Skip to content
Publicly Available Published by De Gruyter December 19, 2016

Non-coding RNAs: the cancer genome dark matter that matters!

  • Hui Ling EMAIL logo , Leonard Girnita , Octavian Buda and George A. Calin EMAIL logo

Abstract

Protein-coding genes comprise only 3% of the human genome, while the genes that are transcribed into RNAs but do not code for proteins occupy majority of the genome. Once considered as biological darker matter, non-coding RNAs are now being recognized as critical regulators in cancer genome. Among the many types of non-coding RNAs, microRNAs approximately 20 nucleotides in length are best characterized and their mechanisms of action are well generalized. microRNA exerts oncogenic or tumor suppressor function by regulation of protein-coding genes via sequence complementarity. The expression of microRNA is aberrantly regulated in all cancer types, and both academia and biotech companies have been keenly pursuing the potential of microRNA as cancer biomarker for early detection, prognosis, and therapeutic response. The key involvement of microRNAs in cancer also prompted interest on exploration of therapeutic values of microRNAs as anticancer drugs and drug targets. MRX34, a liposome-formulated miRNA-34 mimic, developed by Mirna Therapeutics, becomes the first microRNA therapeutic entering clinical trial for the treatment of hepatocellular carcinoma, renal cell carcinoma, and melanoma. In this review, we presented a general overview of microRNAs in cancer biology, the potential of microRNAs as cancer biomarkers and therapeutic targets, and associated challenges.

Introduction

As stated in the central dogma of molecular biology, RNA is an essential component in the flow of genetic information from DNA to protein [1]. Because of the recognized importance of proteins in exerting biological functions, RNAs had been regarded for a long time as merely a mediator in passing genetic codes to final functional protein molecules. As a consequence, the functional activities of RNAs themselves are largely neglected [2]. Recent advances on the sequencing technologies revealed that vast majority of the human genome are transcribed, while the protein-coding genes only occupy 3% of the human genome [3]. The widespread transcription of the genome into non-protein-coding RNAs strongly suggest that RNAs process functions other than mere mediators between DNAs and proteins. Indeed, RNA have been suggested to be the earliest molecule of life and thought to possess both informational and catalytic function [4]. Emerging evidences from the last two decades have unambiguously proved the functional importance of noncoding RNAs in human biology and diseases [5].

Ribosome RNAs and RNAs are among the early-discovered non-coding RNA transcripts. However, because of their roles in facilitating protein translation, they are still considered part of the machinery translating genomic code into protein synthesis. The discovery of microRNAs in early 1990s [6], [7] opened a new chapter of gene regulation by non-coding RNAs and stimulated the scientific interest onto the many RNA molecules that do not code for proteins [8]. Numerous other types of small non-coding RNAs have also been subsequently identified, including endogenous small interfering RNAs (endo-siRNAs), PIWI-associated small RNAs (piRNAs), small nucleolar RNAs (snoRNAs), sno-derived RNAs (sdRNAs), transcription initiation RNAs (tiRNAs), miRNA-offset RNAs (moRNAs), and others [5]. Long non-coding RNAs, defined as non-coding transcripts longer than 200 nucleotides, also emerge as important regulator in a wide range of biological activities and human disease [9]. A systematic classification of long non-coding RNA is missing; currently, long non-coding RNAs are categorized as, according to their genomic localization or other features, natural antisense transcripts, long intergenic non-coding RNAs, transcribed ultraconserved regions (T-UCRs), circular RNAs, enhancer-associated RNAs, promoter-associated RNAs, and others [5].

In this review, we focus on the well-characterized microRNAs by providing a general overview on their production, function, regulatory mechanisms, and their clinical importance as cancer biomarkers and cancer therapeutics. Due to the space limit of this review, we only present the essential and relevant literatures and have to skip many excellent works.

What are microRNAs?

In 1993, Victor Ambros’s research group and Gray Ruvkun’s research team independently reported that a small RNA transcript around 20 nucleotides, known as lin-4, regulates developmental programming of Caenorhabditis elegans by posttranscriptional regulation of the heterochronic gene lin-14 via an antisense RNA-RNA interaction [6], [7]. These reports mark the first discovery of microRNAs, a new type of non-coding RNA with unique regulation mechanism. In 2000, the second microRNA let-7 was discovered from C. elegans [10]. Until now, the miRBase database (version 19) cataloged over 25,000 mature microRNA sequences in 193 species, including 2042 mature microRNAs in human beings.

Mature microRNAs are single-stranded non-coding RNAs with 19–24 nucleotides that are usually evolutionarily conserved [11]. The biogenesis of microRNA is a multi-step event [11] (Figure 1). First, microRNA gene is transcribed by RNA polymerase II into primary microRNA (pri-miRNA) of various lengths (usually 1000–3000 nucleotides). Second, the pri-miRNA is cleaved by the ribonuclease (RNase) III Drosha-DGCR8 nuclear complex into hairpin RNA of 60–100 nucleotides in length named precursor miRNA (pre-miRNA). Third, pre-miRNA is transported from the nucleus to cytoplasm by exportin-5, and further cleaved by the RNase III enzyme Dicer into imperfect microRNA duplex about 22 nucleotides in length. Finally, the microRNA duplex unwinds and the mature single-stranded microRNA incorporates into the RNA-induced silencing complex (RISC). Through Watson-Crick complementarities between the seed sequence of microRNA (positions 2–8) and the 3′ untranslated region (UTR) of its target messenger RNAs (mRNAs), microRNA recognizes and recruits mRNAs onto RISC. The resulting microRNA-mRNA interaction prevents mRNA translation or enhances RNA degradation in either case, altering protein output [1].

Figure 1: microRNA biogenesis and action mechanisms.microRNA genes are initially transcribed as primary microRNAs (pri-miRNAs), which are then processed by Drosha/DGCR8 complex into hairpin-structure microRNA precursors (pre-miRNAs). The precursors are transported from nucleus into cytosol, where Dicer further cleaves them into short microRNA duplex. The duplex unwinds, and the single-stranded mature microRNA loads onto the RNA-induced silencing complex to exert posttranscriptional regulation on target genes. The interaction of microRNA with 3′ untranslated region (UTR) of target messenger RNA based on sequence complementarity is considered as the canonical mechanism. Targeting other regions such as coding sequence or 5′UTR has also been reported.
Figure 1:

microRNA biogenesis and action mechanisms.

microRNA genes are initially transcribed as primary microRNAs (pri-miRNAs), which are then processed by Drosha/DGCR8 complex into hairpin-structure microRNA precursors (pre-miRNAs). The precursors are transported from nucleus into cytosol, where Dicer further cleaves them into short microRNA duplex. The duplex unwinds, and the single-stranded mature microRNA loads onto the RNA-induced silencing complex to exert posttranscriptional regulation on target genes. The interaction of microRNA with 3′ untranslated region (UTR) of target messenger RNA based on sequence complementarity is considered as the canonical mechanism. Targeting other regions such as coding sequence or 5′UTR has also been reported.

