Review For reprint orders, please contact:
[email protected] miRNAs as prognostic and therapeutic tools in epithelial ovarian cancer
Epithelial ovarian cancer (EOC) is the most lethal gynecologic malignancy and is the fifth leading cause of cancer deaths in women. Developing adjuvant therapy to circumvent drug resistance represents an important aspect of current initiatives to improve survival in women with advanced EOC. A regulatory molecule that can act on multiple genes associated with a chemoresistant phenotype will be the ideal target for the development of therapeutics to overcome resistance and miRNAs constitute promising tools in this regard. In this review, we discuss the emerging role of miRNAs in regulating EOC phenotype with a focus on prognostic and therapeutic importance of miRNAs and the possibility of miRNA modulation as a tool to improve efficacy of chemotherapy in EOC.
Anil Belur Nagaraj1, Peronne Joseph1 & Analisa DiFeo*,1 Case Comprehensive Cancer Center, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH 44106, USA *Author for correspondence: Tel.: +1 216 368 3117 Analisa.Difeo@ case.edu 1
Keywords: biomarkers • chemotherapy resistance • epithelial ovarian cancer • miRNA expression profiling • miRNAs • MRX34 • OncomiRs • pharmaco-miR • tumor heterogeneity • tumor suppressor miRs
Barriers in eradicating epithelial ovarian cancer Epithelial ovarian cancer (EOC) is the most lethal gynecologic malignancy and is the fifth leading cause of cancer deaths in women [1] . This year, it is estimated that approximately 22,000 women will be newly diagnosed and 14,200 will succumb to this lethal disease in the USA [1] . This poor overall survival is due to late-stage diagnosis and almost inevitable resistance to chemotherapy. Most cases of EOC are high-grade serous cancers (HGSOC), which are initially highly sensitive to standard treatment including cytoreduction surgery and platinum-based chemotherapy with response rate close to 85% [2] . In spite of such high initial response to platinum, the outcomes are poor with 5-year survival being less than 30% and the majority of women who initially respond to platinum therapy relapse due to development of chemoresistance [2] . Hence, there is immediate need of early diagnostic biomarkers and novel therapeutic tools that can enhance the efficiency of platinum-based therapy by overcoming chemoresistance in EOC.
10.2217/BMM.14.108 © 2015 Future Medicine Ltd
The world of miRNAs miRNAs are approximately 20–22 nucleotide length noncoding RNAs that regulate gene expression mainly by posttranslational mechanisms involving sequence-specific interaction with their targets [3,4] . miRNAs are transcribed as primary transcripts in the nucleus and undergo a series of tightly regulated cropping and cleavage events, which eventually result in functional mature miRNAs in the cytoplasm [3,4] . Being part of such a complex transcription and processing machinery [5] coupled with their nature of sequence-specific recognition of multiple targets, miRNAs are characterized by diverse expression profile and have emerged as important regulators of cell proliferation and differentiation processes [3] . miRNA deregulation is implicated in pathogenesis of a variety of tumors including ovarian cancer and miRNAs can be classified as tumor suppressor miRNAs or oncomiRs based on their functions in regulating tumor phenotype [6–8] . miRNAs have been identified as critical regulators of various aspects
Biomark. Med. (2015) 9(3), 241–257
part of
ISSN 1752-0363
241
Review Nagaraj, Joseph & DiFeo of tumorigenesis including transformation events, metastatic events, cancer stem cell functions, chemoresistant mechanisms and hence miRNA modulation is emerging as an exciting prospect in cancer t herapeutics [9–11] . miRNAs & EOC PubMed search with ‘miRNAs and cancer’ lists around 12843 publications, whereas search with the term ‘miRNAs and epithelial ovarian cancer’ lists around 86 publications; thus, publications in EOC regarding miRNAs constitute a very tiny proportion of ~0.6% of overall publications describing miRNA roles in cancer indicating that there is quite some distance to be covered in this scenario. Around ~70% of these miRNA publications in EOC have been published in the last 2 years, indicating that miRNAs are currently an important focus in understanding mechanisms regulating EOC phenotype. Importance of novel & reliable prognostic markers in EOC
Failure to efficiently diagnose the disease in early stage and emergence of chemoresistance to platinum-based compounds are the major barriers to success in treating EOC. miRNA deregulation has been identified as an important feature in mechanisms underlying EOC [12,13] . A plethora of mechanisms including miRNA deregulation have been implicated in emergence of acquired resistance during the course of standard platinum-based treatment regimens in EOC [14] . The chemoresistance problem in EOC has many dimensions that need to be tackled. To begin with, there is lack of biomarkers that can identify the disease process in early stage in which treatment outcomes are most successful. Second, there is lack of reliable patient stratification strategies for platinum-based chemotherapy in EOC, which limits the application of targeted therapy strategies. Lastly, there is lack of reliable biomarkers that can be employed to efficiently monitor response during chemotherapy enabling early detection of chemoresistance. In spite of being greatly heterogeneous in terms of origin and composition, all EOC tumors are initially treated the same, i.e. with platinum compounds [15] . This is a major problem since platinum treatment is reported to increase the expression of stem-cell markers in tumors during the course of chemotherapy resulting in treatment failure and tumor relapse due to accumulation of stem-like cancer cells, which are drug resistant [16,17] . Hence, identification of potential responders (mostly with good survival outcome) and potential nonresponders (mostly with poor survival outcome) at the beginning of chemotherapy is critical
242
Biomark. Med. (2015) 9(3)
to improve patient outcome and prevent tumor relapse in EOC [18] . Unfortunately, this is a great challenge in current EOC treatment scenario. Even though there are few ongoing-targeted therapeutic approaches in ovarian cancers employing antiangiogenic drugs and PARP inhibitors, the main challenges in these studies are lack of reliable biomarkers that enable efficient patient stratification and also reliably portray response to chemotherapy [19] . Over the last few decades, CA-125 has remained the most extensively employed prognostic factor in EOCs to predict survival, monitor disease progression and response to chemotherapy in ovarian cancer, but varying observations have been documented in different studies [20] . Analysis of The Cancer Genome Atlas (TCGA) gene expression dataset of high-grade serous EOC samples did not support a major effect of CA-125 in predicting overall survival and more importantly no correlation was observed between CA-125 expression levels and resistance to chemotherapy [21] highlighting the need for novel predictors of chemoresistance in EOC. miRNAs as prognostic markers in EOC miRNAs are attractive candidates as biomarkers in cancers including EOC because of their unique properties namely: miRNA deregulation is an established hallmark of cancers [6] ; miRNAs are characterized by tissue-specific expression and miRNA signatures can classify cancers [22] ; miRNAs are known to be very stable in formalin-fixed tissues, which are the common source of samples for biomarker analysis [23] ; and miRNAs are present in body fluids [24] , which makes their analysis possible by less invasive methods and hence more practical for biomarker analysis in clinical settings. miRNA expression profiling is being employed as a powerful tool to study miRNA deregulation in tumors and miRNAs are emerging as reliable prognostic tools in predicting survival outcome in EOC [25] . Various technologies are available to investigate the use of miRNAs as diagnostic entities in clinical settings (as summarized in Figure 1) and each method has its own advantages and disadvantages and the choice of these techniques is mostly dependent on the clinical settings encountered [26,27] . miRNAs & early diagnosis in EOC
miRNAs are known to play an important role in regulating ovarian physiology [28] and hence it is not surprising that miRNA deregulation is a prominent feature in EOCs [7] . Interestingly, miRNAs like let-7 tumor suppressor family, miR-125b and miR-29b that are known to be highly expressed in normal ovarian tissue [28] are often deregulated in EOCs, thus linking
future science group
miRNAs as prognostic & therapeutic tools in epithelial ovarian cancer
Review
Samples Tissue microRNAs
Circulating microRNAs
[Fresh/fixed]
[Serum] RNA isolation ± enrichment
Real-time PCR
microRNA microarray
RNA sequencing
Bead-based flow cytometry
Fast
Slow
Slow
Fast
Input RNA
Moderate
Low-moderate
Moderate
Low-moderate
Sensitivity
High
Low
Moderate
High
Moderate
Moderate
High
Moderate
Low
High
High
Low
[Relative] Speed
Cost Infrastructure
Figure 1. Flowchart illustrating various techniques that are currently being employed in microRNA research in both laboratory- and clinic-based settings, and also summarizes the relative advantages associated with each methodology.
