Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 3764 2024-01-17 23:20:48 |
2 layout & references Meta information modification 3764 2024-01-18 02:16:55 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Wilczyński, J.; Paradowska, E.; Wilczyńska, J.; Wilczyński, M. Cancer Tissue-Derived and Peripheral Blood Biomarkers in HGSOC. Encyclopedia. Available online: https://encyclopedia.pub/entry/53995 (accessed on 09 May 2024).
Wilczyński J, Paradowska E, Wilczyńska J, Wilczyński M. Cancer Tissue-Derived and Peripheral Blood Biomarkers in HGSOC. Encyclopedia. Available at: https://encyclopedia.pub/entry/53995. Accessed May 09, 2024.
Wilczyński, Jacek, Edyta Paradowska, Justyna Wilczyńska, Miłosz Wilczyński. "Cancer Tissue-Derived and Peripheral Blood Biomarkers in HGSOC" Encyclopedia, https://encyclopedia.pub/entry/53995 (accessed May 09, 2024).
Wilczyński, J., Paradowska, E., Wilczyńska, J., & Wilczyński, M. (2024, January 17). Cancer Tissue-Derived and Peripheral Blood Biomarkers in HGSOC. In Encyclopedia. https://encyclopedia.pub/entry/53995
Wilczyński, Jacek, et al. "Cancer Tissue-Derived and Peripheral Blood Biomarkers in HGSOC." Encyclopedia. Web. 17 January, 2024.
Cancer Tissue-Derived and Peripheral Blood Biomarkers in HGSOC
Edit

High-grade serous ovarian cancer (HGSOC) is one of the most lethal tumors generally and the most fatal cancer of the female genital tract. The approved standard therapy consists of surgical cytoreduction and platinum/taxane-based chemotherapy, and of targeted therapy in selected patients. The main therapeutic problem is chemoresistance of recurrent and metastatic HGSOC tumors which results in low survival in the group of FIGO III/IV. Therefore, the prediction and monitoring of chemoresistance seems to be of utmost importance for the improvement of HGSOC management. 

ovarian cancer prediction biomarkers chemoresistance

1. Introduction

High-grade serous ovarian cancer (HGSOC) is the most lethal tumor of the female genital tract due to lack of screening programs for the average-risk population, followed by the delayed diagnosis. It has also a high proliferative potential and recurrence rate. Therefore, the 5-year survival in the advanced patient population (clinical stages III–IV) is unsatisfactory, although recently this has been improved by the introduction of poly-ADP-ribose polymerase (PARP) inhibitors in the group of homologous recombination deficiency (HRD)-positive tumors (data of American Cancer Society 2020. https://www.cancer.org/cancer/ovarian-cancer/detection-diagnosis-staging/survival-rates.html, accessed on 10 August 2023) [1][2]. The approved standard therapy consists of primary cytoreductive surgery followed by standard platinum/taxane-based chemotherapy or, alternatively, in inoperable tumors, of neo-adjuvant platinum/taxane chemotherapy followed by interval cytoreduction. In the case of advanced and sub-optimally operated tumors, anti-VEGF (vascular–endothelial growth factor) humanized monoclonal antibody bevacizumab has been approved, while, in the group of HRD-deficient patients, the poly-ADP ribose polymerase (PARP) inhibitors are used, with PARP-based therapy showing satisfactory efficacy [2][3][4][5][6][7][8]. In phase III, PAOLA-1/ENGOT-ov25 trial patients with the BRCA-mutated tumors benefitted from maintenance therapy using the PARP inhibitor olaparib + bevacizumab versus placebo + bevacizumab (24-month progression-free survival PFS 89% vs. 15%) [9]. In the VELIA study, complete response rates among patients’ residual disease after cytoreduction were 24% in the PARP inhibitor veliparib group compared with 18% in the controls [10]. The PRIMA/ENGOT-ov26/GOG-3012 trial showed that, in newly diagnosed advanced ovarian cancer, therapy with the PARP inhibitor niraparib was effective compared with placebo irrespectively of HRD status (HRD-positive 38% vs. 17%, overall 24% vs. 14%) [8]. However, resistance to PARP inhibitors has unfortunately been observed [11]. High-grade serous ovarian cancer is chemo-responsive to the first line of standard platinum and taxane-based chemotherapy, particularly in the group of BRCA-mutated patients. However, the main therapeutic problem appears when chemotherapy is used to treat the patent or recurrent disease, as well as in the cases of primary chemo-refractoriness. In these cases, treatment is eventually ineffective and the disease is usually lethal. Therefore, identification of the biomarkers of drug resistance is one of the most desirable activities in ovarian cancer surveillance and therapy. Chemoresistance is regulated by many different mechanisms originating both from the cancer cells themselves and from the tumor microenvironment (TME). Recent studies have brought much information about genetic HGSOC heterogeneity, disturbed epigenetic regulation, and the modulating role of TME. Tumors of different genetic or epigenetic signature and TME composition are most probably characterized by diverse drug responses [12][13][14][15]. The prediction concerning the drug resistance of the primary, metastatic, and recurrent tumors should be an indispensable element of the personalized anti-cancer therapy. As chemosensitivity could change depending on both localization (different TME) and time in the course of the disease, repetitive monitoring of the chemosensitivity during therapy should be another step in the management of HGSOC patients.