Comparing with other types of novel non-coding RNAs, functioning mechanisms of microRNAs are well characterized. In the canonical way, microRNAs binds to 3′-untranslated region of the target mRNA based on sequence complementarity and regulates protein output via promoting mRNA degradation or interfering with the translation process [11]. The sequence complementarity does not need to be perfect; the seed sequence, in most cases situated at positions 2–7 from the 5′ end of microRNA, determines the target identification. As such, one microRNA could target tens or hundreds of mRNA candidates, and consequently affects various types of physiological activities. For example, miR-15a and miR-16 regulates B-cell survival; miR-181 regulates B-cell lineage fate; miR-125b and let-7 are involved in cell proliferation; miR-430 controls brain patterning; miR-375 regulates insulin secretion in pancreatic cells; and miR-143 regulates adipocyte development [12]. Of note, although the canonical mechanism applies to most cases of regulation by microRNAs, multiple non-canonical mechanisms have been reported. For instance, the fact that miR-29b is localized in nucleus challenged the canonical mechanism, which could only explains interaction occurring in the cytoplasm [13]; some microRNAs such as miR-10a also bind to 5′-untranslated region or gene body to modulate target gene expression [14]; miR-373 interacts with CDH1 promoter and up-regulates its transcription [15]; miR-369-3p interacts with AU-rich elements in TNF mRNA and recruits the protein complex composed of Ago2 and FXR1 to promote TNF translation during cell cycle arrest [16]; circulating miR-21 and miR-29a act as agonists of Toll-like receptors (TLRs) in the endosome of the recipient cells and modulate secretion of inflammatory cytokines [17].

microRNAs and human cancer

Despite the discovery of microRNAs lin-4 and let-7 in Caenorhabditis elegans in 1993 and 2000, respectively, the importance of microRNAs in human disease was not recognized. In 2002, George Calin and Carlo Croce reported the first microRNA-cancer link, discovering that a microRNA cluster, miR-15a/16-1 is frequently lost in chronic lymphocytic leukemia (CLL) [18]. This finding spurs immediate interest in the scientific society. Following this pioneer study, the role of miR-15a/16-1 as tumor suppressor was proved, and several other tumor suppressor microRNAs including let-7 family and miR-34 were demonstrated [8]. These tumor suppressor microRNAs perform their function by reducing levels of oncogenic protein-coding genes. For instance, the miR-15a/16-1 targets the BCL2, an oncogene associated with apoptotic cell death; let-7 reduces the levels of oncogenes KRAS and MYC; miR-34 mediates p53 signaling by targeting several important oncogenes CDK4, MYC and MET [12].

Studies also revealed several oncogenic microRNAs including the miR17-miR-92 cluster, miR-21, and miR-155 [19]. The miR-17/miR-92 cluster contains several well-studied oncogenic microRNAs and is frequently activated in lymphoma, lung cancer, breast cancer, gastric cancer, colon cancer, and pancreatic cancer [20]. These microRNAs are direct MYC targets and they work synergistically with the MYC oncogene in promoting B cell lymphoma in mice [21]. As a typical oncogenic microRNA, miR-21 expression is upregulated in many cancer types [8]. Overexpression of miR-21 induces pre-B cell lymphoma in mouse model, stimulates lung carcinogenesis by activating the MAPK signaling, and promotes metastasis of colorectal cancers by regulating PDCD4 [22]. Similarly, miR-155 alone was shown to induce lymphoblastic leukemia or high-grade lymphoma in transgenic mice [23]. These studies suggest that microRNAs could be the driving force of carcinogenesis.

Further studies demonstrated the functional role of microRNAs in cancer metastasis. For instance, a panel of microRNAs including miR-9, miR-10b, miR-103, miR373, and miR-520c has been shown to promote cancer metastasis in mouse models [24]. microRNAs including miR-31, miR-34a, miR-126, miR-200, miR-206, and miR-335 were able to block cancer metastasis [24]. In our own study, we identified via screening that miR-224 is a promoter in metastasis of colorectal cancer [25]. Forced expression of miR-224 dramatically increased cancer metastasis from colon to distant organs of lung and liver in orthotopic mouse models. We identified SMAD4 as a direct miR-224 target, as well as a mediator of miR-224′s pro-metastatic effect [25]. Among the metastasis-associated microRNAs, miR-126 is interesting because of its unique effect in impairing metastatic colonization [26]. This was achieved by suppressing recruitment of endothelial cells in the tumor microenvironment into the metastatic site through its targets IGFBP2, PITPNC1, and MERTK [26].

However, the concept of microRNAs as tumor suppressors and oncogenes should not be stereotyped. Depending on the cellular contexts, the same microRNA could play oncogenic role in one tumor type and act as a tumor suppressor in other(s). For instance, miR-200 is considered as a suppressor of tumor invasion because of its inhibitory effect on epithelial-mesenchymal transition (EMT) by targeting ZEB1 and ZEB2 [27], [28]. On the other hand, miR-200 was shown to promote colonization of circulating cancer cells in the distant metastatic organs [29]. Therefore, miR-200 exhibits oncogenic or tumor suppressor function, in different metastatic steps. A single microRNA could also have distinct function in different cancer types. For example, miR-221 targets the KIT oncogene to inhibit inhibits erythroleukemic cell proliferation [30]. In the scenario of hepatocellular carcinoma, miR-221 was shown to regulate DDIT4, a DNA damage-inducible transcript, to promote liver carcinogenesis [31]. The functions of a miRNA should thus be carefully examined and determined in specific biological conditions.

The plethora of reports argue that microRNAs are master regulator of cancer biology, and deregulated expression of microRNAs affects all hallmarks of cancer [32] (Figure 2): (1) sustaining proliferative signaling (miR-21, let-7) [33], [34]; (2) evading growth suppressors (miR-221 and miR-222) [35]; (3) resisting cell death (miR-34a, miR-15a/16-1) [36], [37]; (4) enabling replicative immortality (miR-29, miR-19b) [38], [39]; (5) inducing angiogenesis (miR-210, miR-17-92 cluster) [40], [41]; (6) activating invasion and metastasis (miR-10b, miR-224) [25], [42]; (7) reprogramming energy metabolism (miR-23, miR-103) [43], [44]; (8) evading immune destruction (miR-34a, miR-124a) [45], [46].

