Topic Highlight Open Access
Copyright ©2014 Baishideng Publishing Group Inc. All rights reserved.
World J Clin Oncol. Oct 10, 2014; 5(4): 604-620
Published online Oct 10, 2014. doi: 10.5306/wjco.v5.i4.604
MicroRNAs as lung cancer biomarkers
Valerio Del Vescovo, Margherita Grasso, Michela A Denti, Laboratory of RNA Biology and Biotechnology, Centre for Integrative Biology, University of Trento, 38123 Trento, Italy
Mattia Barbareschi, Unit of Surgical Pathology, Laboratory of Molecular Pathology, Trentino Biobank, S.Chiara Hospital, 38100 Trento, Italy
Author contributions: Del Vescovo V and Grasso M contributed equally to this work, searched the literature material and designed the research; Del Vescovo V, Grasso M and Denti MA wrote the paper; Barbareschi M and Denti MA revised the paper.
Correspondence to: Dr. Michela A Denti, Laboratory of RNA Biology and Biotechnology, Centre for Integrative Biology, University of Trento, via Sommarive 5, 38123 Trento, Italy. denti@science.unitn.it
Telephone: +39-0461-283820 Fax: +39-0461-283937
Received: January 16, 2014
Revised: February 28, 2014
Accepted: May 8, 2014
Published online: October 10, 2014

Abstract

Lung cancer is the leading cause of cancer mortality worldwide. Its high mortality is due to the poor prognosis of the disease caused by a late disease presentation, tumor heterogeneities within histological subtypes, and the relatively limited understanding of tumor biology. Importantly, lung cancer histological subgroups respond differently to some chemotherapeutic substances and side effects of some therapies appear to vary between subgroups. Biomarkers able to stratify for the subtype of lung cancer, prognosticate the course of disease, or predict the response to treatment are in high demand. In the last decade, microRNAs (miRNAs), measured in resected tumor samples or in fine needle aspirate samples have emerged as biomarkers for tumor diagnosis, prognosis and prediction of response to treatment, due to the ease of their detection and in their extreme specificity. Moreover, miRNAs present in sputum, in plasma, in serum or in whole blood have increasingly been explored in the last five years as less invasive biomarkers for the early detection of cancers. In this review we cover the increasing amounts of data that have accumulated in the last ten years on the use of miRNAs as lung cancer biomarkers.

Key Words: Lung cancer, Non-small cell lung carcinomas, MicroRNAs, Biomarkers

Core tip: Biomarkers able to stratify for the subtype of lung cancer, prognosticate the course of disease, or predict the response to treatment are in high demand. In the last decade, microRNAs (miRNAs), measured in resected tumor samples have emerged as biomarkers, due to the ease of their detection and in their extreme specificity. Moreover, miRNAs present in sputum, in plasma, in serum or in whole blood have increasingly been explored in the last five years as less invasive biomarkers for the early detection of cancers.



INTRODUCTION

Lung cancer is the leading cause of cancer mortality worldwide[1]. About 270000 individuals were predicted to die of lung cancer in the European Union in 2013[2]. This high mortality is due to the poor prognosis of the disease caused by a late disease presentation, tumour heterogeneities within histological subtypes, and the relatively limited understanding of tumour biology. Most lung cancer patients are diagnosed at an advanced stage of disease, and, although a small subsets of these patients can be treated with new drugs offering improved survival and reasonable quality of life, the majority of patients can only be treated with palliative chemotherapy. Overall survival remains poor, and many patients die within a few months of diagnosis.

Lung cancer is not one, but rather a group of diseases: small cell lung carcinomas (SCLCs) are high grade neuroendocrine tumours (NET), metastasize earlier and are initially more chemosensitive than the so called non-small cell lung carcinomas (NSCLC). Over the past two decades it has become clear that NSCLC itself is a clinically and biologically heterogeneous group of lung cancers, and should not be treated as a single disease entity. The two main subgroups of NSCLC are adenocarcinoma (AD) and squamous cell carcinoma (SCC), with a remaining third class of carcinomas devoid of histological features of adeno- or squamous- differentiation, named LCC (large cell carcinoma)[3]. The appearance of these tumours at light microscopy differs substantially, suggesting that their aetiology and biology differ as well. Importantly, the histological subgroups of NSCLC respond differently to some chemotherapeutic substances[4-7] and side effects of some therapies appear to vary between subgroups[8].

Beside SCLC, NETs include a spectrum of tumors from the low-grade typical carcinoid (TC) and intermediate-grade atypical carcinoid (AC) to the high-grade large-cell NE carcinoma (LCNEC). The distinction between these various entities may be sometimes difficult on histological grounds, but is of great therapeutic relevance.

Biomarkers able to stratify for the subtype of lung cancer, prognosticate the course of disease, or predict the response to treatment are in increasing demand. Lung cancer subtyping has traditionally relied on the histopathological observation of resected specimens, bronchoscopic biopsies, fine needle aspirations or sputum, which represent samples with decreasing invasiveness for the patient, but also of increasing challenge for the pathologist, as proportionally fewer tumour cells are captured[9-11]. Recently, the introduction of several immunohistochemical markers has rendered lung cancer subtyping more accurate and clinically useful[12].

In the last decade, microRNAs (miRNAs), measured either from tumour samples or in biofluids, have emerged as biomarkers for tumor diagnosis, prognosis and prediction of response to treatment. In the following pages we will review the increasing amounts of data that have accumulated in the last ten years on the use of miRNAs as lung cancer biomarkers.

MIRNAS AND CANCER

microRNAs (miRNAs or miRs) are small non-coding single-stranded RNAs, 19-25 nucleotides (nt) in length, acting as negative regulators of gene expression at the post-transcriptional level. More than 1000 miRNAs are transcribed from miRNA genes in the human genome. A single miRNA is able to modulate hundreds of downstream genes by recognizing complementary sequences in the 3’UTRs of their target mRNAs. It has been estimated that in humans about 30% of messenger RNAs (mRNAs) are under miRNA regulation, but this percentage is likely to grow in the future, as studies have shown that miRNAs can also bind target sequences located in the 5’ UTR or in the open reading frame (ORF)[13]. The biological functions of miRNAs are diverse and include several key cellular processes, such as differentiation, proliferation, cellular development, cell death and metabolism.

In a seminal paper the Croce laboratory showed in 2002 that the genes for miR-15 and miR-16 are deleted or down-regulated in the majority of chronic lymphocytic leukaemia cases[14]. Scott Hammond, Gregory Hannon and collaborators demonstrated that miRNAs can modulate tumour formation and implicated the mir-17-92 cluster as a potential human oncogene in another fundamental paper[15], in which they demonstrated that enforced expression of the mir-17-92 cluster acts with c-myc expression to accelerate tumour development in a mouse B-cell lymphoma model. Since then, evidences have accumulated to indicate that miRNAs play a role in the onset and progression of several human cancers[16]. The transcription or processing of some miRNAs is altered in neoplastic tissues, in respect to their normal counterparts. miRNAs whose levels increase in tumors are referred to as oncogenic miRNAs (“onco-miRs”), sometimes even if there is no evidence for their causative role in tumorigenesis. On the other hand, miRNAs down-regulated in cancer are considered tumor suppressors. From the mechanistic point of view, it is important to understand how these variations may contribute to tumor progression.

In 2005, the Horvitz and Golub labs demonstrated the potential for miRNAs as diagnostic tumor markers, when they were able to indicate the tumor embryonic origin using miRNA expression profiles, successfully classifying poorly differentiated tumors, among which lung tumors[17]. Subsequently, the Croce lab performed a large-scale miRNome analysis on 540 samples including lung, breast, stomach, prostate, colon, and pancreatic tumors, and identified a solid cancer miRNA signature composed by a large portion of over-expressed miRNAs. While some miRNAs were commonly dys-regulated in the six cancer types, several other miRNAs were associated to a particular type of cancer[18]. The utility of miRNAs levels as diagnostic and prognostic biomarkers became clear already from these first studies[19]. Moreover, the effectiveness of miRNAs as biomarkers for tracing the tissue of origin of cancers of unknown primary origin was demonstrated by Rosenfeld and colleagues[20], who constructed a tissue classifier based on the measurement of 48 miRNAs on a microarray, to identify the tissue origin of metastatic tumors. These results were translated into a qRT-PCR platform, to develop a diagnostic test for the identification of tumour tissue origin[21]. The classifier has been further implemented in a second-generation custom microarray based on the measurement of 64 miRNAs[22] the uselfuness of which as a diagnostic tool was very recently confirmed[23].

A different 47-miRNA signature for the identification of cancers with unknown primary tissue-of-origin was identified by other authors using a different microarray platform[24].

MIRNAS AND LUNG CANCER

As far as lung cancers are concerned, the role of miRNAs in lung carcinogenesis was indicated as early as 2004, when the Croce lab demonstrated that more than half of the miRNA genes then known were located in cancer-associated genomic regions or in fragile sites and that several miRNAs located in this deleted regions have low expression levels in lung cancer cell lines as well as in chronic lymphocytic leukaemia samples[25]. In the same year, Takamizawa and colleagues reported reduced expression of the let-7 microRNA in human NSCLC lung cancers[26], followed by several independent studies[27-29]. The let-7 family was later shown to have an onco-suppressor activity in NSCLC tumor development in mice xenografts[30]. One of the consequences of let-7a down-regulation in lung cancer has been demonstrated to be the upregulation of RAS protein[27]. A single nucleotide polymorphism (SNP) in a let-7 complementary site of KRAS mRNA was found to be associated with increased risk of NSCLC in moderate smokers[31]. Based on in vitro experiments and analyses of patient samples the authors concluded that this SNP alters the ability of let-7 to regulate translation of KRAS, leading to overexpression of KRAS and increased lung cancer risk.

Other miRNAs may also interact with RAS. For instance, Wang et al[32] found that miR-451 is downregulated in NSCLC, and that low expression correlated with poor survival. The authors were able to show that miR-451 inhibits the expression of ras-related protein 14 (RAB14), suggesting that lower expression of miR-451 may allow this oncogene to escape regulation.

The oncogenic miR-17-92 cluster is markedly overexpressed in lung cancers, especially with SCLC histology and enhances cell proliferation in vitro, therefore possibly playing a role in the development of lung cancers[33]. On the other hand, deletion of the miR-17-92 cluster, in mice, is lethal and causes lung and lymphoid cell developmental defects[34].

The tumor suppressor protein p53 is mutated in a large number of lung cancer cell lines and tumour specimens from patients with lung cancer[35,36]. There is growing evidence that p53 regulates the expression of several miRNAs[37-42]. p53 directly regulates the expression of miR-34 family members, and the upregulation of these miRNAs result in the downregulation of genes associated with cell cycle control[37] and promotion of apoptosis[40] in cultured lung cancer cells. Further miRNAs, including miR-125a, have more recently also been linked to p53-regulated apoptosis in lung cancer cells[41].

METHODS FOR THE QUANTIFICATION OF MIRNAS

The main advantage of the use of miRNAs as biomarkers resides in the ease of their detection and in their extreme specificity. miRNAs are stable molecules well preserved in formalin fixed, paraffin embedded tissues (FFPE) as well as in fresh snap-frozen specimens, unlike larger RNA molecules as mRNAs[43].

A range of methods has been used for the isolation and profiling of miRNAs. Purification of total RNA is obtained either through several commercially available column filtration protocols, implemented to guarantee recovery of miRNAs, or via the extraction of RNA by variously named “Tri-reagents” (acid phenol in combination with guanidinium-thiocyanate and chloroform), also available from vendors. Given that the interest is focused on the quantification of specific miRNAs in different conditions, the method of choice should exclude any bias in the purification of miRNAs from the samples. Importantly, the Kim laboratory has recently reported that, differently from what everybody in the field has thought for decades, in Tri-reagents-based RNA purification protocols, short structured miRNAs with low GC content are lost when a small number of cells are used[44]. The finding raises warning flags about comparisons of miRNA levels between populations of cells at different densities.

Sequencing-, microarray- and quantitative reverse-transcription polymerase chain reaction (qRT-PCR)-based methods are currently used for miRNA profiling.

Next generation sequencing (NGS)[45], providing accurate and sensitive miRNAs measurements, allows the identification of miRNAs differently expressed in tumor samples and matched healthy tissue. Furthermore, NGS enables scientists to discover novel miRNAs, as opposed to microarrays and qRT-PCR methods, which only can detect already known miRNAs. NGS techniques are rapidly evolving in power and multiplexing capacities, but they remain expensive and labor-consuming, both in sample preparation and data analysis.

