Denli AM, Tops BB, Plasterk RHA, Ketting RF and Hannon GJ: Processing of primary microRNAs by the Microprocessor complex. Nature. 432:231–235. 2004. View Article : Google Scholar : PubMed/NCBI

48 

Han J, Lee Y, Yeom KH, Kim YK, Jin H and Kim VN: The Drosha-DGCR8 complex in primary microRNA processing. Genes Dev. 18:3016–3027. 2004. View Article : Google Scholar : PubMed/NCBI

49 

Yoda M, Kawamata T, Paroo Z, Ye X, Iwasaki S, Liu Q and Tomari Y: ATP-dependent human RISC assembly pathways. Nat Struct Mol Biol. 17:17–23. 2010. View Article : Google Scholar : PubMed/NCBI

50 

Ambros V: The functions of animal microRNAs. Nature. 431:350–355. 2004. View Article : Google Scholar : PubMed/NCBI

51 

Broughton JP, Lovci MT, Huang JL, Yeo GW and Pasquinelli AE: Pairing beyond the seed supports MicroRNA targeting specificity. Mol Cell. 64:320–333. 2016. View Article : Google Scholar : PubMed/NCBI

52 

Braun JE, Huntzinger E and Izaurralde E: The role of GW182 proteins in miRNA-mediated gene silencing. Adv Exp Med Biol. 768:147–163. 2013. View Article : Google Scholar : PubMed/NCBI

53 

Hayder H, O'Brien J, Nadeem U and Peng C: MicroRNAs: Crucial regulators of placental development. Reproduction. 155:R259–R271. 2018. View Article : Google Scholar : PubMed/NCBI

54 

Christie M, Boland A, Huntzinger E, Weichenrieder O and Izaurralde E: Structure of the PAN3 pseudokinase reveals the basis for interactions with the PAN2 deadenylase and the GW182 proteins. Mol Cell. 51:360–373. 2013. View Article : Google Scholar : PubMed/NCBI

55 

Vasudevan S and Steitz JA: AU-rich-element-mediated upregulation of translation by FXR1 and argonaute 2. Cell. 128:1105–1118. 2007. View Article : Google Scholar : PubMed/NCBI

56 

Peng Y and Croce CM: The role of MicroRNAs in human cancer. Signal Transduct Target Ther. 1:150042016. View Article : Google Scholar : PubMed/NCBI

57 

Zhang B, Pan X, Cobb GP and Anderson TA: microRNAs as oncogenes and tumor suppressors. Dev Biol. 302:1–12. 2007. View Article : Google Scholar : PubMed/NCBI

58 

Chou CH, Chang NW, Shrestha S, Hsu SD, Lin YL, Lee WH, Yang CD, Hong HC, Wei TY, Tu SJ, et al: miRTarBase 2016: Updates to the experimentally validated miRNA-target interactions database. Nucleic Acids Res. 44(D1): D239–D247. 2016. View Article : Google Scholar : PubMed/NCBI

59 

Kang M, Tang B, Li J, Zhou Z, Liu K, Wang R, Jiang Z, Bi F, Patrick D, Kim D, et al: Identification of miPEP133 as a novel tumor-suppressor microprotein encoded by miR-34a pri-miRNA. Mol Cancer. 19:1432020. View Article : Google Scholar : PubMed/NCBI

60 

Zhang WC, Chin TM, Yang H, Nga ME, Lunny DP, Lim EK, Sun LL, Pang YH, Leow YN, Malusay SR, et al: Tumour-initiating cell-specific miR-1246 and miR-1290 expression converge to promote non-small cell lung cancer progression. Nat Commun. 7:117022016. View Article : Google Scholar : PubMed/NCBI

61 

Chai S, Ng KY, Tong M, Lau EY, Lee TK, Chan KW, Yuan YF, Cheung TT, Cheung ST, Wang XQ, et al: Octamer 4/ microRNA-1246 signaling axis drives Wnt/β-catenin activation in liver cancer stem cells. Hepatology. 64:2062–2076. 2016. View Article : Google Scholar : PubMed/NCBI

62 

Cooks T, Pateras IS, Jenkins LM, Patel KM, Robles AI, Morris J, Forshew T, Appella E, Gorgoulis VG and Harris CC: Mutant p53 cancers reprogram macrophages to tumor supporting macrophages via exosomal miR-1246. Nat Commun. 9:7712018. View Article : Google Scholar : PubMed/NCBI

63 

Wang W, Jo H, Park S, Kim H, Kim SI, Han Y, Lee J, Seol A, Kim J, Lee M, et al: Integrated analysis of ascites and plasma extracellular vesicles identifies a miRNA-based diagnostic signature in ovarian cancer. Cancer Lett. 542:2157352022. View Article : Google Scholar : PubMed/NCBI

64 

Kanlikilicer P, Bayraktar R, Denizli M, Rashed MH, Ivan C, Aslan B, Mitra R, Karagoz K, Bayraktar E, Zhang X, et al: Exosomal miRNA confers chemo resistance via targeting Cav1/p-gp/M2-type macrophage axis in ovarian cancer. EBioMedicine. 38:100–112. 2018. View Article : Google Scholar : PubMed/NCBI

65 

Yamashita R, Sato M, Kakumu T, Hase T, Yogo N, Maruyama E, Sekido Y, Kondo M and Hasegawa Y: Growth inhibitory effects of miR-221 and miR-222 in non-small cell lung cancer cells. Cancer Med. 4:551–564. 2015. View Article : Google Scholar : PubMed/NCBI

66 

Li J, Li Q, Huang H, Li Y, Li L, Hou W and You Z: Overexpression of miRNA-221 promotes cell proliferation by targeting the apoptotic protease activating factor-1 and indicates a poor prognosis in ovarian cancer. Int J Oncol. 50:1087–1096. 2017. View Article : Google Scholar : PubMed/NCBI

