Abstract
Ovarian cancer (OC) is the third most common gynecological malignancy with the highest mortality worldwide. OC is usually diagnosed at an advanced stage, and the standard treatment is surgery combined with platinum or paclitaxel chemotherapy. However, chemoresistance inevitably appears coupled with the easy recurrence and poor prognosis. Thus, early diagnosis, predicting prognosis, and reducing chemoresistance are of great significance for controlling the progression and improving treatment effects of OC. Recently, much insight has been gained into the non-coding RNA (ncRNA) that is employed for RNAs but does not encode a protein, and many types of ncRNAs have been characterized including long-chain non-coding RNAs, microRNAs, and circular RNAs. Accumulating evidence indicates these ncRNAs play very active roles in OC progression and metastasis. In this review, we briefly discuss the ncRNAs as biomarkers for OC prognosis. We focus on the recent advances of ncRNAs as therapeutic targets in preventing OC metastasis, chemoresistance, immune escape, and metabolism. The novel strategies for ncRNAs-targeted therapy are also exploited for improving the survival of OC patients.
Keywords: circular RNA, long non-coding RNA, microRNA, ovarian cancer, prognosis, therapeutics
Introduction
Ovarian cancer (OC) ranks fifth in the mortality rate among female malignant tumors, which seriously endangers women’s lives and health. 1 In 2020, the worldwide prevalence of OC in females was 3.4% and the mortality rate was 4.7%. 2 According to the International Federation of Obstetrics and Gynecology, approximately 75% of OC patients are diagnosed in stage III or IV with extensive abdominal metastases. 3 The main obstacle to improving the diagnosis of OC is the lack of effective screening methods for early detection. 4 Although the survival rate of OC patients has improved in the past few decades, the 5-year survival rate for women with stage I epithelial ovarian cancer (EOC) is 92%, while the women diagnosed with the advanced stage OC are still less than 30%.5,6 The median progression-free survival (PFS) for OC ranges from 16 to 21 months, and 75% of patients with the advanced disease undergo recurrence within 18–24 months.7–9
The blood cancer antigen 125 (CA-125) test is the most sensitive and specific early detection marker for OC available.10,11 However, this diagnostic method is still sub-optimal, due to the low sensitivity in the early stage of the disease, and its predictive value for screening is limited. Several new studies have demonstrated the promise of improving OC diagnosis. It was reported that the Risk of Ovarian Cancer Algorithm (ROCA) was used to evaluate continuous CA-125 measurements and showed the improved sensitivity for OC early diagnosis.12,13 In addition, the ROCA followed by transvaginal ultrasound indicated excellent specificity and positive predictive value in the American female population at an average risk of OC. 14 However, useful biomarkers are urgently needed for OC early diagnosis and predicting prognosis.
The basic treatment approach for OC is cytoreductive surgery combined with platinum-based chemotherapy, and the early-stage patients often have better curative effects. 15 With the several lines of medications, OC patients gradually develop chemoresistance, which seriously affects patients’ prognosis. 16 In recurrent OC disease, chemotherapy, antiangiogenic agents, and poly (ADP-ribose) polymerase inhibitors (PARPis) are commonly used in combination. At present, bevacizumab and three PARPis (olaparib, niraparib, rucaparib) have been approved by the Food and Drug Administration as the maintenance treatment after the recurrence of specific patients with platinum-sensitive diseases.17–19 Immunotherapy is a very promising treatment option. 20 However, it needs more preclinical and clinical investigations to further confirm its therapeutic effects.
The recent advances in transcriptomics analysis have demonstrated that molecular non-coding RNAs (ncRNAs) play important roles in various aspects of OC. ncRNAs are functional RNA molecules that are not being translated into proteins but are involved in various physiological and pathological processes. 21 It was found that the dysregulation of certain ncRNAs is related to tumorigenesis, neurological, cardiovascular development, and other diseases. 22 In recent decades, a large number of studies have reported the key role of ncRNAs in cancer progression, metastasis, and drug resistance. 23 And gene expression is regulated under physiological conditions both positively and negatively by various subtypes of ncRNAs. 24 At the same time, some small ncRNA molecules are stable in the bloodstream, which can be used as novel biomarkers for the diagnosis and prognosis of OC in clinic.25,26 In addition, ncRNAs are becoming important therapeutic targets for the treatment of OC by delivering RNA interference (RNAi) or oligonucleotide targeting messenger RNA (mRNA).27–29
In this review, we searched the literature from ‘Pubmed’, ‘web of science’, and other websites closely related with the content of this paper from year 1990 to year 2022. Papers selected are pivotal studies in OC and ncRNA area demonstrating the critical evidence presented in the manuscript. This article briefly introduces the ncRNAs as biomarkers for OC prognosis and focuses on the recent progress of ncRNAs in OC therapeutics, especially for novel targeted therapy development.
Functional characterization of ncRNAs in OC
With the revolution of the high-throughput genome sequencing and array-based technology, approximately 90% of the human genome is transcribed. 30 Although the transcribed genome encodes about 20,000 proteins, this accounts for only 2% of the entire genome. In other words, not all RNAs are translated into functional proteins, which are called ncRNAs, including long-chain non-coding RNAs (lncRNAs), microRNAs (miRNAs), circular RNAs (circRNAs), and other RNAs.30,31 lncRNA molecule consists of more than 200 bases in length, transcribed by RNA polymerase II, capped, and polyadenylated at the 5′ and 3′ ends. 32 miRNA is a small RNA molecule with a sequence of 17–22 nucleotides and has been characterized as ‘molecular rheostat’ or ‘fine-tuner’ of gene expression in different tissues and cell types.33–35 The circRNA varies greatly in length from hundreds to thousands of nucleotides and has been ubiquitously discovered in many species in recent years.36,37 In addition, ncRNAs are usually expressed in a specific manner under a certain cell type, tissue, and developmental stage.38–43
miRNAs are usually combined with a short complementary sequence usually located in the three prime untranslated region (3′-UTR) region of mRNA to regulate the expression of target mRNA, prevent the expression of the corresponding mRNA, or make it degrade. 44 While lncRNAs and circRNAs exhibit gene regulatory mechanisms at the transcriptional and post-transcriptional levels, which regulate gene expression by sponging miRNAs, weakening the interactions between miRNAs and mRNAs through a competitive mechanism.45–49 The corresponding biological functions of these ncRNAs are not only realized by a single RNA regulation but also by interacting with each other.23,45
Research in recent years has demonstrated the important function of ncRNAs in cellular activities associated with OC progression, including cell proliferation, apoptosis, invasion, migration, chemoresistance, angiogenesis, and reprogram energy metabolism.50–53 Studying ncRNAs as prognostic biomarkers could help develop precision medicine for OC patients. Understanding the mechanisms of ncRNAs in the regulation of OC metastasis and chemoresistance holds promise for developing novel therapies to improve the patent’s prognosis.
In addition, OC has high variability in histological subtypes. It contains two categories: EOC which takes 90% of all OC and the rest 10% is non-epithelial OC. 54 EOC is also divided into the most common high-grade serous OC (HGSOC, about 52%), endometrioid ovarian cancer (10%), ovarian clear cell carcinoma (6%), mucinous ovarian cancer (MOC, 6%), and low-grade serous ovarian.54,55 In non-epithelial OC, germ cell tumors and sex cord stromal tumors account for only 3% and 2% of all OC, respectively. 55 Current studies on ncRNAs mainly fall into the EOC. A comprehensive miRNA expression profile can help refine the subtype classification in EOC, opening up new opportunities for identifying clinically applicable markers to improve stratification and diagnosis of OC. 56 It was reported that some ncRNAs showed differential expression levels in subtypes of OC. For example, miR-483-5p was found to be differently expressed in serous EOC and non-serous EOC, with an apparent upregulation in serous EOC. 56 Similarly, the expression of exosomal miR-1290 in HGSOC was reported higher compared to other subtypes of OCs, 57 indicating the different expression of profiles in miRNAs may be associated with different histological types of OC and could have the potential to develop personalized medicine for individual diagnosis and treatment. More deep studies are warranted in the area in future investigation.
NcRNAs as prognostic biomarkers for developing OC personalized treatment
Accurate prediction of the prognosis of OC patients is helpful to guide treatment decisions, which may greatly improve the relapse/relapse-free survival of patients. 58 So far, several methods are used in the prognosis of OC in clinics. First, various molecular markers related to the prognosis of OC can be detected by laboratory experiments and bioinformatics analysis. It was reported that high-level expression of phosphoserine aminotransferase 1 in OC tissue samples was associated with a poor prognosis of patients. 59 The machine learning system also provides the diagnosis and prognosis predictions for EOC patients before the initial intervention. 60 In the past decade, several studies have demonstrated the potential application of liquid biopsy in cancer detection and progression monitoring. Liquid biopsy including circulating tumor cells, circulating tumor DNA, and extracellular vesicles (EVs) holds promising as a new tool for improving OC diagnosis and/or prognosis.61,62 However, the current methods and protein molecules used for OC prognosis still encounter certain limitations. Since most of the findings for biomarker studies are based on The Cancer Genome Atlas (TCGA) database, which lacks valid clinical validation studies for following up and does not have a enough large sample size; thus, they are unable to define the specific molecular mechanisms in OC. 63 Due to the high stability and expression pattern in clinical samples, ncRNAs have shown great potential as prognostic biomarkers for OC.
Based on genome-wide copy number variation, lncRNAs including LOC101927151, LINC00861, and LEMD1-AS1 have been identified as new prognostic markers to predict the survival of OC patients. It was found that the lower the expression of LOC101927151 and the higher the expression of LINC00861 and LEMD1-AS1, the worse the prognosis of the OC patients. 64 In addition, the increase in exosomal lncRNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) was found to be highly correlated with the advanced and metastatic phenotype of OC as an independent predictor of the overall survival (OS) of OC patients. 65
Various miRNAs were also reported to be associated with OC prognosis. The miRNA 200 family members provide useful information about prognosis and response to the treatment of HGSOC. Compared with the control, the expression of circulating blood miR-200a, miR-200b, and miR-200c was found to be upregulated in OC patients, which was related to disease stage and reflected tissue expression. 66 It was also reported that using univariate analysis, the higher the concentration of miR-200b in exosomes, the lower the OS in the EOC. 67 Wei et al. 68 showed that OC patients with high expression of miR-199b-3p had longer OS and disease-free survival (DFS). Research by Xie et al. 69 demonstrated that the low miR-1231 expression in tissue indicated a poor prognosis for patients with OC, while upregulation of miR-1231 expression in four human OC cell lines (SKOV3, OVCA433, OV2008, and A2780) inhibited cells growth. A study based on gene expression synthesis (GEO) and bioinformatics analysis found that the prospective pathway signals of miRNA-182 were highly expressed in OC tissues and associated with a poor prognosis. 70 miR-150-5p was reported to be significantly upregulated in recurrent OC tissue specimens compared with primary tissue samples, and its expression was related to the early recurrence and poor survival rate of OC patients. 71 High levels of miR-424 (322) in tumors were found positively to be associated with PFS in patients with OC. 72 A recent clinical study showed that high expression of miR-181a was closely related to the poor prognosis in OC. 73 miR-203 was found to be upregulated and associated with significantly shorter OS and higher risk for OC progression. 74 Furthermore, highly expressed miR-206 was associated with shorter OS in EOC patients who received platinum-based chemotherapy and used to predict chemoresistance to platinum treatment. 75 Some miRNAs are downregulated in OC. For example, the expression level of miR-484 in exosomes was significantly reduced, accompanied with a poor prognosis in OC. 76 Circulating miRNAs were also useful as predictive biomarkers in patients with HGSOC. For example, higher levels of miR-187-5p and miR-6870-5p were associated with both poorer PFS and OS, while miR-1908-5p and miR-6727-5p only acted as prognostic indicators of PFS. 77
Multivariate Cox analysis showed that high expression of circRNA itchy E3 ubiquitin-protein ligase (circ-ITCH) was an independent predictor for the good OS in EOC patients, and high expression of circRNA ABCB10 (circ-ABCB10) was found to be a potential marker for a poor prognosis of OC.78,79 The rates of DFS and OS in patients with low expression of circRNA_LARP4 (circ LARP4) were significantly worse. Therefore, circ LARP4 might be a potential biomarker for the prognosis of OC. 80 The exosomal circular forkhead box protein P1 (circFoxp1) was highly expressed in serum of EOC patients, and through survival analysis, it was found that EOC patients with high exosome circFoxp1 expression had lower OS and DFS, suggesting circFoxp1 was a worse prognostic biomarker. 81
The above-mentioned studies have shown that both tissular and circulating exosome ncRNAs play important roles in the prognosis of OC. Circulating exosome ncRNAs have the potential to be used as biomarkers for predicting the prognosis of OC patients in clinics in a non-invasive manner. With an in-depth understanding of patients’ molecular characteristics using ncRNAs as prognostic markers, the tailored therapies can be developed and applied to those who are expected to benefit the most while limiting ineffective or harmful interventions. 82 And many new therapies that regulate RNAs are being studied extensively, especially miRNAs. 83 Therefore, regulating the expression of certain ncRNAs to change the prognosis of OC might provide the promise for developing the individualized treatment of OC patients and improving their quality of life. The ncRNA biomarkers used for OC prognosis are summarized in Table 1.