2. Analysis of Cancer Tissue-Derived and Peripheral Blood Biomarkers

2.1. Genetic and Proteomic Biomarkers

The problem of chemoresistance concerns not only the chemotherapeutics but also drugs used in the targeted therapy. The classic biomarkers of platinum and PARP sensitivity are the germinal and somatic mutations of BRCA1/2 genes and the HRD status of the tumor, respectively [16]. The reverse mutations in BRCA genes are able to switch chemosensitivity into chemoresistance. BRCA reverse mutations were identified in cfDNA of 18% of pre-treatment patients with platinum-refractory HGSOC cancer and in cfDNA of 13% of platinum-resistant tumors. Patients without reverse mutations had better outcomes during rucaparib therapy [17]. Reverse mutations in other homologous recombination repair (HR) genes like RAD51C, RAD51D, and PALB2 are also responsible for secondary resistance to platinum- and PARP inhibitor-based therapy [17][18]. The highly expressed PARP, Fanconi Anemia complementation group D2 (FANCD2) and p53 proteins are the feature of BRCAness phenotype and are positively correlated with platinum resistance [19]. The drug-resistant and drug-sensitive recurrent ovarian cancers were shown to possess unique genetic alterations when studied by ctDNA liquid biopsy. TP53 was the most frequently mutated gene in both groups. Copy number variations of MYC, RB1, and phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) were noticed in recurrent cancers, while BRCA2 N372H polymorphism was noticed in recurrent drug-resistant tumors [20]. Whole-genome sequencing (WGS) analysis of ctDNA in heavily pre-treated HGSOC patients confirmed that the most mutated gene was TP53 (94% of HGSOC cases). The WGS detected mutated TP53 ctDNA in 88% of cases, but there was a low correlation between plasma and tumor copy number alteration profiles; however, it was high in the subgroup with the highest ctDNA tumor fraction. Both the genome-altered fraction and plasma mutation burden had prognostic value according to chemoresistance and survival [21]. More than half of HGSOC have a defective HR pathway, but in tumors with intact HR, an amplification or gain of cyclin E1 (CCNE1) was observed and was related to chemoresistance and decreased OS [22]. Based on the HRD score, gene insertions and deletions, copy number changes load, duplication load, single nucleotide variants, and mutational signatures, a predictor of platinum-resistance, named DRD score, were established and validated in a cohort of HGSOC patients reaching the sensitivity of 91% [23]. Cytogenetic analysis indicated that several losses (13q32.1 and 8p21.1, 8p and 9q) or gains (1q, 5q14~q23, and 13q21~q32, 9p13.2–13.1, 9q21.2–21.32, 9q22.2–22.31, 9q22.32–22.33, and 9q33.1–34.11) in chromosomal regions were connected to chemoresistance [24][25][26][27]. The presence of several SNPs, between others, rs4910232 (11p15.3), rs2549714 (16q23), and rs6674079 (1q22), was also connected to poor response for first-line platinum-based chemotherapy and unfavorable outcome in ovarian cancer patients [28]. Mutations in the members of the A disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS) family were associated with a significantly higher chemosensitivity (100% for ADAMTS-mutated vs. 64% for ADAMTS wild-type cases), and with significantly better OS and PFS [29]. In the patient-derived xenograft mouse model of ovarian cancer, the differential expression of Sin3A-associated protein 25 (SAP25), major histocompatibility complex, class II, DP alpha 1 (HLA-DPA1), AKT serine/threonine kinase 3 (AKT3), and phosphoinositide-3-kinase regulatory subunit 5 (PIK3R5) genes and mutation of transmembrane protein 205 (TMEM205) and DNA-directed RNA polymerase II subunit RPB1 (POLR2A) coding genes were found to have unfavorable function in the induction of chemoresistance [30]. Conversely, transcriptome analysis identified interferon regulatory and tumor suppression factor 1 (IRF1) transcription factor as a supporter of platinum sensitivity in HGSOC [31]. Study of gene networks and expression of quantitative trait loci (eQTLs) indicated that many of the mapped genes associated to chemoresistance in ovarian cancer were located in chromosome 9, which supported the previous observations of the connection between gene alterations in that region with progression and chemoresistance of ovarian tumors [32][33]. A total of 96% of genes were co-regulated by the same transcription factor organic cation transporter 1 (OCT1) which is engaged in disturbances of platinum-induced apoptosis. Another important protein was valosin-containing protein (VCP) which played a critical role in the disintegration of polypeptide cellular structures. Low expression of VCP is found in ovarian cancer cells, especially from platinum-resistant cell lines and ovarian cancer cohorts [32][34]. Other genes which correlated to chemoresistance were BRCA2 and neighboring NEDD4 binding protein 2-like 1 (N4BP2L1), -like 2 (N4BP2L2), FRY microtubule binding protein (FRY), and StAR related lipid transfer domain containing 13 (STARD13) genes [26]. The BRCA2 and STARD13 are tumor-suppressors, while up-regulation of N4BP2L1 and N4BP2L2 was associated with positive prediction in ovarian cancer patients [35]. The up-regulation of BRCA2 was observed in chemotherapy resistant patients, and the down-regulation of BRCA2 reduced the DNA repair in ovarian cancer cells, sensitizing them to cisplatin [36]. The chemo-refractory HGSOC genotype is also characterized by other exclusive genomic and transcriptional modifications represented by changed expression of several genes, including: hypoxia-induced factors (HIF), tumor necrosis factor (TNF), JUN transcription factor (JUN), FOR proto-oncogene family (FOS), growth arrest and DNA damage inducible beta (GADD45B), induced myeloid leukemia cell differentiation anti-apoptotic protein Mcl-1 (MCL1), C-X-C motif chemokine receptor 4 (CXCR4), snail family zinc finger 1 (SNA1), vimentin (VIM), soluble N-ethylmaleimide-sensitive factor-attachment protein (SNAP) receptors SNAREs, member of RAS oncogene family (Rab), and NF-κB transcription factor [37][38][39]. The genomic and translational changes that drive to acquired chemoresistance in HGSOC are represented by disturbed expression of multi drug resistance 1 (MDR1), glutathione S-transferase Pi 1 (GSTpi), B-cell lymphoma 2 (BCL-2), survivin, SMAD family member 4 (SMAD4) and β-tubulin III, and CpG methylation [16][40][41][42][43][44]. High expression of two genes angiogenic factor with G-Patch and FHA domains 1 (AGGF1), and microfibril associated protein 4 (MFAP4), was observed in HGSOC and correlated with platinum resistance [45]. In advanced stage HGSOC, stemness-associated genes were found to be connected to the resistance to platinum and to combined platinum-taxol therapy. The expression of Aurora A kinase-AURKA, cyclin A2-CCNA2, MYB proto-oncogene-like 2-MYBL2, and origin recognition complex subunit 1-ORC1 affected the survival of platinum resistant patients, while the expression of CCNA2, cyclin-dependent kinase 1-CDK1, ORC1, DNA topoisomerase II alpha-TOP2A, and threonine tyrosine kinase-TTK affected the survival of platinum/taxol-resistant patients [46]. The problem of chemoresistance also concerns the group of patients subjected to neo-adjuvant chemotherapy. The highest incidence of copy number variations was found in three genes: maestro heat-like repeat-containing protein family member 1 (MROH1), transmembrane protein 249 (TMEM249), and heat shock transcription factor 1 (HSF1), and was associated with neo-adjuvant chemoresistance [47].
The chemoresistant group and the chemosensitive group showed differentially expressed plasma proteins. Among them, complement C4-A, IgJ chain, clusterin, α-1-antitrypsin, and carbonic anhydrase 1 were up-regulated, and transthyretin, haptoglobin, β-2-glycoprotein, Ig γ-2 chain C region, Ig γ-1 chain C region, complement factor I light chain, Igκ chain C region, complement C3, and apolipoprotein E were down-regulated in the chemoresistant group when compared with the chemosensitive group [48]. The shed form of cellular desmoglein-2 (DSG2), an epithelial junction protein, was significantly over-expressed in the serum of HGSOC patients with chemo-refractory cancer and worse survival. Shedding of DSG2 is mediated by EGFR followed by a MMP cleavage and accompanies many malignancies [49]. Soluble programmed death receptor ligands sPD-L1 and sPD-L2 were related to reduced OS and platinum resistance, respectively [50]. Secretome proteomics of chemo-resistant and chemo-sensitive ovarian cancer enabled the identification of the proteins that showed different levels between the studied groups. One of them, collagen type XI alpha 1 chain (COL11A1), was found to be a biomarker of chemoresistance and correlated with worse survival [51]. The flow cytometric analysis of the blood of ovarian cancer patients found significant relationship between chemoresistance and both increased the number of CD44+/CD24− stem cells and over-expression of ubiquitin-conjugating enzyme E2 coding gene RAD6 [52]. The population of CD44+/CD117+ stem cells were related to platinum and paclitaxel resistance [53].
Transcriptome-based stemness-related gene signature was used to create the prediction system for platinum sensitivity. The results revealed that four genes were associated with the OS of advanced-stage HGSOC patients, with AURKA, MYBL2, and ORC1 predicting shorter survival, and Polo-like kinase 1-PLK1 longer survival, respectively. The study performed to investigate the potential chemoresistance indicators among the stemness-associated key genes in stage III–IV HGSOC patients receiving platinum or the combination of platinum and taxol showed that the higher expression of AURKA, MYBL2, and ORC1 genes prognosed shorter OS, while the higher expression of CCNA2 gene prognosed longer OS. According to the PFS, the higher expression of AURKA, BIRC5, CCNA2, CCNB2, CDC20, CDK1, PLK1, RRM2, TOP2A, and TTK predicted longer PFS. In the group of advanced HGSOC patients treated with both platinum and taxol, shorter OS was correlated with the higher expression of CCNA2, CDK1, ORC1, TOP2A, and TTK, whereas higher expression of BIRC5, CCNB2, CDC20, MYBL2, PLK1, TOP2A, and TTK predicted longer PFS. The expression of BIRC5, BUB1, CDC20, CDK1, and ORC1 was up-regulated in platinum-sensitive compared with platinum-resistant HGSOC samples. From all these genes, CDC20 was found to be the most relevant gene for tumor progression and drug resistance [46].

2.2. Circulating Tumor Cells (CTCs)

The presence of particular populations of CTCs was also considered a prognostic factor for chemo-resistance. Analysis of EpCAM, mucin (MUC1 and MUC16), and excision repair cross-complementation group 1 (ERCC1) protein positive CTCs indicated that ERCC1+ CTCs were independent prognostic factors for platinum resistance, OS, and PFS in primary ovarian epithelial malignant tumors [54]. The presence of ERCC1+ CTCs correlated with sPD-L2 serum levels, and their persistence, indicated poor post-treatment outcome [50][55]. A stronger concordance of platinum sensitivity was noted for elevated iCTCs than for serum CA125 [56]. Upon platinum-based chemotherapy, CTCs acquired EMT-like phenotype, characterized by a shift towards PI3Kα and Twist-expressing CTCs, which could reflect a clonal tumor evolution towards therapy-resistant phenotype [57]. The analysis of CTCs for prognostic purposes is available even on a level of single CTCs. The CTCs positive for both stem cell (CD44, ALDH1A1, Nanog, Oct4) and EMT markers (N-cadherin, Vimentin, Snai2, CD117, CD146) could account for chemoresistance; however, the interpretation of the results is hampered by an inter-cellular and intra/inter-patient heterogeneity [58].