67 

Liu CH, Jing XN, Liu XL, Qin SY, Liu MW and Hou CH: Tumor-suppressor miRNA-27b-5p regulates the growth and metastatic behaviors of ovarian carcinoma cells by targeting CXCL1. J Ovarian Res. 13:922020. View Article : Google Scholar : PubMed/NCBI

68 

Cancer Genome Atlas Research Network, . Integrated genomic analyses of ovarian carcinoma. Nature. 474:609–615. 2011. View Article : Google Scholar : PubMed/NCBI

69 

Tothill RW, Tinker AV, George J, Brown R, Fox SB, Lade S, Johnson DS, Trivett MK, Etemadmoghadam D, Locandro B, et al: Novel molecular subtypes of serous and endometrioid ovarian cancer linked to clinical outcome. Clin Cancer Res. 14:5198–5208. 2008. View Article : Google Scholar : PubMed/NCBI

70 

Liu R, Hu R, Zeng Y, Zhang W and Zhou HH: Tumour immune cell infiltration and survival after platinum-based chemotherapy in high-grade serous ovarian cancer subtypes: A gene expression-based computational study. EBioMedicine. 51:1026022020. View Article : Google Scholar : PubMed/NCBI

71 

Yang Z, Wang W, Zhao L, Wang X, Gimple RC, Xu L, Wang Y, Rich JN and Zhou S: Plasma cells shape the mesenchymal identity of ovarian cancers through transfer of exosome-derived microRNAs. Sci Adv. 7:eabb07372021. View Article : Google Scholar : PubMed/NCBI

72 

Brannan CI, Dees EC, Ingram RS and Tilghman SM: The product of the H19 gene may function as an RNA. Mol Cell Biol. 10:28–36. 1990. View Article : Google Scholar : PubMed/NCBI

73 

Zhao L, Sun W, Zheng A, Zhang Y, Fang C and Zhang P: Ginsenoside Rg3 suppresses ovarian cancer cell proliferation and invasion by inhibiting the expression of lncRNA H19. Acta Biochim Pol. 68:575–582. 2021.PubMed/NCBI

74 

Hartford CCR and Lal A: When long noncoding becomes protein coding. Mol Cell Biol. 40:e00528–19. 2020. View Article : Google Scholar : PubMed/NCBI

75 

Quinn JJ and Chang HY: Unique features of long non-coding RNA biogenesis and function. Nat Rev Genet. 17:47–62. 2016. View Article : Google Scholar : PubMed/NCBI

76 

Ransohoff JD, Wei Y and Khavari PA: The functions and unique features of long intergenic non-coding RNA. Nat Rev Mol Cell Biol. 19:143–157. 2018. View Article : Google Scholar : PubMed/NCBI

77 

Mattick JS, Amaral PP, Carninci P, Carpenter S, Chang HY, Chen LL, Chen R, Dean C, Dinger ME, Fitzgerald KA, et al: Long non-coding RNAs: Definitions, functions, challenges and recommendations. Nat Rev Mol Cell Biol. 24:430–447. 2023. View Article : Google Scholar : PubMed/NCBI

78 

St Laurent G, Wahlestedt C and Kapranov P: The Landscape of long noncoding RNA classification. Trends Genet. 31:239–251. 2015. View Article : Google Scholar : PubMed/NCBI

79 

Wang H, Meng Q, Qian J, Li M, Gu C and Yang Y: Review: RNA-based diagnostic markers discovery and therapeutic targets development in cancer. Pharmacol Ther. 234:1081232022. View Article : Google Scholar : PubMed/NCBI

80 

Kopp F and Mendell JT: Functional classification and experimental dissection of long noncoding RNAs. Cell. 172:393–407. 2018. View Article : Google Scholar : PubMed/NCBI

81 

Graf J and Kretz M: From structure to function: Route to understanding lncRNA mechanism. Bioessays. 42:e20000272020. View Article : Google Scholar : PubMed/NCBI

82 

Seetin MG and Mathews DH: RNA structure prediction: An overview of methods. Methods Mol Biol. 905:99–122. 2012. View Article : Google Scholar : PubMed/NCBI

83 

Lampropoulou DI, Papadimitriou M, Papadimitriou C, Filippou D, Kourlaba G, Aravantinos G and Gazouli M: The role of EMT-related lncRNAs in ovarian cancer. Int J Mol Sci. 24:100792023. View Article : Google Scholar : PubMed/NCBI

84 

Liang H, Yu T, Han Y, Jiang H, Wang C, You T, Zhao X, Shan H, Yang R, Yang L, et al: LncRNA PTAR promotes EMT and invasion-metastasis in serous ovarian cancer by competitively binding miR-101-3p to regulate ZEB1 expression. Mol Cancer. 17:1192018. View Article : Google Scholar : PubMed/NCBI

85 

Leung D, Price ZK, Lokman NA, Wang W, Goonetilleke L, Kadife E, Oehler MK, Ricciardelli C, Kannourakis G and Ahmed N: Platinum-resistance in epithelial ovarian cancer: An interplay of epithelial-mesenchymal transition interlinked with reprogrammed metabolism. J Transl Med. 20:5562022. View Article : Google Scholar : PubMed/NCBI

86 

Kralj J, Pernar Kovač M, Dabelić S, Polančec DS, Wachtmeister T, Köhrer K and Brozovic A: Transcriptome analysis of newly established carboplatin-resistant ovarian cancer cell model reveals genes shared by drug resistance and drug-induced EMT. Br J Cancer. 128:1344–1359. 2023. View Article : Google Scholar : PubMed/NCBI

