Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Nov 14.
Published in final edited form as: Cell. 2019 Nov 14;179(5):1033–1055. doi: 10.1016/j.cell.2019.10.017

The Role of Non-coding RNAs in Oncology

Frank J Slack 1,2,3,*, Arul M Chinnaiyan 4,5,6,7,8,*
PMCID: PMC7347159  NIHMSID: NIHMS1542053  PMID: 31730848

Abstract

For decades, research into cancer biology focused on the involvement of protein-coding genes. Only recently was it discovered that an entire class of molecules, termed non-coding RNA (ncRNA), plays key regulatory roles in shaping cellular activity. An explosion of studies into ncRNA biology has since shown that they represent a diverse and prevalent group of RNAs including both oncogenic molecules and those that work in a tumor suppressive manner. As a result, hundreds of cancer-focused clinical trials involving ncRNAs as novel biomarkers or therapies have begun, and these are likely just the beginning.

eTOC – Chinnaiyan & Slack

Slack and Chinnaiyan explore the diverse and context-dependent roles of ncRNAs, including circRNAs, lncRNAs, miRNAs, piRNAs, and tsRNAs, in cancer. They provide insight into the prospect of therapeutic targeting and use of ncRNAs as biomarkers with an up-to-date summary of clinical and preclinical studies.


Although now one of the hottest topics in biomedical science, the importance of non-coding RNA (ncRNA) was largely unrecognized until recently. RNA was once thought to mostly serve as a messenger that carried instructions encoded in DNA so that other molecules, like the ribosome, could use the code to make proteins. However, in the last 30 years, researchers have discovered that multiple types of RNA exist, and that among the most important is non-coding RNA- the type that is not involved in producing proteins. The discovery of tens of thousands of ncRNA species has revolutionized the field, altering the way that researchers think about physiology and the development of disease (Adams et al., 2017; Bartel, 2018; Evans et al., 2016; Rupaimoole and Slack, 2017). Non-coding RNAs constitute more than 90% of the RNAs made from the human genome, but most of the >50,000 known ncRNAs have been discovered only in the past 10 years and remain largely unstudied (Deveson et al., 2017; Esposito et al., 2019; Kopp and Mendell, 2018; Ransohoff et al., 2018).

Still, there are many ncRNAs that have since been shown to play key roles in both normal cellular function and disease, including cancer, and this information is being actively translated into the clinic. Some small ncRNAs are so stable that they survive in the bloodstream and could be the basis for accurate and sensitive screens for major human cancers in a few drops of blood (Agaoglu et al., 2011; Imaoka et al., 2016; Toiyama et al., 2013). Additionally, ncRNAs can be therapeutically targeted, and the delivery of ncRNAs can be based on an existing foundation of what has been learned regarding delivery of RNAi and oligonucleotides targeting protein-coding mRNAs (Levin, 2019; Pecot et al., 2011; Wu et al., 2014). In fact, the field of RNA medicine has seen a renaissance (Levin, 2019) with the recent approval of the first RNAi drug Onpattro™ (patisiran; reduces levels of TTR for treatment of the neurodegenerative disease hereditary transthyretin amyloidosis) (Adams et al., 2018) and the clinical success of the RNA-targeting oligonucleotide drug Spinraza® (nusinersen; increases levels of full-length SMN2 for treatment of the neuromuscular disease spinal muscular atrophy) (Wurster et al., 2019). In addition, clinical trials with drugs based on a class of ncRNAs called microRNA (miRNA), either therapies that increase or decrease the target miRNA, have begun for cancer (Beg et al., 2017; Seto et al., 2018; van Zandwijk et al., 2017).

In this review, we will discuss ncRNAs in relation to cancer cell biology and their relevance to current clinical practice. We first examine the intricacies of the different classes of ncRNAs (microRNAs (miRNAs), transfer RNA (tRNA)-derived small RNAs (tsRNAs), PIWI-interacting RNAs (piRNAs), long ncRNAs (lncRNAs), pseudogenes, and circular RNAs (circRNAs), Supplemental Table 1) and provide fundamental examples of the far-reaching roles that these molecules have in affecting cancer processes. We then discuss how these basic science insights in ncRNA biology are being used to develop next-generation diagnostics and therapies in cancer. As the so-called “dark matter” of the genome continues to be brought into the light, it is evident that targeting ncRNA signaling has great potential to impact cancer patient care.

Overview of classes of ncRNAs and their association with cancer

For decades, the miniscule protein-coding portion of the genome was the primary focus of medical research. The sequencing of the human genome showed that only about 2% of our genes ultimately code for proteins, and many in the scientific community believed that the remaining 98% was simply non-functonal “junk” (Mattick and Makunin, 2006; Slack, 2006). However, the ENCODE project revealed that the non-protein coding portion of the genome is copied into thousands of RNA molecules (Djebali et al., 2012; Gerstein et al., 2012) that not only regulate fundamental biological processes such as growth, development, and organ function, but also appear to play a critical role in the whole spectrum of human disease, notably cancer (for recent reviews, see (Adams et al., 2017; Deveson et al., 2017; Rupaimoole and Slack, 2017)). Trailblazing research led to our understanding of how ncRNA molecules perform multiple vital roles in the coding, decoding, regulation, and expression of genes as well as how they communicate with each other (Anastasiadou et al., 2018b; Esquela-Kerscher and Slack, 2006; Gregory and Shiekhattar, 2005; Huarte and Rinn, 2010; Kasinski and Slack, 2011; Krichevsky et al., 2003; Rinn and Huarte, 2011; Tay et al., 2014). This knowledge of ncRNA regulatory roles revealed specific ncRNA networks based on complementary base pairing at work in different cancer types (Anastasiadou et al., 2018b), which in turn, opened up possibilities for scientists in this field to develop specific cancer therapeutic and preventive strategies focused on the ncRNAs made from the human genome (for recent reviews, see (Cieslik and Chinnaiyan, 2018; Rupaimoole and Slack, 2017)).

Cancer is characterized by cells that grow (proliferate) out of control, are able to spread to other tissues (metastasize), and lose the ability to die through the orderly process of cell death (apoptosis). The discovery of ncRNA has added a new dimension to the understanding of how cancer develops, and how it may be treated, by providing a window into the impact of the rest of the genome. Deregulated ncRNA expression, and subsequent downstream signaling processes that we detail in later sections, have been directly implicated in cancer development and progression. Genetic alterations in genes encoding ncRNAs have been found to be associated with cancer; however, compared to protein-coding genes, the list of genetic examples from studies thus far is considerably shorter. The most notable example is likely deletion of 13q14.3 in chronic lymphocytic leukemia (CLL) that deletes the miR-15/16 tumor suppressors (Calin et al., 2002). Conversely, amplification of chromosomal regions encoding oncogenic ncRNAs are also found in cancer, including amplification of lncRNAs FAL1 (Hu et al., 2014) and PVT1 (Tseng et al., 2014). Single nucleotide polymorphisms (SNPs) in the genes of lncRNAs H19 (Hua et al., 2016), ANRIL (Pasmant et al., 2011), and CCAT2 (Ling et al., 2013) have also been associated with varying risks of cancer development. In addition to genetic alterations within transcribed regions, mutations in the promoters of ncRNAs can lead to altered gene expression levels, such as recurrent driver mutations in the promoters of lncRNAs NEAT1 and RMRP in breast cancer (Rheinbay et al., 2017). Besides aberrations in sequences encoding the ncRNA itself, mutations or dysregulation of enzymes involved in the biogenesis of ncRNAs are implicated in cancer, such as Drosha and Dicer involved in miRNA processing (Rupaimoole and Slack, 2017). In addition to these genetic mechanisms, up- or downregulation of ncRNA expression associated with cancer can occur through epigenetic, transcriptional, or post-transcriptional processes (see recent reviews (Adams et al., 2014; Anastasiadou et al., 2018a; Rupaimoole and Slack, 2017)).

Non-coding RNAs can be divided into different classes, broadly based upon their size. The small ncRNAs important in cancer include miRNAs, tsRNAs, and piRNAs. At the opposite end of the size spectrum are the lncRNAs, which are characterized as untranslated RNAs greater than 200 nucleotides in length, and include subclasses such as pseudogenes and circRNAs.

MicroRNAs

Near the turn of the millennium, the first miRNAs, lin-4 and let-7, were identified through developmental studies in C. elegans (Lee et al., 1993; Reinhart et al., 2000). MicroRNAs are short ncRNAs of approximately 22 nucleotides in length that regulate the expression of other RNAs, notably mRNAs through binding between the 5’ end (known as the “seed”) of the miRNA with complementary sequences in target RNAs. Genes encoding miRNAs are transcribed by RNA Pol II and processed through an evolutionarily conserved pathway. In the canonical processing pathway, this longer primary transcript, called the pri-miRNA, forms a characteristic hairpin structure that is recognized by the Microprocessor complex (consisting of Drosha and DGCR8), cleaved into a pre-miRNA about 60 nucleotides in length, and exported to the cytoplasm via an Exportin 5 and Ran-GTP complex. The ends of the pre-miRNA are then cleaved by Dicer to form a miRNA duplex, which consists of 5’ phosphates and 2 nucleotide overhangs on each 3’ end. One strand of the miRNA duplex, the guide strand, is loaded onto an Argonaute protein and selected to form the RNA-induced silencing complex (RISC) containing the mature 22 nucleotide miRNA (see recent reviews by (Anastasiadou et al., 2018a; Bartel, 2018)). The mature miRNA functions by binding to the 3’ untranslated regions (3’UTR) of mRNAs and inhibiting their use by either degradation or translational repression (Bartel, 2018; Esquela-Kerscher and Slack, 2006). Multiple studies have attempted to annotate the number of miRNAs in different species. For humans, higher estimates from miRBase v22 have placed the number of mature miRNAs at 2654, but algorithms from other databases, such as MirGeneDB2.0, decrease this number to 588 high confidence miRNAs (Fromm et al., 2015; Kozomara et al., 2018). Despite discrepancies in their absolute numbers, it is evident that miRNAs have far-reaching effects on downstream processes, as more than 60% of coding genes are potential targets of miRNAs (Friedman et al., 2009). In addition, hundreds of miRNAs are conserved at their seed region across phylogeny (Bartel, 2018), suggesting key roles in developmental or physiological processes in animals.

Regarding cancer, miRNAs provide a powerful new avenue for the discovery of novel genetic risk factors (Ryan et al., 2010). Among the small ncRNA species, miRNAs are by far the most extensively studied in cancer compared to tsRNAs and piRNAs. MicroRNAs have been found to be altered in all cancer types studied (Volinia et al., 2006), and alterations in miRNAs have been demonstrated to play a crucial role in affecting molecular and cellular processes of the cancer state (Esquela-Kerscher and Slack, 2006; Nicoloso et al., 2009). Although researchers are still learning the extent of the contribution of miRNAs to cancer, these small ncRNAs seem to function in one of two ways—as tumor suppressors or oncogenes (commonly referred to as oncomiRs) that promote cancer growth or metastasis (Rupaimoole and Slack, 2017). Although small, miRNAs are powerful, with each molecule often able to regulate more than one target, and, vice versa, mRNAs are frequently targeted by several miRNAs (Bartel, 2018). As such, miRNAs function as master regulators that control the expression of thousands of coding and non-coding genes, including most of the insidious oncogenes, such as RAS, MYC, and EGFR, and the critical tumor suppressors, including TP53, PTEN, and BRCA1. Table 1 provides a list of selected miRNAs (and examples from the other classes of ncRNAs) that have in vivo experimental evidence to either support an oncogenic, tumor suppressive, or context-dependent role. Many of these studies involved creation of transgenic mouse models and/or delivery of miRNA mimetics or antimiRs, synthetic oligoribonucleotides that either replenish or decrease levels of the target miRNA. Table 1 further provides instances of the cancer-related molecular mechanisms that each miRNA has been shown to regulate. Here, we highlight a few examples from each category, with Figure 1 and Figure 2 also providing visual representations of one oncogenic and one tumor suppressive pathway, respectively.

Table 1.

Oncogenic or tumor suppressive non-coding RNAs with in vivo experimental evidence

Name ncRNA Cancer types examined In vivo experimental techniques used Cancer-related mechanisms and/or functions of ncRNA References
Oncogenic ncRNAs

miR-10b miRNA breast, glioblastoma antimiRs, CRISPR-Cas9 knockdown in mouse xenografts and allografts, transgenic knockout mouse models targets several transcripts that encode regulators of cell cycle progression, migration, invasion, and metastasis Ma et al., 2010, Kim et al., 2016, El Fatimy et al., 2017
miR-21 miRNA lung, B cell lymphoma transgenic knockout, overexpression mouse models targets transcripts that encode negative regulators of RAS signaling, leading to increased proliferation and decreased apoptosis Hatley et al., 2010, Medina et al., 2010
miR-31 miRNA lung, breast overexpression in mouse xenografts, transgenic knockout, overexpression mouse models targets transcripts that encode regulators of RAS, WNT, and TGFβ signaling to increase proliferation, stem cell renewal, and metastasis Edmonds et al., 2016, Lv et al., 2017
miR-155 miRNA lymphoma transgenic overexpression mouse model, treatment with antimiRs targets SHIP1 transcript, a negative regulator of AKT, to increase proliferation and survival O’Connell et al., 2009, Babar et al., 2012, Cheng et al., 2015
miR-221 miRNA liver overexpression, treatment with antimiRs in mouse xenografts targets transcripts of tumor suppressors and cell cycle inhibitors (e.g., p27, PTEN) to increase proliferation and decrease apoptosis Pineau et al., 2010, Park et al., 2011
LeuCAG3’tsRNA tsRNA liver LNA knockdown in PDX enhances translation of transcripts encoding ribosomal proteins, leading to increased ribosome biogenesis and proliferation Kim et al., 2017
ARLNC1 lncRNA prostate shRNA or ASO knockdown in mouse xenografts interacts with the mRNA encoding AR, a nuclear receptor, to promote oncogenic AR signaling, proliferation, and suraval Zhang et al., 2018
CamK-A lncRNA breast shRNA, siRNA knockdown in mouse xenografts/PDX interacts with and controls activity of kinases that modulate calcium-induced NF-KB signaling, leading to remodeling of the tumor microenvironment Sang et al., 2018
CCAT1 lncRNA colorectal, esophageal shRNA, siRNA knockdown in mouse xenografts interacts with transcription factors (e.g., SOX2, p63) to activate expression of genes involved in increasing proliferation and decreasing apoptosis Kim et al., 2014, Jiang et al., 2018
CTBP1-AS lncRNA prostate siRNA knockdown, overexpression in mouse xenografts recruits chromatin modifying, splicing factors to promoter of a nuclear receptor corepressor (CTBP) to decrease its expression, leading to increased oncogenic AR activity Takayama et al., 2013
DSCAM-AS1 lncRNA breast shRNA knockdown in mouse xenografts interacts with proteins of the hnRNP complex involved in RNA processing and mediates proliferation, invasion, and metastasis Niknafs et al., 2016
EPIC1 lncRNA breast shRNA knockdown in mouse xenografts interacts with MYC transcription factor and increases its activation of target genes, leading to enhanced cell cycle progression Wang et al., 2018
FAL1 lncRNA ovarian breast shRNA, siRNA knockdown in mouse xenografts stabilizes components of PRC1 chromatin modifying complex to mediate expression of genes involved in proliferation and suraval Hu et al., 2014
HOTAIR lncRNA breast siRNA knockdown, overexpression in mouse xenografts recruits PRC2, LSD1/CoREST/REST chromatin modifying complexes, scaffolds transcription factors at target promoters of genes involved in invasion, metastasis, and proliferation Gupta et al., 2010, Li et al., 2016b
LINK-A lncRNA breast shRNA knockdown in mouse xenografts, transgenic overexpression in mammary gland, LNA knockdown interacts with kinases that control HIF1α activity, glycolysis, enhances degradation of tumor suppressors (RB, p53) and antigen peptide-loading complexes to promote immune evasion Lin et al., 2016, Hu et al., 2019
lncARSR lncRNA RCC shRNA knockdown, overexpression in mouse xenografts interacts with transcriptional coactivator YAP and acts as a ceRNA for miRNAs that target RTK transcripts, leading to enhanced suraval and propagation of tumor-initiating cells Qu et al., 2016a, Qu et al., 2016b
PCAT-1 lncRNA prostate overexpression in mouse xenografts represses expression of BRCA2 tumor suppressor to impact DNA damage repair Prensner et al., 2014a
PVT1 lncRNA colorectal, gastric CRISPR-Cas9 or shRNA knockdown, overexpression in mouse xenografts activates oncogenic signaling (MYC, STAT3) and represses expression of tumor suppressors (p15, p16), resulting in increased proliferation, angiogenesis and decreased apoptosis Tseng et al., 2014, Kong et al., 2015, Zhao et al., 2018
SAMMSON lncRNA melanoma GapmeR knockdown in PDX interacts with and controls subcellular localization of proteins that regulate mitochondrial homeostasis and metabolism Leucci et al., 2016
SChLAP1 lncRNA prostate shRNA knockdown in mouse xenografts interacts with and antagonizes activity of the SWI/SNF chromatin modifying complex to promote invasion and metastasis Prensner et al., 2013
THOR lncRNA lung, melanoma CRISPR-Cas9 knockdown, overexpression in mouse xenografts, transgenic knockout, overexpression in zebrafish binds IGF2BP1 to stabilize interactions with oncogenic target mRNAs, in turn stabilizing those transcripts and promoting proliferation Hosono et al., 2017
BRAFP1 pseudogene B cell lymphoma transgenic overexpression mouse model acts as a ceRNA for miRNAs that target the BRAF transcript, leading to increased BRAF expression, MAPK signaling, and proliferation Karreth et al., 2015
circCCDC66 circRNA colorectal siRNA knockdown in mouse xenografts sponges several miRNAs that target oncogenic transcripts (e.g., MYC), promoting proliferation, migration, and invasion Hsiao et al., 2017
circCTNNB1 circRNA gastric shRNA knockdown, overexpression in mouse xenografts binds a DEAD-box RNA helicase (DDX3) to increase transcription factor (YY1) activation of WNT/p-catenin target genes Yang et al., 2019