Figure 2: microRNAs and cancer hallmarks.microRNAs are functionally associated with all aspects of cancer hallmarks. Typical examples of oncogenic or tumor suppressor microRNAs for each hallmark are shown.
Figure 2:

microRNAs and cancer hallmarks.

microRNAs are functionally associated with all aspects of cancer hallmarks. Typical examples of oncogenic or tumor suppressor microRNAs for each hallmark are shown.

Aberrant microRNA expression in human cancer

Following the initial discovery of low miR-15a/16-1 expression in CLL, frequent deregulation of microRNA expression in many cancer types has been demonstrated. There are several mechanisms underlying the aberrant expression of microRNA in cancer. First, as demonstrated initially, microRNAs located in the frequently altered genomic regions could lost their expression due to chromosomal deletion or increase their expression as result of chromosomal gains. By comprehensive mapping of microRNAs in human genome, it had been discovered that microRNA genes are frequently located at cancer associated fragile sites in the genome, i.e. the chromosomal regions prone to deletions or amplifications [47]. For instance, the 7 mb genomic loci of 3p21.1 – 21.2, where let-7g and miR-135-1 genes are located, is frequently deleted in lung and breast cancer; miR-145 and miR-143 genes at 5q32 is recurrently lost in myelodysplastic syndrome; miR-123 gene at 9q33 is frequently deleted in non-small cell lung cancer; miR-34a-1 and miR-34a-2 at 11q23-q24 are lost in breast and long cancers [47]. On the contrary, many oncogenic microRNAs are genomically amplified in human cancers: the miR-17-92 cluster genes at 13q32-33 are commonly amplified in follicular lymphoma; the miR-21 gene at 17q23 is amplified in several cancer types including neuroblastoma [47].

The expression levels of microRNA are also controlled by other mechanisms. For instance, the miR-34a is transcriptionally activated by p53, and the miR-17-92 cluster is transcriptionally regulated by the MYC oncogene [21], [36]. Epigenetic mechanisms such as CpG island methylation also play important roles in regulating microRNA expression. With a genetic knockout model lacking DNA methyltransferase activity, Lujambio et al. identified that miR-124 is silenced by promoter CpG island hypermethylation in human cancer [48]. This group further reported a DNA methylation signature comprising miR-148a, miR-34b/c, and miR-9 that is associated with the risk of human cancer metastasis [49]. Similarly, the miR-200 family and miR-205 genes, which are key regulators of EMT by targeting ZEB1 and ZEB2, are tightly controlled by promoter CpG islands [50]. Aberrant promoter methylation of these microRNA genes and consequent low microRNA expression are associated with lung cancer initiation and progression [51]. Additionally, defects in microRNA-processing machineries could significantly alter the expression levels of mature microRNAs. Indeed, downregulation of or mutations in microRNA-processing genes including Dicer, TRBP, and Exportin have been observed in many cancer types, and affect a wide range of microRNA expression [52]. While one expects that the general effect of such defects lead to reduced microRNA processing, nucleolin, an element of the Drosha-DGCR8 complex, was shown to promote the processing of several pro-metastatic miRNAs, including miR-221/222, miR-21, and miR-103 [53].

In certain situations, multiple mechanisms work together to regulate a specific microRNA expression. In the case of miR-34a, it is located at 1p36, a frequently lost genomic region in neuroblastoma, transcriptionally regulated by p53 and MYC, and silenced via CpG methylation. The high levels of miR-17-92 cluster microRNAs in cancer are not only related to copy number gains, but also due to transcriptional activation by MYC.

Competitive endogenous RNAs (ceRNAs), which act as microRNA sponges through their microRNA binding sites, could de-repress target genes of the microRNA according to the ceRNA hypothesis [54]. This represents another layer of microRNA regulation in human cancer, with changes in the function but not expression of a microRNA. The debate on the physiological relevance of ceRNA concept is summarized in a recent excellent review [55].

microRNAs and growth factors signaling

As tumor initiation and progression are tightly controlled by growth factors and their intracellular signaling [56], [57], [58], it is not surprising that growth factors regulate microRNA expression while microRNAs orchestrate the expression and function of proteins within growth factor signaling pathways [59]. Over the last decade this bi-directional regulatory networks between microRNAs and signaling complexes coordinated by growth factors receptors have received particular attention revealing some conserved modules [60]. Among RTKs, the IGF and EGF system of ligands, receptors and binding proteins are major player in normal cellular growth and differentiation, as well as in cancer development [60], [61], [62], [63]. Both IGF and EGF systems are organized on three distinct levels: (i) the input layer of ligands, receptors and regulatory proteins of ligand-receptor interaction; (ii) the signal transduction layer, coordinated by adaptors and enzymes of the signaling cascades and the (iii) output layer of transcription factors, controlling the biological response. The receptors bind to their respective ligands with by far the greatest affinity, activating the signaling cascade by two-steps ligand induced dimerization and kinase activation (EGFR) or by inducing conformational changes of the preformed dimers (IGF-1R and IR) [60], [61]. Growth factor signaling pathways in general, including the ones activated by IGF-1 and EGFs are involved in microRNA processing [59], [60]. EGF stimulation of normal mammary epithelial cells dramatically modified microRNA expression profile in less than 1 h [64], [65]. Some of the identified microRNAs were demonstrated to control a defined malignant behavior, for instance miR-15b, upregulated by EGF stimulation, and miR-191, downregulated by EGF, were shown to positively regulates the metastatic phenotype (for extensive review see [60]). Similarly, in the case of IGF-1R stimulation, specific microRNA profiles were found to be associated with signaling activation as related to the malignant phenotype [66], [67]. In breast cancer cell lines, 18-h stimulation with IGF-1 lead to modified microRNA profile with increase of defined oncogenic microRNAs (103/107, 1826, 191, 93) and downregulation of tumor suppressive microRNAs (15b, 98, 195, 200b, let-7c, and let-7g) [67].

On the other hand, the microRNA network regulates the expression of IGF-1, their receptors as well as their regulatory proteins or effectors [68]. For instance miR-486, an IGF-1 inhibitor at transcriptional level was found to be decreased in NSCLC cancer cell lines [69]. In addition several other microRNAs including Let-7, miR-16, miR-122, or miR-196 were demonstrated to control the expression of the IGF-1R in different cancer types (for an extensive review see [68]). Not only the expression of the ligands and receptors are controlled by the microRNA network but also the downstream signaling proteins such as insulin-receptor substrate (IRSs) [70], β-arrestins [71], [72], the IGF-1R ubiquitination enzymes [73], [74], or the components of the P3K/Akt and MAPK pathways [68].