87 

Teschendorff AE, Lee SH, Jones A, Fiegl H, Kalwa M, Wagner W, Chindera K, Evans I, Dubeau L, Orjalo A, et al: HOTAIR and its surrogate DNA methylation signature indicate carboplatin resistance in ovarian cancer. Genome Med. 7:1082015. View Article : Google Scholar : PubMed/NCBI

88 

Gupta RA, Shah N, Wang KC, Kim J, Horlings HM, Wong DJ, Tsai MC, Hung T, Argani P, Rinn JL, et al: Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature. 464:1071–1076. 2010. View Article : Google Scholar : PubMed/NCBI

89 

Bernstein BE, Mikkelsen TS, Xie X, Kamal M, Huebert DJ, Cuff J, Fry B, Meissner A, Wernig M, Plath K, et al: A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell. 125:315–326. 2006. View Article : Google Scholar : PubMed/NCBI

90 

Lee TI, Jenner RG, Boyer LA, Guenther MG, Levine SS, Kumar RM, Chevalier B, Johnstone SE, Cole MF, Isono K, et al: Control of developmental regulators by polycomb in human embryonic stem cells. Cell. 125:301–313. 2006. View Article : Google Scholar : PubMed/NCBI

91 

Teschendorff AE, Menon U, Gentry-Maharaj A, Ramus SJ, Weisenberger DJ, Shen H, Campan M, Noushmehr H, Bell CG, Maxwell AP, et al: Age-dependent DNA methylation of genes that are suppressed in stem cells is a hallmark of cancer. Genome Res. 20:440–446. 2010. View Article : Google Scholar : PubMed/NCBI

92 

Qiu JJ, Lin YY, Ye LC, Ding JX, Feng WW, Jin HY, Zhang Y, Li Q and Hua KQ: Overexpression of long non-coding RNA HOTAIR predicts poor patient prognosis and promotes tumor metastasis in epithelial ovarian cancer. Gynecol Oncol. 134:121–128. 2014. View Article : Google Scholar : PubMed/NCBI

93 

Yang S, Ji J, Wang M, Nie J and Wang S: Construction of ovarian cancer prognostic model based on the investigation of ferroptosis-related lncRNA. Biomolecules. 13:3062023. View Article : Google Scholar : PubMed/NCBI

94 

Jin Y, Qiu J, Lu X, Ma Y and Li G: LncRNA CACNA1G-AS1 up-regulates FTH1 to inhibit ferroptosis and promote malignant phenotypes in ovarian cancer cells. Oncol Res. 31:169–179. 2023. View Article : Google Scholar : PubMed/NCBI

95 

Cai L, Hu X, Ye L, Bai P, Jie Y and Shu K: Long non-coding RNA ADAMTS9-AS1 attenuates ferroptosis by Targeting microRNA-587/solute carrier family 7 member 11 axis in epithelial ovarian cancer. Bioengineered. 13:8226–8239. 2022. View Article : Google Scholar : PubMed/NCBI

96 

Liu F, Cao L, Zhang Y, Xia X and Ji Y: LncRNA LIFR-AS1 overexpression suppressed the progression of serous ovarian carcinoma. J Clin Lab Anal. 36:e254702022. View Article : Google Scholar : PubMed/NCBI

97 

Lin X, Feng D, Li P and Lv Y: LncRNA LINC00857 regulates the progression and glycolysis in ovarian cancer by modulating the Hippo signaling pathway. Cancer Med. 9:8122–8132. 2020. View Article : Google Scholar : PubMed/NCBI

98 

Sanger HL, Klotz G, Riesner D, Gross HJ and Kleinschmidt AK: Viroids are single-stranded covalently closed circular RNA molecules existing as highly base-paired rod-like structures. Proc Natl Acad Sci USA. 73:3852–3856. 1976. View Article : Google Scholar : PubMed/NCBI

99 

Li Y, Zheng Q, Bao C, Li S, Guo W, Zhao J, Chen D, Gu J, He X and Huang S: Circular RNA is enriched and stable in exosomes: A promising biomarker for cancer diagnosis. Cell Res. 25:981–984. 2015. View Article : Google Scholar : PubMed/NCBI

100 

Meng X, Li X, Zhang P, Wang J, Zhou Y and Chen M: Circular RNA: An emerging key player in RNA world. Brief Bioinform. 18:547–557. 2017.PubMed/NCBI

101 

Patop IL, Wüst S and Kadener S: Past, present, and future of circRNAs. EMBO J. 38:e1008362019. View Article : Google Scholar : PubMed/NCBI

102 

Yao T, Chen Q, Fu L and Guo J: Circular RNAs: Biogenesis, properties, roles, and their relationships with liver diseases. Hepatol Res. 47:497–504. 2017. View Article : Google Scholar : PubMed/NCBI

103 

Lasda E and Parker R: Circular RNAs co-precipitate with extracellular vesicles: A possible mechanism for circRNA clearance. PLoS One. 11:e01484072016. View Article : Google Scholar : PubMed/NCBI

104 

Ngo LH, Bert AG, Dredge BK, Williams T, Murphy V, Li W, Hamilton WB, Carey KT, Toubia J, Pillman KA, et al: Nuclear export of circular RNA. Nature. 627:212–220. 2024. View Article : Google Scholar : PubMed/NCBI

105 

Pisignano G, Michael DC, Visal TH, Pirlog R, Ladomery M and Calin GA: Going circular: History, present, and future of circRNAs in cancer. Oncogene. 42:2783–2800. 2023. View Article : Google Scholar : PubMed/NCBI

106 

Jeck WR, Sorrentino JA, Wang K, Slevin MK, Burd CE, Liu J, Marzluff WF and Sharpless NE: Circular RNAs are abundant, conserved, and associated with ALU repeats. RNA. 19:141–157. 2013. View Article : Google Scholar : PubMed/NCBI

