Open Access

MicroRNA‑mediated approaches in ovarian cancer therapy: A comprehensive systematic review

  • Authors:
    • Henny Meitri Andrie Rachmasari Putri
    • Putri Wikie Novianti
    • Heru Pradjatmo
    • Sofia Mubarika Haryana
  • View Affiliations

  • Published online on: August 12, 2024     https://doi.org/10.3892/ol.2024.14624
  • Article Number: 491
  • Copyright: © Putri et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

Ovarian cancer (OC) poses a significant health risk to women worldwide, with late diagnoses and chemotherapy resistance leading to high mortality rates. Despite several histological subtypes, the primary challenge remains the subtle nature of its symptoms, resulting in advanced‑stage diagnosis and reduced treatment success rates. With platinum‑based therapies showing relative efficacy but limited survival enhancements, the emergence of chemotherapy resistance during recurrence remains a critical obstacle. Precision medicine development has aimed to address these challenges in the context of the molecular diversity of OC. The present review explored the landscape of microRNA (miRNA)‑mediated approaches in OC treatment. miRNAs have emerged as regulators of gene expression, serving as both oncogenes and tumor suppressors in OC. Dysregulated miRNAs are associated with disease progression and chemotherapy resistance, underscoring their significance in diagnosis and tailored treatment strategies. The present review extracted 295 publications from the PUBMED database. Out of the 73 eligible studies, 55 miRNAs were assessed. A total of three of these miRNAs were not associated with any disease or cancer, whilst eight were associated with OC, albeit also associated with other diseases. The present review encompassed three dimensions: i) The role of miRNAs in treatment efficacy; ii) the use of miRNAs to enhance therapy outcomes; and iii) adjunctive strategies for improved treatment results. Furthermore, it offered insights into potential avenues for improving OC treatment using miRNA‑based approaches.

Introduction

Ovarian cancer (OC) is a complex and challenging disease that affects women worldwide. As the fifth most common cancer in women (1), it poses several significant health risks, ranging from late diagnosis to chemoresistance, and contributes to ~150,000 global deaths per year, mental health problems and a financial burden for patients (2). OC is not a singular entity; it comprises several histological subtypes, each with distinct characteristics. Epithelial (E)OC is the most common type, accounting for 85–90% of OCs (3,4). It arises from the cells that cover the outer surface of the ovary. EOC includes subtypes such as serous, mucinous, endometrioid and clear cell carcinoma. Hereditary factors also serve a role, with family histories of ovarian or breast cancer increasing the risk. The association between OC and mutations in the BRCA genes, particularly BRCA1 and BRCA2, highlights the genetic contribution to the etiology of the disease. Individuals with familial histories of breast or OC carrying BRCA1/2 mutations have elevated risks, emphasizing the importance of genetic testing and precision medicine approaches for targeted risk assessment and management (57). This risk increases with age, as the median age of diagnosis is ~63 years (8).

The primary challenge in OCs pertains to its diagnosis, which is characterized by an absence of efficient early detection methods as the symptoms of OC are notoriously subtle and are often referred to as ‘stealthy assailant’ (4). This leads to patients being diagnosed in advanced stages (9), rendering surgical intervention more complex due to widespread metastasis. If effective early-stage detection is achievable, the survival rate can potentially increase to 70%. However, with an estimated early-stage detection rate of only 20%, late-stage detection with advanced cancer notably lowers the survival rate to 35% for most patients (10).

The current standard treatment for OC includes surgery and chemotherapy, alongside emerging therapies like anti-angiogenic agents, poly(ADP-ribose) polymerase inhibitors and immunotherapeutic approaches. To mitigate morbidity rates, a strategy involving pre-sensitization of cancer cells with standard therapy has been investigated, particularly effective with platinum-based agents due to its heightened response in relapsed cases, addressing drug resistance. Second-line chemotherapy selection depends on tumor sensitivity to platinum derivatives, often incorporating carboplatin or cisplatin in combination with other drug therapy (such as paclitaxel and gemcitabine) (11). However, the emergence of chemotherapy resistance, especially during disease recurrence, remains a critical challenge. Given the inherent heterogeneity of OC, a precision medicine approach aims to identify specific mutations and customize treatment strategies accordingly. This tailored approach aims to address the challenges of resistance and enhance treatment efficacy in the context of the molecular diversity of OC (12).

Within the last decades, several biomolecular studies focused on trying to address the aforementioned challenges. Following their first discovery in 1993 (13), microRNAs (miRNAs/miRs) have been studied in many diseases, including OC, to understand biological mechanisms of diseases as well as improving the diagnosis and treatment of patients. miRNAs are small RNA molecules that regulate gene expression and can act as oncogenes (promoting cancer) or tumor suppressors (inhibiting cancer) in OC. Research on miRNAs has reported that deregulation of miRNAs is one of the pathogenesis processes found in several types of OC. Furthermore, dysregulation of specific miRNAs has been associated with OC development, progression and chemoresistance (12).

miRNAs have dual roles in OC management: i) The miRNA expression profile can be used for diagnostic and prognostic purposes, forming the basis of a personalized medicine approach. By identifying specific miRNA signatures, clinicians can tailor treatment plans to improve outcomes and minimize exposure to ineffective therapies (12,14); and ii) miRNAs themselves are being explored as therapeutic agents. Synthetic miRNAs, such as miRNA mimics and inhibitors, have shown promise in preclinical studies. These synthetic miRNAs can restore normal gene expression patterns, inhibit tumor growth and enhance the sensitivity of cancer cells to chemotherapeutic agents like cisplatin. Studies have concluded that targeting miRNAs can be a viable therapeutic strategy in OC. By modulating miRNA activity, researchers aim to correct the aberrant gene expression that drives cancer progression and resistance. Therefore, the miRNA profile not only aids in selecting effective therapies but also forms the basis for developing miRNA-targeted treatments. This dual approach shows the potential of miRNA research to revolutionize OC therapy, leading to better patient outcomes (12).