Potential use of microRNA as cancer biomarkers

microRNAs have several advantages as cancer biomarkers [75]. First, microRNAs are stable under harsh environments such as high temperatures, long-term storage, strong acidic or basic conditions, and frequent freeze-thaw. This feature makes possible the detection of microRNA expression from formalin fixed paraffin embedded (FFPE) samples, plasma, serum, urine, and other body fluids. Second, the detection of mature microRNA by either PCR-based method or hybridization-based method is straightforward. Third, the tissue-specific expression pattern of microRNAs makes them also ideal cancer biomarkers.

Cancer of unknown primary origin (CUP) represents around 3%–5% of all newly diagnosed cancers in the United States and remains an unmet medical challenge in cancer diagnosis [76]. The feature of tissue-specific of some microRNAs offers an opportunity to characterize the site and type of cancer origin. Early in 2005, The Golub lab reported that microRNA pattern is more accurate in establishing the right diagnosis than the mRNA criteria, in poorly differentiated metastatic cancer cases with CUP [77]. A large effort of microRNA profiling with 400 FFPE and fresh-frozen samples from Barshack group constructed a microRNA classifier composing 48 microRNAs that can accurately identify origin of cancer tissues [78]. The tissue-specific feature also allows for determination of histological subtypes. For instance, miR-205 has been reported as an accurate marker for squamous non-small cell lung cancer, with high sensitivity (96%) and specificity (90%) [79]. Because of the high performance of microRNA biomarkers in differentiating cancer subtypes, Biotech companies have started developing diagnostic products. Using 8 microRNA biomarkers, mi-LUNG (Rosetta Genomics Ltd.) accurately differentiates the four subtypes of lung cancer (small-cell lung cancer, carcinoid, squamous non-small cell lung cancer, and non-squamous NSCLC) with 94% sensitivity and 98% specificity [80].

Several advantages of microRNAs, including stability and easy detection, allow for potential use of microRNAs in body fluids as convenient and less invasive biomarkers for early cancer detection. Boeri et al. reported that microRNA signatures in plasma are early predictor of lung cancer occurrence, months before the diagnosis by computed tomography [81]. With a large set of lung cancer cohort, Montani et al. further validated that a curated serum microRNA signature, named miR-Test, accurately identify lung cancer cases in high-risk individuals, with 74.9% sensitivity and 74.8% specificity [82]. The diagnostic value of microRNA expression in other body fluids for cancer detection has also been explored. Using Sputum samples, Xing et al. showed that a panel of miR-21, miR-31, and miR-210 is able to differentiate malignant and benign solitary pulmonary nodules, with both high sensitivity and specificity (>80%), in all three cohorts of samples [83]. Hanke et al. reported that the ratio of miR-126 to miR-152, and the ratio of miR-182 to miR-152 are indicators of presence of bladder cancer [84].

Circulating microRNAs are also gold mines for biomarkers indicative of prognosis and therapeutic response. A four-microRNA signature comprising miR-486, miR-30d, miR-1 and miR-499 in the serum independently predicts survival of non-small cell lung cancer, showing potential of microRNAs as noninvasive prognosis predictor [85]. In a recent study, high expression levels of miR-155 were significantly associated with therapeutic resistance of patients with diffuse large B-cell lymphoma to the rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone treatment [86]. These microRNA indicators for therapeutic response may help stratify patients for anticancer treatment to avoid unnecessary treatments.

Potential use of microRNA for cancer therapeutics

As cancer drivers, key promoters, or essential guardians, the therapeutic potential of microRNAs in cancer certainly attracts much attention [5]. microRNA therapeutic has the ability of targeting multiple protein-coding genes from the same pathways at different levels, thus probably preventing compensatory mechanisms such as mutation in the targeted oncogenes that leads to resistance. microRNAs are involved in all hallmarks of cancer, and targeting these microRNAs might offer therapeutic benefits. Identification of specific microRNAs associated with oncogenic IGF-1R signaling may also provide new tools for the molecular-designed treatments of cancer in clinical settings. The function of microRNAs with tumor suppressor activities can be restored with synthetic microRNA mimics, while the activities of oncogenic microRNAs can be blocked by oligonucleotides specifically interfering with the targeted microRNA [5]. Such strategies have been shown to be successful by many in vitro and in vivo studies. More impressively, MRX34, a liposome-based miR-34 mimetic developed by Mirna Therapeutics, has entered the phase of clinical trials for treating patients with liver cancer. The tumor suppressor miR-34a down regulates a number of key oncogenes, including MET, MEK1, CDK4/6, NOTCH, CD44, and immune evasion gene PDL1 [87]. The update on clinical trial with MRX34 American Society of Clinical Oncology (ASCO) Annual meeting 2016 reported that MRX34 has a manageable toxicity profile and evidence of activity in liver cancer, renal cell carcinoma, and melanoma during the phase I study (Hong et al. oral presentation at ASCO annual meeting 2016, abstract # 2508). In September 2016, Mirna Therapeutics voluntarily halted the phase I clinical trial because of multiple immune-related severe adverse events in patients treated with MRX34 (http://www.mirnarx.com/pipeline/mirna-pipeline.html). In another phase I trial in Australia, restoration of miR-16 led to a marked metabolic and radiological response on a patient with malignant pleural mesothelioma [88]. Whether it is an effective and safe therapy for patients with malignant pleural mesothelioma remains to be further evaluated.

Challenges and perspectives

Many challenges exist on the way of translating the preclinical findings into reliable cancer biomarkers and therapeutic agents. First, we still lack understanding on the basic mechanisms of how microRNAs determine their targets, besides their sequence complementarity, in a specific context. In addition, the frequent inconsistency between different reports on plasma microRNA biomarkers is a big concern on furthering such findings to clinical usages [89]. Sample collection time-point, extraction procedure, storage condition, variability in using internal controls, and different ways of data analysis are possible contributors to such discrepancies. microRNA therapeutics is even more challenging in achieving efficient and site-specific delivery of oligonucleotides, while avoiding side effects associated with oligonucleotides and delivery systems [5].

Despite these challenges, it is clear that the impact of microRNAs on cancer biology is tremendous. In two decades, microRNA concept has moved quickly from initial discovery to promising clinical candidates. With standardized, large-size, multiple-centered biomarker study design, it is possible to identify reliable cell-free cancer biomarkers in the near future. Following the initial launch of MRX34, other microRNA-based cancer therapeutics on the pipeline will soon enter the stage of clinical trials. The discovery of enrichment of microRNAs in extracellular vesicles such as exosomes provides further avenue for development of microRNA biomarker. Additionally, the feature of active packaging and release of exosome suggests that this might be an efficient microRNA delivery system for therapeutics [90]. We expect to see in the near future the use of microRNAs as cancer biomarkers, and the use of microRNA therapeutics, in combination with other therapeutic regimens, in cancer treatment.