Table 1.
The summary of ncRNA biomarkers used for OC prognosis.
| ncRNA | Sample type | Expression pattern | Prognosis | References |
|---|---|---|---|---|
| LOC101927151 | TCGA Genomic Data | Downregulated | The lower the expression of LOC101927151, the worse the prognosis of the OC patients | Zheng et al. 64 |
| LINC00861 | TCGA Genomic Data | Upregulated | The higher the expression of LINC00861, the worse the prognosis of the OC patients | Zheng et al. 64 |
| LEMD1-AS1 | TCGA Genomic Data | Upregulated | The higher the expression of LEMD1-AS1, the worse the prognosis of the OC patients | Zheng et al. 64 |
| MALAT1 | Serum (exosome) | Upregulated | Serum exosomal MALAT1 was overexpressed and can predict poor prognosis in EOC | Qiu et al. 65 |
| miR-200a | Serum | Upregulated | The higher miR-200a had the poor prognosis | Kan et al. 66 |
| miR-200b | Serum | Upregulated | The higher miR-200b had the poor prognosis | Kan et al. 66 |
| miR-200b | Serum (exosome) | Upregulated | The higher the concentration of exosomal miR-200b, the lower the OS rate | Pan et al. 67 |
| miR-200c | Serum | Upregulated | The higher miR-200c had the poor prognosis | Kan et al. 66 |
| miR-199b-3p | Tissues | Downregulated | Patients with high-expressing miRNA-199b-3p had longer OS and DFS | Wei et al. 68 |
| miR-1231 | Tissues | Downregulated | The low expression of miR-1231 predicted poor prognosis in OC patients | Xie et al. 69 |
| miRNA-182 | GEO database (Tissues) | Upregulated | The high expression of miR-1231 predicted poor prognosis in OC patients | Li and Li 70 |
| miR-150-5p | Tissues | Upregulated | The high expression of miR-150-5p was related to the early recurrence and poor survival rate of OC patients | Tung et al. 71 |
| miR-424 (322) | Tissues | Downregulated | High level of miR-424 (322) in tumors was positively associated with PFS in patients with OC | Xu et al. 72 |
| miR-181a | Tissues | Upregulated | miR-181a overexpression was unveiled as powerful and independent molecular predictor of patients’ poor survival and higher risk for disease progression following debulking surgery and platinum-based chemotherapy | Panoutsopoulou et al. 73 |
| miR-203 | Tissues | Upregulated | Increased miR-203 level in OC patients was correlated with unfavorable prognosis and higher risk for disease progression, independently of FIGO stage, tumor grade, residual tumor after surgery, chemotherapy response, and age | Panoutsopoulou et al. 74 |
| miR-206 | Tissues | Upregulated | High expression of miR-206 predicted platinum resistance and poor prognosis in patients with EOC | Yu et al. 75 |
| miR-484 | Serum (exosome) | Downregulated | Low serum exosomal miR-484 expression can predict poor prognosis of OC | Zhang et al. 76 |
| miR-187-5p | Serum | Upregulated | Higher level of miR-187-5p was associated with both poorer PFS and OS | Yoshida et al. 77 |
| miR-6870-5p | Serum | Upregulated | Higher level of miR-6870-5p was associated with both poorer PFS and OS | Yoshida et al. 77 |
| miR-1908-5p | Serum | Upregulated | miR-1908-5p was poor prognostic indicators of PFS | Yoshida et al. 77 |
| miR-6727-5p | Serum | Upregulated | miR-6727-5p was poor prognostic indicators of PFS | Yoshida et al. 77 |
| circ-ITCH | Tissues | Downregulated | The high expression of circ-ITCH was an independent predictor of good OS in EOC patients | Luo et al. 78 |
| circ-ABCB10 | Tissues | Upregulated | The high expression of circ-ABCB10 was a potential marker for poor prognosis of OC | Chen et al. 79 |
| circ-LARP4 | Tissues | Downregulated | Lower circ LARP4 expression was associated with poor prognosis of OC patients | Zou et al. 80 |
| circFoxp1 | Serum (exosome) | Upregulated | High circFoxp1 expression had lower OS and lower DFS | Luo et al. 81 |
circ-ABCB10, circular RNA ABCB10; circFoxp1, circular forkhead box protein P1; circ-ITCH, circRNA itchy E3 ubiquitin-protein ligase; circ-LARP4, circular RNA_LARP4; DFS, disease-free survival; EOC, epithelial ovarian cancer; FIGO, Federation of Obstetrics and Gynecology; GEO, gene expression synthesis; MALAT1, metastasis-associated lung adenocarcinoma transcript 1; OC, ovarian cancer; OS, overall survival; PFS, progression-free survival; TCGA, The Cancer Genome Atlas.
NcRNAs as therapeutic targets for OC treatment
Most patients diagnosed with OC undergo surgery first, followed by platinum-based chemotherapy.84,85 However, the majority of women with advanced EOC, fallopian tube cancer, or primary peritoneal cancer relapse and require additional treatment. 86 According to the American Society of Clinical Oncology guidelines, PARPis are also used in the management of OC. 87 It was reported that ncRNAs are involved in the regulation of many aspects of OC progression,24,50,88 so they can be used as potential therapeutic targets for OC treatment with certain advantages. For example, the specificity of lncRNA has been used to selectively kill tumors without affecting normal tissues. 89 The emerging importance of circRNAs in the initiation and progression of OC also makes them an attractive therapeutic option. 90 For tumor suppressor circRNA, due to its stability and long half-life, it might induce expression in specific cancer cells and produce great anticancer effects. 91 In the following sections, we will focus on the potential of ncRNAs as therapeutic targets for OC treatment from the aspects of metastasis, chemoresistance, immune escape, and metabolic regulation. The ncRNAs as potential therapeutic targets for OC therapy are shown in Figure 1 and Table 2.
Figure 1.
ncRNAs as potential therapeutic targets for OC therapy. This diagram illustrates several associated mechanisms by which ncRNAs (lncRNAs, miRNAs, and circRNAs) regulate OC metastasis and chemoresistance, immune escape, and metabolism. Mechanistic studies have shown some ncRNAs that can be used as OC therapeutic targets by inhibiting these potential mechanisms. Treatment of OC by upregulating or downregulating the expression of ncRNAs is a promising way to control OC spread.
CSC, cancer stem cell; circ-CSPP1, circ-centrosome/spindle pole-associated protein; circ-ITCH, circRNA itchy E3 ubiquitin-protein ligase; circRHOBTB3, circular RNA RHOBTB3; DANCR, differentiation antagonizing non-protein coding RNA; EMT, epithelial-to-mesenchymal transition; ESRP1, epithelial splicing regulatory protein-1; FEN1, flap structure-specific endonuclease 1; FOXR2, forkhead box 2; GAS5, growth arrest-specific transcript 5; HOTTIP, lncRNA HOXA transcript at the distal tip; IL-6, interleukin 6; MALAT1, metastasis-associated lung adenocarcinoma transcript 1; MICA/B, MHC class I chain-related molecules A/B; MMP: matrix metalloproteinases; ncRNA, non-coding RNA; NRCP, lncRNA ceruloplasmin; OC, ovarian cancer; PARP1, poly (ADP-ribose) polymerase 1; PD-L1, programmed death-1 ligand 1; PFKFB2, fructose-2,6-biphosphatase 2; PTAR, lncRNA pro-transition-associated RNA; SCAI, suppressor of cancer cell invasion; TP73-AS1, lncRNA P73 antisense RNA 1T; UCA1, urothelial carcinoma associated 1; VEGF, vascular endothelial growth factor; VEGFA, vascular endothelial growth factor A; ZEB1, zinc finger E-box binding homeobox 1.
Table 2.
ncRNAs as therapeutic targets for OC cell migration, invasion, metastasis, chemoresistance, immune escape, and metabolism reprogramming.
| ncRNA | Sample type | Expression pattern | Target gene | Participation | References |
|---|---|---|---|---|---|
| PTAR | Tissues | Upregulated | miR-101-3p-ZEB1 | Invasion, metastasis | Liang et al. 92 |
| WDFY3-AS2 | Cells lines | Upregulated | miR-139-5p-SDC4 | Migration, invasion, chemoresistance | Wu et al. 93 |
| DANCR | Tissues and cell lines | Upregulated | miR-145-VEGF | Angiogenesis, invasion, metastasis | Lin et al. 94 |
| TP73-AS1 | Tissues and cell lines | Upregulated | MMP-2, MMP-9 | Metastasis | Wang et al. 95 |
| UCA1 | Cell lines | Upregulated | miR-129-ABCB1 | Migration, invasion, chemoresistance | Wang et al. 96 |
| MALAT1 | Tissues and cell lines | Upregulated | Notch1 | Invasion, metastasis, apoptosis, chemoresistance | Bai et al. 97 |
| GAS5 | Tissues and cell lines | Downregulated | E2F4-PARP1-MAPK | Chemoresistance, apoptosis | Long et al. 98 |
| HOTTIP | Tissues | Upregulated | IL-6 | Immune escape, chemoresistance | Shang et al. 99 |
| NRCP | Tissues | Upregulated | STAT1 | Metastasis, glycolysis | Rupaimoole et al. 100 |
| lncRNA LINC00092 | Cell lines | Upregulated | PFKFB2 | Metabolism | Zhao et al. 101 |
| miR-424(322) | Cell lines | PD-L1 | Immune escape, chemoresistance | Xu et al. 72 | |
| miR-205 | Tissues and serum (exosomes) | Upregulated | PTEN | Invasion, migration, chemoresistance | He et al. 102 |
| miR-134-3p | Cell lines | Downregulated | FEN1 | Invasion, migration | Zhao et al. 103 |
| miR-495 | Cell lines | ABCB1 | Chemoresistance | Zou et al. 104 | |
| let-7e | Cell lines | PARP1 | Chemoresistance | Xiao et al. 105 | |
| miR-9 | Cell lines | Downregulated | BRCA1 | Migration, invasion, chemoresistance | Sun et al. 106 |
| miR-509-3 | Tissues | HMGA2/RAD51 | Migration, invasion, chemoresistance | Sun et al. 107 | |
| miR-21 | Tissues and cell lines | Chemoresistance | An and Yang 108 | ||
| miR-20a | Tissues and serum | Upregulated | MICA/B | Invasion, chemoresistance, immune escape | Xie et al. 109 |
| miR-29b | Cell lines | Downregulated | AKT2/AKT3 | Chemoresistance, migration, invasion, glucose metabolism | Teng et al. 110 |
| miR-450a | Cell lines | Downregulated | TIMMDC1, MT-ND2, ACO2, ATP5B | Metabolism (glycolysis, glutaminolysis) | Muys et al. 111 |
| miR-145 | Tissues and cell lines | Downregulated | c-myc | Metabolism (glycolysis, glutaminolysis), migration, invasion | Li et al. 112 |
| circ-ITCH | Tissues and cell lines | Downregulated | miR-106a-CDH1 | Invasion, glycolysis, apoptosis | Luo et al. 78 , Lin et al. 113 |
| circ-CSPP1 | Tissues and cell lines | Upregulated | miR-1236-3p-ZEB1 (MMP-2/VEGFA) | Invasion, migration | Li et al. 114 |
| circ_0026123 | Tissues and cell lines | Upregulated | miR-124-3p-EZH2 | Metastasis | Yang et al. 115 |
| circASH2L | Tissues and cell lines | Upregulated | miR-665-VEGFA | Angiogenesis, invasion | Chen et al. 116 |
| circATRNL1 | Tissues and cell lines | Downregulated | miR-378-Smad4 | Metastasis | Wang et al. 117 |
| cicr-Cdr1as | Tissues and cell lines | Downregulated | miR-1270-SCAI | Chemoresistance, invasion, migration | Zhao et al. 118 |
| circCELSR1 | Tissues and cell lines | Upregulated | miR-1252-FOXR2 | Chemoresistance, invasion, metastasis | Zhang et al. 119 |
| circ_0005585 | Cell lines | miR-15a/15b/16/23a/23b-ESRP1 | Immune, suppression | Deng et al. 120 | |
| circRHOBTB3 | Cell lines | Upregulated | Metastasis, metabolism(glycolysis) | Yalan et al. 121 |
circ-CSPP1, circ-centrosome/spindle pole-associated protein; circ-ITCH, circRNA itchy E3 ubiquitin-protein ligase; circRHOBTB3, circular RNA RHOBTB3; DANCR, differentiation antagonizing non-protein coding RNA; ESRP1, epithelial splicing regulatory protein-1; FEN1, flap structure-specific endonuclease 1; FOXR2, forkhead box 2; GAS5, growth arrest-specific transcript 5; HOTTIP, lncRNA HOXA transcript at the distal tip; IL-6, interleukin 6; MALAT1, metastasis-associated lung adenocarcinoma transcript 1; MICA/B, MHC class I chain-related molecules A/B; MMP, matrix metalloproteinases; ncRNA, non-coding RNA; NRCP, lncRNA ceruloplasmin; PARP1, poly (ADP-ribose) polymerase 1; PD-L1, programmed death-1 ligand 1; PFKFB2, fructose-2,6-biphosphatase 2; PTAR, lncRNA pro-transition associated RNA; SCAI, suppressor of cancer cell invasion; SDC, Syndecan; TP73-AS1, lncRNA P73 antisense RNA 1T; UCA1, urothelial carcinoma associated 1; VEGF, vascular endothelial growth factor; VEGFA, vascular endothelial growth factor A; ZEB1, zinc finger E-box binding homeobox 1.