The present review aimed to assess the current research landscape concerning OC treatment within the past decades, and the outcomes of the review are categorized into the following distinct dimensions: i) Understanding the role of miRNAs in influencing treatment outcomes; ii) harnessing miRNAs to improve therapy outcomes; and iii) miRNA-enhanced adjunctive strategies for therapy outcomes. In the former dimension, the review identified the underlying mechanisms of miRNAs that have been elucidated to contribute to therapy, addressing both their impacts on treatment efficacy and the development of resistance. In the two latter dimensions, a review of previously published studies that centered around miRNAs and OC revealed potential avenues for enhancing the current treatment strategies in the clinical practice.

Materials and methods

A systematic search was performed by first searching with keywords that were built around two key terms, namely: ‘Ovarian cancer’ and ‘miR’. More detailed terminology was elaborated on for each term, which further combined to construct comprehensive search keywords, allowing for a broader background while maintaining the study's emphasis (Table SI). To guarantee optimal studies that might suit the aims of the current investigation, high false-positive search results were tolerated, in which ‘therapy’-related terms were not included in the search keywords. The keywords were then used in the PubMed database (https://punmed.ncbi.nlm.nih.gov/), using the PubMed Advanced Search Builder tool. Subsequently, the titles and abstracts of the eligible studies were retrieved from the database.

The systematic review process was as follows: First, two independent reviewers scanned titles and abstracts by applying the following inclusion criteria: i) A focus on human OC; ii) use of and a focus on miRNAs; and iii) a focus on chemotherapy. Subsequently, the full report of all studies that passed the first screening were reviewed. The inclusion criteria for the second screening were as follows: i) Use of synthetic miRNAs; and ii) an available full text in the English language. All discrepancies between two reviewers were resolved by discussions to reach an agreement. The reasons for the exclusion of studies is summarized in Table SII. Finally, the following information was extracted from all the studies that passed the second screening: Year of publication, country of first author, key findings, tissues/cell lines, therapy type and miR of interest (including its regulatory functions). The reporting and workflow of the present systematic review study followed the Preferred Reporting Items for Systematic Reviews and Meta-Analysis reporting guidelines (15).

Results and Discussion

Systematic search results

As of July 2023, the keyword search resulted in 295 publications from the PubMed repository. The two-step screening process resulted in 73 eligible publications for information extraction (Fig. 1). All eligible studies exclusively used tissue samples and/or cell lines. However, this outcome was not predetermined; namely, sample types were not specifically defined during the search process as one of selection criteria. Notably, there was a relatively low annual publication count on the topic of interest, in particular the association of miRNAs with ovarian cancer chemotherapy (Fig. S1), averaging only two publications per year. The countries of the first authors were also mostly centralized to China and the United States (Fig. S2). Furthermore, 36% of the eligible studies focused on cisplatin therapy (Fig. 2). Cisplatin is a potent chemotherapy drug used in the treatment of several cancers, including OC. It is part of a class of drugs known as platinum-containing compounds and it exerts its anticancer effects by binding to and damaging the DNA in cancer cells. This damage interferes with the ability of the cells to divide and grow, ultimately leading to cell death (16).

There was a notable proportion of studies amongst the eligible studies that focused on understanding the role of miRNAs in influencing treatment outcomes (79.45%; Fig. 3), indicating substantial unknown aspects regarding treatment outcomes in OC from the perspective of miRNAs. This hypothesis was strengthened by the wide variety of miRNAs studied in the eligible studies, in which there were cumulatively 66 miRNAs investigated. A total of nine of those miRNAs were identified by the Human Gene Database (17), which were associated with OC, although they were not specific to the disease. Moreover, four miRNAs were not reported to be associated with any diseases (Table SIII).

Understanding the role of miRNAs in influencing treatment outcome

OC is known for its high mortality rate, often due to chemoresistance. This resistance significantly limits the effectiveness of treatment and often leads to the recurrence of the disease and poor patient outcomes (10,18). Table SIV presents the eligible studies that evaluated the association between miRNAs and therapy outcomes in OC, with a focus on studies that used miRNAs to assess why specific therapeutic interventions yielded suboptimal results.

miR-21 is a well-known miR that is associated with therapy resistance in OC. It is known as oncomiR in several other cancers, including cervical, colorectal and breast cancer (19). Abnormally highly expressed miR-21 may regulate drug resistance via augmented apoptotic pathways (20). As such, whether through natural compounds like icariin or by modulating c-MYB expression, targeting miR-21 may hold promise for enhancing the efficacy of therapies, particularly against drug resistance in OC. Icariin was assessed by Li et al (21) as a potential alternative therapy for OC. Icariin, the primary bioactive compound present in epimedium, a traditional Chinese medicinal herb, demonstrates diverse pharmacological properties including bolstered immune function, anticancer potential, cardiovascular enhancement and modulation of endocrine activity (21). Moreover, icariin has been reported to suppress miR-21 expression in OC cells, although the precise mechanisms underlying modulation of miR-21 expression by icariin remains unknown (21). Another study on cisplatin-resistant therapy indicated that miR-21 serves a crucial role in c-MYB-induced cisplatin resistance (22). c-MYB is a transcription factor protein encoded by the MYB gene and dysregulation of c-MYB has been implicated in several cancers, including OC, where it contributes to tumor progression and chemotherapy resistance (22). Silencing c-MYB has been reported to lead to reduced miR-21 levels, decreased epithelial-mesenchymal transmission (EMT), lowered cisplatin resistance and increased β-catenin phosphorylation (22).