2.3. Epigenetic Biomarkers

2.3.1. Histone and Methylation Biomarkers

Several studies identified some mechanisms of epigenetic regulation of chemoresistance in HGSOC. Gene sets associated with H3K27me3/H3K4me3 histone marks at transcription start sites in a HGSOC tumor were investigated in one of the studies. The significantly lower expression for the H3K27me3 and bivalent gene sets in “stem-like cells” and in the chemoresistant cell lines made the point of the role of genetic silencing in HGSOC progression [59]. Another study revealed that by recruiting the DOT1-like histone lysine methyltransferase (DOT1L), CCAAT/enhancer-binding protein beta (C/EBPβ) can maintain an open chromatin state by H3K79 methylation of multi-drug resistance genes, thereby augmenting the chemoresistance of tumor cells [60]. 5-Hydroxymethylcytosine (5hmC) may regulate gene expression or prompt DNA methylation. Loss of 5hmC levels have been associated with resistance to platinum-based therapy and worse patient survival [61]. The aberrant miR-7 methylation followed by changed regulation of MAF BZIP transcription factor G (MAFG) target gene has been involved in the development of platinum resistance and was associated with poor prognosis in ovarian cancer patients [62]. DNA/RNA helicase Schlafen-11 (SLFN11) is one of the strongest predictors of sensitivity to platinum. In tumor-infiltrating immune cells, SLFN11 expression was associated with immune activation in HGSOC by platinum treatment. However, CpG island hypermethylation of SLFN11 promoter was associated with platinum resistance in HGSOC patients [63][64]. Hypomethylating agents are able to resensitize tumor cells to cisplatin [65]. However, hypomethylation could also enhance the expression of plasminogen activator inhibitor type 1 (SERPINE1) and EMT in ovarian cancer, thus supporting chemoresistance [66].

2.3.2. MicroRNA Biomarkers

MiRNAs are known regulators of cancer gene’s expression and function, and several miRNAs were found to play a prognostic and predictive role in ovarian cancer therapy. MiR-335 expression level was observed to be reduced in malignant tissue samples, especially omental implants, and was associated with shorter OS and tumor recurrence [67]. Up-regulation of miR-335-5p expression restored the cisplatin sensitivity of ovarian cancer cells through suppressing the BCL2L2 anti-apoptotic gene, suggesting the potential of miR-335-5p/BCL2L2 signaling as a therapeutic target to overcome the cisplatin resistance [68]. High collagen type XI alpha 1 (COL11A1) levels were found to be associated with tumor progression, chemoresistance, and poor patient survival. MiR-509 and miR-335 were identified as the candidate miRNAs regulating COL11A1 expression. Treatment of ovarian cancer cells with miR-335 mimics decreased COL11A1 expression and suppressed cell proliferation and invasion, simultaneously increasing the cisplatin sensitivity [69]. Moreover, COL11A1 regulates twist family basic helix–loop–helix transcription factor 1-related protein 1 (TWIST1), resulting in the induction of chemoresistance and inhibition of apoptosis in ovarian cancer cells [70].
The set of four miRNA biomarkers (miR-454-3p, miR-98-5p, miR-183-5p, and miR-22-3p) identified in the ovarian tumor were able to discriminate between platinum-sensitive and platinum-resistant HGSOC patients, thus being an indicator of chemoresistance [71]. The set of ten miRNAs (miR-151, miR-301b, miR-505, miR-324, miR-502, miR-421, let-7a, miR-320, miR-146a, and miR-193a) was used to create a 10-miRNA score. The tumor samples were classified into four subtypes: mesenchymal (high expression of stromal components), proliferative (high expression of proliferation markers), immunoreactive (high expression of T-cell chemokine ligands and their receptors), and differentiated (high expression of ovarian tumor markers) [72]. Results indicated that the high 10-miRNA-score group contained more tumors of the proliferative subtype, while the low 10-miRNA-score group contained more tumors of the mesenchymal subtype, respectively. A high 10-miRNA score was associated with extreme genome instability that explained the chemosensitivity, followed by favorable survival [61]. Low tumor suppressor miR-let7g tissue levels in ovarian cancer were associated with chemoresistance, and up-regulation of this miRNA in ovarian cancer cell lines promoted cell cycle arrest, inhibited epithelial-to-mesenchymal transition (EMT), and restored the chemosensitivity [73]. The PCR analysis confirmed the up-regulation of another miRNA, miR-23a-3p, in chemo-resistant tumors from HGSOC patients. MiR-23a-3p suppresses apoptosis of tumor cells and supports platinum chemoresistance by regulating the expression of the apoptotic protease-activating factor 1 APAF1. The miR23a-3p/APAF1 signaling could be a possible target to reverse platinum resistance [74]. Restoration of miR-139-5p in chemo-resistant ovarian cancer cell lines increased the sensitivity to cisplatin treatment and promoted cisplatin-induced mitochondrial apoptosis [75]. Lipid metabolism is important for sustainment of proliferation and stemness of ovarian cancer cells. Solute carrier family 27 member 2 (SLC27A2) is responsible for transporting long-chain and very long-chain fatty acids into the cells and activating intracellular signaling pathways. SLC27A2 can bind to the miR-411 promoter region and change its effects on the drug-transporter ABCG2 target gene. By this mechanism, down-regulated miR-411 expression contributes to ovarian cancer chemoresistance [76]. The epigenetic regulation of insulin growth factor-1 receptor (IGF1R) by another miR-1294 has also been connected to cisplatin resistance [77]. Study of both primary and recurrent BRCA1/2-mutated ovarian cancers, and patient-derived cell lines used in the in vivo BRCA2-mutated mouse model, allowed for identification of miR-493-5p that induced platinum/PARPi resistance exclusively in BRCA2-mutated tumors [78]. Disturbed function of ion channels is one of the features of cancer cells. Expression of potassium channel calcium activated large conductance subfamily M alpha member 1 (KCNMA1) is reduced in chemoresistant ovarian cancer cells and correlates with the over-expression of miR-31 regulatory miRNA [79]. MiRNA-21 is an oncogenic miRNA found to be up-regulated in almost all human cancers. The miR-21 expression was up-regulated in cisplatin-resistant compared with cisplatin-sensitive ovarian cancer cells, and its expression was regulated by c-Jun N-terminal protein kinase 1 (JNK-1)/c-Jun/miR-21 pathway [80]. In vitro experiments indicated that miR-551b enhanced the proliferation and chemoresistance of ovarian cancer stem cells through the suppression of forkhead box O3 (Foxo3) and tripartite motif containing 31 (TRIM31) tumor suppressor genes [81]. However, there is also a group of miRNAs in which over-expression was correlated with increased chemosensitivity, like miR-9, miR-30a, and miR-211 [82][83][84]. The increase in the cancer stem cells population results from the activation of the Hippo/YAP pathway target genes upon myosin phosphatase target subunit 1 (MYPT1) down-regulation, mediated by the over-expression of miR-30b. Combination therapy with cisplatin and YAP inhibitors could potentially suppress MYPT1-induced resistance [85]. Defective function of the DNA repair and genome integrity checkpoints is responsible for the genetic instability of cancer cells. The Chk1 is a serine/threonine kinase which is activated in response to diverse genotoxic signals and retransmits signals from the proximal checkpoint kinases like ATM serine/threonine kinase (ATM), ATR serine/threonine kinase (ATR), and serine/threonine kinase (ATX). The consequence of the signal transmission depends on the route of further signals and could be as follows: a switch to the stress-induced transcription program, initiation of DNA repair, delay or sustained block of cell cycle progression, apoptosis, and modulation of the chromatin remodeling pathway [86]. Dual oxidase maturation factor 1 (DUOXA1) over-expression stimulates reactive oxygen species (ROS) production and activates the ATX-Chk1 pathway [87]. The ROS production and DUOXA1 up-regulation cause the activation of c-Myc/miR-137/enhancer of zeste 2 polycomb repressive complex 2 subunit methylotransferase (EZH2) pathway and enhances platinum resistance [88]. Combination of the EZH2 inhibitor with a RAC1 GTPase inhibitor reduced the expression of stemness and induced inflammatory gene expression, thus promoting the differentiation of subpopulations of HGSOC cells and chemosensitivity [89].

