Open Access

Role of DNA methylation and non‑coding RNAs expression in pathogenesis, detection, prognosis, and therapy‑resistant ovarian carcinoma (Review)

  • Authors:
    • Victor M. Del Castillo Falconi
    • Jenny A. Godinez Rodriguez
    • Verónica Fragoso‑Ontiveros
    • Laura Contreras‑Espinosa
    • Abraham Pedroza‑Torres
    • José Díaz‑Chávez
    • Luis A. Herrera
  • View Affiliations

  • Published online on: April 1, 2025     https://doi.org/10.3892/mmr.2025.13509
  • Article Number: 144
  • Copyright: © Del Castillo Falconi et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Ovarian cancer is the deadliest gynecological cancer globally, with epithelial ovarian cancer (EOC) comprising up to 90% of cases. A molecular characterization linking the histological subtypes with tumor grade in EOC has been suggested. Variations in genetic biomarkers such as BRCA1/2, MSH2, MLH1/6, BRIP1, and RAD51C/D have been studied in EOC. In addition, molecular characteristics, including DNA methylation and RNA transcription, are being explored as potential new biomarkers for the diagnosis and prognosis of this type of neoplasia. The present review focused on the role of DNA methylation and non‑coding RNA expression in the development of ovarian carcinomas and their association with diagnosis, prognosis, and the resistance of cancer cells to radiotherapy and chemotherapy. The present review considered the transition from the DNA structure to the RNA expression in ovarian carcinoma.

Introduction

Ovary cancer (OC) is considered the most lethal malignancy among gynecological cancers. In 2020 worldwide, OC caused 1.6% of all new cancer-related deaths (1). Epithelial ovarian cancer (EOC) is a clinical type of OC that is already diagnosed in 90% of women patients with OC; patients with this OC type are typically diagnosed in the advanced stages of the disease (75%) when cancer has disseminated to a different abdominal tissue or metastases are present. The majority of patients (>70%) with advanced-stage OC do not respond to standard therapies, resulting in a resistant, fatal disease (2). Due to this, the identification and understanding of the molecular characteristics associated with early disease progression, prediction and clinical responses are necessary to improve survival and clinical treatment in women with EOC.

In this regard, differential cancer DNA methylation, compared with the origin tissue cells, is an early event during carcinogenesis. That is, DNA methylation is composed of concomitant global unmethylated DNA and local locus-methylated DNA. Methylated DNA consists of the addition of a methyl group in the fifth carbon of cytosine residues, forming a CpG; this addition forms 5-methylcytosine. Typical examples of DNA-methylation phenotypes have been characterized in certain types of cancer, such as colorectal carcinoma, breast carcinoma and glioma (38). Methylated DNA molecules are more compact than unmethylated DNA molecules (3). It has been proposed that unmethylated DNA is a characteristic of cancer while methylated DNA presents only a variable consequence depending on the locus and on the specific part of the locus in cancer cells (9,10). RNA transcription is strongly influenced by DNA methylation. Methylated loci are silenced and unmethylated loci are transcriptionally activated in ovarian carcinoma tumors. The diagnosis of patients is made by transvaginal ultrasound and detection of cancer antigen (CA)-125 levels; however, the state of DNA methylation and RNA expression in the tumors has been currently associated with the diagnosis of the histological subtypes in the different types of ovarian carcinoma, the advanced stages and, importantly, the survival of the patients.

EOC is a heterogeneous carcinoma type and every EOC subtype has its natural history of development. Specifically, cell subtypes are described by histopathological and molecular characteristics of ovarian carcinoma (1113). However, it is considered that serous ovarian carcinomas are developed as a disease continuum, from low-grade to high-grade serous ovarian carcinomas. Evidence suggests that high-grade and low-grade serous carcinomas develop independently in their natural course of progression and they have different prognoses (12,13).

At the molecular level, the prognosis of spontaneous EOC type I and type II is associated with the EOC sub-type, the age of the patient and the treatment used. Although advances in clinical treatments have increased in the past few decades, the pathology structure remains unaltered (14). Of patients diagnosed with ovarian cancer, ~50% survive five years following diagnosis, including 29% of those with metastases of ovarian carcinoma (1). In ovarian carcinomas, several factors determine survival, including primarily histological subtype, grade, stage, cytoreductive surgery and, secondarily, ethnicity (15,16).

The mortalities associated with ovarian cancer comprise 4% of all cancer-related deaths (1,14). Germinal variations are changes in DNA locus. Germinal variants of ovarian carcinomas are present in 5% of patients with ovarian carcinoma. Ovarian carcinomas are mutated in the following two ways: In germinal DNA (DNA variants of hereditary cancer origin) or in somatic DNA (DNA variants with individual spontaneous tumor cancer origin). Variants in the germ line DNA represent 24% of OC. The majority of the genetic variants are present in BRCA in hereditary breast and ovarian cancer syndrome (HBOC), whereas other DNA repair genes are present in Lynch syndrome (LS), also called hereditary non-polyposis colorectal cancer syndrome (17).

The loci mutations with higher hereditary penetrance to develop ovarian carcinoma are those of BRCA1 or BRCA2. HBOC accounts for ~80% of hereditary ovarian carcinoma and 15% of epithelial OC cases. In HBOC, 65–85% of cancers are due to genomic variants in BRCA1 and BRCA2 genes, which are considered high penetrance for OC (18). These genes encode proteins for homologous recombination to repair DNA double-strand breaks and maintain genomic stability. In addition, germinal carcinoma with BRCA1/2 mutations develops a high-grade serous ovarian carcinoma (HGSOC) subtype (15,16,19). Other loci mutations with moderate familial penetrance involve genes implicated in OC, such as BARD1, BRIP1, PALB2, RAD50, RAD51C, NBN and MRE11A; these mutations are encoded in each gene that has been involved in OC as part of the BRCA2/Fanconi anemia signaling pathway (20). Epithelial OC deficiency in DNA mismatch repair is the second most common cause of HBOC, accounting for 10–15% of this condition. The lifetime risk of developing OC with LS is ~8–12% and the mean age at presentation is ~43 years. OC-associated genes in this pathway are the following: MLH1, MSH2, MSH6 and PMS2. Specifically, BRIP1, MLH1, MSH2, MSH6, PMS2 and EPCAM are moderate penetrance genes in OC (1619).

It is notable that each mutated locus develops different characteristics in the phenotype of tumor cells present in the patients (1923). By contrast, EOCs developed from a somatic spontaneous origin are heterogeneous. At the cellular level, ovarian carcinoma tumors are classified into five different types: HGSOC, low-grade serous ovarian carcinoma (LGSOC), mucinous carcinoma (MC), clear cells carcinoma (CCC) and endometrioid carcinoma (EC). Recently, DNA methylation and RNA transcription have been shown to vary in a defined way to develop EOC tumors, such as hypomethylated DNA and variation in RNA expression. It is notable that germinal variation in the DNA locus of the large non-coding RNA (lncRNA) HOTAIR is a risk cause of developing ovarian carcinoma. In addition, overexpression of HOTAIR has been found in ovarian carcinomas and it has been associated with chemotherapy resistance (24).

Ovarian carcinoma, which is resistant to radiotherapy and chemotherapy is another important problem. For example, in HGSOC, the presence of the TP53 mutation and chromosome instability (CIN) are associated with resistance to radiotherapy and standard chemotherapy, which is a mix of carboplatin and taxane (25). In addition, it has been observed that DNA methylation loci induce sensitivity to therapies. By contrast, the DNA methylation loci of the nuclear RNA transcripts are associated with resistance to treatment. In this regard, it has been proposed that ovarian carcinoma cells could be sensitized to radiotherapy and chemotherapy by the addition of DNA methylation inhibitors such as decitabine (25).

DNA methylation in the diagnoses of ovarian carcinomas

The molecular characteristics validate the natural history of ovarian carcinoma tumors and explain the association between clinical characteristics of ovarian carcinoma, such as mutations in the expression levels of DNA, RNA and proteins with patient survival. The first characterization of ovarian carcinoma is performed by quantifying transvaginal CA-125 levels using ultrasonic waves (26). Subsequently, the characterization of the macroscopic tumors in surgery is required and finally the histological characteristics have to be defined by microscopic observations. Finally, the characterization of the genotype is proposed using nuclear characteristics to improve diagnosis and prognosis, as well as to confirm the natural history of the tumors. This is due to the phenotype of the tumor cells being associated with DNA methylation. Unmethylated DNA has been associated with nuclear size, aneuploidy, carcinoma subtypes and higher proliferation of ovarian carcinoma cells (27,28) [Table I, (2948)].

Table I.

Characterization of DNA methylation loci in ovarian carcinomas.

Table I.

Characterization of DNA methylation loci in ovarian carcinomas.

First author/s, yearOvarian carcinoma subtypeLocus chromosomeGene on the locusMethodsStatus in ovarian carcinoma(Refs.)
Feng et al, 2008EOC3q13.33 and 19q13.43ARH1 and PEG3Sodium bisulfite and pyrosequencing Hypermethylated(29)
Link et al, 2013EOC 20q13.31 BORIS/CTCFLSodium bisulfite and pyrosequencingUnmethylated(30)
Wang et al, 2013EOC 17q21.31BRCA1Sodium bisulfite and RT-qPCR Hypermethylated(31)
Abou-Zeid et al, 2011; Bhagat et al, 2014EOC9p21.3CDKN2ASodium bisulfite and RT-qPCR Hypermethylated(32,33)
Yang et al, 2013EOC8p21.1 ClusterinSodium bisulfite and RT-qPCRUnmethylated(34)
Zhang et al, 2015EOCXq26.3CT45Sodium bisulfite and pyrosequencingUnmethylated(35)
Wang et al, 2017EOC11q25OPCMLSodium bisulfite and RT-qPCR Hypermethylated(36)
Kaur et al, 2016EOC9q21.33 and 13q34DAPK1 and SOX1Sodium bisulfite and RT-qPCR Hypermethylated(37)
Rattanapan et al, 2018HGSOC9q34.3EGFL7Sodium bisulfite and pyrosequencing Hypermethylated(38)
da Conceição Braga et al, 2014EOC18q21.33 and 8p21.3BCL2 and TRAIL2-R2Sodium bisulfite and RT-qPCR Hypermethylated(39)
Bonito et al, 2016EOC4p16.2MSX1Sodium bisulfite and RT-qPCR Hypermethylated(40)
Kardum et al, 2017; Suzuki et al, 2008EOC14q23.2ER-βSodium bisulfite and RT-qPCR. Hypermethylated(41,42)
Sodium bisulfite and pyrosequencing
Baranova et al, 2018HGSOC13q21.1PCDH17Sodium bisulfite and RT-qPCR. Hypermethylated(43)
Sodium bisulfite and pyrosequencing
Ding et al, 2016EOC11p14.3FANCFSodium bisulfite and pyrosequencing Hypermethylated(44)
Gozzi et al, 2016EOC1p12TBX15Sodium bisulfite and pyrosequencing Hypermethylated(45)
Choi et al, 2006EOC3p21.31RASSF1ASodium bisulfite and RT-RT-qPCR Hypermethylated(46)
Häfner et al, 2016EOC1p36.11 and 1p36.12RUNX3 and CAMK2N1Sodium bisulfite and RT-qPCR Hypermethylated(47)
Jin et al, 2018EOC3p14.3Wnt5aSodium bisulfite and RT-qPCR Hypermethylated(48)

[i] EOC, epithelial ovarian cancer; HGSOC, high-grade serous ovarian carcinoma; RT-qPCR, reverse transcription-quantitative PCR.

Currently, the following examples have been demonstrated that indicate the DNA methylation status of ovarian carcinomas and describe the hypomethylated nuclear locus in the tumors compared with that of ovarian epithelial cells: The global loci markers (satellite sequences and ALU repetitive sequences) and the local loci markers. The assays used to discriminate the state of methylation currently available in human tumors are the following: Sodium bisulfite DNA treatment and pyrosequencing, reverse transcription-quantitative PCR (RT-qPCR), or methylation-specific PCR. High-resolution methods to determine cell single DNA methylation are currently available, such as droplet and digital PCR (4350).

DNA methylation and RNA expression are associated with ovarian carcinoma cells. This indicates that RNA transcription could be inhibited due to the DNA methylation status of the locus, except for certain recent paradoxical examples led by a negative correlation in other types of cancers, where intragenic methylation correlates with gene overexpression (Table II) (51,52). In contrast to these observations, RNA expression is activated in the unmethylated DNA status of the locus (9). Therefore, RNA transcription is differentially present in ovarian carcinoma. MicroRNAs (miRs) are a class of small RNA transcripts (19–22 nucleotides) that decrease gene expression via translational inhibition or degradation of target messenger RNA (mRNA). Various miRs are differentially expressed in cancer, suggesting a link between these molecules and the different expression levels of proteins in cancer tissues (53). By contrast, large non-coding RNAs (lncRNAs) function on affecting the nuclear structure and RNA transcription. It is notable that the hypermethylated DNA of RNA loci decrease the presence of miRs and MEG3 (5355), which is a lncRNA, so that the RNA transcripts in the normal ovarian tissue, benign epithelial tumors, benign epithelial ovarian cysts, malignant ovarian carcinoma and serous ovarian carcinoma exhibit differential expression of RNA transcripts (5659).

Table II.

Hyper-methylated miRNAs locus in ovarian carcinoma.

Table II.

Hyper-methylated miRNAs locus in ovarian carcinoma.