The majority of miRNA profiling studies have been carried out so far using microarrays and have provided signatures consisting in few to several (5-30) distinct miRNAs[46]. With time, it has become clear in the field that the use of microarrays for miRNA profiling presents with major problems of cross-hybridisation between members of miRNA families and discrepancies in comparing results obtained with different microarray platforms[47-59]. A common strategy is to validate the microarray data by qRT-PCR, which warrantees high sensitivity and specificity.

Additionally, the analysis of a great number of genes including ones whose changes are not directly or indirectly associated with cancer, in clinical settings is not necessary. Hence, the large amount of data obtained by microarray and NGS profiling needs to be transposed into clinical trials by developing an easily performed and serviceable assay that can analyze the cancer-specific miRNAs for cancer diagnosis and prognosis. Such an analysis has been so far relying on qRT-PCR assays.

Several methods for the analysis of miRNAs by qRT-PCR have been devised[60] and companies are providing multiwell plate-based qRT-PCR assays that promise to substitute microarrays in the high-throughput profiling of miRNAs. Additionally, qRT-PCR is the most easily performed and cost-effective technique and it is therefore the method of choice when it comes to measuring the levels of the restricted number of miRNA biomarkers in cancer samples, in the clinical setting.

qRT-PCR measures the relative levels of variable target miRNAs in comparison with one or more stably expressed reference genes (sometimes called housekeeping or endogenous control genes). This normalization is required to allow for variability in RNA quantity and quality and/or in the efficiency of cDNA synthesis. Despite miRNAs having been intensively studied in cancer research in the last years, suitable reference genes for relative quantification of miRNA levels in qRT-PCR assays have not been satisfyingly identified. Ribosomal RNAs (rRNAs) have been used as a reference RNA in miRNA studies[26,61]. However, concerns have been raised regarding the use of rRNAs in normalization, as they can be expressed at much higher levels than the target RNA, making it challenging to quantify an rRNA and a rare transcript in the same RNA dilution. Moreover, there is evidence of rRNA deregulation in apoptosis[62] and cancer[63]. The most commonly used reference RNA in miRNA qRT-PCR experiments is the small nuclear RNA U6 (U6 snRNA)[55,64]. However, using U6 snRNA to normalise miRNA levels is controversial because being much bigger in size (106-107 nt) it might differ from miRNAs with respect to efficiency of its extraction, reverse transcription and PCR amplification. U6 snRNA has been analysed as a reference gene for miRNA studies in several papers, and most of them came to the conclusion that the variance of U6 expression across tissues is high, therefore making U6 not a suitable reference gene for miRNA quantification[65-67]. Further reference genes commonly used used are snoRNAs but they too might be dys-regulated in cancer[68]. Several authors have suggested to use as reference a combination of miRNAs whose levels do not vary in the specific tumor tissue under investigation. For example, in fresh-frozen lung cancer samples, Peltier and Latham[65] suggest the use of a combination of miR-191 and miR-103.

The use of unvalidated and different reference genes makes it difficult to compare papers describing miRNA-expression cancer profiles and might represent one of the major reasons for the discrepancies in published studies regarding differentially expressed miRNAs in specific cancers.

A recent addition to the detection methods for miRNAs is droplet digital PCR (dPCR)[69], a method especially useful for low abundance miRNAs. In droplet dPCR, single cDNA molecules are partitioned evenly among hundreds of individual droplets in which they are amplified to generate binary calls. With this method an absolute readout of total DNA copy number can be obtained, avoiding the need for an endogenous reference gene.

MIRNAS FROM RESECTED SPECIMENS
MiRNAs as biomarkers for the diagnosis of lung cancer and for stratifying lung cancer subtypes

In 2005, the Horvitz and Golub labs used a bead-based flow cytometric miRNA expression method to identify complex profiles consisting of approximately a hundred of dys-regulated miRNAs, able to classify 11 different tumor types, among which lung tumors[17]. Subsequently, the Croce lab used a custom-made oligonucleotide miRNA microarray to compare lung carcinomas to normal tissue, and identified a group of three downregulated and 35 upregulated miRNAs. Among these, miR-21 was commonly up-regulated in the six cancer types analyzed (lung, breast, stomach, prostate, colon, and pancreatic tumors) and miR-17-5p, miR-128b, miR-155, miR-191 and miR-199a-1 were up-regulated in at least other two cancer types[18].

In 2006 Yanaihara et al[28] compared the miRNAs expression profiles in 104 pairs of lung cancer tissues and corresponding non-cancerous lung tissue, by the same custom-made oligonucleotide miRNA microarray used by Volinia and colleagues. They identified a unique profile made of 43 differently expressed miRNAs (Table 1) allowing the distinction of lung cancer from the non-cancerous lung tissue. Of the 15 upregulated and 28 downregulated miRNAs, miR-21 and miR-205 are located in a chromosomal region amplified and miR-32, miR-126-5p and miR-126-3p in a region deleted in lung cancers, respectively. The authors next validated the microarray results by a solution hybridization detection method and by qRT-PCR, confirming that miR-21 and miR-205 are frequently up-regulated and miR-126-5p is often down-regulated in lung cancer tissues when compared with the corresponding noncancerous lung tissues.

Table 1 MiRNAs from resected samples as biomarkers for the diagnosis of lung cancer.
MiRNAScopeSampleRef.
mir-21, mir-191, mir-210, mir-155, mir-205, mir-24-2, mir-212, mir-214, mir-17-3p, mir-106a, mir-197, mir-192, mir-146, mir-203, mir-150, (UP) mir-126-5p, mir-143, mir-192-prec, mir-224, mir-126, mir-30a-5p, mir-140, mir-9, mir-124a-1, mir-218-2, mir-95, mir-145, mir-198, mir-216-prec, mir-219-1, mir-125a-prec, mir-26a-1-prec, mir-199b-prec, let-7a-2-prec, mir-27b, mir-32, mir-29b-2, mir-220, mir-33, mir-181c-prec, mir-101-1, mir-124a-3, mir-125a (DOWN)Lung cancer vs normalSolid (Not specified)[28]
mir-21, mir-191, mir-155, mir-210, mir-24-2 (UP) mir-126-5p, mir-126-3p, mir-219-1, mir-95, mir-192-prec, mir-220, mir-216-prec, mir-204-prec, mir-188, mir-198, mir-145, mir-224 (DOWN)ADs vs normalSolid (Not specified)[28]
mir-205, mir-191, mir-210, mir-17-3p, mir-203, mir-155, mir-21, mir-214, mir-212, mir-197 (UP) mir-224, mir-126*, mir-140, mir-29b, mir-143, mir-30a-5p (DOWN)SCC vs normalSolid (Not specified)[28]
miR-31 (UP) miR-107, miR-185, let-7a (DOWN)Lung cancer tissue vs normalSolid (Not specified)[70]
miR-26a, let-7g, let-7f, miR-98, miR-29a, let-7c, miR-30b, let-7i, let-7b, miR-29b, miR-26b, let-7a, miR-146b-5p, miR-195, miR-29c, miR-30d, miR-20a, miR-17, miR-19b, miR-106a, miR-16, let-7d, miR-106b, miR-181a, miR-498, miR-103, miR-107, miR-191, mir-663, miR-491-5p, let-7e, mir-654-5p (UP) miR-453, miR-509-3p (DOWN)AD vs SCC; male smokers patientsSolid, formalin-fixed, paraffin-embedded[71]
miR-17-5p, miR-20a, miR-20b, miR-93, miR-106a, miR-106b, miR-182, miR-183, miR-200a, miR-200c, miR-203, miR-210, miR-224 (UP) miR-125a, let7e (DOWN)SCC vs normalSolid, Snap-frozen[72]
miR-30a, miR-140-3p, miR-182, miR-210, miR-486-5pStage I-III vs normalSolid, Snap-frozen[73]
miR-182, miR-200c, miR-141, miR-375, miR-7, miR-429, miR-200a, miR-370, miR-200b, miR-382 (UP) miR-126, miR-451, miR-195, miR-486-5p, miR-214, miR-199a-5p (DOWN)Primary lung tumors vs metastasesSolid, formalin-fixed, paraffin-embedded[74]
miR-205, miR-21 (relative expression)AD vs SCCSolid, formalin-fixed, paraffin-embedded[75]
miR-21, miR-155 (UP)LCNECs and SCLCs vs TCs and ACsSolid, formalin-fixed, paraffin-embedded[79]
miR-205, miR-27a, miR-29a, miR-29b, miR-34a (DOWN in NSCLC) miR-25, miR-375 (UP in NSCLC)SCLC vs NSCLCSolid, formalin-fixed, paraffin-embedded[80]
miR-29a, miR-29b, miR-34a, miR-375 (DOWN in SQ) miR-205, miR-25, miR-27a (UP in SQ)SCC vs ADSolid, formalin-fixed, paraffin-embedded[80]
miR-7, miR-21, miR-29b, miR-106a, miR-125a-5p, miR-129-3p, miR-205, miR-375 (relative expression)Carcinoid, SCLC, and squamous and nonsquamous NSCLCSolid, Fresh Biopsy[81]
miR-21, miR-155, miR-7 (UP)Tumor vs normalSolid, fine-needle aspirate (FNA)[82]
miR-21, miR-155 (UP)NSCLC vs normalSputum[96]
miR-205, miR-210, miR-708 (relative expression)SCC vs normalSputum[97]
miR-21, miR-200b, miR-375 and miR-486 (relative expression)AD vs NormalSputum[98]
miR-31, miR-210 (relative expression)Stage I NSCLC vs normalSputum[99]
miR-31, miR-210 (Relative Expression) + computed tomographyStage I NSCLC vs normalSputum[100]

Comparison analyses between ADC vs noncancerous tissues and SCC vs noncancerous tissues revealed 17 and 16 miRNAs with statistically different expression, respectively (Table 1). Six miRNAs (miR-21, miR-155, miR-191, mir-126-5p, miR-210 and miR-224) were shared in both histological types of NSCLC. Yanaihara and colleagues also directly compared the two most common histological types of NSCLC, identifying two miRNAs (miR-99b and miR-102) that were higher in ADC and 4 miRNAs (miR-202, miR-203, miR-205 and the precursor of miR-204) that were higher in SCC. However, the authors do not explore further the issue of distinguishing ADCs from SCCs, in this paper.

Decreased expression of miR-107, miR-185 and let7a, and the overexpression of miR-31a, was observed by qRT-PCR in lung cancer tissues and cell lines, compared to normal lung tissue[70]. Landi et al[71] reported 34 miRNAs that significantly differentiated SCCs from ADs in male smoker patients, of which 2 were downregulated and 32 upregulated in AD vs SCC. Raponi et al[72] used Ambion microarrays to profile total RNA from 61 SCC samples and 10 matched normal lung samples and identified 15 miRNAs that were differentially expressed between normal lung and SCC (Table 1). Two of these miRNAs were down-regulated in SCCs (miR-125a and let7e) while the remaining 13 miRNAs were upregulated (miR-17-5p, miR-20a, miR-20b, miR-93, miR-106a, miR-106b, miR-182, miR-183, miR-200a, miR-200c, miR-203, miR-210, miR-224). More recently, a 5-miRNA classifier was identified by microarray analysis(miR-30a, miR-140-3p, miR-182, miR-210, miR-486-5p,) that could distinguish stage I-III SCC from normal lung tissues[73]. This classifier had an accuracy of 94,1% in a training cohort (34 patients) and 96,2% in a test cohort (26 patients).

A panel of 16 miRNAs has been reported to differentiate between primary lung tumors and metastases to the lung of various origin (Table 1)[74]. This miRNA profile was identified using microRNA microarray data generated from FFPE samples, and was confirmed by qRT-PCR. The panel includes miR-182, which was most strongly over-expressed in the lung primary tumors, and miR-126, which was over-expressed in the metastatic tumors.

Researchers have also aimed at finding one or few miRNAs that can be used as a convenient tool for lung cancers diagnosis. Lebanony et al[75] used a microarray to measure miRNA levels in AD and SCC FFPE samples, and verified their findings by qRT-PCR. They identified miR-205 as a highly specific marker for SCC, when combined with the measured miR21 levels. The finding was confirmed by other papers[76,77]. Moreover, an algorithm for accurate classification of NSCLC cases, diagnosed as LCC on purely morphologic grounds, was proposed by integrating immunohistochemical markers (Δnp63, DSC3, and napsin A) with miR-205 and miR-21 measurement[78].