107 

Shang Q, Yang Z, Jia R and Ge S: The novel roles of circRNAs in human cancer. Mol Cancer. 18:62019. View Article : Google Scholar : PubMed/NCBI

108 

Ye D, Gong M, Deng Y, Fang S, Cao Y, Xiang Y and Shen Z: Roles and clinical application of exosomal circRNAs in the diagnosis and treatment of malignant tumors. J Transl Med. 20:1612022. View Article : Google Scholar : PubMed/NCBI

109 

Su M, Xiao Y, Ma J, Tang Y, Tian B, Zhang Y, Li X, Wu Z, Yang D, Zhou Y, et al: Circular RNAs in cancer: Emerging functions in hallmarks, stemness, resistance and roles as potential biomarkers. Mol Cancer. 18:902019. View Article : Google Scholar : PubMed/NCBI

110 

Cortes R and Forner MJ: Circular RNAS: Novel biomarkers of disease activity in systemic lupus erythematosus? Clin Sci (Lond). 133:1049–1052. 2019. View Article : Google Scholar : PubMed/NCBI

111 

Liu CX and Chen LL: Circular RNAs: Characterization, cellular roles, and applications. Cell. 185:2016–2034. 2022. View Article : Google Scholar : PubMed/NCBI

112 

Lei M, Zheng G, Ning Q, Zheng J and Dong D: Translation and functional roles of circular RNAs in human cancer. Mol Cancer. 19:302020. View Article : Google Scholar : PubMed/NCBI

113 

Zhao Q, Liu J, Deng H, Ma R, Liao JY, Liang H, Hu J, Li J, Guo Z, Cai J, et al: Targeting mitochondria-located circRNA SCAR alleviates NASH via reducing mROS output. Cell. 183:76–93.e22. 2020. View Article : Google Scholar : PubMed/NCBI

114 

Huang D, Zhu X, Ye S, Zhang J, Liao J, Zhang N, Zeng X, Wang J, Yang B, Zhang Y, et al: Tumour circular RNAs elicit anti-tumour immunity by encoding cryptic peptides. Nature. 625:593–602. 2024. View Article : Google Scholar : PubMed/NCBI

115 

Li H, Lin R, Zhang Y, Zhu Y, Huang S, Lan J, Lu N, Xie C, He S and Zhang W: N6-methyladenosine-modified circPLPP4 sustains cisplatin resistance in ovarian cancer cells via PIK3R1 upregulation. Mol Cancer. 23:52024. View Article : Google Scholar : PubMed/NCBI

116 

Zhou X, Jiang J and Guo S: Hsa_circ_0004712 downregulation attenuates ovarian cancer malignant development by targeting the miR-331-3p/FZD4 pathway. J Ovarian Res. 14:1182021. View Article : Google Scholar : PubMed/NCBI

117 

Camuzard O, Santucci-Darmanin S, Carle GF and Pierrefite-Carle V: Autophagy in the crosstalk between tumor and microenvironment. Cancer Lett. 490:143–153. 2020. View Article : Google Scholar : PubMed/NCBI

118 

Ye J, Wu D, Wu P, Chen Z and Huang J: The cancer stem cell niche: Cross talk between cancer stem cells and their microenvironment. Tumour Biol. 35:3945–3951. 2014. View Article : Google Scholar : PubMed/NCBI

119 

Kaplan RN, Riba RD, Zacharoulis S, Bramley AH, Vincent L, Costa C, MacDonald DD, Jin DK, Shido K, Kerns SA, et al: VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature. 438:820–827. 2005. View Article : Google Scholar : PubMed/NCBI

120 

Yue M, Hu S, Sun H, Tuo B, Jia B, Chen C, Wang W, Liu J, Liu Y, Sun Z and Hu J: Extracellular vesicles remodel tumor environment for cancer immunotherapy. Mol Cancer. 22:2032023. View Article : Google Scholar : PubMed/NCBI

121 

Mei S, Chen X, Wang K and Chen Y: Tumor microenvironment in ovarian cancer peritoneal metastasis. Cancer Cell Int. 23:112023. View Article : Google Scholar : PubMed/NCBI

122 

Lorusso D, Ceni V, Daniele G, Salutari V, Pietragalla A, Muratore M, Nero C, Ciccarone F and Scambia G: Newly diagnosed ovarian cancer: Which first-line treatment? Cancer Treat Rev. 91:1021112020. View Article : Google Scholar : PubMed/NCBI

123 

Mo Y, Leung LL, Mak CSL, Wang X, Chan WS, Hui LMN, Tang HWM, Siu MKY, Sharma R, Xu D, et al: Tumor-secreted exosomal miR-141 activates tumor-stroma interactions and controls premetastatic niche formation in ovarian cancer metastasis. Mol Cancer. 22:42023. View Article : Google Scholar : PubMed/NCBI

124 

Yi H, Ye J, Yang XM, Zhang LW, Zhang ZG and Chen YP: High-grade ovarian cancer secreting effective exosomes in tumor angiogenesis. Int J Clin Exp Pathol. 8:5062–5070. 2015.PubMed/NCBI

125 

Leary N, Walser S, He Y, Cousin N, Pereira P, Gallo A, Collado-Diaz V, Halin C, Garcia-Silva S, Peinado H and Dieterich LC: Melanoma-derived extracellular vesicles mediate lymphatic remodelling and impair tumour immunity in draining lymph nodes. J Extracell Vesicles. 11:e121972022. View Article : Google Scholar : PubMed/NCBI

126 

Chen G, Huang AC, Zhang W, Zhang G, Wu M, Xu W, Yu Z, Yang J, Wang B, Sun H, et al: Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature. 560:382–386. 2018. View Article : Google Scholar : PubMed/NCBI