2.3.3. Long Non-Coding RNA Biomarkers

The panel of seven HGSOC tumor-derived long non-coding RNAs (lncRNAs) including both up-regulated (RP11-126K1.6, ZBED3-AS1, RP11-439E19.10, and RP11-348N5.7) and down-regulated lncRNAs (RNF144A-AS1, GAS5, and F11-AS1) showed high accuracy in predicting chemosensitivity (AUC > 0.8) [90]. Another panel of seven lncRNAs was also shown to predict chemoresistance with LINC01363, AC114401.1, and AL360169.2 indicating chemo-resistance, and LINC01018, LINC02636, AC090625.2, and AC084781.1 indicating at least partial chemosensitivity [91]. Plasmacytoma variant translocation 1 (PVT1) is a lncRNA responsible for dysregulation and down-regulation of tumor suppressors. The PVT1 was significantly up-regulated in ovarian cancer tissues of cisplatin-resistant patients, acting through the regulation of TGF-β1, p-Smad4, and Caspase-3 expression in apoptotic pathways [92]. Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) regulates the expression of metastasis-associated genes in cancer. Its knockdown enhanced platinum-induced apoptosis in vivo and inhibited the Notch1 signaling pathway and ATP binding cassette subfamily C member 1 (ABCC1) drug transport system expression in platinum-resistant ovarian cancer cells [93]. HOX transcript antisense RNA (HOTAIR) lncRNA is a regulator of chromatin state and is over-expressed in several metastatic tumors. It was found in ovarian cancer that the NF-κB-HOTAIR axis contributes during platinum-triggered DNA damage response to cellular senescence and chemotherapy resistance [94].
Long integrated non-coding RNAs (lincRNAs) play an important role in platinum-induced DNA-damage response. LincRNA H19 knockdown in the HGSOC cell line resulted in the recovery of cisplatin sensitivity through the reduction of seven key proteins involved in the glutathione metabolism pathway [95].

2.3.4. Circular RNA Biomarkers

Circular RNAs (circ_RNAs) are responsible for the sponging of miRNAs, a process which could both promote or suppress the proliferation and invasiveness of several cancers. They can also interact with RNA-binding proteins and be involved in protein translation [96]. The serum levels of circ_SETDB1 were associated with ovarian cancer progression, metastases, and primary chemoresistance [97]. In another study, it was found that ovarian cancer platinum sensitivity could be regulated by circ_Cdr1as sponging miR-1270 [98]. In the cisplatin-resistant group of patients, the circulating circ_RNA foxed box protein P1 (circ_FoxP1) was found to be significantly increased and correlated with clinical features of disease advancement and with patients’ survival. Circ_FoxP1-mediated sponging of miR-22 and miR-150-3p regulates chemosensitivity, as the use of inhibitors of these two miRNAs enhanced cisplatin resistance. The regulation through sponged miRNAs involves CCAAT enhancer binding protein gamma (CEBPG) and formic-like 3 (FMNL3) proteins engaged in the response to the cellular stress and regulation of cell morphology and cytoskeletal organization, respectively [99].