Evaluating by qRT-PCR FPPE specimens from NETs, Lee et al[79] found that the levels of miR-21 and miR-155 were significantly higher in high-grade NET carcinomas (LCNECs and SCLCs) than in carcinoid tumors (TCs and ACs).

Two microRNA panels yielded high diagnostic accuracy in discriminating SCLC from NSCLC (miR-29a and miR-375) and in differentiating SCC from AD (miR-205 and miR-34a) in FFPE surgical lung specimens[80]. Moreover, the same microRNA panels accurately differentiated SCLC from NSCLC and SCC from AD in bronchial brushing specimens.

Gilad et al[81] reported a single assay for the classification of the four main types of lung cancer (Table 1): carcinoid, SCLC, and squamous and nonsquamous NSCLC, based on the expression of eight miRNAs (miR-7, miR-21, miR-29b, miR-106a, miR-125a-5p, miR-129-3p, miR-205, miR-375) The assay was effective both on resected and on cytologic (fine-needle aspiration (FNA) and bronchial brushing and washing) lung cancer samples.

A recent study has also evaluated miRNAs in FNA NSCLC biopsies[82]. miR-21, miR-155, and miR-7 showed a higher level in tumoral FNA than in normal FNA specimens, while let7a showed a lower level. A direct comparison of FNAs with resected specimens from the same patients indicated that the measured miRNAs had the same trend in the two types of specimens.

MiRNAs as biomarkers for lung cancers prognosis

Reduced expression of the let-7 family has been correlated with poor postoperative survival in NSCLC[26]. In a later study, AD patients with high expression of either mir-155, mir-17-3p, mir-106a, mir-93, or mir-21 and low expression of either let-7a-2, let-7b, or mir-145 were found to have a significantly worse prognosis (Table 2)[28]. Overexpression of the precursor of miR-155 and reduced expression of let-7-a was especially predictive of poor survival.

Table 2 MiRNAs as biomarkers for the prognosis and to predict response to therapy.
miRNAExperimentScopeSampleRef.
let-7a family (DOWN)lung cancer tissue vs normalNSCLC poor postoperative survivalSolid, (not specified)[26]
mir-155, mir-17-3p, mir-106a, mir-93, mir-21 (UP) let-7a-2, let-7b (DOWN)AD vs SCCAD poor survivalSolid, (not specified)[28]
miR-221, let-7a (DOWN) miR-137, miR-182-3p, miR-372 (UP)NSCLC vs normalNSCLC poor survivalSolid, snap frozen[83]
miR-146b, miR-191, miR-155, miR-15a, miR-511, miR-100, miR-10a, miR-21, miR-126 (UP) miR-206, miR-299-3p, miR-122a, miR-513, miR-184, miR-453, miR-379, miR-202, miR-494, miR-432, miR-370 (DOWN)SCC vs normalSCC Overall SurvivalSolid, snap frozen[72]
miR-34a (DOWN)NSCLC vs normalNSCLC Probability of relapseSolid, (not specified)[84]
mir-124-5p, mir-146b-3p, mir-200b-5p, mir-30c-1-3p, mir-510, mir-585, mir-630, mir-657, mir-708 (relative expression)stage I NSCLC vs normalStage I NSCLC recurrenceSolid, formalin-fixed, paraffin-embedded (FFPE)[64]
miR-16 (UP)NSCLC vs normalNSCLC poor survivalSolid, (not specified)[86]
miR-143 (DOWN)Lung cancer tissue vs normalsmoking statusSolid, snap frozen[88]
miR-21 (UP) miR-181a (DOWN)Lung cancer tissue vs normalNSCLC poor survivalSolid, snap frozen[88]
miR-25, miR-34c-5p, miR-191, let-7e, miR-34a (DOWN)AD vs SCCSCC survivalSolid, formalin-fixed, paraffin-embedded (FFPE)[71]
miR-31 (UP)SCC vs normalSCC poor survivalSolid, snap frozen[73]
miR-325, miR-326, miR-328, miR-329-2-pre, miR-330-3p, miR-500a-3p, miR-370, miR-650-pre (UP)BM − NSCLC vs BM + NSCLCNSCLC, risk for brain metastasisSolid, formalin-fixed, paraffin-embedded (FFPE)[89]
miR-21 (UP)Platinum-based chemotherapy-resistant NSCLC patientsSolid, snap frozen[90]
miR-196a-2 SNPTreatment vs genotypeSevere toxicity after platinum-based chemotherapy of advanced NSCLC patientsGenomic DNA from peripheral leukocytes[91]
miR-128b (UP)NSCLC samples correlated with clinical response and survival following gefitinib treatmentMicrodissected primary surgically resected NSCLC tumors[92]
miR-30b, miR-30cRegulated by EGFR and hepatocyte growth factor (MET) receptor tyrosine kinaseSolid, Lung tumor tissue samples[93]
miR-30b, miR-30cPrognostic predictors in NSCLC patients who underwent first line treatment with TKIsSolid, formalin-fixed, paraffin-embedded (FFPE)[94]

Analyzing frozen resected specimens from NSCLC patients, Yu et al[83] identified a five-microRNA signature that can predict the survival and relapse of patients with lung cancer (Table 2). Two of these miRNAs (miR-221 and let-7a) were protective (i.e., their down-regulation correlated with poor survival and high relapse probability), while the other three (miR-137, miR-182-3p, miR-372) were risky, and their up-regulation was predictive of poor survival and high relapse probability. The authors also demonstrated that miR-221, miR-137, miR-182-3p and miR-372 can alter the invasive ability of lung cancer cells in culture.

In the already described work by Raponi et al[72]. Twenty miRNAs were identified as having a significant association with overall survival in lung SCC patients (Table 2). Among these miRNAs, miR-146b alone was found to have the strongest prediction accuracy as the group with high miR-146b expression had significantly worse overall survival.

The p53-dependent miR-34 family was observed to be down-regulated in surgically resected NSCLC tumor samples compared with normal tissue, and low levels of miR-34a expression were correlated with a high probability of relapse[84]. MicroRNA expression profiles were also identified that may predict recurrence of localized stage I NSCLC after surgical resection[85]. These profiles included mir-124-5p, mir-146b-3p, mir-200b-5p, mir-30c-1-3p, mir-510, mir-585, mir-630, mir-657 and mir-708 (Table 2).

High miR-16 levels, measured by qRT-PCR in resected NSCLC samples, were reported as a prognostic factor for poor disease-free survival and poor overall survival[86]. Low miR-145 and high miR-367 are associated with shorter time to relapse (TTR) in resected NSCLC[87]. Noteworthy, p53 regulates miR-145 expression, which, in turn, inhibits the translation of SRY-related HMG box (SOX)2 and octamer-binding transcription factor (OCT)4. These transcription factors control the expression of the miR-302-367 cluster.

In a panel of 27 miRNAs which were observed by microarray analysis to be deregulated greater than two-fold in NSCLC resected samples compared to normal lung tissue, Gao et al[88] identified three miRNAs whose levels (confirmed by qRT-PCR) were related to clinico-pathologic characteristics or patient prognosis: low levels of miR-143 were significantly correlated with smoking status, high miR-21 expression and low miR-181a expression were associated with poor survival.

The lower expression level of a 5-miRNA signature (miR-25, miR-34c-5p, miR-191, let-7e, and miR-34a) correlated with poor overall survival among SCC patients (Table 2)[71] and high expression of miR-31 was associated with poor survival in Chinese SCC patients[73].

Eight miRNAs were confirmed to be significantly differentially expressed in NSCLC FFPE samples from patients with brain metastases compared with patients without brain metastases (Table 2)[89]. In particular, in this work, the over-expression of miR-328 and miR-330-3p was indicated as a marker for patients at risk for brain metastases, and a role for miR-328 in conferring migratory potential to NSCLC cells was suggested.

MiRNAs as biomarkers to predict response to therapy in lung cancer

miRNAs have also been used as biomarkers predictive of patient’s response to therapy. miR-21 expression was significantly increased in platinum-based chemotherapy-resistant NSCLC patients and increased miR-21 expression was associated with the shorter disease-free survival[90]. A single nucleotide polymorphism in miR-196a-2 gene was reported to be associated with severe toxicity after platinum-based chemotherapy of advanced NSCLC patients in a Chinese population[91].

Recent therapeutic advances for the treatment of NCLSC include the use of epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) including gefitinib and erlotinib. MiRNA-128b was shown to directly regulate EGFR translation and miR-128b LOH was found to be frequent in NSCLC samples and correlated significantly with clinical response and survival following gefitinib treatment[92]. miR-30b and miR-30c expression levels, which are regulated by EGFR and hepatocyte growth factor (MET) receptor tyrosine kinase[93] have been reported to be prognostic predictors in NSCLC patients who underwent first line treatment with TKIs[94].

MIRNAS IN SPUTUM AS NON-INVASIVE LUNG CANCER BIOMARKERS

An early diagnosis of cancer remains a challenge and, in this context, it is important to find a sensitive, non-invasive tool to detect early neoplastic changes. One relatively non-invasive source of miRNAs for the diagnosis of lung cancers is sputum[95]. The Jiang laboratory demonstrated that endogenous miRNAs are stably present in sputum specimens. Using qRT-PCR, miR-21 and miR-155 were detected, of which miR-21 was significantly overexpressed in sputum of NSCLC patients as compared with cancer-free subjects[96]. Furthermore, elevated miR-21 expression was more sensitive (70%) than conventional sputum cytology (48%) in diagnosing lung cancer. The same research group defined miRNA signatures for different histologic types of lung cancer in studies of similar design[97,98]. For the diagnosis of SCC, the combination of miR-205, miR-210 and miR-708 yielded 73% sensitivity and 96% specificity. A panel consisting of miR-21, miR-200b, miR-375 and miR-486 produced 81% sensitivity and 92% specificity in discriminating sputum of AD patients from controls. The authors found no association between miRNA expression and stage of lung cancer, suggesting that the miRNA signatures can be used as a tool in the detection of early lung cancer. The same group recently reported that combined quantification of miR-31 and miR-210 copy number by digital PCR in sputum of the cases and controls provided 65.71 % sensitivity and 85.00 % specificity for stage I NSCLC diagnosis[99]. Very recently, the same authors also reported that combining miR-31 and miR-210 detection by qRT-PCR and Computed Tomography they improved NSCLC diagnosis specificity[100].

In an independent study, a five-miRNA profile (miR-21, miR-143, miR-155, miR-210, miR-372) performed by qRT-PCR on sputum samples detected NSCLC with 83.3% sensitivity and 100% specificity[101].

CIRCULATING MIRNAS
First evidences

The finding that miRNAs have an exceptional stability in several tissues suggested that these tiny molecules were also preserved, detectable and quantifiable in the circulation and in other biofluids.

The first indication of circulating microRNAs with a potential as non-invasive diagnostic biomarkers for diffuse large B-cell lymphoma (DLBCL) and possibly other cancers was found by Lawrie et al[102]. These authors were the first to highlight that miRNAs could be reliably detected in serum and they demonstrated that high levels of miR-21, miR-210 and miR-155 could discriminate cancer patients with (DLBCL) from healthy individuals. Plasma miRNAs were observed to be present in a remarkably stable form that is protected from endogenous RNase activity[103]. These papers established the measurement of tumor-derived miRNAs in serum or plasma as an important approach for the blood-based detection of human cancer. Further studies demonstrated that microRNAs are also preserved and detectable in other biofluids such as urine, saliva, cerebro-spinal fluid and amniotic fluid[104] and their composition and concentrations are measurably different among these fluids[105].

On the origin of circulating miRNAs

miRNAs can be released in the circulation by two different pathways: energy-free passive release or active and selective secretion in response to different stimuli. The first process does not need energy, it occurs after cell breakage in pathological conditions such as tissue damage, apoptosis, metastasis or inflammation but it does not play a major role in the generation of circulating miRNAs[106]. miRNAs active secretion, differently from the passive leakage, is a process ATP- and temperature-dependent. It is similar to the release of hormones and cytokines, with or without cell stimulation[106].

Circulating miRNAs can be packaged in microvesicles (MVs) or apoptotic bodies (ABs) or can be found as microvesicle-free miRNAs, associated with various multiprotein or lipoprotein complexes.

MVs are small vesicles derived from cells, generally including microparticles (MPs) and exosomes. These two kinds of MVs have quite different vesicular structures: exosomes (50-90 nm), with an endocytic origin, are released by fusion of multivesicular bodies (MVBs) with the plasma membrane[107]. They have been identified in various body fluids such as blood, urine, malignant ascites, bronchoalveolar lavage fluid, synovial fluid, breast milk and saliva[108]. MPs, on the other hand, are larger than exosomes (> 100 nm diameter) and are shed from plasma membranes[109]. Almost all cell types can release MVs under normal physiological or pathological conditions.