127 

Kelleher RJ Jr, Balu-Iyer S, Loyall J, Sacca AJ, Shenoy GN, Peng P, Iyer V, Fathallah AM, Berenson CS, Wallace PK, et al: Extracellular vesicles present in human ovarian tumor microenvironments induce a phosphatidylserine-dependent arrest in the T-cell signaling cascade. Cancer Immunol Res. 3:1269–1278. 2015. View Article : Google Scholar : PubMed/NCBI

128 

Arenaccio C, Chiozzini C, Columba-Cabezas S, Manfredi F, Affabris E, Baur A and Federico M: Exosomes from human immunodeficiency virus type 1 (HIV-1)-infected cells license quiescent CD4+ T lymphocytes to replicate HIV-1 through a Nef- and ADAM17-dependent mechanism. J Virol. 88:11529–11539. 2014. View Article : Google Scholar : PubMed/NCBI

129 

Lugini L, Cecchetti S, Huber V, Luciani F, Macchia G, Spadaro F, Paris L, Abalsamo L, Colone M, Molinari A, et al: Immune surveillance properties of human NK cell-derived exosomes. J Immunol. 189:2833–2842. 2012. View Article : Google Scholar : PubMed/NCBI

130 

Luo H, Zhou Y, Zhang J, Zhang Y, Long S, Lin X, Yang A, Duan J, Yang N, Yang Z, et al: NK cell-derived exosomes enhance the anti-tumor effects against ovarian cancer by delivering cisplatin and reactivating NK cell functions. Front Immunol. 13:10876892023. View Article : Google Scholar : PubMed/NCBI

131 

Shiao MS, Chang JM, Lertkhachonsuk AA, Rermluk N and Jinawath N: Circulating exosomal miRNAs as biomarkers in epithelial ovarian cancer. Biomedicines. 9:14332021. View Article : Google Scholar : PubMed/NCBI

132 

Buys SS, Partridge E, Black A, Johnson CC, Lamerato L, Isaacs C, Reding DJ, Greenlee RT, Yokochi LA, Kessel B, et al: Effect of screening on ovarian cancer mortality: The prostate, lung, colorectal and ovarian (PLCO) cancer screening randomized controlled trial. JAMA. 305:2295–2303. 2011. View Article : Google Scholar : PubMed/NCBI

133 

Moss EL, Hollingworth J and Reynolds TM: The role of CA125 in clinical practice. J Clin Pathol. 58:308–312. 2005. View Article : Google Scholar : PubMed/NCBI

134 

Yokoi A, Yoshioka Y, Hirakawa A, Yamamoto Y, Ishikawa M, Ikeda SI, Kato T, Niimi K, Kajiyama H, Kikkawa F and Ochiya T: A combination of circulating miRNAs for the early detection of ovarian cancer. Oncotarget. 8:89811–89823. 2017. View Article : Google Scholar : PubMed/NCBI

135 

Wang S and Song X, Wang K, Zheng B, Lin Q, Yu M, Xie L, Chen L and Song X: Plasma exosomal miR-320d, miR-4479, and miR-6763-5p as diagnostic biomarkers in epithelial ovarian cancer. Front Oncol. 12:9863432022. View Article : Google Scholar : PubMed/NCBI

136 

Pan C, Stevic I, Müller V, Ni Q, Oliveira-Ferrer L, Pantel K and Schwarzenbach H: Exosomal microRNAs as tumor markers in epithelial ovarian cancer. Mol Oncol. 12:1935–1948. 2018. View Article : Google Scholar : PubMed/NCBI

137 

Maeda K, Sasaki H, Ueda S, Miyamoto S, Terada S, Konishi H, Kogata Y, Ashihara K, Fujiwara S, Tanaka Y, et al: Serum exosomal microRNA-34a as a potential biomarker in epithelial ovarian cancer. J Ovarian Res. 13:472020. View Article : Google Scholar : PubMed/NCBI

138 

Qiu JJ, Lin XJ, Tang XY, Zheng TT, Lin YY and Hua KQ: Exosomal metastasis-associated lung adenocarcinoma transcript 1 promotes angiogenesis and predicts poor prognosis in epithelial ovarian cancer. Int J Biol Sci. 14:1960–1973. 2018. View Article : Google Scholar : PubMed/NCBI

139 

Zhang W, Su X, Li S, Liu Z, Wang Q and Zeng H: Low serum exosomal miR-484 expression predicts unfavorable prognosis in ovarian cancer. Cancer Biomark. 27:485–491. 2020. View Article : Google Scholar : PubMed/NCBI

140 

Luo Y and Gui R: Circulating exosomal circFoxp1 confers cisplatin resistance in epithelial ovarian cancer cells. J Gynecol Oncol. 31:e752020. View Article : Google Scholar : PubMed/NCBI

141 

Chowdhury S, Kennedy JJ, Ivey RG, Murillo OD, Hosseini N, Song X, Petralia F, Calinawan A, Savage SR, Berry AB, et al: Proteogenomic analysis of chemo-refractory high-grade serous ovarian cancer. Cell. 186:3476–3498.e35. 2023. View Article : Google Scholar : PubMed/NCBI

142 

Martínez-Greene JA, Gómez-Chavarín M, Ramos-Godínez MDP and Martínez-Martínez E: Isolation of hepatic and adipose-tissue-derived extracellular vesicles using density gradient separation and size exclusion chromatography. Int J Mol Sci. 24:127042023. View Article : Google Scholar : PubMed/NCBI

143 

Bai HH, Wang XF, Zhang BY and Liu W: A comparison of size exclusion chromatography-based tandem strategies for plasma exosome enrichment and proteomic analysis. Anal Methods. 15:6245–6251. 2023. View Article : Google Scholar : PubMed/NCBI