Tumor suppressive ncRNAs

let-7 miRNA lung antimiRs, mimetics in mouse xenografts or transgenic lung cancer mouse models targets several transcripts that encode oncogenes, including RAS, leading to decreased cell cycle progression and proliferation Trang et al., 2010, Trang et al., 2011
miR-15a/16-1 miRNA prostate, leukemia knockdown, overexpression in mouse xenografts, transgenic knockout mouse targets several transcripts that encode cyclins, CDKs, and anti-apoptotic proteins, thereby increasing apoptosis and inhibiting proliferation Bonci et al., 2008, Klein et al., 2010
miR-34a miRNA lung, prostate, breast overexpression, mimetics, antimiRs in mouse xenografts, mimetics in transgenic lung cancer mouse models targets several oncogenic transcripts encoding cyclins, CDKs, cell adhesion molecules, RTKs, and other non-RTKs, resulting in decreased proliferation, invasion, and suraval Liu et al., 2011, Trang et al., 2011, Kasinski et al., 2012, Adams et al., 2016b
miR-122 miRNA liver transgenic knockout mouse models, overexpression in transgenic liver cancer mouse models targets expression of several genes involved in lipid metabolism, proliferation, and inflammation Hsu et al., 2012, Tsai et al., 2012
miR-506 miRNA ovanan mimetics in mouse xenografts targets SNAI2 transcript to decrease its expression and inhibit migration, invasion, and EMT Yang et al., 2013a
GAS5 lncRNA glioblastoma overexpression in mouse xenografts acts as a nuclear receptor response element mimic for GR, decreases oncogenic miRNA expression, leading to enhanced apoptosis, decreased proliferation, invasion, and migration Zhang et al., 2013, Zhao et al., 2015
LET lncRNA liver, colorectal shRNA knockdown, overexpression in mouse xenografts interacts with and destabilizes a dsRNA binding protein (NF90), a key factor involved in regulation of HIF-1a levels and cell invasion Yang et al., 2013b
MEG3 lncRNA lung overexpression in mouse xenografts increases p53 tumor suppressor levels to activate apoptosis and decrease proliferation Lu et al., 2013
PTENP1 pseudogene RCC, bladder overexpression in mouse xenografts acts as a ceRNA for miRNAs that target the PTEN tumor suppressor transcript, leading to increased apoptosis, decreased proliferation, migration, and invasion Yu et al., 2014, Zheng et al., 2018
circHIPK3 circRNA bladder overexpression in mouse xenografts sponges miRNAs to suppress expression of an endoglycosidase (HPSE), thereby decreasing levels of pro-migratory and angiogenic factors Li et al., 2017

Context-dependent ncRNAs

miR-26a miRNA glioma, leukemia, liver, colorectal overexpression in transgenic glioma, liver cancer mouse models, transgenic overexpression mouse models targets both tumor suppressor (PTEN) and cyclin (CCND2/E2) transcripts to either increase or decrease proliferation, depending on the context Huse et al., 2009, Kota et al., 2009, Mavrakis et al., 2011, Zeitels et al., 2014
miR-29 miRNA leukemia, glioblastoma mimetics in mouse xenografts, transgenic overexpression mouse models promotes proliferation in B cells but can also target and decrease expression of transcipts that enhance lipid synthesis (SCAP, SREBP-1), proliferation (CDK6), and apoptosis (MCL-1) Garzon et al., 2009, Santanam et al., 2010, Ru et al., 2016
MALAT1 lncRNA lung, breast genetic knockout, overexpression in mouse xenografts and allografts, transgenic knockout, overexpression mouse models, ASOs in mouse xenografts or transgenic models promotes expression of genes involved in metastasis, conversely, shown to also inhibit metastasis through interaction with/inhibition of a pro-metastatic transcription factor (TEAD) Gutschner et al., 2013, Arun et al., 2016, Kim et al., 2018
NEAT1 lncRNA prostate, skin, pancreatic shRNA knockdown, overexpression in mouse xenografts, transgenic knockout mouse models mediates oncogenic nuclear receptor (ER) signaling, prevents DNA damage and activation of p53 tumor suppressor, leading to proliferation, invasion, and decreased apoptosis, conversely, shown to also prevent transformation and proliferation in other settings Chakravarty et al., 2014, Adriaens et al., 2016, Mello et al., 2017
NKILA lncRNA breast shRNA knockdown, overexpression in mouse xenografts/PDX binds/inhibits NF-KB and downstream inflammation, which increases apoptosis, reduces invasion, promotes activation-induced cell death in CTLs, T>1 cells, leading to immune evasion Liu et al., 2015, Huang et al., 2018

Abbreviations: AR, androgen receptor, ASO, antisense oligonucleotide, CDK, cyclin-dependent kinase, ceRNA, competitive endogenous RNA, circRNA, circular RNA, CTL, cytotoxic T lymphocyte, EMT, epithelial-to-mesenchymal transition, ER, estrogen receptor, GR, glucocorticoid receptor, hnRNP, heterogeneous nuclear ribonucleoprotein, LNA, locked nucleic acid, lncRNA, long non-coding RNA, miRNA, microRNA, ncRNA, non-coding RNA, PDX, patient-derived xenograft, RCC, renal cell carcinoma, RTK, receptor tyrosine kinase, tsRNA, tRNA-derived small RNA

Figure 1. Oncogenic ncRNAs and cancer-promoting mechanisms.

Figure 1.

Simplified examples of pathways towards oncogenesis for the different classes of ncRNAs discussed in the review. (A) miRNA: miR-155 directly targets and decreases expression of SHIP1, a hematopoietic cell-specific phosphatase that hydrolyzes phosphatidylinositol-3,4,5-triphosphate [PI(3,4,5)P3] to phosphatidylinositol-3,4-bisphosphate [PI(3,4)P2]. Decreased SHIP1 levels can drive AKT signaling, proliferation, and survival, leading to lymphoma. As represented in the figure, clinical trials with anti-miR-155 (NCT02580522/NCT03713320) are underway. (Therapeutic clinical trials targeting the other classes of ncRNAs have yet to be initiated, and are, thus, not depicted.) (B) tsRNA: LeuCAG3’tsRNA (abbreviated Leu3’ in the figure) directly binds and enhances translation of RPS15 and RPS28 transcripts by unfolding of their secondary structures. LeuCAG3’tsRNA can, therefore, increase the levels of these small ribosomal proteins and biogenesis of ribosomes, promoting proliferation of hepatocellular carcinoma cells. (C) lncRNA: IGF2BP1 is an RNA binding protein that forms a messenger ribonucleoprotein (mRNP) complex with other proteins. Interaction of the lncRNA THOR with IGF2BP1 within the mRNP complex leads to stabilization and increased translation of IGF2BP1 target mRNAs (represented by gray curved lines). These proteins (represented as different color ovals) include known oncogenes with wide-reaching effects that can lead to cancers such as melanoma. (D) pseudogene: BRAFP1 acts as a competitive endogenous RNA for BRAF by binding shared miRNAs. This leads to increased BRAF expression and downstream proliferative MAPK signaling that can drive cancers like lymphoma. (E) circRNA: circCTNNBI binds to DDX3 and increases its interaction with YY1 transcription factors, resulting in enhanced transactivation of YY1 target promoters. This includes promoters for many genes involved in Wnt/β-catenin signaling.

Figure 2. Tumor suppressive ncRNAs and pathways inhibiting tumor progression.

Figure 2.

Examples of ncRNAs that can function as tumor suppressors and representative functional mechanisms of inhibition. Examples of tumor suppressive tsRNAs have not yet been mechanistically detailed and are, thus, not included in this figure. (A) miRNA: Among the targets of miR-16-1 are several transcripts that encode cyclins and cyclin-dependent kinases important for the G0/G1-S phase transition of the cell cycle. Decreased expression of these cell cycle regulators can prevent cancer development, and the locus encoding miR-16-1 is often deleted in chronic lymphocytic leukemia. A recent clinical trial (NCT02369198) examined the effects of replenishing this tumor suppressive miRNA in malignant pleural mesothelioma and non-small cell lung cancer using a TargomiR strategy. (B) lncRNA: MEG3 enhances the stability of p53 by directly binding to the protein. MEG3 can also inhibit expression of MDM2, which results in stabilization of p53. Both of these pathways lead to increased p53 activity, inhibition of proliferation, and activation of apoptosis. (C) pseudogene: PTENP1 functions as a competitive endogenous RNA for PTEN by sponging shared miRNA, thereby increasing levels of PTEN. Elevated PTEN levels can cause inhibition of AKT signaling, activation of apoptosis, inhibition of proliferation, and prevent malignancies such as renal cell carcinoma. (D) circRNA: circHIPK3 is an example of a tumor suppressive circRNA that acts as a sponge for miRNA (miR-558 in particular). miR-558 has been suggested to associate with the HPSE (heparanase) promoter and increase expression of heparanase. Therefore, sponging of miR-558 by circHIPK3 decreases heparanase levels which in turn leads to decreased MMP9 and VEGF. These changes result in decreased invasion, migration, and angiogenesis and can inhibit bladder cancer progression.

Oncogenes:

Research has shown that miRNAs can function as oncogenes, promoting abnormal cell growth and contributing to tumor formation. These miRNAs may directly inhibit the activity of tumor suppressors or work indirectly by removing the genetic brakes on oncogene activity. For example, miR-155 can promote abnormal B-cell proliferation, setting in process a series of changes that eventually lead to the development of leukemia and lymphoma (Babar et al., 2012; O’Connell et al., 2009). Importantly, delivery of antimiRs targeting miR-155 can inhibit tumor growth (Babar et al., 2012; Cheng et al., 2015) (Figure 1A). Another miRNA, miR-21, is overexpressed in pre B-cell lymphoma and has been implicated in other cancers, such as lung cancer, through targeting negative regulators of Ras signaling (Hatley et al., 2010; Medina et al., 2010). In glioblastomas, an oncogenic miRNA, miR-10b, is expressed at higher levels than in normal brain tissue and is required for tumor growth (El Fatimy et al., 2017). These oncogenic miRNAs exhibit the phenomenon of oncogene addiction (oncomiR addiction), since the tumors are dependent on the continued expression of the miRNAs for survival (Cheng and Slack, 2012), and are, thus, important potential targets in anti-cancer therapy. Recently, the potential for targeting oncomiRs uniquely overexpressed in an individual patient’s tumors was demonstrated, opening the way to personalized miRNA therapy as we discuss at the end of the review (Gilles et al., 2018).

Tumor suppressors:

Research suggests that miRNAs can also act as tumor suppressors, and when their function is lost, so is their protective power. For instance, the conserved miRNA let-7 represses RAS, a family of oncogenes implicated in about one-third of all human cancers (Johnson et al., 2007; Johnson et al., 2005). Consequently, let-7 expression can reduce levels of RAS, suggesting that it acts as a tumor suppressor and can be used as a new and promising therapeutic agent (Trang et al., 2010; Trang et al., 2011). As another example, miR-15a and miR-16-1 typically act as tumor suppressors but, when mutated or deleted, are associated with the development of chronic lymphocytic leukemia (CLL), the most common type of leukemia (Calin et al., 2002; Calin et al., 2005; Klein et al., 2010) (Figure 2A). Furthermore, miR-34a, a member of the conserved, redundant miR-34 family of miRNAs (Concepcion et al., 2012), regulates the expression of several oncogenes and is a direct downstream target of p53 (Adams et al., 2016a; Adams et al., 2016b; Kasinski and Slack, 2012; Liu et al., 2011). These miRNAs are also disabled in other types of cancer, such as multiple myeloma, mangle cell lymphoma, and prostate cancer (Volinia et al., 2006).

Context-dependent:

Some miRNAs function as either tumor suppressors or oncogenes, depending on the context. A notable example is miR-29, which helps prevent disease progression in the indolent form of B-CLL, but is also elevated in acute myeloid leukemia and the more aggressive form of B-CLL, implying that this miRNA can also function as an oncogene (Pekarsky and Croce, 2010).

Transfer RNA-derived small RNAs (also called tRNA fragments)

Just in the last decade, a new group of clinically-relevant small ncRNAs was discovered that are derived from tRNAs, which we will collectively refer to as tRNA-derived small RNAs (tsRNAs) (Cole et al., 2009; Lee et al., 2009); these molecules have also been referred to as tRNA fragments (tRFs) or tRNA-derived stress-induced RNAs (tiRNAs). Many aspects of their biogenesis and downstream mechanisms of action are still being unraveled, along with implementation of a universal nomenclature system. Transfer RNAs are transcribed by RNA Pol III as pre-tRNAs and then undergo modification and processing to produce mature tRNAs. A tRNA can generate different types of tsRNAs, depending on where cleavage occurs and whether it is in the pre-tRNA transcript or mature tRNA. The enzymes responsible for generation of tsRNAs are still being fully elucidated, but roles for ribonucleases such as Angiogenin, Dicer, and RNase Z have been noted in some instances (see recent reviews by (Balatti et al., 2017; Kumar et al., 2016; Saikia and Hatzoglou, 2015)). Several tsRNA databases have already been constructed for this new class of small ncRNAs to assist in their characterization. MINTbase v2.0 analyzed all datasets from The Cancer Genome Atlas (TCGA) and identified 26,531 unique tsRNA sequences from cancerous tissues (only fragments resulting from mature tRNAs were included in the algorithm) (Pliatsika et al., 2018). Despite an impressive number of distinct tsRNA molecules in cancer cells and some unique mechanisms of action, tsRNAs overlap with miRNAs in some aspects. Like miRNAs, tsRNAs have been found associated with Argonaute proteins, can mediate translational repression of mRNAs via binding to target 3’UTRs, and can be either oncogenic or tumor suppressive (Goodarzi et al., 2015; Kuscu et al., 2018; Maute et al., 2013). In a recent, exciting study, it was discovered that a tsRNA derived from a Leucine tRNA plays a role in global protein translation by regulating the expression of genes coding for ribosomal components (Table 1, Figure 1B). Importantly, the researchers show that this tsRNA is upregulated in liver tumors and that targeting the tsRNA with a LNA oligonucleotide in vitro and in vivo causes liver tumor cells to undergo cell death (Kim et al., 2017). These results demonstrate that tsRNAs are another potential new target for anti-cancer therapy.