Larger in size than MPs, ABs are generated in response to apoptotic stimuli[110] and implicated in tissue repair and angiogenesis.

It has been demonstrated that MVs and ABs are involved in the transport of circulating miRNAs[111-114]. Gibbings et al[115] showed that miRNAs loading into exosomes is not a random event, but it is mediated by proteins as Argonaute protein Ago2, a part of RNA-induced silencing complex.

On the other hand, several studies suggested that a significant fraction of extracellular miRNAs resides outside of vesicles and acts in exosome-independent manner, by its association with RNA-binding proteins including Ago2 and Nucleophosmin 1 (NPM1) or lipoprotein complexes such as high-density lipoprotein (HDL). Arroyo et al[116] showed that miRNAs in plasma are predominantly free of exosomes or microvesicles. They demonstrated an association of miRNAs with Ago2 protein, showing that this binding protects and increases the stability of released miRNAs[116]. Turchinovich et al[117] confirmed this hypothesis and demonstrated that also additional Argonaute proteins (Ago1, -3, -4) may be associated with cell-free circulating miRNAs. Wang et al[118] found that other RNA binding proteins, such as NPM1, can protect miRNAs from degradation, playing a role in the packaging and export of circulating miRNAs. Vickers et al[119] in 2011 revealed a potential new role for HDL in gene regulation and intercellular communication, showing that this lipoprotein transports and delivers miRNAs to recipient cells.

Circulating miRNAs as cancer biomarkers

It has been shown that miRNAs present in body fluids can reflect altered physiological conditions, representing new effective biomarkers[104].

A perfect biomarker should have some important features: non-invasivity, specificity, early detection, sensitivity and ease of translatability from model systems to humans.

Proteins used as blood biomarkers (e.g., troponin for cardiovascular conditions, carcinoembryonic antigen (CAE) for various cancers, prostate specific antigen (PSA) for prostate cancer, and aminotransferases for liver function) do not respect all these criteria and are difficult to use in this field. On the contrary, secreted miRNAs have many of these requisites: they are stable in various biofluids, the expression of some miRNAs is specific to tissues or biological stages, and their level can be easily detected by various methods.

However, several challenges have to be overcome in order to successfully usew circulating miRNAs as cancer biomarkers. First, biofluids contain very low amount of RNA, and normal quantification methods are not suitable for these type of samples. Second, it is important to avoid cellular contamination and hemolysis and third, biofluids contain inhibitors of reverse transcriptase and polymerase enzymes used for miRNAs quantification. All these factors are obstacles to consider when circulating miRNAs are isolated and quantified from biofluids such as plasma or serum. Another major challenge for the analysis of circulating miRNAs is the choice of an appropriate reference gene, since some of the small RNA species frequently used as reference genes (such as U6 RNA) are present in extremely low concentrations in serum and plasma as well as in other biofluids. Moreover, normalization controls used to remove variations and increase the accuracy of miRNAs quantification cannot ensure constant expression under all experimental conditions, underlining the importance of the selection of a proper reference gene.

Chen et al[120] reported that U6 and 5S rRNA are degraded in serum samples from lung cancer patients, and miR-16 is inconsistent, choosing to normalize the level of circulating miRNAs to total RNA. In a study on sera from Hepatitis B infected patients and matched controls, snRNA U6-1 is found to have high variability and snRNA U6-2 is not detectable. In this type of samples the combination of miR-26a, miR-221, and miR-22* is recommended as the most stable set of reference genes for circulating miRNAs evaluation[121]. Similarly, U6-2 is inconsistent in serum from gastric cancer patients and healthy controls. In this study, authors recommend the combined use of miR-16 and miR-93 as suitable reference genes[122]. On the contrary, in serum from uro-oncological patients, U6-2 is detectable and rather consistent[123]. snoRNA U44 levels are similar in sera from breast cancer patients and from age-matched healthy women, differently from miR-16 and 5S rRNS that show remarkable variability in the same samples. Surprisingly, snRNA U6-1 serum levels are found consistently higher in breast cancer patients compared to healthy controls, not only confirming that U6 is not an appropriate reference gene, but also indicating an interesting new paradigm in cancer[124]. Finally, Sourvinou and colleagues showed that a combined use of endogenous miR-21 and miR-16 and exogenous cel-miR-39, compensates differences in miRNAs recovery and differences in cDNA synthesis between samples. Using this normalization procedure and miR-21 as a biomarker, it seems possible to clearly discriminate healthy individuals from NSCLC patients[125].

Circulating miRNAs as lung cancer biomarkers

The Jiang laboratory found that miR-155, miR-197 and miR-182 can be potential biomarkers for early detection of lung cancer with 81.33% sensitivity and 86.76% specificity (Table 3). The levels of these miRNAs in plasma of NSCLC patients are elevated compared with healthy controls[126]. The same group demonstrated that another set of plasma miRNAs (miR-21, miR-126, miR-210, and miR-486-5p), had 86.22% sensitivity and 96.55% specificity in distinguishing NSCLC patients from the healthy controls (Table 3). Furthermore, the panel of four miRNAs produced 73.33% sensitivity and 96.55% specificity in identifying stage I NSCLC patients. The miR panel had higher sensitivity (91.67%) in diagnosis of AD compared with SCC (82.35%)[127]. Authors from the Jiang lab recently reported that quantification of the plasma miR-21-5p and miR-335-3p by digital PCR provided 71.8% sensitivity and 80.6% specificity in distinguishing lung cancer patients from cancer-free subjects (Table 3)[128].

Table 3 Circulating miRNAs as biomarkers in lung cancer.
MiRNAFunctionScopeSampleRef.
miR-155, miR-197, miR-182 (UP)DiagnosticLung cancer patients vs healthy controlsPlasma[126]
miR-21, miR-210, miR-126, miR-486-5p (relative expression)DiagnosticNSCLC patients vs healthy controlsPlasma[127]
miR-21–5p (UP) and miR-335–3p (DOWN)DiagnosticLung cancer patients vs healthy controlsPlasma[128]
miR-21, miR-155 (UP), miR-145 (DOWN)DiagnosticLung cancer patient vs healthy smokersPlasma[129]
miR-361-3p, miR-625* (DOWN)DiagnosticLung cancer patients vs healthy controlsSerum[130]
miR-146b, miR-221, let-7a, miR-155, miR-17-5p, miR-27a and miR-106a (DOWN), miR-29c (UP)DiagnosticEarly stage NCSLC vs healthy controlsSerum[131]
miR-92a, miR-484, miR-486-5p, miR-328, miR-191, miR-376a, miR-342, miR-331-3p, miR-30c, miR-28-5p, miR-98, miR-17-5p, miR-26b, miR-374, miR-30b, miR-26a, miR-142-3p, miR-103, miR-126, let-7a, let-7d, let-7b, miR-22, miR-148b, miR-139 (DOWN), miR-32, miR-133b, miR-566, miR-432-3p, miR-223, miR-29a, miR-148a, miR-142-5p, miR-140-5p (UP)DiagnosticAsymptomatic NSCLC patients vs healthy smokersSerum[132]
miR-205-5p, miR-205-3p, and miR-21-3p (UP)DiagnosticNSCLC patients vs benign lung disease and healthy controlsSerum[133]
miR-190b, miR-630, miR-942 and miR-1284 (relative expression)DiagnosticLung cancer patients vs healthy controlsWhole-blood[85]
miR-22, miR-24, and miR-34a (UP)DiagnosticNSCLC patients vs healthy controlsWhole-blood[134]
miR-205, miR-19a, miR-19b, miR-30b, miR-20a (DOWN)DiagnosticPatients after lung cancer surgery vs healthy controlsPlasma[135]
miR-7, miR-21, miR-200b, miR-210, miR-219-1, miR-324 (UP), miR-126, miR-451, miR-30a, miR-486 (DOWN)DiagnosticNSCLC patients vs healthy controlsPlasma[137]
miR-101, miR-106a miR-126, miR-133a, miR-140-3p, miR-140-5p, miR-142-3p, miR-145, miR-148a, miR-15b, miR-16, miR-17, miR-197, miR-19b, miR-21, miR-221, miR-28-3p, miR-30b, miR-30c, miR-320, miR-451, miR-486-5p, miR-660, and miR-92a (relative expression)DiagnosticNSCLC patients vs healthy controlsPlasma[138]
miR-155, miR-197 (UP)PrognosticLung cancer patients with metastasis vs patients without metastasisPlasma[126]
miR-486, miR-30d, miR-1, miR-499 (relative expression)PrognosticNSCLC patients vs healthy controlsSerum[139]
let-7f, miR-30e-3p (DOWN)PrognosticNSCLC patients vs healthy controlsPlasma[140]
miR-125b (relative expression)PrognosticNSCLC patients vs healthy controlsSerum[141]
miR-21 (UP)Response to treatmentPlatinum chemotherapy-resistant patients vs non resistant patientsPlasma[90]
miR-21 and miR-10b (UP)Response to treatmentNSCLC patients with EGFR mutation vs patients without mutationPlasma[142]
miR-22 (UP)Response to treatmentNSCLC patients vs healthy controlsWhole-blood[134]

Tang et al[129] reported that higher plasma miR-21 and miR-155 and lower plasma miR-145 expression levels distinguish lung cancer patients from healthy smokers with 69.4% sensitivity and 78.3% specificity (Table 3). Levels of miR-361-3p and miR-625-3p might have a protective influence on the development of NSCLC, and the quantification of these miRNAs in serum could be useful for the diagnosis of NSCLC, in particular in smokers[130]. A study reported that the expression of miR-146b, miR-221, let-7a, miR-155, miR-17-5p, miR-27a and miR-106a is significantly reduced in sera of NSCLC cases, while miR-29c is significantly increased (Table 3). Unexpectedly, no significant differences were observed in plasma of patients compared with controls[131].

Bianchi et al[132] provided an evidence that some serum-circulating miRNAs are important to identify asymptomathic high-risk individuals with early stage lung cancer (Table 3). Between others, they highlighted the importance of let-7 family, members of miR-17-92 cluster, miR-126 and miR-486 in sera of NSCLC patients[132].

Recently, qRT-PCR was used to assess miR-205-5p, miR-205-3p, and miR-21-3p expressions in serum and tissue samples (Table 3)[133]. The relative expressions of miR-205-5p and miR-205-3p were significantly higher in NSCLC tissues compared with cancer-adjacent paired specimens. In the serum, significantly higher miR-205-5p, miR-205-3p, and miR-21-3p relative expressions were observed in the NSCLC group compared with healthy volunteers or patients diagnosed with a benign lung disease (pulmonary tuberculosis, pneumonia, chronic obstructive pulmonary disease, or interstitial pneumonia). The relative expressions of miR-205-5p and miR-21-3p in NSCLC tissues and serum were significantly correlated, while no significant correlation was observed for miR-205-3p. Expressions of miR-205-5p and miR-205-3p in SCC specimens were significantly higher than in lung adenocarcinoma specimens. Similarly, higher serum miR-205-5p and miR-205-3p levels were observed in SCC patients.

MiRNAs expression profile in whole-blood showed that miR-190b, miR-630, miR-942 and miR-1284 are present in a majority of the classifiers generated during the analyses to distinguish lung cancer cases from controls[64]. In a different study, miR-22, miR-24, and miR-34a were found upregulated in RNA extracted from whole blood of NSCLC patients vs healthy controls (Table 3)[134].

In a recent paper, Aushev et al[135] described a specific panel of miRNAs (miR-205, -19a, -19b, -30b, and -20a) decreasing in plasma of patients after SCC surgery (Table 3). Interestingly, high levels of these miRNA are found in tumor-specific exosomes[135].

Also NGS has been used to depict the differential expression of miRNAs in peripheral blood of lung cancer patients detecting 76 previously unknown miRNAs and 41 novel mature forms of known precursors. In addition, the authors identified 32 annotated and seven unknown miRNAs that were significantly altered in NSCLC patients[136].

A plasma-based 24-miRNA signature classifier with predictive, diagnostic, and prognostic value was described, whose use could reduce the false-positive rate of low-dose computed tomography (LDCT), thus improving the efficacy of lung cancer screening (Table 3)[137,138].