Cisplatin stands as the prominent platinum-based therapy entwined with miR regulation, based on the results of the present systematic review, which revealed that 36% of the total eligible studies focused on cisplatin therapy. The emergence of cisplatin resistance poses a formidable challenge to effective treatment. The following miRNAs were mentioned due to their pivotal roles in orchestrating this resistance phenomenon: i) A high expression of miR-93 has been reported to be associated with cisplatin resistance and it can target genes that regulate apoptosis. L-tetrahydropalmatine (L-THP; an anticancer compound obtained mainly from genera Stephania and Corydalis) has been reported to suppress miR-93 expression whilst increasing PTEN levels, a pivotal tumor suppressor in OC (23). Furthermore, PTEN small interfering (si)RNA-treated cells increased survival, which was reversed by the AKT inhibitor Triciribine. L-THP enhanced OC cell sensitivity to cisplatin by modulating the miR-93/PTEN/AKT pathway (23); ii) in cisplatin-resistant OC cells, miR-1271 is often overexpressed. It can target tumor suppressors and cell cycle regulators like p53 and cyclin B1, promoting cell survival and reducing apoptosis. This, in turn, makes cancer cells more resistant to the cytotoxic effects of cisplatin (24); iii) miR-302 is known to influence cisplatin resistance through multiple mechanisms. It can affect drug transporters and drug-metabolizing enzymes. In certain cases, miR-302 can downregulate copper transporters such as solute carrier family 31 member 1, leading to decreased intracellular cisplatin accumulation. This limits the access of the drug to its target DNA (25); iv) miR-138-5p appears to influence cisplatin resistance through its ability to regulate cellular senescence and stemness. The miR was reported to be downregulated in cisplatin resistance cell-lines. Increasing miR-138-5p levels (either by inducing HOX transcript antisense RNA siRNA or by its mimics) improved chemosensitivity and reduced enhancer of zeste 2 polycomb repressive complex 2 subunit and sirtuin 1 expression, which are key players in cisplatin resistance (26); and v) high expression of miR-149-3p in OC tissues has been reported in cisplatin-resistant OC cells. Knockdown of miR-149-3p inhibited cisplatin resistance and malignant traits in OC cells. Mechanistically, miR-149-3p targeted cyclin dependent kinase inhibitor 1A and TIMP metallopeptidase inhibitor 2 to promote cisplatin resistance and EMT in OC (27). Fig. 4 summarizes key insights on cisplatin resistance from the eligible studies presented in this sub-section.

Notably, there appears to be potential associations between the aforementioned miRNAs, indicating overlapping molecular pathways or biological processes affected in OC. Table I provides a comparative analysis of the effectiveness of those miRNAs: miR-21 and miR-93 are associated with cisplatin resistance by regulating apoptosis pathways in several types of cancer; miR-1271 and miR-149-3p are also associated with cisplatin resistance in ovarian cancer cells by modulating multiple pathways involved; and miR-302 and miR-138-5p are associated with drug transport in cancer cells through the stem pathway and several other pathways. However, the aforementioned miRNAs require further experimental validation to ensure their significance and clinical implications, which further may offer promise for optimizing therapy outcomes in OC management.

Table I.

Summary of potential associations and comparative analysis of the effectiveness of certain microRNAs in enhancing therapy outcomes.

Table I.

Summary of potential associations and comparative analysis of the effectiveness of certain microRNAs in enhancing therapy outcomes.

miRPotential associationComparative analysis
miR-21 and miR-93Associated with cisplatin resistance by regulating apoptotic pathwaysTargeting miR-21 may offer broader applicability in enhancing therapy outcomes due to its involvement in multiple cancer types
miR-1271 and miR-149-3pAssociated with cisplatin resistance in ovarian cancer cellsTargeting miR-149-3p may offer broader implications for overcoming cisplatin resistance and malignant traits in ovarian cancer cells due to its modulation of multiple pathways involved in drug resistance and epithelial-mesenchymal transition
miR-302 and miR-138-5pInfluenced cisplatin resistance, albeit through distinct mechanisms; whilst miR-302 affects drug transporters, miR-138-5p regulates cellular senescence and stemnessTargeting miR-138-5p may offer potential advantages in overcoming cisplatin resistance by modulating stemness-related pathways, thereby sensitizing cancer cells to chemotherapy-induced apoptosis. However, combination strategies targeting both miRNAs may provide synergistic effects in combating cisplatin resistance by simultaneously modulating multiple pathways involved in drug resistance

[i] miRNA/miR, microRNA.

Harnessing miRNAs to improve therapy outcomes

The present review explored studies in which miRs were used to improve standard treatment outcomes via the transfection of anti-miRs or mimic-miRs. This pursuit aimed to both enhance treatment effectiveness and gain more insights into alternative instances where therapeutic interventions failed to achieve the intended outcomes, by either reinstating miR expression in tumor suppressor genes or by inhibiting the activity of oncogenic miRNAs, referred to as ‘mimic-miRs’ and ‘anti-miRs’, respectively. By assessing the scientific evidence, the present review aimed to present how miRNA-based interventions offer their potential for optimizing therapeutic strategies. The present review identified 10 studies that reported that mimic- and/or anti-miRs could help improve therapy outcomes, and cumulatively ten miRNAs of interest were discussed (Table II). Notably, none of the studies were registered in a clinical trials database. Moreover, cisplatin and paclitaxel were the most common type of therapies used in the experimental studies (Fig. 2).