NcRNAs as therapeutic targets for OC via regulating metastasis
Metastasis accounts for the most lethal reason for the high recurrence and poor prognosis of OC patients.122,123 Therefore, inhibiting tumor metastasis can prevent the recurrence of OC, which is also an important part of OC treatment. Since there is no barrier between the primary tumor and the abdominal cavity, aggregating cells with stem cell characteristics can leave the primary tumor and are implanted in the peritoneum in OC metastasis. 124 It was found that epithelial-to-mesenchymal transition (EMT), cell stemness, angiogenesis, matrix metalloproteinases (MMPs), and changes in cell adhesion molecules promote OC metastasis,122,125–128 which is related to the dysregulation of ncRNAs.
miRNAs are directly regulated by lncRNAs/circRNAs to affect the EMT pathway and related molecules, promoting or inhibiting the metastasis of OC. Liang et al. reported that lncRNA pro-transition-associated RNA (PTAR) was significantly upregulated in the mesenchymal subtype samples, which regulates zinc finger E-box binding homeobox 1 (ZEB1) expression by competitively binding miR-101-3p to promote EMT, leading to increased metastasis of serous OC. Thus, PTAR might be an effective target for OC antimetastatic therapy. 92 It was shown that circ-centrosome/spindle pole-associated protein (circ-CSPP1) sponged miR-1236-3p, to impair its inhibitory effect on ZEB1, and regulated EMT to promote distant metastasis of OC. 114 Overexpression of miR-200b/c was also reported to target ZEB1, which inhibited OC metastatic transmission, and miR-200b/c was related to disease stages.129,130
Several lines of evidence indicate that cancer stem cells (CSCs) are involved in tumor invasion and metastasis.131,132 The lncRNA WDFY3-AS2 sponged miR-139-5p to induce traits in CSCs, which regulates migration and invasion of the chemoresistant OC cell line A2780-DDP. 93 In vitro study showed that inhibition of miR-328 held promise for the development of efficient strategies for eliminating CSCs to prevent OC metastasis and recurrence. 133 In vitro and in vivo experiments had shown that silencing has-circ-0026123 inhibited OC cell proliferation and migration as well as inhibited the expression of CSC differentiation-related markers. 115
It was reported that the formation of a large number of microvessels is the basis for OC growth and metastasis.126,127 The angiogenesis-related ncRNAs regulate the expression of certain molecules that modulate OC angiogenesis, which further promotes OC metastasis. The differentiation antagonizing non-protein coding RNA was reported to regulate the miR-145/vascular endothelial growth factor (VEGF) axis to promote OC angiogenesis, facilitating OC metastasis. 94 miR-205 was a metastasis-associated miRNA in OC and its upregulation was found to positively correlate with microvessel density in OC tissues. 102 circASH2L was highly expressed in OC tissues and cell lines (A2780, TOV112D, OVCAR-3, and SKOV-3) and played a key role in regulating tumorigenesis, angiogenesis, and lymphangiogenesis of OC through the miR-665/VEGFA axis, 116 suggesting it might be a useful target for OC therapy. In addition, circATRNL1 was shown to sponge miR-378 and subsequently activate the Smad4 signaling pathway, which inhibits OC angiogenesis and metastasis. 117
MMPs are a family of secreted or transmembrane enzymes that collectively digest almost all extracellular matrix (ECM) and basement membrane components.134,135 More importantly, MMP-2 and MMP-9 degrade collagen IV, a major ECM component of the basement membrane, and have been implicated as key factors for the invasive and metastatic potential of OC. 124 The study by Wang et al. 95 showed that lncRNA P73 antisense RNA 1T (TP73-AS1) promoted OC cell proliferation and metastasis via the modulation of MMP-2 and MMP-9. miR-134-3p mimic transfection inhibited migration and invasion of SKOV-3 and OVCAR-3 cells and decreased the protein expression levels of cyclooxygenase-2, MMP-2, and MMP-9. 103 In addition, knockdown of circ-CSPP1 also caused a decrease in MMP-2 expression to inhibit OC metastasis, while overexpression of circ-CSPP1 had opposite effects. 114
Inhibition of tumor metastasis by regulating the expression of ncRNAs is an important aspect of OC therapy and might provide a new possible avenue for OC treatment.
NcRNAs as therapeutic targets for OC via decreasing chemoresistance
Nearly 75% of OC patients are highly sensitive to initial anticancer therapy; however, most patients encounter tumor relapse within 2 years, and are unable to respond to available chemotherapeutic compounds due to acquired resistance.136,137 Tumor cells develop several mechanisms to reduce the anticancer effects of cisplatin (CIS)/paclitaxel (PTX) through reducing drug uptake, increasing drug efflux, and inducing drug detoxification by covalently binding to glutathione or metalloprotein. 138 Besides, alterations in DNA damage repair, reactivation of homologous recombination (HR) mechanism, the occurrence of CSCs and EMT, methylation, histone acetylation, and other phenotypic changes, immune cell infiltration, angiogenesis, modification of drug targets, and defect sand hypoxia resistance are all possible reasons for OC chemoresistance.139–145 Extensive studies suggest that ncRNAs are involved in regulating the above-mentioned mechanisms and affecting OC chemoresistance. Therefore, altering the expression of ncRNAs to inhibit OC chemoresistance is also an important strategy for the treatment of OC.
The ATP-binding cassette (ABC) transporter protein family is an energy-dependent transport system for substrate-binding proteins. 146 Multidrug resistance protein 1 (MDR1), multidrug resistance-associated protein 1 (MRP1) and breast cancer resistance protein are the most important ABC transporter protein family involved in drug efflux in tumor chemoresistance mechanisms.147–149 There are many reports on ncRNA-associated regulation of ABC family gene expression, which promotes drug efflux and ultimately leads to the onset of chemoresistance. Wang et al. 96 reported that lncRNA urothelial carcinoma associated 1 was highly expressed in PTX-resistant OC cells, sponging miR-129, regulating the high expression of ABCB1, and accelerating PTX drug efflux. lncRNA MALAT1 was highly expressed in CDDP-OC cells, and induced the expression of MRP1 to accelerate CDDP drug efflux.97,150 miR-495 was reported to inhibit MDR1 expression and reduce drug efflux to reverse MDR in OC. 104
Most chemotherapy drugs, such as platinum-based drugs, directly or indirectly induce DNA damage by activating various signaling pathways, ultimately leading to cell death. Enhanced activity of DNA damage repair was found in CIS-resistant OC cells. 151 PARP is a nuclease that plays a key role in the repair of single-stranded DNA damage. 152 Let-7e is expressed in the low level in OC chemoresistant cells, but PARP1 was highly expressed. The high expression of let-7e was used to target PARP1 and thus inhibited DNA damage repair, which improves the chemoresistance in OC cells. 105 BRCA1 is a key component of the error-free HR double-strand DNA repair pathway. 153 One study reported that miR-9 targeted BRCA1, to improve the chemosensitivity of OC. 106 Another study showed that miR-509-3 in OC enhanced the synthetic lethality of PARPi by regulating HR repair in the HGSOC patient-derived-xenograft model. 107
Accumulating studies show that ncRNAs affects apoptosis as well as the cell cycle that affect the chemotherapy sensitivity.154–157 The lncRNA growth arrest-specific transcript 5 was found to affect the MAPK activity, pro-apoptosis, and cycle arrest, thereby improving chemotherapy sensitivity. 98 It was reported that the high expression of miR-21 decreased PTEN expression, promoted the PI3K/Akt activity, and inhibited OC cell apoptosis. 158 The high expression of miR-21 was also shown to regulate M2 macrophage polarization, inhibit apoptosis, and promote chemoresistance. 108 In OC, the overexpression of circ-Cdr1as was found to enhance DDP-induced apoptosis by regulating the suppressor of cancer cell invasion and miR-1270. 118 circCELSR1 was also highly expressed in PTX-resistant OC cell lines (SKOV-3/PTX and HeyA-8/PTX), and circCELSR1 silencing enhanced PTX-induced OC cytotoxicity by increasing G0/G1 blockade and apoptosis in OC cells. 119
Although advances in understanding ncRNAs in OC chemoresistance have been made in the last decade, the mechanism of action of ncRNA in OC chemoresistance is still not well understood and further studies are urgently needed. This research area is very critical to address the unmet clinical challenge of OC chemoresistance, as well as develop novel treatments to control the recurrence of OC patients. Clarifying these mechanisms of action is also important for laying a solid foundation for the clinical translation of ncRNAs in OC therapy. The putative mechanism of ncRNAs in regulating OC metastasis and chemoresistance is shown in Figure 2.
Figure 2.
The putative mechanism of ncRNAs regulates OC metastasis and chemoresistance via target genes and the related pathways. Altered expression of RNAs affects EMT, CSCs, neoangiogenesis, apoptosis pathway, cell cycle, drug efflux, and other biological functions of OC cells, leading to chemoresistance and metastasis.
CSC, cancer stem cell; EMT, epithelial-to-mesenchymal transition; ncRNA, non-coding RNA; OC, ovarian cancer.
NcRNAs as therapeutic targets for OC immunotherapy
Cancer immunotherapy has emerged as a promising therapeutic approach in oncology, characterized by the activation of the immune system and induction of tumor immune surveillance or reversal of tumor immune escape. 159 Marth et al. 160 highlighted the therapeutic benefits of immunotherapy and challenges in OC treatment. Figuring out the specific mechanism of immune escape of ncRNAs in OC is an important option for treating OC.
lncRNA HOXA transcript at the distal tip (HOTTIP) was highly expressed in OC tissues, resulting in increasing interleukin 6 expression by binding to c-jun, and promoting programmed death-1 ligand 1 (PD-L1) expression to inhibit T-cell proliferation. Therefore, HOTTIP might be involved in a potential therapeutic strategy by targeting HOTTIP in OC. 99 Another study shown that miR-424(322) inhibited PD-L1 and CD80 expression, promoted the proliferation and survival of CD8+ cytotoxic T lymphocytes and improved the OC immune response. 72 Besides, miR-20a was reported to bind directly to the 3′UTR of MHC class I chain-related molecules A/B (MICA/B) mRNA, leading to its degradation and reducing MICA/B proteins at the plasma membrane. 109 The membrane-bound MICA/B protein was a ligand for the natural killer (NK) group 2 member D receptor and was found on NK cells, γδ+ T cells, and CD8+ T cells, where its reduction enabled tumor cells to evade immune-mediated killing. 109 circ-0005585 was also found to regulate the overexpression of epithelial splicing regulatory protein-1 (ESRP1) by sponging miR-23a/b and miR-15a/15b/16, and the high expression of ESRP1 was related to immunosuppression in OC, 120 suggesting that ESRP1 was a potential therapeutic target for OC immunotherapy. One example of potential effects of ncRNAs as therapeutic targets in OC immunotherapy is shown in Figure 3.