Regarding the potential of circulating miRNAs as prognostic factors, the levels of miR-155 and miR-197 have been found higher in plasma from lung cancer patients with metastasis than in those without metastasis (Table 3)[126].

Moreover, Hu et al[139] using NGS described that serum levels of miR-486, miR-30d, miR-1 and miR-499 are significantly associated with overall survival (Table 3). NSCLC patients and healthy controls differ in vesicle-related miRNAs in plasma: let-7f and miR-30e-3p levels decreased in plasma vesicles of NSCLC patients and the expression of these miRNAs is associated with poor outcome (Table 3)[140]. Finally, serum miR-125b may represent a biomarker in NSCLC with an independent prognostic potential for overall survival (Table 3)[141].

Circulating miRNAs have also been explored for their ability to predict response to treatment. miR-21 expression has trends similar in plasma and matched resected specimens and was significantly increased in platinum-based chemotherapy-resistant patients, in which increased miR-21 expression was associated with the shorter disease-free survival (Table 3)[90].

The expression of miR-21 and miR-10b was much higher in plasma samples of patients with NSCLCs with EGFR mutation than without mutation (Table 3)[142]. Patients who had up-regulated miR-21 expression had shorter overall survival, but a better response to gefitinib than patients who had low expression of the microRNA. Additionally, miR-10b is highly expressed in progressive disease compared with complete remission or stable disease.

Franchina et al[134] recently reported a correlation between high expression of miR-22 in whole blood and the lack of response in pemetrexed treated NSCLC patients (Table 3).

CONCLUSION

MiRNAs have increasingly been pointed as important players in carcinogenesis and cancer progression, but also as potential diagnostic and prognostic markers.

For the future, circulating miRNAs could open new opportunities in the field of diagnosis and prognosis in various types of human cancers. The difficulties found in traditional therapies, due to insufficient disruption of oncogenic pathways, drug resistance and drug-induced toxicity, require the development of novel therapeutic strategies. The ease, specificity and sensitivity of determining body fluid miRNAs profiles paves the way for several applications and provides hope to accomplish this task. However, from the technical and applicative point of view, there are still several limitations to consider. Further studies are necessary to find the best possible normalization control and to improve the technique, but also to establish panels of miRNAs specific to each type of tumor, taking into account early or advanced cancer stages, response to treatment, patient outcome and recurrence. After the complete validation for several candidates, miRNAs studies could open a new era in cancer treatments, providing improved targeted agents for the cure of patients[143] and constituting the basis for the development of novel therapies.

