Skip to content
Publicly Available Published by De Gruyter January 9, 2016

miRNAs as novel biomarkers in the management of prostate cancer

  • Xavier Filella EMAIL logo and Laura Foj

Abstract

microRNAs (miRNAs) are small non-coding RNAs that control gene expression posttranscriptionally and are part of the giant non codifying genoma. Cumulating data suggest that miRNAs are promising potential biomarkers for many diseases, including cancer. Prostate cancer (PCa) detection is currently based in the serum prostate-specific antigen biomarker and digital rectal examination. However, these methods are limited by a low predictive value and the adverse consequences associated with overdiagnosis and overtreatment. New biomarkers that could be used for PCa detection and prognosis are still needed. Recent studies have demonstrated that aberrant expressions of microRNAs are associated with the underlying mechanisms of PCa. This review attempts to extensively summarize the current knowledge of miRNA expression patterns, as well as their targets and involvement in PCa pathogenesis. We focused our review in the value of circulating and urine miRNAs as biomarkers in PCa patients, highlighting the existing discrepancies between different studies, probably associated with the important methodological issues related to their quantitation and normalization. The majority of studies have been performed in serum or plasma, but urine obtained after prostate massage appears as a new way to explore the usefulness of miRNAs. Large screening studies to select a miRNA profile have been completed, but bioinformatics tools appear as a new approach to select miRNAs that are relevant in PCa development. Promising preliminary results were published concerning miR-141, miR-375 and miR-21, but larger and prospective studies using standardized methodology are necessary to define the value of miRNAs in the detection and prognosis of PCa.

Introduction

New biomarkers have been described during recent years for the management of early prostate cancer (PCa), among them the Prostate Health Index, PCA3 score and the four-kallikrein panel [1–3]. Emerging biomarkers should improve the specificity of prostate specific antigen (PSA) in the detection of PCa, showing a relationship with the aggressiveness of the tumor. Currently, PSA remains the most used biomarker in the detection of PCa despite the controversies around its use as a screening tool. Actually, findings show that the screening based on PSA levels results in a higher incidence of low risk PCa, most of which do require no treatment.

Overdiagnosis and overtreatment has become a major problem in the management of PCa. Because of this fact, active surveillance (AS) has been proposed to palliate these negative effects, delaying any kind of definitive treatment [4]. The selection of patients for AS combines clinical stage, PSA serum levels, Gleason score, the number of positive prostate biopsies and the amount of malignant tissue per core. However, even when these criteria are strictly used, inaccuracies are found [5]. Novel tools are necessary to improve patients selection for AS.

Cumulating data suggest that microRNAs (miRNAs) can be used as potential biomarkers for the detection of PCa, as well as in the selection of patients for AS due to their relationship with the aggressiveness of the tumor. The aim of this paper is to present a systematic review about the role of miRNAs as biomarkers in PCa.

Biogenesis and function of miRNAs

The involvement of protein coding genes in the development of cancer has been extensively studied in recent years. Although 90% of the human genome is transcribed to messenger RNA (mRNA), only 2% encodes for proteins, so there is a large number of RNAs that encodes for no protein. Recent data suggest that this portion of the genome that does not encode for protein also plays a role in carcinogenesis. These non-coding RNAs include small nucleolar RNA, miRNA, small interfering RNA, piwi-interacting RNA and long non-coding RNA [6], which for example includes PTENP1, a pseudogene of the tumor suppressor PTEN, and PCA3, proposed as a useful biomarker in the detection of PCa [7, 8].

miRNAs are small (17–22 nucleotides) single-stranded non-coding RNAs, that negatively regulate the gene expression through the binding to their corresponding mRNA targets. The first miRNA described was lin-4, identified in 1993 by Lee et al. [9] in the nematode Caenorhabditis elegans. Currently, according to the database mirbase.org, 28,645 miRNAs have been identified in different species (miRBase, release 21: June 2014) [10], of which 2588 mature miRNAs have been recognized in humans.

Biogenesis of miRNAs (Figure 1) is a complex process that begins in the cell nucleus with the formation of the double-stranded primary miRNA (pri-miRNA), by the action of RNA polymerase II. Then, the pri-miRNAs are processed by a microprocessor that contains the Drosha RNAse III enzyme and the DGCR8 protein, and allows the formation of the precursor miRNA (pre-miRNA). After that, the pre-miRNA is transferred to the cytoplasm by exportin 5 and Ran GTP cofactor, where the pre-miRNA is processed by Dicer RNAse, forming a double-stranded RNA of approximately 22 nucleotides, which contains two strands of miRNA: one becomes the mature miRNA and the other one the passenger miRNA or 5p miRNA, which is normally degraded. The mature miRNA binds to the Argonaut protein (AGO) and forms the RNA-induced silencing complex (RISC). After being incorporated into the RISC complex, miRNAs can exercise the silencing function on their corresponding mRNA targets.

Figure 1: Schematic representation of miRNA biosynthesis.
Figure 1:

Schematic representation of miRNA biosynthesis.

Extracellular miRNAs are heterogeneous, including miRNAs packaged in microparticles (exosomes, microvesicles and apoptotic bodies) and miRNAs linked to RNA binding proteins (AGO-2, nucleophosmin-1) and to high-density lipoproteins to prevent their degradation. Apoptotic bodies are the largest microparticles (0.5–2 μm) and are produced from cells during apoptosis. Microvesicles are heterogeneous in size (0.1–1 μm) and they are released from all cell types by different mechanisms. Finally, exosomes are small vesicles (50–100 nm) that originate from the endosome. Results are confusing about the proportion between exosomal and non-exosomal miRNAs. Arroyo et al. [11], using differential centrifugation and size-exclusion chromatography to characterize circulating miRNA complexes in human plasma and serum, concluded that the vesicle associated miRNAs represents the minority, whereas around 90% of miRNAs in the circulation is present in a non-membrane-bound form. Additionally, Dijkstra et al. [12] compared PCA3 and TMPRSS2-ERG expression levels in urinary sediments and exosomes and concluded that in exosomes these biomarkers had lower analytical sensitivity, although exosomes seem to be a more robust source of urinary biomarker. On the other hand, according to Gallo et al. [13] exosome isolation improves the sensitivity of miRNA from human biological fluids, including serum and saliva. Moreover, Cheng et al. [14] showed that in urine the highest proportion of miRNA was extracted from exosomes compared with the cell pellet and cell free supernatant.

Advances in technology together with the stability of miRNAs in different kind of samples [15] have allowed to identify the role of miRNAs in several diseases [16]. Specific miRNAs profiles of different cancers can be identified using microarrays. qRT-PCR has been generally used to validate the miRNAs selected by microarrays. The development of digital PCR will improve the performance of qRT-PCR, without the use of endogenous or exogenous controls to normalize the results. In contrast, digital PCR is a direct method for quantifying nucleic acids, particularly useful for samples with low abundant miRNAs, and showing a high degree of sensitivity and precision compared to qRT-PCR [17, 18]. Finally, next generation sequencing (NGS) platforms, such as those from Solexa/Illumina and 454 Life Sciences/Roche, have been developed to find novel miRNAs, generating a complete expression profile, distinguishing sequentially similar miRNAs and identifying point mutations.

miRNAs and cancer

In 2002, Calin et al. [19] demonstrated that a miRNA cluster was frequently deleted or downregulated in chronic lymphocytic leukemia. Subsequently, several miRNAs expression and functional studies showed that miRNAs may be associated in cancer initiation and progression. Abnormal expression of miRNAs in cancer is related to different mechanisms, including genomic mutations, epigenetic changes, chromosomal abnormalities and alterations in miRNA biogenesis, i.e. the altered expression of the miRNA processing endoribonuclease Dicer [20].

Currently, different miRNAs have been involved in human carcinogenesis by acting as “oncomirs” or “tumor suppressive miRNAs” [16], inhibiting suppressor tumoral genes or activating oncogenes, respectively. The study of aberrant miRNA expression profiles in cancer is, in fact, an innovative way to identify new biomarkers for cancer detection and prognosis. An exponential growth of studies has been published since the first circulating miRNA profile was reported by Lawrie et al. [21] in 2008 as a diagnostic tool in oncology. The discovery of miRNAs in other body fluids, including urine, saliva and bronchial lavage, has intensified investigations about the use of miRNAs as non-invasive biomarkers in patients with cancer [15].

Interestingly, according to available results, miRNAs signatures are able to classify human cancer origin [22]. The highly tissue specific origin of miRNAs leads to an efficient identification of metastatic cancers of unknown primary origin. Søkilde et al. [23] reported in a series of 208 tumors, representing 15 different histologies including PCa, that miRNA expression profiling on paraffin tissue can efficiently predict the primary origin of the tumor. The authors developed a new miRNAs classification algorithm, obtaining an overall accuracy of 85% (CI: 79%–89%). When the algorithm was applied to an independent test set of 48 metastases, the primary site was correctly identified in 42 cases (88% accuracy; CI, 75%–94%). Additional studies are necessary to confirm this point.

Preanalytical and analytical variables in miRNA measurement

Several preanalytical factors can influence the levels of miRNAs, including procedures of sample collection, specimen acquisition, handling and storage [24]. Differences in circulating miRNAs levels have been observed between plasma and serum. Wang et al. [25] described higher levels of miRNAs in serum than in plasma, suggesting that additional miRNAs would be produced during the coagulation process. Also, the different kind of anticoagulants used can influence the results. Thus, Kim et al. [26] observed that the levels of miR-16 and miR-223 are lower when heparin is used and higher with oxalate, while EDTA provides similar results to serum. Hemolysis is also a factor that affects the analysis of some miRNAs, including miR-16, miR-451, miR-92a and miR-486-5p that are released by erythrocytes [27, 28]. Recommendations to analyze miRNAs in plasma include platelet removal, while serum separator tubes are recommended to work with serum. Hemolysis can be assessed by visual inspection or using a spectrophotometer at 540 nm. Furthermore, it can be checked measuring the ratio between miR-451 and miR-23a, a highly expressed and stable serum miRNA. A ratio higher than five is an indicative of possible hemolysis contamination, while a ratio higher than eight indicates a high risk of hemolysis affecting thus the obtained data [29].

Time between sample extraction and the aliquotation of plasma or serum also affects results, although according to the majority of authors, miRNAs are stable biomarkers. Mitchell et al. [30] reported that miRNA levels remain stable when plasma is subjected to prolonged room temperature incubation or freeze-thawed multiple times. Besides, McDonald et al. [31] showed that miRNAs were stable at room temperature for 24 h and refrigerated or frozen for up to 72 h. However, Rice et al. [32], obtaining plasma at different time points, showed that time to plasma extraction influences ΔCt values in 5 of 11 miRNAs tested. Based on these results, the authors recommend plasma extraction before 12 h.

There are also differences in the results obtained in relation to the method used for isolating total RNA from different body fluids (mirVana, Exiqon miRCURY, miRNeasy, TRIzol) [14, 33–35]. Unfortunately, results published in these studies do not always support similar conclusions. According to McAlexander et al. [33] Exiqon miRCURY Biofluids Kit outperforms other RNA isolation methods tested working with plasma, including Exiqon Cell and Plant kit, TRIzol, mirVana and miRNeasy. In contrast, Kroh et al. [34] reported that Qiagen miRNeasy kit produces 2–3 fold greater RNA than mirVana kit. On the other hand, Cheng et al. [14] compared six commercially available kits for total RNA extraction from the cell pellets, cell-free urine, and exosomes from urine, including miRNeasy, miRNeasy with RNeasy MinElute Cleanup Kit, mirVana, TRIzol reagent with mirVana, miRCURY, and Urine Exosome RNA Isolation kit. miRNeasy kit using the additional RNeasy MinElute was the most efficient reagent to isolate miRNA from exosomes and cell-free urine, according to these authors, while miRCURY was the most efficient in cell pellets.