Figure 3.
ncRNAs as therapeutic targets for OC immunotherapy. LncRNA HOTTIP is highly expressed in OC, which increases the expression of IL-6 by binding to c-jun and promotes PD-L1 expression in neutrophils to inhibit T-cell proliferation. Therefore, OC can be treated by targeting HOTTIP to increase T-cell proliferation and kill cancer cells.
IL-6, interleukin 6; lncRNAs, long-chain non-coding RNAs; ncRNA, non-coding RNA; OC, ovarian cancer; PD-1, programmed cell death protein 1; PD-L1, programmed death-1 ligand 1.
Immunotherapy is currently a more advanced treatment option for several cancers such as lung cancer, melanoma, and breast cancer. However, very limited reports are present in OC immunotherapy. ncRNAs can regulate immune escape and surveillance, but the specific immune regulatory mechanisms in OC need to be further investigated. ncRNAs’ regulation of immune escape and immune suppression might be used in combination with immunotherapy for better clinical treatment of OC in the future.
NcRNAs as therapeutic targets for OC via regulating metabolism
Reprogramming of energy metabolism is a hallmark of tumors due to genomic instability. 161 Warburg effect in cancer cells is related to aerobic glycolysis in that glucose is metabolized to lactic acid under aerobic conditions. 162
Previous studies demonstrated that lncRNA regulated glycolysis in cancer cells by directly binding to key glycolytic enzymes or by enhancing the transcription of glycolytic enzyme genes activated by lncRNA binding to RNA polymerase II.100,161,163 Mechanistic studies shown that lncRNA LINC00092 binds to a glycolytic enzyme, fructose-2,6-biphosphatase 2, thereby promoting OC metastasis by altering glycolysis and maintaining the local support function of cancer-associated fibroblasts. 101 lncRNA ceruloplasmin was highly expressed in OC tissues, and at the same time, it acted as an intermediate binding partner between STAT1 and RNA polymerase II, leading to increased expression of downstream target genes (such as glucose 6-phosphate isomerase) and regulating glycolysis. 100
miRNAs are also involved in various aspects of tumor metabolism including glucose, lipid, and amino acid metabolism. 164 Overexpression of miR-29b was reported to negatively regulate OC glucose metabolism in vivo. 110 miR-450a was found to reduce amino acid production in OC by regulating targets related to glutamine catabolism. 111 Besides, miR-145 inhibited glutamine metabolism by targeting c-myc. 112 As part of the ncRNA regulatory network, circRNAs were also reported to regulate the metabolism of carbohydrates, lipids, and amino acids by targeting miRNAs or proteins. 165 In OC, high expression of circ-ITCH in A2780 and OVCAR-3 cell lines inhibited glucose consumption and decreased both lactate and ATP content, indicating that circ-ITCH inhibited glycolysis. 113 Furthermore, circRNA RHOBTB3 (circRHOBTB3) overexpression significantly inhibited OC cell glycolysis. 121 So far, research on ncRNAs in metabolic regulation that contributes to OC progression is still very limited. The potential mechanisms of ncRNAs in OC metabolism need to be further explored, and investigating the role of ncRNAs in OC metabolism is a very important and promising area for developing novel OC therapy.
New strategies for ncRNAs-targeted OC therapy
ncRNAs participate in the regulation of multiple molecules in cellular signaling pathways to cause specific reactions. 166 However, many challenges in ncRNA research still exist. In vivo stability, in vivo cell permeability, tissue-specific targeting, and potential off-target effects are the obstacles to the successful conversion of ncRNAs-based compounds from the laboratory to clinic. 167 Given the above reasons, targeted therapy strategies explored in recent years have become hot spots. On the one hand, they can solve the instability of ncRNAs, on the other hand, they hold the potential to aim specific therapeutic targets.
Current research has focused on the development of miRNA nano-formulations to enhance cell uptake, bioavailability, and tumor site accumulation.168,169 The use of nanoparticles (NPs) coupled with antibodies and/or polypeptides can effectively target and sustain the release of miRNAs/anti-miRNAs, which reduces the required therapeutic dose while minimizing systemic and cytotoxicity. 169 Studies have shown that in vivo nanoliposome delivery of miR-15a and miR-16 reduced tumor growth in a preclinical chemoresistant OC orthotopic mouse model to support combination therapy. 170 Another in vivo study using xenograft models has shown that NP-mediated miR-124 reduced OC growth and induced cells sensitive to etoposide. 171 Since the overexpression of miR-21 was related to OC chemoresistance, 169 AS1411 anti-nucleolin aptamer-decorated PEGylated poly(lactic-co-glycolic acid) NPs containing CIS (Ap-CIS-NPs) were used to infect A2780 chemoresistant cells through nucleolar protein-mediated endocytosis and inhibited endogenous miR-21. 172 The targeted delivery of CIS using Ap-CIS-NPs into the miR-21-inhibited cells caused enhanced cell death. 172 The tumor-associated antigen, folate receptor alpha is a GPI membrane protein that is overexpressed in OC. 173 Covalently bound octahedral DNA nanocapsules were functionalized with folate molecules and used as scaffolds to engineer four chelating units with miR-21 complementary sequences to obtain biocompatible Fol-miR-21-NC non-toxic nanostructures, enabling the selective recognition of folate receptor alpha overexpressing cancer cells and sequestration of oncogenic miR-21. 174 Co-administration of doxorubicin and anti-miR-21 exhibited an additive cytotoxicity on tumor cells, laying the foundation for its use as a selective nucleic acid drug. 174 miR-155 was reported to be downregulated in OC-associated dendritic cells (DC) and is essential for DC optimal antigen presentation and T-cell activation. 175 Therefore, the use of polyethylenimine-based nano complexes to deliver miR-155 to tumor-associated DCs increased the expression of miR-155 in vitro and led to enhanced antitumor immunity, thereby increasing the survival rate of mice. 175 A novel nano-targeted co-delivery system modified with hyaluronic acid (HA) was prepared by coating functionalized mesoporous silica NPs (HA-PTX/Let-7a-GNR@MSN) with gold nanorods. 176 This drug delivery system was used in combination with hydrophobic chemotherapeutic drugs PTX and Let-7a and bound to the CD44 receptor, which is highly expressed on the surface of the SKOV3/SKOV3TR cells membrane to overcome MDR in OC. 176 However, as synthesized particles, these NPs might have obvious disadvantages after administration, such as toxicity, loss of targeting ability, and/or rapid clearance from blood circulation.177–179
Exosomes are a subtype of EVs (40–150 nm in diameter), which are considered to be a new generation of nano-scale drug delivery system. Exosomes secreted by different types of cells carry different signal molecules (such as RNAs and proteins), so they have great potential in targeted drug delivery and therapy.180,181 ncRNAs are selectively enriched and stable in exosomes. 182 Exosome ncRNAs play an important role in cell-cell communications, affecting key processes of tumor development such as tumorigenesis, metastasis, angiogenesis, immune regulation, and drug resistance in OC.65,183 Given the important biological functions of exosomal ncRNAs in OC, a strategy specifically targeting exosomes or their cargo might be a promising option for OC treatment. 184 Exosomal miR-21 was demonstrated to transfer from adjacent stromal cells to OC cells, conferring chemoresistance and aggressive phenotype to OC cells, which indicates that preventing exosomal miR-21 transfer from stromal cells was a new way to inhibit the growth of OC. 185 Exosomal miR-146a derived from human umbilical cord mesenchymal stem cells was reported to increase the sensitivity of OC cells to docetaxel and taxane. 186
Although the clinical application of exosomal ncRNAs has a long way to go, new research can help to find cost and time-saving nanotechnology to achieve large-scale production of exosomes. 187 Studies have shown that exosomes engineered by overexpressing miR-92b-3p had the stronger abilities of antiangiogenesis and antitumor than parental OC-derived exosomes, providing a new approach for antiangiogenic therapy of OC. 188 In addition, targeted delivery of miR-484 via RGD-modified exosomes induced normalization of tumor blood vessels in OC, increased tumor sensitivity to chemotherapy, and prolonged survival time after chemotherapy in tumor-bearing mice. 189 The potential of ncRNAs combined with exogenous NPs and endogenous exosomes for OC targeted therapy are shown in Figure 4. The new strategies for ncRNAs-targeted OC therapy are summarized in Table 3.
Figure 4.
The potential of ncRNAs combined with exogenous nanomaterials and endogenous exosomes for OC targeted therapy. miR-155 is downregulated in DCs and is essential for DC optimal antigen presentation and T-cell activation. Therefore, the use of PEI-based nano complexes to deliver miR-155 to tumor-associated DCs increases the expression of miR-155 in vitro and leads to enhanced antitumor immunity. Exosomal miR-146a derived from hUCMSCs increases the sensitivity of OC cells to docetaxel and taxane. Both nanomaterials and exosomes containing ncRNAs combined with chemotherapy drugs greatly induce OC cell death.
DCs, dendritic cells; hUCMSCs, human umbilical cord mesenchymal stem cells; ncRNA, non-coding RNA; NP, nanoparticle; OC, ovarian cancer; PEI, polyethylenimine.
Table 3.
New strategies for ncRNAs-targeted OC therapy.
| New strategy | ncRNA | Material/source | References |
|---|---|---|---|
| NPs | miR-15a | Neutral liposome DOPC | Dwivedi et al. 170 |
| miR-16 | Neutral liposome DOPC | Dwivedi et al. 170 | |
| miR-124 | DOPC | Seviour et al. 171 | |
| miR-21 | Ap-CIS-NPs | Vandghanooni et al. 172 | |
| miR-21 | Covalently bound octahedral DNA nanocages were functionalized with folate molecules and utilized as scaffolds to engineer four sequestering units with a miR-21 complementary sequence for obtaining biocompatible Fol-miR-21-NC non-toxic nanostructures | Xing et al. 173 | |
| miR-155 | PEI-based nano complexes | Cubillos-Ruiz et al. 175 | |
| Let-7a | HA-PTX/Let-7a-GNR@MSN | Wang et al. 176 | |
| exosome | miR-21 | Adjacent stromal cells | Au Yeung et al. 185 |
| miR-146a | hUCMSCs | Qiu et al. 186 | |
| miR-92b-3p | IOSE-80, SKOV-3, and A2780 | Wang et al. 188 | |
| miR-484 | HEK293T | Zhao et al. 189 |
Ap-CIS-NPs, AS1411 anti-nucleolin aptamer-decorated PEGylated poly (lactic-co-glycolic acid) nanoparticles containing CIS; CIS, cisplatin; DOPC, 1,2-dioleoyl-sn-glycero-3-phosphatidylcholine; HA-PTX/Let-7a-GNR@MSN, hyaluronic acid (HA) was developed employing GNRs coated with functionalized MSNs (GNR@MSN); hUCMSCs, human umbilical cord mesenchymal stem cells; ncRNA, non-coding RNA; NP, nanoparticle; PEI, polyethylenimine.
Both nanomaterials and exosomes provide a good chance for the application of ncRNAs in OC treatment. As above mentioned, some studies have been conducted on ncRNAs in OC, combined with exosomes/nanomaterials to achieve targeted transport. However, there are still many challenges for overcoming, including the toxicity of NPs, the evasion of the phagocytic system, the inhibition of physiological barriers, and the immune response to the body. 190 In addition, off-target exosomes and nanomaterials that deliver ncRNAs might also exist. However, due to the species specificity of ncRNAs, the results of animal experiments are still facing a huge test before being used in clinical application, and more in-depth exploration is needed.191,192 The current research on ncRNAs targeted therapy mainly focuses on miRNAs, and there are few reports on other types of ncRNAs. More broad research in ncRNAs is an urgent need in the future.