EOC pathogenesis to potential miRNA deregulation events during normal ovarian functioning. Functional delineation of miRNAs involved in early transformation events occurring in normal ovarian tissue is necessary to realize the possibility of employing miRNAs as tools for early diagnosis of the disease process leading to EOC. To date only one miRNA has been implicated in EOC in vitro transformation, namely miR-182 [29] . One of the major challenges in this context is the availability of reliable genetically engineered in vivo models that can faithfully recapitulate early events of EOC pathogenesis. EOC pathogenesis is highly heterogeneous both in terms of origin and composition of the tumor, which makes the generation of such oncogenic in vivo models a tough task, and this is further complicated by the fact that there is lack of understanding regarding specific genetic drivers of EOC phenotype. One potentially efficient model in this context was described recently wherein a de novo mouse model of HGSOC was generated through the deletion of p53, Brca1 or Brca2 and PTEN within the fallopian tube using the Pax8 promoter [30] . Combination of such genetically engineered EOC models with miRNA knockin or knockout mouse models should shed more light into understanding the role of miRNAs in regulating early transformation events in EOC pathogenesis and thus could pave the way for the development of reliable miRNA-based early diagnostic strategies in EOC.
future science group
The most common genetic alterations in EOC pathogenesis are reported to revolve around the two major tumor suppressor genes BRCA1 and TP53 [31] and interestingly both BRCA1 and TP53 are known to extensively cross talk with miRNA machinery, which is important for their functioning; thus, suggesting miRNA deregulation could be involved in the pathogenesis of EOC [32,33] . miR-29a and miR-29b expression was reported to be upregulated in high-grade serous EOC with BRCA1/2 abnormalities [33] . Loss of miR-31 is reported to be associated with defects in p53 pathway in EOC and overexpression of miR31 in p53-deficient background showed therapeutic effects [34] . Thus, miRNA deregulation events could potentially underlie the early transformation events in EOC pathogenesis and further studies are required in this context to develop a reliable miRNA diagnostic platform for early diagnosis of EOC. miRNAs predicting survival in EOC
One of the earliest reports describing miRNA deregulation in EOC was from the laboratory of Carlo Croce [7] in which miRNA signatures in EOC were analyzed and 39 miRNAs were found to be significantly deregulated in tumor tissues as compared with normal ovary [7] . The tumor suppressor miR-200 family was one of the earliest identified miRNA prognostic markers in EOC even though there are differences in observations from different groups about this family of miRNA,
www.futuremedicine.com
243
Review Nagaraj, Joseph & DiFeo which is probably due to the differences in methodology employed and cohorts investigated [7,35] . Since these initial findings in EOC, several other groups have reported differential miRNA expression in EOC, including miR-21 upregulation and miR-125b downregulation correlation with poor patient outcome in HGSOC [36] ; miR-34c was identified as sole independent predictor of recurrence-free survival with higher miR-34c levels favoring good prognosis in EOC [33] and Kim et al. reported that significant upregulation of miR-519a is associated with poor survival outcome in HGSOC [37] . In perhaps the most extensive analysis of miRNA signatures in EOC, TCGA provided a comprehensive analysis of HGSOC by integrated analyses of various parameters including miRNA expression profiles and identified four molecular subtypes of ovarian cancer [31] . miRNA expression clustering in this analysis revealed three subtypes with subtype 1 being associated with poor survival. Further refinement of these data identified a miRNA regulatory network that defined a mesenchymal subtype associated with poor survival [38] . Interestingly, only eight downregulated miRNAs identified in this analysis (miR-25, miR506, miR-29c, miR-182, miR-128, miR-101, miR-141 and miR-200a) were predicted to regulate 89% of the altered miRNA-associated genes, thus suggesting the importance of miRNA networks as predictors of survival in EOC. miRNAs predicting chemoresistance in EOC
Predicting potential chemoresistant tumors both at the beginning and during the course of chemotherapy is critical to the planning and success of targeted therapy strategies and one of the critical constraints in this context is the requirement of reliable biomarkers that reliably reflect the response to chemotherapy [39] . miRNA deregulation is a characteristic feature of chemotherapy resistance in ovarian tumors [12] . In one of the earliest reports in this regard, Sorrentini et al. identified expression of five miRNAs (let-7e, miR-30c, miR125b, miR-130a and miR-335) to be altered in drug resistant ovarian tumor cell lines [40] . let-7i was the first miRNA reported to be significantly decreased in chemoresistant primary EOC samples and it was further observed in this study that lower let-7i levels correlated with shorter survival in EOC [41] . Eitan et al. reported miRNA signature predicting the response of EOC patients to platinum-based chemotherapy and this also correlated with survival outcome [39] . In addition, a cluster of eight miRNAs located at chrXq27.3 locus that were consistently downregulated in EOC patients who experienced early relapse and lower expression of this cluster correlated with shorter survival time
244
Biomark. Med. (2015) 9(3)
in these patients [42] . Furthermore, Vecchione et al. identified a signature of 23 miRNAs associated with chemoresistance in EOC and from this set further validated that three miRNAs (miR-484, miR-642 and miR-217) that were able to predict chemoresistance in these tumors. Our lab recently identified miR-181a as a predictor of tumor recurrence and chemoresistance in HGSOC [43] . miR-181a levels were associated with poor outcome and shorter time to recurrence in advanced-stage EOC; higher expression predicting poor survival and early recurrence thus indicating that miR-181a could be an important prognostic tool in EOC. Circulating miRNAs as prognostic markers in EOC
One of the unique features of miRNAs is that they are present in body fluids and can be detected in serum samples, which is the one of the least invasive sample available for biomarker analysis. Hence, circulating miRNAs are increasingly gaining importance as biomarkers in various cancers even though there are some preprofiling factors that have to be taken into consideration that can significantly affect such diagnostic approaches [44,45] . miRNAs that are secreted from cells into circulation are known to be very stable because they exist in different forms like in complex with AGO2 protein and within vesicles called exosomes that confer resistance to degradation by RNAses [44] . One of the very first studies identifying circulating miRNAs as potential diagnostic entities in cancer was reported in 2008 by the laboratory of Muneesh Tewari [24] . In the same year, Resnick et al. identified miR-92 to be consistently overexpressed in serum of EOC patients as compared with healthy controls [46] . Following these studies, it was reported that miR-200 family miRNAs were greatly elevated in the serum of EOC patients as compared with age-matched healthy controls [47] . This observation was consistent with previous reports identifying miR-200 family miRNAs to be upregulated in EOC tissue samples. miRNAs in serum of EOC patients have been shown to correlate with survival outcome suggesting that circulating miRNAs could be of prognostic importance in EOC. High serum expression of miR-221 was identified as an independent unfavorable prognostic factor in EOC showing that serum miRNA expression can correlate with survival outcome in EOC [48] . miR21 was another miRNA to be identified in serum of EOC patients whose expression correlated with survival outcome; higher expression being associated with shortened overall survival [49] . A higher miR-92 level in serum EOC patients was observed to correlate with advanced disease stage [50] . In an interesting study that
future science group
miRNAs as prognostic & therapeutic tools in epithelial ovarian cancer
Review
Table 1. Important studies describing the effect of miRNA modulation on platinum resistance in epithelial ovarian cancer. miRNA
Modulation
Platinum response
Targets
Study type
let-7e
Overexpression knockdown
Sensitivity upon overexpression
EZH2 CCND1
In vitro and in vivo
[95]
miR-29
Overexpression knockdown
Sensitivity upon overexpression
COL1A1
In vitro and in vivo
[96]
miR-152
Overexpression
Sensitivity
DNMT1
In vitro and in vivo
[97]
miR-185
Overexpression
Sensitivity
DNMT1
In vitro and in vivo
[97]
miR-199b-5p
Overexpression
Sensitivity
JAG1
In vitro and in vivo
[98]
miR-449a
Overexpression
Sensitivity
NOTCH1
In vitro and in vivo
[99]
miR-484
Overexpression
Sensitivity
VEGFB VEGFR2
In vitro and in vivo
[100]
miR-9
Overexpression knockdown
Sensitivity upon overexpression
BRCA1
In vitro
[101]
miR-21
Knockdown
Sensitivity
PDCD4
In vitro
[102]
miR-21
Overexpression knockdown
Sensitivity upon knockdown
PDCD4
In vitro
[103]
miR-30c-2–3p Overexpression knockdown
Sensitivity upon overexpression
BCL9
In vitro
[104]
miR-30d
Overexpression
Sensitivity
ABCD2
In vitro
[105]
miR-93
Overexpression and knockdown
Resistance upon overexpression
PTEN
In vitro
[106]
miR-107
Overexpression
Sensitivity
RAD51 RAD51 D
In vitro
[107]
miR-103
Overexpression
Sensitivity
RAD51 RAD51 D
In vitro
[107]
miR-106a
Overexpression and knockdown
Sensitivity upon overexpression
Mcl-1
In vitro
[108]
miR-106a
Overexpression and knockdown
Sensitivity upon knockdown
PDCD4
In vitro
[108]
miR-125b
Overexpression and knockdown
Resistance upon overexpression
Bak1
In vitro
[109]
miR-130a
Overexpression and knockdown
Resistance upon overexpression
PTEN
In vitro
[110]
miR-130a
Overexpression
Sensitivity
XIAP
In vitro
[111]
miR-130b
Overexpression and knockdown
Sensitivity upon overexpression
CSF-1
In vitro
[112]
miR-141
Overexpression
Resistance
KEAP1
In vitro
[113]
miR-193b-3p
Overexpression
Resistance
CRIM1
In vitro
[114]
miR-199a
Overexpression
Sensitivity
CD44
In vitro
[115]
miR-199a
Overexpression and knockdown
Sensitivity upon overexpression
IKKb
In vitro
[116]
miR-199a
Overexpression knockdown
Sensitivity upon overexpression
mTOR
In vitro
[117]
miR-214
Overexpression knockdown
Resistance upon overexpression
PTEN
In vitro
[118]
miR-302b
Overexpression
Sensitivity
HDAC4
In vitro
[119]
miR-376c
Overexpression
Resistance
ALK7
In vitro
[120]
miR-429
Overexpression
Sensitivity
Not tested
In vitro
[121]
miR-489
Overexpression knockdown
Sensitivity upon overexpression
Akt3
In vitro
[122]
miR-519d
Overexpression knockdown
Sensitivity upon overexpression
XIAP
In vitro
[123]
could potentially support the application of circulating miRNAs in early diagnosis of EOC events, Zheng et al. reported that combination of miR-205 and let-7f expression in plasma samples of EOC patients provided
future science group
Ref.