  1. Author contributions: All the authors have accepted responsibility for the entire content of this submitted manuscript and approved submission.

  2. Research funding: Dr. Calin is The Alan M. Gewirtz Leukemia & Lymphoma Society Scholar. Work in Dr. Calin’s laboratory is supported in part by the NIH/NCI grants 1UH2TR00943-01 and 1 R01 CA182905-01, the UT MD Anderson Cancer Center SPORE in Melanoma grant from NCI (P50 CA093459), Aim at Melanoma Foundation and the Miriam and Jim Mulva research funds, the UT MD Anderson Cancer Center Brain SPORE (2P50CA127001), a Developmental Research award from Leukemia SPORE, a CLL Moonshot Flagship project, a 2015 Knowledge GAP MDACC grant, an Owens Foundation grant, and the Estate of C. G. Johnson, Jr.

  3. Employment or leadership: None declared.

  4. Honorarium: None declared.

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

References

1. Crick F. Central dogma of molecular biology. Nature 1970;227:561–3.10.1038/227561a0Search in Google Scholar

2. Morris KV, Mattick JS. The rise of regulatory RNA. Nat Rev Genet 2014;15:423–37.10.1038/nrg3722Search in Google Scholar

3. Ling H, Vincent K, Pichler M, Fodde R, Berindan-Neagoe I, Slack FJ, et al. Junk DNA and the long non-coding RNA twist in cancer genetics. Oncogene 2015;34:5003–11.10.1038/onc.2014.456Search in Google Scholar

4. Higgs PG, Lehman N. The RNA World: molecular cooperation at the origins of life. Nat Rev Genet 2015;16:7–17.10.1038/nrg3841Search in Google Scholar

5. Ling H, Fabbri M, Calin GA. MicroRNAs and other non-coding RNAs as targets for anticancer drug development. Nat Rev Drug Discov 2013;12:847–65.10.1038/nrd4140Search in Google Scholar

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

7. Wightman B, Ha I, Ruvkun G. Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C. elegans. Cell 1993;75:855–62.10.1016/0092-8674(93)90530-4Search in Google Scholar

8. Calin GA, Croce CM. MicroRNA signatures in human cancers. Nat Rev Cancer 2006;6:857–66.10.1038/nrc1997Search in Google Scholar PubMed

9. Mercer TR, Dinger ME, Mattick JS. Long non-coding RNAs: insights into functions. Nat Rev Genet 2009;10:155–9.10.1038/nrg2521Search in Google Scholar PubMed

10. Pasquinelli AE, Reinhart BJ, Slack F, Martindale MQ, Kuroda MI, Maller B, et al. Conservation of the sequence and temporal expression of let-7 heterochronic regulatory RNA. Nature 2000;408:86–9.10.1038/35040556Search in Google Scholar PubMed

11. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 2004;116:281–97.10.1016/S0092-8674(04)00045-5Search in Google Scholar

12. Ling H, Zhang W, Calin GA. Principles of microRNA involvement in human cancers. Chin J Cancer 2011;30:739–48.10.5732/cjc.011.10243Search in Google Scholar PubMed PubMed Central

13. Hwang HW, Wentzel EA, Mendell JT. A hexanucleotide element directs microRNA nuclear import. Science 2007;315:97–100.10.1126/science.1136235Search in Google Scholar PubMed

14. Orom UA, Nielsen FC, Lund AH. MicroRNA-10a binds the 5′UTR of ribosomal protein mRNAs and enhances their translation. Mol Cell 2008;30:460–71.10.1016/j.molcel.2008.05.001Search in Google Scholar PubMed

15. Place RF, Li LC, Pookot D, Noonan EJ, Dahiya R. MicroRNA-373 induces expression of genes with complementary promoter sequences. Proc Natl Acad Sci USA 2008;105:1608–13.10.1073/pnas.0707594105Search in Google Scholar PubMed PubMed Central

16. Vasudevan S, Tong Y, Steitz JA. Switching from repression to activation: microRNAs can up-regulate translation. Science 2007;318:1931–4.10.1126/science.1149460Search in Google Scholar PubMed

17. Fabbri M, Paone A, Calore F, Galli R, Gaudio E, Santhanam R, et al. MicroRNAs bind to Toll-like receptors to induce prometastatic inflammatory response. Proc Natl Acad Sci USA 2012;109:E2110–6.10.1073/pnas.1209414109Search in Google Scholar PubMed PubMed Central

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

19. Esquela-Kerscher A, Slack FJ. Oncomirs – microRNAs with a role in cancer. Nat Rev Cancer 2006;6:259–69.10.1038/nrc1840Search in Google Scholar PubMed

20. Mendell JT. miRiad roles for the miR-17-92 cluster in development and disease. Cell 2008;133:217–22.10.1016/j.cell.2008.04.001Search in Google Scholar PubMed PubMed Central

21. Sandhu SK, Fassan M, Volinia S, Lovat F, Balatti V, Pekarsky Y, et al. B-cell malignancies in microRNA Emu-miR-17~92 transgenic mice. Proc Natl Acad Sci USA 2013;110:18208–13.10.1073/pnas.1315365110Search in Google Scholar PubMed PubMed Central

22. Melnik BC. MiR-21: an environmental driver of malignant melanoma? J Transl Med 2015;13:202.10.1186/s12967-015-0570-5Search in Google Scholar PubMed PubMed Central

23. Costinean S, Zanesi N, Pekarsky Y, Tili E, Volinia S, Heerema N, et al. Pre-B cell proliferation and lymphoblastic leukemia/high-grade lymphoma in E(mu)-miR155 transgenic mice. Proc Natl Acad Sci USA 2006;103:7024–9.10.1073/pnas.0602266103Search in Google Scholar PubMed PubMed Central

24. Pencheva N, Tavazoie SF. Control of metastatic progression by microRNA regulatory networks. Nat Cell Biol 2013;15:546–54.10.1038/ncb2769Search in Google Scholar PubMed PubMed Central

25. Ling H, Pickard K, Ivan C, Isella C, Ikuo M, Mitter R, et al. The clinical and biological significance of MIR-224 expression in colorectal cancer metastasis. Gut 2016;65:977–89.10.1136/gutjnl-2015-309372Search in Google Scholar PubMed PubMed Central