Conclusions and future perspectives
This review mainly focuses on the role of ncRNAs in the prognosis and treatment of OC. We investigate pivotal studies to bring new insights into the clinical application of ncRNAs in OC for future research. The network in which ncRNAs function affects multiple molecular targets that control the biological fate of cells and their responses to oncogenic effects. ncRNAs affect all aspects of OC cells, including proliferation, metastasis, chemoresistance, immune escape, and metabolism, which are in a good position to be used in clinics. 193
It is known that a network effect involves the complex regulatory mechanisms in ncRNAs’ functions. lncRNAs, circRNAs, and miRNAs are competing endogenous RNAs. lncRNAs/cicrRNAs compete with miRNAs to bind to mRNAs and regulate the expression of the corresponding molecules. The interactions among these molecules require further investigations to elucidate the related mechanisms in OC.
And it is important to understand the mechanisms performed by ncRNAs in OC metastasis, chemoresistance, immune escape, and metabolism for developing therapeutic targets. Since the combination of advanced technologies (next-generation sequencing) and new materials (exosome and nanomaterial) has great potential in the application of OC patients. The use of ncRNAs with these advanced technologies enables dual targeting of specific cells and specific genes.
Some nanoscale drug delivery systems, including NPs, liposomes, and nanocapsules, can be used to overcome the shortcomings of traditional drug delivery, preferentially target OC cells and amplify the therapeutic potential. 194 A large number of studies have shown that ncRNAs in exosomes are differentially expressed in serum, plasma, and ascites samples between OC patients and normal controls, and these ncRNAs hold great potential as biomarkers to improve the efficiency of diagnosis, prognosis, and treatment, such as the low expression of miR-484 in OC serum exosomes.76,195 However, there are few studies on targeted therapies combined with ncRNAs in OC treatment, which are worthy of further study.
Transfer RNA-derived fragments (tRFs) are another novel class of small ncRNAs produced through enzymatic cleavage of tRNAs and have been shown to play key regulatory roles similar to miRNAs.196,197 Accumulating evidence has shown that tRFs regulate gene expression at transcriptional and post-transcriptional levels. 196 It was also found that tRFs are expressed in HGSOC and normal ovarian tissues with significant differences, which might provide potential biomarkers for the diagnosis and treatment of HGSOC. 198 Although there have been many studies on tRF in other tumor types, 196 it has rarely been reported in OC research. Plant-derived tRFs-T11 was found to interact with AGO2 to suppress TRPA1 via a RNAi pathway in the A2780 cell, which, in turn, suppresses OC proliferation. 199 Silico analyses and expression profiling were performed using the TCGA-OC database, the GEO dataset, and two institutionally independent cohorts. This analysis highlighted a tRNA GlyGCC-derived internal fragment as a novel molecular predictor of EOC prognosis and supported tRNA may have bright future in the precision medicine decisions in EOC treatment. Like other ncRNAs, tRFs also play important roles in OC progression 200 and need great efforts to deeply investigate their functions and mechanism.
Besides, some small ncRNAs are so stable that they can survive in blood and form the basis for accurate and sensitive screening of major human cancers in a few drops of blood.25,26,201 In addition, the development of diagnostic biomarkers envisaged to be found in urine or blood is ideal to spare patients from the non-invasive procedures usually associated with tissue collection. It is important to find appropriate and effective ncRNAs to improve early diagnosis of OC as well as to serve as prognostic markers, and also as target molecules for therapy.
Acknowledgments
None.
Footnotes
ORCID iD: Mengyu Chen
https://orcid.org/0000-0002-4600-4889
Contributor Information
Mengyu Chen, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
Ningjing Lei, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
Wanjia Tian, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
Yong Li, Cancer Care Centre, St George Hospital, Level 2, Research and Education Centre, 4-10 South Street, Kogarah, NSW 2217, Australia; St George and Sutherland Clinical School, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia.
Lei Chang, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, No. 1 East Jianshe Road, Erqi District, Zhengzhou, Henan 450000, China.
Declarations
Ethics approval and consent to participate: Not applicable.
Consent for publication: Not applicable.
Author contribution(s): Mengyu Chen: Formal analysis; Writing – original draft.
Ningjing Lei: Data curation; Writing – original draft.
Wanjia Tian: Writing – review & editing.
Yong Li: Conceptualization; Writing – review & editing.
Lei Chang: Conceptualization; Data curation; Funding acquisition; Resources; Supervision; Writing – review & editing.
Funding: The authors disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This work was supported by Henan Provincial Medical Science and Technology Research Plan Joint Provincial and Ministry Youth Project (No. SB201902013), Scientific and Technological Project of Henan Province (No. 182102310325 and 192102310069), Henan Province Colleges and Universities Innovative Talent Support Program (No. 21HASTIT044), Henan Medical Education Research Project (No. Wjlx2020062), Key Research and Development Project of Henan Province (No. 202102310539), Henan Province Young and Middle-aged Health Science and Technology Innovative Talent Training Project (No. YXKC2020039), Central Plains Youth Top Talent Project, and High-level Talents Return to China for Research Funding Projects.
Conflict of interest statement: The authors declare that there is no conflict of interest.
Availability of data and materials: Not applicable.
References
- 1. Siegel RL, Miller KD, Fuchs HE, et al. Cancer statistics, 2022. CA Cancer J Clin 2022; 72: 7–33. [DOI] [PubMed] [Google Scholar]
- 2. Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2021; 71: 209–249. [DOI] [PubMed] [Google Scholar]
- 3. Lengyel E. Ovarian cancer development and metastasis. Am J Pathol 2010; 177: 1053–1064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Yokoi A, Matsuzaki J, Yamamoto Y, et al. Integrated extracellular microRNA profiling for ovarian cancer screening. Nat Commun 2018; 9: 4319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Lheureux S, Braunstein M, Oza AM. Epithelial ovarian cancer: evolution of management in the era of precision medicine. CA Cancer J Clin 2019; 69: 280–304. [DOI] [PubMed] [Google Scholar]
- 6. Doubeni CA, Doubeni AR, Myers AE. Diagnosis and management of ovarian cancer. Am Fam Physician 2016; 93: 937–944. [PubMed] [Google Scholar]
- 7. Markman M, Liu PY, Wilczynski S, et al. Phase III randomized trial of 12 versus 3 months of maintenance paclitaxel in patients with advanced ovarian cancer after complete response to platinum and paclitaxel-based chemotherapy: a Southwest Oncology Group and Gynecologic Oncology Group trial. J Clin Oncol 2003; 21: 2460–2465. [DOI] [PubMed] [Google Scholar]
- 8. Jayson GC, Kohn EC, Kitchener HC, et al. Ovarian cancer. Lancet 2014; 384: 1376–1388. [DOI] [PubMed] [Google Scholar]
- 9. Ushijima K. Treatment for recurrent ovarian cancer-at first relapse. J Oncol 2010; 2010: 497429. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Terry KL, Schock H, Fortner RT, et al. A prospective evaluation of early detection biomarkers for ovarian cancer in the European EPIC cohort. Clin Cancer Res 2016; 22: 4664–4675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Buys SS, Partridge E, Black A, et al. Effect of screening on ovarian cancer mortality: the Prostate, Lung, Colorectal and Ovarian (PLCO) Cancer Screening Randomized Controlled Trial. JAMA 2011; 305: 2295–2303. [DOI] [PubMed] [Google Scholar]
- 12. Jacobs IJ, Menon U, Ryan A, et al. Ovarian cancer screening and mortality in the UK Collaborative Trial of Ovarian Cancer Screening (UKCTOCS): a randomised controlled trial. Lancet 2016; 387: 945–956. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Skates SJ, Greene MH, Buys SS, et al. Early detection of ovarian cancer using the risk of ovarian cancer algorithm with frequent CA125 testing in women at increased familial risk – combined results from two screening trials. Clin Cancer Res 2017; 23: 3628–3637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Lu KH, Skates S, Hernandez MA, et al. A 2-stage ovarian cancer screening strategy using the risk of ovarian cancer algorithm (ROCA) identifies early-stage incident cancers and demonstrates high positive predictive value. Cancer 2013; 119: 3454–3461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Hennessy BT, Coleman RL, Markman M. Ovarian cancer. Lancet 2009; 374: 1371–1382. [DOI] [PubMed] [Google Scholar]
- 16. Xing Y, Cui D, Wang S, et al. Oleuropein represses the radiation resistance of ovarian cancer by inhibiting hypoxia and microRNA-299-targeted heparanase expression. Food Funct 2017; 8: 2857–2864. [DOI] [PubMed] [Google Scholar]
- 17. Pujade-Lauraine E, Ledermann JA, Selle F, et al. Olaparib tablets as maintenance therapy in patients with platinum-sensitive, relapsed ovarian cancer and a BRCA1/2 mutation (SOLO2/ENGOT-Ov21): a double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol 2017; 18: 1274–1284. [DOI] [PubMed] [Google Scholar]
- 18. Mirza MR, Monk BJ, Herrstedt J, et al. Niraparib maintenance therapy in platinum-sensitive, recurrent ovarian cancer. N Engl J Med 2016; 375: 2154–2164. [DOI] [PubMed] [Google Scholar]
- 19. Coleman RL, Oza AM, Lorusso D, et al. Rucaparib maintenance treatment for recurrent ovarian carcinoma after response to platinum therapy (ARIEL3): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017; 390: 1949–1961. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Odunsi K. Immunotherapy in ovarian cancer. Ann Oncol 2017; 28: viii1–viii7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Song X, Guo Y, Song P, et al. Non-coding RNAs in regulating tumor angiogenesis. Front Cell Dev Biol 2021; 9: 751578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Esteller M. Non-coding RNAs in human disease. Nat Rev Genet 2011; 12: 861–874. [DOI] [PubMed] [Google Scholar]
- 23. Anastasiadou E, Jacob LS, Slack FJ. Non-coding RNA networks in cancer. Nat Rev Cancer 2018; 18: 5–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Goodall GJ, Wickramasinghe VO. RNA in cancer. Nat Rev Cancer 2021; 21: 22–36. [DOI] [PubMed] [Google Scholar]
- 25. Imaoka H, Toiyama Y, Fujikawa H, et al. Circulating microRNA-1290 as a novel diagnostic and prognostic biomarker in human colorectal cancer. Ann Oncol 2016; 27: 1879–1886. [DOI] [PubMed] [Google Scholar]
- 26. Toiyama Y, Takahashi M, Hur K, et al. Serum miR-21 as a diagnostic and prognostic biomarker in colorectal cancer. J Natl Cancer Inst 2013; 105: 849–859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Levin AA. Treating disease at the RNA level with oligonucleotides. N Engl J Med 2019; 380: 57–70. [DOI] [PubMed] [Google Scholar]
- 28. Pecot CV, Calin GA, Coleman RL, et al. RNA interference in the clinic: challenges and future directions. Nat Rev Cancer 2011; 11: 59–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Wu SY, Lopez-Berestein G, Calin GA, et al. RNAi therapies: drugging the undruggable. Sci Transl Med 2014; 6: 240ps7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Consortium EP, Birney E, Stamatoyannopoulos JA, et al. Identification and analysis of functional elements in 1% of the human genome by the ENCODE pilot project. Nature 2007; 447: 799–816. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Lee YS, Dutta A. MicroRNAs in cancer. Annu Rev Pathol 2009; 4: 199–227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Quinn JJ, Chang HY. Unique features of long non-coding RNA biogenesis and function. Nat Rev Genet 2016; 17: 47–62. [DOI] [PubMed] [Google Scholar]