Evaluation of miRNA requires normalization to minimize data variation that can mask or exaggerate biologically meaningful changes. Variation in the measurement of miRNAs among experiments can be attributed to different sources, such as the RNA isolation, the quantity and quality of RNA, or the efficiency of the reverse transcription and qRT-PCR. Normalization of results is essential to obtain homogeneous results between experiments and convert raw data into valid results. Different strategies have been described to normalize miRNA expression levels, including the normalization to the average Cq value of all measured miRNA assays, the normalization to an endogenous miRNA, and the normalization to an external spike-in oligoRNA added during the RNA extraction. The normalization to the average Cq value, also called the mean centering method, is restricted to studies which assay a large number of miRNAs [36]. One variation of this method is the mean centering restricted method, which has been developed for experiments where a substantial fraction of miRNA data values is missing, using the mean obtained of only the miRNAs expressed across all samples [37].

Several protocols for the normalization using endogenous miRNAs controls have been implemented. The ideal normalizer should be independent of biological variation, disease stage or treatments, showing similar storage stability, extraction properties and quantification efficiency compared to the miRNA target. Stable small non-coding RNAs are often used as reference genes to quantify cellular miRNAs, for instance, the small nucleolar RNAs, like SNORD43, SNORD44, SNORD48 and Mamm U6. Also, some miRNAs have been used as reference genes, like let-7a-1 or miR-130b. Several algorithms, particularly geNorm and NormFinder software, can be used to select the most stable reference material from a list of candidates. GeNorm and NormFinder software are application tools for Microsoft Excel that offer the stability value based on the intra- and intergroup expression variations of the reference genes studied. A low combined intra- and intergroup variation proves high expression stability. Nonetheless, this ideal normalizer, particularly working with plasma/serum or other body fluids, does not exist [38] and the use of endogenous reference genes is controversial.

Actually, there are no validated reference controls for the analysis of the expression of miRNAs in PCa, showing the published studies contradictory results. Sanders et al. [39] proposed SNORD43 as a suitable reference gene for the analysis of circulating miRNAs in patients with urological malignancies, reporting a high stability, although the stability of RNU6-2, RNU1-4, miR-106a, let-7a and SNORD48 was also remarkably high according to these authors. On the other hand, Schaefer et al. [40] recommended the use of miR-130b or the geometric mean of miR-130b and RNU6-2 to normalize miRNA expression in PCa tissues studies. However, according to Carlsson et al. [41], RNU24 and RNU44 would be suitable control genes for miRNA expression studies in prostate tissues, but not miR-130b, because its stability was not appropriate to be a normalizer control.

Quantifying miRNAs in plasma/serum or other body fluids using endogenous controls is particularly difficult since invariant reference control has not been found. The use of a spike-in exogenous control appears as a solution to this problem [42]. The addition of an exogenous synthetic miRNA (e.g. the derived C. elegans cel-miR-39, cel-miR-54 and cel-miR-238, or the Arabidopsis thaliana ath-miR-159a) in the phenolic phase allows to monitorize the efficiency of the reverse transcription and qRT-PCR reactions. Mitchell et al. [30] proposed the combined use of cel-miR-39, cel-miR-54 and cel-miR-238 to quantify circulating miRNAs in plasma. However, Sourvinou et al. [43] concluded that results improve when normalization is based on combined exogenous (cel-miR-39) and endogenous (miR-16) miRNAs controls, because differences in miRNA recovery and differences in cDNA synthesis between samples are compensated. In contrast, McDonald et al. [31] showed that imprecision was higher using the endogenous control miR-16 than using exogenous cel-miR-39, because miR-16 is susceptible to hemolysis. According to these authors, results did not improve when the exogenous controls cel-miR-54 and cel-miR-238 to cel-miR-39 were added. Exogenous miRNAs controls, such as ath-miR-159a, cel-miR-54, cel-miR-238 and particularly cel-miR-39, appear as the optimal way to normalize miRNAs expression in absence of an ideal endogenous control [44, 45], although biological variability should be taken into account to improve miRNAs measurement.

Role of miRNAs in prostate cancer

Altered miRNA expression in PCa has been reported by different authors, showing their deregulation in several processes, including cell proliferation, differentiation and progression [46–48]. The miRNAs involved in PCa pathogenesis and their signaling pathways are shown in Figure 2. Deregulation of the cell cycle leads to aberrant cell proliferation that characterizes cancer. Usually, miRNAs interact with cell cycle proteins, conferring thus the potential to regulate cellular division and cell cycle progression. For example, Lewis et al. [49] reported that miR-888 inhibits the target retinoblastoma-like 1, which blocks first gap phase to synthetic phase cell cycle progression by binding and inhibiting the E2F transcription factors.

Figure 2: Role of miRNAs in PCa pathogenesis.
Figure 2:

Role of miRNAs in PCa pathogenesis.

Cancer stem cells (CSC) are a subset of cancer cell subpopulations in the tumor mass and play an important role in tumor progression and metastasis in several types of cancers, including PCa. PCa stem cells keep the self-renewal and differentiation properties and can lead to more cancer cells. miRNAs are involved in promoting or inhibiting the stemness of CSCs. Aberrantly expressed miRNAs can cause deregulation of specific signaling pathways that are associated with the functions of CSCs. Particularly, miR-34 family or let-7 family [50, 51] have been involved in the regulation of PCa CSC properties.

miRNAs have been also implicated in the resistance to apoptosis in PCa through different mechanisms. miR-21 is usually upregulated in PCa and directly targets PTEN, whose downregulation results in significantly reduced apoptosis in PCa. miR-21 also regulates the myristoylated alanine-rich protein kinase c substrate (MARCKS), conferring apoptosis resistance and leading to aberrant proliferation [52, 53] (Figure 3). Another mechanism of regulating apoptosis is through targeting B-cell lymphoma 2 (Bcl-2) family members; for instance some miR-34 family members or the tumor suppressor miR-205, which directly target Bcl-2 in PCa [54, 55].

Figure 3: miR-21, miR-141, miR-143, miR-145, miR-221 and miR-222 in PCa pathogenesis: summary of their roles and interactions with targets.
Figure 3:

miR-21, miR-141, miR-143, miR-145, miR-221 and miR-222 in PCa pathogenesis: summary of their roles and interactions with targets.

The epithelial-mesenchymal transition (EMT) is a biologic process that allows a polarized epithelial cell to submit to multiple biochemical changes to assume a mesenchymal cell phenotype, which confers enhanced migratory capacity, invasiveness and stem cell properties [56]. PCa cells are subjected to EMT, then invade surrounding tissues and colonize several tissues via blood or lymphatic routes to generate metastases. Metastatic cells can then revert through mesenchymal-epithelial transition (MET) to reacquire epithelial characteristics similar to cells in the primary tumor. Some miRNAs are regulators of EMT and MET events through the regulation of the expression of key proteins of these processes. miRNA-200 family members (miR-200a, miR-200b, miR-200c, miR-141 and miR-429) play a critical role in the regulation of the EMT process by targeting and repressing mesenchymal markers, such as vimentin, and up-regulating epithelial markers, such as E-cadherin, by directly targeting ZEB1 and ZEB2, which is a direct target of miR-145, too [57, 58] (Figure 3). Ren et al. [59] reported that ZEB2 also represses the transcription of miR-145, creating a double-negative feedback loop between ZEB2 and miR-145, which is a key in the control of EMT and stem cell properties during the bone metastasis of PCa. Besides, upregulation of miR-21 in PCa also plays an important role in EMT (Figure 3) by decreasing the BTG2 levels, initiating thus the acquisition of luminal markers and EMT [60, 61]. Another invasion-related gene regulated by miR-21 is RECK, a matrix metalloproteinase inhibitor. Neutralizing miR-21 represses the matrix metalloproteinase levels and reverses the invasive phenotype [62]. Recently, Wang et al. [63] found that miR-573 expression was significantly lower in metastatic tissues than matched primary PCa. They demonstrated that miR-573 modulates EMT through the direct target of fibroblast growth factor receptor 1 (FGFR1) gene, promoting the metastasis of PCa cells, and suggested miR-573 as a potential biomarker and/or therapeutic target for PCa management.

Androgen signaling through the androgen receptor (AR) is an important oncogenic pathway for PCa progression. Patients eventually acquire resistance and progress to castration resistant PCa (CRPC). Furthermore, the interactions of miRNAs with AR play a determinant role in the progression from castration sensitive PCa to an incurable CRPC [64]. Several miRNAs (miR-21, miR-31, miR-34 and miR-124) can regulate the androgen receptor expression, and simultaneously, AR can regulate the expression of several miRNAs (miR-21, miR-27a, miR-34, miR-125b, miR-221 and let-7) [65]. Goto et al. [66] recently demonstrated that miR-221 and 222 were significantly downregulated in CRPC specimens, even when this cluster was previously described to be upregulated (Figure 3) [67, 68]. This fact shows the dynamic status of miRNAs in the development of PCa, regulating the same miRNA different targets depending on the point of the cancer progression. Loss of the tumor-suppressive miR-221/222 cluster enhanced migration and invasion in PCa cells targeting Ecm29, whose precise mechanisms in cancer cells remain unknown but it is involved in cancer cells invasion. Ottman et al. [69], comparing androgen sensitive and resistant PCa cell lines, identified 43 miRNAs that are significantly altered during progression of PCa cells to treatment resistance, suggesting their involvement in the development of CRPC. Specific changes in miRNA expression in CRPC patients could be used as a new predictive tool for monitoring the susceptibility of development of CRPC. Additionally, new therapeutic strategies based on the use of miRNA as drugs or drug targets could be explored in the future for the management of CRPC patients.

Functional enrichment resources for miRNA analysis

In order to understand the effects of miRNAs on the regulation of gene expression it is crucial to predict the targets of miRNAs. In this way, multiple bioinformatic tools have been developed to predict possible targets of miRNAs and their functions or their implication in signal pathways, because in vitro validation of all the possibilities of miRNA-targets regulation would be too costly and time consuming. So it is necessary for scientists to provide some criteria to hierarchize trials, then validate in vitro the predicted networks and discover novel biomarkers related with PCa aggressiveness [70, 71].

The tools to predict miRNAs targets are based on certain assumptions, such as the base complementarity in the 3′UTR, thermodynamic stability, target-site accessibility, and evolutionary conservation of miRNA binding sites. PicTar [72] is a combinatorial miRNA target prediction tool, which allows to search miRNA targets on mRNA transcripts according to the alignments of 3′UTRs matching to miRNA seeds and their thermodynamic stabilities. PicTar minimizes false positive results by using sequence alignment to eight vertebrate species and the candidate genes of each species are scored to create a combined score for a gene. TargetScan [73–75] is a web server similar to PicTar, but it also provides conserved targeting sites in the open reading frames of vertebrate genomes. TargetScan only considers stringent seeds, and ignores many potential targets. The intersection of PicTar and TargetScan predictions is recommended in order to achieve both high sensitivity and high specificity.

miRanda [76, 77] was one of the first miRNA target prediction algorithms and one of the most heavily used for target prediction by multiple interfaces, including microRNA.org and MicroCosm. The algorithm ranks the likelihood of each gene to be a miRNA target and the likelihood of each miRNA to target a gene. miRanda can predict more target sites and detect non-conserved seed match because it allows occasional mismatches and G:U base-pairs.

Enrichment analysis of miRNA targets is a statistical method to identify functional modular changes and to elucidate underlying functional mechanisms. After predicting the genes targeted by selected miRNAs using the aforementioned tools, the next step for enrichment analysis is annotating target genes for their participation in pathways and processes. Different pathways are commonly used for this step. Gene ontology (GO) [78] annotates genes to biological/cellular/molecular terms in a hierarchically structured way, whereas Kyoto encyclopedia of genes and genomes (KEGG) [79] assigns genes to functional pathways. The final step in enrichment analysis is the statistical testing for over-representation of a biological process in the set of targeted genes. The hypergeometric distribution or Fisher’s exact test is used to test for enrichment.

Baglioni et al. [80] using an enrichment analysis strategy developed the miRable method to identify new potentially relevant genes and their interaction networks associate to a specific pathology. miRable combines medical knowledge and gene expression data to find new interesting protein-coding genes and miRNAs involved in the disease. The method predicted some genes that have been already studied and others less known in relation to PCa. The three new predicted genes were estrogen receptor 1 (ESR1), miR-548c-3p and miR-494. The role of ESR1, a potential candidate target of miR-182, in PCa has not been investigated adequately although a high number of similarities between the AR and ESR1 signaling pathways are known.