Footnotes

P- Reviewer: Wu DF, Vetvicka V S- Editor: Song XX L- Editor: A E- Editor: Lu YJ

References
1.  Ferlay J, Parkin DM, Steliarova-Foucher E. Estimates of cancer incidence and mortality in Europe in 2008. Eur J Cancer. 2010;46:765-781.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1579]  [Cited by in F6Publishing: 1596]  [Article Influence: 114.0]  [Reference Citation Analysis (0)]
2.  Malvezzi M, Bertuccio P, Levi F, La Vecchia C, Negri E. European cancer mortality predictions for the year 2013. Ann Oncol. 2013;24:792-800.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 233]  [Cited by in F6Publishing: 252]  [Article Influence: 22.9]  [Reference Citation Analysis (0)]
3.  Rossi G, Mengoli MC, Cavazza A, Nicoli D, Barbareschi M, Cantaloni C, Papotti M, Tironi A, Graziano P, Paci M. Large cell carcinoma of the lung: clinically oriented classification integrating immunohistochemistry and molecular biology. Virchows Arch. 2014;464:61-68.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 77]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
4.  Scagliotti GV, Selvaggi G. New data integrating multitargeted antifolates into treatment of first-line and relapsed non-small-cell lung cancer. Clin Lung Cancer. 2008;9 Suppl 3:S122-S128.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 9]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
5.  Hanna N, Shepherd FA, Fossella FV, Pereira JR, De Marinis F, von Pawel J, Gatzemeier U, Tsao TC, Pless M, Muller T. Randomized phase III trial of pemetrexed versus docetaxel in patients with non-small-cell lung cancer previously treated with chemotherapy. J Clin Oncol. 2004;22:1589-1597.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1861]  [Cited by in F6Publishing: 2016]  [Article Influence: 100.8]  [Reference Citation Analysis (0)]
6.  Ciuleanu T, Brodowicz T, Zielinski C, Kim JH, Krzakowski M, Laack E, Wu YL, Bover I, Begbie S, Tzekova V. Maintenance pemetrexed plus best supportive care versus placebo plus best supportive care for non-small-cell lung cancer: a randomised, double-blind, phase 3 study. Lancet. 2009;374:1432-1440.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 843]  [Cited by in F6Publishing: 831]  [Article Influence: 55.4]  [Reference Citation Analysis (0)]
7.  Ardizzoni A, Boni L, Tiseo M, Fossella FV, Schiller JH, Paesmans M, Radosavljevic D, Paccagnella A, Zatloukal P, Mazzanti P, Bisset D, Rosell R; CISCA (CISplatin versus CArboplatin) Meta-analysis Group. Cisplatin- versus carboplatin-based chemotherapy in first-line treatment of advanced non-small-cell lung cancer: an individual patient data meta-analysis. J Natl Cancer Inst. 2007;99:847-57.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 445]  [Cited by in F6Publishing: 440]  [Article Influence: 25.9]  [Reference Citation Analysis (0)]
8.  Sandler AB, Schiller JH, Gray R, Dimery I, Brahmer J, Samant M, Wang LI, Johnson DH. Retrospective evaluation of the clinical and radiographic risk factors associated with severe pulmonary hemorrhage in first-line advanced, unresectable non-small-cell lung cancer treated with Carboplatin and Paclitaxel plus bevacizumab. J Clin Oncol. 2009;27:1405-1412.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
9.  Rekhtman N, Brandt SM, Sigel CS, Friedlander MA, Riely GJ, Travis WD, Zakowski MF, Moreira AL. Suitability of thoracic cytology for new therapeutic paradigms in non-small cell lung carcinoma: high accuracy of tumor subtyping and feasibility of EGFR and KRAS molecular testing. J Thorac Oncol. 2011;6:451-458.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
10.  Nizzoli R, Tiseo M, Gelsomino F, Bartolotti M, Majori M, Ferrari L, De Filippo M, Rindi G, Silini EM, Guazzi A. Accuracy of fine needle aspiration cytology in the pathological typing of non-small cell lung cancer. J Thorac Oncol. 2011;6:489-493.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 78]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
11.  Travis WD, Rekhtman N. Pathological diagnosis and classification of lung cancer in small biopsies and cytology: strategic management of tissue for molecular testing. Semin Respir Crit Care Med. 2011;32:22-31.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 102]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
12.  Rossi G, Pelosi G, Barbareschi M, Graziano P, Cavazza A, Papotti M. Subtyping non-small cell lung cancer: relevant issues and operative recommendations for the best pathology practice. Int J Surg Pathol. 2013;21:326-336.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 28]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
13.  Lytle JR, Yario TA, Steitz JA. Target mRNAs are repressed as efficiently by microRNA-binding sites in the 5’ UTR as in the 3’ UTR. Proc Natl Acad Sci USA. 2007;104:9667-9672.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 820]  [Cited by in F6Publishing: 859]  [Article Influence: 50.5]  [Reference Citation Analysis (0)]
14.  Calin GA, Dumitru CD, Shimizu M, Bichi R, Zupo S, Noch E, Aldler H, Rattan S, Keating M, Rai K. Frequent deletions and down-regulation of micro- RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci USA. 2002;99:15524-15529.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3675]  [Cited by in F6Publishing: 3636]  [Article Influence: 165.3]  [Reference Citation Analysis (0)]
15.  He L, Thomson JM, Hemann MT, Hernando-Monge E, Mu D, Goodson S, Powers S, Cordon-Cardo C, Lowe SW, Hannon GJ. A microRNA polycistron as a potential human oncogene. Nature. 2005;435:828-833.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2729]  [Cited by in F6Publishing: 2776]  [Article Influence: 146.1]  [Reference Citation Analysis (0)]
16.  Farazi TA, Spitzer JI, Morozov P, Tuschl T. miRNAs in human cancer. J Pathol. 2011;223:102-115.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 686]  [Cited by in F6Publishing: 746]  [Article Influence: 57.4]  [Reference Citation Analysis (0)]
17.  Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, Sweet-Cordero A, Ebert BL, Mak RH, Ferrando AA. MicroRNA expression profiles classify human cancers. Nature. 2005;435:834-838.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7124]  [Cited by in F6Publishing: 7177]  [Article Influence: 377.7]  [Reference Citation Analysis (0)]
18.  Volinia S, Calin GA, Liu CG, Ambs S, Cimmino A, Petrocca F, Visone R, Iorio M, Roldo C, Ferracin M. A microRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci USA. 2006;103:2257-2261.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4162]  [Cited by in F6Publishing: 4420]  [Article Influence: 245.6]  [Reference Citation Analysis (0)]
19.  Calin GA, Croce CM. MicroRNA signatures in human cancers. Nat Rev Cancer. 2006;6:857-866.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5705]  [Cited by in F6Publishing: 5849]  [Article Influence: 324.9]  [Reference Citation Analysis (0)]
20.  Rosenfeld N, Aharonov R, Meiri E, Rosenwald S, Spector Y, Zepeniuk M, Benjamin H, Shabes N, Tabak S, Levy A. MicroRNAs accurately identify cancer tissue origin. Nat Biotechnol. 2008;26:462-469.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 728]  [Cited by in F6Publishing: 698]  [Article Influence: 43.6]  [Reference Citation Analysis (0)]
21.  Rosenwald S, Gilad S, Benjamin S, Lebanony D, Dromi N, Faerman A, Benjamin H, Tamir R, Ezagouri M, Goren E. Validation of a microRNA-based qRT-PCR test for accurate identification of tumor tissue origin. Mod Pathol. 2010;23:814-823.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 95]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
22.  Meiri E, Mueller WC, Rosenwald S, Zepeniuk M, Klinke E, Edmonston TB, Werner M, Lass U, Barshack I, Feinmesser M. A second-generation microRNA-based assay for diagnosing tumor tissue origin. Oncologist. 2012;17:801-812.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 106]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
23.  Pentheroudakis G, Pavlidis N, Fountzilas G, Krikelis D, Goussia A, Stoyianni A, Sanden M, St Cyr B, Yerushalmi N, Benjamin H. Novel microRNA-based assay demonstrates 92% agreement with diagnosis based on clinicopathologic and management data in a cohort of patients with carcinoma of unknown primary. Mol Cancer. 2013;12:57.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 38]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
24.  Ferracin M, Pedriali M, Veronese A, Zagatti B, Gafà R, Magri E, Lunardi M, Munerato G, Querzoli G, Maestri I. MicroRNA profiling for the identification of cancers with unknown primary tissue-of-origin. J Pathol. 2011;225:43-53.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 106]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
25.  Calin GA, Sevignani C, Dumitru CD, Hyslop T, Noch E, Yendamuri S, Shimizu M, Rattan S, Bullrich F, Negrini M. Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers. Proc Natl Acad Sci USA. 2004;101:2999-3004.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2908]  [Cited by in F6Publishing: 3027]  [Article Influence: 151.4]  [Reference Citation Analysis (0)]
26.  Takamizawa J, Konishi H, Yanagisawa K, Tomida S, Osada H, Endoh H, Harano T, Yatabe Y, Nagino M, Nimura Y. Reduced expression of the let-7 microRNAs in human lung cancers in association with shortened postoperative survival. Cancer Res. 2004;64:3753-3756.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1813]  [Cited by in F6Publishing: 1812]  [Article Influence: 90.6]  [Reference Citation Analysis (0)]
27.  Johnson SM, Grosshans H, Shingara J, Byrom M, Jarvis R, Cheng A, Labourier E, Reinert KL, Brown D, Slack FJ. RAS is regulated by the let-7 microRNA family. Cell. 2005;120:635-647.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2664]  [Cited by in F6Publishing: 2651]  [Article Influence: 139.5]  [Reference Citation Analysis (0)]
28.  Yanaihara N, Caplen N, Bowman E, Seike M, Kumamoto K, Yi M, Stephens RM, Okamoto A, Yokota J, Tanaka T. Unique microRNA molecular profiles in lung cancer diagnosis and prognosis. Cancer Cell. 2006;9:189-198.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2314]  [Cited by in F6Publishing: 2327]  [Article Influence: 129.3]  [Reference Citation Analysis (0)]
29.  Inamura K, Togashi Y, Nomura K, Ninomiya H, Hiramatsu M, Satoh Y, Okumura S, Nakagawa K, Ishikawa Y. let-7 microRNA expression is reduced in bronchioloalveolar carcinoma, a non-invasive carcinoma, and is not correlated with prognosis. Lung Cancer. 2007;58:392-396.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 63]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
30.  Kumar MS, Erkeland SJ, Pester RE, Chen CY, Ebert MS, Sharp PA, Jacks T. Suppression of non-small cell lung tumor development by the let-7 microRNA family. Proc Natl Acad Sci USA. 2008;105:3903-3908.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 627]  [Cited by in F6Publishing: 656]  [Article Influence: 41.0]  [Reference Citation Analysis (0)]
31.  Chin LJ, Ratner E, Leng S, Zhai R, Nallur S, Babar I, Muller RU, Straka E, Su L, Burki EA. A SNP in a let-7 microRNA complementary site in the KRAS 3’ untranslated region increases non-small cell lung cancer risk. Cancer Res. 2008;68:8535-8540.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 475]  [Cited by in F6Publishing: 502]  [Article Influence: 31.4]  [Reference Citation Analysis (0)]
32.  Wang R, Wang ZX, Yang JS, Pan X, De W, Chen LB. MicroRNA-451 functions as a tumor suppressor in human non-small cell lung cancer by targeting ras-related protein 14 (RAB14). Oncogene. 2011;30:2644-2658.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 225]  [Cited by in F6Publishing: 245]  [Article Influence: 18.8]  [Reference Citation Analysis (0)]
33.  Hayashita Y, Osada H, Tatematsu Y, Yamada H, Yanagisawa K, Tomida S, Yatabe Y, Kawahara K, Sekido Y, Takahashi T. A polycistronic microRNA cluster, miR-17-92, is overexpressed in human lung cancers and enhances cell proliferation. Cancer Res. 2005;65:9628-9632.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1174]  [Cited by in F6Publishing: 1187]  [Article Influence: 62.5]  [Reference Citation Analysis (0)]
34.  Ventura A, Young AG, Winslow MM, Lintault L, Meissner A, Erkeland SJ, Newman J, Bronson RT, Crowley D, Stone JR. “Targeted deletion reveals essential and overlapping functions of the miR-17~92 family of miRNA clusters.”. Cell. 2008;132:875–86.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1236]  [Cited by in F6Publishing: 1268]  [Article Influence: 79.3]  [Reference Citation Analysis (1)]
35.  Takahashi T, Nau MM, Chiba I, Birrer MJ, Rosenberg RK, Vinocour M, Levitt M, Pass H, Gazdar AF, Minna JD. p53: a frequent target for genetic abnormalities in lung cancer. Science. 1989;246:491-494.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 792]  [Cited by in F6Publishing: 828]  [Article Influence: 23.7]  [Reference Citation Analysis (0)]
36.  Ding L, Getz G, Wheeler DA, Mardis ER, McLellan MD, Cibulskis K, Sougnez C, Greulich H, Muzny DM, Morgan MB. Somatic mutations affect key pathways in lung adenocarcinoma. Nature. 2008;455:1069-1075.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2058]  [Cited by in F6Publishing: 2027]  [Article Influence: 126.7]  [Reference Citation Analysis (0)]
37.  He L, He X, Lim LP, de Stanchina E, Xuan Z, Liang Y, Xue W, Zender L, Magnus J, Ridzon D. A microRNA component of the p53 tumour suppressor network. Nature. 2007;447:1130-1134.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2001]  [Cited by in F6Publishing: 2046]  [Article Influence: 120.4]  [Reference Citation Analysis (0)]
38.  Chang TC, Wentzel EA, Kent OA, Ramachandran K, Mullendore M, Lee KH, Feldmann G, Yamakuchi M, Ferlito M, Lowenstein CJ. Transactivation of miR-34a by p53 broadly influences gene expression and promotes apoptosis. Mol Cell. 2007;26:745-752.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1481]  [Cited by in F6Publishing: 1531]  [Article Influence: 90.1]  [Reference Citation Analysis (0)]
39.  Corney DC, Flesken-Nikitin A, Godwin AK, Wang W, Nikitin AY. MicroRNA-34b and MicroRNA-34c are targets of p53 and cooperate in control of cell proliferation and adhesion-independent growth. Cancer Res. 2007;67:8433-8438.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 500]  [Cited by in F6Publishing: 524]  [Article Influence: 30.8]  [Reference Citation Analysis (0)]
40.  Raver-Shapira N, Marciano E, Meiri E, Spector Y, Rosenfeld N, Moskovits N, Bentwich Z, Oren M. Transcriptional activation of miR-34a contributes to p53-mediated apoptosis. Mol Cell. 2007;26:731-743.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1014]  [Cited by in F6Publishing: 1007]  [Article Influence: 59.2]  [Reference Citation Analysis (0)]
41.  Jiang L, Huang Q, Chang J, Wang E, Qiu X. MicroRNA HSA-miR-125a-5p induces apoptosis by activating p53 in lung cancer cells. Exp Lung Res. 2011;37:387-398.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 70]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
42.  Bisio A, De Sanctis V, Del Vescovo V, Denti MA, Jegga AG, Inga A, Ciribilli Y. Identification of new p53 target microRNAs by bioinformatics and functional analysis. BMC Cancer. 2013;13:552.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 45]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
43.  Xi Y, Nakajima G, Gavin E, Morris CG, Kudo K, Hayashi K, Ju J. Systematic analysis of microRNA expression of RNA extracted from fresh frozen and formalin-fixed paraffin-embedded samples. RNA. 2007;13:1668-1674.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 444]  [Cited by in F6Publishing: 454]  [Article Influence: 26.7]  [Reference Citation Analysis (0)]
44.  Kim YK, Yeo J, Kim B, Ha M, Kim VN. Short structured RNAs with low GC content are selectively lost during extraction from a small number of cells. Mol Cell. 2012;46:893-895.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 160]  [Cited by in F6Publishing: 136]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
45.  Metzker ML. Sequencing technologies - the next generation. Nat Rev Genet. 2010;11:31-46.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4795]  [Cited by in F6Publishing: 3964]  [Article Influence: 264.3]  [Reference Citation Analysis (0)]
46.  Shen J, Stass SA, Jiang F. MicroRNAs as potential biomarkers in human solid tumors. Cancer Lett. 2013;329:125-136.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 168]  [Cited by in F6Publishing: 186]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