144 

Takov K, Yellon DM and Davidson SM: Comparison of small extracellular vesicles isolated from plasma by ultracentrifugation or size-exclusion chromatography: Yield, purity and functional potential. J Extracell Vesicles. 8:15608092018. View Article : Google Scholar : PubMed/NCBI

145 

Tian Y, Gong M, Hu Y, Liu H, Zhang W, Zhang M, Hu X, Aubert D, Zhu S, Wu L and Yan X: Quality and efficiency assessment of six extracellular vesicle isolation methods by nano-flow cytometry. J Extracell Vesicles. 9:16970282019. View Article : Google Scholar : PubMed/NCBI

146 

Aliakbari F, Stocek NB, Cole-André M, Gomes J, Fanchini G, Pasternak SH, Christiansen G, Morshedi D, Volkening K and Strong MJ: A methodological primer of extracellular vesicles isolation and characterization via different techniques. Biol Methods Protoc. 9:bpae0092024. View Article : Google Scholar : PubMed/NCBI

147 

Knarr M, Avelar RA, Sekhar SC, Kwiatkowski LJ, Dziubinski ML, McAnulty J, Skala S, Avril S, Drapkin R and DiFeo A: miR-181a initiates and perpetuates oncogenic transformation through the regulation of innate immune signaling. Nat Commun. 11:32312020. View Article : Google Scholar : PubMed/NCBI

148 

Wu M, Qiu Q, Zhou Q, Li J, Yang J, Zheng C, Luo A, Li X, Zhang H, Cheng X, et al: circFBXO7/miR-96-5p/MTSS1 axis is an important regulator in the Wnt signaling pathway in ovarian cancer. Mol Cancer. 21:1372022. View Article : Google Scholar : PubMed/NCBI

149 

Luo L, Gao YQ and Sun XF: Circular RNA ITCH suppresses proliferation and promotes apoptosis in human epithelial ovarian cancer cells by sponging miR-10a-α. Eur Rev Med Pharmacol Sci. 22:8119–8126. 2018.PubMed/NCBI

150 

Ying X, Wu Q, Wu X, Zhu Q and Wang X, Jiang L, Chen X and Wang X: Epithelial ovarian cancer-secreted exosomal miR-222-3p induces polarization of tumor-associated macrophages. Oncotarget. 7:43076–43087. 2016. View Article : Google Scholar : PubMed/NCBI

151 

Cappellesso R, Tinazzi A, Giurici T, Simonato F, Guzzardo V, Ventura L, Crescenzi M, Chiarelli S and Fassina A: Programmed cell death 4 and microRNA 21 inverse expression is maintained in cells and exosomes from ovarian serous carcinoma effusions. Cancer Cytopathol. 122:685–693. 2014. View Article : Google Scholar : PubMed/NCBI

152 

Meng X, Müller V, Milde-Langosch K, Trillsch F, Pantel K and Schwarzenbach H: Diagnostic and prognostic relevance of circulating exosomal miR-373, miR-200a, miR-200b and miR-200c in patients with epithelial ovarian cancer. Oncotarget. 7:16923–16935. 2016. View Article : Google Scholar : PubMed/NCBI

153 

Zavesky L, Jandakova E, Turyna R, Langmeierova L, Weinberger V and Minar L: Supernatant versus exosomal urinary microRNAs. Two fractions with different outcomes in gynaecological cancers. Neoplasma. 63:121–132. 2016. View Article : Google Scholar : PubMed/NCBI

154 

Zhou J, Gong G, Tan H, Dai F, Zhu X, Chen Y, Wang J, Liu Y, Chen P, Wu X and Wen J: Urinary microRNA-30a-5p is a potential biomarker for ovarian serous adenocarcinoma. Oncol Rep. 33:2915–2923. 2015. View Article : Google Scholar : PubMed/NCBI

155 

Hong JS, Son T, Castro CM and Im H: CRISPR/Cas13a-based MicroRNA detection in tumor-derived extracellular vesicles. Adv Sci (Weinh). 10:e23017662023. View Article : Google Scholar : PubMed/NCBI

156 

Zhang L, Zhou Q, Qiu Q, Hou L, Wu M, Li J, Li X, Lu B, Cheng X, Liu P, et al: CircPLEKHM3 acts as a tumor suppressor through regulation of the miR-9/BRCA1/DNAJB6/KLF4/AKT1 axis in ovarian cancer. Mol Cancer. 18:1442019. View Article : Google Scholar : PubMed/NCBI

157 

Li L, Zhang F, Zhang J, Shi X, Wu H, Chao X, Ma S, Lang J, Wu M, Zhang D and Liang Z: Identifying serum small extracellular vesicle MicroRNA as a noninvasive diagnostic and prognostic biomarker for ovarian cancer. ACS Nano. 17:19197–19210. 2023. View Article : Google Scholar : PubMed/NCBI

158 

Yu CC, Liao YW, Hsieh PL and Chang YC: Targeting lncRNA H19/miR-29b/COL1A1 axis impedes myofibroblast activities of precancerous oral submucous fibrosis. Int J Mol Sci. 22:22162021. View Article : Google Scholar : PubMed/NCBI

159 

Záveský L, Jandáková E, Turyna R, Langmeierová L, Weinberger V, Záveská Drábková L, Hůlková M, Hořínek A, Dušková D, Feyereisl J, et al: Evaluation of cell-free urine microRNAs expression for the use in diagnosis of ovarian and endometrial cancers. A pilot study. Pathol Oncol Res. 21:1027–1035. 2015. View Article : Google Scholar : PubMed/NCBI

160 

Wang LL, Sun KX, Wu DD, Xiu YL, Chen X, Chen S, Zong ZH, Sang XB, Liu Y and Zhao Y: DLEU1 contributes to ovarian carcinoma tumourigenesis and development by interacting with miR-490-3p and altering CDK1 expression. J Cell Mol Med. 21:3055–3065. 2017. View Article : Google Scholar : PubMed/NCBI