Likewise, Zhang et al. [81] identified 39 novel miRNAs candidates as PCa biomarkers analyzing Gene Expression Omnibus datasets for PCa and benign prostatic hyperplasia (BPH) using bioinformatics tools, considering the regulatory power of miRNAs. Results were verified by qRT-PCR concluding that miRNA-648 could be a novel biomarker of PCa. The authors underlined the implication of miRNA-648 in PCa progression, predicting 10 target genes, including eight that have not been previously described in PCa. Using a similar procedure, Feng et al. [82] found the overexpression of miR-31, proposing this biomarker to distinguish PCa tissues from benign tissues.

miRNA expression in PCa tissue

The first miRNA expression study in PCa tissue was reported by Volinia et al. [83] analyzing 228 miRNAs in 56 PCa tissues and six normal prostate tissues. The authors reported that 39 miRNAs were upregulated, while six were downregulated. In contrast, Porkka et al. [84] comparing 319 miRNAs in nine PCa and four BPH tissues showed that eight miRNAs were upregulated and 22 were downregulated. More recently, Carlsson et al. [85], evaluating 667 miRNA, found nine miRNAs that consistently differ between malignant prostate tissues and the adjacent normal tissue from each case. Srivastava et al. [86] showed that miR-205, miR-214, miR-221 and miR-99b were significantly downregulated in PCa tissues compared to adjacent normal tissue. Areas under the curve (AUCs) obtained by receiver operating characteristics were 0.83, 0.92, 0.75 and 0.86, respectively.

Interestingly, Hellwinkel et al. [87] reported seven miRNAs differentially expressed in normal prostate tissue from patients with PCa compared with normal prostate tissue from patients with negative biopsy. The content of four of these miRNAs (miR-185, miR-16, let-7a and let-7b) in normal prostate tissue remained significantly different comparing patients with PCa and patients with negative biopsy and high PSA levels. The authors suggested that the identification of these cancer-typical miRNA patterns in unsuspicious prostate tissue could lead to the assessment of PCa risk by the evaluation of apparently tumor-free biopsy specimens. Finally, Schaefer et al. [40] identified 15 differently expressed miRNAs comparing 470 miRNAs in matched tumor and adjacent normal tissues obtained after radical prostatectomy. Table 1 shows results regarding miRNAs expression in PCa tissue.

Table 1

Expression of miRNAs in tissue.

ReferencesmiRNAs analyzedUpregulated miRNAsDownregulated miRNAsPatients
Volinia et al. [83]228 miRNAslet-7d, let-7i, miR-101-1 prec, -106a, -124a-1, -135-2, -146, -148, -16-1, -17-5p, -181b-1, -181b-1 prec, -184 prec, -187, -191, -195, -196-1, -196-1, -197, -198, -199a-1, -199a-2, -203, -206, -20a, -21, -214, -223, -25, -26a-1, -27a, -29a, -29b-2, -30c, -32, -34a, -92-2, -93-1, -95let-7a-2 prec, miR-128a prec, -218-2, -29a prec, -149, -24-156 PCa and six normal prostatic tissues
Porkka et al. [84]319 miRNAsmiR-202, -210, -296, -320, -370, -373*, -498, -503let-7a, let-7b, let-7c, let-7d, let-7g, miR-16, -23a, 23b, 26a, -92, 99a, -103, -125a, 125b, -143, -145, -195, -199a, -199a*, -221, -222, -497Nine PCa and four BPH tissues
Schaefer et al. [40]470 miRNAsmiR-375, -182, -96, -183, -182*miR-16, -31, -149, -181b, -221, -222, -205, -125b, -145, -18476 Matched tumor and adjacent normal tissues obtained after radical prostatectomy
Carlsson et al. [41]667 miRNAmiR622, -30d, -425*, -342-3pmiR126*, -34a*, -195, -26a, -29a*19 PCa and 19 normal tissues
Hellwinkel et al. [87]154 miRNAsmiR-124a, miR-146a & b, miR-185, miR-16, let-7a, let-7b31 Normal tissue from PCa patients, 17 normal tissues from patients with negative biopsy and low PSA levels and 14 normal tissues from patients with negative biopsy and high PSA levels
Srivastava et al. [86]32 miRNAsmiR-367*, -758*, -190*miR-221*, -205*, -212*, -99b*, -214*, miR-203, -127-3p, -130a, -335, -376, -10a, -589, -422a, -10b, -25, -210, -99a, -429, -92a, -100, -222, -484, -125b, -574-3p, -328, -483-5p, -331-3p, -135a, let-7c40 Formalin-fixed, paraffin-embedded tissue specimen blocks, comparing PCa tissues to adjacent normal tissue

Circulating miRNAs in prostate cancer

In 2008, Mitchell et al. [30] demonstrated that miRNAs originated in human PCa xenografts enter into the circulation and can be identified in plasma. Furthermore, the authors reported that miR-141 was significantly higher in patients with advanced PCa than in the matched controls, showing an AUC of 0.907. This study was the first to report the presence of miRNAs in the serum of PCa patients. More recently, other studies have shown the usefulness of circulating miRNAs in the detection and prognosis of PCa, demonstrating their presence in serum and plasma as well as in exosomes and in blood mononuclear cells.

Several studies have identified specific circulating miRNA signatures for PCa detection and prognosis using microarrays. Differences between panels are substantial and only miR-141, miR-375 and miR-21 appear repeatedly. Therefore, the selected panels differ substantially, probably due to the various types of sample and platforms used, the age of specimens, the small patient size studied, ethnic differences between series, and the different balance between local and advance PCa in every study. These differences are also obvious in the miRNAs suggested as PCa biomarkers by studies based on qRT-PCR. In consequence, while 74 miRNAs have been suggested as circulating biomarker candidates for PCa, only 25 of these miRNAs have been congruently associated with PCa in more than one study (Figure 4). Remarkably, 11 studies suggested the value of miR-141 and miR-375, while 10 studies suggested the utility of miR-21. Discordant results have been published for six miRNAs. Finally, no clinical value for miRNAs was reported in two studies [39, 88].

Figure 4: Reported concordant and discordant results on circulating miRNAs.
Figure 4:

Reported concordant and discordant results on circulating miRNAs.

Chen et al. [89] initially evaluated 1146 miRNAs in the plasma from 25 PCa (15 nonmetastatic, 10 metastatic) and 17 BPH patients, identifying a panel of five miRNAs (miR-622, miR-1285, let-7e, let-7c, and miR-30c) that could differentiate PCa from BPH and from healthy controls with high accuracy, showing AUCs of 0.924 and 0.860, respectively. Additionally, accuracy increased combining miRNAs with routine PSA test. miR-30c was especially useful in patients with PSA ≥4 μg/L, with an AUC of 0.908, whereas the AUC was of 0.509 for patients with PSA <4 μg/L. Similarly, the diagnostic performance of let-7e was better in patients with PSA <4 μg/L, reporting an AUC of 0.969, while the AUC was 0.773 when PSA was ≥4 μg/L.

Diagnostic value of miRNAs in serum was confirmed by Srivastava et al. [90] selecting a signature of three miRNAs (miR-25, miR-101 and miR-628-5p) from a profile analysis of 667 miRNAs. The validation of selected miRNAs demonstrated a good performance, discriminating PCa patients and healthy age matched controls. AUCs were of 0.66, 0.80 and 0.94 for miR-25, miR-101 and miR-628-5p, respectively.

Moltzahn et al. [91] identified in serum a panel of 12 miRNAs from a 384 miRNAs microarray. The validation study found AUCs from 0.812 to 0.928 for miR-106a, miR-1274, miR-93, miR-223, miR-874 and miR-1207, showing a consistent relationship with CAPRA score, a risk assessment score based on patient age, PSA serum level, clinical tumor stage, Gleason score and percentage of positive biopsy cores. The prognostic value of miRNAs was also demonstrated by Nguyen et al. [92], studying the differential expression of 669 miRNAs in 84 serum samples from patients with PCa. The authors showed that miR-375, miR-378* and miR-141 had an increasing trend with disease progression, while miR-409-3p increased in the high-risk group compared with those in the low-risk group, but significantly declined in the metastatic CRPC, which could be in relation with the androgen deprivation therapy.

This study also demonstrated that miR-375 and miR-141 were significantly expressed in prostate tumor tissues, but not in peripheral blood mononuclear cells, suggesting that their increased expression in serum of advanced PCa patients may result from the selective exocytosis of these miRNAs from PCa cells into the circulation system. This hypothesis was confirmed by Cheng et al. [93] validating the significance of a miRNA panel based on the analysis of 365 miRNAs in serum, comparing metastatic CRPC and age-matched healthy controls. A signature composed by miR-141, miR-200a, miR-200c, miR-375 and miR-210 was significantly expressed in metastatic CRPC patients. The authors found that these miRNAs were also overexpressed in PCa tissue as well as in lymph metastases, suggesting that circulating miRNAs could be plausibly cancer cell-derived.

Watahiki et al. [94] analyzed 742 miRNAs in plasma samples from 50 PCa patients. Eight miRNAs were significantly upregulated in metastatic CRPC patients compared with those with localized PCa, while two were downregulated. The authors proposed a panel composed by miR-141, miR-151-3p and miR-16, showing an AUC of 0.944. Also, the authors described that a panel including miR-141, 151-3p, 152 and 423-3p was associated with a poorer outcome and/or a higher Gleason score. Additionally, miR-141 and miR-152 identified patients with a high probability of recurrence after radical prostatectomy.

At the moment, the widest study about the clinical usefulness of circulating miRNAs has been performed by Mihelich et al. [95], measuring the levels of 21 miRNAs in 50 BPH patients and 100 PCa patients in stages T1–T2, classified according to the Gleason score. The authors proposed a miRNA signature (let7a, miR-103, miR-451, miR-24, miR-26b, miR-30c, miR-93, miR-106a, miR-223, miR-874, miR-146a, miR-125b, miR-100, miR-107 and miR-130b) to predict with high accuracy the absence of high-grade PCa among the BPH and low-grade PCa patients. Seven of these miRNAs (miR-451, miR-106a, miR-223, miR-107, miR-130b, let-7a and miR-26b) were also significantly lower in PCa patients with biochemical recurrence after radical prostatectomy compared with those without biochemical recurrence. Interestingly, according to a logistic regression analysis, both miRNAs signatures were better predictors of high Gleason score or biochemical recurrence than other pre-surgical factors, including PSA, age and percentage of positive cores. Similarly, Shen et al. [96] found a plasma miRNA signature composed by miR-20a, miR-21, miR-145 and miR-221 that predicted the aggressiveness of PCa, distinguishing low and high-risk PCa patients according to D’Amico criteria, with an AUC of 0.824. However, the panels proposed for these groups do not overlap in miRNAs composition. On the other hand, downregulated results reported by Mihelich et al. [95] concerning miR-93, miR-106a, miR-107, miR-130b and miR-451 were discordant with those reported in the studies made by Moltzahn et al. [91], Li et al. [97] and Bryant et al. [98], which indicated that these miRNAs were upregulated in PCa patients.

Candidates for AS include patients with a biopsy Gleason score of six or less, PSA lower than 10 μg/L and clinical stages T1 or T2. Misclassification rates are around 20% or higher [99], remaining a severe limitation to identify accurately patients with low risk PCa. Notably, Wang et al. [100] showed that miR-19, miR-345 and miR-519c-5p serum levels independently predicted adverse pathology in PCa patients eligible for AS. The discriminatory ability to distinguish patients with adverse pathology increased from 0.77 to 0.94 when these miRNAs were added to a model including age, PSA, clinical stage and biopsy involvement. Measurement of miRNAs appears as a new way to better select patients for AS.

Contributions using microarrays and RNA-sequencing technologies are summarized in Table 2 [88–94, 98, 100–104], while Table 3 shows contributions based on qRT-PCR [30, 39, 95–97, 105–116].

Table 2

Circulating miRNAs studies in PCa patients using microarrays and RNA-sequencing technologies.