high diagnostic accuracy for stage I disease in EOC [51] . More interestingly, the accuracy of this miRNAbased prediction increased when combined with CA-125 levels suggesting that circulating miRNAs can
www.futuremedicine.com
245
Review Nagaraj, Joseph & DiFeo complement CA-125 as biomarkers in EOC. This is very promising since the less invasive nature of circulating miRNA analysis could make them an important integral component of early diagnostic screening platforms in EOC and also could enhance the accuracy of CA-125-based prediction of the disease. Exosomes are an important source of circulating miRNAs and could constitute important tools in miRNA-based diagnostic modalities in cancers including EOC because exosomes are enriched source of miRNAs in circulation. In one of the first reports characterizing exosomal miRNAs in EOC, Taylor et al. conducted miRNA expression profiling of exosomes isolated from serum samples of highgrade serous ovarian cancer patients and compared them with expression profile of tumor cell-derived miRNAs from the same patients [52] . Some miRNAs were unique to the exosomes and tumor cells, but the global miRNA expression profile of exosomes was remarkably similar to the cell-derived miRNAs suggesting that exosomes faithfully recapitulate the disease process. Improvised protocols for isolation and enrichment of exosomes from serum samples are available now and hence this field is rapidly emerging as a diagnostic tool in cancers including EOC [53] . However, a very recent report by Chevillet et al. has shown that quantitative analysis of miRNAs in exosomes needs better models of evaluation and standard protocols and analysis methods currently in practice may not efficiently reflect miRNA biology associated with exosomes. Therefore, further studies are necessary to delineate efficient methods of exploring exosome-associated miRNAs [54] . miRNAs as therapeutic tools in EOC Since miRNA deregulation is a characteristic feature of cancers, this presents with a miRNA-based cancer therapeutics possibility to correct this deregulation either by replacing the downregulated tumor suppressor miRNAs using miRNA mimics (miRNA replacement therapy) or by inhibiting the upregulated oncomiRs using antisense miRs (miRNA inhibition therapy) [55,56] . Such miRNA-based therapeutics approach has the potential to have drastic effects on the tumor phenotype because of the fact that a single miRNA can regulate hundreds of targets and multiple targets can be regulated by a single miRNA, but at the same time this therapeutic approach would be pathway specific too. Since cells already express these miRNAs, exogenous delivery of miRNAs potentially would not result in adverse effects. Also, since miRNA deregulation drives tumor phenotype, correcting this deregulation would specifically affect cancer cells while potentially sparing normal cells. Hence,
246
Biomark. Med. (2015) 9(3)
miRNA modulation is emerging as an exciting cancer therapeutics option. The first and the only ‘anticancer miRNA drug’ MRX34 that is a miR-34 miRNA mimic has entered Phase I clinical trials in patients with advanced hepatocellular carcinoma heralding the entry of miRNAs into cancer therapeutics in clinical settings. Interestingly, miR-34 family of miRNAs was found to be frequently downregulated in EOC and this downregulation was more evident in EOC patients with mutations in TP53 and was associated with metastatic clinical stage [57] . In the same study, it was further shown that miR-34 replacement therapy in the metastatic SKOV3 ovarian tumor cells significantly decreased invasion, migration and proliferation. Even though there is currently no miRNA drug in clinical settings in EOC, MRX34 could provide with an exciting possibility of clinical translation in EOC patients in future. miRNA modulation as a tool to enhance efficacy of chemotherapy in EOC
Numerous studies in preclinical models over the last few years have established that miRNA modulation is an exciting approach to target EOC mechanisms and improve chemosensitivity. Some of the important studies in this regard are summarized in Table 1. One of the major exciting observations in miRNA therapeutics is the ability of miRNAs to determine the efficacy of drugs [58] . Figure 2 illustrates some of the possible mechanisms by which miRNAs can improve the efficacy of chemotherapy and overcome c hemotherapy resistance. Combination of kinase inhibitors & miRNA replacement therapy
A classic example of this case would be the tumor suppressive miRNA, miR-34a, which is known to directly target EGFR [59] . The combination of miR-34a and the EGFR inhibitor Erlotinib exerted synergistic effect in hepatocellular carcinoma cells, which were resistant to Erlotinib alone [60] . miR-34a decreased the IC50 of Erlotinib by 46-fold; the reason behind this dramatic effect being miR-34a negatively targets multiple oncogenes known to confer resistance to Erlotinib. EGFR signaling pathway is known to be an important regulator of EOC phenotype; monoclonal antibodies targeting EGFR and small-molecule inhibitors of EGFR are in clinical trials in ovarian cancer but with limited success outcome [61] . This leads to an interesting speculation that miR-34 replacement therapy could greatly improve the efficacy of EGFR inhibitors in EOC, especially since the initial clinical trials of these drugs in EOC were met with limited success. EOC patients with low miR-34a expression along with altered EGFR
future science group
miRNAs as prognostic & therapeutic tools in epithelial ovarian cancer
pathway components could be the subset that could greatly benefit from such combination studies. Apart from the exciting possibility of combining with drugs, miRNAs are also reported to enhance siRNA-based therapeutic approach. Tumor suppressor miRNAs can be combined with siRNAs targeting oncogenes and such combination therapies could have additional beneficial effects as compared with miRNAs or siRNAs employed alone. One such combination was described in EOC by Nishimura et al., wherein the tumor suppressor miRNA miR-520d-3p exhibited synergistic effect with siRNA against the oncogene EphA2 [62] . This advantage conferred by miR-520d-3p addition was found to be due to the transcriptional repression both EphA2 and EphB2 by miR-520d-3p. This further highlights the possibility that the unique nature of miRNAs, i.e ability to target multiple genes could have great application potential in designing miRNA-based combination therapeutic approaches either with drugs or siRNAs. Combination of platinum-based compounds with tumor suppressor miRNA mimics or antisense oligonucleotides against oncomiRs
A variety of mechanisms have been implicated in emergence of resistance to platinum-based compounds [63] . Some of the important constituents in this aspect are decreased drug accumulation due to upregulated ABC transporter family efflux pumps; upregulated DNA repair enzymes and upregulated antiapoptotic proteins that decrease cell death and potentially confer survival advantage to tumor cells in response to platinum-based compounds [14] . Supplementing platinum-based therapy with miRNA mimics targeting these drug resistance regulators or with antisense oligonucleotides that inhibit miRNAs promoting drug resistance could greatly improve the efficiency of cell death induced by platinum compounds. A couple of possibilities can be discussed in this context. The tumor suppressor miR-34 family that is frequently downregulated in EOC is known to promote apoptosis in cancer cells by directly targeting a variety of antiapoptotic genes [64] and hence it can be speculated that MRX34 addition could potentially improve the efficacy of platinum-based therapy in EOC. Similarly, miR-181a that is upregulated in chemoresistant EOC tumors [43] is reported to exert antiapoptotic effects by targeting the proapoptotic protein Bim [65] and Bim downregulation is known to contribute to cisplatin resistance in ovarian cancer [66] . Hence, antisense oligonucleotides inhibiting miR-181a could potentially improve the efficacy of platinum-based drugs by enhancing apoptotic cell death in response to cisplatin.
future science group
Review
Combining platinum therapy with miRNAs targeting tumor-initiating cells in EOC
Emerging evidence supports the concept that platinum-based therapies are able to eliminate the bulk of differentiated cancer cells but are unable to eliminate tumor-initiating cells (TICs; also termed cancer stem cells) that are resistant to cytotoxic therapies [16,17,67] . These rare populations of TICs are characterized by their ability to self-renew indefinitely in an undifferentiated state and also result in differentiated progeny of cells with high proliferative and invasive capacity, thus driving the expansion of drug resistant tumors. Various stem cell markers have been reported to enrich for ovarian TICs and ALDH is the most convincing marker reported till date in this regard [68] . CD133 in combination with ALDH is reported to further increase the enrichment of TIC population in EOC [69] . The proven inability of platinum compounds to eradicate TICs in EOC provides an opportunity for miRNAs targeting TICs in EOC to be used in combination with cisplatin to eradicate TICs and overcome chemoresistance. Only very few miRNAs have been implicated to date in regulation of TICs in EOC [70] . One of the first miRNAs to be identified in this context was miR-214. miR-214 knockdown was shown to increase sensitivity to cisplatin and doxorubicin in ALDH-positive cells isolated from ovarian tumor cell lines [71] . Park et al. analyzed the miRNA expression profile of ALDH-positive chemoresistant SKOV3 ovarian tumor cells and found that miR-23b, miR-27a, miR-27b, miR-346, miR424 and miR-503 were overexpressed in ALDH-positive cells and these miRNAs were also upregulated in chemoresistant EOC tumors as compared with the chemosensitive group [72] . These reports suggest that modulation of miRNAs could be an exciting prospect in TIC-based therapeutic strategies in EOC in coming days. Predicting novel miRNAs that could improve drug efficacy in EOC
The observation that miRNAs can determine the efficacy of drugs has given rise to the field termed as ‘miRNA pharmacogenomics’ and these miRNAs are termed as ‘pharmaco-miRs’ [58] . As discussed earlier in Figure 2, the basis of this effect is that targeting miRNAs that regulate genes/pathways that are known to be involved in drug resistance/inactivation mechanisms could enhance drug efficacy by overcoming the ‘roadblocks’ affecting the drug action. Pharmaco-miRs provide with an exciting window of opportunity in overcoming chemoresistance in EOCs. Since platinum-based therapy is the mainstay of EOC treatment and most of the patients initially respond very well
www.futuremedicine.com
247
Review Nagaraj, Joseph & DiFeo
A Mutations
Kinase inhibitors or Phosphatase activators
Kinase
Mutations
Kinase
Kinase inhibitors or Phosphatase activators
Multiple targets in the pathway downregulated Chemo sensitivity
Down + regulation
+ Drug resistance Tumor suppressor microRNA mimics B
microRNA mimics Anti cancer drugs DNA-repair and anti-apoptotic proteins
+++ oncomiRs
Cell death Drug resistance
microRNA mimics
Efflux pumps
oncomiRs
Anti-sense miRs
++++ Cell death Chemo sensitivity
C Differentiated
EOC
TICs
p Tumor relapse TICs survive Cisplatin alone
Tum mor regression reg gressio Tumor Eradicates TICs Cisplatin + miRs t geting TICs tar targeting
Figure 2. Illustration of potential scenarios in which microRNAs could function as adjuvants to chemotherapy and enhance chemosensitivity. (A) Mutations in kinases could confer resistance to kinase inhibitors or phosphatase activators. Direct targeting of these kinases by microRNAs could overcome this resistance and enhance the efficacy in combination with these drugs. (B) Drug efflux pumps and mutations in DNA repair/anti-apoptotic proteins confer chemoresistance and hence microRNAs directly targeting these could confer chemosensitivity. (C) TICs may survive after platinum-based chemotherapy and constitute tumor recurrence and chemoresistance. Hence, microRNAs that directly target TICs could form an effective combination with cisplatin in eradicating TICs. TIC: Tumor-initiating cell.