PIWI-interacting RNAs

PIWI-interacting RNAs are a class of small RNAs approximately 21-35 nucleotides in length normally associated with the PIWI subfamily of Argonaute proteins and transposon silencing in germline development (Aravin et al., 2006; Girard et al., 2006). Species-specific pathways exist for production of piRNA precursors, and piRNA sequences are not very conserved. In mammals, piRNAs are processed from long, single-stranded transcripts whose genomic loci are clustered throughout the genome and transcribed by RNA Pol II, and approximately 20,000 piRNAs exist in the human genome (Ng et al., 2016). As noted above, to mediate their effects, miRNAs associate with the ubiquitously expressed AGO family of Argonaute proteins. In contrast, as their name implies, piRNAs are loaded onto proteins of the PIWI family of Argonaute proteins, which are often restricted to gonadal cells. The classic function of piRNAs is to silence transposons, and they do this in two ways. In the first mechanism, piRNAs guide PIWI proteins to nascent transposon transcripts and generate repressive chromatin states at target transposon promoters to silence their transcription. In the second scenario, piRNAs guide the PIWI complex to the transposon mRNA, where it cleaves the transcript (see (Ng et al., 2016; Ozata et al., 2019) for recent reviews). Although classically thought of as functioning only in gonadal cells, recent work shows that some of the tens of thousands of piRNAs are expressed in somatic tissues, albeit it at very low levels, and misexpressed in cancers, suggesting that these small ncRNAs might also be useful biomarkers (Martinez et al., 2015; Mei et al., 2015; Ng et al., 2016). However, the functional roles of piRNAs in somatic tissue and cancer are still being elucidated.

Long ncRNAs

Some of the earliest lncRNAs to be discovered were XIST and H19 (Bartolomei et al., 1991; Brown et al., 1991). Although initially identified through studies characterizing X-chromosome inactivation and embryonic development, respectively, these two genes are now among a long list of lncRNAs to be mechanistically linked to several types of cancers. Long ncRNAs are characterized as non-coding transcripts greater than 200 base pairs in length transcribed by RNA Pol II from independent promoters. Similar to protein-coding genes, their genomic locations are marked by enrichment of H3K4 trimethylation at the transcriptional start site and H3K36 trimethylation throughout the gene body. Long ncRNA transcripts consist of multiple exons that are spliced through canonical mechanisms into a mature transcript and usually include 5’caps and 3’poly(A) tails. However, lncRNAs have fewer exons and are expressed at lower levels overall compared to protein-coding transcripts (Cabili et al., 2011; Derrien et al., 2012; Iyer et al., 2015). Interestingly, lncRNAs are also not highly evolutionarily conserved, with only 5-6% of lncRNAs harboring conserved sequences (Iyer et al., 2015). In regards to this, there is a line of thinking that highly conserved IncRNAs may be more likely to be functional. However, there are primate-specific lncRNAs which likely could be involved in disease processes. RNA-sequencing (RNA-seq) has shown that there are much higher absolute numbers of unique lncRNA transcripts compared to protein-coding genes. A recent extensive cancer-centric study, MiTranscriptome, analyzed 7256 RNA-seq libraries primarily from human tissues (5298 primary tumors, 281 metastases, and 701 normal/benign) and identified 58,648 lncRNAs. Furthermore, these datasets allowed for an in-depth exploration of the landscape of lncRNAs in cancer and led to the identification of 7941 lncRNAs that were cancer- and/or lineage-specific (Iyer et al., 2015).

Compared to small ncRNAs, lncRNAs exhibit extensive mechanistic diversity to carry out their functional roles and, therefore, require a lengthier discussion in this area. Long ncRNAs can function either in cis or trans, meaning they mediate local effects near their own sites of transcription (cis), or they operate at distant genomic or cellular locations (trans). Different lncRNAs have also been shown to be able to influence gene expression at all levels-epigenetic, transcriptional, and post-transcriptional. Multiple lncRNAs bring other regulatory molecules (e.g., mRNAs, miRNAs, DNA) into proximity with one another and with proteins (e.g., chromatin modifying complexes, transcription factors, E3 ligases, RNA-binding proteins (RBPs)), essentially creating a flexible molecular scaffold that fosters the chemical interactions necessary to sustain cellular activity (see recent reviews, (Anastasiadou et al., 2018b; Kopp and Mendell, 2018)).

In terms of lncRNAs functioning by directly binding to protein complexes, one of the most well-characterized mechanisms is guiding of chromatin modifying complexes to target gene promoters to influence transcriptional repression/activation. Noted examples include the following: HOTAIR binds PRC2 and LSD1/CoREST/REST on its 5’ and 3’ ends, respectively, to target the complex to promoters and modulate histone methylation levels (Tsai et al., 2010); SChLAP1, an aggressive prostate cancer-specific lncRNA, interacts directly with the SWI/SNF nucleosome remodeling complex (Prensner et al., 2013); ANRIL interacts with components of PRC1 and PRC2 to silence genes in the CDKN2B/2A tumor suppressor gene cluster (Yap et al., 2010). Long ncRNAs also commonly bind transcription factors, which can have broad downstream effects on cellular transcriptional programs. Intriguingly, the GAS5 lncRNA acts as a mimic of a glucocorticoid response element (GRE), directly binding the DNA-binding domain of the glucocorticoid receptor (GR) and preventing it from activating its target genes, including those that prevent apoptosis (Hudson et al., 2014; Kino et al., 2010). Other direct interaction partners include PANDA and NF-YA transcription factors that are critical activators of p53-targeted cell death genes (Hung et al., 2011), PCGEM1 enhancement of c-Myc activity as a master regulator of metabolism (Hung et al., 2014), NKILA directly binding to the NF-KB/IKB complex to block phosphorylation of IKB by IKK (Liu et al., 2015), and DINO interacting with and stabilizing p53 following DNA damage (Schmitt et al., 2016). Long ncRNAs are often also found in direct contact with RBPs that regulate mRNA processing and stability. The effects of HOTAIR are mediated by binding to hnRNPA2/B1, which function as “matchmakers” to target HOTAIR/PRC2 to mRNA transcripts (Meredith et al., 2016). Recently, the novel, ultraconserved lncRNA THOR was discovered and shown to function as an oncogene by stabilizing the binding of IGF2BP1 to target mRNAs (Hosono et al., 2017) (Figure 1C). Finally, lncRNAs can function as scaffolds for regulatory molecules found in nuclear speckles and paraspeckles, exemplified by lncRNAs NEAT1 and MALAT1 (Clemson et al., 2009; Tripathi et al., 2010).

In addition to proteins, lncRNAs can directly bind to nucleic acids to mediate their molecular mechanisms of action. Two interesting examples of this include the recently identified lncRNA ARLNC1 that interacts with AR mRNA to regulate its cytoplasmic levels in prostate cancer (Zhang et al., 2018), and LincRNA-p21, which directly binds JUNB and CTNNB1 transcripts to repress their translation (Yoon et al., 2012). Another common mechanism involves lncRNA acting as a competitive endogenous RNA (ceRNA) or “sponge” for miRNAs. One of the first lncRNAs to be identified, H19, acts as a decoy for several tumor suppressor miRNAs, with let-7 as one notable example (Kallen et al., 2013). TUG1 has been shown to act as a sponge for miRNAs that target PTEN in prostate cancer (Du et al., 2016), as well as those that can target SOX2 and MYC in glioma cells (Katsushima et al., 2016). Long ncRNAs have furthermore been found to interact directly with DNA. GA-rich motifs in target promoters of genes involved in the TGFβ pathway attract GA-rich sequences of the MEG3 lncRNA via RNA-DNA triplex formation, and PRC2 is pulled along via interaction with MEG3 (Mondal et al., 2015).

Given the wide range of regulatory mechanisms and the diversity of downstream pathways affected, it is not surprising that many lncRNAs, including several of those highlighted above, have been found through in vivo experiments to be important contributors to cancer progression and represent possible therapeutic targets (Table 1). Due to their poor species conservation, most of these studies have relied upon modulating expression of lncRNAs in human cancer cell lines xenografted in mice. Like miRNAs, lncRNAs have been found to function either as tumor suppressors, oncogenes, or exhibit context-dependent roles (Table 1).

Oncogenes:

Many lncRNAs whose cellular roles have been elucidated function as oncogenes that promote tumor growth and are often overexpressed in cancer. HOTAIR is one of the most well-studied oncogenic lncRNAs that was initially characterized as a regulator of the HOX family of genes, which help control cellular identity (Rinn et al., 2007). However, a more global role for HOTAIR in controlling gene repression through targeting of PRC2 and LSD1/CoREST/REST was quickly noted (Gupta et al., 2010; Tsai et al., 2010). HOTAIR overexpression has been associated with poor outcomes in breast and several other cancers, possibly by increasing metastasis and tumor invasiveness (Balas and Johnson, 2018; Bhan et al., 2017; Gupta et al., 2010; Li et al., 2016b). Several novel oncogenic lncRNAs have been functionally described only in the past few years, including THOR (Hosono et al., 2017) (Figure 1C) and ARLNC1 (Zhang et al., 2018) mentioned previously, as well as SAMMSON (Leucci et al., 2016), DSCAM-AS1 (Niknafs et al., 2016), lncARSR (Qu et al., 2016a), CamK-A (Sang et al., 2018), and EPIC1 (Wang et al., 2018) (Table 1), and this list will undoubtedly continue to grow. SAMMSON has garnered much attention in particular lately as a melanoma-specific IncRNA that is required for growth and survival of cells, regardless of TP53, BRAF, or NRAS mutational status (Leucci et al., 2016). Mechanistically, SAMMSON promotes cell growth through interactions with p32, CARF, and XRN2 proteins that help to balance ribosomal RNA (rRNA) maturation and protein synthesis in the cytosol and mitochondria (Leucci et al., 2016; Vendramin et al., 2018). Importantly, GapmeR silencing of SAMMSON in patient-derived xenograft (PDX) models inhibits tumor growth, demonstrating the therapeutic potential of this lncRNA (Leucci et al., 2016).

Tumor suppressors:

Some lncRNAs act as safeguards against cancer development by preventing proliferation, activating apoptosis, maintaining genomic stability, or promoting tumor suppressor expression. For instance, MEG3 is one of the most well-characterized tumor suppressive lncRNAs. In addition to regulating the TGFβ pathway noted above, MEG3 downregulates MDM2 expression and increases p53 protein levels (Figure 2B); regulation of these and other pathways leads to decreased cell proliferation (Lu et al., 2013; Mondal et al., 2015; Zhang et al., 2010; Zhou et al., 2007). Consistent with its status as a tumor suppressor, copy loss of MEG3 and increased CpG methylation of its promoter is often noted in cancers (Balas and Johnson, 2018; Zhang et al., 2010). GAS5 is another lncRNA with known tumor suppressive roles often downregulated in cancers and with direct in vivo experimental evidence in breast and glioblastoma models (Bhan et al., 2017; Zhang et al., 2013; Zhao et al., 2015). In addition to affecting GR signaling, GAS5 is also involved in interactions with miRNAs that ultimately can lead to decreased cell proliferation and increased apoptosis, as well as decreased migratory potential (Hudson et al., 2014; Kino et al., 2010; Zhang et al., 2013; Zhao et al., 2015).

Context-dependent:

Many lncRNAs display both tumor suppressive and oncogenic functions. For example, in breast cancer, the lncRNA NKILA negatively regulates NF-KB signaling and downstream inflammation. In mouse xenograft models with human breast cancer cell lines overexpressing NKILA, this translates into reduced metastasis and increased survival, suggesting a tumor suppressive role (Liu et al., 2015). However, it was also recently shown that increased NKILA can promote tumor immune evasion, an oncogenic property, through activation-induced cell death of cytotoxic T lymphocytes (CTLs) and TH1 cells (Huang et al., 2018).

Pseudogenes

Pseudogenes are a subclass of lncRNA transcripts that are similar in sequence to coding genes but have become inactivated and no longer result in functional proteins. RNA-seq studies have found the expression of thousands of pseudogenes from across the genome, with many of these shown to be specific to particular cancer types (Kalyana-Sundaram et al., 2012). Although pseudogenes are sometimes dismissed as useless evolutionary relics, research suggests that they perform vital functions and can both protect against or contribute to the development of cancer. One way that pseudogenes function is to act as decoys that divert miRNAs and other molecules away from their coding gene counterparts. The PTENP1 pseudogene is almost identical in size and structure to a coding gene, PTEN, which is a powerful tumor suppressor. When functioning normally, the pseudogene PTENP1 can act as a decoy to sponge miRNAs that would otherwise reduce the production of PTEN (Poliseno et al., 2010; Yu et al., 2014; Zheng et al., 2018) (Figure 2C). By acting in this manner as a ceRNA, the PTENP1 pseudogene functions as a tumor suppressor (Table 1). Other pseudogene ceRNAs can promote cancer growth. Recently, abnormal levels of the pseudogene BRAFP1 were shown to cause aggressive B-cell lymphoma in a mouse model. When overexpressed, the pseudogene operated as a miRNA decoy that increased the amount of BRAF protein (Figure 1D). In fact, the mice with the abnormal pseudogene developed cancer as rapidly as those with the protein-coding counterpart BRAF oncogene (Karreth et al., 2015). This suggests that far from being “dead” genes, pseudogenes may contribute directly to cancer development. Although the research is just beginning, it is becoming clearer that pseudogenes are more active in physiology and cancer than previously appreciated.

Circular RNAs

Within the IncRNA group are also circRNAs, which, as their name implies, are single-stranded covalently closed RNA molecules. These molecules are often generated through a back-splicing process, but the exact mechanisms of circRNA biogenesis remain to be fully elucidated. However, it has been shown that the parent gene expression of circRNAs is not a predictor of circRNA expression. Factors that do seem to be associated with circRNAs are longer flanking intron lengths and increased presence of intronic repetitive and reverse complement elements (Chen et al., 2019; Ivanov et al., 2015; Jeck et al., 2013; Vo et al., 2019). Several databases of circRNAs found in cell lines have emerged over the past few years, but studies centered on the direct identification of circRNA species in clinical samples were only recently conducted. In one study, a global analysis of circRNAs using clinical tumor samples (2000+) was performed across more than 40 cancer sites and led to the generation of the MiOncoCirc database. Notably, these efforts identified over 160,000 significantly expressed circRNAs (Vo et al., 2019). Another recent study, specifically in localized prostate cancer, identified 76,311 circRNAs through RNA-seq of prostate tumor specimens (Chen et al., 2019). Combined, these studies demonstrate the high prevalence of circRNAs in cancer. Interestingly, overall global circRNA abundance has been demonstrated to be negatively correlated with cell proliferation (Bachmayr-Heyda et al., 2015; Vo et al., 2019).

Despite widespread expression of circRNAs, most mechanisms of action and functional roles are still being elucidated. For the handful of circRNAs that have been functionally studied, mechanisms similar to other lncRNAs have been described, as well as the ability to function as oncogenes or tumor suppressors (Table 1). As one mechanism of action, like lncRNAs, circRNAs have been noted to sponge miRNAs, as is the case for circCCDC66, which has oncogenic functions in colorectal cancer (Hsiao et al., 2017) and circHIPK3, which has tumor suppressive roles in bladder cancer (Li et al., 2017) (Figure 2D). Interactions of circRNAs with miRNAs may also function to stabilize the miRNA binding partner, which was recently suggested to occur with circCSNK1G3 and miR-181 in prostate cancer, leading to enhanced cell proliferation (Chen et al., 2019). Circular RNAs have also been proposed to bind directly to proteins to either act as a scaffold for complex assembly, such as for circCTNNBI and DDX3 (Yang et al., 2019) (Figure 1E), or to sponge the protein of interest to prevent it from mediating its actions (Kristensen et al., 2018). It is also important to note that while we have discussed circRNAs in the context of ncRNAs, there have also been studies showing that some circRNAs may be translated (Legnini et al., 2017; Pamudurti et al., 2017). While functional studies and assessments of the therapeutic potential of circRNAs are only in their infancy, it is clear that circRNAs possess enhanced stability (Vo et al., 2019), making them prime candidates for novel cancer biomarkers, discussed further below.