Table II.

List of observed miRNAs from studies focused on improving therapy outcomes through miRNAs and other substances.

Table II.

List of observed miRNAs from studies focused on improving therapy outcomes through miRNAs and other substances.

A, Downregulated miR can improve therapy sensitivity

First author/s, yearmiRNA of interestanti-miRmimic-miRType of therapy(Refs.)
Jin and Wei, 2015miR-23aAntagomiR-23axCisplatin(28)
Rupaimoole et al, 2016miR-630Anti-miR 630amiR-630 mimicsAnti-vascular endothelial growth factor antibody and anti-miR-630(43)
García-Vázquez et al, 2018let-7d-3plet-7d-3p inhibitor and let-7d-3p antagomiRxCarboplatin and paclitaxel(30)
Ye et al, 2011miR-376canti miR-376cxCisplatin(44)
Echevarría-Vargas et al, 2014miR-21miR-21 inhibitorsxCisplatin(45)
Vandghanooni et al, 2018 anti miR-21xCisplatin(46)
Suardi et al, 2020miR-324-5pAntagonist miR-324-5paxChitosan(47)

B, Upregulated miR can improve therapy sensitivity

First author/s, yearmiRNA of interest anti-miR mimic-miRType of therapy(Refs.)

Wu et al, 2016miR-873anti-miR-873miR-630 mimicsCisplatin and paclitaxel(35)
Bieg et al, 2019miR-424-3pxmiR-424-3p mimicsCisplatin(36)
Sun et al, 2015miR-186anti-miR-186xCisplatin and paclitaxel(37)
Suardi et al, 2020miR-155-5pxmiR-155-5p mimicsChitosan(47)

a Synthetic miRs may improve therapy sensitivity, based on the corresponding study. miR/miRNA, microRNA; x, not employed.

Jin and Wei (28) indicated that when miR-23a inhibition was coupled with cisplatin treatment in OC cells, there was a significant increase in the effectiveness of cisplatin in limiting cell proliferation. miR-23a had been previously reported to be upregulated in a chemotherapy-resistant OC cell and this led to a marked increase in the rate of apoptosis in cancer cells. However, this effect was not observed when cells were treated with cisplatin alone, emphasizing the role of miR-23a in suppressing cell death and promoting drug resistance (29). Another study reported how the downregulated miR let-7d-3p could improve therapy outcomes (30). let-7d-3p is a miR associated with tumor progression and chemotherapy resistance in OC, where it is reported to be upregulated. let-7d-3p is the passenger strand and used to be considered ‘non-functional’ (30). Moreover, it is part of the let-7 family, which is comprised of 13 members that are highly conserved across species. let-7 is a key regulator of differentiation, pluripotency and apoptosis in eukaryotic cells, and has a role in cell cycle deregulation, cell division, proliferation, angiogenesis and apoptosis (31). Thus, let-7 can be used as a molecular tool and marker in cancer therapy (3134). Previous studies have reported that miR-let-7d-3p can be used as a diagnostic biomarker for non-invasive screening of ovarian cancer and its precursors (3134). To observe the potential of the miR to enhance treatment outcomes, García-Vázquez et al (30) used a two-fold strategy: i) let-7d-3p was inhibited using an antagomiR, an antagonist designed to suppress the activity of miRNAs. This inhibition led to a marked decrease in cell proliferation and the activation of apoptosis. These outcomes suggest that inhibiting let-7d-3p with the antagomiR sensitizes OC cells to therapy and increases their susceptibility to cell death; and ii) carboplatin was introduced. let-7d-3p affects the Ras pathway, which is associated with therapy resistance; therefore, targeting let-7d-3p with an antagomiR may modulate key cellular processes and signaling pathways, including ErbB and hypoxia-inducible factor-1, which are implicated in tumor progression and drug resistance (30). As such, the antagomiR for let-7d-3p inhibited the activity of carboplatin, leading to reduced cell proliferation and increased apoptosis. Suppressing let-7d-3p using an antagomiR sensitized the OC cells to carboplatin and enhanced the effectiveness of the treatment by promoting apoptosis. This two-pronged approach, using an antagomiR and chemotherapy, offers a promising strategy to improve therapy outcomes and combat the challenges of drug resistance in OC (3034).

The present review also investigated studies that focused on reinstating the expression of miRNAs which are associated as tumor suppressor genes, indicating that the injection of mimic-miR to OC cells (possibly in combination with another treatment) may yield an improved therapy outcome (35). miR-873 mimics were introduced into OC cells and the results revealed that the overexpression of miR-873 led to a marked increase in the sensitivity of the cells to cisplatin and paclitaxel. In another study, researchers aimed to enhance therapy outcomes for OC by using miR-424-3p mimics. This miR-based approach targeted the expression of galectin-3, an anti-apoptotic protein that is often overexpressed in OC and associated with resistance to chemotherapy, especially cisplatin (28). miR-424-3p mimics were transfected into OC cells to assess its impact on the expression of galectin-3. The results demonstrated that miR-424-3p mimics notably suppressed the expression of galectin-3 at the protein level. This downregulation of galectin-3 is critical as this protein is associated with inhibiting apoptosis, a process that serves a central role in the response of the cell to chemotherapy. Following that, the cell viability and proliferation rates decreased, indicating a reduced capacity for the cells to grow and divide. Moreover, apoptosis was enhanced, with the cells treated with miR-424-3p mimics and cisplatin demonstrating a marked increase in apoptosis compared with the control group. Another similar study that assessed the enhancement of platinum-based-therapy through mimic-miRs, reported similar results, although they used miR-186 as their main object of observation (33).