to these drugs, employing pharmaco-miRs in combination with cisplatin could increase the efficiency of platinum-based therapy and more importantly could prevent the emergence of chemoresistance. A novel prediction database is available for identifying and characterizing potential pharmaco-miRs in the context of the drug of interest (http://www. pharmaco-mir.org/) [58] . Figure 3 lists one such prediction of potential pharmaco-miRs that can modulate cisplatin efficacy. Using this database, miRNAs that are known to be associated with targets that can affect cisplatin function can be identified and these miRNAs can be selected for further functional validation as tools to enhance cisplatin effectiveness. The miRNA association listed in this database includes both predicted miRNA binding sites in the 3’UTR of the targets and also experimentally verified targets (by 3’UTR reporter assays). As illustrated in Figure 3, several targets that are known to modulate cisplatin
248
Biomark. Med. (2015) 9(3)
function and form important components of cisplatin resistance. For example, Pharmaco-miR associates the tumor suppressor miR-200c, which is commonly deregulated in EOCs with ABCB1 (MDR1) and very recently it has been shown that miR-200c antagonizes MDR1-dependent drug resistance in cancer cells [73] . These data show that pharmaco-miR prediction can reliably identify miRNAs that could modulate drug efficacy. miRNA modulation to target metastasis in EOC
At time of EOC diagnosis, the majority of patients present with extensive metastatic dissemination and malignant ascites, which usually correlates with poor survival. Hence, targeting metastatic ovarian tumor cells is critical in improving patient response to chemotherapy and to prolong survival. Metastatic process is thought to be initiated by epithelial-mesenchymal transition (EMT) where in epithelial tumor cells acquire
future science group
miRNAs as prognostic & therapeutic tools in epithelial ovarian cancer
mesenchymal features enabling them to migrate, invade and metastasize to distant organs [74,75] . miRNAs are important regulators of EMT in cancers [76] . The tumor suppressor miR-200 family that is one of the most commonly deregulated miRNAs in EOC and was one of the very first identified inhibitors of EMT process by directly targeting ZEB1, ZEB2 and vimentin, which are positive regulators of EMT [70,77,78] . miR-125a and miR-138 were later shown to inhibit EMT in ovarian cancer cells [79,80] . Furthermore, a comprehensive clustering analyses of 459 serous EOC samples identified a miRNA regulatory network, which consisted of miRNAs that were downregulated and inhibited EMT, correlated with the more aggressive mesenchymal ovarian cancer phenotype. Specifically, they found that miR-506 was an EMT inhibitor and positive prognostic predictor in ovarian cancer by targeting SNAI2 [38] . Enhanced expression of miR506 was found to upregulate E-cadherin, suppress EMT and reduced cancer cell proliferation, migration and invasion in EOC. Clearly, these reports identify a potential role for tumor suppressor miRNA mimics in targeting metastasis by inhibiting EMT processes. In the first report identifying miRNAs as promoters of EMT and metastasis in EOC, our lab showed that miR-181a is a critical regulator of metastasis in EOC by promoting EMT transition via activation of TGF-β signaling pathway [43] . We found that miR181a directly targets SMAD7, the critical inhibitor of TGF-β signaling pathway and promotes EMT transition in epithelial ovarian tumor cells and also miR181a inhibition decreased survival and migration in metastatic ovarian tumor cells. Our study suggests that antisense oligonucleotides targeting miR-181a could be efficient tools in targeting metastatic cells and improve survival in EOC. miRNA modulation targeting tumor microenvironment in EOC
Tumor microenvironment is an integral component of tumor heterogeneity, which greatly complicates diagnostic and therapeutic modalities [81] . This is one of the active areas of research in cancers including EOC, which will form one of the most important arms of EOC translational research aimed at overcoming chemoresistance [82] . miRNAs are known to play important functional roles in tumor microenvironment and hence miRNA modulation is emerging as an exciting tool to target tumor microenvironment in order to overcome chemoresistance [83] . Defining tumor microenvironment in EOC is more complicated as compared with other tumors because of the fact that the origin of EOC is greatly heterogeneous and identifying the site of origin of EOC is still a matter of intensive research.
future science group
Review
However, one of the components of tumor microenvironment, namely cancer-associated fibroblasts (CAFs) has been linked to EOC pathogenesis and miRNAs have been identified as regulators of these components of the microenvironment. In the first study reporting the regulation of CAFs in EOC by miRNAs, Mitra et al. identified three miRNAs as necessary and sufficient to reprogram normal fibroblasts into CAFs [84] . miR-214 and miR-31 were downregulated and miR-155 was upregulated in CAFs as compared with normal fibroblasts. Accordingly, miRNA modulation mimicking this deregulation-converted normal fibroblasts to CAFs and vice versa. This study highlights the importance of employing miRNAs as tools to target microenvironment; however, further studies are required to understand how miRNAs regulate other components of tumor stroma in EOC. Efficient miRNA delivery methods are important for success of miRNA therapeutics in EOC miRNA delivery methods in vivo are the most practical determinants or roadblocks of realizing miRNA transition into clinical settings because delivering small RNA molecules into the site of action in the human body bypassing all the tissue barriers is easier said than done. This concept is extensively gaining importance currently since this is the major obstacle in miRNA therapeutics. Systemic administration of miRNA mimics (miRNA replacement therapy) and antisense miRNA oligonucleotides (miRNA inhibition therapy) in vivo aimed at targeting tumor mechanisms could be a challenging task. A variety of miRNA modification methodologies are being tested in preclinical models for their stability and tissue-specific effects in vivo [55,85] . MRX34, which is the anticancer miRNA drug in clinical trial, is a liposome-based method of delivering miRNA in vivo [55] . Yang et al. reported nanoliposome-based method of delivering miR-506 intraperitoneally in EOC xenograft models and hence liposome-based miRNA delivery approaches could be important in EOC [38] . It is not surprising that the two miRNAs in clinical trials currently (miR-34 [MRX34] for hepatocellular carcinoma and miR-122 [miravirsen] for HCV infection) [55] are targeted against hepatic pathology. The first major barrier any therapeutic molecule has to successfully overcome after systemic administration is the hepatic circulation and this may not be a major problem in targeting hepatic pathology because accumulation of the ‘therapeutic miRNAs’ at the site of action is maximal in this scenario since most of the systemically administered miRNA molecules are known to be taken up by liver and kidney [55] .
www.futuremedicine.com
249
Review Nagaraj, Joseph & DiFeo
Cisplatin resistance-associated targets ABC transporters DNA repair ABCB1
miR-222 miR-221 miR-200c miR-27a miR-331-5p miR-451
ABCC1
miR-217 miR-133b miR-133a miR-145 miR-873 miR-199a-5p miR-199b-5p miR-185 miR-7 miR-326 miR-330-5p miR-186 miR-224 miR-345
ERCC4
miR-544 let-7g miR-98 let-7c let-7a let-7i let-7b let-7e let-7d let-7f miR-202
miR-520h miR-520g miR-93 miR-519d miR-20b miR-20a miR-106b miR-106a miR-17 miR-205 miR-361-5p miR-200c miR-429 miR-200b miR-302e miR-302d miR-302b miR-302a miR-302c miR-372 miR-520a-3p
Angiogenesis
Apoptosis
VEGFA
miR-29b miR-520e miR-29a miR-373 miR-520c-3p miR-15b miR-520b miR-497 miR-424 miR-520d-3p miR-16 miR-378 miR-195 miR-203 miR-140-5p miR-15a miR-543 miR-613 miR-503 miR-1 miR-410 miR-206 miR-377 miR-199a-5p miR-199b-5p miR-299-3p miR-150 miR-185 miR-34b miR-186 miR-504 miR-125a-5p miR-134 miR-34a miR-126 miR-330-3p miR-383 miR-147 miR-107 miR-29c
XIAP
MCL1
miR-193a-3p miR-193b miR-582-5p miR-181a miR-181c miR-181d miR-181b miR-133b miR-133a miR-93 miR-519d miR-20b miR-20a miR-106b miR-106a miR-17 miR-153 miR-661 miR-892b miR-302e miR-302d
miR-302b miR-302a miR-302c miR-372 miR-520a-3p miR-520e miR-373 miR-520c-3p miR-520b miR-520d-3p miR-367 miR-363 miR-32 miR-92a miR-92b miR-25 miR-135a miR-135b miR-101 miR-125b miR-125a-5p
miR-302a miR-518a-5p miR-29c miR-29b miR-29a miR-16 miR-15a miR-876-3p miR-512-5p miR-886-3p miR-148b* miR-148* miR-193a miR-518 miR-582
miR-96 miR-181a miR-340 miR-181c miR-181d miR-23a miR-181b miR-23b miR-93 miR-7 miR-186 miR-519d miR-320b miR-20b miR-320c miR-20a miR-106b miR-320d miR-106a miR-320a miR-17 miR-219-5p miR-495 miR-136 miR-421 miR-200c miR-216a miR-429 miR-410 miR-200b miR-377 miR-27b miR-27a miR-494 miR-374a miR-371-5p miR-374b miR-1271
microRNAs associated with cisplatin resistance-associated targets
Figure 3. Pharmaco-miR database based prediction of microRNAs that could modulate the efficacy of cisplatin-based chemotherapy. ABC transporters (ABCB1, ABCC1), DNA repair components (ERCC4), angiogenesis regulators (VEGFA) and apoptosis regulators (MCL1, XIAP) constitute some of the important components of cisplatin resistance in EOC and hence microRNAs directly targeting these drug resistance regulators could be employed as effective tools in overcoming cisplatin resistance in EOC. Important microRNAs deregulated in EOCs are highlighted.