161 

Dong L and Hui L: HOTAIR promotes proliferation, migration, and invasion of ovarian cancer SKOV3 cells through regulating PIK3R3. Med Sci Monit. 22:325–331. 2016. View Article : Google Scholar : PubMed/NCBI

162 

Zhang Y, Dun Y, Zhou S and Huang XH: LncRNA HOXD-AS1 promotes epithelial ovarian cancer cells proliferation and invasion by targeting miR-133a-3p and activating Wnt/β-catenin signaling pathway. Biomed Pharmacother. 96:1216–1221. 2017. View Article : Google Scholar : PubMed/NCBI

163 

Wang J, Tian Y, Zheng H, Ding Y and Wang X: An integrated analysis reveals the oncogenic function of lncRNA LINC00511 in human ovarian cancer. Cancer Med. 8:3026–3035. 2019. View Article : Google Scholar : PubMed/NCBI

164 

An J, Lv W and Zhang Y: LncRNA NEAT1 contributes to paclitaxel resistance of ovarian cancer cells by regulating ZEB1 expression via miR-194. Onco Targets Ther. 10:5377–5390. 2017. View Article : Google Scholar : PubMed/NCBI

165 

Cao Y, Shi H, Ren F, Jia Y and Zhang R: Long non-coding RNA CCAT1 promotes metastasis and poor prognosis in epithelial ovarian cancer. Exp Cell Res. 359:185–194. 2017. View Article : Google Scholar : PubMed/NCBI

166 

Yiwei T, Hua H, Hui G, Mao M and Xiang L: HOTAIR interacting with MAPK1 regulates ovarian cancer skov3 cell proliferation, migration, and invasion. Med Sci Monit. 21:1856–1863. 2015. View Article : Google Scholar : PubMed/NCBI

167 

Yan H, Silva MA, Li H, Zhu L, Li P, Li X, Wang X, Gao J, Wang P and Zhang Z: Long noncoding RNA DQ786243 interacts with miR-506 and promotes progression of ovarian cancer through targeting cAMP responsive element binding protein 1. J Cell Biochem. 119:9764–9780. 2018. View Article : Google Scholar : PubMed/NCBI

168 

Duan M, Fang M, Wang C, Wang H and Li M: LncRNA EMX2OS induces proliferation, invasion and sphere formation of ovarian cancer cells via regulating the miR-654-3p/AKT3/PD-L1 axis. Cancer Manag Res. 12:2141–2154. 2020. View Article : Google Scholar : PubMed/NCBI

169 

Yang M, Zhai Z, Zhang Y and Wang Y: Clinical significance and oncogene function of long noncoding RNA HAGLROS overexpression in ovarian cancer. Arch Gynecol Obstet. 300:703–710. 2019. View Article : Google Scholar : PubMed/NCBI

170 

Wang Y, Zhang W, Wang Y and Wang S: HOXD-AS1 promotes cell proliferation, migration and invasion through miR-608/FZD4 axis in ovarian cancer. Am J Cancer Res. 8:170–182. 2018.PubMed/NCBI

171 

Jin Y, Feng SJ, Qiu S, Shao N and Zheng JH: LncRNA MALAT1 promotes proliferation and metastasis in epithelial ovarian cancer via the PI3K-AKT pathway. Eur Rev Med Pharmacol Sci. 21:3176–3184. 2017.PubMed/NCBI

172 

Liu Y, Wang Y, Fu X and Lu Z: Long non-coding RNA NEAT1 promoted ovarian cancer cells' metastasis through regulation of miR-382-3p/ROCK1 axial. Cancer Sci. 109:2188–2198. 2018. View Article : Google Scholar : PubMed/NCBI

173 

Ding C, Wei R, Rodríguez RA and Del Mar Requena Mullor M: LncRNA PCAT-1 plays an oncogenic role in epithelial ovarian cancer by modulating cyclinD1/CDK4 expression. Int J Clin Exp Pathol. 12:2148–2156. 2019.PubMed/NCBI

174 

Özeş AR, Miller DF, Özeş ON, Fang F, Liu Y, Matei D, Huang T and Nephew KP: NF-κB-HOTAIR axis links DNA damage response, chemoresistance and cellular senescence in ovarian cancer. Oncogene. 35:5350–5361. 2016. View Article : Google Scholar : PubMed/NCBI

175 

Song R, Liu Z, Lu L, Liu F and Zhang B: Long noncoding RNA SCAMP1 targets miR-137/CXCL12 axis to boost cell invasion and angiogenesis in ovarian cancer. DNA Cell Biol. 39:1041–1050. 2020. View Article : Google Scholar : PubMed/NCBI

176 

Chen S, Wang LL, Sun KX, Xiu YL, Zong ZH, Chen X and Zhao Y: The role of the long non-coding RNA TDRG1 in epithelial ovarian carcinoma tumorigenesis and progression through miR-93/RhoC pathway. Mol Carcinog. 57:225–234. 2018. View Article : Google Scholar : PubMed/NCBI

177 

Mu Y, Li N and Cui YL: The lncRNA CCAT1 upregulates TGFβR1 via sponging miR-490-3p to promote TGFβ1-induced EMT of ovarian cancer cells. Cancer Cell Int. 18:1452018. View Article : Google Scholar : PubMed/NCBI

178 

Li J, Li J, Huang Y, Deng X, Luo M, Wang X, Hu H, Liu C and Zhong M: Long noncoding RNA H19 promotes transforming growth factor-β-induced epithelial-mesenchymal transition by acting as a competing endogenous RNA of miR-370-3p in ovarian cancer cells. Onco Targets Ther. 11:427–440. 2018. View Article : Google Scholar : PubMed/NCBI