References

  1. Ovarian Cancer Survival Rates|Ovarian Cancer Prognosis|American Cancer Society. Available online: https://www.cancer.org/cancer/types/ovarian-cancer/detection-diagnosis-staging/survival-rates.html (accessed on 11 December 2023).
  2. Ray-Coquard, I.; Leary, A.; Pignata, S.; Cropet, C.; González-Martín, A.; Marth, C.; Nagao, S.; Vergote, I.; Colombo, N.; Mäenpää, J.; et al. Olaparib plus Bevacizumab First-Line Maintenance in Ovarian Cancer: Final Overall Survival Results from the PAOLA-1/ENGOT-Ov25 Trial. Ann. Oncol. 2023, 34, 681–692.
  3. Pujade-Lauraine, E.; Hilpert, F.; Weber, B.; Reuss, A.; Poveda, A.; Kristensen, G.; Sorio, R.; Vergote, I.; Witteveen, P.; Bamias, A.; et al. Bevacizumab Combined With Chemotherapy for Platinum-Resistant Recurrent Ovarian Cancer: The AURELIA Open-Label Randomized Phase III Trial. JCO 2014, 32, 1302–1308.
  4. Aghajanian, C.; Blank, S.V.; Goff, B.A.; Judson, P.L.; Teneriello, M.G.; Husain, A.; Sovak, M.A.; Yi, J.; Nycum, L.R. OCEANS: A Randomized, Double-Blind, Placebo-Controlled Phase III Trial of Chemotherapy With or Without Bevacizumab in Patients With Platinum-Sensitive Recurrent Epithelial Ovarian, Primary Peritoneal, or Fallopian Tube Cancer. JCO 2012, 30, 2039–2045.
  5. Oza, A.M.; Cook, A.D.; Pfisterer, J.; Embleton, A.; Ledermann, J.A.; Pujade-Lauraine, E.; Kristensen, G.; Carey, M.S.; Beale, P.; Cervantes, A.; et al. Standard Chemotherapy with or without Bevacizumab for Women with Newly Diagnosed Ovarian Cancer (ICON7): Overall Survival Results of a Phase 3 Randomised Trial. Lancet Oncol. 2015, 16, 928–936.
  6. DiSilvestro, P.; Banerjee, S.; Colombo, N.; Scambia, G.; Kim, B.-G.; Oaknin, A.; Friedlander, M.; Lisyanskaya, A.; Floquet, A.; Leary, A.; et al. Overall Survival With Maintenance Olaparib at a 7-Year Follow-Up in Patients With Newly Diagnosed Advanced Ovarian Cancer and a BRCA Mutation: The SOLO1/GOG 3004 Trial. JCO 2023, 41, 609–617.
  7. Harter, P.; Mouret-Reynier, M.A.; Pignata, S.; Cropet, C.; González-Martín, A.; Bogner, G.; Fujiwara, K.; Vergote, I.; Colombo, N.; Nøttrup, T.J.; et al. Efficacy of Maintenance Olaparib plus Bevacizumab According to Clinical Risk in Patients with Newly Diagnosed, Advanced Ovarian Cancer in the Phase III PAOLA-1/ENGOT-Ov25 Trial. Gynecol. Oncol. 2022, 164, 254–264.
  8. González-Martín, A.; Pothuri, B.; Vergote, I.; Graybill, W.; Lorusso, D.; McCormick, C.C.; Freyer, G.; Backes, F.; Heitz, F.; Redondo, A.; et al. Progression-Free Survival and Safety at 3.5 Years of Follow-up: Results from the Randomised Phase 3 PRIMA/ENGOT-OV26/GOG-3012 Trial of Niraparib Maintenance Treatment in Patients with Newly Diagnosed Ovarian Cancer. Eur. J. Cancer 2023, 189, 112908.
  9. Labidi-Galy, S.I.; Rodrigues, M.; Sandoval, J.L.; Kurtz, J.E.; Heitz, F.; Mosconi, A.M.; Romero, I.; Denison, U.; Nagao, S.; Vergote, I.; et al. Association of Location of BRCA1 and BRCA2 Mutations with Benefit from Olaparib and Bevacizumab Maintenance in High-Grade Ovarian Cancer: Phase III PAOLA-1/ENGOT-Ov25 Trial Subgroup Exploratory Analysis. Ann. Oncol. 2023, 34, 152–162.
  10. Swisher, E.M.; Aghajanian, C.; O’Malley, D.M.; Fleming, G.F.; Kaufmann, S.H.; Levine, D.A.; Birrer, M.J.; Moore, K.N.; Spirtos, N.M.; Shahin, M.S.; et al. Impact of Homologous Recombination Status and Responses with Veliparib Combined with First-Line Chemotherapy in Ovarian Cancer in the Phase 3 VELIA/GOG-3005 Study. Gynecol. Oncol. 2022, 164, 245–253.
  11. Li, H.; Liu, Z.-Y.; Wu, N.; Chen, Y.-C.; Cheng, Q.; Wang, J. PARP Inhibitor Resistance: The Underlying Mechanisms and Clinical Implications. Mol. Cancer 2020, 19, 107.
  12. Vanderstichele, A.; Busschaert, P.; Olbrecht, S.; Lambrechts, D.; Vergote, I. Genomic Signatures as Predictive Biomarkers of Homologous Recombination Deficiency in Ovarian Cancer. Eur. J. Cancer 2017, 86, 5–14.
  13. Micek, H.M.; Visetsouk, M.R.; Fleszar, A.J.; Kreeger, P.K. The Many Microenvironments of Ovarian Cancer. In Tumor Microenvironments in Organs; Birbrair, A., Ed.; Advances in Experimental Medicine and Biology; Springer International Publishing: Cham, Switzerland, 2020; Volume 1296, pp. 199–213.
  14. Waldron, L.; Haibe-Kains, B.; Culhane, A.C.; Riester, M.; Ding, J.; Wang, X.V.; Ahmadifar, M.; Tyekucheva, S.; Bernau, C.; Risch, T.; et al. Comparative Meta-Analysis of Prognostic Gene Signatures for Late-Stage Ovarian Cancer. NCI J. Natl. Cancer Inst. 2014, 106, dju049.
  15. Ledermann, J.A.; Drew, Y.; Kristeleit, R.S. Homologous Recombination Deficiency and Ovarian Cancer. Eur. J. Cancer 2016, 60, 49–58.
  16. Patch, A.-M.; Christie, E.L.; Etemadmoghadam, D.; Garsed, D.W.; George, J.; Fereday, S.; Nones, K.; Cowin, P.; Alsop, K.; Bailey, P.J.; et al. Whole–Genome Characterization of Chemoresistant Ovarian Cancer. Nature 2015, 521, 489–494.
  17. Lin, K.K.; Harrell, M.I.; Oza, A.M.; Oaknin, A.; Ray-Coquard, I.; Tinker, A.V.; Helman, E.; Radke, M.R.; Say, C.; Vo, L.-T.; et al. BRCA Reversion Mutations in Circulating Tumor DNA Predict Primary and Acquired Resistance to the PARP Inhibitor Rucaparib in High-Grade Ovarian Carcinoma. Cancer Discov. 2019, 9, 210–219.
  18. Kondrashova, O.; Nguyen, M.; Shield-Artin, K.; Tinker, A.V.; Teng, N.N.H.; Harrell, M.I.; Kuiper, M.J.; Ho, G.-Y.; Barker, H.; Jasin, M.; et al. Secondary Somatic Mutations Restoring RAD51C and RAD51D Associated with Acquired Resistance to the PARP Inhibitor Rucaparib in High-Grade Ovarian Carcinoma. Cancer Discov. 2017, 7, 984–998.
  19. Wysham, W.Z.; Mhawech-Fauceglia, P.; Li, H.; Hays, L.; Syriac, S.; Skrepnik, T.; Wright, J.; Pande, N.; Hoatlin, M.; Pejovic, T. BRCAness Profile of Sporadic Ovarian Cancer Predicts Disease Recurrence. PLoS ONE 2012, 7, e30042.
  20. Du, Z.-H.; Bi, F.-F.; Wang, L.; Yang, Q. Next-Generation Sequencing Unravels Extensive Genetic Alteration in Recurrent Ovarian Cancer and Unique Genetic Changes in Drug-Resistant Recurrent Ovarian Cancer. Mol. Genet. Genomic. Med. 2018, 6, 638–647.
  21. Sabatier, R.; Garnier, S.; Guille, A.; Carbuccia, N.; Pakradouni, J.; Adelaide, J.; Provansal, M.; Cappiello, M.; Rousseau, F.; Chaffanet, M.; et al. Whole-Genome/Exome Analysis of Circulating Tumor DNA and Comparison to Tumor Genomics from Patients with Heavily Pre-Treated Ovarian Cancer: Subset Analysis of the PERMED-01 Trial. Front. Oncol. 2022, 12, 946257.
  22. Nakayama, N.; Nakayama, K.; Shamima, Y.; Ishikawa, M.; Katagiri, A.; Iida, K.; Miyazaki, K. Gene Amplification CCNE1 Is Related to Poor Survival and Potential Therapeutic Target in Ovarian Cancer. Cancer 2010, 116, 2621–2634.