Delivering miRNA therapeutic molecules targeting EOC cells is a great challenge since the site of action is very distant from systemic administration as compared with hepatic tissue and also the miRNA molecules have to survive in the circulation long enough to reach the destination in sufficient concentration. Hence, intraperitoneal administration of miRNA therapeutic molecules could be an improvised way of targeted delivery especially in EOC since intraperitoneal administration of platinum-based compounds is often employed in EOC chemotherapy [86] . Importantly, this method could be of specific importance in targeting metastatic cells in EOC since EOC rarely disseminates outside of the abdominal cavity. Thus, it would be interesting to speculate that intraperitoneal administration of tumor suppressor miR-200 mimics and antisense oligonucleotides against miR-181a could emerge as important tools in miRNA-based EOC therapy by targeting metastatic ovarian tumor cells. Since the tumor suppressor family miR-34 is frequently downregulated
250
Biomark. Med. (2015) 9(3)
in EOC, MRX34 could have potential implications too in miRNA-based EOC chemotherapy in future. Nanoparticle-based intraperitoneal delivery of therapeutic molecules specifically targeted at ovarian tumor cells have been reported recently [87] . Combination of miRNAs with such nanoparticle-based ovarian tumor cell-targeted therapy administered intraperitoneally could greatly improve the application potential of miRNAs in EOC therapeutics. Current challenges & future directions of miRNA applications in EOC clinical settings The first miRNA anticancer drug MRX34 entered clinical trial approximately a decade after the discovery that miRNAs are deregulated in cancers. This is an impressive time period for a basic science discovery to translate into therapeutics in clinical settings considering that these are small RNA molecules that offer tremendous hope for cancer therapeutics in coming times. However, the dream of miRNA therapeutics becoming
future science group
miRNAs as prognostic & therapeutic tools in epithelial ovarian cancer
realistic in cancer treatment still has to overcome various obstacles and this is more so in EOC considering that EOC is greatly heterogeneous both in terms of origin and composition of tumors and still is the most lethal gynecological malignancy. Some important perspectives in this regard can be summarized as below: miRNAs & early diagnosis of EOC pathogenesis
The role of miRNAs in early transformation events in EOC pathogenesis remains largely unexplored even though it is known the miRNAs regulate ovarian physiology. Reliable biomarkers for diagnosis of EOC pathogenesis could greatly improve patient outcome in EOC because chemotherapy intervention at early stage is highly successful with low recurrence. This is one important scenario where miRNAs could dramatically improve patient outcome in EOC. To date, there is lack of knockin or knockout miRNA mouse models specific for ovarian or fallopian tube tissue. These kinds of models are crucial in understanding the role of miRNAs in early transformation events in EOC. The utility of these models can be greatly enhanced in combination with the recently reported BRCA-TP53-PTEN targeting EOC mouse model that can be used to study the effects of miRNA modulation on early EOC events. Candidate miRNA knockin or knockout in the background of this BRCA-TP53-PTEN model can be tested to identify miRNAs that aggravate or reduce the occurrence of tumors in these mice and thus could be selected for diagnostic and therapeutic considerations. miRNAs & intratumoral heterogeneity in EOC
Expression analysis of different EOC cohorts has reported different miRNA signatures in EOCs, thus complicating the identification of common miRNA signatures across EOC patients to enable translation into clinical settings. Even though these kinds of variations are generally attributed to the differences in the methodologies employed and nature of cohorts studied, tumor geneity appears to play an important role in these conditions. Intratumoral heterogeneity presents one of the greatest challenges in clinical translation of novel diagnostic and therapeutic modalities that are successful in preclinical models [88,89] . This could further complicate the identification of common miRNA prognostic and therapeutic signatures across EOC patients since intratumoral heterogeneity is beginning to be identified in cancer patients [90] . This also leads to the interesting possibility of developing patientspecific miRNA signatures that will enable patient stratification and individualized miRNA therapeutics approach (Figure 4) . This kind of targeted miRNA prognostic and therapeutic approach is more likely to be successful in clinical settings and could offer
future science group
Review
maximal utility toward improving patient outcome in EOC. One of the greatest challenges in understanding intratumoral heterogeneity in tumors is the selection of right kind of samples that need to be used for analysis. Classical approach has been acquiring biopsies from multiple tumor sites in patients but this technique relies more on rules of probability and chance than specificity and hence, the accuracy of these samples in reflecting true heterogeneity can sometimes be questionable. Circulating tumor cells (CTCs) are increasingly gaining importance as potential reflectors of intratumoral heterogeneity [91] . Since CTCs shed by multiple tumor lesions in a patient enter a common streamline in the circulation, they are more likely to offer a dynamic view of disease pathogenesis in real time as compared with tumor biopsies that provide a more static outlook of the disease. CTC identification in ovarian cancers has not been very successful and the results have been disappointing due to lot of technical limitations that make sample range insufficient for analysis [92] . This provides a great opportunity for circulating miRNAs in EOCs to be employed as biomarkers to develop individualized therapeutic approaches. Since miRNAs that are secreted from multiple tumor lesions in the same patient are channeled into a common pool in circulation, these miRNAs can provide a reliable outlook of EOC pathogenesis events in real-time specific for each patient. Since isolation and analysis of circulating miRNAs in EOC patients are very well established, these circulating miRNAs could greatly revolutionize individualized prognostic and therapeutic approaches in EOCs in future. miRNAs as translational activators
The classic outlook of miRNA functioning has been that miRNAs negatively regulate gene expression mainly by posttranslational mechanisms. In a breakthrough finding few years ago, Vasudevan et al. reported that miRNAs could also function as translational activators [93] and since then concept of regulation of gene expression by miRNAs has taken a new look. Recently, one such scenario was identified in cancer cells suggesting translational activation by miRNAs could be important regulators of tumor phenotype [94] . It is very likely that all these years, important miRNAs could have been missed out being identified as regulators of tumorigenesis since miRNA-cancer studies are most commonly based on the outlook of negative regulation of gene expression by miRNAs. Even though this phenomenon is yet to be identified in EOCs, miRNAs that function as translation activators could play an important role in translation of miRNA application in EOC clinical settings in future.
www.futuremedicine.com
251
Review Nagaraj, Joseph & DiFeo
EOC tumor samples (biopsies, serum etc.)
Predicted good survival microRNA expression profiling
(potential chemo sensitive)
Standard chemotherapy
Predicted poor survival (potential chemoresistant) Patient stratification
Targeted therapy personalized
microRNA inhibition therapy
microRNA replacement therapy
Tumors with upregulated oncomiRs
Tumors with downregulated tumor suppressor miRs
miR-A miR-B Anti-sense miRs + standard chemotherapy
miR-C miR-D miR-E microRNA mimics + standard chemotherapy
Figure 4. A futuristic outlook of microRNA application in EOC diagnostics and therapeutics. microRNA signatures could be employed to predict patient survival and plan relevant treatment strategies in EOC clinical settings. microRNA signatures could form an integral component of targeted therapeutic approaches in EOC by enabling reliable patient stratification. microRNA inhibition therapy aimed at downregulating onco-miRs and microRNA replacement therapy aimed at upregulating tumor suppressor microRNAs could constitute future EOC therapeutics aimed at eradicating EOC.
To summarize, the discovery of miRNAs and the subsequent finding that they are deregulated in cancers a decade ago ushered in astronomical hopes that these small RNA molecules could be the magic bullets that mankind was in search of to eradicate cancer and the review of the progress in this regard in the last decade has been promising. If MRX34 shows favorable results in the ongoing clinical trial it could start a new era of miRNA cancer therapeutics and these tiny RNAs could greatly strengthen our fight in eradicating cancer.
in way of realistic application of miRNAs in oncology clinics on a daily basis. The hurdles to overcome are tough if not impossible. These tiny RNA molecules could go a long way in revolutionizing cancer diagnostics and therapeutics in coming years. Personalized therapeutic strategies could well form the basis of cancer diagnostics and therapeutics in the future and miRNAs will have a significant role to play in realizing this possibility. Financial & competing interests disclosure
Future perspective miRNAs have made a significant leap into translational research settings within a decade of being discovered to be deregulated in cancers. A lot of enthusiasm revolves around the success of MRX34, which is the miRNA currently in clinical trial for the treatment of cancer. miRNA and cancer research field has exploded considerably over the last few years and the main focus of research is now to overcome the challenges that stand
252
Biomark. Med. (2015) 9(3)
The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties. No writing assistance was utilized in the production of this manuscript.
future science group
miRNAs as prognostic & therapeutic tools in epithelial ovarian cancer
Review
Executive summary Unique nature of gene expression regulation by miRNAs • miRNAs are small RNAs that regulate cell proliferation and differentiation by associating with their targets in a sequence-specific manner and have been identified as critical regulators of tumorigenesis mechanisms.
miRNA expression deregulation in cancers • miRNA expression profiling and miRNA modulation strategies are emerging as promising entities in cancer diagnostics and therapeutics. • The Cancer Genome Atlas (TCGA) provided a comprehensive analysis of high-grade serous EOCs by integrated analyses of various parameters including miRNA expression profiles and identified four molecular subtypes of ovarian cancer and miRNA expression clustering in this analysis revealed three subtypes with subtype 1 being associated with poor survival.
miRNA functioning in ovarian physiology & tumorigenesis processes • miRNAs are known to play an important role in regulating ovarian physiology and hence it is not surprising that miRNAs have been implicated in regulating EOC mechanisms. • miRNA deregulation is a characteristic feature of chemotherapy resistance and metastasis in ovarian tumors.