ReferencesmiRNAs analyzedUpregulated miRNAsDownregulated miRNAsMethodologyqRT-PCR NormalizerBody fluidPatientsMost relevant clinical value
Brase et al. [101]667 miRs

Selected miRs: miR-375, -9*, -141, -200b and -516a-3p
miR-375, -9*, -141, -200b and -516a-3pTaqMan human microRNA array and qRT-PCRcel-miR-39, cel-miR-54 and cel-miR-238SerumScreening set: 7 metastatic PCa and 14 localized PCa

1st Validation set: 45 PCa

2nd Validation set: 71 PCa (48 N1 vs. 23 N0; 29 Gleason ≥8 vs. 42 Gleason 7)
miR-375 and -141 levels correlated with a high Gleason score and lymph-node positive status
Moltzahn et al. [91]384 miRs

12 miRs selected for validation study
Comparing PCa vs. healthy group: miR-874, -1274a, -1207-5p, -93 and -106a

Comparing high risk vs. healthy group: miR-451
Comparing PCa vs. healthy group: miR- 223, -26b, -30c and -24Multiplex qRT-PCRMedian ΔCtSerum12 Low-risk PCa, 12 intermediate-risk PCa, and 12 high-risk PCa

12 Healthy male blood donors
AUCs: miR-106a, 0.928; miR-1274, 0.928; miR-93, 0.907; miR-223, 0.876; miR-874, 0.845; miR-1207, 0.812; miR-24: 0.778.

miR-93, -106a and -24 differentiate healthy and metastatic groups
Bryant et al. [98]742 miRs (cMV)

12 Selected miRs
miR-107, -130b, -141, -2110, -301a, -326, -331-3p, -432, -484, -574-3p, and -625*miR-181a-2*Exiqon qRT-PCR microarray and qRT-PCR

cMV isolated by: exoMir kit
cel-miR-39Plasma cMV and serumSet 1

Plasma: 78 PCa and 28 healthy men

Set 2

Serum: 119 PCa following radical prostatectomy, 72 without recurrence and 47 with metastasis
miR-141 and -375 are significantly increased in plasma exosomes and cMV in metastatic PCa compared with non-recurrent PCa

miR-107 and -574-3p were increased in cMVs of men with non-metastatic

PCa vs. healthy men
Chen et al. [89]1146 miRs

Eight selected miRs for validation study
miR-622 and -1285let-7e, let-7c, and miR-30cIllumina’s Human v2 miRNA microarray and qRT-PCRU6PlasmaScreening set: 17 BPH and 25 CaP

Validation set: 44 BPH, 54 healthy controls and 80 CaP
Combined miR-622, -1285, -30c, let-7e and let-7c discriminate

CaP vs. BPH or healthy controls

AUC of 0.924 and 0.860, respectively
Nguyen et al. [92]669 miRs

Selected miRs: -375, -378*, -141, -409-3p
Comparing metastatic vs. low-risk PCa: miR-375, -378*, and -141Comparing low-risk vs. metastatic PCa: miR-409-3pTaqMan Human MicroRNA Array and qRT-PCRU6Serum28 Low-risk PCa, 30 high-risk PCa and 26 metastatic castration resistant PCamiR-375, -378*, and -141 increase with disease progression
Cheng et al. [93]365 miRs

Selected miRs: -100, -141, -148a, -200a, -200c, -210, -375, -425-5p, -222
miR-141, -200a, -200c, -210, -375TaqMan human microRNA array and qRT-PCRcel-miR-39, cel-miR-54 and cel-miR-238SerumScreening set: 25 metastatic castration resistant PCa and 25 age-matched controls

Validation set:

21 metastatic castration resistant PCa and 20 age-matched healthy controls
AUCs: miR-141, 0.842; miR-200a, 0.638; miR-200c, 0.645; miR-210, 0.652; miR-375, 0.660 (validation set)

miR-210 levels are related to PSA response in metastatic castration resistant PCa
Watahiki et al. [94]742 miRs

Selected

miRs: -141, -152, -375, -16, -21, -126, -151-3p, -200c, -205, -423-3p
Comparing metastatic vs. localized PCa: miR-141, -375, -200c, -126, -21, -151-3p, -152 and -423-3pComparing metastatic vs. localized PCa: miR-205 and -16Exiqon qRT-PCR microarray and qRT-PCRmir-30ePlasma25 Localized PCa and 25 metastatic castration resistant PCamiR-141, -151-3p, -152 and -423-3p are associated with a poorer outcome and/or a higher Gleason score.

Combined miR141, -151-3p, -16 differentiate localized and metastatic PCa

AUC: 0.944; sensitivity: 84%; specificity: 96%
Sapre et al. [88]miRs -16, -20a, -21, -34a, -145, -106b, -182, -205, -221, -222, -331, -375TaqMan human microRNA array and qRT-PCRGeometric meanPlasma37 Low-risk PCa and 33 high-risk PCaNo separation between high and low-risk PCa was obtained using a 12 miRs profile. The better AUC was for miR16 (0.62)
Srivastava et al. [90]667 miRs

Selected miRs: -25, 101, -628-5p
Comparing PCa vs. healthy controls: miR-25, -101, and -628-5pTaqMan® array human microRNA and qRT-PCRmiR-223SerumScreening study: 12 PCa

Confirmation study: 40 PCa and 32 healthy age matched controls
AUCs: miR-25, 0.66; miR-101, 0.80; miR-628-5p, 0.94
Wang et al. [100]672 miRs

36 Selected miRs for the validation study
miR-19a, -19bMicrofluidic-based multiplex qRT-PCRMedian CtSerumDiscovery set:

48 low-risk PCa and 48 PCa with pathologic Gleason score ≥7

Validation set: 25 low-risk PCa and 35 PCa with pathologic Gleason score ≥7
miR-19a/b, -345 and -519c-5p independently predict post-surgical pathological upgrade
Lin et al. [102]46 miRsTaqMan human microRNA array and qRT-PCROverall mean miRNA expression valuePlasma/serum (14 serum and 26 plasma samples)97 Metastatic castration resistant PCa patients receiving docetaxel chemotherapy12 miRNAs are associated with overall survival according to their pre-docetaxel levels or the direction of change in their post-docetaxel levels
Haldrup et al. [103]732 miRsComparing PCa vs. BPH: miR-17*, -200b*, -210, -297, -375, -501-3p, -551b and -562Exiqon’s microRNA ready-to-use PCR panels and qRT-PCRmiR-320a and UniSp3Serum13 BPH, 11 localized PCa, nine lymph node or distant metastases PCa, and 11 castration resistant PCaCombined miR-562, -210, -501-3p, -375 and -551b offer a sensitivity of 84% and 100% specificity (AUC: 0.919)
Huang et al. [104]miR-30a/e-

5p, -99a-5p, let-7c, -1246, -1290, -16-5p, -125a-5p, -375
miR-1290 and -375RNA sequencing and qRT-PCR

Exosomes isolation: ExoQuick exosome precipitation solution
miR-30a-5p, -30e-5p and geometric mean of bothPlasma exosomesScreening set: 23 PCa at the time of androgen deprivation therapy failure

Follow-up set: 100 PCa at the time of androgen deprivation therapy failure
Association of miR-1290, -1246, and -375 with overall survival

Higher levels of miR-1290 and -375 were significantly associated with poor overall survival in the follow-up cohort.
Table 3

Circulating miRNAs studies in PCa patients based on qRT-PCR.

ReferencesmiRNAs analyzedUpregulated miRNAsDownregulated miRNAsMethodologyqRT-PCR NormalizerBody fluidPatientsMost relevant clinical value
Mitchell et al. [30]miR-100, -125b, -141, -143, -205, and -296miR-141qRT-PCRcel-miR-39, cel-miR-54 and cel-miR-238Serum25 Metastatic PCa and 25 matched healthy donorsmiR-141

AUC: 0.907

(PCa vs. healthy)
Zhang et al. [105]miR-21miR-21qRT-PCRU6Serum20 Localized PCa, 20

androgen-dependent PCa, 10 hormone-refractory PCa and six BPH
Elevated levels of miR-21 in hormone-refractory PCa
Yaman Agaoglu et al. [106]miR-21, -141, and -221miR-21 and -221qRT-PCRRNU1APlasma51 PCa (25 with metastasis) and 20 healthy individualsAUCs: miR-21, 0.88; miR-221, 0.83

miR-21 and -221 did not reach the power of PSA to discriminate metastatic vs. localized PCa
Mahn et al. [107]miR-26a, -32, -195, -let7iComparing PCa vs. BPH: miR-26a, -195, and -let7iqRT-PCRcel-miR-39Serum20 Healthy controls, 18 BPH, 37 localized PCa, eight metastatic PCamiR-26a differenciates PCa and BPH with 89% sensitivity and 56% specificity

AUC: 0.703.

Combined miR-26a, miR-32, miR-195, miR-let7i showed a sensitivity of 78% and specificity of 67%.

AUC 0.758 (PCa vs. BPH).
Gonzales et al. [108]miR-141miR-141qRT-PCRNot availablePlasma21 Patients with CaPLongitudinal changes of miR-141 correlates with clinical course
Selth et al. [109]miR-141, mmu-miR-298, mmu-miR-346 and mmu-miR-375miR-141, -298 and -375qRT-PCRcel-miR-39Serum25 Metastatic castration-resistant PCa and 25 healthy menLevels of miR-141, -298 and -375 increased in PCa
Shen et al. [96]Comparing intermediate vs. high risk PCa: miR-21 and -145qRT-PCRSpecific RNA oligonucleotides were used to generate standard curvesPlasma82 PCa (38 low risk, 27 intermediate risk, 17 high risk according to D’Amico criteria)Combined miR-20a, -21, -145 and miR-221 distinguish low and high risk PCa (AUC: 0.824)
Sanders et al. [39]miR-21qRT-PCRcel-miR-39

SNORD43
Serum24 PCa, 24 bladder cancer, 24 renal cell carcinoma, 48 control subjectsmiR-21 was similar in cancer patients and healthy controls
Westermann et al. [110]miR-26a-1 and -141qRT-PCRRNU1-4 and SNORD43Serum54 PCa and 79 non-malignantmiR-26a-1 and -141 are not helpful to detect PCa.

miR-141 increased in patients with Gleason score 9
Singh et al. [111]43 miRsmiR-125b, and -222miR-103qRT-PCROverall mean miRNA expression valueSerum62 Non progressors PCa and 31 progressors PCamiR-103, -125b, and -222 predict progression risk in PCa
Santos et al. [112]miR-7, -145, -199b, -200b, -221, -222, -1233Comparing early vs. late castration-resistant PCa: miR-7 and -221qRT-PCRRNU44Peripheral whole-blood45 PCamiR-7 and -221 predict castration-resistant PCa development
Kotb et al. [113]miR-21 and -221miR-21 and -221qRT-PCRSNORD47Serum10 PCa and 10 BPHmiR-21 and -221 predict

PCa (sensitivity 90% and 80%; specificity 90% and 80%, respectively)
Mihelich et al. [95]21 miRsComparing high vs. low-risk PCa or vs. BPH: let-7a, miR-24, -26b, -30c, -93, -103, -106a, -107, -130b, -146a, -223, and -451qRT-PCRSp3, Sp6,

cel-miR-39 and total RNA input normalization
Serum100 No treated PCa (50 low-grade, 50 high-grade) and 50 BPHCombined let-7a, miR-103, -451, -24, -26b, -30c, -93, -106a, -223, -874, -146a, -125b, -100, -107 and -130b distinguish low-grade PCa and BPH vs. high-grade PCa patients

Combined miR-451, -106a, -223, -107, -130b, let-7a and miR-26b correlates with BCR and predicts disease-free survival
Li et al. [97]miR-16, -20a, -21, -96,

-107, -141, -145, -183, -221, -375, -409 and -574
miR-375, -21, and -574qRT-PCRResults expressed as average Ct values

For exosomes isolation: total exosome rna and protein isolation kit
Serum exosomesEight metastatic PCa, six PCa treated with radical prostatectomy, 10 healthy controls.miR-375, -21 and -574 discriminate between groups
Kelly et al. [114]miR-16, -21, -34a, -141, -143, -145, -155, -125b, -221, -375, -425 and let-7amiR-141, -145 and -155let-7aqRT-PCRmiR-16 and -425Whole blood75 PCa and 27 patients with negative biopsyCombined let-7a, miR-141, -145 and -155 offers an AUC of 0.783 and a PPV of 80%
Kachakova et al. [115]let-7c, miR-30c, miR-141, and miR-375Comparing PCa vs. BPH: miR-375qRT-PCRU6Plasma59 PCa,16 BPH and 11 young asymptomatic menmiR-375 AUC of 0.809.