Locus chromosomeNon-coding RNA expressed in locusDNA status methylation in locusRNA expression of locus
11q24.1 miR-125-1 HypermethylatedDownregulated
14q32.2miR-127 HypermethylatedDownregulated
11p11.2 miR-129-2 HypermethylatedDownregulated
1p21.3miR-137 HypermethylatedDownregulated
17q11.2 miR-193a HypermethylatedDownregulated
14q32.2MEG3 HypermethylatedDownregulated

[i] miR, microRNA. All dates referenced in this table from (53).

It remains to be determined why DNA methylation in ovarian carcinomas is heterogeneous. In 2014, the World Health Organization classification guidelines for female reproductive tumors defined the ovarian carcinoma type in cell-level characterized ovarian carcinoma subtypes HGSOC, LGSOC, CCC, MC and EC (1113,21,60). It was proposed that at the molecular level, the natural history of ovarian carcinomas is HGSOC. A fallopian tube epithelial origin was found when DNA methylation was compared (61,62). HGSOC has locally DNA methylations in 6 loci that differentiate HGSOC from the OSE DNA methylation pattern (ARMCX1, ICAM4, LOC134466, PEG3, PYCARD and SGNE1) (41); HGSOC overexpresses miR-223, miR-551b-3p, miR-30a-5p, miR-9 and miR-30a-5p (6370). Other studies using miR microarrays have described the different expression patterns of serous ovarian carcinoma (70) and clear cell carcinoma (CCC), specifically the SFRP1 methylated locus (41,71,72). By contrast, DNA methylation in low-grade serous ovarian carcinoma has not been reported to date compared with other ovarian carcinoma subtypes or epithelial ovarian cells. By contrast, it has been shown that endometrioid carcinoma (EC) has similarities with endometrial and ovarian carcinomas in the promoter hypermethylated locus (7376). CCC has been characterized by the HNF1 pathway to be unmethylated, whereas the RE alpha pathway is unmethylated, similar to OSE (71). Finally, mucinous carcinomas (MUC) exhibit 81 unmethylated genes that are different from those of HGSOC. It is notable that MUC-DNA methylation is more similar to colorectal and stomach carcinoma than HGSOC, providing additional information on the MUC origins from colorectal metaplasia (12,77). Downregulation of miR-192 and miR-2215 levels is also noted in MUC (78) (Fig. 1).

In conclusion, DNA methylation is a characteristic that varies early in the development of ovarian carcinoma cells. The vestige of a methylated locus in the origin tissues of ovarian carcinomas marks a directional methylation of every ovarian carcinoma subtype. DNA methylation and RNA expression are associated. Finally, DNA methylation and RNA transcripts are associated and differentially presented by the subtype cells.

DNA methylation and ncRNA expression as prognostic biomarkers of ovarian carcinomas

The following factors are associated with the prognosis of patients with ovarian carcinoma: The variations in DNA syndromes, the ethnic origin, the origin of the gynecological pathologies, the subtypes, the advanced stages of the tumors (metastasis to lymph node, or metastasis to distant tissues), the size of residual tumor following cytoreductive surgery and the resistance of cancer cells to radiotherapy and chemotherapy. It is notable that 75% of ovarian carcinomas are of HGSOC sub-type and the patient 5-year survival following diagnosis with ovarian carcinoma is ~47%. In comparison, the survival of the women diagnosed with metastasis of ovarian carcinoma is only 29% (11,15,2123,79,80).

The RNA expression could be driven by a random accumulation or by a directional and defined development as determined by the stages of International Federation of Gynecology and Obstetrics (FIGO) (81) in every molecular ovarian carcinoma subtype. Several pieces of evidence have concluded that RNA transcripts are overexpressed and downregulated in a directional way (Tables II and III). First, this has been presented in the diagnosis biomarkers without poor prognosis. Except for the BRCA1/2 mutation and the DNA methylation locus associated with response to chemotherapy, the RNA transcripts are not related to the prognosis of the patients or the single nucleotide polymorphism in HOXA11 that protects cells from developing HGSOC (82). This suggests that biologically, hereditary mutations have a higher risk weight than DNA methylation or RNA expression, which are more sensitive to alterations of the phenotype state but not of the patient's outcome. The nuclear characteristics determined of the advanced FIGO stages of ovarian carcinoma are CIN, DNA methylated locus in genes, such as BRCA1, FANCF, RASSF1A and Wnt5A, the downregulated levels of TUBA14B and the overexpressed RNA transcripts of the following genes and lncRNAs: MGMT, OSMR, ESR1 and FOXL2 and long non-coding (lnc)BRM, HOTAIR, HOXDAS-1 and lncSOX4 and CPS1-IT1 (8387) (Table IV).

Table III.

microRNAs expression associated with cancer functions derived from locus and target locus RNAs probed in specific ovarian carcinoma subtypes.

Table III.

microRNAs expression associated with cancer functions derived from locus and target locus RNAs probed in specific ovarian carcinoma subtypes.

First author/s, yearOvarian carcinoma subtypeAssociated withLocus of the miRNAmicroRNARNA regulationLocus of the targetRNA from the locus target(Refs.)
Nymoen et al, 2016EOCPoor prognosis7q32.329aDown2p23.3DNMT3A(66)
Arts et al, 2017HGSOCHGSOCXq12223Up 12q13.12SMARCD1(67)
Ying et al, 2016EOCPoor prognosis, clinical stage III and IV clinical grade11q24.1125bDown9q34.11SET(99)
Zhu et al, 2017EOCII and III lymph node metastasis distant metastasis11q24.1125bUp9q34.11SET(100)
Teng et al, 2015EOCPoor prognosis1p36.3329bDown1q21.2, 19q13.2 and 1q43Mcl-1, AKT2 and AKT3(101)
Cao et al, 2015; Katepanakis et al, 2015; Meng et al, 2016EOCPoor prognosis stage grade1p36.33200a/200b/200cUp (102104)
Du et al, 2017EOCEOC 12p13.31551aDown 22q11.22/14q32.33MAPK/AKT(105)
Chaluvally-Raghavan et al, 2016EOCPoor prognosis1p36.32551b-3pUp17q21.2STAT3(68)
Chen et al, 2015EOCAdvanced stages High grade3q26.2490-3pUp10q21.2CDK1(106)
Shuang et al, 2015EOCChemotherapy resistant 14q32.31134Down3q29Pak2(107)
Zou et al, 2015EOCStage histology15q24.1630Down 10q23.31PTEN(108)
Zhang et al, 2016EOCEOC5q32143-3pDown 18p11.22RALBP1(109)
Zhang et al, 2018EOCStage lymph node metastasis21q21.1Let-7cDown3p21.31CDC25a(110)
Liu et al, 2014EOCPoor prognosis stage9q34.11199b-5pDown 20p12.2-9q34.3JAG1-NOTCH1(111)
Ma et al, 2016EOCEOC8p11.21486-5pDown13q14.3OLFM4(65)
Kobayashi et al, 2018HGSOCAdvanced stages1p36.131290Up-Serum(112)
Zhao, et al, 2015.EOCComplete response14q32.2136Up-DNA repair and apoptosis(69)
Zhao et al, 2014EOCComplete/poor responseXq25224-5pDown3p21.1PRKCD(113)
Wang et al, 2018HGSOCEOC6q13 13p.3330a-5p 200a-5pUp6p21.2P21(64)
Chen et al, 2016EOCStage grade relapse17q23.121Up20q13.2HE4(114)
Li et al, 2014EOCStage III and IV11p15.5210Up14q23.2HIF(115)
Zhu et al, 2017EOCResistance9p21.12204Up--(116)
Fan et al, 2015EOCPoor prognosis stage III and IV tumor size metastasis 17q21.32196aUp 21q22.12RUNX1(117)
Koukorakis et al, 2018HGSOCHGSOC1p36.22 1p36.3334a 200Down Up9p24.1PD-L1(118)
Liu et al, 2016EOCEOC8p22383Up7q34Caspase-2(119)
Dai et al, 2014EOCStages III and IV relapse7q32.329bDown4q21.3MAPK10(120)
Xiao et al, 2017EOCTherapy sensible poor prognosis 19q13.41Let-7eDown17q21.31 and 15q15.1BRCA1 and RAD51(121)
Li et al, 2015EOCTherapy sensible poor prognosis metastasis1q229Down Up5q34 and 16q22.1CCNG1 and E-cadherin(122)
Paudel et al, 2016EOCStages III and IV 22q11.21130bDown1q36.11RUNX3(123)
Duan et al, 2018EOCPoor prognosis3p21.2135a-3pDown3p21.31CCR2(124)
Chen et al, 2015EOCEOC5q32145Down4q31.3TRIM2(125)
Qin et al, 2015EOCPoor prognosis stages III and IV15q25.1184Down--(126)
Liang et al, 2016EOCEOC1q41194Up7q11.23PTPN12(127)
Wei et al, 2017EOCEOC6p21.32219-5pDown7p21.1Twist(128)
Fu et al, 2016EOCGrades II and III poor prognosisXp11.3222-3pDown14q32.33 and 10q23.31AKT and PTEN(129)
Wu et al, 2017CCCCCC 424Down13q13.3DCLK1(130)
Chen et al, 2015EOCAdvanced stages Grades II and IIIXq26.3490-3pDown10q21.2CDK1(106)
Zhang et al, 2016EOCPoor prognosis stage III and IV ascites lymph node metastasis grade III tumor size chemoresistance 19q13.42520g- 15q22.31DAPK2(131)
Zhang et al, 2016EOCStage III and IV lymph node metastasis1p21.3137Down--(132)
Liu et al, 2017EOCEOC11q13.4139Down1q23.1HDGF(133)
Xu et al, 2017EOCStage III and IV tumor size lymph node metastasis9q32455Down9q34.3NOTCH1(134)
Yan et al, 2016EOCAge9q22.3223bDown5q34CCNG1(135)
Lin et al, 2015EOCPoor prognosis stages III and IV distant metastasis recurrence2q3526bDown17q24.2KPNA2(136)
Xu et al, 2017EOCEOC3q2828-5pUp16q12.1N4BP1(137)
Wang et al, 2017HGSOCAdvanced stages11q21.1130aDown9q34TSC1(138)
Wang et al, 2016EOCStages III and IV Grades II and III lymph node metastasis5q32143Down6q23.2CTGF(139)
Dong et al, 2015ECEC3p21.31191Up9q21.33DAPK1(140)
Niu et al, 2015EOCStages III and IV High grade1q32.2205Up 10p11.22ZEB1(141)
Dai et al, 2018EOCEOC6p12.2206Down1p36.22mTOR(142)
Xia et al, 2015HGSOC, CCCTumors15q13.3211Down11q13.3 and 7q21.2Cyclin D1 and CDK6(143)
Wu et al, 2018EOCPoor prognosis deathXp11.3221-3pDown3p14.3ARF4(144)
Cao et al, 2018EOCResistantXq26.2363Down20q13.3Snail(145)
Xia et al, 2016EOCStages III and IV8p22383Down14q32.2YY1(146)
Grades II and III
Yuan et al, 2016; Li et al 2016EOCStages III and IV Grades II and III lymph node metastasis 14q32.31494Down8q24.21 and 15q26.3c-Myc and IGF1R(147,148)
Zhou et al, 2017EOCStage III and IV Grade II and III distant metastasis7q36.3595Down--(149)
Zhang et al, 2017EOCEOC15q24.1630Up10p15.2KLF6(150)
Shi et al, 2016EOCEOC1p32.3761Down 12q24.31MSI1(151)
Xie et al, 2018EOCEOCXp11.3221Up15q15.1BMF(152)
Wen et al, 2015EOCStage III and IV Grade II and III lymph node metastasis17q25.3338-3pDown6p21.1RUNX2(153)
Salem et al, 2018EOCGrades II and III7q11.23590-3pUp 20p11.21FOXA2(154)
Lin et al, 2016EOCEOC17p13.1497Down 10q24.31PAX2(155)
Lin et al, 2018EOCStage grade lymph node metastasis 19q13.32330-5pUp 22q11.22MAPK(156)
Chen et al, 2016EOCEOC 12p13.31141Down21q22.3SIK1(157)
Zuberi et al, 2016EOCStage lymph node metastasis19p13.2199aDown--(158)
Agostini et al, 2018MCMC11q13.1 and 1q41192 and 215Down--(78)
Guan et al, 2017EOCEOC 19q13.42372Down8q24.13, 13q12.11, 5q35.3, 10q21.1 and 13q13.3ATAD2, LATS2, P62, DKK1 and cyclinA1(159)
Li et al, 2016EOCAdvanced stages High grade lymph node metastasis2p16.1217Down15q26.3IGF1R(160)
Zhang et al, 2015EOCPoor prognosis4p15.33572Up16p13.13 and 6p21.2SOCS1 and P21(161)
Zhou et al, 2015SOCPreoperative6q1330a-5pUp--(162)

[i] EOC, epithelial ovarian cancer; HGSOC, high grade serous ovarian carcinoma; CCC, clear cells carcinoma; MC, mucinous carcinoma; EC, endometrioid carcinoma. High grade, grades II and III. Advanced stages, III and IV.

Table IV.

Long non-coding RNAs expression associated to cancer functions derived from locus probed in specific ovarian carcinoma subtypes.

Table IV.

Long non-coding RNAs expression associated to cancer functions derived from locus probed in specific ovarian carcinoma subtypes.