- 33. Wang S, Wu W, Claret FX. Mutual regulation of microRNAs and DNA methylation in human cancers. Epigenetics 2017; 12: 187–197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Wahid F, Shehzad A, Khan T, et al. MicroRNAs: synthesis, mechanism, function, and recent clinical trials. Biochim Biophys Acta 2010; 1803: 1231–1243. [DOI] [PubMed] [Google Scholar]
- 35. Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell 2009; 136: 215–233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Chen LL. The biogenesis and emerging roles of circular RNAs. Nat Rev Mol Cell Biol 2016; 17: 205–211. [DOI] [PubMed] [Google Scholar]
- 37. Jeck WR, Sharpless NE. Detecting and characterizing circular RNAs. Nat Biotechnol 2014; 32: 453–461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Cabili MN, Trapnell C, Goff L, et al. Integrative annotation of human large intergenic noncoding RNAs reveals global properties and specific subclasses. Genes Dev 2011; 25: 1915–1927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Derrien T, Johnson R, Bussotti G, et al. The GENCODE v7 catalog of human long noncoding RNAs: analysis of their gene structure, evolution, and expression. Genome Res 2012; 22: 1775–1789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Guttman M, Amit I, Garber M, et al. Chromatin signature reveals over a thousand highly conserved large non-coding RNAs in mammals. Nature 2009; 458: 223–227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Zhou M, Hara H, Dai Y, et al. Circulating organ-specific MicroRNAs serve as biomarkers in organ-specific diseases: implications for organ allo- and xeno-transplantation. Int J Mol Sci 2016; 17: 1232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Chen LL, Yang L. Regulation of circRNA biogenesis. RNA Biol 2015; 12: 381–388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Shabaninejad Z, Vafadar A, Movahedpour A, et al. Circular RNAs in cancer: new insights into functions and implications in ovarian cancer. J Ovarian Res 2019; 12: 84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Ambros V. The functions of animal microRNAs. Nature 2004; 431: 350–355. [DOI] [PubMed] [Google Scholar]
- 45. Panni S, Lovering RC, Porras P, et al. Non-coding RNA regulatory networks. Biochim Biophys Acta Gene Regul Mech 2020; 1863: 194417. [DOI] [PubMed] [Google Scholar]
- 46. Salmena L, Poliseno L, Tay Y, et al. A ceRNA hypothesis: the Rosetta stone of a hidden RNA language? Cell 2011; 146: 353–358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Dykes IM, Emanueli C. Transcriptional and post-transcriptional gene regulation by long non-coding RNA. Genom Proteom Bioinf 2017; 15: 177–186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Hansen TB, Jensen TI, Clausen BH, et al. Natural RNA circles function as efficient microRNA sponges. Nature 2013; 495: 384–388. [DOI] [PubMed] [Google Scholar]
- 49. Cheng DL, Xiang YY, Ji LJ, et al. Competing endogenous RNA interplay in cancer: mechanism, methodology, and perspectives. Tumour Biol 2015; 36: 479–488. [DOI] [PubMed] [Google Scholar]
- 50. Wang JY, Lu AQ, Chen LJ. LncRNAs in ovarian cancer. Clin Chim Acta 2019; 490: 17–27. [DOI] [PubMed] [Google Scholar]
- 51. Ghafouri-Fard S, Shoorei H, Taheri M. miRNA profile in ovarian cancer. Exp Mol Pathol 2020; 113: 104381. [DOI] [PubMed] [Google Scholar]
- 52. Berindan-Neagoe I, Monroig Pdel C, Pasculli B, et al. MicroRNAome genome: a treasure for cancer diagnosis and therapy. CA Cancer J Clin 2014; 64: 311–336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Yang X, Mei J, Wang H, et al. The emerging roles of circular RNAs in ovarian cancer. Cancer Cell Int 2020; 20: 265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Mota A, Oltra SS, Moreno-Bueno G. Insight updating of the molecular hallmarks in ovarian carcinoma. Eur J Cancer Suppl 2020; 15: 16–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Torre LA, Trabert B, DeSantis CE, et al. Ovarian cancer statistics, 2018. CA Cancer J Clin 2018; 68: 284–296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Rattanapan Y, Korkiatsakul V, Kongruang A, et al. MicroRNA expression profiling of epithelial ovarian cancer identifies new markers of tumor subtype. Microrna 2020; 9: 289–294. [DOI] [PubMed] [Google Scholar]
- 57. Kobayashi M, Sawada K, Nakamura 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 2018; 11: 81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Sheng R, Li X, Wang Z, et al. Circular RNAs and their emerging roles as diagnostic and prognostic biomarkers in ovarian cancer. Cancer Lett 2020; 473: 139–147. [DOI] [PubMed] [Google Scholar]
- 59. Zheng MJ, Li X, Hu YX, et al. Identification of molecular marker associated with ovarian cancer prognosis using bioinformatics analysis and experiments. J Cell Physiol 2019; 234: 11023–11036. [DOI] [PubMed] [Google Scholar]
- 60. Kawakami E, Tabata J, Yanaihara N, et al. Application of artificial intelligence for preoperative diagnostic and prognostic prediction in epithelial ovarian cancer based on blood biomarkers. Clin Cancer Res 2019; 25: 3006–3015. [DOI] [PubMed] [Google Scholar]
- 61. Asante DB, Calapre L, Ziman M, et al. Liquid biopsy in ovarian cancer using circulating tumor DNA and cells: ready for prime time? Cancer Lett 2020; 468: 59–71. [DOI] [PubMed] [Google Scholar]
- 62. Li X, Wang X. The emerging roles and therapeutic potential of exosomes in epithelial ovarian cancer. Mol Cancer 2017; 16: 92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Xu H, Wang H, Li G, et al. The immune-related gene ELF3 is a novel biomarker for the prognosis of ovarian cancer. Int J Gen Med 2021; 14: 5537–5548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Zheng M, Hu Y, Gou R, et al. Identification three LncRNA prognostic signature of ovarian cancer based on genome-wide copy number variation. Biomed Pharmacother 2020; 124: 109810. [DOI] [PubMed] [Google Scholar]
- 65. Qiu JJ, Lin XJ, Tang XY, et al. Exosomal metastasis associated lung adenocarcinoma transcript 1 promotes angiogenesis and predicts poor prognosis in epithelial ovarian cancer. Int J Biol Sci 2018; 14: 1960–1973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Kan CW, Hahn MA, Gard GB, et al. Elevated levels of circulating microRNA-200 family members correlate with serous epithelial ovarian cancer. BMC Cancer 2012; 12: 627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Pan C, Stevic I, Muller V, et al. Exosomal microRNAs as tumor markers in epithelial ovarian cancer. Mol Oncol 2018; 12: 1935–1948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Wei L, He Y, Bi S, et al. miRNA199b3p suppresses growth and progression of ovarian cancer via the CHK1/E-cadherin/EMT signaling pathway by targeting ZEB1. Oncol Rep 2021; 45: 569–581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Xie JR, Jiang YY, Xu W, et al. MiR-1231 correlates tumor prognosis and inhibits cell growth in ovarian cancer. Eur Rev Med Pharmacol Sci 2020; 24: 8308–8313. [DOI] [PubMed] [Google Scholar]
- 70. Li Y, Li L. Prognostic values and prospective pathway signaling of MicroRNA-182 in ovarian cancer: a study based on gene expression omnibus (GEO) and bioinformatics analysis. J Ovarian Res 2019; 12: 106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Tung CH, Kuo LW, Huang MF, et al. MicroRNA-150-5p promotes cell motility by inhibiting c-Myb-mediated Slug suppression and is a prognostic biomarker for recurrent ovarian cancer. Oncogene 2020; 39: 862–876. [DOI] [PubMed] [Google Scholar]
- 72. Xu S, Tao Z, Hai B, et al. miR-424(322) reverses chemoresistance via T-cell immune response activation by blocking the PD-L1 immune checkpoint. Nat Commun 2016; 7: 11406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Panoutsopoulou K, Avgeris M, Magkou P, et al. miR-181a overexpression predicts the poor treatment response and early-progression of serous ovarian cancer patients. Int J Cancer 2020; 147: 3560–3573. [DOI] [PubMed] [Google Scholar]
- 74. Panoutsopoulou K, Avgeris M, Mavridis K, et al. miR-203 is an independent molecular predictor of prognosis and treatment outcome in ovarian cancer: a multi-institutional study. Carcinogenesis 2020; 41: 442–451. [DOI] [PubMed] [Google Scholar]
- 75. Yu X, Zhang X, Wang G, et al. miR-206 as a prognostic and sensitivity biomarker for platinum chemotherapy in epithelial ovarian cancer. Cancer Cell Int 2020; 20: 534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Zhang W, Su X, Li S, et al. Low serum exosomal miR-484 expression predicts unfavorable prognosis in ovarian cancer. Cancer Biomark 2020; 27: 485–491. [DOI] [PubMed] [Google Scholar]
- 77. Yoshida K, Yokoi A, Matsuzaki J, et al. Extracellular microRNA profiling for prognostic prediction in patients with high-grade serous ovarian carcinoma. Cancer Sci 2021; 112: 4977–4986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Luo L, Gao Y, Sun X. Circ-ITCH correlates with small tumor size, decreased FIGO stage and prolonged overall survival, and it inhibits cells proliferation while promotes cells apoptosis in epithelial ovarian cancer. Cancer Biomark 2018; 23: 505–513. [DOI] [PubMed] [Google Scholar]
- 79. Chen Y, Ye X, Xia X, et al. Circular RNA ABCB10 correlates with advanced clinicopathological features and unfavorable survival, and promotes cell proliferation while reduces cell apoptosis in epithelial ovarian cancer. Cancer Biomark 2019; 26: 151–161. [DOI] [PubMed] [Google Scholar]
- 80. Zou T, Wang PL, Gao Y, et al. Circular RNA_LARP4 is lower expressed and serves as a potential biomarker of ovarian cancer prognosis. Eur Rev Med Pharmacol Sci 2018; 22: 7178–7182. [DOI] [PubMed] [Google Scholar]
- 81. Luo Y, Gui R. Circulating exosomal circFoxp1 confers cisplatin resistance in epithelial ovarian cancer cells. J Gynecol Oncol 2020; 31: e75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. de Gonzalo-Calvo D, Vea A, Bar C, et al. Circulating non-coding RNAs in biomarker-guided cardiovascular therapy: a novel tool for personalized medicine? Eur Heart J 2019; 40: 1643–1650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Chakraborty C, Sharma AR, Sharma G, et al. Therapeutic advances of miRNAs: a preclinical and clinical update. J Adv Res 2021; 28: 127–138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Ledermann JA, Raja FA, Fotopoulou C, et al. Newly diagnosed and relapsed epithelial ovarian carcinoma: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol 2013; 246: vi24–vi32. [DOI] [PubMed] [Google Scholar]
- 85. Coleman RL, Monk BJ, Sood AK, et al. Latest research and treatment of advanced-stage epithelial ovarian cancer. Nat Rev Clin Oncol 2013; 10: 211–224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Lheureux S, Gourley C, Vergote I, et al. Epithelial ovarian cancer. Lancet 2019; 393: 1240–1253. [DOI] [PubMed] [Google Scholar]
- 87. Tew WP, Lacchetti C, Ellis A, et al. PARP inhibitors in the management of ovarian cancer: ASCO guideline. J Clin Oncol 2020; 38: 3468–3493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Deb B, Uddin A, Chakraborty S. miRNAs and ovarian cancer: an overview. J Cell Physiol 2018; 233: 3846–3854. [DOI] [PubMed] [Google Scholar]
- 89. Chandra Gupta S, Nandan Tripathi Y. Potential of long non-coding RNAs in cancer patients: from biomarkers to therapeutic targets. Int J Cancer 2017; 140: 1955–1967. [DOI] [PubMed] [Google Scholar]
- 90. Li F, Yang Q, He AT, et al. Circular RNAs in cancer: limitations in functional studies and diagnostic potential. Semin Cancer Biol 2021; 75: 49–61. [DOI] [PubMed] [Google Scholar]