179 

Xu M, Zhou K, Wu Y, Wang L and Lu S: Linc00161 regulated the drug resistance of ovarian cancer by sponging microRNA-128 and modulating MAPK1. Mol Carcinog. 58:577–587. 2019. View Article : Google Scholar : PubMed/NCBI

180 

Pan L, Meng Q, Li H, Liang K and Li B: LINC00339 promotes cell proliferation, migration, and invasion of ovarian cancer cells via miR-148a-3p/ROCK1 axes. Biomed Pharmacother. 120:1094232019. View Article : Google Scholar : PubMed/NCBI

181 

Zou A, Liu R and Wu X: Long non-coding RNA MALAT1 is up-regulated in ovarian cancer tissue and promotes SK-OV-3 cell proliferation and invasion. Neoplasma. 63:865–872. 2016. View Article : Google Scholar : PubMed/NCBI

182 

Chang L, Guo R, Yuan Z, Shi H and Zhang D: LncRNA HOTAIR regulates CCND1 and CCND2 expression by sponging miR-206 in ovarian cancer. Cell Physiol Biochem. 49:1289–1303. 2018. View Article : Google Scholar : PubMed/NCBI

183 

Sun Q, Li Q and Xie F: LncRNA-MALAT1 regulates proliferation and apoptosis of ovarian cancer cells by targeting miR-503-5p. Onco Targets Ther. 12:6297–6307. 2019. View Article : Google Scholar : PubMed/NCBI

184 

An Q, Liu T, Wang MY, Yang YJ, Zhang ZD, Lin ZJ and Yang B: circKRT7-miR-29a-3p-COL1A1 axis promotes ovarian cancer cell progression. Onco Targets Ther. 13:8963–8976. 2020. View Article : Google Scholar : PubMed/NCBI

185 

Wang F, Niu Y, Chen K, Yuan X, Qin Y, Zheng F, Cui Z, Lu W, Wu Y and Xia D: Extracellular vesicle-packaged circATP2B4 mediates M2 macrophage polarization via miR-532-3p/SREBF1 axis to promote epithelial ovarian cancer metastasis. Cancer Immunol Res. 11:199–216. 2023. View Article : Google Scholar : PubMed/NCBI

186 

Wu Y, Xu M, Feng Z, Wu H, Wu J, Ha X, Wu Y, Chen S, Xu F, Wen H, et al: AUF1-induced circular RNA hsa_circ_0010467 promotes platinum resistance of ovarian cancer through miR-637/LIF/STAT3 axis. Cell Mol Life Sci. 80:2562023. View Article : Google Scholar : PubMed/NCBI

187 

Li H, Luo F, Jiang X, Zhang W, Xiang T, Pan Q, Cai L, Zhao J, Weng D, Li Y, et al: CircITGB6 promotes ovarian cancer cisplatin resistance by resetting tumor-associated macrophage polarization toward the M2 phenotype. J Immunother Cancer. 10:e0040292022. View Article : Google Scholar : PubMed/NCBI

188 

Zou T, Wang PL, Gao Y and Liang WT: Circular RNA_LARP4 is lower expressed and serves as a potential biomarker of ovarian cancer prognosis. Eur Rev Med Pharmacol Sci. 22:7178–7182. 2018.PubMed/NCBI

Related Articles

  • Abstract
  • View
  • Download
  • Twitter
Copy and paste a formatted citation
Spandidos Publications style
Wang X, Yang M, Zhu J, Zhou Y and Li G: Role of exosomal non‑coding RNAs in ovarian cancer (Review). Int J Mol Med 54: 87, 2024.
APA
Wang, X., Yang, M., Zhu, J., Zhou, Y., & Li, G. (2024). Role of exosomal non‑coding RNAs in ovarian cancer (Review). International Journal of Molecular Medicine, 54, 87. https://doi.org/10.3892/ijmm.2024.5411
MLA
Wang, X., Yang, M., Zhu, J., Zhou, Y., Li, G."Role of exosomal non‑coding RNAs in ovarian cancer (Review)". International Journal of Molecular Medicine 54.4 (2024): 87.
Chicago
Wang, X., Yang, M., Zhu, J., Zhou, Y., Li, G."Role of exosomal non‑coding RNAs in ovarian cancer (Review)". International Journal of Molecular Medicine 54, no. 4 (2024): 87. https://doi.org/10.3892/ijmm.2024.5411
Copy and paste a formatted citation
x
Spandidos Publications style
Wang X, Yang M, Zhu J, Zhou Y and Li G: Role of exosomal non‑coding RNAs in ovarian cancer (Review). Int J Mol Med 54: 87, 2024.
APA
Wang, X., Yang, M., Zhu, J., Zhou, Y., & Li, G. (2024). Role of exosomal non‑coding RNAs in ovarian cancer (Review). International Journal of Molecular Medicine, 54, 87. https://doi.org/10.3892/ijmm.2024.5411
MLA
Wang, X., Yang, M., Zhu, J., Zhou, Y., Li, G."Role of exosomal non‑coding RNAs in ovarian cancer (Review)". International Journal of Molecular Medicine 54.4 (2024): 87.
Chicago
Wang, X., Yang, M., Zhu, J., Zhou, Y., Li, G."Role of exosomal non‑coding RNAs in ovarian cancer (Review)". International Journal of Molecular Medicine 54, no. 4 (2024): 87. https://doi.org/10.3892/ijmm.2024.5411
Follow us
  • Twitter
  • LinkedIn
  • Facebook
About
  • Spandidos Publications
  • Careers
  • Cookie Policy
  • Privacy Policy
How can we help?
  • Help
  • Live Chat
  • Contact
  • Email to our Support Team