47.  Ach RA, Wang H, Curry B. Measuring microRNAs: comparisons of microarray and quantitative PCR measurements, and of different total RNA prep methods. BMC Biotechnol. 2008;8:69.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 133]  [Cited by in F6Publishing: 137]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
48.  Chen Y, Gelfond JA, McManus LM, Shireman PK. Reproducibility of quantitative RT-PCR array in miRNA expression profiling and comparison with microarray analysis. BMC Genomics. 2009;10:407.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 237]  [Cited by in F6Publishing: 232]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
49.  Willenbrock H, Salomon J, Søkilde R, Barken KB, Hansen TN, Nielsen FC, Møller S, Litman T. Quantitative miRNA expression analysis: comparing microarrays with next-generation sequencing. RNA. 2009;15:2028-2034.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 128]  [Cited by in F6Publishing: 104]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
50.  Sato F, Tsuchiya S, Terasawa K, Tsujimoto G. Intra-platform repeatability and inter-platform comparability of microRNA microarray technology. PLoS One. 2009;4:e5540.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 158]  [Cited by in F6Publishing: 167]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
51.  Git A, Dvinge H, Salmon-Divon M, Osborne M, Kutter C, Hadfield J, Bertone P, Caldas C. Systematic comparison of microarray profiling, real-time PCR, and next-generation sequencing technologies for measuring differential microRNA expression. RNA. 2010;16:991-1006.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 526]  [Cited by in F6Publishing: 503]  [Article Influence: 35.9]  [Reference Citation Analysis (0)]
52.  Sah S, McCall MN, Eveleigh D, Wilson M, Irizarry RA. Performance evaluation of commercial miRNA expression array platforms. BMC Res Notes. 2010;3:80.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 26]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
53.  Pradervand S, Weber J, Lemoine F, Consales F, Paillusson A, Dupasquier M, Thomas J, Richter H, Kaessmann H, Beaudoing E. Concordance among digital gene expression, microarrays, and qPCR when measuring differential expression of microRNAs. Biotechniques. 2010;48:219-222.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 80]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
54.  Yauk CL, Rowan-Carroll A, Stead JD, Williams A. Cross-platform analysis of global microRNA expression technologies. BMC Genomics. 2010;11:330.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 41]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
55.  Wang B, Howel P, Bruheim S, Ju J, Owen LB, Fodstad O, Xi Y. Systematic evaluation of three microRNA profiling platforms: microarray, beads array, and quantitative real-time PCR array. PLoS One. 2011;6:e17167.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 87]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
56.  Callari M, Dugo M, Musella V, Marchesi E, Chiorino G, Grand MM, Pierotti MA, Daidone MG, Canevari S, De Cecco L. Comparison of microarray platforms for measuring differential microRNA expression in paired normal/cancer colon tissues. PLoS One. 2012;7:e45105.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 49]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
57.  Leshkowitz D, Horn-Saban S, Parmet Y, Feldmesser E. Differences in microRNA detection levels are technology and sequence dependent. RNA. 2013;19:527-538.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 82]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
58.  Kolbert CP, Feddersen RM, Rakhshan F, Grill DE, Simon G, Middha S, Jang JS, Simon V, Schultz DA, Zschunke M. Multi-platform analysis of microRNA expression measurements in RNA from fresh frozen and FFPE tissues. PLoS One. 2013;8:e52517.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 81]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
59.  Del Vescovo V, Meier T, Inga A, Denti MA, Borlak J. A cross-platform comparison of affymetrix and Agilent microarrays reveals discordant miRNA expression in lung tumors of c-Raf transgenic mice. PLoS One. 2013;8:e78870.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 33]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
60.  Benes V, Castoldi M. Expression profiling of microRNA using real-time quantitative PCR, how to use it and what is available. Methods. 2010;50:244-249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 268]  [Cited by in F6Publishing: 249]  [Article Influence: 17.8]  [Reference Citation Analysis (0)]
61.  Kulshreshtha R, Ferracin M, Wojcik SE, Garzon R, Alder H, Agosto-Perez FJ, Davuluri R, Liu CG, Croce CM, Negrini M. A microRNA signature of hypoxia. Mol Cell Biol. 2007;27:1859-1867.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 743]  [Cited by in F6Publishing: 829]  [Article Influence: 46.1]  [Reference Citation Analysis (0)]
62.  Nadano D, Sato TA. Caspase-3-dependent and -independent degradation of 28 S ribosomal RNA may be involved in the inhibition of protein synthesis during apoptosis initiated by death receptor engagement. J Biol Chem. 2000;275:13967-13973.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 44]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
63.  Chan MW, Wei SH, Wen P, Wang Z, Matei DE, Liu JC, Liyanarachchi S, Brown R, Nephew KP, Yan PS. Hypermethylation of 18S and 28S ribosomal DNAs predicts progression-free survival in patients with ovarian cancer. Clin Cancer Res. 2005;11:7376-7383.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 58]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
64.  Patnaik SK, Kannisto E, Knudsen S, Yendamuri S. Evaluation of microRNA expression profiles that may predict recurrence of localized stage I non-small cell lung cancer after surgical resection. Cancer Res. 2010;70:36-45.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 180]  [Cited by in F6Publishing: 190]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
65.  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;14:844-852.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 621]  [Cited by in F6Publishing: 636]  [Article Influence: 39.8]  [Reference Citation Analysis (0)]
66.  Schaefer A, Jung M, Miller K, Lein M, Kristiansen G, Erbersdobler A, Jung K. Suitable reference genes for relative quantification of miRNA expression in prostate cancer. Exp Mol Med. 2010;42:749-758.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 92]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
67.  Wotschofsky Z, Meyer HA, Jung M, Fendler A, Wagner I, Stephan C, Busch J, Erbersdobler A, Disch AC, Mollenkopf HJ. Reference genes for the relative quantification of microRNAs in renal cell carcinomas and their metastases. Anal Biochem. 2011;417:233-241.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 72]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
68.  Gee HE, Buffa FM, Camps C, Ramachandran A, Leek R, Taylor M, Patil M, Sheldon H, Betts G, Homer J. The small-nucleolar RNAs commonly used for microRNA normalisation correlate with tumour pathology and prognosis. Br J Cancer. 2011;104:1168-1177.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 205]  [Cited by in F6Publishing: 220]  [Article Influence: 16.9]  [Reference Citation Analysis (0)]
69.  Hindson CM, Chevillet JR, Briggs HA, Gallichotte EN, Ruf IK, Hindson BJ, Vessella RL, Tewari M. Absolute quantification by droplet digital PCR versus analog real-time PCR. Nat Methods. 2013;10:1003-1005.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 927]  [Cited by in F6Publishing: 987]  [Article Influence: 89.7]  [Reference Citation Analysis (0)]
70.  Takahashi Y, Forrest AR, Maeno E, Hashimoto T, Daub CO, Yasuda J. MiR-107 and MiR-185 can induce cell cycle arrest in human non small cell lung cancer cell lines. PLoS One. 2009;4:e6677.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 150]  [Cited by in F6Publishing: 169]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
71.  Landi MT, Zhao Y, Rotunno M, Koshiol J, Liu H, Bergen AW, Rubagotti M, Goldstein AM, Linnoila I, Marincola FM. MicroRNA expression differentiates histology and predicts survival of lung cancer. Clin Cancer Res. 2010;16:430-441.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 248]  [Cited by in F6Publishing: 272]  [Article Influence: 19.4]  [Reference Citation Analysis (0)]
72.  Raponi M, Dossey L, Jatkoe T, Wu X, Chen G, Fan H, Beer DG. MicroRNA classifiers for predicting prognosis of squamous cell lung cancer. Cancer Res. 2009;69:5776-5783.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Tan X, Qin W, Zhang L, Hang J, Li B, Zhang C, Wan J, Zhou F, Shao K, Sun Y. A 5-microRNA signature for lung squamous cell carcinoma diagnosis and hsa-miR-31 for prognosis. Clin Cancer Res. 2011;17:6802-6811.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Barshack I, Lithwick-Yanai G, Afek A, Rosenblatt K, Tabibian-Keissar H, Zepeniuk M, Cohen L, Dan H, Zion O, Strenov Y. MicroRNA expression differentiates between primary lung tumors and metastases to the lung. Pathol Res Pract. 2010;206:578-584.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 53]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
75.  Lebanony D, Benjamin H, Gilad S, Ezagouri M, Dov A, Ashkenazi K, Gefen N, Izraeli S, Rechavi G, Pass H. Diagnostic assay based on hsa-miR-205 expression distinguishes squamous from nonsquamous non-small-cell lung carcinoma. J Clin Oncol. 2009;27:2030-2037.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 304]  [Cited by in F6Publishing: 310]  [Article Influence: 20.7]  [Reference Citation Analysis (0)]
76.  Bishop JA, Benjamin H, Cholakh H, Chajut A, Clark DP, Westra WH. Accurate classification of non-small cell lung carcinoma using a novel microRNA-based approach. Clin Cancer Res. 2010;16:610-619.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
77.  Del Vescovo V, Cantaloni C, Cucino A, Girlando S, Silvestri M, Bragantini E, Fasanella S, Cuorvo LV, Palma PD, Rossi G. miR-205 Expression levels in nonsmall cell lung cancer do not always distinguish adenocarcinomas from squamous cell carcinomas. Am J Surg Pathol. 2011;35:268-275.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 44]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
78.  Barbareschi M, Cantaloni C, Del Vescovo V, Cavazza A, Monica V, Carella R, Rossi G, Morelli L, Cucino A, Silvestri M. Heterogeneity of large cell carcinoma of the lung: an immunophenotypic and miRNA-based analysis. Am J Clin Pathol. 2011;136:773-782.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 28]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
79.  Lee HW, Lee EH, Ha SY, Lee CH, Chang HK, Chang S, Kwon KY, Hwang IS, Roh MS, Seo JW. Altered expression of microRNA miR-21, miR-155, and let-7a and their roles in pulmonary neuroendocrine tumors. Pathol Int. 2012;62:583-591.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 41]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
80.  Huang W, Hu J, Yang DW, Fan XT, Jin Y, Hou YY, Wang JP, Yuan YF, Tan YS, Zhu XZ. Two microRNA panels to discriminate three subtypes of lung carcinoma in bronchial brushing specimens. Am J Respir Crit Care Med. 2012;186:1160-1167.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 39]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
81.  Gilad S, Lithwick-Yanai G, Barshack I, Benjamin S, Krivitsky I, Edmonston TB, Bibbo M, Thurm C, Horowitz L, Huang Y. Classification of the four main types of lung cancer using a microRNA-based diagnostic assay. J Mol Diagn. 2012;14:510-517.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 86]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
82.  Petriella D, Galetta D, Rubini V, Savino E, Paradiso A, Simone G, Tommasi S. Molecular profiling of thin-prep FNA samples in assisting clinical management of non-small-cell lung cancer. Mol Biotechnol. 2013;54:913-919.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 19]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
83.  Yu SL, Chen HY, Chang GC, Chen CY, Chen HW, Singh S, Cheng CL, Yu CJ, Lee YC, Chen HS. MicroRNA signature predicts survival and relapse in lung cancer. Cancer Cell. 2008;13:48-57.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 616]  [Cited by in F6Publishing: 661]  [Article Influence: 41.3]  [Reference Citation Analysis (0)]
84.  Gallardo E, Navarro A, Viñolas N, Marrades RM, Diaz T, Gel B, Quera A, Bandres E, Garcia-Foncillas J, Ramirez J. miR-34a as a prognostic marker of relapse in surgically resected non-small-cell lung cancer. Carcinogenesis. 2009;30:1903-1909.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 257]  [Cited by in F6Publishing: 279]  [Article Influence: 18.6]  [Reference Citation Analysis (0)]
85.  Patnaik SK, Yendamuri S, Kannisto E, Kucharczuk JC, Singhal S, Vachani A. MicroRNA expression profiles of whole blood in lung adenocarcinoma. PLoS One. 2012;7:e46045.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 84]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
86.  Navarro A, Diaz T, Gallardo E, Viñolas N, Marrades RM, Gel B, Campayo M, Quera A, Bandres E, Garcia-Foncillas J. Prognostic implications of miR-16 expression levels in resected non-small-cell lung cancer. J Surg Oncol. 2011;103:411-415.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 50]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
87.  Campayo M, Navarro A, Viñolas N, Diaz T, Tejero R, Gimferrer JM, Molins L, Cabanas ML, Ramirez J, Monzo M. Low miR-145 and high miR-367 are associated with unfavourable prognosis in resected nonsmall cell lung cancer. Eur Respir J. 2013;41:1172-1178.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 59]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
88.  Gao W, Yu Y, Cao H, Shen H, Li X, Pan S, Shu Y. Deregulated expression of miR-21, miR-143 and miR-181a in non small cell lung cancer is related to clinicopathologic characteristics or patient prognosis. Biomed Pharmacother. 2010;64:399-408.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 187]  [Cited by in F6Publishing: 201]  [Article Influence: 14.4]  [Reference Citation Analysis (0)]
89.  Arora S, Ranade AR, Tran NL, Nasser S, Sridhar S, Korn RL, Ross JT, Dhruv H, Foss KM, Sibenaller Z. MicroRNA-328 is associated with (non-small) cell lung cancer (NSCLC) brain metastasis and mediates NSCLC migration. Int J Cancer. 2011;129:2621-2631.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
90.  Gao W, Lu X, Liu L, Xu J, Feng D, Shu Y. MiRNA-21: a biomarker predictive for platinum-based adjuvant chemotherapy response in patients with non-small cell lung cancer. Cancer Biol Ther. 2012;13:330-340.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 114]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
91.  Zhan X, Wu W, Han B, Gao G, Qiao R, Lv J, Zhang S, Zhang W, Fan W, Chen H. Hsa-miR-196a2 functional SNP is associated with severe toxicity after platinum-based chemotherapy of advanced nonsmall cell lung cancer patients in a Chinese population. J Clin Lab Anal. 2012;26:441-446.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 21]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
92.  Weiss GJ, Bemis LT, Nakajima E, Sugita M, Birks DK, Robinson WA, Varella-Garcia M, Bunn PA, Haney J, Helfrich BA. EGFR regulation by microRNA in lung cancer: correlation with clinical response and survival to gefitinib and EGFR expression in cell lines. Ann Oncol. 2008;19:1053-1059.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 172]  [Cited by in F6Publishing: 185]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
93.  Garofalo M, Romano G, Di Leva G, Nuovo G, Jeon YJ, Ngankeu A, Sun J, Lovat F, Alder H, Condorelli G. EGFR and MET receptor tyrosine kinase-altered microRNA expression induces tumorigenesis and gefitinib resistance in lung cancers. Nat Med. 2012;18:74-82.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 296]  [Cited by in F6Publishing: 324]  [Article Influence: 24.9]  [Reference Citation Analysis (0)]
94.  Gu YF, Zhang H, Su D, Mo ML, Song P, Zhang F, Zhang SC. miR-30b and miR-30c expression predicted response to tyrosine kinase inhibitors as first line treatment in non-small cell lung cancer. Chin Med J (Engl). 2013;126:4435-4439.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 3]  [Reference Citation Analysis (0)]