26. Zhang Y, Yang P, Sun T, Li D, Xu X, Rui Y, et al. miR-126 and miR-126* repress recruitment of mesenchymal stem cells and inflammatory monocytes to inhibit breast cancer metastasis. Nat Cell Biol 2013;15:284–94.10.1038/ncb2690Search in Google Scholar PubMed PubMed Central

27. Park SM, Gaur AB, Lengyel E, Peter ME. The miR-200 family determines the epithelial phenotype of cancer cells by targeting the E-cadherin repressors ZEB1 and ZEB2. Genes Dev 2008;22:894–907.10.1101/gad.1640608Search in Google Scholar PubMed PubMed Central

28. Pichler M, Ress AL, Winter E, Stiegelbauer V, Karbiener M, Schwarzenbacher D, et al. MiR-200a regulates epithelial to mesenchymal transition-related gene expression and determines prognosis in colorectal cancer patients. Br J Cancer 2014;110:1614–21.10.1038/bjc.2014.51Search in Google Scholar PubMed PubMed Central

29. Korpal M, Ell BJ, Buffa FM, Ibrahim T, Blanco MA, Celia-Terrassa T, et al. Direct targeting of Sec23a by miR-200s influences cancer cell secretome and promotes metastatic colonization. Nat Med 2011;17:1101–8.10.1038/nm.2401Search in Google Scholar PubMed PubMed Central

30. Felli N, Fontana L, Pelosi E, Botta R, Bonci D, Facchiano F, et al. MicroRNAs 221 and 222 inhibit normal erythropoiesis and erythroleukemic cell growth via kit receptor down-modulation. Proc Natl Acad Sci USA 2005;102:18081–6.10.1073/pnas.0506216102Search in Google Scholar PubMed PubMed Central

31. Pineau P, Volinia S, McJunkin K, Marchio A, Battiston C, Terris B, et al. miR-221 overexpression contributes to liver tumorigenesis. Proc Natl Acad Sci USA 2010;107:264–9.10.1073/pnas.0907904107Search in Google Scholar PubMed PubMed Central

32. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011;144:646–74.10.1016/j.cell.2011.02.013Search in Google Scholar PubMed

33. Meng F, Henson R, Wehbe-Janek H, Ghoshal K, Jacob ST, Patel T. MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer. Gastroenterology 2007;133:647–58.10.1053/j.gastro.2007.05.022Search in Google Scholar PubMed PubMed Central

34. Johnson CD, Esquela-Kerscher A, Stefani G, Byrom M, Kelnar K, Ovcharenko D, et al. The let-7 microRNA represses cell proliferation pathways in human cells. Cancer Res 2007;67:7713–22.10.1158/0008-5472.CAN-07-1083Search in Google Scholar PubMed

35. le Sage C, Nagel R, Egan DA, Schrier M, Mesman E, Mangiola A, et al. Regulation of the p27(Kip1) tumor suppressor by miR-221 and miR-222 promotes cancer cell proliferation. EMBO J 2007;26:3699–708.10.1038/sj.emboj.7601790Search in Google Scholar PubMed PubMed Central

36. Yamakuchi M, Ferlito M, Lowenstein CJ. miR-34a repression of SIRT1 regulates apoptosis. Proc Natl Acad Sci USA 2008;105:13421–6.10.1073/pnas.0801613105Search in Google Scholar PubMed PubMed Central

37. Cimmino A, Calin GA, Fabbri M, Iorio MV, Ferracin M, Shimizu M, et al. miR-15 and miR-16 induce apoptosis by targeting BCL2. Proc Natl Acad Sci USA 2005;102:13944–9.10.1073/pnas.0506654102Search in Google Scholar PubMed PubMed Central

38. Martinez I, Cazalla D, Almstead LL, Steitz JA, DiMaio D. miR-29 and miR-30 regulate B-Myb expression during cellular senescence. Proc Natl Acad Sci USA 2011;108:522–7.10.1073/pnas.1017346108Search in Google Scholar PubMed PubMed Central

39. Ohira T, Naohiro S, Nakayama Y, Osaki M, Okada F, Oshimura M, et al. miR-19b regulates hTERT mRNA expression through targeting PITX1 mRNA in melanoma cells. Sci Rep 2015;5:8201.10.1038/srep08201Search in Google Scholar PubMed PubMed Central

40. Lou YL, Guo F, Liu F, Gao FL, Zhang PQ, Niu X, et al. miR-210 activates notch signaling pathway in angiogenesis induced by cerebral ischemia. Mol Cell Biochem 2012;370:45–51.10.1007/s11010-012-1396-6Search in Google Scholar PubMed

41. Kuhnert F, Kuo CJ. miR-17-92 angiogenesis micromanagement. Blood 2010;115:4631–3.10.1182/blood-2010-03-276428Search in Google Scholar PubMed

42. Ma L, Teruya-Feldstein J, Weinberg RA. Tumour invasion and metastasis initiated by microRNA-10b in breast cancer. Nature 2007;449:682–8.10.1038/nature06174Search in Google Scholar PubMed

43. Gao P, Tchernyshyov I, Chang TC, Lee YS, Kita K, Ochi T, et al. c-Myc suppression of miR-23a/b enhances mitochondrial glutaminase expression and glutamine metabolism. Nature 2009;458:762–5.10.1038/nature07823Search in Google Scholar PubMed PubMed Central

44. Rottiers V, Naar AM. MicroRNAs in metabolism and metabolic disorders. Nat Rev Mol Cell Biol 2012;13:239–50.10.1038/nrm3313Search in Google Scholar PubMed PubMed Central

45. Cortez MA, Ivan C, Valdecanas D, Wang X, Peltier HJ, Ye Y, et al. PDL1 Regulation by p53 via miR-34. J Natl Cancer Inst 2016:108. DOI: 10.1093/jnci/djv303.Search in Google Scholar PubMed PubMed Central

46. Wei J, Wang F, Kong LY, Xu S, Doucette T, Ferguson SD, et al. miR-124 inhibits STAT3 signaling to enhance T cell-mediated immune clearance of glioma. Cancer Res 2013;73:3913–26.10.1158/0008-5472.CAN-12-4318Search in Google Scholar PubMed PubMed Central

47. Calin GA, Sevignani C, Dumitru CD, Hyslop T, Noch E, Yendamuri S, et al. Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers. Proc Natl Acad Sci USA 2004;101:2999–3004.10.1073/pnas.0307323101Search in Google Scholar PubMed PubMed Central

48. Lujambio A, Ropero S, Ballestar E, Fraga MF, Cerrato C, Setien F, et al. Genetic unmasking of an epigenetically silenced microRNA in human cancer cells. Cancer Res 2007;67:1424–9.10.1158/0008-5472.CAN-06-4218Search in Google Scholar PubMed