Clinical relevance of ncRNAs

A search through the Clinicaltrials.gov database with keywords associated with ncRNAs shows hundreds of clinical trials involving ncRNAs (Supplemental Table 1), with many of these being cancer trials and the vast majority, over 800 trials, involving miRNAs. Given the high specificity of detection methods for ncRNAs and their tissue-specificity, as well as a plethora of supporting data in discovery studies, it is not surprising that the vast majority of these trials are diagnostic in nature, where the ncRNAs are used as biomarkers for a disease state or a prognostic marker for disease outcome. However, given the recent success of RNAi-based and oligo-based drugs (Levin, 2019; Pecot et al., 2011), a number of therapeutic clinical trials with ncRNAs have begun (Rupaimoole and Slack, 2017), albeit with mixed success. Below, we highlight notable ncRNA diagnostic and therapeutic cancer trials that are currently active, or just recently completed, and have the potential to impact patient care.

Non-coding RNAs as cancer biomarkers

Non-coding RNAs are useful molecular biomarkers since deregulated ncRNA expression is observed across diverse cancers. Numerous large-scale discovery studies in the pre-clinical setting have shown the promise of ncRNAs as diagnostic and prognostic biomarkers, with prominent examples outlined in Table 2. Notable examples can be found across both small (miRNAs) and long ncRNA (lncRNAs, circRNAs) classes. Although they are only beginning to be functionally described, circRNAs are a particularly intriguing class of biomarkers due to their enhanced stability, resulting from covalently closed ends resistant to exoribonucleases (Schwanhausser et al., 2013; Vo et al., 2019). Importantly, as shown in Table 2, several biomarkers can be found in blood or urine, where acquisition is much less invasive than that required for obtaining tumor biopsies. Expression of a given biomarker can detect the presence of cancer compared to normal samples, or predict survival, metastasis, or therapy response once cancer has been found.

Table 2.

Non-coding RNA cancer biomarkers

Name ncRNA class Cancer Example of biomarker utility Source material References
miR-10b miRNA pancreatic detection; prognostic for MFS and OS tissue Preis et al., 2011
miR-16 miRNA lung prognostic for OS blood Wang et al., 2013
miR-21 miRNA breast prognostic for OS tissue Yan et al., 2008
colorectal detection; prognostic for OS blood; tissue Toiyama et al., 2013
leukemia prognostic for OS blood Rossi et al., 2010
lung prognostic for CSS and RFS tissue Saito et al., 2011
prostate detection blood Agaoglu et al., 2011
miR-34a miRNA lung prognostic for RFS tissue Gallardo et al., 2009
miR-155 miRNA leukemia detection; prognostic for OS blood Ferrajoli et al., 2013
miR-221 miRNA prostate detection blood Agaoglu et al., 2011
miR-375 miRNA prostate prognostic for OS blood Huang et al., 2015
miR-506 miRNA gastric prognostic for OS tissue Sakimura et al., 2015
ovarian prognostic for OS and PFS tissue Yang et al., 2013a
pancreatic prognostic for OS tissue Li et al., 2016a
miR-1290 miRNA colorectal detection; prognostic for OS and RFS blood; tissue Imaoka et al., 2016
prostate prognostic for OS blood Huang et al., 2015
CamK-A lncRNA breast prognostic for OS and RFS tissue Sang et al., 2018
CCAT1 lncRNA colorectal prognostic for OS, CSS, and RFS tissue McCleland et al., 2016; Ozawa et al., 2017
CCAT2 lncRNA colorectal prognostic for OS and RFS tissue Ozawa et al., 2017
EPIC1 lncRNA breast prognostic for OS tissue Wang et al., 2018
FAL1 lncRNA ovarian prognostic for OS tissue Hu et al., 2014
H19 lncRNA gastric detection blood Zhou et al., 2015
HOTAIR lncRNA breast prognostic for OS and MFS tissue Gupta et al., 2010
colorectal detection; prognostic for OS blood; tissue Kogo et al., 2011; Svoboda et al., 2014
ESCC prognostic for OS tissue Li et al., 2013
ovarian prognostic for OS in carboplatin-treated patients tissue Teschendorff et al., 2015
pancreatic prognostic for OS tissue Kim et al., 2013
HOTTIP lncRNA liver prognostic for OS tissue Quagliata et al., 2014
HULC lncRNA liver detection blood; tissue Panzitt et al., 2007; Xie et al., 2013
LINK-A lncRNA breast prognostic for RFS tissue Lin et al., 2016
lncARSR lncRNA RCC prognostic for OS and RFS; prognostic for PFS in sunitinib-treated patients blood; tissue Qu et al., 2016a; Qu et al., 2016b
MALAT1 lncRNA lung prognostic for OS in early-stage disease tissue Ji et al., 2003
prostate detection blood; urine Ren et al., 2013; Wang et al., 2014
NEAT1 lncRNA prostate detection; prognostic for MFS, BCR, and CSS tissue Chakravarty et al., 2014
PCA3 lncRNA prostate detection urine Hessels et al., 2003
PCAT-1 lncRNA prostate detection tissue Prensner et al., 2011
PCAT-14 lncRNA prostate detection; prognostic for OS, BCR, MFS, and CSS tissue Shukla et al., 2016; White et al., 2017
SChLAP1 lncRNA prostate prognostic for BCR, MFS, and CSS tissue Prensner et al., 2014b; Mehra et al., 2016
UCA1 lncRNA bladder detection urine Wang et al., 2006
circAR circRNA prostate detection of mCRPC versus primary prostate cancer tissue Vo et al., 2019
circCCDC66 circRNA colorectal detection; prognostic for OS tissue Hsiao et al., 2017
circCTNNB1 circRNA gastric prognostic for OS tissue Yang et al., 2019
ciRS-7 circRNA colorectal detection; prognostic for OS tissue Weng et al., 2017

Abbreviations: BCR, biochemical recurrence; circRNA, circular RNA; CSS, cancer-specific survival; ESCC, esophageal squamous cell carcinoma; IncRNA, long non-coding RNA; mCRPC, metastatic castration-resistant prostate cancer; MFS, metastasis-free survival; OS, overall survival; PFS, progression-free survival; RCC, renal cell carcinoma; RFS, recurrence-free survival

Of the biomarkers listed in Table 2, PCA3 is the first and only ncRNA to receive FDA-approval as a cancer biomarker test to date. PCA3 is a prostate-specific marker often overexpressed in prostate cancer, and importantly, can be detected easily through non-invasive urine collection (Hessels et al., 2003). When combined with urine TMPRSS2:ERG levels, another transcript often overexpressed in prostate cancer, the diagnostic power of PCA3 is even stronger (Tomlins et al., 2011; Tomlins et al., 2016). With expression data from thousands of ncRNAs now available across disease sites, the potential to identify new single biomarkers (as in Table 2) or panels of biomarkers is endless. The four highlighted below involve compelling science and are illustrative examples of the hundreds of ongoing clinical trials centered on ncRNA-related biomarkers in cancer.

“Addition of microRNA Blood Test to Lung Cancer Screening Low Dose CT” (NCT03452514)

A trial sponsored by Hummingbird Diagnostics and conducted at Harvard Medical School and Lahey Clinic is currently enrolling to determine whether a miRNA profile (HMBDx, proprietary to Hummingbird) is superior to the specificity of low dose computed tomography (LDCT) for the diagnosis of lung cancer. Target enrollment is 400 participants who meet the criteria for lung cancer screening. Here, a blood draw will be used to screen for a serum miRNA signature (Keller et al., 2011a; Keller et al., 2011b; Leidinger et al., 2011) prior to LDCT and correlated with future outcome after a minimum 12 months of follow-up. Although the ability of CT to detect lung nodules is high, there are the caveats of false positives and repeated exposure to radiation. Being able to use a simple, non-invasive blood-based miRNA test would be a significant step forward for lung cancer diagnostics.

“Evaluating the Expression Levels of MicroRNA-10b in Patients with Gliomas” (NCT01849952)

Glioblastoma is one of the most aggressive types of cancer, and the development of novel early biomarkers and therapeutic targets is critically needed. As mentioned above, the oncogenic miR-10b is highly expressed in glioblastomas and required for tumor growth (El Fatimy et al., 2017). This multi-site clinical trial is being sponsored by and conducted at Dartmouth-Hitchcock Medical Center, in addition to Tufts Medical Center, Massachusetts General Hospital, and University of Vermont. The trial will test whether miR-10b expression levels in primary tumor, blood, and cerebrospinal fluid samples from glioblastoma patients can serve as a prognostic and diagnostic marker of glioblastoma. Patient survival, as well as tumor grade and genotypic variations, will be correlated to miR-10b expression levels in an anticipated 200 participants with brain tumors of glial origin.

“DICER1-related Pleuropulmonary Blastoma Cancer Predisposition Syndrome: A Natural History Study” (NCT01247597)

This large observational study, aiming to enroll 1500 participants, is sponsored by the National Cancer Institute (NCI). As mentioned above, Dicer1 is a ribonuclease involved in the miRNA biogenesis pathway. (Hill et al., 2009). This trial seeks to identify the specific types of cancer and benign neoplasms associated with an inherited mutation in DICER1, in particular in pleuropulmonary blastoma (PPB), a rare lung tumor that appears in young children. Researchers are collecting further medical history and genetic information on individuals and close relatives of individuals who have PPB or other rare tumors associated with PPB (e.g., cystic nephroma, nasal chondromesenchymal hamartoma, ovarian Sertoli-Leydig cell tumors, ocular medulloepithelioma) to better understand this disease.

“Non-coding RNA in the Exosome of the Epithelia Ovarian Cancer” (NCT03738319)

This case-control observational study sponsored by Peking Union Medical College Hospital will use next-generation sequencing to analyze the expression of miRNAs and lncRNAs in blood samples from 160 patients with high grade serous ovarian cancer (HGSOC) and benign gynecologic diseases. Differential expression of miRNAs and lncRNAs will be compared between cancer and control groups, and candidate ncRNAs will be validated as biomarkers for the detection and prognosis of ovarian cancer. Completion of this trial will hopefully lead to promising clinical ncRNA biomarkers that can be used to aide in early detection of this disease, which is often not diagnosed until an advanced stage (Giannopoulou et al., 2018).

Therapeutic targeting of ncRNAs in cancer clinical trials

As discussed throughout this review, ncRNAs are important regulatory molecules that can either drive or prevent oncogenic processes (Table 1, Figures 1 and 2). Accordingly, development of effective therapies to silence (for oncogenes) or overexpress (for tumor suppressors) ncRNAs has been an active area of investigation in recent years (Adams et al., 2017; Arun et al., 2018; Rupaimoole and Slack, 2017). In vivo inhibition of ncRNAs often requires oligonucleotide targeting of the ncRNA as well as a delivery method. For targeting of miRNA in which overexpression is desired, synthetic oligoribonucleotides consisting of a miRNA duplex are used (miRNA mimetics). Conversely, for achieving inhibition of oncogenic miRNA, single-stranded antisense RNAs are employed (antimiRs or antagomiRs) (Rupaimoole and Slack, 2017). Chemical modifications are often made to RNA drugs in order to increase stability and affinity in vivo. Common examples include locked nucleic acid (LNA) modification to lock the confirmation of the sugar phosphate backbone of the RNA and substitutions to the 2’-OH of the ribose sugar, such as 2’-O-methyl (2’-O-Me) or 2’-O-methoxyethyl (2’-O-MOE) modifications (Adams et al., 2017). In addition to optimization of targeting design, RNA drugs often require an in vivo delivery method, with lipid-based nanocarriers being the most well-established, and exciting new advances in peptide and polymer delivery systems also being developed (Adams et al., 2017; Rupaimoole and Slack, 2017). Long ncRNAs can be inhibited in vivo through approaches like antisense oligonucleotide (ASO) technology, which results in RNase H-dependent degradation of the target lncRNA (Arun et al., 2018). Specific chemical modifications can also be made to lncRNA-targeting ASOs to increase their stability, such as LNA, 2’-O-Me, or 2’-O-MOE mentioned above, S-constrained ethyl (cEt) modifications, or “GapmeR” RNA-DNA-RNA hybrid ASOs. This optimization has even allowed for development of the next-generation of ASOs with increased potency that are freely uptaken in vivo, without a delivery method (Hong et al., 2015; Yamamoto et al., 2015).

To date, most studies employing ncRNA-targeting drugs in the cancer setting have been in the pre-clinical stage, but these studies have nonetheless highlighted the promise of this strategy. For example, ASO-based depletion of the lncRNA MALAT1 can impact growth and metastasis of lung and breast cancer cells in murine models (Arun et al., 2016; Gutschner et al., 2013). Below, we expand upon the two therapeutic clinical trials that have been completed, and one which has reported preliminary results, all of which targeted miRNAs. Since no therapeutic trials directly targeting the other classes of ncRNAs have been completed, an alternative strategy harnessing the H19 lncRNA promoter in a clinical trial in bladder cancer is discussed. As research and technology progresses, we are sure to see more ncRNA-based therapies advance to the clinic.

“A Multicenter Phase 1 Study of MRX34, MicroRNA miR-RX34 Liposomal Injection” (NCT01829971/NCT02862145)

This international study was sponsored by Mirna Therapeutics (now Synlogic Therapeutics) and conducted at multiple sites in the USA and Korea. It evaluated the safety of MRX34 in patients with unresectable primary liver cancer, select advanced or metastatic cancer with or without liver involvement, or hematologic malignancies. The drug was a liposomal miR-34 mimetic given intravenously. As mentioned above, miR-34 is a tumor suppressive miRNA that is a direct target of p53 and regulates the expression of several oncogenes (Adams et al., 2016a; Adams et al., 2016b; Kasinski and Slack, 2012; Liu et al., 2011). Forty-seven patients received drug and a maximum tolerated dose (MTD) was determined, while a handful of patients experienced partial responses (Beg et al., 2017). From this study, the authors concluded that MRX34 showed some anti-tumor activity and was safe. Unfortunately, in a Phase 1b study, multiple patients experienced immune-related toxicities, some patients experienced severe (Grade 4) cytokine release syndrome, and the trial was terminated. Since the final results of the trial have not been published, it is unclear why these patients experienced severe adverse events (SAEs), but it should be noted that both lipid-based nanoparticles and small double-stranded RNAs have been reported to cause immune toxicity (discussed in (Levin, 2019)). A clinical trial (NCT02716012) using the same lipid nanoparticle, known as a Smarticle, but with a different dsRNA (targeting the protein-coding gene CEBPA (Reebye et al., 2018; Voutila et al., 2017)) from MiNA Therapeutics recently reported no SAEs at the 2019 European Society for Medical Oncology Congress.

“MesomiR 1: A Phase 1 Study of TargomiRs as 2nd or 3rd Line Treatment for Patients with Recurrent MPM and NSCLC” (NCT02369198)

This study, sponsored by the Asbestos Diseases Research Foundation and EnGeneIC Limited and conducted at the University of Sydney, evaluated the MTD of TargomiRs in patients with recurrent malignant pleural mesothelioma (MPM) and non-small cell lung cancer (NSCLC). The intravenously-delivered TargomiR drug was a minicell containing a double-stranded, 23 base pair, synthetic miRNA mimic based on tumor suppressive miR-16, discussed previously (Reid et al., 2013) (Figure 2A). The drug delivery vehicle was constructed from nonliving bacterial minicells coated with a targeting moiety, an anti-EGFR antibody. This specifically targeted the particle and its miRNA payload to EGFR-expressing cancer cells. 26 patients were treated with TargomiRs, and a MTD was determined (van Zandwijk et al., 2017). One patient showed a partial response to therapy, leading the investigators to conclude that their studies supported further trials, perhaps in combination with other current therapies.