  23. Li, Y.; Zhang, X.; Gao, Y.; Shang, C.; Yu, B.; Wang, T.; Su, J.; Huang, C.; Wu, Y.; Guo, H.; et al. Development of a Genomic Signatures-Based Predictor of Initial Platinum-Resistance in Advanced High-Grade Serous Ovarian Cancer Patients. Front. Oncol. 2021, 10, 625866.
  24. Österberg, L.; Levan, K.; Partheen, K.; Helou, K.; Horvath, G. Cytogenetic Analysis of Carboplatin Resistance in Early-Stage Epithelial Ovarian Carcinoma. Cancer Genet. Cytogenet. 2005, 163, 144–150.
  25. Kim, S.W.; Kim, J.W.; Kim, Y.T.; Kim, J.H.; Kim, S.; Yoon, B.S.; Nam, E.J.; Kim, H.Y. Analysis of Chromosomal Changes in Serous Ovarian Carcinoma Using High-Resolution Array Comparative Genomic Hybridization: Potential Predictive Markers of Chemoresistant Disease. Genes Chromosom. Cancer 2007, 46, 1–9.
  26. Osterberg, L.; Levan, K.; Partheen, K.; Delle, U.; Olsson, B.; Sundfeldt, K.; Horvath, G. Specific Copy Number Alterations Associated with Docetaxel/Carboplatin Response in Ovarian Carcinomas. Anticancer Res. 2010, 30, 4451–4458.
  27. Partheen, K.; Levan, K.; Österberg, L.; Helou, K.; Horvath, G. Analysis of Cytogenetic Alterations in Stage III Serous Ovarian Adenocarcinoma Reveals a Heterogeneous Group Regarding Survival, Surgical Outcome, and Substage: CGH of Stage III Serous Ovarian Carcinoma. Genes Chromosom. Cancer 2004, 40, 342–348.
  28. Johnatty, S.E.; Tyrer, J.P.; Kar, S.; Beesley, J.; Lu, Y.; Gao, B.; Fasching, P.A.; Hein, A.; Ekici, A.B.; Beckmann, M.W.; et al. Genome-Wide Analysis Identifies Novel Loci Associated with Ovarian Cancer Outcomes: Findings from the Ovarian Cancer Association Consortium. Clin. Cancer Res. 2015, 21, 5264–5276.
  29. Liu, Y.; Yasukawa, M.; Chen, K.; Hu, L.; Broaddus, R.R.; Ding, L.; Mardis, E.R.; Spellman, P.; Levine, D.A.; Mills, G.B.; et al. Association of Somatic Mutations of ADAMTS Genes With Chemotherapy Sensitivity and Survival in High-Grade Serous Ovarian Carcinoma. JAMA Oncol. 2015, 1, 486.
  30. Li, L.Y.; Kim, H.J.; Park, S.A.; Lee, S.H.; Kim, L.K.; Lee, J.Y.; Kim, S.; Kim, Y.T.; Kim, S.W.; Nam, E.J. Genetic Profiles Associated with Chemoresistance in Patient-Derived Xenograft Models of Ovarian Cancer. Cancer Res. Treat. 2019, 51, 1117–1127.
  31. Cohen, S.; Mosig, R.; Moshier, E.; Pereira, E.; Rahaman, J.; Prasad-Hayes, M.; Halpert, R.; Billaud, J.-N.; Dottino, P.; Martignetti, J.A. Interferon Regulatory Factor 1 Is an Independent Predictor of Platinum Resistance and Survival in High-Grade Serous Ovarian Carcinoma. Gynecol. Oncol. 2014, 134, 591–598.
  32. Choi, J.; Topouza, D.G.; Tarnouskaya, A.; Nesdoly, S.; Koti, M.; Duan, Q.L. Gene Networks and Expression Quantitative Trait Loci Associated with Adjuvant Chemotherapy Response in High-Grade Serous Ovarian Cancer. BMC Cancer 2020, 20, 413.
  33. Devlin, J.; Elder, P.; Gabra, H.; Steel, C.; Knowles, M. High Frequency of Chromosome 9 Deletion in Ovarian Cancer: Evidence for Three Tumour-Suppressor Loci. Br. J. Cancer 1996, 73, 420–423.
  34. Etemadmoghadam, D.; Weir, B.A.; Au-Yeung, G.; Alsop, K.; Mitchell, G.; George, J.; Davis, S.; D’Andrea, A.D.; Simpson, K.; Hahn, W.C.; et al. Synthetic Lethality between CCNE1 Amplification and Loss of BRCA1. Proc. Natl. Acad. Sci. USA 2013, 110, 19489–19494.
  35. Koussounadis, A.; Langdon, S.P.; Harrison, D.J.; Smith, V.A. Chemotherapy-Induced Dynamic Gene Expression Changes in Vivo Are Prognostic in Ovarian Cancer. Br. J. Cancer 2014, 110, 2975–2984.
  36. Wan, B.; Dai, L.; Wang, L.; Zhang, Y.; Huang, H.; Qian, G.; Yu, T. Knockdown of BRCA2 Enhances Cisplatin and Cisplatin-Induced Autophagy in Ovarian Cancer Cells. Endocr.-Relat. Cancer 2018, 25, 69–82.
  37. Zhang, K.; Erkan, E.P.; Jamalzadeh, S.; Dai, J.; Andersson, N.; Kaipio, K.; Lamminen, T.; Mansuri, N.; Huhtinen, K.; Carpén, O.; et al. Longitudinal Single-Cell RNA-Seq Analysis Reveals Stress-Promoted Chemoresistance in Metastatic Ovarian Cancer. Sci. Adv. 2022, 8, eabm1831.
  38. Xu, L.; Xie, K.; Mukaida, N.; Matsushima, K.; Fidler, I.J. Hypoxia-Induced Elevation in Interleukin-8 Expression by Human Ovarian Carcinoma Cells. Cancer Res. 1999, 59, 5822–5829.
  39. Wang, T.; Gilkes, D.M.; Takano, N.; Xiang, L.; Luo, W.; Bishop, C.J.; Chaturvedi, P.; Green, J.J.; Semenza, G.L. Hypoxia-Inducible Factors and RAB22A Mediate Formation of Microvesicles That Stimulate Breast Cancer Invasion and Metastasis. Proc. Natl. Acad. Sci. USA 2014, 111, E3234–E3242.
  40. Wang, Y.; Niu, X.L.; Qu, Y.; Wu, J.; Zhu, Y.Q.; Sun, W.J.; Li, L.Z. Autocrine Production of Interleukin-6 Confers Cisplatin and Paclitaxel Resistance in Ovarian Cancer Cells. Cancer Lett. 2010, 295, 110–123.
  41. Abubaker, K.; Latifi, A.; Luwor, R.; Nazaretian, S.; Zhu, H.; Quinn, M.A.; Thompson, E.W.; Findlay, J.K.; Ahmed, N. Short-Term Single Treatment of Chemotherapy Results in the Enrichment of Ovarian Cancer Stem Cell-like Cells Leading to an Increased Tumor Burden. Mol. Cancer 2013, 12, 24.
  42. Wu, C.-J.; Sundararajan, V.; Sheu, B.-C.; Huang, R.Y.-J.; Wei, L.-H. Activation of STAT3 and STAT5 Signaling in Epithelial Ovarian Cancer Progression: Mechanism and Therapeutic Opportunity. Cancers 2019, 12, 24.
  43. Crow, J.; Atay, S.; Banskota, S.; Artale, B.; Schmitt, S.; Godwin, A.K. Exosomes as Mediators of Platinum Resistance in Ovarian Cancer. Oncotarget 2017, 8, 11917–11936.
  44. Zeller, C.; Dai, W.; Steele, N.L.; Siddiq, A.; Walley, A.J.; Wilhelm-Benartzi, C.S.M.; Rizzo, S.; Van Der Zee, A.; Plumb, J.A.; Brown, R. Candidate DNA Methylation Drivers of Acquired Cisplatin Resistance in Ovarian Cancer Identified by Methylome and Expression Profiling. Oncogene 2012, 31, 4567–4576.
  45. Zhao, H.; Sun, Q.; Li, L.; Zhou, J.; Zhang, C.; Hu, T.; Zhou, X.; Zhang, L.; Wang, B.; Li, B.; et al. High Expression Levels of AGGF1 and MFAP4 Predict Primary Platinum-Based Chemoresistance and Are Associated with Adverse Prognosis in Patients with Serous Ovarian Cancer. J. Cancer 2019, 10, 397–407.
  46. Sun, X.; Liu, Q.; Huang, J.; Diao, G.; Liang, Z. Transcriptome-Based Stemness Indices Analysis Reveals Platinum-Based Chemo-Theraputic Response Indicators in Advanced-Stage Serous Ovarian Cancer. Bioengineered 2021, 12, 3753–3771.
  47. Sharbatoghli, M.; Fattahi, F.; Aboulkheyr Es, H.; Akbari, A.; Akhavan, S.; Ebrahimi, M.; Asadi-Lari, M.; Totonchi, M.; Madjd, Z. Copy Number Variation of Circulating Tumor DNA (ctDNA) Detected Using NIPT in Neoadjuvant Chemotherapy-Treated Ovarian Cancer Patients. Front. Genet. 2022, 13, 938985.