49. Lujambio A, Calin GA, Villanueva A, Ropero S, Sanchez-Cespedes M, Blanco D, et al. A microRNA DNA methylation signature for human cancer metastasis. Proc Natl Acad Sci USA 2008;105:13556–61.10.1073/pnas.0803055105Search in Google Scholar PubMed PubMed Central

50. Davalos V, Moutinho C, Villanueva A, Boque R, Silva P, Carneiro F, et al. Dynamic epigenetic regulation of the microRNA-200 family mediates epithelial and mesenchymal transitions in human tumorigenesis. Oncogene 2012;31:2062–74.10.1038/onc.2011.383Search in Google Scholar PubMed PubMed Central

51. Tellez CS, Juri DE, Do K, Bernauer AM, Thomas CL, Damiani LA, et al. EMT and stem cell-like properties associated with miR-205 and miR-200 epigenetic silencing are early manifestations during carcinogen-induced transformation of human lung epithelial cells. Cancer Res 2011;71:3087–97.10.1158/0008-5472.CAN-10-3035Search in Google Scholar PubMed PubMed Central

52. Ohtsuka M, Ling H, Doki Y, Mori M, Calin GA. MicroRNA Processing and Human Cancer. J Clin Med 2015;4:1651–67.10.3390/jcm4081651Search in Google Scholar PubMed PubMed Central

53. Pichiorri F, Palmieri D, De Luca L, Consiglio J, You J, Rocci A, et al. In vivo NCL targeting affects breast cancer aggressiveness through miRNA regulation. J Exp Med 2013;210:951–68.10.1084/jem.20120950Search in Google Scholar PubMed PubMed Central

54. Salmena L, Poliseno L, Tay Y, Kats L, Pandolfi PP. A ceRNA hypothesis: the Rosetta Stone of a hidden RNA language? Cell 2011;146:353–8.10.1016/j.cell.2011.07.014Search in Google Scholar PubMed PubMed Central

55. Thomson DW, Dinger ME. Endogenous microRNA sponges: evidence and controversy. Nat Rev Genet 2016;17:272–83.10.1038/nrg.2016.20Search in Google Scholar PubMed

56. Girnita L, Worrall C, Takahashi S, Seregard S, Girnita A. Something old, something new and something borrowed: emerging paradigm of insulin-like growth factor type 1 receptor (IGF-1R) signaling regulation. Cell Mol Life Sci 2014;71:2403–27.10.1007/s00018-013-1514-ySearch in Google Scholar PubMed PubMed Central

57. Economou MA, Wu J, Vasilcanu D, Rosengren L, All-Ericsson C, van der Ploeg I, et al. Inhibition of VEGF secretion and experimental choroidal neovascularization by picropodophyllin (PPP), an inhibitor of the insulin-like growth factor-1 receptor. Invest Ophthalmol Vis Sci 2008;49:2620–6.10.1167/iovs.07-0742Search in Google Scholar PubMed

58. Girnita A, Zheng H, Gronberg A, Girnita L, Stahle M. Identification of the cathelicidin peptide LL-37 as agonist for the type I insulin-like growth factor receptor. Oncogene 2012;31:352–65.10.1038/onc.2011.239Search in Google Scholar PubMed PubMed Central

59. Donzelli S, Cioce M, Muti P, Strano S, Yarden Y, Blandino G. MicroRNAs: non-coding fine tuners of receptor tyrosine kinase signalling in cancer. Semin Cell Dev Biol 2016;50:133–42.10.1016/j.semcdb.2015.12.020Search in Google Scholar PubMed

60. Kedmi M, Sas-Chen A, Yarden Y. MicroRNAs and Growth Factors: an Alliance Propelling Tumor Progression. J Clin Med 2015;4:1578–99.10.3390/jcm4081578Search in Google Scholar PubMed PubMed Central

61. Crudden C, Ilic M, Suleymanova N, Worrall C, Girnita A, Girnita L. The dichotomy of the Insulin-like growth factor 1 receptor: RTK and GPCR: friend or foe for cancer treatment? Growth Horm IGF Res 2015;25:2–12.10.1016/j.ghir.2014.10.002Search in Google Scholar PubMed

62. Zheng H, Shen H, Oprea I, Worrall C, Stefanescu R, Girnita A, et al. beta-Arrestin-biased agonism as the central mechanism of action for insulin-like growth factor 1 receptor-targeting antibodies in Ewing’s sarcoma. Proc Natl Acad Sci USA 2012;109:20620–5.10.1073/pnas.1216348110Search in Google Scholar PubMed PubMed Central

63. Vasilcanu R, Vasilcanu D, Sehat B, Yin S, Girnita A, Axelson M, et al. Insulin-like growth factor type-I receptor-dependent phosphorylation of extracellular signal-regulated kinase 1/2 but not Akt (protein kinase B) can be induced by picropodophyllin. Mol Pharmacol 2008;73:930–9.10.1124/mol.107.040014Search in Google Scholar PubMed

64. Kedmi M, Ben-Chetrit N, Korner C, Mancini M, Ben-Moshe NB, Lauriola M, et al. EGF induces microRNAs that target suppressors of cell migration: miR-15b targets MTSS1 in breast cancer. Sci Signal 2015;8:ra29.10.1126/scisignal.2005866Search in Google Scholar PubMed

65. Katz M, Amit I, Citri A, Shay T, Carvalho S, Lavi S, et al. A reciprocal tensin-3-cten switch mediates EGF-driven mammary cell migration. Nat Cell Biol 2007;9:961–9.10.1038/ncb1622Search in Google Scholar PubMed

66. Shi B, Prisco M, Calin G, Liu CG, Russo G, Giordano A, et al. Expression profiles of micro RNA in proliferating and differentiating 32D murine myeloid cells. J Cell Physiol 2006;207:706–10.10.1002/jcp.20613Search in Google Scholar PubMed

67. Martin EC, Bratton MR, Zhu Y, Rhodes LV, Tilghman SL, Collins-Burow BM, et al. Insulin-like growth factor-1 signaling regulates miRNA expression in MCF-7 breast cancer cell line. PLoS One 2012;7:e49067.10.1371/journal.pone.0049067Search in Google Scholar PubMed PubMed Central

68. Jung HJ, Suh Y. Regulation of IGF -1 signaling by microRNAs. Front Genet 2014;5:472.10.3389/fgene.2014.00472Search in Google Scholar PubMed PubMed Central