Combined let-7c, miR-30c, -141, -375 and PSA offers an AUC of 0.877 (sensitivity: 86.8%, specificity 81.8%)
Huang et al. [116]miR-21Comparing PCa patients with recurrence or metastasis vs. PCa patients without them and to healthy controls: miR-21qRT-PCRU6Peripheral blood mononuclear cells75 PCa and 75 healthy subjectsmiR-21 AUC of 0.9 (sensitivity 87.5%, specificity 85.7%)

miR-21 was associated with tumor differentiation, clinical stage, and lymph node metastasis

miRNAs in urine in prostate cancer

Several authors have demonstrated the role of urinary biomarkers in the management of PCa. In theory, urine is a potential way to obtain new PCa biomarkers, because of the anatomical localization of the prostate in relation to the urethra, obtaining exfoliated cancer cells or secreted products through the prostatic ductal system. According to Weber et al. [15], the concentration and number of detectable miRNAs in urine is lower than in plasma, suggesting that they are cleared in the kidneys or destroyed in the urine. However, the enrichment of urine with prostatic cells performing a prostate massage is a way to increase the presence of prostate biomarkers, improving test accuracy [117]. Additionally, the protein content is lower in urine than in serum and plasma, reducing interferences in the miRNAs isolation [118]. Nevertheless, the measurement of miRNAs in urine has the inconvenience derived of the low nucleic acid concentration in this fluid. Methods to obtain RNA based on the enrichment of short RNAs and preamplification before qRT-PCR could be necessary to adequately measure miRNAs in urine.

Most studies used the urine pellet, but total urine and the cell-free fraction are also used, generally after digital rectal examination. According to Cheng et al. [14], miRNAs were found to be significantly enriched and intact in urine-derived exosomes compared with cell-free urine. In fact, exosomes have been found as a robust source of urinary biomarkers, showing the presence after digital rectal examination of high levels of PCA3 and TMPRSS2-ERG in urinary exosomes [12]. However, no studies are currently available about miRNAs in urinary exosomes in PCa, even when they have been widely explored in benign nephropathies [119, 120]. Table 4 presents published results about miRNAs in urine, showing AUCs between 0.639 and 0.769 [49, 86, 88, 98, 121–128]. Unfortunately, there is not a consensus in miRNAs selected for these studies neither congruence with results published in blood. On the other hand, miRNAs were not associated with biochemical recurrence [123], PSA serum levels [126] or Gleason score [122, 126], although according to Lewis et al. [49], miR-888 measured in supernatant, but not in pellet, is associated with high-grade PCa. Finally, the value of miRNAs in the detection of PCa could increase in association with other tests. Egidi et al. [127] showed an AUC of 0.880 combining human kallikrein 3 mRNA with miR-19a-3p, while Yun et al. [128] reported AUCs of 0.761 and 0.738 for hsv1-miR-H18 and hsv2-miR-H9-5p, respectively, for patients in the PSA gray area. These promising preliminary results are worthy of larger studies.

Table 4

Urine miRNAs studies in PCa patients.

ReferencesmiRNAs analyzedUpregulated miRNAsDownregulated miRNAsMethodologyqRT-PCR NormalizerUrine fractionPatientsMost relevant clinical value
Ahumada-Tamayo et al. [121]373 miRsmiR-196b, -574-3p, let-7b, -7c, -7d, -7e, -7g, miR-200b, -149, -20b, -17, -184, -20a, -106a, -671-3p, -148a, -429, -31, -100miR-150, -328qRT-PCREndogenous controlUrine pellet obtained after prostate massageNine PCa and nine BPHDifferential expression of 21 miRNAs between PCa and BPH
Bryant et al. [98]miR-107, -141, -200b, -375, -574-3pComparing Pca vs. controls: miR-107 and -574-3pqRT-PCRRNU44

RNU48
Urine pellet after DRE17 Controls, 70 localized PCa, 48 advanced PCamiR-107, AUC: 0.74 and -574-3p, AUC: 0.66 detect PCa
Srivastava et al. [86]miR-205, -214, -221 and -99bmiR-205 and -214qRT-PCRRNU48Total urine samples36 PCa and 12 age and ethnicity matched healthy donorsmiR-205, AUC: 0.708

miR-214, AUC: 0.743

Combined miR-205 and miR-214

improve detection of PCa: 89% sensitivity and 80% specificity
Korzeniewski et al. [122]miR-483-5p, -1275, and -1290Comparing PCa vs. negative biopsy patients: miR-483-5pqRT-PCRcel-miR-39Cell-free fraction of freely voided urine18 Negative biopsy and 71 PCamiR-483-5p, AUC: 0.694

3-signature miRNA panel did not improve the detection of PCa over PSA level

miR-483-5p is not associated with Gleason score
Corcoran et al. [123]miR-598, miR-34a, miR-146a, miR-148aComparing PCa vs. BPH: miR-148aNANAPublically available datasetNine PCa and eight BPH Publically available datasetmiR-598, miR-148a, and miR-146a are not associated with biochemical recurrence
Sapre et al. [88]miR-21, -20a, -375, -331, -205, -218, -16, -222, -221, -34a, -106b, -182, -145

Selected miRs: -16, -21, -222.
Comparing high risk vs. indolent PCa group in the discovery set: miR-16, -20a, -21, -34a, -145, -106b, -182, -205, -221, -222, -331 and -375

miR-16, -21, -222 were upregulated in the discovery set using Taqman array and qRT-PCR, but failed in the validation set using qRT-PCR
TaqMan human microRNA array and qRT-PCRGeometric meanTotal urine obtained after DREDiscovery set: 33 patients prior to RP (17 indolent PCa and 16 high risk).

Validation set: 22 with high risk

PCa and 14 indolent PCa
Combined miR-16, -21, -222, AUC: 0.75 predict high-risk PCa. However, in the validation set: AUC 0.35
Haj-Ahmad et al. [124]894 miRNAs assayed

Selected miRs: miR-1234, -1238, -1913, -486-5p, -1825, -484 and -483-5p
Comparing PCa vs. healthy: miR-1825Comparing PCa vs. healthy and BPH vs. healthy: miR-484LC Science’s μParaflo™ miRNA microarray and qRT-PCR5S rRNAUrine samples collected with Norgen’s urine preservative solutionEight PCa, 12

BPH and 10 healthy males
Comparing PCa vs. BPH

miR-1825 sensitivity 60%; specificity 69%

miR-484 sensitivity: 80%; specificity: 19%

Combined miR-1825 and 484 sensitivity 45%; specificity 75%
Casanova-Salas et al. [125]miR-182 and 187miR-187qRT-PCRRNU44 and RNU48Urine pellet obtained after DRE45 Negative biopsy and 47 PCamiR-187 provided an independent predictive value for PCa

Combined PSA, PCA3 and miR-187 provided 88.6% sensitivity and 50% specificity (AUC 0.711)
Lewis et al. [49]50 miRNAs in the supernatant of EPS

miR-888, -200b, let-7c, in the pellet fraction of EPS
Comparing high vs. low-risk PCa in the supernatant of EPS: miR-888, -34a, -205Comparing high vs. low-risk PCa in the supernatant of EPS: let-7c, -7d and miR-200b, 92a, -99a, -141, -375qRT-PCRSupernatant of EPS: cel-miR39

Pellet fraction of EPS: RNU44 and RNU48
Supernatant and pellet fractions of EPS (urine) obtained after DRE24 Non-cancer subjects, 25 PCa Gleason 6–7, one PCa Gleason

8, six PCa Gleason 9–10
miR-888 in the supernatant EPS is associated with high-grade PCa

Pellets of EPS failed to correlate with clinical grade or detect PCa
Stephan et al. [126]miR-183, -205qRT-PCRmiR-130b, total RNA and PSA mRNA normalizationUrine pellet after DRE38 PCa and 38 no evidence of malignancymiR-183 and miR-205 failed to detect early and aggressive PCa

miRNAs are not associated with Gleason score and serum PSA
Egidi et al. [127]miR-9-3p and miR-19a-3pComparing Pca vs. BPH: miR-9-3p and miR-19a-3pqRT-PCRmiR-191Urine pellet after DRE41 PCa and 38 age-matched subjects with BPHmiR-19a-3p, AUC: 0.723

miR-9-3p, AUC: 0.769

Highest AUC (0.880) was reached combining KLK3 with miR-19a-3p
Yun et al. [128]1205 Human and 144 viral miRs (discovery set)

Selected miRs: hsa-miR-615-3p, ebv-miRBART4, hsv1-miR-H18, hsv2-miR-H9-5p, and hsa-miR-4316. (validation set 1)

hsv1-miR-H18 and hsv2-miR-H9-5p (validation set 2 and 3)
Comparing PCa vs. BPH controls. (validation set 1): hsa-miR-615-3p, hsv1-miR-H18, hsv2-miR-H9-5p, and hsa-miR-4316

Comparing PCa vs. BPH controls (validation set 2 and 3): hsv1-miR-H18 and hsv2-miR-H9-5p
Agilent human miRNA microarray and

qRT-PCR
Synthesized RNA oligonucleotides were used to generate standard curves.

Normalization using total RNA concentration
Supernatant urineDiscovery set: 14 PCa and 5 BPH

Validation set 1: 173 PCa, 108 BPH

Validation set 2: 191 PCa, 143 BPH

Validation set 3: 46 PCa, 104 negative biopsy
hsv1-miR-H18 and hsv2-miR-H9-5p, AUCs: 0.639 and 0.760, respectively.

AUC for PSA: 0.71

hsv1-miR-H18 and hsv2-miR-H9-5p, AUCs: 0.761 and 0.738, respectively, in the PSA gray area

Conclusions

Cumulative results underlined the role of deregulated miRNAs in the development of PCa. This is a complex process that includes the dynamic participation of several miRNAs in the successive PCa steps. Multiple studies showed the clinical usefulness of miRNAs in the management of PCa; however, results differ regarding their accuracy and the relationship with the aggressiveness of the tumor. Besides, results offer a large variation in the miRNAs included in selected profile. Contradictory results could be associated with important methodological issues related to their quantitation and normalization. In fact, even if miRNAs appear as inherently stable molecules, optimization and standardization of methods are necessary to obtain high quality and reproducible results. The best published results were obtained using miR-141, miR-375 and miR-21. In fact, the selection of the most appropriate miRNAs is a key challenge to obtain outstanding results. Large screening studies to select a miRNA profile result laborious and expensive, whereas bioinformatics tools offer a new approach to select miRNAs that are relevant in PCa development. Currently, the majority of studies concerning the evaluation of the clinical usefulness of miRNAs have been performed in serum or plasma, but urine obtained after prostate massage appears as a new way to describe new PCa biomarkers. Therefore, new efforts are necessary to define the value of miRNAs in the management of PCa.


Corresponding author: Dr. Xavier Filella, Department of Biochemistry and Molecular Genetics (CDB), Hospital Clínic, IDIBAPS, C/Villarroel, 170, 08036 Barcelona, Catalonia, Spain, Phone: 0034 93 227 93 75, Fax: 0034 93 227 93 76, E-mail:
aXavier Filella and Laura Foj contributed equally to this work, and both should be considered first author.
  1. Author contributions: All the authors have accepted responsibility for the entire content of this submitted manuscript and approved submission.

  2. Research funding: None declared.

  3. Employment or leadership: None declared.

  4. Honorarium: None declared.

  5. Competing interests: The funding organization(s) played no role in the study design; in the collection, analysis, and interpretation of data; in the writing of the report; or in the decision to submit the report for publication.