  48. Zhang, Z.; Qin, K.; Zhang, W.; Yang, B.; Zhao, C.; Zhang, X.; Zhang, F.; Zhao, L.; Shan, B. Postoperative Recurrence of Epithelial Ovarian Cancer Patients and Chemoresistance Related Protein Analyses. J. Ovarian Res. 2019, 12, 29.
  49. Kim, J.; Beidler, P.; Wang, H.; Li, C.; Quassab, A.; Coles, C.; Drescher, C.; Carter, D.; Lieber, A. Desmoglein-2 as a Prognostic and Biomarker in Ovarian Cancer. Cancer Biol. Ther. 2020, 21, 1154–1162.
  50. Buderath, P.; Schwich, E.; Jensen, C.; Horn, P.A.; Kimmig, R.; Kasimir-Bauer, S.; Rebmann, V. Soluble Programmed Death Receptor Ligands sPD-L1 and sPD-L2 as Liquid Biopsy Markers for Prognosis and Platinum Response in Epithelial Ovarian Cancer. Front. Oncol. 2019, 9, 1015.
  51. Teng, P.-N.; Wang, G.; Hood, B.L.; Conrads, K.A.; Hamilton, C.A.; Maxwell, G.L.; Darcy, K.M.; Conrads, T.P. Identification of Candidate Circulating Cisplatin-Resistant Biomarkers from Epithelial Ovarian Carcinoma Cell Secretomes. Br. J. Cancer 2014, 110, 123–132.
  52. Sihombing, U.H.M.; Purwoto, G.; Gandamihardja, S.; Harahap, A.R.; Rustamadji, P.; Kekalih, A.; Widyawati, R.; Fuady, D.R. Expression of CD44+/CD24-, RAD6 and DDB2 on Chemotherapy Response in Ovarian Cancer: A Prospective Flow Cytometry Study. Gynecol. Oncol. Rep. 2022, 42, 101005.
  53. Li, S.-S.; Ma, J.; Wong, A.S.T. Chemoresistance in Ovarian Cancer: Exploiting Cancer Stem Cell Metabolism. J. Gynecol. Oncol. 2018, 29, e32.
  54. Kuhlmann, J.D.; Wimberger, P.; Bankfalvi, A.; Keller, T.; Schöler, S.; Aktas, B.; Buderath, P.; Hauch, S.; Otterbach, F.; Kimmig, R.; et al. ERCC1-Positive Circulating Tumor Cells in the Blood of Ovarian Cancer Patients as a Predictive Biomarker for Platinum Resistance. Clin. Chem. 2014, 60, 1282–1289.
  55. Chebouti, I.; Kuhlmann, J.D.; Buderath, P.; Weber, S.; Wimberger, P.; Bokeloh, Y.; Hauch, S.; Kimmig, R.; Kasimir-Bauer, S. ERCC1-Expressing Circulating Tumor Cells as a Potential Diagnostic Tool for Monitoring Response to Platinum-Based Chemotherapy and for Predicting Post-Therapeutic Outcome of Ovarian Cancer. Oncotarget 2017, 8, 24303–24313.
  56. Pearl, M.L.; Zhao, Q.; Yang, J.; Dong, H.; Tulley, S.; Zhang, Q.; Golightly, M.; Zucker, S.; Chen, W.-T. Prognostic Analysis of Invasive Circulating Tumor Cells (iCTCs) in Epithelial Ovarian Cancer. Gynecol. Oncol. 2014, 134, 581–590.
  57. Chebouti, I.; Kasimir-Bauer, S.; Buderath, P.; Wimberger, P.; Hauch, S.; Kimmig, R.; Kuhlmann, J.D. EMT-like Circulating Tumor Cells in Ovarian Cancer Patients Are Enriched by Platinum-Based Chemotherapy. Oncotarget 2017, 8, 48820–48831.
  58. Blassl, C.; Kuhlmann, J.D.; Webers, A.; Wimberger, P.; Fehm, T.; Neubauer, H. Gene Expression Profiling of Single Circulating Tumor Cells in Ovarian Cancer—Establishment of a Multi-Marker Gene Panel. Mol. Oncol. 2016, 10, 1030–1042.
  59. Chapman-Rothe, N.; Curry, E.; Zeller, C.; Liber, D.; Stronach, E.; Gabra, H.; Ghaem-Maghami, S.; Brown, R. Chromatin H3K27me3/H3K4me3 Histone Marks Define Gene Sets in High-Grade Serous Ovarian Cancer That Distinguish Malignant, Tumour-Sustaining and Chemo-Resistant Ovarian Tumour Cells. Oncogene 2013, 32, 4586–4592.
  60. Liu, D.; Zhang, X.-X.; Li, M.-C.; Cao, C.-H.; Wan, D.-Y.; Xi, B.-X.; Tan, J.-H.; Wang, J.; Yang, Z.-Y.; Feng, X.-X.; et al. C/EBPβ Enhances Platinum Resistance of Ovarian Cancer Cells by Reprogramming H3K79 Methylation. Nat. Commun. 2018, 9, 1739.
  61. Wang, T.; Wang, G.; Zhang, X.; Wu, D.; Yang, L.; Wang, G.; Hao, D. The Expression of miRNAs Is Associated with Tumour Genome Instability and Predicts the Outcome of Ovarian Cancer Patients Treated with Platinum Agents. Sci. Rep. 2017, 7, 14736.
  62. Vera, O.; Jimenez, J.; Pernia, O.; Rodriguez-Antolin, C.; Rodriguez, C.; Sanchez Cabo, F.; Soto, J.; Rosas, R.; Lopez-Magallon, S.; Esteban Rodriguez, I.; et al. DNA Methylation of miR-7 Is a Mechanism Involved in Platinum Response through MAFG Overexpression in Cancer Cells. Theranostics 2017, 7, 4118–4134.
  63. Winkler, C.; King, M.; Berthe, J.; Ferraioli, D.; Garuti, A.; Grillo, F.; Rodriguez-Canales, J.; Ferrando, L.; Chopin, N.; Ray-Coquard, I.; et al. SLFN11 Captures Cancer-Immunity Interactions Associated with Platinum Sensitivity in High-Grade Serous Ovarian Cancer. JCI Insight 2021, 6, e146098.
  64. Nogales, V.; Reinhold, W.C.; Varma, S.; Martinez-Cardus, A.; Moutinho, C.; Moran, S.; Heyn, H.; Sebio, A.; Barnadas, A.; Pommier, Y.; et al. Epigenetic Inactivation of the Putative DNA/RNA Helicase SLFN11 in Human Cancer Confers Resistance to Platinum Drugs. Oncotarget 2016, 7, 3084–3097.
  65. Fang, F.; Munck, J.; Tang, J.; Taverna, P.; Wang, Y.; Miller, D.F.B.; Pilrose, J.; Choy, G.; Azab, M.; Pawelczak, K.S.; et al. The Novel, Small-Molecule DNA Methylation Inhibitor SGI-110 as an Ovarian Cancer Chemosensitizer. Clin. Cancer Res. 2014, 20, 6504–6516.
  66. Pan, J.-X.; Qu, F.; Wang, F.-F.; Xu, J.; Mu, L.-S.; Ye, L.-Y.; Li, J.-J. Aberrant SERPINE1 DNA Methylation Is Involved in Carboplatin Induced Epithelial-Mesenchymal Transition in Epithelial Ovarian Cancer. Arch. Gynecol. Obset. 2017, 296, 1145–1152.
  67. Cao, J.; Cai, J.; Huang, D.; Han, Q.; Chen, Y.; Yang, Q.; Yang, C.; Kuang, Y.; Li, D.; Wang, Z. miR-335 Represents an Independent Prognostic Marker in Epithelial Ovarian Cancer. Am. J. Clin. Pathol. 2014, 141, 437–442.
  68. Liu, R.; Guo, H.; Lu, S. MiR-335-5p Restores Cisplatin Sensitivity in Ovarian Cancer Cells through Targeting BCL2L2. Cancer Med. 2018, 7, 4598–4609.
  69. Wu, Y.-H.; Huang, Y.-F.; Chang, T.-H.; Wu, P.-Y.; Hsieh, T.-Y.; Hsiao, S.-Y.; Huang, S.-C.; Chou, C.-Y. miR-335 Restrains the Aggressive Phenotypes of Ovarian Cancer Cells by Inhibiting COL11A1. Cancers 2021, 13, 6257.
  70. Wu, Y.; Huang, Y.; Chang, T.; Chou, C. Activation of TWIST1 by COL11A1 Promotes Chemoresistance and Inhibits Apoptosis in Ovarian Cancer Cells by Modulating NF-κB-mediated IKKβ Expression. Intl. J. Cancer 2017, 141, 2305–2317.
  71. Qi, X.; Yu, C.; Wang, Y.; Lin, Y.; Shen, B. Network Vulnerability-based and Knowledge-guided Identification of microRNA Biomarkers Indicating Platinum Resistance in High-grade Serous Ovarian Cancer. Clin. Transl. Med. 2019, 8, 28.
  72. The Cancer Genome Atlas Research Network. Integrated Genomic Analyses of Ovarian Carcinoma. Nature 2011, 474, 609–615.