“Safety, Tolerability, and Pharmacokinetics of MRG-106 in Patients with Mycosis Fungoides (MF), CLL, DLBCL (Diffuse Large B-Cell Lymphoma), or ATLL (Adult T-Cell Leukemia/Lymphoma)” (NCT02580552/NCT03713320)

The objectives of these studies, sponsored by miRagen Therapeutics and conducted at multiple sites in the USA and Canada, are to evaluate the safety and potential efficacy of cobomarsen, also known as MRG-106, in patients diagnosed with a subset of lymphomas and leukemias. Cobomarsen is a LNA designed to inhibit the activity of miR-155 (Figure 1A), an oncogenic miRNA found at high levels in these types of cancers and important for the growth and survival of MF cancer cells (Seto et al., 2018). In preliminary results presented at the American Society for Hematology in 2018, the authors showed that cobomarsen was well tolerated in 43 patients treated either intratumorally or systemically. Furthermore, some patients had durable partial responses.

“Codex: Study of Inodiftagene Vixteplasmid (BC-819) in Unresponsive NMIBC” (NCT03719300)

H19 is a lncRNA that possesses several oncogenic mechanisms of action and is often upregulated in different types of cancer (Bhan et al., 2017). Due to its upregulation in cancer cells, researchers have used its promoter to deliver cancer-specific expression of downstream sequences. One compound, BC-819 (also known as DTA-H19), is a DNA plasmid expressing diphtheria toxin A (DTA) under the regulation of the H19 promoter, delivered as a complex with polyethyleneimine (PEI). BC-819 has completed Phase 1/2a trials in pancreatic, ovarian, and bladder cancers, with current clinical development focused on bladder cancer, where H19 is often suppressed in normal cells but upregulated in tumor cells (Gofrit et al., 2014). This Phase 2, multi-site trial sponsored by Anchiano Therapeutics will examine 140 patients with NMIBC (non-muscle-invasive bladder cancer) who are unresponsive to Bacillus Calmette-Guerin (BCG) therapy, a standard treatment. BC-819 will be directly instilled into the bladder, and several measures of clinical efficacy will be assessed throughout the study. While not directly targeting a ncRNA, this is an interesting example of how properties of ncRNAs, like H19 cancer-specific expression, can be harnessed for novel cancer therapeutic interventions.

Perspectives and future opportunities

Looking forward, ncRNAs are likely to continue to better enable researchers and clinicians to discover molecular signatures that help fine-tune diagnoses and differentiate malignant from benign tissue, as well as one type of cancer from another. These tests will also likely be prescribed to determine prognosis, such as determining risk of metastasis or probability of responding to treatments like chemotherapy. Although the therapeutic applications are still at an early stage, it is expected that drugs based on ncRNAs involved in cancer pathways will one day be prescribed for cancer patients. For identifying candidate biomarkers or therapeutic targets, focusing on lineage and disease-specific ncRNAs seems to be the ideal approach, as is the case with PCA3 mentioned above in prostate cancer (Hessels et al., 2003). Level of expression is also a key factor for developing ncRNA-based diagnostics and therapeutics. As noted previously, lncRNAs are often expressed at lower levels compared to protein-coding genes (Derrien et al., 2012; Iyer et al., 2015), so it is important to identify those candidates that can be readily detected. Furthermore, development of diagnostic biomarkers found in urine or blood is ideal to spare patients the non-invasive procedures often associated with tissue collection.

Compared to biologics or small-molecules, there are important benefits and challenges associated with RNA-based therapeutics that should be taken into consideration. RNA drugs are based on nucleotide hybridization, so targets can be easily approached; one just needs to identify the sequence and test candidate therapeutics for activity. With small-molecules or biologics, lengthier and laborious screens or structure-based design approaches must be employed. There is also the potential to design RNA-based approaches that can easily target multiple combinations of RNAs. Despite these advantages, the delivery method for RNA-based drugs has to be taken into account and optimized, and this can often be a challenging process. Oral formulations are also more difficult for RNA-based therapeutics compared to small-molecules and biologics. As noted above, there have further been issues with immune-related toxicity and other adverse events from RNA-based drugs. Improvement of oligonucleotide chemistry and delivery methods are continuously being pursued to mitigate these issues in future studies. Finally, the cost of gram quantity small-molecules can be much less than RNA-based drugs. We point the reader to excellent recent reviews (and references within) on the topics of the preceding two paragraphs for more discussion (Adams et al., 2017; Levin, 2019; Rupaimoole and Slack, 2017; Setten et al., 2019).

Members of our group recently showed the potential of targeting oncomiRs that were upregulated in an individual cancer patient’s tumor using patient-derived organoids (PDO) and xenografts (PDX) (Gilles et al., 2018), opening up the exciting possibility of personalized RNA medicine. Currently, each cancer patient is assigned to a dedicated care team consisting of a specialized oncology surgeon, medical oncologist, radiation oncologist, and pathologist. It is not too far-fetched to envision that tumor boards will add an integrated ncRNA workflow to the standard of care for every cancer patient. This could include the following components: a complete RNA-seq (including ncRNAs) performed on the cancer patient’s tumor and serum; a complete whole-genome sequence on the tumor and germline genome; an integrative bioinformatics and pathology analysis to quantify malignant pathologic phenotypes in the patient’s cancer and to identify significantly altered genomic and ncRNA alterations; a pathology report that includes an interpretation of the tumor pathology, DNA and RNA data, and potential therapeutic vulnerabilities, which would be presented at tumor boards to aid in therapeutic decisions; PDO and PDX models grown and therapeutic approaches tested on the PDO and PDX (e.g., traditional drug testing and ncRNA-based drug screening centered on the RNA profiles); recommendations for possible ncRNA-based therapy; a searchable file of the patient’s DNA/RNA and prognostic data placed in the electronic medical record of the patient for later reanalysis if necessary and if additional ncRNA information becomes known. The discovery of ncRNAs has opened up a new chapter in the history of medicine, one that promises to revolutionize the way that cancer and other diseases (discussed in (Adams et al., 2017; Levin, 2019; Rupaimoole and Slack, 2017)) are diagnosed and treated, and we have just begun to scratch the surface.

Supplementary Material

1

ACKNOWLEDGMENTS

We gratefully acknowledge all of the other researchers and work in this field that we were unable to cite due to space restrictions. We thank Stephanie Ellison and Bonnie Prescott for their outstanding efforts in the preparation of this manuscript and Robin Kunkel for generation of figures. F.J.S. acknowledges the NIH-YALE SPORE in Lung Cancer (P50CA196530) and the NCI Outstanding Investigator Award (R35CA232105). Additional support to F.J.S. was provided by P50CA196530-03S1, R01CA212649, and support from the Ludwig Center at Harvard, the Lungevity Foundation, and the V Foundation for Cancer Research. A.M.C. is a Howard Hughes Medical Institute Investigator, A. Alfred Taubman Scholar, and American Cancer Society Professor. A.M.C. is also supported by the NCI Outstanding Investigator Award (R35CA231996), Early Detection Research Network (UO1CA214170), NCI Prostate SPORE (P50CA186786), and Prostate Cancer Foundation.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

DECLARATION OF INTERESTS

F.J.S. is co-founder of two companies, MiraDx and 28/7 Therapeutics, and is or has been on the scientific advisory boards of additional companies, including Mirna Therapeutics, miRagen Therapeutics, The RNA Medicines Company, Mirxes, and Precision Nanosystems. A.M.C. is a co-founder of Oncopia, Esanik, Medsyn, and Lynx Dx and serves on the scientific advisory boards of Tempus, Ascentage, and GenePath.