95.  Hubers AJ, Prinsen CF, Sozzi G, Witte BI, Thunnissen E. Molecular sputum analysis for the diagnosis of lung cancer. Br J Cancer. 2013;109:530-537.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 78]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
96.  Xie Y, Todd NW, Liu Z, Zhan M, Fang H, Peng H, Alattar M, Deepak J, Stass SA, Jiang F. Altered miRNA expression in sputum for diagnosis of non-small cell lung cancer. Lung Cancer. 2010;67:170-176.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 221]  [Cited by in F6Publishing: 248]  [Article Influence: 16.5]  [Reference Citation Analysis (0)]
97.  Xing L, Todd NW, Yu L, Fang H, Jiang F. Early detection of squamous cell lung cancer in sputum by a panel of microRNA markers. Mod Pathol. 2010;23:1157-1164.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 194]  [Cited by in F6Publishing: 206]  [Article Influence: 14.7]  [Reference Citation Analysis (0)]
98.  Yu L, Todd NW, Xing L, Xie Y, Zhang H, Liu Z, Fang H, Zhang J, Katz RL, Jiang F. Early detection of lung adenocarcinoma in sputum by a panel of microRNA markers. Int J Cancer. 2010;127:2870-2878.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 260]  [Cited by in F6Publishing: 287]  [Article Influence: 22.1]  [Reference Citation Analysis (0)]
99.  Li N, Ma J, Guarnera MA, Fang H, Cai L, Jiang F. Digital PCR quantification of miRNAs in sputum for diagnosis of lung cancer. J Cancer Res Clin Oncol. 2014;140:145-150.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 76]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
100.  Shen J, Liao J, Guarnera MA, Fang H, Cai L, Stass SA, Jiang F. Analysis of MicroRNAs in sputum to improve computed tomography for lung cancer diagnosis. J Thorac Oncol. 2014;9:33-40.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 76]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
101.  Roa WH, Kim JO, Razzak R, Du H, Guo L, Singh R, Gazala S, Ghosh S, Wong E, Joy AA. Sputum microRNA profiling: a novel approach for the early detection of non-small cell lung cancer. Clin Invest Med. 2012;35:E271.  [PubMed]  [DOI]  [Cited in This Article: ]
102.  Lawrie CH, Gal S, Dunlop HM, Pushkaran B, Liggins AP, Pulford K, Banham AH, Pezzella F, Boultwood J, Wainscoat JS. Detection of elevated levels of tumour-associated microRNAs in serum of patients with diffuse large B-cell lymphoma. Br J Haematol. 2008;141:672-675.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1258]  [Cited by in F6Publishing: 1278]  [Article Influence: 79.9]  [Reference Citation Analysis (0)]
103.  Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-Agadjanyan EL, Peterson A, Noteboom J, O’Briant KC, Allen A. Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci USA. 2008;105:10513-10518.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5636]  [Cited by in F6Publishing: 6041]  [Article Influence: 377.6]  [Reference Citation Analysis (0)]
104.  Gilad S, Meiri E, Yogev Y, Benjamin S, Lebanony D, Yerushalmi N, Benjamin H, Kushnir M, Cholakh H, Melamed N. Serum microRNAs are promising novel biomarkers. PLoS One. 2008;3:e3148.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 996]  [Cited by in F6Publishing: 1042]  [Article Influence: 65.1]  [Reference Citation Analysis (0)]
105.  Weber JA, Baxter DH, Zhang S, Huang DY, Huang KH, Lee MJ, Galas DJ, Wang K. The microRNA spectrum in 12 body fluids. Clin Chem. 2010;56:1733-1741.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1810]  [Cited by in F6Publishing: 1937]  [Article Influence: 138.4]  [Reference Citation Analysis (0)]
106.  Zen K, Zhang CY. Circulating microRNAs: a novel class of biomarkers to diagnose and monitor human cancers. Med Res Rev. 2012;32:326-348.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 337]  [Cited by in F6Publishing: 355]  [Article Influence: 25.4]  [Reference Citation Analysis (0)]
107.  Théry C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis and function. Nat Rev Immunol. 2002;2:569-579.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3512]  [Cited by in F6Publishing: 3746]  [Article Influence: 170.3]  [Reference Citation Analysis (0)]
108.  Simpson RJ, Lim JW, Moritz RL, Mathivanan S. Exosomes: proteomic insights and diagnostic potential. Expert Rev Proteomics. 2009;6:267-283.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 736]  [Cited by in F6Publishing: 788]  [Article Influence: 52.5]  [Reference Citation Analysis (0)]
109.  Boulanger CM, Amabile N, Tedgui A. Circulating microparticles: a potential prognostic marker for atherosclerotic vascular disease. Hypertension. 2006;48:180-186.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 275]  [Cited by in F6Publishing: 281]  [Article Influence: 15.6]  [Reference Citation Analysis (0)]
110.  Beyer C, Pisetsky DS. The role of microparticles in the pathogenesis of rheumatic diseases. Nat Rev Rheumatol. 2010;6:21-29.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 197]  [Cited by in F6Publishing: 202]  [Article Influence: 13.5]  [Reference Citation Analysis (0)]
111.  Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ, Lötvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007;9:654-659.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8246]  [Cited by in F6Publishing: 9018]  [Article Influence: 530.5]  [Reference Citation Analysis (0)]
112.  Hunter MP, Ismail N, Zhang X, Aguda BD, Lee EJ, Yu L, Xiao T, Schafer J, Lee ML, Schmittgen TD. Detection of microRNA expression in human peripheral blood microvesicles. PLoS One. 2008;3:e3694.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1078]  [Cited by in F6Publishing: 1094]  [Article Influence: 68.4]  [Reference Citation Analysis (0)]
113.  Zernecke A, Bidzhekov K, Noels H, Shagdarsuren E, Gan L, Denecke B, Hristov M, Köppel T, Jahantigh MN, Lutgens E. Delivery of microRNA-126 by apoptotic bodies induces CXCL12-dependent vascular protection. Sci Signal. 2009;2:ra81.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 959]  [Cited by in F6Publishing: 1005]  [Article Influence: 67.0]  [Reference Citation Analysis (0)]
114.  Zampetaki A, Willeit P, Drozdov I, Kiechl S, Mayr M. Profiling of circulating microRNAs: from single biomarkers to re-wired networks. Cardiovasc Res. 2012;93:555-562.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 194]  [Cited by in F6Publishing: 198]  [Article Influence: 15.2]  [Reference Citation Analysis (0)]
115.  Gibbings DJ, Ciaudo C, Erhardt M, Voinnet O. Multivesicular bodies associate with components of miRNA effector complexes and modulate miRNA activity. Nat Cell Biol. 2009;11:1143-1149.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 741]  [Cited by in F6Publishing: 762]  [Article Influence: 50.8]  [Reference Citation Analysis (0)]
116.  Arroyo JD, Chevillet JR, Kroh EM, Ruf IK, Pritchard CC, Gibson DF, Mitchell PS, Bennett CF, Pogosova-Agadjanyan EL, Stirewalt DL. Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma. Proc Natl Acad Sci USA. 2011;108:5003-5008.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2345]  [Cited by in F6Publishing: 2488]  [Article Influence: 191.4]  [Reference Citation Analysis (0)]
117.  Turchinovich A, Weiz L, Langheinz A, Burwinkel B. Characterization of extracellular circulating microRNA. Nucleic Acids Res. 2011;39:7223-7233.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1356]  [Cited by in F6Publishing: 1426]  [Article Influence: 109.7]  [Reference Citation Analysis (0)]
118.  Wang K, Zhang S, Weber J, Baxter D, Galas DJ. Export of microRNAs and microRNA-protective protein by mammalian cells. Nucleic Acids Res. 2010;38:7248-7259.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 750]  [Cited by in F6Publishing: 776]  [Article Influence: 55.4]  [Reference Citation Analysis (0)]
119.  Vickers KC, Palmisano BT, Shoucri BM, Shamburek RD, Remaley AT. MicroRNAs are transported in plasma and delivered to recipient cells by high-density lipoproteins. Nat Cell Biol. 2011;13:423-433.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1953]  [Cited by in F6Publishing: 2061]  [Article Influence: 158.5]  [Reference Citation Analysis (0)]
120.  Chen X, Ba Y, Ma L, Cai X, Yin Y, Wang K, Guo J, Zhang Y, Chen J, Guo X. Characterization of microRNAs in serum: a novel class of biomarkers for diagnosis of cancer and other diseases. Cell Res. 2008;18:997-1006.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3218]  [Cited by in F6Publishing: 3380]  [Article Influence: 211.3]  [Reference Citation Analysis (0)]
121.  Zhu X, Li H, Long L, Hui L, Chen H, Wang X, Shen H, Xu W. miR-126 enhances the sensitivity of non-small cell lung cancer cells to anticancer agents by targeting vascular endothelial growth factor A. Acta Biochim Biophys Sin (Shanghai). 2012;44:519-526.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 92]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
122.  Song J, Bai Z, Han W, Zhang J, Meng H, Bi J, Ma X, Han S, Zhang Z. Identification of suitable reference genes for qPCR analysis of serum microRNA in gastric cancer patients. Dig Dis Sci. 2012;57:897-904.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 204]  [Cited by in F6Publishing: 212]  [Article Influence: 17.7]  [Reference Citation Analysis (0)]
123.  Sanders I, Holdenrieder S, Walgenbach-Brünagel G, von Ruecker A, Kristiansen G, Müller SC, Ellinger J. 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;19:1017-1025.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 78]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
124.  Appaiah HN, Goswami CP, Mina LA, Badve S, Sledge GW, Liu Y, Nakshatri H. Persistent upregulation of U6: SNORD44 small RNA ratio in the serum of breast cancer patients. Breast Cancer Res. 2011;13:R86.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 76]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
125.  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;15:827-834.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 157]  [Cited by in F6Publishing: 170]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
126.  Zheng D, Haddadin S, Wang Y, Gu LQ, Perry MC, Freter CE, Wang MX. Plasma microRNAs as novel biomarkers for early detection of lung cancer. Int J Clin Exp Pathol. 2011;4:575-586.  [PubMed]  [DOI]  [Cited in This Article: ]
127.  Shen J, Todd NW, Zhang H, Yu L, Lingxiao X, Mei Y, Guarnera M, Liao J, Chou A, Lu CL. Plasma microRNAs as potential biomarkers for non-small-cell lung cancer. Lab Invest. 2011;91:579-587.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 286]  [Cited by in F6Publishing: 308]  [Article Influence: 23.7]  [Reference Citation Analysis (0)]
128.  Ma J, Li N, Guarnera M, Jiang F. Quantification of Plasma miRNAs by Digital PCR for Cancer Diagnosis. Biomark Insights. 2013;8:127-136.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 79]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
129.  Tang D, Shen Y, Wang M, Yang R, Wang Z, Sui A, Jiao W, Wang Y. Identification of plasma microRNAs as novel noninvasive biomarkers for early detection of lung cancer. Eur J Cancer Prev. 2013;22:540-548.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 95]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
130.  Roth C, Stuckrath I, Pantel K, Izbicki JR, Tachezy M, Schwarzenbach H. Low levels of cell-free circulating miR-361-3p and miR-625* as blood-based markers for discriminating malignant from benign lung tumors. PloS One. 2012;7:e38248.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 63]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
131.  Heegaard NH, Schetter AJ, Welsh JA, Yoneda M, Bowman ED, Harris CC. Circulating micro-RNA expression profiles in early stage nonsmall cell lung cancer. Int J Cancer. 2012;130:1378-1386.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 200]  [Cited by in F6Publishing: 230]  [Article Influence: 17.7]  [Reference Citation Analysis (0)]
132.  Bianchi F, Nicassio F, Marzi M, Belloni E, Dall’olio V, Bernard L, Pelosi G, Maisonneuve P, Veronesi G, Di Fiore PP. A serum circulating miRNA diagnostic test to identify asymptomatic high-risk individuals with early stage lung cancer. EMBO Mol Med. 2011;3:495-503.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 259]  [Cited by in F6Publishing: 274]  [Article Influence: 21.1]  [Reference Citation Analysis (0)]
133.  Jiang M, Zhang P, Hu G, Xiao Z, Xu F, Zhong T, Huang F, Kuang H, Zhang W. Relative expressions of miR-205-5p, miR-205-3p, and miR-21 in tissues and serum of non-small cell lung cancer patients. Mol Cell Biochem. 2013;383:67-75.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 42]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
134.  Franchina T, Amodeo V, Bronte G, Savio G, Ricciardi GR, Picciotto M, Russo A, Giordano A, Adamo V. Circulating miR-22, miR-24 and miR-34a as novel predictive biomarkers to pemetrexed-based chemotherapy in advanced non-small cell lung cancer. J Cell Physiol. 2014;229:97-99.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 52]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
135.  Aushev VN, Zborovskaya IB, Laktionov KK, Girard N, Cros MP, Herceg Z, Krutovskikh V. Comparisons of microRNA patterns in plasma before and after tumor removal reveal new biomarkers of lung squamous cell carcinoma. PLoS One. 2013;8:e78649.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 102]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
136.  Keller A, Backes C, Leidinger P, Kefer N, Boisguerin V, Barbacioru C, Vogel B, Matzas M, Huwer H, Katus HA. Next-generation sequencing identifies novel microRNAs in peripheral blood of lung cancer patients. Mol Biosyst. 2011;7:3187-3199.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 54]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
137.  Boeri M, Verri C, Conte D, Roz L, Modena P, Facchinetti F, Calabrò E, Croce CM, Pastorino U, Sozzi G. MicroRNA signatures in tissues and plasma predict development and prognosis of computed tomography detected lung cancer. Proc Natl Acad Sci USA. 2011;108:3713-3718.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 543]  [Cited by in F6Publishing: 549]  [Article Influence: 42.2]  [Reference Citation Analysis (0)]
138.  Sozzi G, Boeri M, Rossi M, Verri C, Suatoni P, Bravi F, Roz L, Conte D, Grassi M, Sverzellati N. Clinical utility of a plasma-based miRNA signature classifier within computed tomography lung cancer screening: a correlative MILD trial study. J Clin Oncol. 2014;32:768-773.  [PubMed]  [DOI]  [Cited in This Article: ]
139.  Hu Z, Chen X, Zhao Y, Tian T, Jin G, Shu Y, Chen Y, Xu L, Zen K, Zhang C. Serum microRNA signatures identified in a genome-wide serum microRNA expression profiling predict survival of non-small-cell lung cancer. J Clin Oncol. 2010;28:1721-1726.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 591]  [Cited by in F6Publishing: 625]  [Article Influence: 44.6]  [Reference Citation Analysis (0)]
140.  Silva J, García V, Zaballos Á, Provencio M, Lombardía L, Almonacid L, García JM, Domínguez G, Peña C, Diaz R. Vesicle-related microRNAs in plasma of nonsmall cell lung cancer patients and correlation with survival. Eur Respir J. 2011;37:617-623.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 203]  [Cited by in F6Publishing: 222]  [Article Influence: 15.9]  [Reference Citation Analysis (0)]
141.  Yuxia M, Zhennan T, Wei Z. Circulating miR-125b is a novel biomarker for screening non-small-cell lung cancer and predicts poor prognosis. J Cancer Res Clin Oncol. 2012;138:2045-2050.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 85]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
142.  Shen Y, Tang D, Yao R, Wang M, Wang Y, Yao Y, Li X, Zhang H. microRNA expression profiles associated with survival, disease progression, and response to gefitinib in completely resected non-small-cell lung cancer with EGFR mutation. Med Oncol. 2013;30:750.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 41]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
143.  Bader AG, Brown D, Stoudemire J, Lammers P. Developing therapeutic microRNAs for cancer. Gene Ther. 2011;18:1121-1126.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 222]  [Cited by in F6Publishing: 241]  [Article Influence: 18.5]  [Reference Citation Analysis (0)]