First author/s, yearOvarian cancer subtypeAssociated withLocusLncRNARegulation(Refs.)
Xi et al, 2017EOCPoor prognosis stage grade lymph node metastasis5q11.2lncBRMUp(92)
Zhang et al, 2017EOCEOC1p21.2 NR_026689Up(163)
Wang et al, 2017EOCFavorable prognosis stage Lymph node metastasis2q34 CPS1-IT1Down(97)
Zhu et al, 2018EOCMetastasis CA-125 levels2p25.1 CTD2020K17.1Up(164)
Qiu et al, 2017EOCEOC1q32.1 ElncRNA1Up(165)
Gao et al, 2015EOCEOC10q23.1HOST2Up(166)
Wang et al, 2015EOCPoor prognosis Stage Histological grade Residual tumor Lymph node metastasis 12q13.13HOTAIRUp(167)
Lu et al, 2018HGSOCPoor prognosis7p15.2 HOXA11-ASUp(168)
Zhang et al, 2017EOCPoor prognostic stage lymph node metastasis2q31.1 HOXD-AS1Up(93)
Du et al, 2018EOCPoor prognosis21q22.3 LINC00319Up(169)
Shu et al, 2018EOCPoor prognosis stage grade lymph metastasis distant metastasis9q21.31ARSRUp(170)
Chen et al, 2017EOCEOC6p24.3HULCUp(171)
Liu et al, 2018EOCStage tumor size distant metastasis6p21LncSox4Up(94)
Qunbo et al, 2018; Lin et al, 2018EOCPoor prognostic11q13.1MALAT1Up(172,173)
Yan et al, 2018EOCPoor prognostic stage depth of invasion lymph node metastasis distant metastasis2q31.1 MLK7-AS1Up(95)
Yan et al, 2017EOCFavorable prognosis tumor size6p22.3NBAT-1Down(174)
Liu et al, 2018EOCPoor prognostic stage grade residual tumor metastasis11q13.1NEAT1Up(175)
Chen et al, 2018EOCGrade2q32.3PCGEM1Up(176)
Huang et al, 2018EOCStage grade tumor size1p13.2 RP11-552M11.4Up(177)
Li et al, 2017EOCPoor prognosis stage grade lymph node metastasis5q31.3 SPRY4-IT1Up(178)
Zhu et al, 2017EOCFavorable prognosis stage grade lymph node metastasis CA-125 levels2q35 lncRNA-TUBA4BDown(98)
Li et al, 2018EOCEOC poor prognosis stage grade tumor size22q12.2 lncRNA-TUG1Up(179)
Hong et al, 2016EOCPoor prognostic stage lymph node metastasis chemotherapy response 19p13.12 lncRNA-UCA1Up(180,181)
Qiu et al, 2016EOCPoor prognosis stage grade9p21.3ANRILUp(182)
Zhang et al, 2018EOCEOC 10q26.11CASC2Down(90)
Cao et al, 2017EOCEOC8q24.21CCAT1Up(183)
Hua et al, 2018EOCEOC8q24.21CCAT2Up(184)

[i] EOC, epithelial ovarian cancer; HGSOC, high-grade serous ovarian carcinoma.

The assays used to analyze RNAs are transcriptomics or expression analysis of single locus transcription detected by RT-qPCR (88,89). It is notable that the overexpressed and downregulated transcripts associated with tumor development classified by FIGO stages could probably result in equilibration of their levels in the nuclear structure; in addition, the variation in transcript levels within patients has to be taken into account; for example, the levels of MLK7-AS1 and TUG1 were increased 2.5- and 2.2-fold, respectively; the levels of CASC2 were diminished 0.6-fold compared with the relative expression noted in advanced stage ovarian carcinoma cells (9098). These types of variation in the RNA expression levels are individual assessments in ovarian carcinoma subtypes derived from a population, which are defined by comparison of the expression levels of their corresponding counterparts. However, in the majority of the studies, the ovarian cancer cells are used as a point of calibration (66,67,98162).

Patients with advanced stages have methylated loci DNA on ER-b, RUNX3, and CAMK2N1; these alterations have been associated with poor prognosis (4143). The lncRNAs SPRY4-IT1 and HOXA11 are associated with ovarian carcinoma transformation; they are overexpressed in ovarian carcinoma compared with ovarian untransformed cells (107,108,163184). In addition, overexpression of the miR-200 family members and MALAT1 have been associated with poor prognosis in advanced stages with a sensitivity of 88%; specifically miR-125b overexpression exhibits a sensitivity of 75.6% (108,172,181185). In addition, miR-199a exhibits a sensitivity of 72% related to positive lymph node metastasis (154,186189). Moreover, miR-125b overexpression exhibits a sensitivity of 72%, which is characteristic of ovarian carcinoma, associated with 67% grade, 77% positive lymph node metastasis and 89% metastasis (190). The upregulation of the expression of CPST1-IT1 is also associated with cancer, stage and lymph node metastasis with a hazard ratio of 3.257 (P=0.004) (97). This suggests a variation in the gradual overexpression during the development of ovarian carcinomas. RNA transcripts have a directional variation of expression during the development of the natural history of ovarian carcinomas, which is initiated from the tissues of origin (Fig. 2).

The molecular-resistant ovarian carcinoma

Molecular and cellular biology allows the improved understanding of the microscopic and macroscopic observations of ovarian carcinomas. The selection of the therapeutic methods, such as surgery, radiotherapy and chemotherapy, depends on several factors, such as the carcinoma grade, FIGO stage and patient characteristics, namely age (81). Surgery is performed to obtain the tumor via cytoreduction, which involves extracting carcinoma cells from the patient. Chemotherapy is subsequently administered following cytoreduction in cases of advanced ovarian carcinoma. By contrast, radiation therapy uses high-energy particles to destroy tumor cells, either directly or indirectly, to inhibit further cell growth. Concomitantly, radiotherapy has been commonly used as a first-line treatment for ovarian carcinoma until the 1990s. It is now rarely used alone and is typically used with surgery (191). However, radiotherapy can still be beneficial in certain ways, such as reducing tumor size prior to surgery, treating areas where cancer has spread and providing palliative care (192,193). Ovarian carcinoma cells are radiosensitive at the early stages of development (194), notably in low-grade carcinoma subtypes such as EC (195).

Chemotherapy presents a challenging environment for cells, aiming to eliminate tumor cells and improve the prognosis for patients with cancer. The standard chemotherapy treatment for ovarian carcinoma combines carboplatin and paclitaxel. Chemotherapy can alter the nuclear structure, DNA methylation and RNA expression in ovarian carcinoma cells (203205). To date, the association of methylation with the incidence of cancer in patients is not directly known. However, DNA methylation is associated with chromosomal instability of high-grade serous ovarian carcinoma. This is the most aggressive subtype of ovarian cancer. It can be inferred that by understanding the relationship between methylation and CIN, the progression of ovarian cancer can be predicted. For example, it is known that the HGSOC subtype with TP53 mutation and CIN is associated with DNA hypermethylation in patients with poorer prognosis (206213). HGSOC is characterized by TP53 mutation and CIN and is linked to chemotherapy resistance; it is also considered to be the more resistant and heterogeneous subtype of ovarian carcinomas (214). For example, treatment with paclitaxel in specific cell line models induces overexpression of mdr1 and the lncRNAs UCA1 and long intergenic non-coding RNA linc00312 (215).

Paclitaxel is a drug that inhibits depolymerization of microtubules. It arrests cells in the G2/M phase. It also induces CIN, leading to cell death. However, certain ovarian carcinoma cells are resistant to paclitaxel. Certain miR biomarkers, such as miR-134 and miR-224-5p, are associated with EOC resistance to paclitaxel chemotherapy with 85 and 90% sensitivity, respectively (94,102). Studies in ovarian carcinoma have shown that RNA transcript levels are altered during paclitaxel treatment in ovarian cancer cells. For example, treatment of A2780, OVCAR3, SKOV3, and SW626 cells with paclitaxel induces overexpression of mdr1 (215), UCA1 and lincRNA00312.

Previous studies have shown that cisplatin increases the expression of ZEB1 and MP63 (216,217). It is notable that the use of array expression and PCR validation in the A2780 cisplatin-resistant cell line revealed that the expression levels of the following six miRs were upregulated: miR-1064, miR-300, miR-193b, miR-642 and miR-1299; however, the expression levels of the following five miRs were downregulated: miR-625, miR-20b, miRPlus-F1147, let-7c, miR-1231 and miR-542-3p (214,215,218,219). This evidence suggests that RNA transcripts are differentially expressed in chemotherapy-resistant cells compared with sensitive cells, providing a solid basis for further research. It also indicates the implications for the survival outcomes of the patients with ovarian cancer (220225) (Fig. 3).

Discussion and conclusions

Ovarian carcinoma is a type of cancer resulting from tissue transformation and can occur both in hereditary (20%) and sporadic (80%) forms. DNA methylations repress RNA transcription differentially in ovarian carcinoma subtypes. This suggests that DNA methylation of ovarian carcinoma subtypes is more similar to their tissue of origin with regard to their nuclear characteristics. Ovarian carcinoma presents a complex molecular landscape where DNA methylation and RNA expression play crucial roles in the disease development, progression and treatment response. DNA methylation serves as both an early event in carcinogenesis and a key regulator of gene expression, either silencing or activating RNA transcription based on the methylation status of specific loci.

The characteristics of nuclear vestiges vary in a directional range of RNA transcripts. In addition, the directional way of variation of the RNA transcripts is directed by FIGO stages observed in every carcinoma subtype. The molecular ovarian carcinoma subtypes are five and can be classified as follows: High-grade serous, low-grade serous, endometrioid, mucinous and clear cell; however, there are other subtypes to be described since they belong in the five subtypes or other similar subtypes, such as carcinosarcoma (analogous to malignant mixed Mullerian/mesodermal tumors) and malignant Brenner tumors. Other cellular subtypes are currently in discovery. This is particularly relevant in HGSOC, where distinct methylation patterns have been identified, linking them to tumor origin, subtype differentiation and patient prognosis. The heterogeneity of ovarian carcinoma is further exemplified by the differential expression of miRs and lncRNAs, which are involved in gene regulation.

The molecular prognosis of ovarian carcinoma varies. Firstly, hereditary ovarian carcinoma exhibits a greater effect than sporadic ovarian carcinoma. By contrast, the survival of each patient with spontaneous ovarian carcinoma depends on the carcinoma development and the subtype. At a molecular level, deviations in the DNA methylation and RNA transcription have been associated with metastasis stages and the development of therapy-resistant cancer cells. These findings suggest that molecular signatures, including miR and lncRNA profiles, could serve as valuable biomarkers for diagnosis, prognosis and therapeutic targeting in ovarian carcinomas. Despite the advances in the understanding of the molecular underpinnings of ovarian carcinomas, the disease prognosis for patients, notably those diagnosed with advanced-stage or metastatic disease remains poor, with survival rates being markedly lower among these groups.

Current diagnostic and therapeutic strategies are increasingly incorporating molecular data, including RNA transcriptomics and DNA methylation analysis, to improve the precision of treatment approaches. While hereditary mutations, such as those noted in BRCA1/2, confer a significant risk, epigenetic changes, notably in the later stages of the disease, are critical in shaping the tumor phenotype and response to therapy. In clinical practice, evidence leads to the hypothesis that certain genes could be used as a combination to adjust cancer treatments. However, a pair of primers may not provide a clinical solution. Therefore, practical techniques, such as PCR and sequencing, are used for validation of the next biological characterization of chromosomes. In the present review, it was hypothesized that chromosome instability, which is characterized by gain of chromosomes in cancer cells and the loss of specific chromosomes or their translocation, should be the focus of future research. For example, changes in DNA methylation of chromosomes 9 and 19 (Figs. 2 and 3) in high-grade serous ovarian carcinoma are associated with ovarian carcinoma progression and resistance to treatments.

Ultimately, a comprehensive understanding of the molecular heterogeneity in ovarian carcinoma, including the roles of DNA methylation and RNA transcription into the nucleus, is essential to develop a vision for more effective treatments, increase the understanding of disease progression and improve long-term outcomes for patients. The continued exploration of these molecular pathways holds the potential not only to revolutionize but also to reform the clinical management of ovarian carcinomas.

Acknowledgements

This article is part of the productivity of LCE as a PhD student of Biological Sciences Postgrade Program, in the Biomedicine Field.

Funding

This article was supported by the Programa de Posgrado en Ciencias Biológicas, UNAM. This article is part of the productivity work of the Ph.D. student LCE in the Programa de Posgrado en Ciencias Biológicas, UNAM, and received a fellowship from CONACYT Currículum Vitae Único (CVU)- 1003211. National Cancer Institute, México (INCAN) and CONAHCYT supported this project (grant nos. CMIC: 295466 and CBF 2023-2024-4004). The content is solely the authors´s responsibility and does not necessarily represent the official views of the National Cancer Institute of México. Institutional Review Board Statement.

Availability of data and materials

Not applicable.

Authors' contributions

Conceptualization and original draft preparation: VMDCF, VFO, JDC, LAH and LCE. Writing and revising the manuscript JAGR, APT, VFO, LAH and VMDCF. Supervision, project administration and funding acquisition: JDC and LAH. All authors have read and approved the final version of the manuscript. Data authentication is not applicable.

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.