REFERENCES

  1. Adams BD, Kasinski AL, and Slack FJ (2014). Aberrant regulation and function of microRNAs in cancer. Curr Biol 24, R762–776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Adams BD, Parsons C, and Slack FJ (2016a). The tumor-suppressive and potential therapeutic functions of miR-34a in epithelial carcinomas. Expert Opin Ther Targets 20, 737–753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Adams BD, Parsons C, Walker L, Zhang WC, and Slack FJ (2017). Targeting noncoding RNAs in disease. J Clin Invest 127, 761–771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Adams BD, Wali VB, Cheng CJ, Inukai S, Booth CJ, Agarwal S, Rimm DL, Gyorffy B, Santarpia L, Pusztai L, et al. (2016b). miR-34a Silences c-SRC to attenuate tumor growth in triple-negative breast cancer. Cancer Res 76, 927–939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Adams D, Gonzalez-Duarte A, O’Riordan WD, Yang CC, Ueda M, Kristen AV, Tournev I, Schmidt HH, Coelho T, Berk JL, et al. (2018). Patisiran, an RNAi therapeutic, for hereditary transthyretin amyloidosis. N Engl J Med 379, 11–21. [DOI] [PubMed] [Google Scholar]
  6. Adriaens C, Standaert L, Barra J, Latil M, Verfaillie A, Kalev P, Boeckx B, Wijnhoven PWG, Radaelli E, Vermi W, et al. (2016). p53 induces formation of NEAT1 lncRNA-containing paraspeckles that modulate replication stress response and chemosensitivity. Nat Med 22, 861–868. [DOI] [PubMed] [Google Scholar]
  7. Agaoglu F, Kovancilar M, Dizdar Y, Darendeliler E, Holdenrieder S, Dalay N, and Gezer U (2011). Investigation of miR-21, miR-141, and miR-221 in blood circulation of patients with prostate cancer. Tumor Biol 32, 583–588. [DOI] [PubMed] [Google Scholar]
  8. Anastasiadou E, Faggioni A, Trivedi P, and Slack FJ (2018a). The nefarious nexus of noncoding RNAs in cancer. Int J Mol Sci 19, 2072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Anastasiadou E, Jacob LS, and Slack FJ (2018b). Non-coding RNA networks in cancer. Nat Rev Cancer 18, 5–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Aravin A, Gaidatzis D, Pfeffer S, Lagos-Quintana M, Landgraf P, Iovino N, Morris P, Brownstein MJ, Kuramochi-Miyagawa S, Nakano T, et al. (2006). A novel class of small RNAs bind to MILI protein in mouse testes. Nature 442, 203–207. [DOI] [PubMed] [Google Scholar]
  11. Arun G, Diermeier S, Akerman M, Chang KC, Wilkinson JE, Hearn S, Kim Y, MacLeod AR, Krainer AR, Norton L, et al. (2016). Differentiation of mammary tumors and reduction in metastasis upon Malat1 lncRNA loss. Genes Dev 30, 34–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Arun G, Diermeier SD, and Spector DL (2018). Therapeutic targeting of long non-coding RNAs in cancer. Trends Mol Med 24, 257–277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Babar IA, Cheng CJ, Booth CJ, Liang X, Weidhaas JB, Saltzman WM, and Slack FJ (2012). Nanoparticle-based therapy in an in vivo microRNA-155 (miR-155)-dependent mouse model of lymphoma. Proc Natl Acad Sci USA 109, E1695–1704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Bachmayr-Heyda A, Reiner AT, Auer K, Sukhbaatar N, Aust S, Bachleitner-Hofmann T, Mesteri I, Grunt TW, Zeillinger R, and Pils D (2015). Correlation of circular RNA abundance with proliferation--exemplified with colorectal and ovarian cancer, idiopathic lung fibrosis, and normal human tissues. Sci Rep 5, 8057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Balas MM, and Johnson AM (2018). Exploring the mechanisms behind long noncoding RNAs and cancer. Noncoding RNA Res 3, 108–117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Balatti V, Pekarsky Y, and Croce CM (2017). Role of the tRNA-derived small RNAs in cancer: new potential biomarkers and target for therapy. Adv Cancer Res 135, 173–187. [DOI] [PubMed] [Google Scholar]
  17. Bartel DP (2018). Metazoan microRNAs. Cell 173, 20–51 [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Bartolomei MS, Zemel S, and Tilghman SM (1991). Parental imprinting of the mouse H19 gene. Nature 351, 153–155. [DOI] [PubMed] [Google Scholar]
  19. Beg MS, Brenner AJ, Sachdev J, Borad M, Kang YK, Stoudemire J, Smith S, Bader AG, Kim S, and Hong DS (2017). Phase I study of MRX34, a liposomal miR-34a mimic, administered twice weekly in patients with advanced solid tumors. Invest New Drugs 35, 180–188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Bhan A, Soleimani M, and Mandal SS (2017). Long noncoding RNA and cancer: a new paradigm. Cancer Res 77, 3965–3981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Bonci D, Coppola V, Musumeci M, Addario A, Giuffrida R, Memeo L, D’Urso L, Pagliuca A, Biffoni M, Labbaye C, et al. (2008). The miR-15a-miR-16–1 cluster controls prostate cancer by targeting multiple oncogenic activities. Nat Med 14, 1271–1277. [DOI] [PubMed] [Google Scholar]
  22. Brown CJ, Ballabio A, Rupert JL, Lafreniere RG, Grompe M, Tonlorenzi R, and Willard HF (1991). A gene from the region of the human X inactivation centre is expressed exclusively from the inactive X chromosome. Nature 349, 38–44. [DOI] [PubMed] [Google Scholar]
  23. Cabili MN, Trapnell C, Goff L, Koziol M, Tazon-Vega B, Regev A, and Rinn JL (2011). Integrative annotation of human large intergenic noncoding RNAs reveals global properties and specific subclasses. Genes Dev 25, 1915–1927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Calin GA, Dumitru CD, Shimizu M, Bichi R, Zupo S, Noch E, Aldler H, Rattan S, Keating M, Rai K, et al. (2002). Frequent deletions and down-regulation of micro-RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci USA 99, 15524–15529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Calin GA, Ferracin M, Cimmino A, Di Leva G, Shimizu M, Wojcik SE, Iorio MV, Visone R, Sever NI, Fabbri M, et al. (2005). A microRNA signature associated with prognosis and progression in chronic lymphocytic leukemia. N Engl J Med 353, 1793–1801. [DOI] [PubMed] [Google Scholar]
  26. Chakravarty D, Sboner A, Nair SS, Giannopoulou E, Li R, Hennig S, Mosquera JM, Pauwels J, Park K, Kossai M, et al. (2014). The oestrogen receptor alpha-regulated lncRNA NEAT1 is a critical modulator of prostate cancer. Nat Commun 5, 5383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Chen S, Huang V, Xu X, Livingstone J, Soares F, Jeon J, Zeng Y, Hua JT, Petricca J, Guo H, et al. (2019). Widespread and functional RNA circularization in localized prostate cancer. Cell 176, 831–843. [DOI] [PubMed] [Google Scholar]
  28. Cheng CJ, Bahal R, Babar IA, Pincus Z, Barrera F, Liu C, Svoronos A, Braddock DT, Glazer PM, Engelman DM, et al. (2015). MicroRNA silencing for cancer therapy targeted to the tumour microenvironment. Nature 518, 107–110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Cheng CJ, and Slack FJ (2012). The duality of OncomiR addiction in the maintenance and treatment of cancer. Cancer J 18, 232–237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Cieslik M, and Chinnaiyan AM (2018). Cancer transcriptome profiling at the juncture of clinical translation. Nat Rev Genet 19, 93–109. [DOI] [PubMed] [Google Scholar]
  31. Clemson CM, Hutchinson JN, Sara SA, Ensminger AW, Fox AH, Chess A, and Lawrence JB (2009). An architectural role for a nuclear noncoding RNA: NEAT1 RNA is essential for the structure of paraspeckles. Mol Cell 33, 717–726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Cole C, Sobala A, Lu C, Thatcher SR, Bowman A, Brown JWS, Green PJ, Barton GJ, and Hutvagner G (2009). Filtering of deep sequencing data reveals the existence of abundant Dicer-dependent small RNAs derived from tRNAs. RNA 15, 2147–2160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Concepcion CP, Han YC, Mu P, Bonetti C, Yao E, D’Andrea A, Vidigal JA, Maughan WP, Ogrodowski P, and Ventura A (2012). Intact p53-dependent responses in miR-34-deficient mice. PLoS Genet 8, e1002797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Derrien T, Johnson R, Bussotti G, Tanzer A, Djebali S, Tilgner H, Guernec G, Martin D, Merkel A, Knowles DG, et al. (2012). The GENCODE v7 catalog of human long noncoding RNAs: analysis of their gene structure, evolution, and expression. Genome Res 22, 1775–1789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Deveson IW, Hardwick SA, Mercer TR, and Mattick JS (2017). The dimensions, dynamics, and relevance of the mammalian noncoding transcriptome. Trends Genet 33, 464–478. [DOI] [PubMed] [Google Scholar]
  36. Djebali S, Davis CA, Merkel A, Dobin A, Lassmann T, Mortazavi A, Tanzer A, Lagarde J, Lin W, Schlesinger F, et al. (2012). Landscape of transcription in human cells. Nature 489, 101–108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Du Z, Sun T, Hacisuleyman E, Fei T, Wang X, Brown M, Rinn JL, Lee MG, Chen Y, Kantoff PW, et al. (2016). Integrative analyses reveal a long noncoding RNA-mediated sponge regulatory network in prostate cancer. Nat Commun 7, 10982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Edmonds MD, Boyd KL, Moyo T, Mitra R, Duszynski R, Arrate MP, Chen X, Zhao Z, Blackwell TS, Andl T, et al. (2016). MicroRNA-31 initiates lung tumorigenesis and promotes mutant KRAS-driven lung cancer. J Clin Invest 126, 349–364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. El Fatimy R, Subramanian S, Uhlmann EJ, and Krichevsky AM (2017). Genome editing reveals glioblastoma addiction to microRNA-10b. Mol Ther 25, 368–378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Esposito R, Bosch N, Lanzos A, Polidori T, Pulido-Quetglas C, and Johnson R (2019). Hacking the cancer genome: profiling therapeutically actionable long non-coding RNAs using CRISPR-Cas9 screening. Cancer Cell 35, 545–557. [DOI] [PubMed] [Google Scholar]
  41. Esquela-Kerscher A, and Slack FJ (2006). OncomiRs - microRNAs with a role in cancer. Nat Rev Cancer 6, 259–269. [DOI] [PubMed] [Google Scholar]
  42. Evans JR, Feng FY, and Chinnaiyan AM (2016). The bright side of dark matter: lncRNAs in cancer. J Clin Invest 126, 2775–2782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Ferrajoli A, Shanafelt TD, Ivan C, Shimizu M, Rabe KG, Nouraee N, Ikuo M, Ghosh AK, Lerner S, Rassenti LZ, et al. (2013). Prognostic value of miR-155 in individuals with monoclonal B-cell lymphocytosis and patients with B chronic lymphocytic leukemia. Blood 122, 1891–1899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Friedman RC, Farh KK, Burge CB, and Bartel DP (2009). Most mammalian mRNAs are conserved targets of microRNAs. Genome Res 19, 92–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Fromm B, Billipp T, Peck LE, Johansen M, Tarver JE, King BL, Newcomb JM, Sempere LF, Flatmark K, Hovig E, et al. (2015). A uniform system for the annotation of vertebrate microRNA genes and the evolution of the human microRNAome. Annu Rev Genet 49, 213–242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Gallardo E, Navarro A, Vinolas N, Marrades RM, Diaz T, Gel B, Quera A, Bandres E, Garcia-Foncillas J, Ramirez J, et al. (2009). miR-34a as a prognostic marker of relapse in surgically resected non-small-cell lung cancer. Carcinogenesis 30, 1903–1909. [DOI] [PubMed] [Google Scholar]
  47. Garzon R, Heaphy CE, Havelange V, Fabbri M, Volinia S, Tsao T, Zanesi N, Kornblau SM, Marcucci G, Calin GA, et al. (2009). MicroRNA 29b functions in acute myeloid leukemia. Blood 114, 5331–5341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Gerstein MB, Kundaje A, Hariharan M, Landt SG, Yan KK, Cheng C, Mu XJ, Khurana E, Rozowsky J, Alexander R, et al. (2012). Architecture of the human regulatory network derived from ENCODE data. Nature 489, 91–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Giannopoulou L, Zavridou M, Kasimir-Bauer S, and Lianidou ES (2018). Liquid biopsy in ovarian cancer: the potential of circulating miRNAs and exosomes. Transl Res 205, 77–91. [DOI] [PubMed] [Google Scholar]
  50. Gilles ME, Hao L, Huang L, Rupaimoole R, Lopez-Casas PP, Pulver E, Jeong JC, Muthuswamy SK, Hidalgo M, Bhatia SN, et al. (2018). Personalized RNA-medicine for pancreatic cancer. Clin Cancer Res 7, 1734–1747. [DOI] [PubMed] [Google Scholar]
  51. Girard A, Sachidanandam R, Hannon GJ, and Carmell MA (2006). A germline-specific class of small RNAs binds mammalian Piwi proteins. Nature 442, 199–202. [DOI] [PubMed] [Google Scholar]
  52. Gofrit ON, Benjamin S, Halachmi S, Leibovitch I, Dotan Z, Lamm DL, Ehrlich N, Yutkin V, Ben-Am M, and Hochberg A (2014). DNA based therapy with diphtheria toxin-A BC-819: a phase 2b marker lesion trial in patients with intermediate risk nonmuscle invasive bladder cancer. J Urol 191, 1697–1702. [DOI] [PubMed] [Google Scholar]
  53. Goodarzi H, Liu X, Nguyen HC, Zhang S, Fish L, and Tavazoie SF (2015). Endogenous tRNA-derived fragments suppress breast cancer progression via YBX1 displacement. Cell 161, 790–802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Gregory RI, and Shiekhattar R (2005). MicroRNA biogenesis and cancer. Cancer Res 65, 3509–3512. [DOI] [PubMed] [Google Scholar]
  55. Gupta RA, Shah N, Wang KC, Kim J, Horlings HM, Wong DJ, Tsai MC, Hung T, Argani P, Rinn JL, et al. (2010). Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature 464, 1071–1076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Gutschner T, Hammerle M, Eissmann M, Hsu J, Kim Y, Hung G, Revenko A, Arun G, Stentrup M, Gross M, et al. (2013). The noncoding RNA MALAT1 is a critical regulator of the metastasis phenotype of lung cancer cells. Cancer Res 73, 1180–1189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Hatley ME, Patrick DM, Garcia MR, Richardson JA, Bassel-Duby R, van Rooij E, and Olson EN (2010). Modulation of K-Ras-dependent lung tumorigenesis by MicroRNA-21. Cancer Cell 18, 282–293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Hessels D, Klein Gunnewiek JM, van Oort I, Karthaus HF, van Leenders GJ, van Balken B, Kiemeney LA, Witjes JA, and Schalken JA (2003). DD3(PCA3)-based molecular urine analysis for the diagnosis of prostate cancer. Eur Urol 44, 8–15. [DOI] [PubMed] [Google Scholar]
  59. Hill DA, Ivanovich J, Priest JR, Gurnett CA, Dehner LP, Desruisseau D, Jarzembowski JA, Wikenheiser-Brokamp KA, Suarez BK, Whelan AJ, et al. (2009). DICER1 mutations in familial pleuropulmonary blastoma. Science 325, 965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Hong D, Kurzrock R, Kim Y, Woessner R, Younes A, Nemunaitis J, Fowler N, Zhou T, Schmidt J, Jo M, et al. (2015). AZD9150, a next-generation antisense oligonucleotide inhibitor of STAT3 with early evidence of clinical activity in lymphoma and lung cancer. Sci Transl Med 7, 314ra185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Hosono Y, Niknafs YS, Prensner JR, Iyer MK, Dhanasekaran SM, Mehra R, Pitchiaya S, Tien J, Escara-Wilke J, Poliakov A, et al. (2017). Oncogenic role of THOR, a conserved cancer/testis long non-coding RNA. Cell 171, 1559–1572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Hsiao KY, Lin YC, Gupta SK, Chang N, Yen L, Sun HS, and Tsai SJ (2017). Noncoding effects of circular RNA CCDC66 promote colon cancer growth and metastasis. Cancer Res 77, 2339–2350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Hsu SH, Wang B, Kota J, Yu J, Costinean S, Kutay H, Yu L, Bai S, La Perle K, Chivukula RR, et al. (2012). Essential metabolic, anti-inflammatory, and anti-tumorigenic functions of miR-122 in liver. J Clin Invest 122, 2871–2883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Hu Q, Ye Y, Chan LC, Li Y, Liang K, Lin A, Egranov SD, Zhang Y, Xia W, Gong J, et al. (2019). Oncogenic lncRNA downregulates cancer cell antigen presentation and intrinsic tumor suppression. Nat Immunol 20, 835–851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Hu X, Feng Y, Zhang D, Zhao SD, Hu Z, Greshock J, Zhang Y, Yang L, Zhong X, Wang LP, et al. (2014). A functional genomic approach identifies FAL1 as an oncogenic long noncoding RNA that associates with BMI1 and represses p21 expression in cancer. Cancer Cell 26, 344–357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Hua Q, Lv X, Gu X, Chen Y, Chu H, Du M, Gong W, Wang M, and Zhang Z (2016). Genetic variants in lncRNA H19 are associated with the risk of bladder cancer in a Chinese population. Mutagenesis 31, 531–538. [DOI] [PubMed] [Google Scholar]
  67. Huang D, Chen J, Yang L, Ouyang Q, Li J, Lao L, Zhao J, Liu J, Lu Y, Xing Y, et al. (2018). NKILA lncRNA promotes tumor immune evasion by sensitizing T cells to activation-induced cell death. Nat Immunol 19, 1112–1125. [DOI] [PubMed] [Google Scholar]