- 91. Li J, Sun D, Pu W, et al. Circular RNAs in cancer: biogenesis, function, and clinical significance. Trends Cancer 2020; 6: 319–336. [DOI] [PubMed] [Google Scholar]
- 92. Liang H, Yu T, Han Y, et al. LncRNA PTAR promotes EMT and invasion-metastasis in serous ovarian cancer by competitively binding miR-101-3p to regulate ZEB1 expression. Mol Cancer 2018; 17: 119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Wu Y, Wang T, Xia L, et al. LncRNA WDFY3-AS2 promotes cisplatin resistance and the cancer stem cell in ovarian cancer by regulating hsa-miR-139-5p/SDC4 axis. Cancer Cell Int 2021; 21: 284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Lin X, Yang F, Qi X, et al. LncRNA DANCR promotes tumor growth and angiogenesis in ovarian cancer through direct targeting of miR-145. Mol Carcinog 2019; 58: 2286–2296. [DOI] [PubMed] [Google Scholar]
- 95. Wang X, Yang B, She Y, et al. The lncRNA TP73-AS1 promotes ovarian cancer cell proliferation and metastasis via modulation of MMP2 and MMP9. J Cell Biochem 2018; 119: 7790–7799. [DOI] [PubMed] [Google Scholar]
- 96. Wang J, Ye C, Liu J, et al. UCA1 confers paclitaxel resistance to ovarian cancer through miR-129/ABCB1 axis. Biochem Biophys Res Commun 2018; 501: 1034–1040. [DOI] [PubMed] [Google Scholar]
- 97. Bai L, Wang A, Zhang Y, et al. Knockdown of MALAT1 enhances chemosensitivity of ovarian cancer cells to cisplatin through inhibiting the Notch1 signaling pathway. Exp Cell Res 2018; 366: 161–171. [DOI] [PubMed] [Google Scholar]
- 98. Long X, Song K, Hu H, et al. Long non-coding RNA GAS5 inhibits DDP-resistance and tumor progression of epithelial ovarian cancer via GAS5-E2F4-PARP1-MAPK axis. J Exp Clin Cancer Res 2019; 38: 345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Shang A, Wang W, Gu C, et al. Long non-coding RNA HOTTIP enhances IL-6 expression to potentiate immune escape of ovarian cancer cells by upregulating the expression of PD-L1 in neutrophils. J Exp Clin Cancer Res 2019; 38: 411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Rupaimoole R, Lee J, Haemmerle M, et al. Long noncoding RNA ceruloplasmin promotes cancer growth by altering glycolysis. Cell Rep 2015; 13: 2395–2402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Zhao L, Ji G, Le X, et al. Long noncoding RNA LINC00092 acts in cancer-associated fibroblasts to drive glycolysis and progression of ovarian cancer. Cancer Res 2017; 77: 1369–1382. [DOI] [PubMed] [Google Scholar]
- 102. He L, Zhu W, Chen Q, et al. Ovarian cancer cell-secreted exosomal miR-205 promotes metastasis by inducing angiogenesis. Theranostics 2019; 9: 8206–8220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Zhao M, Ji H, Fu Q, et al. microRNA-134-3p inhibits ovarian cancer progression by targeting flap structure-specific endonuclease 1 in vitro. Oncol Rep 2021; 45: 119–128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Zou Z, Zou R, Zong D, et al. miR-495 sensitizes MDR cancer cells to the combination of doxorubicin and taxol by inhibiting MDR1 expression. J Cell Mol Med 2017; 21: 1929–1943. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Xiao M, Guo J, Xie L, et al. Let-7e suppresses DNA damage repair and sensitizes ovarian cancer to cisplatin through targeting PARP1. Mol Cancer Res 2020; 18: 436–447. [DOI] [PubMed] [Google Scholar]
- 106. Sun C, Li N, Yang Z, et al. miR-9 regulation of BRCA1 and ovarian cancer sensitivity to cisplatin and PARP inhibition. J Natl Cancer Inst 2013; 105: 1750–1758. [DOI] [PubMed] [Google Scholar]
- 107. Sun C, Cao W, Qiu C, et al. miR-509-3 augments the synthetic lethality of PARPi by regulating HR repair in PDX model of HGSOC. J Hematol Oncol 2020; 13: 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. An Y, Yang Q. miR-21 modulates the polarization of macrophages and increases the effects of M2 macrophages on promoting the chemoresistance of ovarian cancer. Life Sci 2020; 242: 117162. [DOI] [PubMed] [Google Scholar]
- 109. Xie J, Liu M, Li Y, et al. Ovarian tumor-associated microRNA-20a decreases natural killer cell cytotoxicity by downregulating MICA/B expression. Cell Mol Immunol 2014; 11: 495–502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Teng Y, Zhang Y, Qu K, et al. MicroRNA-29B (mir-29b) regulates the Warburg effect in ovarian cancer by targeting AKT2 and AKT3. Oncotarget 2015; 6: 40799–40814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Muys BR, Sousa JF, Placa JR, et al. miR-450a acts as a tumor suppressor in ovarian cancer by regulating energy metabolism. Cancer Res 2019; 79: 3294–3305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Li J, Li X, Wu L, et al. miR-145 inhibits glutamine metabolism through c-myc/GLS1 pathways in ovarian cancer cells. Cell Biol Int 2019; 43: 921–930. [DOI] [PubMed] [Google Scholar]
- 113. Lin C, Xu X, Yang Q, et al. Circular RNA ITCH suppresses proliferation, invasion, and glycolysis of ovarian cancer cells by up-regulating CDH1 via sponging miR-106a. Cancer Cell Int 2020; 20: 336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Li QH, Liu Y, Chen S, et al. circ-CSPP1 promotes proliferation, invasion and migration of ovarian cancer cells by acting as a miR-1236-3p sponge. Biomed Pharmacother 2019; 114: 108832. [DOI] [PubMed] [Google Scholar]
- 115. Yang X, Wang J, Li H, et al. Downregulation of hsa_circ_0026123 suppresses ovarian cancer cell metastasis and proliferation through the miR1243p/EZH2 signaling pathway. Int J Mol Med 2021; 47: 668–676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Chen J, Li X, Yang L, et al. CircASH2L promotes ovarian cancer tumorigenesis, angiogenesis, and lymphangiogenesis by regulating the miR-665/VEGFA axis as a competing endogenous RNA. Front Cell Dev Biol 2020; 8: 595585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Wang J, Li Y, Zhou JH, et al. CircATRNL1 activates Smad4 signaling to inhibit angiogenesis and ovarian cancer metastasis via miR-378. Mol Oncol 2021; 15: 1217–1233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Zhao Z, Ji M, Wang Q, et al. Circular RNA Cdr1as upregulates SCAI to suppress cisplatin resistance in ovarian cancer via miR-1270 suppression. Mol Ther Nucleic Acids 2019; 18: 24–33. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 119. Zhang S, Cheng J, Quan C, et al. circCELSR1 (hsa_circ_0063809) contributes to paclitaxel resistance of ovarian cancer cells by regulating FOXR2 expression via miR-1252. Mol Ther Nucleic Acids 2020; 19: 718–730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Deng G, Zhou X, Chen L, et al. High expression of ESRP1 regulated by circ-0005585 promotes cell colonization in ovarian cancer. Cancer Cell Int 2020; 20: 174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Yalan S, Yanfang L, He C, et al. Circular RNA circRHOBTB3 inhibits ovarian cancer progression through PI3K/AKT signaling pathway. Panminerva Med. Epub ahead of print 27 July 2020. DOI: 10.23736/S0031-0808.20.03957-9. [DOI] [PubMed] [Google Scholar]
- 122. Chen Y, Wang DD, Wu YP, et al. MDM2 promotes epithelial-mesenchymal transition and metastasis of ovarian cancer SKOV3 cells. Br J Cancer 2017; 117: 1192–1201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Hoskins PJ, Gotlieb WH. Missed therapeutic and prevention opportunities in women with BRCA-mutated epithelial ovarian cancer and their families due to low referral rates for genetic counseling and BRCA testing: a review of the literature. CA Cancer J Clin 2017; 67: 493–506. [DOI] [PubMed] [Google Scholar]
- 124. Braga EA, Fridman MV, Moscovtsev AA, et al. LncRNAs in ovarian cancer progression, metastasis, and main pathways: ceRNA and alternative mechanisms. Int J Mol Sci 2020; 21: 8855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Zong X, Nephew KP. Ovarian cancer stem cells: role in metastasis and opportunity for therapeutic targeting. Cancers (Basel) 2019; 11: 934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Dhani NC, Oza AM. Targeting angiogenesis: taming the medusa of ovarian cancer. Hematol Oncol Clin North Am 2018; 32: 1041–1055. [DOI] [PubMed] [Google Scholar]
- 127. Duan P, Fan L, Gao Q, et al. Targeted therapy of ovarian cancer with angiogenesis inhibitors. Curr Drug Targets 2017; 18: 1171–1178. [DOI] [PubMed] [Google Scholar]
- 128. Cheung LW, Leung PC, Wong AS. Gonadotropin-releasing hormone promotes ovarian cancer cell invasiveness through c-Jun NH2-terminal kinase-mediated activation of matrix metalloproteinase (MMP)-2 and MMP-9. Cancer Res 2006; 66: 10902–10910. [DOI] [PubMed] [Google Scholar]
- 129. Sestito R, Cianfrocca R, Tocci P, et al. Targeting endothelin 1 receptor-miR-200b/c-ZEB1 circuitry blunts metastatic progression in ovarian cancer. Commun Biol 2020; 3: 677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Pendlebury A, Hannan NJ, Binder N, et al. The circulating microRNA-200 family in whole blood are potential biomarkers for high-grade serous epithelial ovarian cancer. Biomed Rep 2017; 6: 319–322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Wang L, Liu Y, Zhou Y, et al. Zoledronic acid inhibits the growth of cancer stem cell derived from cervical cancer cell by attenuating their stemness phenotype and inducing apoptosis and cell cycle arrest through the Erk1/2 and Akt pathways. J Exp Clin Cancer Res 2019; 38: 93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Cao S, Wang Z, Gao X, et al. FOXC1 induces cancer stem cell-like properties through upregulation of beta-catenin in NSCLC. J Exp Clin Cancer Res 2018; 37: 220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Srivastava AK, Banerjee A, Cui T, et al. Inhibition of miR-328-3p impairs cancer stem cell function and prevents metastasis in ovarian cancer. Cancer Res 2019; 79: 2314–2326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Mignatti P, Rifkin DB. Biology and biochemistry of proteinases in tumor invasion. Physiol Rev 1993; 73: 161–195. [DOI] [PubMed] [Google Scholar]
- 135. Fishman DA, Liu Y, Ellerbroek SM, et al. Lysophosphatidic acid promotes matrix metalloproteinase (MMP) activation and MMP-dependent invasion in ovarian cancer cells. Cancer Res 2001; 61: 3194–3199. [PubMed] [Google Scholar]
- 136. Banno K, Yanokura M, Iida M, et al. Application of microRNA in diagnosis and treatment of ovarian cancer. Biomed Res Int 2014; 2014: 232817. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Stronach EA, Cunnea P, Turner C, et al. The role of interleukin-8 (IL-8) and IL-8 receptors in platinum response in high grade serous ovarian carcinoma. Oncotarget 2015; 6: 31593–31603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Zisowsky J, Koegel S, Leyers S, et al. Relevance of drug uptake and efflux for cisplatin sensitivity of tumor cells. Biochem Pharmacol 2007; 73: 298–307. [DOI] [PubMed] [Google Scholar]
- 139. Marchetti C, De Felice F, Romito A, et al. Chemotherapy resistance in epithelial ovarian cancer: mechanisms and emerging treatments. Semin Cancer Biol 2021; 77: 144–166. [DOI] [PubMed] [Google Scholar]
- 140. Steg AD, Bevis KS, Katre AA, et al. Stem cell pathways contribute to clinical chemoresistance in ovarian cancer. Clin Cancer Res 2012; 18: 869–881. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Marchini S, Fruscio R, Clivio L, et al. Resistance to platinum-based chemotherapy is associated with epithelial to mesenchymal transition in epithelial ovarian cancer. Eur J Cancer 2013; 49: 520–530. [DOI] [PubMed] [Google Scholar]
- 142. Chebouti I, Kasimir-Bauer S, Buderath P, et al. EMT-like circulating tumor cells in ovarian cancer patients are enriched by platinum-based chemotherapy. Oncotarget 2017; 8: 48820–48831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Wei SH, Balch C, Paik HH, et al. Prognostic DNA methylation biomarkers in ovarian cancer. Clin Cancer Res 2006; 12: 2788–2794. [DOI] [PubMed] [Google Scholar]