Circulating miRNAs as biomarkers • miRNAs are present in body fluids and can be detected in serum samples, which are one of the least invasive samples available for biomarker analysis and hence, circulating miRNAs are increasingly gaining importance as biomarkers in various cancers including EOCs. • Circulating miRNAs can provide a reliable outlook of EOC pathogenesis events in real-time specific for each patient and hence could revolutionize individualized therapy approach in EOC in future.
Application of miRNAs as therapeutic tools in EOC • The two main components of miRNA-based cancer therapeutics are replacing the downregulated tumor suppressor miRNAs using miRNA mimics (miRNA replacement therapy) or by inhibiting the upregulated oncomiRs using antisense miRs (miRNA inhibition therapy). • miRNA modulation can enhance efficacy of chemotherapy and also target metastatic cells in EOC. • miRNA delivery methods in vivo are the most practical determinants or roadblocks of realizing miRNA transition into clinical settings. • MRX34, which is the anticancer miRNA drug in clinical trial, is a liposome-based method of delivering miRNA in vivo. • Nanoliposome-based method of delivering miR-506 intraperitoneally in EOC xenograft models is reported to inhibit metastasis.
miRNAs & early diagnosis of EOC pathogenesis • The role of miRNAs in early transformation events in EOC pathogenesis is an important area to be investigated and miRNA knockin and knockout mouse models are critical requisites in this context.
miRNA in clinical trial • The progress of miRNA cancer research over the last decade is promising and miRNAs could be well set to enter clinical settings and there are great hopes about the success of MRX34 in clinical trial.
References
7
Papers of special note have been highlighted as: • of interest; •• of considerable interest 1
Siegel R, Ma J, Zou Z, Jemal A. Cancer statistics, 2014. CA-Cancer J. Clin. 64(1), 9–29 (2014).
2
Cooke SL, Brenton JD. Evolution of platinum resistance in high-grade serous ovarian cancer. Lancet Oncol. 12(12), 1169–1174 (2011).
3
He L, Hannon GJ. MicroRNAs: small RNAs with a big role in gene regulation. Nat. Rev. Genet. 5(7), 522–531 (2004).
4
Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116(2), 281–297 (2004).
5
Kim VN. MicroRNA biogenesis: coordinated cropping and dicing. Nat. Rev. Mol. Cell Biol. 6(5), 376–385 (2005).
6
Calin GA, Croce CM. MicroRNA signatures in human cancers. Nat. Rev. Cancer 6(11), 857–866 (2006).
future science group
Iorio MV, Visone R, Di Leva G et al. MicroRNA signatures in human ovarian cancer. Cancer Res. 67(18), 8699–8707 (2007).
•
First report showing miRNA deregulation in ovarian cancer.
8
Mezzanzanica D, Bagnoli M, De Cecco L, Valeri B, Canevari S. Role of microRNAs in ovarian cancer pathogenesis and potential clinical implications. Int. J. Biochem. Cell B. 42(8), 1262–1272 (2010).
9
Pencheva N, Tavazoie SF. Control of metastatic progression by microRNA regulatory networks. Nat. Cell Biol. 15(6), 546–554 (2013).
10
Sun X, Jiao X, Pestell TG et al. MicroRNAs and cancer stem cells: the sword and the shield. Oncogene 33(42), 4967–4977 (2014).
11
Garofalo M, Croce CM. MicroRNAs as therapeutic targets in chemoresistance. Drug Resist. Updat. 16(3–5), 47–59 (2013).
www.futuremedicine.com
253
Review Nagaraj, Joseph & DiFeo 12
Van Jaarsveld MT, Helleman J, Berns EM, Wiemer EA. MicroRNAs in ovarian cancer biology and therapy resistance. Int. J. Biochem. Cell B. 42(8), 1282–1290 (2010).
28
Donadeu FX, Schauer SN, Sontakke SD. Involvement of miRNAs in ovarian follicular and luteal development. J. Endocrinol. 215(3), 323–334 (2012).
13
Frederick PJ, Green HN, Huang JS et al. Chemoresistance in ovarian cancer linked to expression of microRNAs. Biotech Histochem. 88(7), 403–409 (2013).
•
Detailed review highlighting functional roles of miRs in ovarian physiology.
29
14
Shen DW, Pouliot LM, Hall MD, Gottesman MM. Cisplatin resistance: a cellular self-defense mechanism resulting from multiple epigenetic and genetic changes. Pharmacol. Rev. 64(3), 706–721 (2012).
Liu Z, Liu J, Segura MF et al. MiR-182 overexpression in tumourigenesis of high-grade serous ovarian carcinoma. J. Pathol. 228(2), 204–215 (2012).
30
Perets R, Wyant GA, Muto KW et al. Transformation of the fallopian tube secretory epithelium leads to high-grade serous ovarian cancer in Brca;Tp53;Pten models. Cancer Cell 24(6), 751–765 (2013).
••
Generation of efficient genetic model of high-grade serous ovarian cancer pathogenesis. Important for identifying regulators of early events in EOC pathogenesis.
31
Cancer Genome Atlas Research Network. Integrated genomic analyses of ovarian carcinoma. Nature 474(7353), 609–615 (2011).
••
The most comprehensive genetic profiling analysis of ovarian tumors.
32
Krell J, Frampton AE, Colombo T et al. The p53 miRNA interactome and its potential role in the cancer clinic. Epigenomics 5(4), 417–428 (2013).
33
Lee CH, Subramanian S, Beck AH et al. MicroRNA profiling of BRCA1/2 mutation-carrying and non-mutationcarrying high-grade serous carcinomas of ovary. PLoS ONE 4(10), e7314 (2009).
34
Creighton CJ, Fountain MD, Yu Z et al. Molecular profiling uncovers a p53-associated role for microRNA-31 in inhibiting the proliferation of serous ovarian carcinomas and other cancers. Cancer Res. 70(5), 1906–1915 (2010).
35
Hu X, Macdonald DM, Huettner PC et al. A miR-200 microRNA cluster as prognostic marker in advanced ovarian cancer. Gynecol. Oncol. 114(3), 457–464 (2009).
36
Nam EJ, Yoon H, Kim SW et al. MicroRNA expression profiles in serous ovarian carcinoma. Clin. Cancer Res. 14(9), 2690–2695 (2008).
37
Kim TH, Kim YK, Kwon Y et al. Deregulation of miR519a, 153, and 485–5p and its clinicopathological relevance in ovarian epithelial tumours. Histopathol. 57(5), 734–743 (2010).
38
Yang D, Sun Y, Hu L et al. Integrated analyses identify a master microRNA regulatory network for the mesenchymal subtype in serous ovarian cancer. Cancer Cell 23(2), 186–199 (2013).
39
Eitan R, Kushnir M, Lithwick-Yanai G et al. Tumor microRNA expression patterns associated with resistance to platinum based chemotherapy and survival in ovarian cancer patients. Gynecol. Oncol. 114(2), 253–259 (2009).
15
Cooke SL, Ng CK, Melnyk N et al. Genomic analysis of genetic heterogeneity and evolution in high-grade serous ovarian carcinoma. Oncogene 29(35), 4905–4913 (2010).
16
Latifi A, Abubaker K, Castrechini N et al. Cisplatin treatment of primary and metastatic epithelial ovarian carcinomas generates residual cells with mesenchymal stem cell-like profile. J Cell. Biochem. 112(10), 2850–2864 (2011).
17
Abubaker K, Latifi A, Luwor R et al. Short-term single treatment of chemotherapy results in the enrichment of ovarian cancer stem cell-like cells leading to an increased tumor burden. Mol. Cancer 12, 24 (2013).
18
Weigelt B, Reis-Filho JS, Swanton C. Genomic analyses to select patients for adjuvant chemotherapy: trials and tribulations. Ann. Oncol. 23(Suppl. 10), x211–x218 (2012).
19
Banerjee S, Gore M. The future of targeted therapies in ovarian cancer. Oncologist 14(7), 706–716 (2009).
20
Gupta D, Lis CG. Role of CA125 in predicting ovarian cancer survival: a review of the epidemiological literature. J. Ovarian Res. 2, 13 (2009).
21
Felder M, Kapur A, Gonzalez-Bosquet J et al. MUC16 (CA125): tumor biomarker to cancer therapy, a work in progress. Mol. Cancer 13, 129 (2014).
22
Lu J, Getz G, Miska EA et al. MicroRNA expression profiles classify human cancers. Nature 435(7043), 834–838 (2005).
23
Xi Y, Nakajima G, Gavin E et al. Systematic analysis of microRNA expression of RNA extracted from fresh frozen and formalin-fixed paraffin-embedded samples. RNA 13(10), 1668–1674 (2007).
24
254
Mitchell PS, Parkin RK, Kroh EM et al. Circulating microRNAs as stable blood-based markers for cancer detection. Proc. Natl Acad. Sci. USA 105(30), 10513–10518 (2008).
•
One of the early reports describing detection of miRNAs in circulation in cancer patients. Gave a significant boost to miRNA application as diagnostic tools.
25
Llaurado M, Majem B, Altadill T et al. MicroRNAs as prognostic markers in ovarian cancer. Mol. Cell. Endocrinol. 390(1–2), 73–84 (2014).
26
Pritchard CC, Cheng HH, Tewari M. MicroRNA profiling: approaches and considerations. Nat. Rev. Genet. 13(5), 358–369 (2012).