References

1. Melichar B. PSA, PCA3 and the philosophy of prostate cancer management. Clin Chem Lab Med 2013;51:707–12.10.1515/cclm-2013-0156Search in Google Scholar

2. Filella X, Foj L. Emerging biomarkers in the detection and prognosis of prostate cancer. Clin Chem Lab Med 2015;53:963–73.10.1515/cclm-2014-0988Search in Google Scholar

3. Stephan C, Jung K, Ralla B. Current biomarkers for diagnosing of prostate cancer. Future Oncol 2015;11:2743–55.10.2217/fon.15.203Search in Google Scholar

4. Klotz L. Prostate cancer overdiagnosis and overtreatment. Curr Opin Endocrinol Diabetes Obes 2013;20:204–9.10.1097/MED.0b013e328360332aSearch in Google Scholar

5. Alcover J, Filella X. Identification of candidates for active surveillance: should we change the current paradigm? Clin Genitourin Cancer 2015;13:499–504.10.1016/j.clgc.2015.06.001Search in Google Scholar

6. Esteller M. Non-coding RNAs in human disease. Nat Rev Genet 2011;12:861–74.10.1038/nrg3074Search in Google Scholar

7. Sun T, Ye H, Wu C-L, Lee G-S, Kantoff PW. Emerging players in prostate cancer: long non-coding RNAs. Am J Clin Exp Urol 2014;2:294.Search in Google Scholar

8. Filella X, Foj L, Milà M, Augé JM, Molina R, Jiménez W. PCA3 in the detection and management of early prostate cancer. Tumour Biol 2013;34:1337–47.10.1007/s13277-013-0739-6Search in Google Scholar

9. Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 1993;75:843–54.10.1016/0092-8674(93)90529-YSearch in Google Scholar

10. Griffiths-Jones S. miRBase: the microRNA sequence database. Methods Mol Biol Clifton NJ 2006;342:129–38.10.1385/1-59745-123-1:129Search in Google Scholar

11. Arroyo JD, Chevillet JR, Kroh EM, Ruf IK, Pritchard CC, Gibson DF, et al. Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma. Proc Natl Acad Sci USA 2011;108:5003–8.10.1073/pnas.1019055108Search in Google Scholar PubMed PubMed Central

12. Dijkstra S, Birker IL, Smit FP, Leyten GH, de Reijke TM, van Oort IM, et al. Prostate cancer biomarker profiles in urinary sediments and exosomes. J Urol 2014;191:1132–8.10.1016/j.juro.2013.11.001Search in Google Scholar PubMed

13. Gallo A, Tandon M, Alevizos I, Illei GG. The majority of microRNAs detectable in serum and saliva is concentrated in exosomes. PLoS One 2012;7:e30679.10.1371/journal.pone.0030679Search in Google Scholar PubMed PubMed Central

14. Cheng L, Sun X, Scicluna BJ, Coleman BM, Hill AF. Characterization and deep sequencing analysis of exosomal and non-exosomal miRNA in human urine. Kidney Int 2014;86:433–44.10.1038/ki.2013.502Search in Google Scholar PubMed

15. Weber JA, Baxter DH, Zhang S, Huang DY, How Huang K, Jen Lee M, et al. The microRNA spectrum in 12 body fluids. Clin Chem 2010;56:1733–41.10.1373/clinchem.2010.147405Search in Google Scholar PubMed PubMed Central

16. Lan H, Lu H, Wang X, Jin H. MicroRNAs as potential biomarkers in cancer: opportunities and challenges. BioMed Res Int 2015;2015:1–17.10.1155/2015/125094Search in Google Scholar PubMed PubMed Central

17. Whale AS, Huggett JF, Cowen S, Speirs V, Shaw J, Ellison S, et al. Comparison of microfluidic digital PCR and conventional quantitative PCR for measuring copy number variation. Nucleic Acids Res 2012;40:e82.10.1093/nar/gks203Search in Google Scholar PubMed PubMed Central

18. Ma J, Li N, Guarnera M, Jiang F. Quantification of plasma miRNAs by digital PCR for cancer diagnosis. Biomark Insights 2013;8:127–36.10.4137/BMI.S13154Search in Google Scholar PubMed PubMed Central

19. Calin GA, Dumitru CD, Shimizu M, Bichi R, Zupo S, Noch E, et al. Frequent deletions and down-regulation of micro-RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci USA 2002;99:15524–9.10.1073/pnas.242606799Search in Google Scholar PubMed PubMed Central

20. Fletcher CE, Dart DA, Bevan CL. Interplay between steroid signalling and microRNAs: implications for hormone-dependent cancers. Endocr Relat Cancer 2014;21:R409–29.10.1530/ERC-14-0208Search in Google Scholar PubMed

21. Lawrie CH, Gal S, Dunlop HM, Pushkaran B, Liggins AP, Pulford K, et al. Detection of elevated levels of tumour-associated microRNAs in serum of patients with diffuse large B-cell lymphoma. Br J Haematol 2008;141:672–5.10.1111/j.1365-2141.2008.07077.xSearch in Google Scholar PubMed

22. Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, et al. MicroRNA expression profiles classify human cancers. Nature 2005;435:834–8.10.1038/nature03702Search in Google Scholar PubMed

23. Søkilde R, Vincent M, Møller AK, Hansen A, Høiby PE, Blondal T, et al. Efficient identification of miRNAs for classification of tumor origin. J Mol Diagn 2014;16: 106–15.10.1016/j.jmoldx.2013.10.001Search in Google Scholar PubMed

24. Sun Y, Zhang K, Fan G, Li J. Identification of circulating microRNAs as biomarkers in cancers: what have we got? Clin Chem Lab Med 2012;50:2121–6.10.1515/cclm-2012-0360Search in Google Scholar PubMed

25. Wang K, Yuan Y, Cho JH, McClarty S, Baxter D, Galas DJ. Comparing the microRNA spectrum between serum and plasma. PLoS One 2012;7:e41561.10.1371/journal.pone.0041561Search in Google Scholar PubMed PubMed Central

26. Kim D-J, Linnstaedt S, Palma J, Park JC, Ntrivalas E, Kwak-Kim JY, et al. Plasma components affect accuracy of circulating cancer-related microRNA quantitation. J Mol Diagn 2012;14:71–80.10.1016/j.jmoldx.2011.09.002Search in Google Scholar PubMed PubMed Central

27. Kirschner MB, Kao SC, Edelman JJ, Armstrong NJ, Vallely MP, van Zandwijk N, et al. Haemolysis during sample preparation alters microRNA content of plasma. PLoS One 2011;6:e24145.10.1371/journal.pone.0024145Search in Google Scholar PubMed PubMed Central

28. Pritchard CC, Kroh E, Wood B, Arroyo JD, Dougherty KJ, Miyaji MM, et al. Blood cell origin of circulating microRNAs: a cautionary note for cancer biomarker studies. Cancer Prev Res (Phila Pa) 2012;5:492–7.10.1158/1940-6207.CAPR-11-0370Search in Google Scholar PubMed PubMed Central

29. Blondal T, Jensby Nielsen S, Baker A, Andreasen D, Mouritzen P, Wrang Teilum M, et al. Assessing sample and miRNA profile quality in serum and plasma or other biofluids. Methods 2013;59:S1–6.10.1016/j.ymeth.2012.09.015Search in Google Scholar PubMed

30. Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-Agadjanyan EL, et al. Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci 2008;105:10513–8.10.1073/pnas.0804549105Search in Google Scholar PubMed PubMed Central

31. McDonald JS, Milosevic D, Reddi HV, Grebe SK, Algeciras-Schimnich A. Analysis of circulating microRNA: preanalytical and analytical challenges. Clin Chem 2011;57:833–40.10.1373/clinchem.2010.157198Search in Google Scholar PubMed

32. Rice J, Roberts H, Burton J, Pan J, States V, Rai SN, et al. Assay reproducibility in clinical studies of plasma miRNA. PLoS One 2015;10:e0121948.10.1371/journal.pone.0121948Search in Google Scholar PubMed PubMed Central

33. McAlexander MA, Phillips MJ, Witwer KW. Comparison of methods for miRNA extraction from plasma and quantitative recovery of RNA from cerebrospinal fluid. Front Genet 2013;4:83.10.3389/fgene.2013.00083Search in Google Scholar PubMed PubMed Central

34. Kroh EM, Parkin RK, Mitchell PS, Tewari M. Analysis of circulating microRNA biomarkers in plasma and serum using quantitative reverse transcription-PCR (qRT-PCR). Methods 2010;50:298–301.10.1016/j.ymeth.2010.01.032Search in Google Scholar PubMed PubMed Central

35. Channavajjhala SK, Rossato M, Morandini F, Castagna A, Pizzolo F, Bazzoni F, et al. Optimizing the purification and analysis of miRNAs from urinary exosomes. Clin Chem Lab Med 2014;52:345–54.10.1515/cclm-2013-0562Search in Google Scholar PubMed

36. Mestdagh P, Hartmann N, Baeriswyl L, Andreasen D, Bernard N, Chen C, et al. Evaluation of quantitative miRNA expression platforms in the microRNA quality control (miRQC) study. Nat Methods 2014;11:809–15.10.1038/nmeth.3014Search in Google Scholar PubMed

37. Wylie D, Shelton J, Choudhary A, Adai AT. A novel mean-centering method for normalizing microRNA expression from high-throughput RT-qPCR data. BMC Res Notes 2011; 4:555.10.1186/1756-0500-4-555Search in Google Scholar PubMed PubMed Central

38. Peltier HJ, Latham GJ. Normalization of microRNA expression levels in quantitative RT-PCR assays: identification of suitable reference RNA targets in normal and cancerous human solid tissues. RNA 2008;14:844–52.10.1261/rna.939908Search in Google Scholar PubMed PubMed Central

39. Sanders I, Holdenrieder S, Walgenbach-Brünagel G, von Ruecker A, Kristiansen G, Müller SC, et al. Evaluation of reference genes for the analysis of serum miRNA in patients with prostate cancer, bladder cancer and renal cell carcinoma. Int J Urol 2012;19:1017–25.10.1111/j.1442-2042.2012.03082.xSearch in Google Scholar PubMed

40. Schaefer A, Jung M, Mollenkopf H-J, Wagner I, Stephan C, Jentzmik F, et al. Diagnostic and prognostic implications of microRNA profiling in prostate carcinoma. Int J Cancer 2010;126:1166–76.10.1002/ijc.24827Search in Google Scholar PubMed

41. Carlsson J, Helenius G, Karlsson M, Lubovac Z, Andrén O, Olsson B, et al. Validation of suitable endogenous control genes for expression studies of miRNA in prostate cancer tissues. Cancer Genet Cytogenet 2010;202:71–5.10.1016/j.cancergencyto.2010.06.009Search in Google Scholar PubMed

42. Moldovan L, Batte KE, Trgovcich J, Wisler J, Marsh CB, Piper M. Methodological challenges in utilizing miRNAs as circulating biomarkers. J Cell Mol Med 2014;18:371–90.10.1111/jcmm.12236Search in Google Scholar PubMed PubMed Central

43. Sourvinou IS, Markou A, Lianidou ES. Quantification of circulating miRNAs in plasma: effect of preanalytical and analytical parameters on their isolation and stability. J Mol Diagn 2013;15:827–34.10.1016/j.jmoldx.2013.07.005Search in Google Scholar PubMed

44. Roberts TC, Coenen-Stass AM, Betts CA, Wood MJ. Detection and quantification of extracellular microRNAs in murine biofluids. Biol Proced Online 2014;16:1–15.10.1186/1480-9222-16-5Search in Google Scholar PubMed PubMed Central

45. Srivastava A, Goldberger H, Afzal Z, Suy S, Collins SP, Kumar D. Detection of circulatory microRNAs in prostate cancer. Methods Mol Biol Clifton NJ 2015;1238:523–38.10.1007/978-1-4939-1804-1_27Search in Google Scholar PubMed

46. Pang Y, Young CY, Yuan H. MicroRNAs and prostate cancer. Acta Biochim Biophys Sin 2010;42:363–9.10.1093/abbs/gmq038Search in Google Scholar PubMed

47. Goto Y, Kurozumi A, Enokida H, Ichikawa T, Seki N. Functional significance of aberrantly expressed microRNAs in prostate cancer. Int J Urol 2015;22:242–52.10.1111/iju.12700Search in Google Scholar PubMed

48. Fang Y-X, Gao W-Q. Roles of microRNAs during prostatic tumorigenesis and tumor progression. Oncogene 2014;33: 135–47.10.1038/onc.2013.54Search in Google Scholar PubMed

49. Lewis H, Lance R, Troyer D, Beydoun H, Hadley M, Orians J, et al. miR-888 is an expressed prostatic secretions-derived microRNA that promotes prostate cell growth and migration. Cell Cycle Georget Tex 2014;13:227–39.10.4161/cc.26984Search in Google Scholar PubMed PubMed Central