  68. Huang X, Yuan T, Liang M, Du M, Xia S, Dittmar R, Wang D, See W, Costello BA, Quevedo F, et al. (2015). Exosomal miR-1290 and miR-375 as prognostic markers in castration-resistant prostate cancer. Eur Urol 67, 33–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Huarte M, and Rinn JL (2010). Large non-coding RNAs: missing links in cancer? Hum Mol Genet 19, R152–161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Hudson WH, Pickard MR, de Vera IM, Kuiper EG, Mourtada-Maarabouni M, Conn GL, Kojetin DJ, Williams GT, and Ortlund EA (2014). Conserved sequence-specific lincRNA-steroid receptor interactions drive transcriptional repression and direct cell fate. Nat Commun 5, 5395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Hung CL, Wang LY, Yu YL, Chen HW, Srivastava S, Petrovics G, and Kung HJ (2014). A long noncoding RNA connects c-Myc to tumor metabolism. Proc Natl Acad Sci USA 111, 18697–18702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Hung T, Wang Y, Lin MF, Koegel AK, Kotake Y, Grant GD, Horlings HM, Shah N, Umbricht C, Wang P, et al. (2011). Extensive and coordinated transcription of noncoding RNAs within cell-cycle promoters. Nat Genet 43, 621–629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Huse JT, Brennan C, Hambardzumyan D, Wee B, Pena J, Rouhanifard SH, Sohn-Lee C , le Sage C, Agami R, Tuschl T, et al. (2009). The PTEN-regulating microRNA miR-26a is amplified in high-grade glioma and facilitates gliomagenesis in vivo. Genes Dev 23, 1327–1337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Imaoka H, Toiyama Y, Fujikawa H, Hiro J, Saigusa S, Tanaka K, Inoue Y, Mohri Y, Mori T, Kato T, et al. (2016). Circulating microRNA-1290 as a novel diagnostic and prognostic biomarker in human colorectal cancer. Ann Oncol 27, 1879–1886. [DOI] [PubMed] [Google Scholar]
  75. Ivanov A, Memczak S, Wyler E, Torti F, Porath HT, Orejuela MR, Piechotta M, Levanon EY, Landthaler M, Dieterich C, et al. (2015). Analysis of intron sequences reveals hallmarks of circular RNA biogenesis in animals. Cell Rep 10, 170–177. [DOI] [PubMed] [Google Scholar]
  76. Iyer MK, Niknafs YS, Malik R, Singhal U, Sahu A, Hosono Y, Barrette TR, Prensner JR, Evans JR, Zhao S, et al. (2015). The landscape of long noncoding RNAs in the human transcriptome. Nat Genet 47, 199–208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Jeck WR, Sorrentino JA, Wang K, Slevin MK, Burd CE, Liu J, Marzluff WF, and Sharpless NE (2013). Circular RNAs are abundant, conserved, and associated with ALU repeats. RNA 19, 141–157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Ji P, Diederichs S, Wang W, Boing S, Metzger R, Schneider PM, Tidow N, Brandt B, Buerger H, Bulk E, et al. (2003). MALAT-1, a novel noncoding RNA, and thymosin beta4 predict metastasis and survival in early-stage non-small cell lung cancer. Oncogene 22, 8031–8041. [DOI] [PubMed] [Google Scholar]
  79. Jiang Y, Jiang Y-Y, Xie J-J, Mayakonda A, Hazawa M, Chen L, Xiao J-F, Li C-Q, Huang M-L, Ding L-W, et al. (2018). Co-activation of super-enhancer-driven CCAT1 by TP63 and SOX2 promotes squamous cancer progression. Nat Commun 9, 3619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Johnson CD, Esquela-Kerscher A, Stefani G, Byrom M, Kelnar K, Ovcharenko D, Wilson M, Wang X, Shelton J, Shingara J, et al. (2007). The let-7 microRNA represses cell proliferation pathways in human cells. Cancer Res 67, 7713–7722. [DOI] [PubMed] [Google Scholar]
  81. Johnson SM, Grosshans H, Shingara J, Byrom M, Jarvis R, Cheng A, Labourier E, Reinert KL, Brown D, and Slack FJ (2005). RAS is regulated by the let-7 microRNA family. Cell 120, 635–647. [DOI] [PubMed] [Google Scholar]
  82. Kallen AN, Zhou XB, Xu J, Qiao C, Ma J, Yan L, Lu L, Liu C, Yi JS, Zhang H, et al. (2013). The imprinted H19 lncRNA antagonizes let-7 microRNAs. Mol Cell 52, 101–112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Kalyana-Sundaram S, Kumar-Sinha C, Shankar S, Robinson DR, Wu YM, Cao X, Asangani IA, Kothari V, Prensner JR, Lonigro RJ, et al. (2012). Expressed pseudogenes in the transcriptional landscape of human cancers. Cell 149, 1622–1634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Karreth FA, Reschke M, Ruocco A, Ng C, Chapuy B, Leopold V, Sjoberg M, Keane TM, Verma A, Ala U, et al. (2015). The BRAF pseudogene functions as a competitive endogenous RNA and induces lymphoma in vivo. Cell 161, 319–332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Kasinski AL, and Slack FJ (2011). Epigenetics and genetics. MicroRNAs en route to the clinic: progress in validating and targeting microRNAs for cancer therapy. Nat Rev Cancer 11, 849–864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Kasinski AL, and Slack FJ (2012). miRNA-34 prevents cancer initiation and progression in a therapeutically resistant K-ras and p53-induced mouse model of lung adenocarcinoma. Cancer Res 72, 5576–5587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Katsushima K, Natsume A, Ohka F, Shinjo K, Hatanaka A, Ichimura N, Sato S, Takahashi S, Kimura H, Totoki Y, et al. (2016). Targeting the Notch-regulated non-coding RNA TUG1 for glioma treatment. Nat Commun 7, 13616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Keller A, Backes C, Leidinger P, Kefer N, Boisguerin V, Barbacioru C, Vogel B, Matzas M, Huwer H, Katus HA, et al. (2011a). Next-generation sequencing identifies novel microRNAs in peripheral blood of lung cancer patients. Mol Biosyst 7, 3187–3199. [DOI] [PubMed] [Google Scholar]
  89. Keller A, Leidinger P, Gislefoss R, Haugen A, Langseth H, Staehler P, Lenhof HP, and Meese E (2011b). Stable serum miRNA profiles as potential tool for non-invasive lung cancer diagnosis. RnA Biol 8, 506–516. [DOI] [PubMed] [Google Scholar]
  90. Kim HK, Fuchs G, Wang S, Wei W, Zhang Y, Park H, Roy-Chaudhuri B, Li P, Xu J, Chu K, et al. (2017). A transfer-RNA-derived small RNA regulates ribosome biogenesis. Nature 552, 57–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Kim J, Piao H-L, Kim B-J, Yao F, Han Z, Wang Y, Xiao Z, Siverly AN, Lawhon SE, Ton BN, et al. (2018). Long noncoding RNA MALAT1 suppresses breast cancer metastasis. Nat Genet 50, 1705–1715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Kim J, Siverly AN, Chen D, Wang M, Yuan Y, Wang Y, Lee H, Zhang J, Muller WJ, Liang H, et al. (2016). Ablation of miR-10b suppresses oncogene-induced mammary tumorigenesis and metastasis and reactivates tumor-suppressive pathways. Cancer Res 76, 6424–6435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Kim K, Jutooru I, Chadalapaka G, Johnson G, Frank J, Burghardt R, Kim S, and Safe S (2013). HOTAIR is a negative prognostic factor and exhibits pro-oncogenic activity in pancreatic cancer. Oncogene 32, 1616–1625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Kim T, Cui R, Jeon YJ, Lee JH, Lee JH, Sim H, Park JK, Fadda P, Tili E, Nakanishi H, et al. (2014). Long-range interaction and correlation between MYC enhancer and oncogenic long noncoding RNA CARLo-5. Proc Natl Acad Sci USA 111, 4173–4178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Kino T, Hurt DE, Ichijo T, Nader N, and Chrousos GP (2010). Noncoding RNA gas5 is a growth arrest- and starvation-associated repressor of the glucocorticoid receptor. Sci Signal 3, ra8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Klein U, Lia M, Crespo M, Siegel R, Shen Q, Mo T, Ambesi-Impiombato A, Califano A, Migliazza A, Bhagat G, et al. (2010). The DLEU2/miR-15a/16–1 cluster controls B cell proliferation and its deletion leads to chronic lymphocytic leukemia. Cancer Cell 17, 28–40. [DOI] [PubMed] [Google Scholar]
  97. Kogo R, Shimamura T, Mimori K, Kawahara K, Imoto S, Sudo T, Tanaka F, Shibata K, Suzuki A, Komune S, et al. (2011). Long noncoding RNA HOTAIR regulates polycomb-dependent chromatin modification and is associated with poor prognosis in colorectal cancers. Cancer Res 71, 6320–6326. [DOI] [PubMed] [Google Scholar]
  98. Kong R, Zhang EB, Yin DD, You LH, Xu TP, Chen WM, Xia R, Wan L, Sun M, Wang ZX, et al. (2015). Long noncoding RNA PVT1 indicates a poor prognosis of gastric cancer and promotes cell proliferation through epigenetically regulating p15 and p16. Mol Cancer 14, 82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Kopp F, and Mendell JT (2018). Functional classification and experimental dissection of long noncoding RNAs. Cell 172, 393–407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Kota J, Chivukula RR, O’Donnell KA, Wentzel EA, Montgomery CL, Hwang HW, Chang TC, Vivekanandan P, Torbenson M, Clark KR, et al. (2009). Therapeutic microRNA delivery suppresses tumorigenesis in a murine liver cancer model. Cell 137, 1005–1017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Kozomara A, Birgaoanu M, and Griffiths-Jones S (2018). miRBase: from microRNA sequences to function. Nucleic Acids Res 47, D155–D162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Krichevsky AM, King KS, Donahue CP, Khrapko K, and Kosik KS (2003). A microRNA array reveals extensive regulation of microRNAs during brain development. RNA 9, 1274–1281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Kristensen LS, Hansen TB, Veno MT, and Kjems J (2018). Circular RNAs in cancer: opportunities and challenges in the field. Oncogene 37, 555–565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Kumar P, Kuscu C, and Dutta A (2016). Biogenesis and function of transfer RNA-related fragments (tRFs). Trends Biochem Sci 41, 679–689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Kuscu C, Kumar P, Kiran M, Su Z, Malik A, and Dutta A (2018). tRNA fragments (tRFs) guide Ago to regulate gene expression post-transcriptionally in a Dicer-independent manner. RNA 24, 1093–1105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Lee RC, Feinbaum RL, and Ambros V (1993). The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 75, 843–854. [DOI] [PubMed] [Google Scholar]
  107. Lee YS, Shibata Y, Malhotra A, and Dutta A (2009). A novel class of small RNAs: tRNA-derived RNA fragments (tRFs). Genes Dev 23, 2639–2649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Legnini I, Di Timoteo G, Rossi F, Morlando M, Briganti F, Sthandier O, Fatica A, Santini T, Andronache A, Wade M, et al. (2017). Circ-ZNF609 is a circular RNA that can be translated and functions in myogenesis. Mol Cell 66, 22–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Leidinger P, Keller A, and Meese E (2011). MicroRNAs - important molecules in lung cancer research. Front Genet 2, 104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Leucci E, Vendramin R, Spinazzi M, Laurette P, Fiers M, Wouters J, Radaelli E, Eyckerman S, Leonelli C, Vanderheyden K, et al. (2016). Melanoma addiction to the long non-coding RNA SAMMSON. Nature 531, 518–522. [DOI] [PubMed] [Google Scholar]
  111. Levin AA (2019). Treating disease at the RNA level with oligonucleotides. N Engl J Med 380, 57–70. [DOI] [PubMed] [Google Scholar]
  112. Li J, Wu H, Li W, Yin L, Guo S, Xu X, Ouyang Y, Zhao Z, Liu S, Tian Y, et al. (2016a). Downregulated miR-506 expression facilitates pancreatic cancer progression and chemoresistance via SPHK1/Akt/NF-kappaB signaling. Oncogene 35, 5501–5514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Li X, Wu Z, Mei Q, Li X, Guo M, Fu X, and Han W (2013). Long non-coding RNA HOTAIR, a driver of malignancy, predicts negative prognosis and exhibits oncogenic activity in oesophageal squamous cell carcinoma. Br J Cancer 109, 2266–2278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Li Y, Wang Z, Shi H, Li H, Li L, Fang R, Cai X, Liu B, Zhang X, and Ye L (2016b). HBXIP and LSD1 scaffolded by lncRNA HOTAIR mediate transcriptional activation by c-Myc. Cancer Res 76, 293–304. [DOI] [PubMed] [Google Scholar]
  115. Li Y, Zheng F, Xiao X, Xie F, Tao D, Huang C, Liu D, Wang M, Wang L, Zeng F, et al. (2017). CircHIPK3 sponges miR-558 to suppress heparanase expression in bladder cancer cells. EMBO Rep 18, 1646–1659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Lin A, Li C, Xing Z, Hu Q, Liang K, Han L, Wang C, Hawke DH, Wang S, Zhang Y, et al. (2016). The LINK-A lncRNA activates normoxic HIF1alpha signalling in triple-negative breast cancer. Nat Cell Biol 18, 213–224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Ling H, Spizzo R, Atlasi Y, Nicoloso M, Shimizu M, Redis RS, Nishida N, Gafa R, Song J, Guo Z, et al. (2013). CCAT2, a novel noncoding RNA mapping to 8q24, underlies metastatic progression and chromosomal instability in colon cancer. Genome Res 23, 1446–1461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Liu B, Sun L, Liu Q, Gong C, Yao Y, Lv X, Lin L, Yao H, Su F, Li D, et al. (2015). A cytoplasmic NF-kappaB interacting long noncoding RNA blocks IkappaB phosphorylation and suppresses breast cancer metastasis. Cancer Cell 27, 370–381. [DOI] [PubMed] [Google Scholar]
  119. Liu C, Kelnar K, Liu B, Chen X, Calhoun-Davis T, Li H, Patrawala L, Yan H, Jeter C, Honorio S, et al. (2011). The microRNA miR-34a inhibits prostate cancer stem cells and metastasis by directly repressing CD44. Nat Med 17, 211–215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Lu KH, Li W, Liu XH, Sun M, Zhang ML, Wu WQ, Xie WP, and Hou YY (2013). Long non-coding RNA MEG3 inhibits NSCLC cells proliferation and induces apoptosis by affecting p53 expression. BMC Cancer 13, 461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Lv C, Li F, Li X, Tian Y, Zhang Y, Sheng X, Song Y, Meng Q, Yuan S, Luan L, et al. (2017). MiR-31 promotes mammary stem cell expansion and breast tumorigenesis by suppressing Wnt signaling antagonists. Nat Commun 8, 1036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Ma L, Reinhardt F, Pan E, Soutschek J, Bhat B, Marcusson EG, Teruya-Feldstein J, Bell GW, and Weinberg RA (2010). Therapeutic silencing of miR-10b inhibits metastasis in a mouse mammary tumor model. Nat Biotechnol 28, 341–347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Martinez VD, Vucic EA, Thu KL, Hubaux R, Enfield KS, Pikor LA, Becker-Santos DD, Brown CJ, Lam S, and Lam WL (2015). Unique somatic and malignant expression patterns implicate PIWI-interacting RNAs in cancer-type specific biology. Sci Rep 5, 10423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Mattick JS, and Makunin IV (2006). Non-coding RNA. Hum Mol Genet 15, R17–29. [DOI] [PubMed] [Google Scholar]
  125. Maute RL, Schneider C, Sumazin P, Holmes A, Califano A, Basso K, and Dalla-Favera R (2013). tRNA-derived microRNA modulates proliferation and the DNA damage response and is down-regulated in B cell lymphoma. Proc Natl Acad Sci USA 110, 1404–1409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Mavrakis KJ, Van Der Meulen J, Wolfe AL, Liu X, Mets E, Taghon T, Khan AA, Setty M, Rondou P, Vandenberghe P, et al. (2011). A cooperative microRNA-tumor suppressor gene network in acute T-cell lymphoblastic leukemia (T-ALL). Nat Genet 43, 673–678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. McCleland ML, Mesh K, Lorenzana E, Chopra VS, Segal E, Watanabe C, Haley B, Mayba O, Yaylaoglu M, Gnad F, et al. (2016). CCAT1 is an enhancer-templated RnA that predicts BET sensitivity in colorectal cancer. J Clin Invest 126, 639–652. [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Medina PP, Nolde M, and Slack FJ (2010). OncomiR addiction in an in vivo model of microRNA-21-induced pre-B-cell lymphoma. Nature 467, 86–90. [DOI] [PubMed] [Google Scholar]
  129. Mehra R, Udager AM, Ahearn TU, Cao X, Feng FY, Loda M, Petimar JS, Kantoff P, Mucci LA, and Chinnaiyan AM (2016). Overexpression of the long non-coding RNA SChLAP1 independently predicts lethal prostate cancer. Eur Urol 70, 549–552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Mei Y, Wang Y, Kumari P, Shetty AC, Clark D, Gable T, MacKerell AD, Ma MZ, Weber DJ, Yang AJ, et al. (2015). A piRNA-like small RNA interacts with and modulates pERM proteins in human somatic cells. Nat Commun 6, 7316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Mello SS, Sinow C, Raj N, Mazur PK, Bieging-Rolett K, Broz DK, Imam JFC, Vogel H, Wood LD, Sage J, et al. (2017). Neat1 is a p53-inducible lincRNA essential for transformation suppression. Genes Dev 31, 1095–1108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Meredith EK, Balas MM, Sindy K, Haislop K, and Johnson AM (2016). An RNA matchmaker protein regulates the activity of the long noncoding RNA HOTAIR. RNA 22, 995–1010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Mondal T, Subhash S, Vaid R, Enroth S, Uday S, Reinius B, Mitra S, Mohammed A, James AR, Hoberg E, et al. (2015). MEG3 long noncoding RNA regulates the TGF-beta pathway genes through formation of RNA-DNA triplex structures. Nat Commun 6, 7743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Ng KW, Anderson C, Marshall EA, Minatel BC, Enfield KS, Saprunoff HL, Lam WL, and Martinez VD (2016). Piwi-interacting RNAs in cancer: emerging functions and clinical utility. Mol Cancer 15, 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Nicoloso MS, Spizzo R, Shimizu M, Rossi S, and Calin GA (2009). MicroRNAs--the micro steering wheel of tumour metastases. Nat Rev Cancer 9, 293–302. [DOI] [PubMed] [Google Scholar]
  136. Niknafs YS, Han S, Ma T, Speers C, Zhang C, Wilder-Romans K, Iyer MK, Pitchiaya S, Malik R, Hosono Y, et al. (2016). The lncRNA landscape of breast cancer reveals a role for DSCAM-AS1 in breast cancer progression. Nat Commun 7, 12791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. O’Connell RM, Chaudhuri AA, Rao DS, and Baltimore D (2009). Inositol phosphatase SHIP1 is a primary target of miR-155. Proc Natl Acad Sci USA 106, 7113–7118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Ozata DM, Gainetdinov I, Zoch A, O’Carroll D, and Zamore PD (2019). PIWI-interacting RNAs: small RNAs with big functions. Nat Rev Genet 2, 89–108. [DOI] [PubMed] [Google Scholar]