40
Sorrentino A, Liu CG, Addario A, Peschle C, Scambia G, Ferlini C. Role of microRNAs in drug-resistant ovarian cancer cells. Gynecol. Oncol. 111(3), 478–486 (2008).
27
Baker M. MicroRNA profiling: separating signal from noise. Nat. Methods 7(9), 687–692 (2010).
41
•
Comprehensive review of techniques that can enable miRNA diagnostic application in clinical settings.
Yang N, Kaur S, Volinia S et al. MicroRNA microarray identifies Let-7i as a novel biomarker and therapeutic target in human epithelial ovarian cancer. Cancer Res. 68(24), 10307–10314 (2008).
Biomark. Med. (2015) 9(3)
future science group
miRNAs as prognostic & therapeutic tools in epithelial ovarian cancer
42
Bagnoli M, De Cecco L, Granata A et al. Identification of a chrXq27.3 microRNA cluster associated with early relapse in advanced stage ovarian cancer patients. Oncotarget 2(12), 1265–1278 (2011).
43
Parikh A, Lee C, Peronne J et al. microRNA-181a has a critical role in ovarian cancer progression through the regulation of the epithelial–mesenchymal transition. Nat. Comms. 5, 2977 (2014).
44
Schwarzenbach H, Nishida N, Calin GA, Pantel K. Clinical relevance of circulating cell-free microRNAs in cancer. Nat. Rev. Clin. Oncol. 11(3), 145–156 (2014).
45
Maclellan SA, Macaulay C, Lam S, Garnis C. Pre-profiling factors influencing serum microRNA levels. BMC Clin. Pathol. 14, 27 (2014).
46
Resnick KE, Alder H, Hagan JP, Richardson DL, Croce CM, Cohn DE. The detection of differentially expressed microRNAs from the serum of ovarian cancer patients using a novel real-time PCR platform. Gynecol. Oncol. 112(1), 55–59 (2009).
47
Kan CW, Hahn MA, Gard GB et al. Elevated levels of circulating microRNA-200 family members correlate with serous epithelial ovarian cancer. BMC Cancer 12, 627 (2012).
48
Hong F, Li Y, Xu Y, Zhu L. Prognostic significance of serum microRNA-221 expression in human epithelial ovarian cancer. J. Int. Med. Res. 41(1), 64–71 (2013).
49
Xu YZ, Xi QH, Ge WL, Zhang XQ. Identification of serum microRNA-21 as a biomarker for early detection and prognosis in human epithelial ovarian cancer. Asian Pac. J. Cancer Prev. 14(2), 1057–1060 (2013).
50
Guo F, Tian J, Lin Y, Jin Y, Wang L, Cui M. Serum microRNA-92 expression in patients with ovarian epithelial carcinoma. J. Int. Med. Res. 41(5), 1456–1461 (2013).
51
Zheng H, Zhang L, Zhao Y et al. Plasma miRNAs as diagnostic and prognostic biomarkers for ovarian cancer. PLoS ONE 8(11), e77853 (2013).
52
Taylor DD, Gercel-Taylor C. MicroRNA signatures of tumor-derived exosomes as diagnostic biomarkers of ovarian cancer. Gynecol. Oncol. 110(1), 13–21 (2008).
53
Moret I, Sanchez-Izquierdo D, Iborra M et al. Assessing an improved protocol for plasma microRNA extraction. PLoS ONE 8(12), e82753 (2013).
54
Chevillet JR, Kang Q, Ruf IK et al. Quantitative and stoichiometric analysis of the microRNA content of exosomes. Proc. Natl Acad. Sci. USA 111(41), 14888–14893 (2014).
55
Li Z, Rana TM. Therapeutic targeting of microRNAs: current status and future challenges. Nat. Rev. Drug Discov. 13(8), 622–638 (2014).
56
Nana-Sinkam SP, Croce CM. MicroRNA dysregulation in cancer: opportunities for the development of microRNAbased drugs. IDrugs 13(12), 843–846 (2010).
57
Corney DC, Hwang CI, Matoso A et al. Frequent downregulation of miR-34 family in human ovarian cancers. Clin. Cancer Res. 16(4), 1119–1128 (2010).
58
Rukov JL, Wilentzik R, Jaffe I, Vinther J, Shomron N. Pharmaco-miR: linking microRNAs and drug effects. Brief. Bioinf. 15(4), 648–659 (2014).
future science group
59
Yin D, Ogawa S, Kawamata N et al. miR-34a functions as a tumor suppressor modulating EGFR in glioblastoma multiforme. Oncogene 32(9), 1155–1163 (2013).
60
Zhao J, Kelnar K, Bader AG. In-depth analysis shows synergy between erlotinib and miR-34a. PLoS ONE 9(2), e89105 (2014).
•
Describes the synergy between miR-34 and erlotinib. Greatly widens the application potential of miRs as combination therapies with drugs.
61
Siwak DR, Carey M, Hennessy BT et al. Targeting the epidermal growth factor receptor in epithelial ovarian cancer: current knowledge and future challenges. J. Oncol. 2010, 568938 (2010).
62
Nishimura M, Jung EJ, Shah MY et al. Therapeutic synergy between microRNA and siRNA in ovarian cancer treatment. Cancer Discov. 3(11), 1302–1315 (2013).
63
Galluzzi L, Senovilla L, Vitale I et al. Molecular mechanisms of cisplatin resistance. Oncogene 31(15), 1869–1883 (2012).
64
Hermeking H. The miR-34 family in cancer and apoptosis. Cell Death Differ. 17(2), 193–199 (2010).
65
Taylor MA, Sossey-Alaoui K, Thompson CL, Danielpour D, Schiemann WP. TGF-beta upregulates miR-181a expression to promote breast cancer metastasis. J. Clin. Invest. 123(1), 150–163 (2013).
66
Wang J, Zhou JY, Wu GS. Bim protein degradation contributes to cisplatin resistance. J. Biol. Chem. 286(25), 22384–22392 (2011).
67
Steg AD, Bevis KS, Katre AA et al. Stem cell pathways contribute to clinical chemoresistance in ovarian cancer. Clin. Cancer Res. 18(3), 869–881 (2012).
68
Deng S, Yang X, Lassus H et al. Distinct expression levels and patterns of stem cell marker, aldehyde dehydrogenase isoform 1 (ALDH1), in human epithelial cancers. PLoS ONE 5(4), e10277 (2010).
69
Silva IA, Bai S, Mclean K et al. Aldehyde dehydrogenase in combination with CD133 defines angiogenic ovarian cancer stem cells that portend poor patient survival. Cancer Res. 71(11), 3991–4001 (2011).
70
Wang Y, Kim S, Kim IM. Regulation of metastasis by microRNAs in ovarian cancer. Front. Oncol. 4, 143 (2014).
71
Xu CX, Xu M, Tan L et al. MicroRNA miR-214 regulates ovarian cancer cell stemness by targeting p53/Nanog. J. Biol. Chem. 287(42), 34970–34978 (2012).
72
Park YT, Jeong JY, Lee MJ et al. MicroRNAs overexpressed in ovarian ALDH1-positive cells are associated with chemoresistance. J. Ovarian Res. 6(1), 18 (2013).
73
Sui H, Cai GX, Pan SF et al. miR-200c attenuates P-gp mediated MDR and metastasis by targeting JNK2/cJun signaling pathway in colorectal cancer. Mol. Cancer Ther.13(12), 3137–3151 (2014).
74
Lengyel E. Ovarian cancer development and metastasis. Am. J. Pathol. 177(3), 1053–1064 (2010).
75
Lamouille S, Xu J, Derynck R. Molecular mechanisms of epithelial-mesenchymal transition. Nat. Rev. Mol. Cell Biol. 15(3), 178–196 (2014).
www.futuremedicine.com
Review
255
Review Nagaraj, Joseph & DiFeo 76
Zhang J, Ma L. MicroRNA control of epithelialmesenchymal transition and metastasis. Cancer Mestat. Rev 31(3–4), 653–662 (2012).
77
Park SM, Gaur AB, Lengyel E, Peter ME. The miR-200 family determines the epithelial phenotype of cancer cells by targeting the E-cadherin repressors ZEB1 and ZEB2. Genes Dev. 22(7), 894–907 (2008).
78
79
Cowden Dahl KD, Dahl R, Kruichak JN, Hudson LG. The epidermal growth factor receptor responsive miR-125a represses mesenchymal morphology in ovarian cancer cells. Neoplasia 11(11), 1208–1215 (2009).
80
Yeh YM, Chuang CM, Chao KC, Wang LH. MicroRNA-138 suppresses ovarian cancer cell invasion and metastasis by targeting SOX4 and HIF-1alpha. Int. J. Cancer 133(4), 867–878 (2013).
81
Junttila MR, De Sauvage FJ. Influence of tumour microenvironment heterogeneity on therapeutic response. Nature 501(7467), 346–354 (2013).
82
Musrap N, Diamandis EP. Revisiting the complexity of the ovarian cancer microenvironment – clinical implications for treatment strategies. Mol. Cancer Res. 10(10), 1254–1264 (2012).
83
Suzuki HI, Katsura A, Matsuyama H, Miyazono K. MicroRNA regulons in tumor microenvironment. Oncogene doi:10.1038/onc.2014.254 (2014) (Epub ahead of print).
Liu JF, Kindelberger D, Doyle C, Lowe A, Barry WT, Matulonis UA. Predictive value of circulating tumor cells (CTCs) in newly-diagnosed and recurrent ovarian cancer patients. Gynecol. Oncol. 131(2), 352–356 (2013).
93
Vasudevan S, Tong Y, Steitz JA. Switching from repression to activation: microRNAs can up-regulate translation. Science 318(5858), 1931–1934 (2007).
••
Breakthrough finding that miRs can function as translational activators.