miRNA-enhanced adjunctive strategies for therapy outcomes

Finally, the present study investigated adjunctive substances that have the potential to enhance therapy outcomes in OC, with a focus on their interactions with anti- and mimic miRs. The aim was to provide insights into emerging strategies that leverage miRNAs in conjunction with other compounds to optimize the effectiveness of OC treatments. For example, nanoparticles have been used for targeted miR therapy as drug delivery systems for miRNA-based cancer therapy (3638) (Table III). These nanoparticles were engineered to efficiently deliver anti-miR or miR-mimics payloads to OC cells, specifically either targeting overexpressed oncogenic miRNAs or reinstating miR expression in tumor suppressor genes. The five studies presented in Table III demonstrated innovative approaches to OC therapy by emphasizing the use of nanotechnology and miRNA-based strategies to target oncogenic miRNAs, sensitize drug-resistant cells, and enhance the precision and effectiveness of treatment.

Table III.

Observed miRNAs from studies which focused on improving therapy outcomes through microRNAs and other substances.

Table III.

Observed miRNAs from studies which focused on improving therapy outcomes through microRNAs and other substances.

First author/s, yearmiRNA of interest anti-/mimic-miRsSubstance(Refs.)
Bertucci et al, 2019miR-21anti-miR-21CGKRK-pSiNP(38)
Javanmardi et al, 2020 PEG2k-CMPEI-ss(39)
Vandghanooni et al, 2020miR-214anti-miR-214Ap-CIS-PCL NPs(40)
Gandham et al, 2022miR-let-7bmiR-let-7b mimicsHyaluronic acid-based nanoparticle(41)
Zhao et al, 2022miR-484miR-484 mimicsRGD-modified exosomes(42)

[i] miR/miRNA, microRNA; CGKRK-pSiNP, intravesically-administered cell-penetrating peptide (Cys-Gly-Lys-Arg-Lys)-porous silicon nanoparticles; PEG2k-CMPEI-ss, PEGylation 2000-subsequent carboxymethylation reaction; Ap-CIS-PCL NPs, Ap-functionalized polyethylene gylated-polycaprolactone encapsulating cisplatin nanoparticles.

miR-21 serves a role in therapy resistance in OCs, indicating its potential to regulate drug resistance via apoptosis. Furthermore, it serves an important role in the oncogenic process as indicated by its association with the high proliferation, low apoptosis, high invasion and metastatic potential of cancer cells. Whilst evaluating the potential of anti-miR-21 as a therapeutic strategy for countering the oncogenic effects of miR-21 in OC, studies have concurrently explored the enhancement of treatment outcomes through the efficient nanoparticle-based delivery of anti-miR-21 to cancer cells whilst minimizing off-target effects. Biodegradable porous silicon nanoparticles were engineered to encapsulate an anti-miR-21 locked nucleic acid payload (38). Additionally, these nanoparticles displayed a tumor-homing peptide that enabled targeted distribution. This targeting peptide, CGKRK, ensured that the nanoparticles accumulated primarily in the tumor environment. CGKRK is a tumor-tracking peptide discovered by phage display techniques and it shows high selective binding affinity to neovascular endothelial cells and tumor tissue, but not to non-tumorigenic cells (39). Another nanoparticle, redox-sensitive, Polyethylene (PEG)-shielded carboxymethyl polyethylenimine (PEI) nanogels were used as a delivery system for anti-miR-21 (39). These nanogels were designed to efficiently deliver the anti-miR-21 to the OC cells by modifying branched PEI, which involved PEGylation (PEG2k-PEI) for steric shielding, redox-sensitive crosslinking for nanogel synthesis (PEG2k-PEI-ss nanogels) and carboxymethylation (PEG2k-CMPEI-ss) to modulate the properties of the polymer. Another study aimed to develop a novel polymeric drug delivery system (DDS) using star-shaped glucose-core polycaprolactone-polyethylene glycol (Glu-PCL-PEG) block copolymer nanoparticles. This system was used to deliver cisplatin and locked nucleic acid anti-miR-214 to OC cells. The study confirmed that nucleolin-mediated endocytosis of the targeted polymeric DDS containing both cisplatin and anti-miR-214 A2780 R cells led to enhanced apoptosis (40).

Other studies with a specific focus on addressing relapse and multidrug resistance issues have assessed let-7b (41) and miR-484 (42). Both studies recognized tumor suppressors with the capacity to target a range of oncogenes and chemoresistance-related genes. let-7b has an important role in growth and proliferation and induces apoptosis of cancer cells by inhibiting cell cycle progression through pathways such as CYP2J2 regulation and Wnt/B-catenin pathway. Downregulation of let-7b levels in clinical OC has been linked to chemotherapy resistance, heightened proliferation, invasion and relapse in EOCs. The restoration of let-7b expression was pivotal as it can significantly heighten sensitivity to chemotherapy whilst inhibiting oncogenic pathways and thwarting chemoresistance mechanisms (41). To ensure effective delivery of let-7b to tumor cells, innovative nanoparticle systems (namely, hyaluronic acid-based nanoparticles and Glu-PCL-PEG nanoparticles) were used to act as versatile delivery vehicles adept at transporting nucleic acids like miRNA. The delivery of let-7b to tumor cells involved using HA-PEI as a delivery vehicle, which is a versatile system known for efficient nucleic acid delivery, including miR (41). The strategic combination of let-7b with these nanoparticles represents a multifaceted approach aimed at reprogramming the miR profile within tumor cells.