- 144. McMullen M, Karakasis K, Madariaga A, et al. Overcoming platinum and PARP-inhibitor resistance in ovarian cancer. Cancers (Basel) 2020; 12: 1607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. Vadlapatla RK, Vadlapudi AD, Pal D, et al. Mechanisms of drug resistance in cancer chemotherapy: coordinated role and regulation of efflux transporters and metabolizing enzymes. Curr Pharm Des 2013; 19: 7126–7140. [DOI] [PubMed] [Google Scholar]
- 146. Theodoulou FL, Kerr ID. ABC transporter research: going strong 40 years on. Biochem Soc Trans 2015; 43: 1033–1040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Dean M, Annilo T. Evolution of the ATP-binding cassette (ABC) transporter superfamily in vertebrates. Annu Rev Genomics Hum Genet 2005; 6: 123–142. [DOI] [PubMed] [Google Scholar]
- 148. Chen J, Ding Z, Peng Y, et al. HIF-1alpha inhibition reverses multidrug resistance in colon cancer cells via downregulation of MDR1/P-glycoprotein. PLoS One 2014; 9: e98882. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Hellsberg E, Montanari F, Ecker GF. The ABC of phytohormone translocation. Planta Med 2015; 81: 474–487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Cole SP, Bhardwaj G, Gerlach JH, et al. Overexpression of a transporter gene in a multidrug-resistant human lung cancer cell line. Science 1992; 258: 1650–1654. [DOI] [PubMed] [Google Scholar]
- 151. Holohan C, Van Schaeybroeck S, Longley DB, et al. Cancer drug resistance: an evolving paradigm. Nat Rev Cancer 2013; 13: 714–726. [DOI] [PubMed] [Google Scholar]
- 152. Sancar A, Lindsey-Boltz LA, Unsal-Kacmaz K, et al. Molecular mechanisms of mammalian DNA repair and the DNA damage checkpoints. Annu Rev Biochem 2004; 73: 39–85. [DOI] [PubMed] [Google Scholar]
- 153. Wang B, Matsuoka S, Ballif BA, et al. Abraxas and RAP80 form a BRCA1 protein complex required for the DNA damage response. Science 2007; 316: 1194–1198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Huang W, Su G, Huang X, et al. Long noncoding RNA PCAT6 inhibits colon cancer cell apoptosis by regulating anti-apoptotic protein ARC expression via EZH2. Cell Cycle 2019; 18: 69–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Wu K, Xu K, Liu K, et al. Long noncoding RNA BC200 regulates cell growth and invasion in colon cancer. Int J Biochem Cell Biol 2018; 99: 219–225. [DOI] [PubMed] [Google Scholar]
- 156. Ong ALC, Ramasamy TS. Role of Sirtuin1-p53 regulatory axis in aging, cancer and cellular reprogramming. Ageing Res Rev 2018; 43: 64–80. [DOI] [PubMed] [Google Scholar]
- 157. Hermeking H. The miR-34 family in cancer and apoptosis. Cell Death Differ 2010; 17: 193–199. [DOI] [PubMed] [Google Scholar]
- 158. Liu HY, Zhang YY, Zhu BL, et al. miR-21 regulates the proliferation and apoptosis of ovarian cancer cells through PTEN/PI3K/AKT. Eur Rev Med Pharmacol Sci 2019; 23: 4149–4155. [DOI] [PubMed] [Google Scholar]
- 159. Masucci GV, Cesano A, Eggermont A, et al. The need for a network to establish and validate predictive biomarkers in cancer immunotherapy. J Transl Med 2017; 15: 223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160. Marth C, Wieser V, Tsibulak I, et al. Immunotherapy in ovarian cancer: fake news or the real deal? Int J Gynecol Cancer 2019; 29: 201–211. [DOI] [PubMed] [Google Scholar]
- 161. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011; 144: 646–674. [DOI] [PubMed] [Google Scholar]
- 162. Warburg O. On the origin of cancer cells. Science 1956; 123: 309–314. [DOI] [PubMed] [Google Scholar]
- 163. Ma MZ, Zhang Y, Weng MZ, et al. Long noncoding RNA GCASPC, a target of miR-17-3p, negatively regulates pyruvate carboxylase-dependent cell proliferation in gallbladder cancer. Cancer Res 2016; 76: 5361–5371. [DOI] [PubMed] [Google Scholar]
- 164. Alamoudi AA, Alnoury A, Gad H. miRNA in tumour metabolism and why could it be the preferred pathway for energy reprograming. Brief Funct Genomics 2018; 17: 157–169. [DOI] [PubMed] [Google Scholar]
- 165. Yu T, Wang Y, Fan Y, et al. circRNAs in cancer metabolism: a review. J Hematol Oncol 2019; 12: 90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. Winkle M, El-Daly SM, Fabbri M, et al. Noncoding RNA therapeutics – challenges and potential solutions. Nat Rev Drug Discov 2021; 20: 629–651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. Ratti M, Lampis A, Ghidini M, et al. MicroRNAs (miRNAs) and long non-coding RNAs (LncRNAs) as new tools for cancer therapy: first steps from bench to bedside. Target Oncol 2020; 15: 261–278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168. Ganju A, Khan S, Hafeez BB, et al. miRNA nanotherapeutics for cancer. Drug Discov Today 2017; 22: 424–432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169. Kafshdooz L, Pourfathi H, Akbarzadeh A, et al. The role of microRNAs and nanoparticles in ovarian cancer: a review. Artif Cells Nanomed Biotechnol 2018; 46: 241–247. [DOI] [PubMed] [Google Scholar]
- 170. Dwivedi SK, Mustafi SB, Mangala LS, et al. Therapeutic evaluation of microRNA-15a and microRNA-16 in ovarian cancer. Oncotarget 2016; 7: 15093–15104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171. Seviour EG, Sehgal V, Lu Y, et al. Functional proteomics identifies miRNAs to target a p27/Myc/phospho-Rb signature in breast and ovarian cancer. Oncogene 2016; 35: 691–701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172. Vandghanooni S, Eskandani M, Barar J, et al. AS1411 aptamer-decorated cisplatin-loaded poly(lactic-co-glycolic acid) nanoparticles for targeted therapy of miR-21-inhibited ovarian cancer cells. Nanomedicine (Lond) 2018; 13: 2729–2758. [DOI] [PubMed] [Google Scholar]
- 173. Xing L, Xu Y, Sun K, et al. Identification of a peptide for folate receptor alpha by phage display and its tumor targeting activity in ovary cancer xenograft. Sci Rep 2018; 8: 8426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Raniolo S, Unida V, Vindigni G, et al. Combined and selective miR-21 silencing and doxorubicin delivery in cancer cells using tailored DNA nanostructures. Cell Death Dis 2021; 12: 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175. Cubillos-Ruiz JR, Baird JR, Tesone AJ, et al. Reprogramming tumor-associated dendritic cells in vivo using miRNA mimetics triggers protective immunity against ovarian cancer. Cancer Res 2012; 72: 1683–1693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176. Wang X, Xiong T, Cui M, et al. A novel targeted co-delivery nanosystem for enhanced ovarian cancer treatment via multidrug resistance reversion and mTOR-mediated signaling pathway. J Nanobiotechnology 2021; 19: 444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177. Peng Q, Zhang S, Yang Q, et al. Preformed albumin corona, a protective coating for nanoparticles based drug delivery system. Biomaterials 2013; 34: 8521–8530. [DOI] [PubMed] [Google Scholar]
- 178. Salvati A, Pitek AS, Monopoli MP, et al. Transferrin-functionalized nanoparticles lose their targeting capabilities when a biomolecule corona adsorbs on the surface. Nat Nanotechnol 2013; 8: 137–143. [DOI] [PubMed] [Google Scholar]
- 179. Ahn J, Cho CS, Cho SW, et al. Investigation on vascular cytotoxicity and extravascular transport of cationic polymer nanoparticles using perfusable 3D microvessel model. Acta Biomater 2018; 76: 154–163. [DOI] [PubMed] [Google Scholar]
- 180. Liao W, Du Y, Zhang C, et al. Exosomes: the next generation of endogenous nanomaterials for advanced drug delivery and therapy. Acta Biomater 2019; 86: 1–14. [DOI] [PubMed] [Google Scholar]
- 181. Tian W, Lei N, Zhou J, et al. Extracellular vesicles in ovarian cancer chemoresistance, metastasis, and immune evasion. Cell Death Dis 2022; 13: 64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182. Bullock MD, Silva AM, Kanlikilicer-Unaldi P, et al. Exosomal non-coding RNAs: diagnostic, prognostic and therapeutic applications in cancer. Noncoding RNA 2015; 1: 53–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183. Kobayashi M, Salomon C, Tapia J, et al. Ovarian cancer cell invasiveness is associated with discordant exosomal sequestration of Let-7 miRNA and miR-200. J Transl Med 2014; 12: 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184. Zhang Y, Wei YJ, Zhang YF, et al. Emerging functions and clinical applications of exosomal ncRNAs in ovarian cancer. Front Oncol 2021; 11: 765458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185. Au Yeung CL, Co NN, Tsuruga T, et al. Exosomal transfer of stroma-derived miR21 confers paclitaxel resistance in ovarian cancer cells through targeting APAF1. Nat Commun 2016; 7: 11150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186. Qiu L, Wang J, Chen M, et al. Exosomal microRNA146a derived from mesenchymal stem cells increases the sensitivity of ovarian cancer cells to docetaxel and taxane via a LAMC2mediated PI3K/Akt axis. Int J Mol Med 2020; 46: 609–620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187. Zhang Y, Liu Y, Liu H, et al. Exosomes: biogenesis, biologic function and clinical potential. Cell Biosci 2019; 9: 19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188. Wang J, Wang C, Li Y, et al. Potential of peptide-engineered exosomes with overexpressed miR-92b-3p in anti-angiogenic therapy of ovarian cancer. Clin Transl Med 2021; 11: e425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189. Zhao Z, Shuang T, Gao Y, et al. Targeted delivery of exosomal miR-484 reprograms tumor vasculature for chemotherapy sensitization. Cancer Lett 2022; 530: 45–58. [DOI] [PubMed] [Google Scholar]
- 190. Ferrari M. Cancer nanotechnology: opportunities and challenges. Nat Rev Cancer 2005; 5: 161–171. [DOI] [PubMed] [Google Scholar]
- 191. Koufaris C, Gooderham NJ. Are differences in microRNA regulation implicated in species-dependent response to toxicological exposures? Toxicol Sci 2013; 131: 337–342. [DOI] [PubMed] [Google Scholar]
- 192. Mor E, Shomron N. Species-specific microRNA regulation influences phenotypic variability: perspectives on species-specific microRNA regulation. Bioessays 2013; 35: 881–888. [DOI] [PubMed] [Google Scholar]
- 193. Slack FJ, Chinnaiyan AM. The role of non-coding RNAs in oncology. Cell 2019; 179: 1033–1055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194. Barani M, Bilal M, Sabir F, et al. Nanotechnology in ovarian cancer: diagnosis and treatment. Life Sci 2021; 266: 118914. [DOI] [PubMed] [Google Scholar]
- 195. Si L, Bai J, Fu H, et al. The functions and potential roles of extracellular vesicle noncoding RNAs in gynecological malignancies. Cell Death Discov 2021; 7: 258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196. Yu M, Lu B, Zhang J, et al. tRNA-derived RNA fragments in cancer: current status and future perspectives. J Hematol Oncol 2020; 13: 121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197. Zeng T, Hua Y, Sun C, et al. Relationship between tRNA-derived fragments and human cancers. Int J Cancer 2020; 147: 3007–3018. [DOI] [PubMed] [Google Scholar]
- 198. Chen B, Liu S, Wang H, et al. Differential expression profiles and function prediction of transfer RNA-derived fragments in high-grade serous ovarian cancer. Biomed Res Int 2021; 2021: 5594081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199. Cao KY, Yan TM, Zhang JZ, et al. A tRNA-derived fragment from Chinese yew suppresses ovarian cancer growth via targeting TRPA1. Mol Ther Nucleic Acids 2022; 27: 718–732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200. Panoutsopoulou K, Dreyer T, Dorn J, et al. tRNA(GlyGCC)-Derived internal fragment (i-tRF-GlyGCC) in ovarian cancer treatment outcome and progression. Cancers (Basel) 2021; 14: 24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201. Yaman Agaoglu F, Kovancilar M, Dizdar Y, et al. Investigation of miR-21, miR-141, and miR-221 in blood circulation of patients with prostate cancer. Tumour Biol 2011; 32: 583–588. [DOI] [PubMed] [Google Scholar]