69. Peng Y, Dai Y, Hitchcock C, Yang X, Kassis ES, Liu L, et al. Insulin growth factor signaling is regulated by microRNA-486, an underexpressed microRNA in lung cancer. Proc Natl Acad Sci USA 2013;110:15043–8.10.1073/pnas.1307107110Search in Google Scholar PubMed PubMed Central

70. Yoshihara H, Fukushima T, Hakuno F, Saeki Y, Tanaka K, Ito A, et al. Insulin/insulin-like growth factor (IGF) stimulation abrogates an association between a deubiquitinating enzyme USP7 and insulin receptor substrates (IRSs) followed by proteasomal degradation of IRSs. Biochem Biophys Res Commun 2012;423:122–7.10.1016/j.bbrc.2012.05.093Search in Google Scholar PubMed

71. Girnita L, Shenoy SK, Sehat B, Vasilcanu R, Girnita A, Lefkowitz RJ, et al. {beta}-Arrestin is crucial for ubiquitination and down-regulation of the insulin-like growth factor-1 receptor by acting as adaptor for the MDM2 E3 ligase. J Biol Chem 2005;280:24412–9.10.1074/jbc.M501129200Search in Google Scholar

72. Girnita L, Shenoy SK, Sehat B, Vasilcanu R, Vasilcanu D, Girnita A, et al. Beta-arrestin and Mdm2 mediate IGF-1 receptor-stimulated ERK activation and cell cycle progression. J Biol Chem 2007;282:11329–38.10.1074/jbc.M611526200Search in Google Scholar

73. Sehat B, Andersson S, Girnita L, Larsson O. Identification of c-Cbl as a new ligase for insulin-like growth factor-I receptor with distinct roles from Mdm2 in receptor ubiquitination and endocytosis. Cancer Res 2008;68:5669–77.10.1158/0008-5472.CAN-07-6364Search in Google Scholar

74. Girnita L, Takahashi SI, Crudden C, Fukushima T, Worrall C, Furuta H, et al. When phosphorylation encounters ubiquitination: a balanced perspective on IGF-1R signaling. Prog Mol Biol Transl Sci 2016;141:277–311.10.1016/bs.pmbts.2016.04.001Search in Google Scholar

75. Cortez MA, Bueso-Ramos C, Ferdin J, Lopez-Berestein G, Sood AK, Calin GA. MicroRNAs in body fluids–the mix of hormones and biomarkers. Nat Rev Clin Oncol 2011;8:467–77.10.1038/nrclinonc.2011.76Search in Google Scholar

76. Hammar SP. Metastatic adenocarcinoma of unknown primary origin. Hum Pathol 1998;29:1393–402.10.1016/S0046-8177(98)90007-7Search in Google Scholar

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

78. Rosenfeld N, Aharonov R, Meiri E, Rosenwald S, Spector Y, Zepeniuk M, et al. MicroRNAs accurately identify cancer tissue origin. Nat Biotechnol 2008;26:462–9.10.1038/nbt1392Search in Google Scholar PubMed

79. Lebanony D, Benjamin H, Gilad S, Ezagouri M, Dov A, Ashkenazi K, et al. Diagnostic assay based on hsa-miR-205 expression distinguishes squamous from nonsquamous non-small-cell lung carcinoma. J Clin Oncol 2009;27:2030–7.10.1200/JCO.2008.19.4134Search in Google Scholar PubMed

80. Gilad S, Lithwick-Yanai G, Barshack I, Benjamin S, Krivitsky I, Edmonston TB, et al. Classification of the four main types of lung cancer using a microRNA-based diagnostic assay. J Mol Diagn 2012;14:510–7.10.1016/j.jmoldx.2012.03.004Search in Google Scholar PubMed

81. Boeri M, Verri C, Conte D, Roz L, Modena P, Facchinetti F, et al. MicroRNA signatures in tissues and plasma predict development and prognosis of computed tomography detected lung cancer. Proc Natl Acad Sci USA 2011;108:3713–8.10.1073/pnas.1100048108Search in Google Scholar PubMed PubMed Central

82. Montani F, Marzi MJ, Dezi F, Dama E, Carletti RM, Bonizzi G, et al. miR-Test: a blood test for lung cancer early detection. J Natl Cancer Inst 2015;107:djv063.10.1093/jnci/djv063Search in Google Scholar PubMed

83. Xing L, Su J, Guarnera MA, Zhang H, Cai L, Zhou R, et al. Sputum microRNA biomarkers for identifying lung cancer in indeterminate solitary pulmonary nodules. Clin Cancer Res 2015;21:484–9.10.1158/1078-0432.CCR-14-1873Search in Google Scholar PubMed PubMed Central

84. Hanke M, Hoefig K, Merz H, Feller AC, Kausch I, Jocham D, et al. A robust methodology to study urine microRNA as tumor marker: microRNA-126 and microRNA-182 are related to urinary bladder cancer. Urol Oncol 2010;28:655–61.10.1016/j.urolonc.2009.01.027Search in Google Scholar PubMed

85. Hu Z, Chen X, Zhao Y, Tian T, Jin G, Shu Y, et al. Serum microRNA signatures identified in a genome-wide serum microRNA expression profiling predict survival of non-small-cell lung cancer. J Clin Oncol 2010;28:1721–6.10.1200/JCO.2009.24.9342Search in Google Scholar PubMed

86. Iqbal J, Shen Y, Huang X, Liu Y, Wake L, Liu C, et al. Global microRNA expression profiling uncovers molecular markers for classification and prognosis in aggressive B-cell lymphoma. Blood 2015;125:1137–45.10.1182/blood-2014-04-566778Search in Google Scholar PubMed PubMed Central

87. Agostini M, Knight RA. miR-34: from bench to bedside. Oncotarget 2014;5:872–81.10.18632/oncotarget.1825Search in Google Scholar PubMed PubMed Central

88. Kao SC, Fulham M, Wong K, Cooper W, Brahmbhatt H, MacDiarmid J, et al. A Significant Metabolic and Radiological Response after a Novel Targeted MicroRNA-based Treatment Approach in Malignant Pleural Mesothelioma. Am J Respir Crit Care Med 2015;191:1467–9.10.1164/rccm.201503-0461LESearch in Google Scholar PubMed

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

90. Chen X, Liang H, Zhang J, Zen K, Zhang CY. Secreted microRNAs: a new form of intercellular communication. Trends Cell Biol 2012;22:125–32.10.1016/j.tcb.2011.12.001Search in Google Scholar PubMed

Received: 2016-8-19
Accepted: 2016-11-10
Published Online: 2016-12-19
Published in Print: 2017-5-1

©2017 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 19.4.2024 from https://www.degruyter.com/document/doi/10.1515/cclm-2016-0740/html
Scroll to top button