References

1 

Momenimovahed Z, Tiznobaik A, Taheri S and Salehiniya H: Ovarian cancer in the world: epidemiology and risk factors. Int J Womens Health. 11:287–299. 2019. View Article : Google Scholar : PubMed/NCBI

2 

Siegel RL, Miller KD and Jemal A: Cancer Statistics, 2016. CA Cancer J Clin. 66:7–30. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Luo Y, Huang J, Tang Y, Luo X, Ge L, Sheng X, Sun X, Chen Y and Zhu D: Regional methylome profiling reveals dynamic epigenetic heterogeneity and convergent hypomethylation of stem cell quiescence-associated genes in breast cancer following neoadjuvant chemotherapy. Cell Biosci. 9:162019. View Article : Google Scholar : PubMed/NCBI

4 

Sandhu R, Roll JD, Rivenbark AG and Coleman WB: Dysregulation of the Epigenome in Human Breast Cancer': Contributions of gene-specific DNA hypermethylation to breast cancer pathobiology and targeting the breast cancer methylome for improved therapy. Am J Pathol. 185:282–292. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Maire CL, Fuh MM, Kaulich K, Fita KD, Stevic I, Heiland DH, Welsh JA, Jones JC, Görgens A, Ricklefs T, et al: Genome-wide methylation profiling of glioblastoma cell-derived extracellular vesicle DNA allows tumor classification. Neuro Oncol. 23:1087–1099. 2021. View Article : Google Scholar : PubMed/NCBI

6 

Wang Z, Cui Y, Wang F, Xu L, Yan Y, Tong X and Yan H: DNA methylation-regulated LINC02587 inhibits ferroptosis and promotes the progression of glioma cells through the CoQ-FSP1 pathway. BMC Cancer. 23:9892023. View Article : Google Scholar : PubMed/NCBI

7 

Wielandt AM, Villarroel C, Hurtado C, Simian D, Zamorano D, Martínez M, Castro M, Vial MT, Kronberg U and López-Kostner F: Characterization of patients with sporadic colorectal cancer following the new Consensus Molecular Subtypes (CMS). Rev Méd Chile. 145:419–430. 2017.(In Spanish). View Article : Google Scholar : PubMed/NCBI

8 

Moreno-Ortiz JM, Jiménez-García J, Gutiérrez-Angulo M, Ayala-Madrigal MD, González-Mercado A, González-Villaseñor CO, Flores-López BA, Alvizo-Rodríguez C, Hernández-Sandoval JA, Fernández-Galindo MA, et al: High frequency of MLH1 promoter methylation mediated by gender and age in colorectal tumors from Mexican patients. GMM. 157:638–644. 2021.(In Spanish).

9 

Del Castillo Falconi VM, Torres-Arciga K, Matus-Ortega G, Díaz-Chávez J and Herrera LA: DNA methyltransferases: From evolution to clinical applications. Int J Mol Sci. 23:89942022. View Article : Google Scholar : PubMed/NCBI

10 

Li E, Bestor TH and Jaenisch R: Targeted mutation of the DNA methyltransferase gene results in embryonic lethality. Cell. 69:915–926. 1992. View Article : Google Scholar : PubMed/NCBI

11 

Shih IeM and Kurman RJ: Ovarian tumorigenesis: A proposed model based on morphological and molecular genetic analysis. Am J Pathol. 164:1511–1518. 2004. View Article : Google Scholar : PubMed/NCBI

12 

Kurman RJ and Shih IeM: Pathogenesis of ovarian cancer: Lessons from morphology and molecular biology and their clinical implications. Int J Gynecol Pathol. 27:151–160. 2018.PubMed/NCBI

13 

Kurman RJ and Shih IeM: The origin and pathogenesis of epithelial ovarian cancer: A proposed unifying theory. Am J Surg Pathol. 34:433–443. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Samuel D, Diaz-Barbe A, Pinto A, Schlumbrecht M and George S: Hereditary ovarian carcinoma: Cancer pathogenesis looking beyond BRCA1 and BRCA2. Cells. 11:5392022. View Article : Google Scholar : PubMed/NCBI

15 

Ramus SJ, Harrington PA, Pye C, DiCioccio RA, Cox MJ, Garlinghouse-Jones K, Oakley-Girvan I, Jacobs IJ, Hardy RM, Whittemore AS, et al: Contribution of BRCA1 and BRCA2 mutations to inherited ovarian cancer. Hum Mutat. 28:1207–1215. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Menon U, Karpinskyj C and Gentry-Maharaj A: Ovarian cancer prevention and screening. Obstet Gynecol. 131:909–927. 2018. View Article : Google Scholar : PubMed/NCBI

17 

Lavoro A, Scalisi A, Candido S, Zanghì GN, Rizzo R, Gattuso G, Caruso G, Libra M and Falzone L: Identification of the most common BRCA alterations through analysis of germline mutation databases: Is droplet digital PCR an additional strategy for the assessment of such alterations in breast and ovarian cancer families? Int J Oncol. 60:582022. View Article : Google Scholar : PubMed/NCBI

18 

Kansuttiviwat C, Lertwilaiwittaya P, Roothumnong E, Nakthong P, Dungort P, Meesamarnpong C, Tansa-Nga W, Pongsuktavorn K, Wiboonthanasarn S, Tititumjariya W, et al: Germline mutations of 4567 patients with hereditary breast-ovarian cancer spectrum in Thailand. NPJ Genom Med. 9:92024. View Article : Google Scholar : PubMed/NCBI

19 

Andrikopoulou A, Zografos E, Apostolidou K, Kyriazoglou A, Papatheodoridi AM, Kaparelou M, Koutsoukos K, Liontos M, Dimopoulos MA and Zagouri F: Germline and somatic variants in ovarian carcinoma: A next-generation sequencing (NGS) analysis. Front Oncol. 12:10307862022. View Article : Google Scholar : PubMed/NCBI

20 

Ghose A, Bolina A, Mahajan I, Raza SA, Clarke M, Pal A, Sanchez E, Rallis KS and Boussios S: Hereditary ovarian cancer: Towards a cost-effective prevention strategy. Int J Environ Res Public Health. 19:120572022. View Article : Google Scholar : PubMed/NCBI

21 

McCluggage WG: Morphological subtypes of ovarian carcinoma: A review with emphasis on new developments and pathogenesis. Pathology. 43:420–432. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Andrews L and Mutch DG: Hereditary ovarian cancer and risk reduction. Best Pract Res Clin Obstet Gynaecol. 41:31–48. 2017. View Article : Google Scholar : PubMed/NCBI

23 

Lynch HT and Lynch JF: Hereditary nonpolyposis colorectal cancer. Semin Surg Oncol. 18:305–313. 2000. View Article : Google Scholar : PubMed/NCBI

24 

Wu H, Shang X, Shi Y, Yang Z, Zhao J, Yang M, Li Y and Xu S: Genetic variants of lncRNA HOTAIR and risk of epithelial ovarian cancer among Chinese women. Oncotarget. 7:41047–41052. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Bronder D, Tighe A, Wangsa D, Zong D, Meyer TJ, Wardenaar R, Minshall P, Hirsch D, Heselmeyer-Haddad K, Nelson L, et al: TP53 loss initiates chromosomal instability in fallopian tube epithelial cells. Dis Model Mech. 14:dmm0490012021. View Article : Google Scholar : PubMed/NCBI

26 

Goff BA, Mandel L, Muntz HG and Melancon CH: Ovarian carcinoma diagnosis. Cancer. 89:2068–2075. 2000. View Article : Google Scholar : PubMed/NCBI

27 

Zeimet AG, Fiegl H, Goebel G, Kopp F, Allasia C, Reimer D, Steppan I, Mueller-Holzner E, Ehrlich M and Marth C: DNA ploidy, nuclear size, proliferation index and DNA-hypomethylation in ovarian cancer. Gynecol Oncol. 121:24–31. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Widschwendter M, Jiang G, Woods C, Müller HM, Fiegl H, Goebel G, Marth C, Müller-Holzner E, Zeimet AG, Laird PW and Ehrlich M: DNA hypomethylation and ovarian cancer biology. Cancer Res. 64:4472–4480. 2004. View Article : Google Scholar : PubMed/NCBI

29 

Feng W, Marquez RT, Lu Z, Liu J, Lu KH, Issa JP, Fishman DM, Yu Y and Bast RC Jr: Imprinted tumor suppressor genesARHI andPEG3 are the most frequently down-regulated in human ovarian cancers by loss of heterozygosity and promoter methylation. Cancer. 112:1489–1502. 2008. View Article : Google Scholar : PubMed/NCBI

30 

Link PA, Zhang W, Odunsi K and Karpf AR: BORIS/CTCFL mRNA isoform expression and epigenetic regulation in epithelial ovarian cancer. Cancer Immun. 13:62013.PubMed/NCBI

31 

Wang YQ, Yan Q, Zhang JR, Li SD, Yang YX and Wan XP: Epigenetic inactivation of BRCA1 through promoter hypermethylation in ovarian cancer progression. J Obstet Gynaecol Res. 39:549–554. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Abou-Zeid AA, Azzam AZ and Kamel NA: Methylation status of the gene promoter of cyclin-dependent kinase inhibitor 2A (CDKN2A) in ovarian cancer. Scand J Clin Lab Invest. 71:542–547. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Bhagat R, Kumar SS, Vaderhobli S, Premalata CS, Pallavi VR, Ramesh G and Krishnamoorthy L: Epigenetic alteration of p16 and retinoic acid receptor beta genes in the development of epithelial ovarian carcinoma. Tumour Biol. 35:9069–9078. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Yang G, Zhang H, Liu Y, Zhou J, He W, Quick CM, Xie D, Smoller BR and Fan CY: Epigenetic and immunohistochemical characterization of the Clusterin gene in ovarian tumors. Arch Gynecol Obstet. 287:989–995. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Zhang W, Barger CJ, Link PA, Mhawech-Fauceglia P, Miller A, Akers SN, Odunsi K and Karpf AR: DNA hypomethylation-mediated activation of Cancer/Testis Antigen 45 (CT45) genes is associated with disease progression and reduced survival in epithelial ovarian cancer. Epigenetics. 10:736–748. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Wang B, Yu L, Luo X, Huang L, Li QS, Shao XS, Liu Y, Fan Y and Yang GZ: Detection of OPCML methylation, a possible epigenetic marker, from free serum circulating DNA to improve the diagnosis of early-stage ovarian epithelial cancer. Oncol Lett. 14:217–223. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Kaur M, Singh A, Singh K, Gupta S and Sachan M: Development of a multiplex MethyLight assay for the detection of DAPK1 and SOX1 methylation in epithelial ovarian cancer in a north Indian population. Genes Genet Syst. 91:175–181. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Rattanapan Y, Korkiatsakul V, Kongruang A, Chareonsirisuthigul T, Rerkamnuaychoke B, Wongkularb A and Wilailak S: EGFL7 and RASSF1 promoter hypermethylation in epithelial ovarian cancer. Cancer Genet. 224–225. 37–40. 2018.PubMed/NCBI

39 

da Conceição Braga C, Silva LM, Piedade JB, Traiman P and da Silva Filho AL: Epigenetic and expression analysis of TRAIL-R2 and BCL2: On the TRAIL to knowledge of apoptosis in ovarian tumors. Arch Gynecol Obstet. 289:1061–1069. 2014. View Article : Google Scholar : PubMed/NCBI

40 

Bonito NA, Borley J, Wilhelm-Benartzi CS, Ghaem-Maghami S and Brown R: Epigenetic regulation of the homeobox gene MSX1 associates with platinum-resistant disease in high-grade serous epithelial ovarian cancer. Clin Cancer Res. 22:3097–3104. 2016. View Article : Google Scholar : PubMed/NCBI

41 

Kardum V, Karin V, Glibo M, Skrtic A, Martic TN, Ibisevic N, Skenderi F, Vranic S and Serman L: Methylation-associated silencing of SFRP1 gene in high-grade serous ovarian carcinomas. Ann Diagn Pathol. 31:45–49. 2017. View Article : Google Scholar : PubMed/NCBI

42 

Suzuki F, Akahira J, Miura I, Suzuki T, Ito K, Hayashi S, Sasano H and Yaegashi N: Loss of estrogen receptor beta isoform expression and its correlation with aberrant DNA methylation of the 5′-untranslated region in human epithelial ovarian carcinoma. Cancer Sci. 99:2365–2372. 2008. View Article : Google Scholar : PubMed/NCBI

43 

Baranova I, Kovarikova H, Laco J, Dvorak O, Sedlakova I, Palicka V and Chmelarova M: Aberrant methylation of PCDH17 gene in high-grade serous ovarian carcinoma. Cancer Biomark. 23:125–133. 2018. View Article : Google Scholar : PubMed/NCBI

44 

Ding JJ, Wang G, Shi WX, Zhou HH and Zhao EF: Promoter hypermethylation of FANCF and susceptibility and prognosis of epithelial ovarian cancer. Reprod Sci. 23:24–30. 2016. View Article : Google Scholar : PubMed/NCBI

45 

Gozzi G, Chelbi ST, Manni P, Alberti L, Fonda S, Saponaro S, Fabbiani L, Rivasi F, Benhattar J and Losi L: Promoter methylation and downregulated expression of the TBX15 gene in ovarian carcinoma. Oncol Lett. 12:2811–2819. 2016. View Article : Google Scholar : PubMed/NCBI

46 

Choi YL, Kang SY, Shin YK, Choi JS, Kim SH, Lee SJ, Bae DS and Ahn G: Aberrant hypermethylation of RASSF1A promoter in ovarian borderline tumors and carcinomas. Virchows Archiv. 448:331–336. 2006. View Article : Google Scholar : PubMed/NCBI

47 

Häfner N, Steinbach D, Jansen L, Diebolder H, Dürst M and Runnebaum IB: RUNX3 and CAMK2N1 hypermethylation as prognostic marker for epithelial ovarian cancer. Int J Cancer. 138:217–228. 2016. View Article : Google Scholar : PubMed/NCBI

48 

Jin P, Song Y and Yu G: The role of abnormal methylation of Wnt5a gene promoter regions in human epithelial ovarian cancer: A clinical and experimental study. Anal Cell Pathol (Amst). 2018:65670812018.PubMed/NCBI