Another study that also assessed a tumor suppressor miRNA, provided mechanistic insights into how the delivery of miR-484 via exosomes impacted the tumor vasculature and chemotherapy sensitization (42). It revealed that miR-484 may serve a role in vessel normalization, which enhances the response of cancer cells to chemotherapy-induced apoptosis. Mechanistically, miR-484 may achieve this effect by simultaneously inhibiting the expression of VEGF-A in cancer cells and its corresponding receptors in endothelial cells. The research demonstrates how miR-484 and exosomal delivery contributed to vascular normalization and chemotherapy sensitization in OC.

Limitations

Despite the comprehensive nature of this systematic review, several limitations may affect the generalizability and applicability of the findings. First, this study was limited to the PubMed database, potentially excluding relevant studies indexed in other databases, such as Scopus, Web of Science or Embase. Second, the included studies varied widely in their methodologies, miRNAs investigated and therapeutic interventions used. This heterogeneity makes it challenging to draw definitive conclusions and compare results across studies. Finally, while the review highlighted associations between miRNAs and treatment outcomes, detailed mechanistic insights were often lacking. Understanding the precise molecular mechanisms is crucial for the development of effective therapeutic strategies.

Conclusion

The present systematic review highlights the role of miRNAs in enhancing OC treatment outcomes. Half of the selected studies, which focused on understanding the impact of miRNAs on treatment outcomes, highlighted the presence of numerous unanswered questions, indicating significant unknowns about OC therapy from the perspective of miRNAs and the need for more comprehensive and ongoing research in this area. The present analysis reveals potential associations between miRNAs that may imply those miRNAs affect similar molecular pathways or biological processes in OC. However, further research is needed to confirm their significance and clinical implications. Furthermore, the innovative use of nanotechnology for targeted miR delivery represents a significant advancement in treatment precision. The findings of the present review demonstrate the intricate relationship between miRNAs and therapy outcomes, providing valuable insights for future research directions and the development of miRNA-based therapeutic interventions in OC management.

Supplementary Material

Supporting Data
Supporting Data

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

The data generated in the present study may be requested from the corresponding author.

Authors' contributions

HMARP designed the study, formulated the strategy for the literature search, performed the review and information extraction, confirmed the authenticity of the data, wrote and reviewed the article. PWN performed a systematic search, reviews and information extraction, confirmed the authenticity of the data, drafted and wrote the article. HP and SMH analyzed the data, drafted and reviewed the article. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI

2 

Webb PM and Jordan SJ: Epidemiology of epithelial ovarian cancer. Best Pract Res Clin Obstet Gynaecol. 41:3–14. 2017. View Article : Google Scholar : PubMed/NCBI

3 

Bast RC Jr, Hennessy B and Mills GB: The biology of ovarian cancer: New opportunities for translation. Nat Rev Cancer. 9:415–428. 2009. View Article : Google Scholar : PubMed/NCBI

4 

Shao C, Guo H, Chen L, Chen J, Wang L and Wang H: Prognostic factors and clinic-pathologic characteristics of ovarian tumor with different histologic subtypes-a SEER database population study of 41,376 cases. Transl Cancer Res. 12:1937–1950. 2023. View Article : Google Scholar : PubMed/NCBI

5 

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

6 

Sekine M, Nishino K and Enomoto T: Differences in ovarian and other cancers risks by population and BRCA mutation location. Genes (Basel). 12:10502021. View Article : Google Scholar : PubMed/NCBI

7 

Vos S, van Diest PJ and Moelans CB: A systematic review on the frequency of BRCA promoter methylation in breast and ovarian carcinomas of BRCA germline mutation carriers: Mutually exclusive, or not? Crit Rev Oncol Hematol. 127:29–41. 2018. View Article : Google Scholar : PubMed/NCBI

8 

Kurman RJ and Shih IM: The dualistic model of ovarian carcinogenesis: Revisited, revised, and expanded. Am J Pathol. 186:733–747. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Doubeni CA, Doubeni AR and Myers AE: Diagnosis and management of ovarian cancer. Am Fam Physician. 93:937–944. 2016.PubMed/NCBI

10 

Chandra A, Pius C, Nabeel M, Nair M, Vishwanatha JK, Ahmad S and Basha R: Ovarian cancer: Current status and strategies for improving therapeutic outcomes. Cancer Med. 8:7018–7031. 2019. View Article : Google Scholar : PubMed/NCBI

11 

Cortez AJ, Tudrej P, Kujawa KA and Lisowska KM: Advances in ovarian cancer therapy. Cancer Chemother Pharmacol. 81:17–38. 2018. View Article : Google Scholar : PubMed/NCBI

12 

Alshamrani AA: Roles of microRNAs in ovarian cancer tumorigenesis: Two decades later, what have we learned? Front Oncol. 10:10842020. View Article : Google Scholar : PubMed/NCBI

13 

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

14 

Kinose Y, Sawada K, Nakamura K and Kimura T: The role of microRNAs in ovarian cancer. Biomed Res Int. 2014:2493932014. View Article : Google Scholar : PubMed/NCBI

15 

Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC and Mulrow CD: PRISMA_2020_flow_diagram_new_SRs_v1 [Internet]. BMJ. 3722017.Available from:. http://www.prisma-statement.org/PRISMAStatement/FlowDiagram