50. Liu C, Kelnar K, Liu B, Chen X, Calhoun-Davis T, Li H, et al. The microRNA miR-34a inhibits prostate cancer stem cells and metastasis by directly repressing CD44. Nat Med 2011;17:211–5.10.1038/nm.2284Search in Google Scholar PubMed PubMed Central

51. Kong D, Heath E, Chen W, Cher ML, Powell I, Heilbrun L, et al. Loss of let-7 up-regulates EZH2 in prostate cancer consistent with the acquisition of cancer stem cell signatures that are attenuated by BR-DIM. PLoS One 2012;7:e33729.10.1371/journal.pone.0033729Search in Google Scholar PubMed PubMed Central

52. Folini M, Gandellini P, Longoni N, Profumo V, Callari M, Pennati M, et al. miR-21: an oncomir on strike in prostate cancer. Mol Cancer 2010;9:12.10.1186/1476-4598-9-12Search in Google Scholar PubMed PubMed Central

53. Li T, Li D, Sha JJ, Sun P, Huang YR. MicroRNA-21 directly targets MARCKS and promotes apoptosis resistance and invasion in prostate cancer cells. Biochem Biophys Res Commun 2009;383:280–5.10.1016/j.bbrc.2009.03.077Search in Google Scholar PubMed

54. Kojima K, Fujita Y, Nozawa Y, Deguchi T, Ito M. MiR-34a attenuates paclitaxel resistance of hormone-refractory prostate cancer PC3 cells through direct and indirect mechanisms. Prostate 2010;70:1501–12.10.1002/pros.21185Search in Google Scholar PubMed

55. Verdoodt B, Neid M, Vogt M, Kuhn V, Liffers ST, Palisaar RJ, et al. MicroRNA-205, a novel regulator of the anti-apoptotic protein Bcl2, is downregulated in prostate cancer. Int J Oncol 2013;43:307–14.10.3892/ijo.2013.1915Search in Google Scholar PubMed

56. Kalluri R, Weinberg RA. The basics of epithelial-mesenchymal transition. J Clin Invest 2009;119:1420–8.10.1172/JCI39104Search in Google Scholar PubMed PubMed Central

57. Gregory PA, Bert AG, Paterson EL. The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat Cell Biol 2008;10:593–601.10.1038/ncb1722Search in Google Scholar PubMed

58. Vandewalle C, Comijn J, De Craene B, Vermassen P, Bruyneel E, Andersen H, et al. SIP1/ZEB2 induces EMT by repressing genes of different epithelial cell–cell junctions. Nucl Acids Res 2005;33:6566–78.10.1093/nar/gki965Search in Google Scholar PubMed PubMed Central

59. Ren D, Wang M, Guo W, Huang S, Wang Z, Zhao X, et al. Double-negative feedback loop between ZEB2 and miR-145 regulates epithelial-mesenchymal transition and stem cell properties in prostate cancer cells. Cell Tissue Res 2014;358:763–78.10.1007/s00441-014-2001-ySearch in Google Scholar PubMed

60. Coppola V, Musumeci M, Patrizii M, Cannistraci A, Addario A, Maugeri-Saccà M, et al. BTG2 loss and miR-21 upregulation contribute to prostate cell transformation by inducing luminal markers expression and epithelial–mesenchymal transition. Oncogene 2013;32:1843–53.10.1038/onc.2012.194Search in Google Scholar PubMed

61. Lawson DA, Zong Y, Memarzadeh S, Xin L, Huang J, Witte ON. Basal epitelial stem cells are efficient targets for prostate cancer initiation. Proc Natl Acad Sci USA 2010;107:2610–5.10.1073/pnas.0913873107Search in Google Scholar PubMed PubMed Central

62. Reis ST, Pontes-Junior J, Antunesnes AA, Dall Oglio MF, Dip N, Passerotti CC, et al. miR-21 may acts as an oncomir by targeting RECK, a matrix metalloproteinase regulator, in prostate cancer. BMC Urol 2012;12:14.10.1186/1471-2490-12-14Search in Google Scholar PubMed PubMed Central

63. Wang L, Song G, Tan W, Qi M, Zhang L, Chan J, et al. miR-573 inhibits prostate cancer metastasis by regulating epithelial-mesenchymal transition. Oncotarget 2015;6:35978–90.10.18632/oncotarget.5427Search in Google Scholar PubMed PubMed Central

64. ChunJiao S, Huan C, ChaoYang X, GuoMei R. Uncovering the roles of miRNAs and their relationship with androgen receptor in prostate cancer. IUBMB Life 2014;66:379–86.10.1002/iub.1281Search in Google Scholar PubMed PubMed Central

65. Ayub SG, Kaul D, Ayub T. Microdissecting the role of microRNAs in the pathogenesis of prostate cancer. Cancer Genet 2015;208:289–302.10.1016/j.cancergen.2015.02.010Search in Google Scholar PubMed

66. Goto Y, Kojima S, Nishikawa R, Kurozumi A, Kato M, Enokida H, et al. MicroRNA expression signature of castration-resistant prostate cancer: the microRNA-221/222 cluster functions as a tumour suppressor and disease progression marker. Br J Cancer 2015;113:1055–65.10.1038/bjc.2015.300Search in Google Scholar PubMed PubMed Central

67. Sun T, Wang X, He HH, Sweeney CJ, Liu SX, Brown M, et al. MiR-221 promotes the development of androgen independence in prostate cancer cells via downregulation of HECTD2 and RAB1A. Oncogene 2014;33:2790–800.10.1038/onc.2013.230Search in Google Scholar PubMed PubMed Central

68. Yang X, Yang Y, Gan R, Zhao L, Li W, Zhou H, et al. Down-regulation of mir-221 and mir-222 restrain prostate cancer cell proliferation and migration that is partly mediated by activation of SIRT1. PLoS One 2014;9:e98833.10.1371/journal.pone.0098833Search in Google Scholar PubMed PubMed Central

69. Ottman R, Nguyen C, Lorch R, Chakrabarti R. MicroRNA expressions associated with progression of prostate cancer cells to antiandrogen therapy resistance. Mol Cancer 2014;13:1.10.1186/1476-4598-13-1Search in Google Scholar PubMed PubMed Central

70. Ekimler S, Sahin K. Computational methods for microRNA target prediction. Genes (Basel) 2014;5:671–83.10.3390/genes5030671Search in Google Scholar PubMed PubMed Central

71. Deng JH, Deng Q, Kuo CH, Delaney SW, Ying SY. MiRNA targets of prostate cancer. Methods Mol Biol 2013;936:357–69.10.1007/978-1-62703-083-0_27Search in Google Scholar PubMed

72. Krek A, Grün D, Poy MN, Wolf R, Rosenberg L, Epstein EJ, et al. Combinatorial microRNA target predictions. Nat Genet 2005;37:495–500.10.1038/ng1536Search in Google Scholar

73. Lewis BP, Shih IH, Jones-Rhoades MW, Bartel DP, Burge CB. Prediction of mammalian microRNA targets. Cell 2003;115: 787–98.10.1016/S0092-8674(03)01018-3Search in Google Scholar

74. Kiriakidou M, Nelson PT, Kouranov A, Fitziev P, Bouyioukos C, Mourelatos Z, et al. A combined computational-experimental approach predicts human microRNA targets. Genes Dev 2004;18:1165–78.10.1101/gad.1184704Search in Google Scholar PubMed PubMed Central

75. Miranda KC, Huynh T, Tay Y, Ang YS, Tam WL, Thomson AM, et al. A pattern-based method for the identification of MicroRNA binding sites and their corresponding heteroduplexes. Cell 2006;126:1203–17.10.1016/j.cell.2006.07.031Search in Google Scholar PubMed

76. Rehmsmeier M, Steffen P, Hochsmann M, Giegerich R. Fast and effective prediction of microRNA/target duplexes. RNA 2004;10:1507–17.10.1261/rna.5248604Search in Google Scholar PubMed PubMed Central

77. John B, Enright AJ, Aravin A, Tuschl T, Sander C, Marks DS. Human MicroRNA targets. PLoS Biol 2004;2:e363.10.1371/journal.pbio.0020363Search in Google Scholar PubMed PubMed Central

78. Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, et al. Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat Genet 2000;25:25–9.10.1038/75556Search in Google Scholar PubMed PubMed Central

79. Kanehisa M. The KEGG database. Novartis Found Symp 2002;247:91–101.10.1002/0470857897.ch8Search in Google Scholar

80. Baglioni M, Russo F, Geraci F, Rizzo M, Rainaldi G, Pellegrini M. A new method for discovering disease-specific MiRNA-target regulatory networks. PLoS One 2015;10:e0122473.10.1371/journal.pone.0122473Search in Google Scholar PubMed PubMed Central

81. Zhang W, Zang J, Jing X, Sun Z, Yan W, Yang D, et al. Identification of candidate miRNA biomarkers from miRNA regulatory network with application to prostate cancer. J Transl Med 2014;12:66.10.1186/1479-5876-12-66Search in Google Scholar PubMed PubMed Central

82. Feng J, Huang C, Diao X, Fan M, Wang P, Xiao Y, et al. Screening biomarkers of prostate cancer by integrating microRNA and mRNA microarrays. Genet Test Mol Biomarkers 2013;17: 807–13.10.1089/gtmb.2013.0226Search in Google Scholar PubMed PubMed Central

83. Volinia S, Calin GA, Liu CG, Ambs S, Cimmino A, Petrocca F, et al. A microRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci USA 2006;103:2257–61.10.1073/pnas.0510565103Search in Google Scholar PubMed PubMed Central

84. Porkka KP, Pfeiffer MJ, Waltering KK, Vessella RL, Tammela TL, Visakorpi T. MicroRNA expression profiling in prostate cancer. Cancer Res 2007;67:6130–5.10.1158/0008-5472.CAN-07-0533Search in Google Scholar PubMed

85. Carlsson J, Davidsson S, Helenius G, Karlsson M, Lubovac Z, Andrén O, et al. A miRNA expression signature that separates between normal and malignant prostate tissues. Cancer Cell Int 2011;11:14.10.1186/1475-2867-11-14Search in Google Scholar PubMed PubMed Central

86. Srivastava A, Goldberger H, Dimtchev A, Ramalinga M, Chijioke J, Marian C, et al. MicroRNA profiling in prostate cancer – the diagnostic potential of urinary miR-205 and miR-214. PLoS One 2013;8:e76994.10.1371/journal.pone.0076994Search in Google Scholar PubMed PubMed Central

87. Hellwinkel OJ, Sellier C, Sylvester Y-M, Brase JC, Isbarn H, Erbersdobler A, et al. A cancer-indicative microRNA pattern in normal prostate tissue. Int J Mol Sci 2013;14:5239–49.10.3390/ijms14035239Search in Google Scholar PubMed PubMed Central

88. Sapre N, Hong MK, Macintyre G, Lewis H, Kowalczyk A, Costello AJ, et al. Curated microRNAs in urine and blood fail to validate as predictive biomarkers for high-risk prostate cancer. PLoS One 2014;9:e91729.10.1371/journal.pone.0091729Search in Google Scholar PubMed PubMed Central

89. Chen Z-H, Zhang G-L, Li H-R, Luo J-D, Li Z-X, Chen G-M, et al. A panel of five circulating microRNAs as potential biomarkers for prostate cancer. Prostate 2012;72:1443–52.10.1002/pros.22495Search in Google Scholar PubMed

90. Srivastava A, Goldberger H, Dimtchev A, Marian C, Soldin O, Li X, et al. Circulatory miR-628-5p is downregulated in prostate cancer patients. Tumour Biol 2014;35:4867–73.10.1007/s13277-014-1638-1Search in Google Scholar PubMed PubMed Central

91. Moltzahn F, Olshen AB, Baehner L, Peek A, Fong L, Stöppler H, et al. Microfluidic-based multiplex qRT-PCR identifies diagnostic and prognostic microRNA signatures in the sera of prostate cancer patients. Cancer Res 2011;71: 550–60.10.1158/0008-5472.CAN-10-1229Search in Google Scholar PubMed PubMed Central