49 

Khodadadi E, Fahmideh L, Khodadadi E, Dao S, Yousefi M, Taghizadeh S, Asgharzadeh M, Yousefi B and Kafil HS: Current advances in DNA methylation analysis methods. Biomed Res Int. 2021:88275162021. View Article : Google Scholar : PubMed/NCBI

50 

Gattuso G, Lavoro A, Caltabiano R, Madonna G, Capone M, Ascierto PA, Falzone L, Libra M and Candido S: Methylation-sensitive restriction enzyme-droplet digital PCR assay for the one-step highly sensitive analysis of DNA methylation hotspots. Int J Mol Med. 53:422024. View Article : Google Scholar : PubMed/NCBI

51 

Falzone L, Salemi R, Travali S, Scalisi A, McCubrey JA, Candido S and Libra M: MMP-9 overexpression is associated with intragenic hypermethylation of MMP9 gene in melanoma. Aging (Albany NY). 8:933–944. 2016. View Article : Google Scholar : PubMed/NCBI

52 

Singer M, Kosti I, Pachter L and Mandel-Gutfreund Y: A diverse epigenetic landscape at human exons with implication for expression. Nucleic Acids Res. 43:3498–3508. 2015. View Article : Google Scholar : PubMed/NCBI

53 

Davidson B, Tropé CG and Reich R: The clinical and diagnostic role of microRNAs in ovarian carcinoma. Gynecol Oncol. 133:640–646. 2014. View Article : Google Scholar : PubMed/NCBI

54 

Sheng X and Li J, Yang L, Chen Z, Zhao Q, Tan L, Zhou Y and Li J: Promoter hypermethylation influences the suppressive role of maternally expressed 3, a long non-coding RNA, in the development of epithelial ovarian cancer. Oncol Rep. 32:277–285. 2014. View Article : Google Scholar : PubMed/NCBI

55 

Loginov VI, Pronina IV, Burdennyy AM, Filippova EA, Kazubskaya TP, Kushlinsky DN, Utkin DO, Khodyrev DS, Kushlinskii NE, Dmitriev AA and Braga EA: Novel miRNA genes deregulated by aberrant methylation in ovarian carcinoma are involved in metastasis. Gene. 662:28–36. 2018. View Article : Google Scholar : PubMed/NCBI

56 

Filippov-Levy N, Cohen-Schussheim H, Tropé CG, Hetland Falkenthal TE, Smith Y, Davidson B and Reich R: Expression and clinical role of long non-coding RNA in high-grade serous carcinoma. Gynecol Oncol. 148:559–566. 2018. View Article : Google Scholar : PubMed/NCBI

57 

Liu X, Dai C, Jia G, Xu S, Fu Z, Xu J, Li Q, Ruan H and Xu P: Microarray analysis reveals differentially expressed lncRNAs in benign epithelial ovarian cysts and normal ovaries. Oncol Rep. 38:799–808. 2017. View Article : Google Scholar : PubMed/NCBI

58 

Lu YM, Wang Y, Liu SQ, Zhou MY and Guo YR: Profile and validation of dysregulated long non-coding RNAs and mRNAs in ovarian cancer. Oncol Rep. 40:2964–2976. 2018.PubMed/NCBI

59 

Wang H, Fu Z, Dai C, Cao J, Liu X, Xu J, Lv M, Gu Y, Zhang J, Hua X, et al: LncRNAs expression profiling in normal ovary, benign ovarian cyst and malignant epithelial ovarian cancer. Sci Rep. 6:389832016. View Article : Google Scholar : PubMed/NCBI

60 

Boyd C and McCluggage WG: Low-grade ovarian serous neoplasms (low-grade serous carcinoma and serous borderline tumor) associated with high-grade serous carcinoma or undifferentiated carcinoma: Report of a series of cases of an unusual phenomenon. Am J Surg Pathol. 36:368–375. 2012. View Article : Google Scholar : PubMed/NCBI

61 

Pisanic TR II, Cope LM, Lin SF, Yen TT, Athamanolap P, Asaka R, Nakayama K, Fader AN, Wang TH, Shih IM and Wang TL: Methylomic analysis of ovarian cancers identifies tumor-specific alterations readily detectable in early precursor lesions. Clin Cancer Res. 24:6536–6547. 2018. View Article : Google Scholar : PubMed/NCBI

62 

Klinkebiel D, Zhang W, Akers SN, Odunsi K and Karpf AR: DNA Methylome analyses implicate fallopian tube epithelia as the origin for high-grade serous ovarian cancer. Mol Cancer Res. 14:787–794. 2016. View Article : Google Scholar : PubMed/NCBI

63 

Givel AM, Kieffer Y, Scholer-Dahirel A, Sirven P, Cardon M, Pelon F, Magagna I, Gentric G, Costa A, Bonneau C, Mieulet V, et al: miR200-regulated CXCL12β promotes fibroblast heterogeneity and immunosuppression in ovarian cancers. Nat Commun. 9:10562018. View Article : Google Scholar : PubMed/NCBI

64 

Wang Y, Qiu C, Lu N, Liu Z, Jin C, Sun C, Bu H, Yu H, Dongol S and Kong B: FOXD1 is targeted by miR-30a-5p and miR-200a-5p and suppresses the proliferation of human ovarian carcinoma cells by promoting p21 expression in a p53-independent manner. Int J Oncol. 52:2130–2142. 2018.PubMed/NCBI

65 

Ma H, Tian T, Liang S, Liu X, Shen H, Xia M, Liu X, Zhang W, Wang L, Chen S and Yu L: Estrogen receptor-mediated miR-486-5p regulation of OLFM4 expression in ovarian cancer. Oncotarget. 7:10594–1605. 2016. View Article : Google Scholar : PubMed/NCBI

66 

Nymoen DA, Slipicevic A, Holth A, Emilsen E, Hetland Falkenthal TE, Tropé CG, Reich R, Flørenes VA and Davidson B: MiR-29a is a candidate biomarker of better survival in metastatic high-grade serous carcinoma. Hum Pathol. 54:74–81. 2016. View Article : Google Scholar : PubMed/NCBI

67 

Arts FA, Keogh L, Smyth P, O'Toole S, Ta R, Gleeson N, O'Leary JJ, Flavin R and Sheils O: miR-223 potentially targets SWI/SNF complex protein SMARCD1 in atypical proliferative serous tumor and high-grade ovarian serous carcinoma. Hum Pathol. 70:98–104. 2017. View Article : Google Scholar : PubMed/NCBI

68 

Chaluvally-Raghavan P, Jeong KJ, Pradeep S, Silva AM, Yu S, Liu W, Moss T, Rodriguez-Aguayo C, Zhang D, Ram P, et al: Direct upregulation of STAT3 by MicroRNA-551b-3p deregulates growth and metastasis of ovarian cancer. Cell Rep. 15:1493–1504. 2016. View Article : Google Scholar : PubMed/NCBI

69 

Zhao H, Liu S, Wang G, Wu X, Ding Y, Guo G, Jiang J and Cui S: Expression of miR-136 is associated with the primary cisplatin resistance of human epithelial ovarian cancer. Oncol Rep. 33:591–598. 2015. View Article : Google Scholar : PubMed/NCBI

70 

Kuznetsov VA, Tang Z and Ivshina AV: Identification of common oncogenic and early developmental pathways in the ovarian carcinomas controlling by distinct prognostically significant microRNA subsets. BMC Genomics. 18 (Suppl 6):6922017. View Article : Google Scholar : PubMed/NCBI

71 

Zhang X, Guo G, Wang G, Zhao J, Wang B, Yu X and Ding Y: Profile of differentially expressed miRNAs in high-grade serous carcinoma and clear cell ovarian carcinoma, and the expression of miR-510 in ovarian carcinoma. Mol Med Rep. 12:8021–8031. 2015. View Article : Google Scholar : PubMed/NCBI

72 

Yanaihara N, Noguchi Y, Saito M, Takenaka M, Takakura S, Yamada K and Okamoto A: MicroRNA gene expression signature driven by miR-9 overexpression in ovarian clear cell carcinoma. PLoS One. 11:e01625842016. View Article : Google Scholar : PubMed/NCBI

73 

Furlan D, Carnevali I, Marcomini B, Cerutti R, Dainese E, Capella C and Riva C: The high frequency of de novo promoter methylation in synchronous primary endometrial and ovarian carcinomas. Clin Cancer Res. 12:3329–3336. 2006. View Article : Google Scholar : PubMed/NCBI

74 

Niskakoski A, Pasanen A, Porkka N, Eldfors S, Lassus H, Renkonen-Sinisalo L, Kaur S, Mecklin JP, Bützow R and Peltomäki P: Converging endometrial and ovarian tumorigenesis in Lynch syndrome: Shared origin of synchronous carcinomas. Gynecol Oncol. 150:92–98. 2018. View Article : Google Scholar : PubMed/NCBI

75 

Kolbe DL, DeLoia JA, Porter-Gill P, Strange M, Petrykowska HM, Guirguis A, Krivak TC, Brody LC and Elnitski L: Differential analysis of ovarian and endometrial cancers identifies a methylator phenotype. PLoS One. 7:e329412012. View Article : Google Scholar : PubMed/NCBI

76 

Guo C, Ren F, Wang D, Li Y, Liu K, Liu S and Chen P: RUNX3 is inactivated by promoter hypermethylation in malignant transformation of ovarian endometriosis. Oncol Rep. 32:2580–2588. 2014. View Article : Google Scholar : PubMed/NCBI

77 

Liew PL, Huang RL, Weng YC, Fang CL, Hui-Ming Huang T and Lai HC: Distinct methylation profile of mucinous ovarian carcinoma reveals susceptibility to proteasome inhibitors: Methylation profile of MuOC and PSMB8. Int J Cancer. 143:355–367. 2018. View Article : Google Scholar : PubMed/NCBI

78 

Agostini A, Brunetti M, Davidson B, Tropé CG, Eriksson AGZ, Heim S, Panagopoulos I and Micci F: The microRNA miR-192/215 family is upregulated in mucinous ovarian carcinomas. Sci Rep. 8:110692018. View Article : Google Scholar : PubMed/NCBI

79 

Vang R, Shih IeM and Kurman RJ: Ovarian low-grade and high-grade serous carcinoma: Pathogenesis, clinicopathologic and molecular biologic features, and diagnostic problems. Adv Anat Pathol. 16:267–282. 2009. View Article : Google Scholar : PubMed/NCBI

80 

Bowtell DD: The genesis and evolution of high-grade serous ovarian cancer. Nat Rev Cancer. 10:803–808. 2010. View Article : Google Scholar : PubMed/NCBI

81 

O'Shea AS: Clinical staging of ovarian cancer. Methods Mol Biol. 2424:3–10. 2022. View Article : Google Scholar : PubMed/NCBI

82 

Richards EJ, Permuth-Wey J, Li Y, Chen YA, Coppola D, Reid BM, Lin HY, Teer JK, Berchuck A, Birrer MJ, et al: A functional variant in HOXA11-AS, a novel long non-coding RNA, inhibits the oncogenic phenotype of epithelial ovarian cancer. Oncotarget. 6:34745–34757. 2015. View Article : Google Scholar : PubMed/NCBI

83 

Zhang T, Wu D, Deng S, Han R, Liu T, Li J and Xu Y: Integrated analysis reveals that long non-coding RNA TUBA4B can be used as a prognostic biomarker in various cancers. Cell Physiol Biochem. 49:530–544. 2018. View Article : Google Scholar : PubMed/NCBI

84 

Meryet-Figuière M, Lambert B, Gauduchon P, Vigneron N, Brotin E, Poulain L and Denoyelle C: An overview of long non-coding RNAs in ovarian cancers. Oncotarget. 7:44719–44734. 2016. View Article : Google Scholar : PubMed/NCBI

85 

Zhong Y, Gao D, He S, Shuai C and Peng S: Dysregulated expression of long noncoding RNAs in ovarian cancer. Int J Gynecol Cancer. 26:1564–1570. 2016. View Article : Google Scholar : PubMed/NCBI

86 

Ma Y, Lu Y and Lu B: MicroRNA and Long Non-Coding RNA in ovarian carcinoma: Translational insights and potential clinical applications. Cancer Invest. 34:465–476. 2016. View Article : Google Scholar : PubMed/NCBI

87 

Lin X, Qiu J and Hua K: Long non-coding RNAs as emerging regulators of epithelial to mesenchymal transition in gynecologic cancers. Biosci Trends. 12:342–353. 2018. View Article : Google Scholar : PubMed/NCBI

88 

Micheel J, Safrastyan A and Wollny D: Advances in non-coding RNA sequencing. Noncoding RNA. 7:702021.PubMed/NCBI

89 

Zhang N, Hu G, Myers TG and Williamson PR: Protocols for the analysis of microRNA expression, biogenesis, and function in immune cells. Curr Protoc Immunol. 126:e782019. View Article : Google Scholar : PubMed/NCBI

90 

Zhang S, Leng T, Zhang Q, Zhao Q, Nie X and Yang L: Sanguinarine inhibits epithelial ovarian cancer development via regulating long non-coding RNA CASC2-EIF4A3 axis and/or inhibiting NF-κB signaling or PI3K/AKT/mTOR pathway. Biomed Pharmacother. 102:302–308. 2018. View Article : Google Scholar : PubMed/NCBI

91 

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

92 

Xi J, Feng J and Zeng S: Long noncoding RNA lncBRM facilitates the proliferation, migration and invasion of ovarian cancer cells via upregulation of Sox4. Am J Cancer Res. 7:2180–2189. 2017.PubMed/NCBI

93 

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

94 

Liu Y, Wang Y, Yao D and Cui D: LncSOX4 serves an oncogenic role in the tumorigenesis of epithelial ovarian cancer by promoting cell proliferation and inhibiting apoptosis. Mol Med Rep. 17:8282–8288. 2018.PubMed/NCBI