16 

Aldossary SA: Review on pharmacology of cisplatin: Clinical use, toxicity and mechanism of resistance of cisplatin. Biomed Pharmacol J. 12:7–15. 2019. View Article : Google Scholar

17 

Stelzer G, Rosen N, Plaschkes I, Zimmerman S, Twik M, Fishilevich S, Stein TI, Nudel R, Lieder I, Mazor Y, et al: The GeneCards suite: From gene data mining to disease genome sequence analyses. Curr Protoc Bioinformatics. 54:1.30.1–1.30.33. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Wang S, Li MY, Liu Y, Vlantis AC, Chan JYK, Xue L, Hu BG, Yang S, Chen MX, Zhou S, et al: The role of microRNA in cisplatin resistance or sensitivity. Expert Opin Ther Targets. 24:885–897. 2020. View Article : Google Scholar : PubMed/NCBI

19 

Wang Y, Zeng G and Jiang Y: The emerging Roles of miR-125b in cancers. Cancer Manag Res. 12:1079–1088. 2020. View Article : Google Scholar : PubMed/NCBI

20 

Chan JK, Blansit K, Kiet T, Sherman A, Wong G, Earle C and Bourguignon LY: The inhibition of miR-21 promotes apoptosis and chemosensitivity in ovarian cancer. Gynecol Oncol. 132:739–744. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Li J, Jiang K and Zhao F: Icariin regulates the proliferation and apoptosis of human ovarian cancer cells through microRNA-21 by targeting PTEN, RECK and Bcl-2. Oncol Rep. 33:2829–2836. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Zhang XY, Li YF, Ma H and Gao YH: Regulation of MYB mediated cisplatin resistance of ovarian cancer cells involves miR-21-wnt signaling axis. Sci Rep. 10:68932020. View Article : Google Scholar : PubMed/NCBI

23 

Gong J, Xing C, Wang LY, Xie SS and Xiong WD: L-Tetrahydropalmatine enhances the sensitivity of human ovarian cancer cells to cisplatin via microRNA-93/PTEN/Akt cascade. J BUON. 24:701–708. 2019.PubMed/NCBI

24 

Chen Y, Wang L and Zhou J: Effects of microRNA-1271 on ovarian cancer via inhibition of epithelial-mesenchymal transition and cisplatin resistance. J Obstet Gynaecol Res. 45:2243–2254. 2019. View Article : Google Scholar : PubMed/NCBI

25 

Ge T, Liu T, Guo L, Chen Z and Lou G: MicroRNA-302 represses epithelial-mesenchymal transition and cisplatin resistance by regulating ATAD2 in ovarian carcinoma. Exp Cell Res. 396:1122412020. View Article : Google Scholar : PubMed/NCBI

26 

Zhang Y, Ai H, Fan X, Chen S, Wang Y and Liu L: Knockdown of long non-coding RNA HOTAIR reverses cisplatin resistance of ovarian cancer cells through inhibiting miR-138-5p-regulated EZH2 and SIRT1. Biol Res. 53:182020. View Article : Google Scholar : PubMed/NCBI

27 

Wang J and Liu L: MiR-149-3p promotes the cisplatin resistance and EMT in ovarian cancer through downregulating TIMP2 and CDKN1A. J Ovarian Res. 14:1652021. View Article : Google Scholar : PubMed/NCBI

28 

Jin AH and Wei ZL: Molecular mechanism of increased sensitivity of cisplatin to ovarian cancer by inhibition of microRNA-23a expression. Int J Clin Exp Med. 8:13329–13334. 2015.PubMed/NCBI

29 

Chhabra R, Dubey R and Saini N: Cooperative and individualistic functions of the microRNAs in the miR-23a~27a~24-2 cluster and its implication in human diseases. Mol Cancer. 9:2322010. View Article : Google Scholar : PubMed/NCBI

30 

García-Vázquez R, Gallardo Rincón D, Ruiz-García E, Meneses García A, Hernández De La Cruz ON, Astudillo-De La Vega H, Isla-Ortiz D, Marchat LA, Salinas-Vera YM, Carlos-Reyes Á, et al: let-7d-3p is associated with apoptosis and response to neoadjuvant chemotherapy in ovarian cancer. Oncol Rep. 39:3086–3094. 2018.PubMed/NCBI

31 

Zheng H, Zhang L, Zhao Y, Yang D, Song F, Wen Y, Hao Q, Hu Z, Zhang W and Chen K: Plasma miRNAs as diagnostic and prognostic biomarkers for ovarian cancer. PLoS One. 8:e778532013. View Article : Google Scholar : PubMed/NCBI

32 

Langhe R, Norris L, Saadeh FA, Blackshields G, Varley R, Harrison A, Gleeson N, Spillane C, Martin C, O'Donnell DM, et al: A novel serum microRNA panel to discriminate benign from malignant ovarian disease. Cancer Lett. 356:628–636. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Kobayashi M, Salomon C, Tapia J, Illanes SE, Mitchell MD and Rice GE: Ovarian cancer cell invasiveness is associated with discordant exosomal sequestration of Let-7 miRNA and miR-200. J Transl Med. 12:42014. View Article : Google Scholar : PubMed/NCBI

34 

Chung YW, Bae HS, Song JY, Lee JK, Lee NW, Kim T and Lee KW: Detection of microRNA as novel biomarkers of epithelial ovarian cancer from the serum of ovarian cancer patients. Int J Gynecol Cancer. 23:673–679. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Wu DD, Li XS, Meng XN, Yan J and Zong ZH: MicroRNA-873 mediates multidrug resistance in ovarian cancer cells by targeting ABCB1. Tumor Biol. 37:10499–10506. 2016. View Article : Google Scholar