  73. Biamonte, F.; Santamaria, G.; Sacco, A.; Perrone, F.M.; Di Cello, A.; Battaglia, A.M.; Salatino, A.; Di Vito, A.; Aversa, I.; Venturella, R.; et al. MicroRNA Let-7g Acts as Tumor Suppressor and Predictive Biomarker for Chemoresistance in Human Epithelial Ovarian Cancer. Sci. Rep. 2019, 9, 5668.
  74. Todeschini, P.; Salviato, E.; Romani, C.; Raimondi, V.; Ciccarese, F.; Ferrari, F.; Tognon, G.; Marchini, S.; D’Incalci, M.; Zanotti, L.; et al. Comprehensive Profiling of Hypoxia-Related miRNAs Identifies miR-23a-3p Overexpression as a Marker of Platinum Resistance and Poor Prognosis in High-Grade Serous Ovarian Cancer. Cancers 2021, 13, 3358.
  75. Jiang, Y.; Jiang, J.; Jia, H.; Qiao, Z.; Zhang, J. Recovery of miR-139-5p in Ovarian Cancer Reverses Cisplatin Resistance by Targeting C-Jun. Cell Physiol. Biochem. 2018, 51, 129–141.
  76. Chen, F.D.; Chen, H.H.; Ke, S.C.; Zheng, L.R.; Zheng, X.Y. SLC27A2 Regulates miR-411 to Affect Chemo-Resistance in Ovarian Cancer. Neo 2018, 65, 915–924.
  77. Zhang, Y.; Huang, S.; Guo, Y.; Li, L. MiR-1294 Confers Cisplatin Resistance in Ovarian Cancer Cells by Targeting IGF1R. Biomed. Pharmacother. 2018, 106, 1357–1363.
  78. Meghani, K.; Fuchs, W.; Detappe, A.; Drané, P.; Gogola, E.; Rottenberg, S.; Jonkers, J.; Matulonis, U.; Swisher, E.M.; Konstantinopoulos, P.A.; et al. Multifaceted Impact of MicroRNA 493-5p on Genome-Stabilizing Pathways Induces Platinum and PARP Inhibitor Resistance in BRCA2-Mutated Carcinomas. Cell Rep. 2018, 23, 100–111.
  79. Samuel, P.; Pink, R.C.; Caley, D.P.; Currie, J.M.S.; Brooks, S.A.; Carter, D.R.F. Over-Expression of miR-31 or Loss of KCNMA1 Leads to Increased Cisplatin Resistance in Ovarian Cancer Cells. Tumor Biol. 2016, 37, 2565–2573.
  80. Echevarría-Vargas, I.M.; Valiyeva, F.; Vivas-Mejía, P.E. Upregulation of miR-21 in Cisplatin Resistant Ovarian Cancer via JNK-1/c-Jun Pathway. PLoS ONE 2014, 9, e97094.
  81. Wei, Z.; Liu, Y.; Wang, Y.; Zhang, Y.; Luo, Q.; Man, X.; Wei, F.; Yu, X. Downregulation of Foxo3 and TRIM31 by miR-551b in Side Population Promotes Cell Proliferation, Invasion, and Drug Resistance of Ovarian Cancer. Med. Oncol. 2016, 33, 126.
  82. Sun, C.; Li, N.; Yang, Z.; Zhou, B.; He, Y.; Weng, D.; Fang, Y.; Wu, P.; Chen, P.; Yang, X.; et al. miR-9 Regulation of BRCA1 and Ovarian Cancer Sensitivity to Cisplatin and PARP Inhibition. JNCI J. Natl. Cancer Inst. 2013, 105, 1750–1758.
  83. Sestito, R.; Cianfrocca, R.; Rosanò, L.; Tocci, P.; Semprucci, E.; Di Castro, V.; Caprara, V.; Ferrandina, G.; Sacconi, A.; Blandino, G.; et al. miR-30a Inhibits Endothelin A Receptor and Chemoresistance in Ovarian Carcinoma. Oncotarget 2016, 7, 4009–4023.
  84. Wang, T.; Hao, D.; Yang, S.; Ma, J.; Yang, W.; Zhu, Y.; Weng, M.; An, X.; Wang, X.; Li, Y.; et al. miR-211 Facilitates Platinum Chemosensitivity by Blocking the DNA Damage Response (DDR) in Ovarian Cancer. Cell Death Dis. 2019, 10, 495.
  85. Muñoz-Galván, S.; Felipe-Abrio, B.; Verdugo-Sivianes, E.M.; Perez, M.; Jiménez-García, M.P.; Suarez-Martinez, E.; Estevez-Garcia, P.; Carnero, A. Downregulation of MYPT1 Increases Tumor Resistance in Ovarian Cancer by Targeting the Hippo Pathway and Increasing the Stemness. Mol. Cancer 2020, 19, 7.
  86. Bartek, J.; Lukas, J. Chk1 and Chk2 Kinases in Checkpoint Control and Cancer. Cancer Cell 2003, 3, 421–429.
  87. Meng, Y.; Chen, C.-W.; Yung, M.M.H.; Sun, W.; Sun, J.; Li, Z.; Li, J.; Li, Z.; Zhou, W.; Liu, S.S.; et al. DUOXA1-Mediated ROS Production Promotes Cisplatin Resistance by Activating ATR-Chk1 Pathway in Ovarian Cancer. Cancer Lett. 2018, 428, 104–116.
  88. Sun, J.; Cai, X.; Yung, M.M.; Zhou, W.; Li, J.; Zhang, Y.; Li, Z.; Liu, S.S.; Cheung, A.N.Y.; Ngan, H.Y.S.; et al. miR-137 Mediates the Functional Link between c-Myc and EZH2 That Regulates Cisplatin Resistance in Ovarian Cancer. Oncogene 2019, 38, 564–580.
  89. Sriramkumar, S.; Metcalfe, T.X.; Lai, T.; Zong, X.; Fang, F.; O’Hagan, H.M.; Nephew, K.P. Single-Cell Analysis of a High-Grade Serous Ovarian Cancer Cell Line Reveals Transcriptomic Changes and Cell Subpopulations Sensitive to Epigenetic Combination Treatment. PLoS ONE 2022, 17, e0271584.
  90. Song, J.; Zhang, W.; Wang, S.; Liu, K.; Song, F.; Ran, L. A Panel of 7 Prognosis-Related Long Non-Coding RNAs to Improve Platinum-Based Chemoresistance Prediction in Ovarian Cancer. Int. J. Oncol. 2018, 53, 866–876.
  91. Cardillo, N.; Russo, D.; Newtson, A.; Reyes, H.; Lyons, Y.; Devor, E.; Bender, D.; Goodheart, M.J.; Gonzalez-Bosquet, J. Identification of Novel lncRNAs in Ovarian Cancer and Their Impact on Overall Survival. IJMS 2021, 22, 1079.
  92. Liu, E.; Liu, Z.; Zhou, Y.; Mi, R.; Wang, D. Overexpression of Long Non-Coding RNA PVT1 in Ovarian Cancer Cells Promotes Cisplatin Resistance by Regulating Apoptotic Pathways. Int. J. Clin. Exp. Med. 2015, 8, 20565–20572.
  93. Bai, L.; Wang, A.; Zhang, Y.; Xu, X.; Zhang, X. Knockdown of MALAT1 Enhances Chemosensitivity of Ovarian Cancer Cells to Cisplatin through Inhibiting the Notch1 Signaling Pathway. Exp. Cell Res. 2018, 366, 161–171.
  94. Özeş, A.R.; Miller, D.F.; Özeş, O.N.; Fang, F.; Liu, Y.; Matei, D.; Huang, T.; Nephew, K.P. NF-κB-HOTAIR Axis Links DNA Damage Response, Chemoresistance and Cellular Senescence in Ovarian Cancer. Oncogene 2016, 35, 5350–5361.
  95. Zheng, Z.-G.; Xu, H.; Suo, S.-S.; Xu, X.-L.; Ni, M.-W.; Gu, L.-H.; Chen, W.; Wang, L.-Y.; Zhao, Y.; Tian, B.; et al. The Essential Role of H19 Contributing to Cisplatin Resistance by Regulating Glutathione Metabolism in High-Grade Serous Ovarian Cancer. Sci. Rep. 2016, 6, 26093.
  96. Shi, Y.; He, R.; Yang, Y.; He, Y.; Shao, K.; Zhan, L.; Wei, B. Circular RNAs: Novel Biomarkers for Cervical, Ovarian and Endometrial Cancer (Review). Oncol. Rep. 2020, 44, 1787–1798.
  97. Wang, W.; Wang, J.; Zhang, X.; Liu, G. Serum circSETDB1 Is a Promising Biomarker for Predicting Response to Platinum-Taxane-Combined Chemotherapy and Relapse in High-Grade Serous Ovarian Cancer. OncoTargets Ther. 2019, 12, 7451–7457.
  98. Zhao, Z.; Ji, M.; Wang, Q.; He, N.; Li, Y. Circular RNA Cdr1as Upregulates SCAI to Suppress Cisplatin Resistance in Ovarian Cancer via miR-1270 Suppression. Mol. Ther. Nucleic Acids 2019, 18, 24–33.
  99. Luo, Y.; Gui, R. Circulating Exosomal circFoxp1 Confers Cisplatin Resistance in Epithelial Ovarian Cancer Cells. J. Gynecol. Oncol. 2020, 31, e75.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , ,
View Times: 76
Revisions: 2 times (View History)
Update Date: 18 Jan 2024
1000/1000