95 

Yan H, Li H, Li P, Li X, Lin J, Zhu L, Silva MA, Wang X, Wang P and Zhang Z: Long noncoding RNA MLK7-AS1 promotes ovarian cancer cells progression by modulating miR-375/YAP1 axis. J Exp Clin Cancer Res. 37:2372018. View Article : Google Scholar : PubMed/NCBI

96 

Li T, Chen Y, Zhang J and Liu S: LncRNA TUG1 promotes cells proliferation and inhibits cells apoptosis through regulating AURKA in epithelial ovarian cancer cells. Medicine (Baltimore). 97:e121312018. View Article : Google Scholar : PubMed/NCBI

97 

Wang YS, Ma LN, Sun JX, Liu N and Wang H: Long non-coding CPS1-IT1 is a positive prognostic factor and inhibits epithelial ovarian cancer tumorigenesis. Eur Rev Med Pharmacol Sci. 21:3169–3175. 2017.PubMed/NCBI

98 

Zhu FF, Zheng FY, Wang HO, Zheng JJ and Zhang Q: Downregulation of lncRNA TUBA4B is associated with poor prognosis for epithelial ovarian cancer. Pathol Oncol Res. 24:419–425. 2018. View Article : Google Scholar : PubMed/NCBI

99 

Ying X, Wei K, Lin Z, Cui Y, Ding J, Chen Y and Xu B: MicroRNA-125b suppresses ovarian cancer progression via suppression of the epithelial-mesenchymal transition pathway by targeting the SET protein. Cell Physiol Biochem. 39:501–510. 2016. View Article : Google Scholar : PubMed/NCBI

100 

Zhu T, Gao W, Chen X, Zhang Y, Wu M, Zhang P and Wang S: A pilot study of circulating MicroRNA-125b as a diagnostic and prognostic biomarker for epithelial ovarian cancer. Int J Gynecol Cancer. 27:3–10. 2017. View Article : Google Scholar : PubMed/NCBI

101 

Teng Y, Zhang Y, Qu K, Yang X, Fu J, Chen W and Li X: MicroRNA-29B (mir-29b) regulates the Warburg effect in ovarian cancer by targeting AKT2 and AKT3. Oncotarget. 6:40799–40814. 2015. View Article : Google Scholar : PubMed/NCBI

102 

Cao Q, Lu K, Dai S, Hu Y and Fan W: Clinicopathological and prognostic implications of the miR-200 family in patients with epithelial ovarian cancer. Int J Clin Exp Pathol. 7:2392–2401. 2014.PubMed/NCBI

103 

Kapetanakis NI, Uzan C, Jimenez-Pailhes AS, Gouy S, Bentivegna E, Morice P, Caron O, Gourzones-Dmitriev C, Le Teuff G and Busson P: Plasma miR-200b in ovarian carcinoma patients: Distinct pattern of pre/post-treatment variation compared to CA-125 and potential for prediction of progression-free survival. Oncotarget. 6:36815–36824. 2015. View Article : Google Scholar : PubMed/NCBI

104 

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

105 

Du Z and Sha X: Demethoxycurcumin inhibited human epithelia ovarian cancer cells' growth via up-regulating miR-551a. Tumour Biol. 39:10104283176943022017. View Article : Google Scholar : PubMed/NCBI

106 

Chen S, Chen X, Xiu YL, Sun KX and Zhao Y: MicroRNA-490-3P targets CDK1 and inhibits ovarian epithelial carcinoma tumorigenesis and progression. Cancer Lett. 362:122–130. 2015. View Article : Google Scholar : PubMed/NCBI

107 

Shuang T, Wang M, Shi C, Zhou Y and Wang D: Down-regulated expression of miR-134 contributes to paclitaxel resistance in human ovarian cancer cells. FEBS Lett. 589:3154–3164. 2015. View Article : Google Scholar : PubMed/NCBI

108 

Zou YT, Gao JY, Wang HL, Wang Y, Wang H and Li PL: Downregulation of microRNA-630 inhibits cell proliferation and invasion and enhances chemosensitivity in human ovarian carcinoma. Genet Mol Res. 14:8766–8777. 2015. View Article : Google Scholar : PubMed/NCBI

109 

Zhang H and Li W: Dysregulation of micro-143-3p and BALBP1 contributes to the pathogenesis of the development of ovarian carcinoma. Oncol Rep. 36:3605–3610. 2016. View Article : Google Scholar : PubMed/NCBI

110 

Zhang W, Zeng Q, Ban Z, Cao J, Chu T, Lei D, Liu C, Guo W and Zeng X: Effects of let-7c on the proliferation of ovarian carcinoma cells by targeted regulation of CDC25a gene expression. Oncol Lett. 16:5543–5550. 2018.PubMed/NCBI

111 

Liu MX, Siu MK, Liu SS, Yam JW, Ngan HY and Chan DW: Epigenetic silencing of microRNA-199b-5p is associated with acquired chemoresistance via activation of JAG1-Notch1 signaling in ovarian cancer. Oncotarget. 5:944–958. 2014. View Article : Google Scholar : PubMed/NCBI

112 

Kobayashi M, Sawada K, Nakamura K, Yoshimura A, Miyamoto M, Shimizu A, Ishida K, Nakatsuka E, Kodama M, Hashimoto K, et al: Exosomal miR-1290 is a potential biomarker of high-grade serous ovarian carcinoma and can discriminate patients from those with malignancies of other histological types. J Ovarian Res. 11:812018. View Article : Google Scholar : PubMed/NCBI

113 

Zhao H, Bi T, Qu Z, Jiang J, Cui S and Wang Y: Expression of miR-224-5p is associated with the original cisplatin resistance of ovarian papillary serous carcinoma. Oncol Rep. 32:1003–1012. 2014. View Article : Google Scholar : PubMed/NCBI

114 

Chen Y, Chen Q, Liu Q and Gao F: Human epididymis protein 4 expression positively correlated with miR-21 and served as a prognostic indicator in ovarian cancer. Tumour Biol. 37:8359–8365. 2016. View Article : Google Scholar : PubMed/NCBI

115 

Li L, Huang K, You Y, Fu X, Hu L, Song L and Meng Y: Hypoxia-induced miR-210 in epithelial ovarian cancer enhances cancer cell viability via promoting proliferation and inhibiting apoptosis. Int J Oncol. 44:2111–2120. 2014. View Article : Google Scholar : PubMed/NCBI

116 

Zhu X, Shen H, Yin X, Long L, Chen X, Feng F, Liu Y, Zhao P, Xu Y, Li M, et al: IL-6R/STAT3/miR-204 feedback loop contributes to cisplatin resistance of epithelial ovarian cancer cells. Oncotarget. 8:39154–39166. 2017. View Article : Google Scholar : PubMed/NCBI

117 

Fan Y, Fan J, Huang L, Ye M, Huang Z, Wang Y, Li Q and Huang J: Increased expression of microRNA-196a predicts poor prognosis in human ovarian carcinoma. Int J Clin Exp Pathol. 8:4132–4137. 2015.PubMed/NCBI

118 

Koukourakis MI, Kontomanolis E, Sotiropoulou M, Mitrakas A, Dafa E, Pouliliou S, Sivridis E and Giatromanolaki A: Increased soluble PD-L1 levels in the plasma of patients with epithelial ovarian cancer correlate with plasma levels of miR34a and miR200. Anticancer Res. 38:5739–5745. 2018. View Article : Google Scholar : PubMed/NCBI

119 

Liu J, Dou Y and Sheng M: Inhibition of microRNA-383 has tumor suppressive effect in human epithelial ovarian cancer through the action on caspase-2 gene. Biomed Pharmacother. 83:1286–1294. 2016. View Article : Google Scholar : PubMed/NCBI

120 

Dai F, Zhang Y and Chen Y: Involvement of miR-29b signaling in the sensitivity to chemotherapy in patients with ovarian carcinoma. Hum Pathol. 45:1285–1293. 2014. View Article : Google Scholar : PubMed/NCBI

121 

Xiao M, Cai J, Cai L, Jia J, Xie L, Zhu Y, Huang B, Jin D and Wang Z: Let-7e sensitizes epithelial ovarian cancer to cisplatin through repressing DNA double strand break repair. J Ovarian Res. 10:242017. View Article : Google Scholar : PubMed/NCBI

122 

Li X, Pan Q, Wan X, Mao Y, Lu W, Xie X and Cheng X: Methylation-associated Has-miR-9 deregulation in paclitaxel-resistant epithelial ovarian carcinoma. BMC Cancer. 15:5092015. View Article : Google Scholar : PubMed/NCBI

123 

Paudel D, Zhou W, Ouyang Y, Dong S, Huang Q, Giri R, Wang J and Tong X: MicroRNA-130b functions as a tumor suppressor by regulating RUNX3 in epithelial ovarian cancer. Gene. 586:48–55. 2016. View Article : Google Scholar : PubMed/NCBI

124 

Duan S, Dong X, Hai J, Jiang J, Wang W, Yang J, Zhang W and Chen C: MicroRNA-135a-3p is downregulated and serves as a tumour suppressor in ovarian cancer by targeting CCR2. Biomed Pharmacother. 107:712–720. 2018. View Article : Google Scholar : PubMed/NCBI

125 

Chen X, Dong C, Law PT, Chan MT, Su Z, Wang S, Wu WK and Xu H: MicroRNA-145 targets TRIM2 and exerts tumor-suppressing functions in epithelial ovarian cancer. Gynecol Oncol. 139:513–519. 2015. View Article : Google Scholar : PubMed/NCBI

126 

Qin CZ, Lou XY, Lv QL, Cheng L, Wu NY, Hu L and Zhou HH: MicroRNA-184 acts as a potential diagnostic and prognostic marker in epithelial ovarian cancer and regualtes cell proliferation, apoptosis and inflammation. Pharmazie. 70:668–673. 2015.PubMed/NCBI

127 

Liang T, Li L, Cheng Y, Ren C and Zhang G: MicroRNA-194 promotes the growth, migration, and invasion of ovarian carcinoma cells by targeting protein tyrosine phosphatase nonreceptor type 12. Onco Targets Ther. 9:4307–4315. 2016. View Article : Google Scholar : PubMed/NCBI

128 

Wei C, Zhang X, He S, Liu B, Han H and Sun X: MicroRNA-219-5p inhibits the proliferation, migration, and invasion of epithelial ovarian cancer cells by targeting the Twist/Wnt/β-catenin signaling pathway. Gene. 637:25–32. 2017. View Article : Google Scholar : PubMed/NCBI

129 

Fu X, Li Y, Alvero A, Li J, Wu Q, Xiao Q, Peng Y, Hu Y, Li X, Yan W, et al: MicroRNA-222-3p/GNAI2/AKT axis inhibits epithelial ovarian cancer cell growth and associates with good overall survival. Oncotarget. 7:80633–80654. 2016. View Article : Google Scholar : PubMed/NCBI

130 

Wu X, Ruan Y, Jiang H and Xu C: MicroRNA-424 inhibits cell migration, invasion, and epithelial mesenchymal transition by downregulating doublecortin-like kinase 1 in ovarian clear cell carcinoma. Int J Biochem Cell Biol. 85:66–74. 2017. View Article : Google Scholar : PubMed/NCBI

131 

Zhang J, Liu L, Sun Y, Xiang J, Zhou D, Wang L, Xu H, Yang X, Du N, Zhang M, et al: MicroRNA-520g promotes epithelial ovarian cancer progression and chemoresistance via DAPK2 repression. Oncotarget. 7:26516–26534. 2016. View Article : Google Scholar : PubMed/NCBI

132 

Zhang L, Li Z, Gai F and Wang Y: MicroRNA-137 suppresses tumor growth in epithelial ovarian cancer in vitro and in vivo. Mol Med Rep. 12:3107–3114. 2015. View Article : Google Scholar : PubMed/NCBI

133 

Liu J, Jin S and Wang R: MicroRNA-139 suppressed tumor cell proliferation, migration and invasion by directly targeting HDGF in epithelial ovarian cancer. Mol Med Rep. 16:3379–3386. 2017. View Article : Google Scholar : PubMed/NCBI

134 

Xu L, Li H, Su L, Lu Q and Liu Z: MicroRNA-455 inhibits cell proliferation and invasion of epithelial ovarian cancer by directly targeting Notch1. Mol Med Rep. 16:9777–9785. 2017. View Article : Google Scholar : PubMed/NCBI

135 

Yan J, Jiang J, Meng XN, Xiu YL and Zong ZH: MiR-23b targets cyclin G1 and suppresses ovarian cancer tumorigenesis and progression. J Exp Clin Cancer Res. 35:312016. View Article : Google Scholar : PubMed/NCBI

136 

Lin J, Zhang L, Huang H, Huang Y, Huang L, Wang J, Huang S, He L, Zhou Y, Jia W, et al: MiR-26b/KPNA2 axis inhibits epithelial ovarian carcinoma proliferation and metastasis through downregulating OCT4. Oncotarget. 6:23793–23806. 2015. View Article : Google Scholar : PubMed/NCBI

137 

Xu J, Jiang N, Shi H, Zhao S, Yao S and Shen H: miR-28-5p promotes the development and progression of ovarian cancer through inhibition of N4BP1. Int J Oncol. 50:1383–1391. 2017. View Article : Google Scholar : PubMed/NCBI

138 

Wang Y, Zhang X, Tang W, Lin Z, Xu L, Dong R, Li Y, Li J, Zhang Z, Li X, et al: miR-130a upregulates mTOR pathway by targeting TSC1 and is transactivated by NF-κB in high-grade serous ovarian carcinoma. Cell Death Differ. 24:2089–2100. 2017. View Article : Google Scholar : PubMed/NCBI