94
Liu M, Roth A, Yu M et al. The IGF2 intronic miR-483 selectively enhances transcription from IGF2 fetal promoters and enhances tumorigenesis. Genes Dev. 27(23), 2543–2548 (2013).
95
Cai J, Yang C, Yang Q et al. Deregulation of let-7e in epithelial ovarian cancer promotes the development of resistance to cisplatin. Oncogenesis 2, e75 (2013).
96
Yu PN, Yan MD, Lai HC et al. Downregulation of miR-29 contributes to cisplatin resistance of ovarian cancer cells. Int. J. Cancer 134(3), 542–551 (2014).
97
Xiang Y, Ma N, Wang D et al. MiR-152 and miR-185 co-contribute to ovarian cancer cells cisplatin sensitivity by targeting DNMT1 directly: a novel epigenetic therapy independent of decitabine. Oncogene 33(3), 378–386 (2014).
98
Liu MX, Siu MK, Liu SS, Yam JW, Ngan HY, Chan DW. Epigenetic silencing of microRNA-199b-5p is associated with acquired chemoresistance via activation of JAG1-Notch1 signaling in ovarian cancer. Oncotarget 5(4), 944–958 (2014).
99
Zhou Y, Chen Q, Qin R, Zhang K, Li H. MicroRNA449a reduces cell survival and enhances cisplatin-induced cytotoxicity via downregulation of NOTCH1 in ovarian cancer cells. Tumour Biol. 35(12), 12369–12378 (2014).
84
Mitra AK, Zillhardt M, Hua Y et al. MicroRNAs reprogram normal fibroblasts into cancer-associated fibroblasts in ovarian cancer. Cancer Discov. 2(12), 1100–1108 (2012).
85
Ling H, Fabbri M, Calin GA. MicroRNAs and other noncoding RNAs as targets for anticancer drug development. Nat. Rev. Drug Discov. 12(11), 847–865 (2013).
86
Armstrong DK, Bundy B, Wenzel L et al. Intraperitoneal cisplatin and paclitaxel in ovarian cancer. N. Engl. J. Med. 354(1), 34–43 (2006).
101 Sun C, Li N, Yang Z et al. miR-9 regulation of BRCA1 and
Zhang Y, Kenny HA, Swindell EP et al. Urokinase plasminogen activator system-targeted delivery of nanobins as a novel ovarian cancer therapy. Mol. Cancer Ther. 12(12), 2628–2639 (2013).
102 Chan JK, Blansit K, Kiet T et al. The inhibition of miR-21
87
88
Bedard PL, Hansen AR, Ratain MJ, Siu LL. Tumour heterogeneity in the clinic. Nature 501(7467), 355–364 (2013).
89
Bashashati A, Ha G, Tone A et al. Distinct evolutionary trajectories of primary high-grade serous ovarian cancers revealed through spatial mutational profiling. J. Pathol. 231(1), 21–34 (2013).
90
Raychaudhuri M, Schuster T, Buchner T et al. Intratumoral heterogeneity of microRNA expression in breast cancer. J. Mol. Diagn. 14(4), 376–384 (2012).
91
256
Chen D, Zhang Y, Wang J et al. MicroRNA-200c overexpression inhibits tumorigenicity and metastasis of CD117+ CD44 + ovarian cancer stem cells by regulating epithelial-mesenchymal transition. J. Ovarian Res. 6(1), 50 (2013).
92
Onstenk W, Gratama JW, Foekens JA, Sleijfer S. Towards a personalized breast cancer treatment approach guided by circulating tumor cell (CTC) characteristics. Cancer Treat. Rev. 39(7), 691–700 (2013).
Biomark. Med. (2015) 9(3)
100 Vecchione A, Belletti B, Lovat F et al. A microRNA
signature defines chemoresistance in ovarian cancer through modulation of angiogenesis. Proc. Natl Acad. Sci. USA 110(24), 9845–9850 (2013). ovarian cancer sensitivity to cisplatin and PARP inhibition. J. Natl Cancer Inst. 105(22), 1750–1758 (2013). promotes apoptosis and chemosensitivity in ovarian cancer. Gynecol. Oncol. 132(3), 739–744 (2014). 103 Liu SG, Fang Y, Shen HL, Xu WL, Li H. Berberine sensitizes
ovarian cancer cells to cisplatin through miR-21/PDCD4 axis. Acta Bioch. Bioph. Sin. 45(9), 756–762 (2013). 104 Jia W, Eneh JO, Ratnaparkhe S, Altman MK, Murph
MM. MicroRNA-30c-2* expressed in ovarian cancer cells suppresses growth factor-induced cellular proliferation and downregulates the oncogene BCL9. Mol. Cancer Res. 9(12), 1732–1745 (2011). 105 Lacroix B, Gamazon ER, Lenkala D et al. Integrative
analyses of genetic variation, epigenetic regulation, and the transcriptome to elucidate the biology of platinum sensitivity. BMC Genomics 15, 292 (2014). 106 Fu X, Tian J, Zhang L, Chen Y, Hao Q. Involvement of
microRNA-93, a new regulator of PTEN/Akt signaling pathway, in regulation of chemotherapeutic drug cisplatin
future science group
miRNAs as prognostic & therapeutic tools in epithelial ovarian cancer
chemosensitivity in ovarian cancer cells. FEBS Lett. 586(9), 1279–1286 (2012). 107 Huang JW, Wang YM, Dhillon KK et al. Systematic screen
identifies miRNAs that target RAD51 and RAD51D to enhance chemosensitivity. Mol. Cancer Res. 11(12), 1564–1573 (2013). 108 Rao YM, Shi HR, Ji M, Chen CH. MiR-106a targets Mcl-1
to suppress cisplatin resistance of ovarian cancer A2780 cells. J. Huazhong Univ. Sci. Technol. Med. Sci. 33(4), 567–572 (2013). 109 Kong F, Sun C, Wang Z et al. miR-125b confers resistance
of ovarian cancer cells to cisplatin by targeting pro-apoptotic Bcl-2 antagonist killer 1. J. Huazhong Univ. Sci. Technol. Med. Sci. 31(4), 543–549 (2011). 110 Li NW, Wang HJ, Yang LY, Jia XB, Chen C, Wang X.
[Regulatory effects and associated mechanisms of miR-130a molecules on cisplatin resistance in ovarian cancer A2780 cell lines]. Sichuan Da Xue Xue Bao Yi Xue Ban 44(6), 865–870 (2013). 111 Zhang X, Huang L, Zhao Y, Tan W. Downregulation of
miR-130a contributes to cisplatin resistance in ovarian cancer cells by targeting X-linked inhibitor of apoptosis (XIAP) directly. Acta Biochim. Biophys. Sin. (Shanghai) 45(12), 995–1001 (2013). 112 Yang C, Cai J, Wang Q et al. Epigenetic silencing of
miR-130b in ovarian cancer promotes the development of multidrug resistance by targeting colony-stimulating factor 1. Gynecol. Oncol. 124(2), 325–334 (2012). 113 Van Jaarsveld MT, Helleman J, Boersma AW et al. miR-141
regulates KEAP1 and modulates cisplatin sensitivity in ovarian cancer cells. Oncogene 32(36), 4284–4293 (2013). 114 Ziliak D, Gamazon ER, Lacroix B, Im HK, Wen YJ,
Huang RS. Genetic variation that predicts platinum sensitivity reveals the role of miR-193b* in chemotherapeutic susceptibility. Mol. Cancer Ther. 11(9), 2054–2061 (2012).
future science group
Review
115 Cheng W, Liu T, Wan X, Gao Y, Wang H. MicroRNA-199a
targets CD44 to suppress the tumorigenicity and multidrug resistance of ovarian cancer-initiating cells. FEBS J. 279(11), 2047–2059 (2012). 116 Chen R, Alvero AB, Silasi DA et al. Regulation of IKKbeta
by miR-199a affects NF-kappaB activity in ovarian cancer cells. Oncogene 27(34), 4712–4723 (2008). 117 Wang ZX, Ting Z, Li Y, Chen G, Lu YP, Hao X. microRNA-
199a is able to reverse cisplatin resistance in human ovarian cancer cells through the inhibition of mammalian target of rapamycin. Oncol. Lett. 6(3), 789–794 (2013). 118 Yang H, Kong W, He L et al. MicroRNA expression profiling
in human ovarian cancer: miR-214 induces cell survival and cisplatin resistance by targeting PTEN. Cancer Res. 68(2), 425–433 (2008). 119 De Cecco L, Berardi M, Sommariva M et al. Increased
sensitivity to chemotherapy induced by CpG-ODN treatment is mediated by microRNA modulation. PLoS ONE 8(3), e58849 (2013). 120 Ye G, Fu G, Cui S et al. MicroRNA 376c enhances ovarian
cancer cell survival by targeting activin receptor-like kinase 7: implications for chemoresistance. J. Cell Sci. 124(Pt 3), 359–368 (2011). 121 Wang L, Mezencev R, Svajdler M, Benigno BB,
Mcdonald JF. Ectopic over-expression of miR-429 induces mesenchymal-to-epithelial transition (MET) and increased drug sensitivity in metastasizing ovarian cancer cells. Gynecol. Oncol. 134(1), 96–103 (2014). 122 Wu H, Xiao Z, Zhang H, Wang K, Liu W, Hao Q. MiR-489
modulates cisplatin resistance in human ovarian cancer cells by targeting Akt3. Anti-Cancer Drugs 25(7), 799–809 (2014). 123 Pang YX, Mao HL, Shen L, Zhao Z, Liu RH, Liu PS. MiR-
519d represses ovarian cancer cell proliferation and enhances cisplatin-mediated cytotoxicity in vitro by targeting XIAP. Oncotargets Ther. 7, 587–597 (2014).
www.futuremedicine.com
257