92. Nguyen HC, Xie W, Yang M, Hsieh C-L, Drouin S, Lee G-S, et al. Expression differences of circulating microRNAs in metastatic castration resistant prostate cancer and low-risk, localized prostate cancer. Prostate 2013;73:346–54.10.1002/pros.22572Search in Google Scholar PubMed PubMed Central

93. Cheng HH, Mitchell PS, Kroh EM, Dowell AE, Chéry L, Siddiqui J, et al. Circulating microRNA profiling identifies a subset of metastatic prostate cancer patients with evidence of cancer-associated hypoxia. PLoS One 2013;8:e69239.10.1371/journal.pone.0069239Search in Google Scholar PubMed PubMed Central

94. Watahiki A, Macfarlane RJ, Gleave ME, Crea F, Wang Y, Helgason CD, et al. Plasma miRNAs as biomarkers to identify patients with castration-resistant metastatic prostate cancer. Int J Mol Sci 2013;14:7757–70.10.3390/ijms14047757Search in Google Scholar PubMed PubMed Central

95. Mihelich BL, Maranville JC, Nolley R, Peehl DM, Nonn L. Elevated serum microRNA levels associate with absence of high-grade prostate cancer in a retrospective cohort. PLoS One 2015;10:e0124245.10.1371/journal.pone.0124245Search in Google Scholar PubMed PubMed Central

96. Shen J, Hruby GW, McKiernan JM, Gurvich I, Lipsky MJ, Benson MC, et al. Dysregulation of circulating microRNAs and prediction of aggressive prostate cancer. Prostate 2012;72:1469–77.10.1002/pros.22499Search in Google Scholar PubMed PubMed Central

97. Li M, Rai AJ, Joel de Castro G, Zeringer E, Barta T, Magdaleno S, et al. An optimized procedure for exosome isolation and analysis using serum samples: application to cancer biomarker discovery. Methods 2015;87:26–30.10.1016/j.ymeth.2015.03.009Search in Google Scholar PubMed

98. Bryant RJ, Pawlowski T, Catto JW, Marsden G, Vessella RL, Rhees B, et al. Changes in circulating microRNA levels associated with prostate cancer. Br J Cancer 2012;106:768–74.10.1038/bjc.2011.595Search in Google Scholar PubMed PubMed Central

99. Palisaar JR, Noldus J, Löppenberg B, von Bodman C, Sommerer F, Eggert T. Comprehensive report on prostate cancer misclassification by 16 currently used low-risk and active surveillance criteria. BJU Int 2012;110:E172–81.10.1111/j.1464-410X.2012.10935.xSearch in Google Scholar PubMed

100. Wang SY, Shiboski S, Belair CD, Cooperberg MR, Simko JP, Stoppler H, et al. miR-19, miR-345, miR-519c-5p serum levels predict adverse pathology in prostate cancer patients eligible for active surveillance. PLoS One 2014;9:e98597.10.1371/journal.pone.0098597Search in Google Scholar PubMed PubMed Central

101. Brase JC, Johannes M, Schlomm T, Fälth M, Haese A, Steuber T, et al. Circulating miRNAs are correlated with tumor progression in prostate cancer. Int J Cancer 2011;128:608–16.10.1002/ijc.25376Search in Google Scholar PubMed

102. Lin HM, Castillo L, Mahon KL, Chiam K, Lee BY, Nguyen Q, et al. Circulating microRNAs are associated with docetaxel chemotherapy outcome in castration-resistant prostate cancer. Br J Cancer 2014;110:2462–71.10.1038/bjc.2014.181Search in Google Scholar PubMed PubMed Central

103. Haldrup C, Kosaka N, Ochiya T, Borre M, Høyer S, Orntoft TF, et al. Profiling of circulating microRNAs for prostate cancer biomarker discovery. Drug Deliv Transl Res 2014;4:19–30.10.1007/s13346-013-0169-4Search in Google Scholar PubMed

104. Huang X, Yuan T, Liang M, Du M, Xia S, Dittmar R, et al. Exosomal miR-1290 and miR-375 as prognostic markers in castration-resistant prostate cancer. Eur Urol 2015;67:33–41.10.1016/j.eururo.2014.07.035Search in Google Scholar PubMed PubMed Central

105. Zhang HL, Yang LF, Zhu Y, Yao XD, Zhang SL, Dai B, et al. Serum miRNA-21: elevated levels in patients with metastatic hormone-refractory prostate cancer and potential predictive factor for the efficacy of docetaxel-based chemotherapy. Prostate 2011;71:326–31.10.1002/pros.21246Search in Google Scholar PubMed

106. Yaman Agaoglu F, Kovancilar M, Dizdar Y, Darendeliler E, Holdenrieder S, Dalay N, et al. Investigation of miR-21, miR-141, and miR-221 in blood circulation of patients with prostate cancer. Tumour Biol 2011;32:583–8.10.1007/s13277-011-0154-9Search in Google Scholar PubMed

107. Mahn R, Heukamp LC, Rogenhofer S, von Ruecker A, Müller SC, Ellinger J. Circulating microRNAs (miRNA) in serum of patients with prostate cancer. Urology 2011;77:1265.e9–16.10.1016/j.urology.2011.01.020Search in Google Scholar PubMed

108. Gonzales JC, Fink LM, Goodman OB Jr, Symanowski JT, Vogelzang NJ, Ward DC. Comparison of circulating MicroRNA 141 to circulating tumor cells, lactate dehydrogenase, and prostate-specific antigen for determining treatment response in patients with metastatic prostate cancer. Clin Genitourin Cancer 2011;9:39–45.10.1016/j.clgc.2011.05.008Search in Google Scholar PubMed

109. Selth LA, Townley S, Gillis JL, Ochnik AM, Murti K, Macfarlane RJ, et al. Discovery of circulating microRNAs associated with human prostate cancer using a mouse model of disease. Int J Cancer 2012;131:652–61.10.1002/ijc.26405Search in Google Scholar PubMed

110. Westermann AM, Schmidt D, Holdenrieder S, Moritz R, Semjonow A, Schmidt M, et al. Serum microRNAs as biomarkers in patients undergoing prostate biopsy: results from a prospective multi-center study. Anticancer Res 2014;34:665–9.Search in Google Scholar

111. Singh PK, Preus L, Hu Q, Yan L, Long MD, Morrison CD, et al. Serum microRNA expression patterns that predict early treatment failure in prostate cancer patients. Oncotarget 2014;5:824–40.10.18632/oncotarget.1776Search in Google Scholar PubMed PubMed Central

112. Santos JI, Teixeira AL, Dias F, Maurício J, Lobo F, Morais A, et al. Influence of peripheral whole-blood microRNA-7 and microRNA-221 high expression levels on the acquisition of castration-resistant prostate cancer: evidences from in vitro and in vivo studies. Tumour Biol 2014;35:7105–13.10.1007/s13277-014-1918-9Search in Google Scholar PubMed

113. Kotb S, Mosharafa A, Essawi M, Hassan H, Meshref A, Morsy A. Circulating miRNAs 21 and 221 as biomarkers for early diagnosis of prostate cancer. Tumour Biol 2014;35:12613–7.10.1007/s13277-014-2584-7Search in Google Scholar

114. Kelly BD, Miller N, Sweeney KJ, Durkan GC, Rogers E, Walsh K, et al. A circulating microRNA signature as a biomarker for prostate cancer in a high risk group. J Clin Med 2015;4:1369–79.10.3390/jcm4071369Search in Google Scholar

115. Kachakova D, Mitkova A, Popov E, Popov I, Vlahova A, Dikov T, et al. Combinations of serum prostate-specific antigen and plasma expression levels of let-7c, miR-30c, miR-141, and miR-375 as potential better diagnostic biomarkers for prostate cancer. DNA Cell Biol 2015;34:189–200.10.1089/dna.2014.2663Search in Google Scholar

116. Huang W, Kang XL, Cen S, Wang Y, Chen X. High-level expression of microRNA-21 in peripheral blood mononuclear cells is a diagnostic and prognostic marker in prostate cancer. Genet Test Mol Biomarkers 2015;19:469–75.10.1089/gtmb.2015.0088Search in Google Scholar

117. Hessels D, Klein Gunnewiek JM, van Oort I, Karthaus HF, van Leenders GJ, van Balken B, et al. DD3(PCA3)-based molecular urine analysis for the diagnosis of prostate cancer. Eur Urol 2003;44:8–15.10.1016/S0302-2838(03)00201-XSearch in Google Scholar

118. Mlcochova H, Hezova R, Meli AC, Slaby O. Urinary MicroRNAs as a new class of noninvasive biomarkers in oncology, nephrology, and cardiology. Methods Mol Biol 2015;1218:439–63.10.1007/978-1-4939-1538-5_26Search in Google Scholar PubMed

119. Alvarez ML, Khosroheidari M, Kanchi Ravi R, DiStefano JK. Comparison of protein, microRNA, and mRNA yields using different methods of urinary exosome isolation for the discovery of kidney disease biomarkers. Kidney Int 2012;82:1024–32.10.1038/ki.2012.256Search in Google Scholar PubMed

120. Barutta F, Tricarico M, Corbelli A, Annaratone L, Pinach S, Grimaldi S, et al. Urinary exosomal microRNAs in incipient diabetic nephropathy. PLoS One 2013;8:e73798.10.1371/journal.pone.0073798Search in Google Scholar PubMed PubMed Central

121. Ahumada-Tamayo S, Saavedra-Briones D, Cantellano-Orozco M, Salido-Guadarrama A, Rodríguez-Dorantes M, Urdiales-Ortiz A, et al. MicroRNA determination in urine for prostate cancer detection in Mexican patients at the Hospital General “Dr.Manuel Gea González”. Rev Mex Urol 2011;71: 213–7.Search in Google Scholar

122. Korzeniewski N, Tosev G, Pahernik S, Hadaschik B, Hohenfellner M, Duensing S. Identification of cell-free microRNAs in the urine of patients with prostate cancer. Urol Oncol 2015;33:16.e17–22.10.1016/j.urolonc.2014.09.015Search in Google Scholar PubMed

123. Corcoran C, Rani S, O’Driscoll L. miR-34a is an intracellular and exosomal predictive biomarker for response to docetaxel with clinical relevance to prostate cancer progression. Prostate 2014;74:1320–34.10.1002/pros.22848Search in Google Scholar PubMed

124. Haj-Ahmad TA, Abdalla MA, Haj-Ahmad Y. Potential urinary miRNA biomarker candidates for the accurate detection of prostate cancer among benign prostatic hyperplasia patients. J Cancer 2014;5:182–91.10.7150/jca.6799Search in Google Scholar PubMed PubMed Central

125. Casanova-Salas I, Rubio-Briones J, Calatrava A, Mancarella C, Masiá E, Casanova J, et al. Identification of miR-187 and miR-182 as biomarkers of early diagnosis and prognosis in patients with prostate cancer treated with radical prostatectomy. J Urol 2014;192:252–9.10.1016/j.juro.2014.01.107Search in Google Scholar PubMed

126. Stephan C, Jung M, Rabenhorst S, Kilic E, Jung K. Urinary miR-183 and miR-205 do not surpass PCA3 in urine as predictive markers for prostate biopsy outcome despite their highly dysregulated expression in prostate cancer tissue. Clin Chem Lab Med 2015;53:1109–18.10.1515/cclm-2014-1000Search in Google Scholar PubMed

127. Egidi MG, Guelfi G, Cochetti G, Poli G, Barillaro F, Zampini D, et al. Characterization of kallireins and microRNAs in urine sediment for the discrimination of prostate cancer from benign prostatic hyperplasia. J Cancer Sci Ther 2015;7:130–6.Search in Google Scholar

128. Yun SJ, Jeong P, Kang HW, Kim Y-H, Kim E-A, Yan C, et al. Urinary microRNAs of prostate cancer: virus-encoded hsv1-miRH18 and hsv2-miR-H9-5p could be valuable diagnostic markers. Int Neurourol J 2015;19:74–84.10.5213/inj.2015.19.2.74Search in Google Scholar PubMed PubMed Central

Received: 2015-11-3
Accepted: 2015-11-29
Published Online: 2016-1-9
Published in Print: 2017-5-1

©2017 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 25.4.2024 from https://www.degruyter.com/document/doi/10.1515/cclm-2015-1073/html
Scroll to top button