139 

Wang L, He J, Xu H, Xu L and Li N: MiR-143 targets CTGF and exerts tumor-suppressing functions in epithelial ovarian cancer. Am J Transl Res. 8:2716–2726. 2016.PubMed/NCBI

140 

Dong M, Yang P and Hua F: miR-191 modulates malignant transformation of endometriosis through regulating TIMP3. Med Sci Monit. 21:915–920. 2015. View Article : Google Scholar : PubMed/NCBI

141 

Niu K, Shen W, Zhang Y, Zhao Y and Lu Y: MiR-205 promotes motility of ovarian cancer cells via targeting ZEB1. Gene. 574:330–336. 2015. View Article : Google Scholar : PubMed/NCBI

142 

Dai C, Xie Y, Zhuang X and Yuan Z: MiR-206 inhibits epithelial ovarian cancer cells growth and invasion via blocking c-Met/AKT/mTOR signaling pathway. Biomed Pharmacother. 104:763–770. 2018. View Article : Google Scholar : PubMed/NCBI

143 

Xia B, Yang S, Liu T and Lou G: miR-211 suppresses epithelial ovarian cancer proliferation and cell-cycle progression by targeting Cyclin D1 and CDK6. Mol Cancer. 14:572015. View Article : Google Scholar : PubMed/NCBI

144 

Wu Q, Ren X, Zhang Y, Fu X, Li Y, Peng Y, Xiao Q, Li T, Ouyang C, Hu Y, et al: MiR-221-3p targets ARF4 and inhibits the proliferation and migration of epithelial ovarian cancer cells. Biochem Biophys Res Commun. 497:1162–1170. 2018. View Article : Google Scholar : PubMed/NCBI

145 

Cao L, Wan Q, Li F and Tang C: MiR-363 inhibits cisplatin chemoresistance of epithelial ovarian cancer by regulating snail-induced epithelial-mesenchymal transition. BMB Rep. 51:456–461. 2018. View Article : Google Scholar : PubMed/NCBI

146 

Xia B, Li H, Yang S, Liu T and Lou G: MiR-381 inhibits epithelial ovarian cancer malignancy via YY1 suppression. Tumour Biol. 37:9157–9167. 2016. View Article : Google Scholar : PubMed/NCBI

147 

Yuan J, Wang K and Xi M: MiR-494 inhibits epithelial ovarian cancer growth by targeting c-Myc. Med Sci Monit. 22:617–624. 2016. View Article : Google Scholar : PubMed/NCBI

148 

Li N, Zhao X, Wang L, Zhang S, Cui M and He J: miR-494 suppresses tumor growth of epithelial ovarian carcinoma by targeting IGF1R. Tumour Biol. 37:7767–7776. 2016. View Article : Google Scholar : PubMed/NCBI

149 

Zhou QH, Zhao YM, JIA LL and Zhang Y: Mir-595 is a significant indicator of poor patient prognosis in epithelial ovarian cancer. Eur Rev Med Pharmacol Sci. 21:4278–4282. 2017.PubMed/NCBI

150 

Zhang S, Zhang JY, Lu LJ, Wang CH and Wang LH: MiR-630 promotes epithelial ovarian cancer proliferation and invasion via targeting KLF6. Eur Rev Med Pharmacol Sci. 21:4542–4547. 217.PubMed/NCBI

151 

Shi C and Zhang Z: miR-761 inhibits tumor progression by targeting MSI1 in ovarian carcinoma. Tumour Biol. 37:5437–5443. 2016. View Article : Google Scholar : PubMed/NCBI

152 

Xie X, Huang Y, Chen L and Wang J: miR-221 regulates proliferation and apoptosis of ovarian cancer cells by targeting BMF. Oncol Lett. 16:6697–6704. 2018.PubMed/NCBI

153 

Wen C, Liu X, Ma H, Zhang W and Li H: miR-338-3p suppresses tumor growth of ovarian epithelial carcinoma by targeting Runx2. Int J Oncol. 46:2277–2285. 2015. View Article : Google Scholar : PubMed/NCBI

154 

Salem M, O'Brien JA, Bernaudo S, Shawer H, Ye G, Brkić J, Amleh A, Vanderhyden BC, Refky B, Yang BB, et al: miR-590-3p promotes ovarian cancer growth and metastasis via a Novel FOXA2-versican pathway. Cancer Res. 78:4175–4190. 2018. View Article : Google Scholar : PubMed/NCBI

155 

Lin Z, Zhao J, Wang X, Zhu X and Gong L: Overexpression of microRNA-497 suppresses cell proliferation and induces apoptosis through targeting paired box 2 in human ovarian cancer. Oncol Rep. 36:2101–2107. 2016. View Article : Google Scholar : PubMed/NCBI

156 

Lin M, Xia B, Qin L, Chen H and Lou G: S100A7 regulates ovarian cancer cell metastasis and chemoresistance through MAPK signaling and is targeted by miR-330-5p. DNA Cell Biol. 37:491–500. 2018. View Article : Google Scholar : PubMed/NCBI

157 

Chen JL, Chen F, Zhang TT and Liu NF: Suppression of SIK1 by miR-141 in human ovarian cancer cell lines and tissues. Int J Mol Med. 37:1601–1610. 2016. View Article : Google Scholar : PubMed/NCBI

158 

Zuberi M, Khan I, Gandhi G, Ray PC and Saxena A: The conglomeration of diagnostic, prognostic and therapeutic potential of serum miR-199a and its association with clinicopathological features in epithelial ovarian cancer. Tumour Biol. 37:11259–11266. 2016. View Article : Google Scholar : PubMed/NCBI

159 

Guan X, Zong ZH, Chen S, Sang XB, Wu DD, Wang LL, Liu Y and Zhao Y: The role of miR-372 in ovarian carcinoma cell proliferation. Gene. 624:14–20. 2017. View Article : Google Scholar : PubMed/NCBI

160 

Li J, Li D and Zhang W: Tumor suppressor role of miR-217 in human epithelial ovarian cancer by targeting IGF1R. Oncol Rep. 35:1671–1679. 2016. View Article : Google Scholar : PubMed/NCBI

161 

Zhang X, Liu J, Zang D, Wu S, Liu A, Zhu J, Wu G, Li J and Jiang L: Upregulation of miR-572 transcriptionally suppresses SOCS1 and p21 and contributes to human ovarian cancer progression. Oncotarget. 6:15180–15193. 2015. View Article : Google Scholar : PubMed/NCBI

162 

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

163 

Zhang X, Li S, Dong C, Xie X and Zhang Y: Knockdown of long noncoding RNA NR_026689 inhibits proliferation and invasion and increases apoptosis in ovarian carcinoma HO-8910PM cells. Oncol Res. 25:259–265. 2017. View Article : Google Scholar : PubMed/NCBI

164 

Zhu L, Guo Q, Lu X, Zhao J, Shi J, Wang Z and Zhou X: CTD-2020K17.1, a novel long non-coding RNA, promotes migration, invasion, and proliferation of serous ovarian cancer cells in vitro. Med Sci Monit. 24:1329–1339. 2018. View Article : Google Scholar : PubMed/NCBI

165 

Qiu JJ, Zhang XD, Tang XY, Zheng TT, Zhang Y and Hua KQ: ElncRNA1, a long non-coding RNA that is transcriptionally induced by oestrogen, promotes epithelial ovarian cancer cell proliferation. Int J Oncol. 51:507–514. 2017. View Article : Google Scholar : PubMed/NCBI

166 

Gao Y, Meng H, Liu S, Hu J, Zhang Y, Jiao T, Liu Y, Ou J, Wang D, Yao L, et al: LncRNA-HOST2 regulates cell biological behaviors in epithelial ovarian cancer through a mechanism involving microRNA let-7b. Hum Mol Genet. 24:841–852. 2015. View Article : Google Scholar : PubMed/NCBI

167 

Wang Y, Wang H, Song T, Zou Y, Jiang J, Fang L and Li P: HOTAIR is a potential target for the treatment of cisplatin-resistant ovarian cancer. Mol Med Rep. 12:2211–2216. 2015. View Article : Google Scholar : PubMed/NCBI

168 

Lu CW, Zhou DD, Xie T, Hao JL, Pant OP, Lu CB and Liu XF: HOXA11 antisense long noncoding RNA (HOXA11-AS): A promising lncRNA in human cancers. Cancer Med. 7:3792–3799. 2018. View Article : Google Scholar : PubMed/NCBI

169 

Du W, Feng Z and Sun Q: LncRNA LINC00319 accelerates ovarian cancer progression through miR-423-5p/NACC1 pathway. Biochem Biophys Res Commun. 507:198–202. 2018. View Article : Google Scholar : PubMed/NCBI

170 

Shu C, Yan D, Mo Y, Gu J, Shah N and He J: Long noncoding RNA lncARSR promotes epithelial ovarian cancer cell proliferation and invasion by association with HuR and miR-200 family. Am J Cancer Res. 8:981–992. 2018.PubMed/NCBI

171 

Chen S, Wu DD, Sang XB, Wang LL, Zong ZH, Sun KX, Liu BL and Zhao Y: The lncRNA HULC functions as an oncogene by targeting ATG7 and ITGB1 in epithelial ovarian carcinoma. Cell Death Dis. 8:e31182017. View Article : Google Scholar : PubMed/NCBI

172 

Qnbo L, Guan W, Ren W, Zhang L, Zhang J and Xu G: MALAT1 affects ovarian cancer cell behavior and patient survival. Oncol Rep. 39:2644–2652. 2018.PubMed/NCBI

173 

Lin Q, Guan W, Ren W, Zhang L, Zhang J and Xu G: MALAT1 affects ovarian cancer cell behavior and patient survival. Oncol Rep. 39:2644–2652. 2018.PubMed/NCBI

174 

Yan C, Jiang Y, Wan Y, Zhang L, Liu J, Zhou S and Cheng W: Long noncoding RNA NBAT-1 suppresses tumorigenesis and predicts favorable prognosis in ovarian cancer. Onco Targets Ther. 10:1993–2002. 2017. View Article : Google Scholar : PubMed/NCBI

175 

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

176 

Chen S, Wang LL, Sun KX, Liu Y, Guan X, Zong ZH and Zhao Y: LncRNA PCGEM1 induces ovarian carcinoma tumorigenesis and progression through RhoA pathway. Cell Physiol Biochem. 47:1578–1588. 2018. View Article : Google Scholar : PubMed/NCBI

177 

Huang K, Geng J and Wang J: Long non-coding RNA RP11-552M11.4 promotes cells proliferation, migration and invasion by targeting BRCA2 in ovarian cancer. Cancer Sci. 109:1428–1446. 2018. View Article : Google Scholar : PubMed/NCBI

178 

Li H, Liu C, Lu Z, Chen L, Wang J, Li Y and Ma H: Upregulation of the long non-coding RNA SPRY4-IT1 indicates a poor prognosis and promotes tumorigenesis in ovarian cancer. Biomed Pharmacother. 88:529–534. 2017. View Article : Google Scholar : PubMed/NCBI

179 

Li TH, Zhang JJ, Liu SX and Chen Y: Long non-coding RNA taurine-upregulated gene 1 predicts unfavorable prognosis, promotes cells proliferation, and inhibits cells apoptosis in epithelial ovarian cancer. Medicine (Baltimore). 97:e05752018. View Article : Google Scholar : PubMed/NCBI

180 

Hong HH, Hou LK, Pan X, Wu CY, Huang H, Li B and Nie W: Long non-coding RNA UCA1 is a predictive biomarker of cancer. Oncotarget. 7:44442–44447. 2016. View Article : Google Scholar : PubMed/NCBI

181 

Zhang L, Cao X, Zhang L, Zhang X, Sheng H and Tao K: UCA1 overexpression predicts clinical outcome of patients with ovarian cancer receiving adjuvant chemotherapy. Cancer Chemother Pharmacol. 77:629–634. 2016. View Article : Google Scholar : PubMed/NCBI

182 

Qiu JJ, Wang Y, Liu YL, Zhang Y, Ding JX and Hua KQ: The long non-coding RNA ANRIL promotes proliferation and cell cycle progression and inhibits apoptosis and senescence in epithelial ovarian cancer. Oncotarget. 7:32478–32492. 2016. View Article : Google Scholar : PubMed/NCBI

183 

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

184 

Hua F, Li CH, Chen XG and Liu XP: Long Noncoding RNA CCAT2 knockdown suppresses tumorous progression by sponging miR-424 in epithelial ovarian cancer. Oncol Res. 26:241–247. 2018. View Article : Google Scholar : PubMed/NCBI

185 

Yim GW, Kim HJ, Kim LK, Kim SW, Kim S, Nam EJ and Kim YT: Long Non-coding RNA HOXA11 antisense promotes cell proliferation and invasion and predicts patient prognosis in serous ovarian cancer. Cancer Res Treat. 49:656–668. 2017. View Article : Google Scholar : PubMed/NCBI

186 

Koutsaki M, Spandidos DA and Zaravinos A: Epithelial-mesenchymal transition-associated miRNAs in ovarian carcinoma, with highlight on the miR-200 family: Prognostic value and prospective role in ovarian cancer therapeutics. Cancer Lett. 351:173–181. 2014. View Article : Google Scholar : PubMed/NCBI

187 

Sulaiman SA, Ab Mutalib NS and Jamal R: miR-200c regulation of metastases in ovarian cancer: Potential role in epithelial and mesenchymal transition. Front Pharmacol. 7:2712016. View Article : Google Scholar : PubMed/NCBI