36 

Bieg D, Sypniewski D, Nowak E and Bednarek I: MiR-424-3p suppresses galectin-3 expression and sensitizes ovarian cancer cells to cisplatin. Arch Gynecol Obstet. 299:1077–1087. 2019. View Article : Google Scholar : PubMed/NCBI

37 

Sun KX, Jiao JW, Chen S, Liu BL and Zhao Y: MicroRNA-186 induces sensitivity of ovarian cancer cells to paclitaxel and cisplatin by targeting ABCB1. J Ovarian Res. 8:802015. View Article : Google Scholar : PubMed/NCBI

38 

Bertucci A, Kim KH, Kang J, Zuidema JM, Lee SH, Kwon EJ, Kim D, Howell SB, Ricci F, Ruoslahti E, et al: Tumor-targeting, MicroRNA-silencing porous silicon nanoparticles for ovarian cancer therapy. ACS Appl Mater Interfaces. 11:23926–23937. 2019. View Article : Google Scholar : PubMed/NCBI

39 

Javanmardi S, Tamaddon AM, Aghamaali MR, Ghahramani L and Abolmaali SS: Redox-sensitive, PEG-shielded carboxymethyl PEI nanogels silencing MicroRNA-21, sensitizes resistant ovarian cancer cells to cisplatin. Asian J Pharm Sci. 15:69–82. 2020. View Article : Google Scholar : PubMed/NCBI

40 

Vandghanooni S, Eskandani M, Barar J and Omidi Y: Antisense LNA-loaded nanoparticles of star-shaped glucose-core PCL-PEG copolymer for enhanced inhibition of oncomiR-214 and nucleolin-mediated therapy of cisplatin-resistant ovarian cancer cells. Int J Pharm. 573:1187292020. View Article : Google Scholar : PubMed/NCBI

41 

Gandham SK, Rao M, Shah A, Trivedi MS and Amiji MM: Combination microRNA-based cellular reprogramming with paclitaxel enhances therapeutic efficacy in a relapsed and multidrug-resistant model of epithelial ovarian cancer. Mol Ther Oncolytics. 25:57–68. 2022. View Article : Google Scholar : PubMed/NCBI

42 

Zhao Z, Shuang T, Gao Y, Lu F, Zhang J, He W, Qu L, Chen B and Hao Q: Targeted delivery of exosomal miR-484 reprograms tumor vasculature for chemotherapy sensitization. Cancer Lett. 530:45–58. 2022. View Article : Google Scholar : PubMed/NCBI

43 

Rupaimoole R, Ivan C, Yang D, Gharpure KM, Wu SY, Pecot CV, Previs RA, Nagaraja AS, Armaiz-Pena GN, McGuire M, et al: Hypoxia-upregulated microRNA-630 targets Dicer, leading to increased tumor progression. Oncogene. 35:4312–4320. 2016. View Article : Google Scholar : PubMed/NCBI

44 

Ye G, Fu G, Cui S, Zhao S, Bernaudo S, Bai Y, Ding Y, Zhang Y, Yang BB and Peng C: MicroRNA 376c enhances ovarian cancer cell survival by targeting activin receptor-like kinase 7: Implications for chemoresistance. J Cell Sci. 124:359–368. 2011. View Article : Google Scholar : PubMed/NCBI

45 

Echevarría-Vargas IM, Valiyeva F and Vivas-Mejía PE: Upregulation of miR-21 in cisplatin resistant ovarian cancer via JNK-1/c-Jun pathway. PLoS One. 9:e970942014. View Article : Google Scholar : PubMed/NCBI

46 

Vandghanooni S, Eskandani M, Barar J and Omidi Y: AS1411 aptamer-decorated cisplatin-loaded poly(lactic-co-glycolic acid) nanoparticles for targeted therapy of miR-21-inhibited ovarian cancer cells. Nanomedicine (Lond). 13:2729–2758. 2018. View Article : Google Scholar : PubMed/NCBI

47 

Suardi RB, Ysrafil Y, Sesotyosari SL, Martien R, Wardana T, Astuti I and Haryana SM: The effects of combination of mimic miR-155-5p and antagonist miR-324-5p encapsulated chitosan in ovarian cancer SKOV3. Asian Pac J Cancer Prev. 21:2603–2608. 2020. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2024
Volume 28 Issue 4

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Putri HR, Novianti PW, Pradjatmo H and Haryana SM: MicroRNA‑mediated approaches in ovarian cancer therapy: A comprehensive systematic review. Oncol Lett 28: 491, 2024.
APA
Putri, H.R., Novianti, P.W., Pradjatmo, H., & Haryana, S.M. (2024). MicroRNA‑mediated approaches in ovarian cancer therapy: A comprehensive systematic review. Oncology Letters, 28, 491. https://doi.org/10.3892/ol.2024.14624
MLA
Putri, H. R., Novianti, P. W., Pradjatmo, H., Haryana, S. M."MicroRNA‑mediated approaches in ovarian cancer therapy: A comprehensive systematic review". Oncology Letters 28.4 (2024): 491.
Chicago
Putri, H. R., Novianti, P. W., Pradjatmo, H., Haryana, S. M."MicroRNA‑mediated approaches in ovarian cancer therapy: A comprehensive systematic review". Oncology Letters 28, no. 4 (2024): 491. https://doi.org/10.3892/ol.2024.14624