188 

Teng Y, Su X, Zhang X, Zhang Y, Li C, Niu W, Liu C and Qu K: miRNA-200a/c as potential biomarker in epithelial ovarian cancer (EOC): Evidence based on miRNA meta-signature and clinical investigations. Oncotarget. 7:81621–81633. 2016. View Article : Google Scholar : PubMed/NCBI

189 

Muralidhar G and Barbolina M: The miR-200 Family: Versatile players in epithelial ovarian cancer. Int J Mol Sci. 16:16833–16847. 2015. View Article : Google Scholar : PubMed/NCBI

190 

Zuberi M, Khan I, Mir R, Gandhi G, Ray PC and Saxena A: Utility of serum miR-125b as a diagnostic and prognostic indicator and its alliance with a panel of tumor suppressor genes in epithelial ovarian cancer. PLoS One. 11:e01539022016. View Article : Google Scholar : PubMed/NCBI

191 

Faul C, Gerszten K, Edwards R, Land S, D'Angelo G, Kelley J III and Price F: A phase I/II study of hypofractionated whole abdominal radiation therapy in patients with chemoresistant ovarian carcinoma: Karnofsky score determines treatment outcome. Int J Radiat Oncol Biol Phys. 47:749–754. 2000. View Article : Google Scholar : PubMed/NCBI

192 

Iorio GC, Martini S, Arcadipane F, Ricardi U and Franco P: The role of radiotherapy in epithelial ovarian cancer: A literature overview. Med Oncol. 36:642019. View Article : Google Scholar : PubMed/NCBI

193 

Sorbe B: Consolidation treatment of advanced ovarian carcinoma with radiotherapy after induction chemotherapy. Int J Gynecol Cancer. 13 (Suppl 2):S192–S195. 2003. View Article : Google Scholar

194 

Pang L and Guo Z: Differences in characteristics and outcomes between large-cell neuroendocrine carcinoma of the ovary and high-grade serous ovarian cancer: A retrospective observational cohort study. Front Oncol. 12:8916992022. View Article : Google Scholar : PubMed/NCBI

195 

Patel SC, Frandsen J, Bhatia S and Gaffney D: Impact on survival with adjuvant radiotherapy for clear cell, mucinous, and endometriod ovarian cancer: The SEER experience from 2004 to 2011. J Gynecol Oncol. 27:e452016. View Article : Google Scholar : PubMed/NCBI

196 

Pestell KE, Medlow CJ, Titley JC, Kelland LR and Walton MI: Characterisation Of The P53 Status, Bcl-2 expression and radiation and platinum drug sensitivity of a panel of human ovarian cancer cell lines. Int J Cancer. 77:913–918. 1998. View Article : Google Scholar : PubMed/NCBI

197 

Zielske SP: Epigenetic DNA methylation in radiation biology: On the field or on the sidelines? J Cell Biochem. 116:212–217. 2015. View Article : Google Scholar : PubMed/NCBI

198 

Kurrey NK, Jalgaonkar SP, Joglekar AV, Ghanate AD, Chaskar PD, Doiphode RY and Bapat SA: Snail and slug mediate radioresistance and chemoresistance by antagonizing p53-Mediated apoptosis and acquiring a stem-like phenotype in ovarian cancer cells. Stem Cells. 27:2059–2068. 2009. View Article : Google Scholar : PubMed/NCBI

199 

Liu WJ, Huang YX, Wang W, Zhang Y, Liu BJ, Qiu JG, Jiang BH and Liu LZ: NOX4 signaling mediates cancer development and therapeutic resistance through HER3 in ovarian cancer cells. Cells. 10:16472021. View Article : Google Scholar : PubMed/NCBI

200 

Chen J, Jia Y, Jia ZH, Zhu Y and Jin YM: Silencing the expression of MTDH increases the radiation sensitivity of SKOV3 ovarian cancer cells and reduces their proliferation and metastasis. Int J Oncol. 53:2180–2190. 2018.PubMed/NCBI

201 

Zhao Y, Liu S, Wen Y and Zhong L: Effect of MicroRNA-210 on the growth of ovarian cancer cells and the efficacy of radiotherapy. Gynecol Obstet Invest. 86:71–80. 2021. View Article : Google Scholar : PubMed/NCBI

202 

Xing Y, Cui D, Wang S, Wang P, Xing X and Li H: Oleuropein represses the radiation resistance of ovarian cancer by inhibiting hypoxia and microRNA-299-targetted heparanase expression. Food Funct. 8:2857–2864. 2017. View Article : Google Scholar : PubMed/NCBI

203 

Marques C, Ferreira da Silva F, Sousa I and Nave M: Chemotherapy-free treatment of recurrent advanced ovarian cancer: Myth or reality? Int J Gynecol Cancer. 33:607–618. 2023. View Article : Google Scholar : PubMed/NCBI

204 

Marchetti C, De Felice F, Romito A, Iacobelli V, Sassu CM, Corrado G, Ricci C, Scambia G and Fagotti A: Chemotherapy resistance in epithelial ovarian cancer: Mechanisms and emerging treatments. Semin Cancer Biol. 77:144–166. 2021. View Article : Google Scholar : PubMed/NCBI

205 

Falzone L, Bordonaro R and Libra M: SnapShot: Cancer chemotherapy. Cell. 186:1816–1816.e1. 2023. View Article : Google Scholar

206 

Raab M, Sanhaji M, Zhou S, Rödel F, El-Balat A, Becker S and Strebhardt K: Blocking mitotic exit of ovarian cancer cells by pharmaceutical inhibition of the anaphase-promoting complex reduces chromosomal instability. Neoplasia. 21:363–375. 2019. View Article : Google Scholar : PubMed/NCBI

207 

Swanton C, Nicke B, Schuett M, Eklund AC, Ng C, Li Q, Hardcastle T, Lee A, Roy R, East P and Kschischo M: Chromosomal instability determines taxane response. Proc Natl Acad Sci USA. 106:8671–8676. 2009. View Article : Google Scholar : PubMed/NCBI

208 

Pradhan M, Risberg BÅ, Tropé CG, van de Rijn M, Gilks CB and Lee CH: Gross genomic alterations and gene expression profiles of high-grade serous carcinoma of the ovary with and without BRCA1 inactivation. BMC Cancer. 10:4932010. View Article : Google Scholar : PubMed/NCBI

209 

Tang Z, Yang J, Wang X, Zeng M, Wang J, Wang A, Zhao M, Guo L, Liu C, Li D and Chen J: Active DNA end processing in micronuclei of ovarian cancer cells. BMC Cancer. 18:4262018. View Article : Google Scholar : PubMed/NCBI

210 

Morden CR, Farrell AC, Sliwowski M, Lichtensztejn Z, Altman AD, Nachtigal MW and McManus KJ: Chromosome instability is prevalent and dynamic in high-grade serous ovarian cancer patient samples. Gynecol Oncol. 161:769–778. 2021. View Article : Google Scholar : PubMed/NCBI

211 

Gorringe KL, Chin SF, Pharoah P, Staines JM, Oliveira C, Edwards PA and Caldas C: Evidence that both genetic instability and selection contribute to the accumulation of chromosome alterations in cancer. Carcinogenesis. 26:923–930. 2005. View Article : Google Scholar : PubMed/NCBI

212 

Bayani J, Paderova J, Murphy J, Rosen B, Zielenska M and Squire JA: Distinct patterns of structural and numerical chromosomal instability characterize sporadic ovarian cancer. Neoplasia. 10:1057–1065. 2008. View Article : Google Scholar : PubMed/NCBI

213 

Birkbak NJ, Eklund AC, Li Q, McClelland SE, Endesfelder D, Tan P, Tan IB, Richardson AL, Szallasi Z and Swanton C: Paradoxical relationship between chromosomal instability and survival outcome in cancer. Cancer Res. 71:3447–3452. 2011. View Article : Google Scholar : PubMed/NCBI

214 

Hille S, Rein DT, Riffelmann M, Neumann R, Sartorius J, Pfützner A, Kurbacher CM, Schöndorf T and Breidenbach M: Anticancer drugs induce mdr1 gene expression in recurrent ovarian cancer. Anticancer Drugs. 17:1041–1044. 2006. View Article : Google Scholar : PubMed/NCBI

215 

Zhang C, Wang M, Shi C, Shi F and Pei C: Long non-coding RNA Linc00312 modulates the sensitivity of ovarian cancer to cisplatin via the Bcl-2/Caspase-3 signaling pathway. Biosci Trends. 12:309–316. 2018. View Article : Google Scholar : PubMed/NCBI

216 

Cui Y, Qin L, Tian D, Wang T, Fan L, Zhang P and Wang Z: ZEB1 Promotes chemoresistance to cisplatin in ovarian cancer cells by suppressing SLC3A2. Chemotherapy. 63:262–271. 2018. View Article : Google Scholar : PubMed/NCBI

217 

Sen T, Sen N, Brait M, Begum S, Chatterjee A, Hoque MO, Ratovitski E and Sidransky D: Np63 confers tumor cell resistance to cisplatin through the AKT1 transcriptional regulation. Cancer Res. 71:1167–1176. 2011. View Article : Google Scholar : PubMed/NCBI

218 

Kumar S, Kumar A, Shah PP, Rai SN, Panguluri SK and Kakar SS: MicroRNA signature of cis-platin resistant vs. cis-platin sensitive ovarian cancer cell lines. J Ovarian Res. 4:172011. View Article : Google Scholar : PubMed/NCBI

219 

Leung AWY, Veinotte CJ, Melong N, Melong N, Oh MH, Chen K, Enfield KSS, Backstrom I, Warburton C, Yapp D, et al: In vivo validation of PAPSS1 (3′-phosphoadenosine 5′-phosphosulfate synthase 1) as a cisplatin-sensitizing therapeutic target. Clin Cancer Res. 23:6555–6566. 2017. View Article : Google Scholar : PubMed/NCBI

220 

Kritsch D, Hoffmann F, Steinbach D, Jansen L, Mary Photini S, Gajda M, Mosig AS, Sonnemann J, Peters S, Melnikova M, et al: Tribbles 2 mediates cisplatin sensitivity and DNA damage response in epithelial ovarian cancer. Int J Cancer. 141:1600–1614. 2017. View Article : Google Scholar : PubMed/NCBI

221 

Nam EJ, Kim S, Lee TS, Kim HJ, Lee JY, Kim SW, Kim JH and Kim YT: Primary and recurrent ovarian high-grade serous carcinomas display similar microRNA expression patterns relative to those of normal ovarian tissue. Oncotarget. 7:70524–70534. 2016. View Article : Google Scholar : PubMed/NCBI

222 

Chong GO, Jeon HS, Han HS, Son JW, Lee YH, Hong DG, Lee YS and Cho Y: Differential MicroRNA expression profiles in primary and recurrent epithelial ovarian cancer. Anticancer Res. 7:2611–2617. 2015.

223 

Chong GO, Jeon HS, Han HS, Son JW, Lee YH, Hong DG, Park HJ, Lee YS and Cho YL: Overexpression of microRNA-196b accelerates invasiveness of cancer cells in recurrent epithelial ovarian cancer through regulation of homeobox A9. Cancer Genomics Proteomics. 14:137–142. 2017. View Article : Google Scholar : PubMed/NCBI

224 

Zhou Y, Wang M, Wu J, Jie Z, Chang S and Shuang T: The clinicopathological significance of miR-1307 in chemotherapy resistant epithelial ovarian cancer. J Ovarian Res. 8:232015. View Article : Google Scholar : PubMed/NCBI

225 

Chen C, Hu Y and Li L: NRP1 is targeted by miR-130a and miR-130b, and is associated with multidrug resistance in epithelial ovarian cancer based on integrated gene network analysis. Mol Med Rep. 13:188–196. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2025
Volume 31 Issue 6

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Del Castillo Falconi VM, Godinez Rodriguez JA, Fragoso‑Ontiveros V, Contreras‑Espinosa L, Pedroza‑Torres A, Díaz‑Chávez J and Herrera LA: Role of DNA methylation and non‑coding RNAs expression in pathogenesis, detection, prognosis, and therapy‑resistant ovarian carcinoma (Review). Mol Med Rep 31: 144, 2025.
APA
Del Castillo Falconi, V.M., Godinez Rodriguez, J.A., Fragoso‑Ontiveros, V., Contreras‑Espinosa, L., Pedroza‑Torres, A., Díaz‑Chávez, J., & Herrera, L.A. (2025). Role of DNA methylation and non‑coding RNAs expression in pathogenesis, detection, prognosis, and therapy‑resistant ovarian carcinoma (Review). Molecular Medicine Reports, 31, 144. https://doi.org/10.3892/mmr.2025.13509
MLA
Del Castillo Falconi, V. M., Godinez Rodriguez, J. A., Fragoso‑Ontiveros, V., Contreras‑Espinosa, L., Pedroza‑Torres, A., Díaz‑Chávez, J., Herrera, L. A."Role of DNA methylation and non‑coding RNAs expression in pathogenesis, detection, prognosis, and therapy‑resistant ovarian carcinoma (Review)". Molecular Medicine Reports 31.6 (2025): 144.
Chicago
Del Castillo Falconi, V. M., Godinez Rodriguez, J. A., Fragoso‑Ontiveros, V., Contreras‑Espinosa, L., Pedroza‑Torres, A., Díaz‑Chávez, J., Herrera, L. A."Role of DNA methylation and non‑coding RNAs expression in pathogenesis, detection, prognosis, and therapy‑resistant ovarian carcinoma (Review)". Molecular Medicine Reports 31, no. 6 (2025): 144. https://doi.org/10.3892/mmr.2025.13509