  139. Ozawa T, Matsuyama T, Toiyama Y, Takahashi N, Ishikawa T, Uetake H, Yamada Y, Kusunoki M, Calin G, and Goel A (2017). CCAT1 and CCAT2 long noncoding RNAs, located within the 8q.24.21 ‘gene desert’, serve as important prognostic biomarkers in colorectal cancer. Ann Oncol 28, 1882–1888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Pamudurti NR, Bartok O, Jens M, Ashwal-Fluss R, Stottmeister C, Ruhe L, Hanan M, Wyler E, Perez-Hernandez D, Ramberger E, et al. (2017). Translation of circRNAs. Mol Cell 66, 9–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Panzitt K, Tschernatsch MM, Guelly C, Moustafa T, Stradner M, Strohmaier HM, Buck CR, Denk H, Schroeder R, Trauner M, et al. (2007). Characterization of HULC, a novel gene with striking up-regulation in hepatocellular carcinoma, as noncoding RNA. Gastroenterology 132, 330–342. [DOI] [PubMed] [Google Scholar]
  142. Park JK, Kogure T, Nuovo GJ, Jiang J, He L, Kim JH, Phelps MA, Papenfuss TL, Croce CM, Patel T, et al. (2011). miR-221 silencing blocks hepatocellular carcinoma and promotes survival. Cancer Res 71, 7608–7616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Pasmant E, Sabbagh A, Vidaud M, and Bieche I (2011). ANRIL, a long, noncoding RNA, is an unexpected major hotspot in GWAS. FASEB J 25, 444–448. [DOI] [PubMed] [Google Scholar]
  144. Pecot CV, Calin GA, Coleman RL, Lopez-Berestein G, and Sood AK (2011). RNA interference in the clinic: challenges and future directions. Nat Rev Cancer 11, 59–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Pekarsky Y, and Croce CM (2010). Is miR-29 an oncogene or tumor suppressor in CLL? Oncotarget 1, 224–227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Pineau P, Volinia S, McJunkin K, Marchio A, Battiston C, Terris B, Mazzaferro V, Lowe SW, Croce CM, and Dejean A (2010). miR-221 overexpression contributes to liver tumorigenesis. Proc Natl Acad Sci USA 107, 264–269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Pliatsika V, Loher P, Magee R, Telonis AG, Londin E, Shigematsu M, Kirino Y, and Rigoutsos I (2018). MINTbase v2.0: a comprehensive database for tRNA-derived fragments that includes nuclear and mitochondrial fragments from all The Cancer Genome Atlas projects. Nucleic Acids Res 46, D152–D159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Poliseno L, Salmena L, Zhang J, Carver B, Haveman WJ, and Pandolfi PP (2010). A coding-independent function of gene and pseudogene mRNAs regulates tumour biology. Nature 465, 1033–1038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Preis M, Gardner TB, Gordon SR, Pipas JM, Mackenzie TA, Klein EE, Longnecker DS, Gutmann EJ, Sempere LF, and Korc M (2011). MicroRNA-10b expression correlates with response to neoadjuvant therapy and survival in pancreatic ductal adenocarcinoma. Clin Cancer Res 17, 5812–5821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Prensner JR, Chen W, Iyer MK, Cao Q, Ma T, Han S, Sahu A, Malik R, Wilder-Romans K, Navone N, et al. (2014a). PCAT-1, a long noncoding RNA, regulates BRCA2 and controls homologous recombination in cancer. Cancer Res 74, 1651–1660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Prensner JR, Iyer MK, Balbin OA, Dhanasekaran SM, Cao Q, Brenner JC, Laxman B , Asangani IA, Grasso CS, Kominsky HD, et al. (2011). Transcriptome sequencing across a prostate cancer cohort identifies PCAT-1, an unannotated lincRNA implicated in disease progression. Nat Biotechnol 29, 742–749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Prensner JR, Iyer MK, Sahu A, Asangani IA, Cao Q, Patel L, Vergara IA, Davicioni E , Erho N, Ghadessi M, et al. (2013). The long noncoding RNA SChLAP1 promotes aggressive prostate cancer and antagonizes the SWI/SNF complex. Nat Genet 45, 1392–1398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Prensner JR, Zhao S, Erho N, Schipper M, Iyer MK, Dhanasekaran SM, Magi-Galluzzi C , Mehra R, Sahu A, Siddiqui J, et al. (2014b). RNA biomarkers associated with metastatic progression in prostate cancer: a multi-institutional high-throughput analysis of SChLAP1. Lancet Oncol 15, 1469–1480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Qu L, Ding J, Chen C, Wu ZJ, Liu B, Gao Y, Chen W, Liu F, Sun W, Li XF, et al. (2016a). Exosome-transmitted lncARSR promotes sunitinib resistance in renal cancer by acting as a competing endogenous RNA. Cancer Cell 29, 653–668. [DOI] [PubMed] [Google Scholar]
  155. Qu L, Wu Z, Li Y, Xu Z, Liu B, Liu F, Bao Y, Wu D, Liu J, Wang A, et al. (2016b). A feed-forward loop between lncARSR and YAP activity promotes expansion of renal tumour-initiating cells. Nat Commun 7, 12692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Quagliata L, Matter MS, Piscuoglio S, Arabi L, Ruiz C, Procino A, Kovac M, Moretti F, Makowska Z, Boldanova T, et al. (2014). Long noncoding RNA HOTTIP/HOXA13 expression is associated with disease progression and predicts outcome in hepatocellular carcinoma patients. Hepatology 59, 911–923. [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Ransohoff JD, Wei Y, and Khavari PA (2018). The functions and unique features of long intergenic non-coding RNA. Nat Rev Mol Cell Biol 19, 143–157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Reebye V, Huang KW, Lin V, Jarvis S, Cutilas P, Dorman S, Ciriello S, Andrikakou P, Voutila J, Saetrom P, et al. (2018). Gene activation of CEBPA using saRNA: preclinical studies of the first in human saRNA drug candidate for liver cancer. Oncogene 37, 3216–3228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Reid G, Pel ME, Kirschner MB, Cheng YY, Mugridge N, Weiss J, Williams M, Wright C, Edelman JJ, Vallely MP, et al. (2013). Restoring expression of miR-16: a novel approach to therapy for malignant pleural mesothelioma. Ann Oncol 24, 3128–3135. [DOI] [PubMed] [Google Scholar]
  160. Reinhart BJ, Slack FJ, Basson M, Pasquinelli AE, Bettinger JC, Rougvie AE, Horvitz HR, and Ruvkun G (2000). The 21-nucleotide let-7 RNA regulates developmental timing in Caenorhabditis elegans. Nature 403, 901–906. [DOI] [PubMed] [Google Scholar]
  161. Ren S, Wang F, Shen J, Sun Y, Xu W, Lu J, Wei M, Xu C, Wu C, Zhang Z, et al. (2013). Long non-coding RNA metastasis associated in lung adenocarcinoma transcript 1 derived miniRNA as a novel plasma-based biomarker for diagnosing prostate cancer. Eur J Cancer 49, 2949–2959. [DOI] [PubMed] [Google Scholar]
  162. Rheinbay E, Parasuraman P, Grimsby J, Tiao G, Engreitz JM, Kim J, Lawrence MS, Taylor-Weiner A, Rodriguez-Cuevas S, Rosenberg M, et al. (2017). Recurrent and functional regulatory mutations in breast cancer. Nature 547, 55–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Rinn JL, and Huarte M (2011). To repress or not to repress: this is the guardian’s question. Trends Cell Biol 21, 344–353. [DOI] [PubMed] [Google Scholar]
  164. Rinn JL, Kertesz M, Wang JK, Squazzo SL, Xu X, Brugmann SA, Goodnough LH, Helms JA, Farnham PJ, Segal E, et al. (2007). Functional demarcation of active and silent chromatin domains in human HOX loci by noncoding RNAs. Cell 129, 1311–1323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Rossi S, Shimizu M, Barbarotto E, Nicoloso MS, Dimitri F, Sampath D, Fabbri M, Lerner S, Barron LL, Rassenti LZ, et al. (2010). microRNA fingerprinting of CLL patients with chromosome 17p deletion identify a miR-21 score that stratifies early survival. Blood 116, 945–952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  166. Ru P, Hu P, Geng F, Mo X, Cheng C, Yoo JY, Cheng X, Wu X, Guo JY, Nakano I, et al. (2016). Feedback loop regulation of SCAP/SREBP-1 by miR-29 modulates EGFR signaling-driven glioblastoma growth. Cell Rep 16, 1527–1535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  167. Rupaimoole R, and Slack FJ (2017). MicroRNA therapeutics: towards a new era for the management of cancer and other diseases. Nat Rev Drug Discov 16, 203–222. [DOI] [PubMed] [Google Scholar]
  168. Ryan BM, Robles AI, and Harris CC (2010). Genetic variation in microRNA networks: the implications for cancer research. Nat Rev Cancer 10, 389–402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Saikia M, and Hatzoglou M (2015). The many virtues of tRNA-derived stress-induced RNAs (tiRNAs): discovering novel mechanisms of stress response and effect on human health. J Biol Chem 290, 29761–29768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Saito M, Schetter AJ, Mollerup S, Kohno T, Skaug V, Bowman ED, Mathe EA, Takenoshita S, Yokota J, Haugen A, et al. (2011). The association of microRNA expression with prognosis and progression in early-stage, non-small cell lung adenocarcinoma: a retrospective analysis of three cohorts. Clin Cancer Res 17, 1875–1882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  171. Sakimura S, Sugimachi K, Kurashige J, Ueda M, Hirata H, Nambara S, Komatsu H, Saito T, Takano Y, Uchi R, et al. (2015). The miR-506-induced epithelial-mesenchymal transition is involved in poor prognosis for patients with gastric cancer. Ann Surg Oncol 22 Suppl 3, S1436–1443. [DOI] [PubMed] [Google Scholar]
  172. Sang LJ, Ju HQ, Liu GP, Tian T, Ma GL, Lu YX, Liu ZX, Pan RL, Li RH, Piao HL, et al. (2018). LncRNA CamK-A regulates Ca(2+)-signaling-mediated tumor microenvironment remodeling. Mol Cell 72, 71–83. [DOI] [PubMed] [Google Scholar]
  173. Santanam U, Zanesi N, Efanov A, Costinean S, Palamarchuk A, Hagan JP, Volinia S, Alder H, Rassenti L, Kipps T, et al. (2010). Chronic lymphocytic leukemia modeled in mouse by targeted miR-29 expression. Proc Natl Acad Sci USA 107, 12210–12215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  174. Schmitt AM, Garcia JT, Hung T, Flynn RA, Shen Y, Qu K, Payumo AY, Peres-da-Silva A, Broz DK, Baum R, et al. (2016). An inducible long noncoding RNA amplifies DNA damage signaling. Nat Genet 48, 1370–1376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  175. Schwanhausser B, Busse D, Li N, Dittmar G, Schuchhardt J, Wolf J, Chen W, and Selbach M (2013). Global quantification of mammalian gene expression control. Nature 495, 126–127. [DOI] [PubMed] [Google Scholar]
  176. Seto AG, Beatty X, Lynch JM, Hermreck M, Tetzlaff M, Duvic M, and Jackson AL (2018). Cobomarsen, an oligonucleotide inhibitor of miR-155, co-ordinately regulates multiple survival pathways to reduce cellular proliferation and survival in cutaneous T-cell lymphoma. Br J Haematol 183, 428–444. [DOI] [PubMed] [Google Scholar]
  177. Setten RL, Rossi JJ, and Han S. p. (2019). The current state and future directions of RNAi-based therapeutics. Nat Rev Drug Discov 18, 421–446. [DOI] [PubMed] [Google Scholar]
  178. Shukla S, Zhang X, Niknafs YS, Xiao L, Mehra R, Cieslik M, Ross A, Schaeffer E, Malik B, Guo S, et al. (2016). Identification and validation of PCAT14 as prognostic biomarker in prostate cancer. Neoplasia 18, 489–499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  179. Slack FJ (2006). Regulatory RNAs and the demise of ‘junk’ DNA. Genome Biol 7, 328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  180. Svoboda M, Slyskova J, Schneiderova M, Makovicky P, Bielik L, Levy M, Lipska L, Hemmelova B, Kala Z, Protivankova M, et al. (2014). HOTAIR long non-coding RnA is a negative prognostic factor not only in primary tumors, but also in the blood of colorectal cancer patients. Carcinogenesis 35, 1510–1515. [DOI] [PubMed] [Google Scholar]
  181. Takayama K, Horie-Inoue K, Katayama S, Suzuki T, Tsutsumi S, Ikeda K, Urano T, Fujimura T, Takagi K, Takahashi S, et al. (2013). Androgen-responsive long noncoding RNA CTBP1-AS promotes prostate cancer. EMBO J 32, 1665–1680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  182. Tay Y, Rinn J, and Pandolfi PP (2014). The multilayered complexity of ceRNA crosstalk and competition. Nature 505, 344–352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  183. Teschendorff AE, Lee SH, Jones A, Fiegl H, Kalwa M, Wagner W, Chindera K, Evans I, Dubeau L, Orjalo A, et al. (2015). HOTAIR and its surrogate DNA methylation signature indicate carboplatin resistance in ovarian cancer. Genome Med 7, 108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  184. Toiyama Y, Takahashi M, Hur K, Nagasaka T, Tanaka K, Inoue Y, Kusunoki M, Boland CR, and Goel A (2013). Serum miR-21 as a diagnostic and prognostic biomarker in colorectal cancer. J Natl Cancer Inst 105, 849–859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. Tomlins SA, Aubin SM, Siddiqui J, Lonigro RJ, Sefton-Miller L, Miick S, Williamsen S, Hodge P, Meinke J, Blase A, et al. (2011). Urine TMPRSS2:ERG fusion transcript stratifies prostate cancer risk in men with elevated serum PSA. Sci Transl Med 3, 94ra72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  186. Tomlins SA, Day JR, Lonigro RJ, Hovelson DH, Siddiqui J, Kunju LP, Dunn RL, Meyer S, Hodge P, Groskopf J, et al. (2016). Urine TmPrSs2:ERG plus PcA3 for individualized prostate cancer risk assessment. Eur Urol 70, 45–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  187. Trang P, Medina PP, Wiggins JF, Ruffino L, Kelnar K, Omotola M, Homer R, Brown D , Bader AG, Weidhaas JB, et al. (2010). Regression of murine lung tumors by the let-7 microRNA. Oncogene 29, 1580–1587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  188. Trang P, Wiggins JF, Daige CL, Cho C, Omotola M, Brown D, Weidhaas JB, Bader AG, and Slack FJ (2011). Systemic delivery of tumor suppressor microRNA mimics using a neutral lipid emulsion inhibits lung tumors in mice. Mol Ther 19, 1116–1122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  189. Tripathi V, Ellis JD, Shen Z, Song DY, Pan Q, Watt AT, Freier SM, Bennett CF, Sharma A, Bubulya PA, et al. (2010). The nuclear-retained noncoding RNA MALAT1 regulates alternative splicing by modulating SR splicing factor phosphorylation. Mol Cell 39, 925–938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  190. Tsai MC, Manor O, Wan Y, Mosammaparast N, Wang JK, Lan F, Shi Y, Segal E, and Chang HY (2010). Long noncoding RNA as modular scaffold of histone modification complexes. Science 329, 689–693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  191. Tsai WC, Hsu SD, Hsu CS, Lai TC, Chen SJ, Shen R, Huang Y, Chen HC, Lee CH, Tsai TF, et al. (2012). MicroRNA-122 plays a critical role in liver homeostasis and hepatocarcinogenesis. J Clin Invest 122, 2884–2897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  192. Tseng YY, Moriarity BS, Gong W, Akiyama R, Tiwari A, Kawakami H, Ronning P, Reuland B, Guenther K, Beadnell TC, et al. (2014). PVT1 dependence in cancer with MYC copy-number increase. Nature 512, 82–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  193. van Zandwijk N, Pavlakis N, Kao SC, Linton A, Boyer MJ, Clarke S, Huynh Y, Chrzanowska A, Fulham MJ, Bailey DL, et al. (2017). Safety and activity of microRNA-loaded minicells in patients with recurrent malignant pleural mesothelioma: a first-in-man, phase 1, open-label, dose-escalation study. Lancet Oncol 18, 1386–1396. [DOI] [PubMed] [Google Scholar]
  194. Vendramin R, Verheyden Y, Ishikawa H, Goedert L, Nicolas E, Saraf K, Armaos A, Delli Ponti R, Izumikawa K, Mestdagh P, et al. (2018). SAMMSON fosters cancer cell fitness by concertedly enhancing mitochondrial and cytosolic translation. Nat Struct Mol Biol 25, 1035–1046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  195. Vo JN, Cieslik M, Zhang Y, Shukla S, Xiao L, Zhang Y, Wu YM, Dhanasekaran SM, Engelke CG, Cao X, et al. (2019). The landscape of circular RNA in cancer. Cell 176, 869–881. [DOI] [PMC free article] [PubMed] [Google Scholar]
  196. Volinia S, Calin GA, Liu CG, Ambs S, Cimmino A, Petrocca F, Visone R, Iorio M, Roldo C, Ferracin M, et al. (2006). A microRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci USA 103, 2257–2261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  197. Voutila J, Reebye V, Roberts TC, Protopapa P, Andrikakou P, Blakey DC, Habib R, Huber H, Saetrom P, Rossi JJ, et al. (2017). Development and mechanism of small activating RNA targeting CEBPA, a novel therapeutic in clinical trials for liver cancer. Mol Ther 25, 2705–2714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  198. Wang Y, Gu J, Roth JA, Hildebrandt MA, Lippman SM, Ye Y, Minna JD, and Wu X (2013). Pathway-based serum microRNA profiling and survival in patients with advanced stage non-small cell lung cancer. Cancer Res 73, 4801–4809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  199. Wang Z, Yang B, Zhang M, Guo W, Wu Z, Wang Y, Jia L, Li S, Xie W, and Yang D (2018). lncRNA epigenetic landscape analysis identifies EPIC1 as an oncogenic lncRNA that interacts with mYc and promotes cell-cycle progression in cancer. Cancer Cell 33, 706–720.e709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  200. Weng W, Wei Q, Toden S, Yoshida K, Nagasaka T, Fujiwara T, Cai S, Qin H, Ma Y, and Goel A (2017). Circular RNA ciRS-7-a promising prognostic biomarker and a potential therapeutic target in colorectal cancer. Clin Cancer Res 23, 3918–3928. [DOI] [PMC free article] [PubMed] [Google Scholar]
  201. White NM, Zhao SG, Zhang J, Rozycki EB, Dang HX, McFadden SD, Eteleeb AM, Alshalalfa M, Vergara IA, Erho N, et al. (2017). Multi-institutional analysis shows that low PCAT-14 expression associates with poor outcomes in prostate cancer. Eur Urol 71, 257–266. [DOI] [PubMed] [Google Scholar]
  202. Wu SY, Lopez-Berestein G, Calin GA, and Sood AK (2014). RNAi therapies: drugging the undruggable. Sci Transl Med 6, 240ps247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  203. Wurster CD, Winter B, Wollinsky K, Ludolph AC, Uzelac Z, Witzel S, Schocke M, Schneider R, and Kocak T (2019). Intrathecal administration of nusinersen in adolescent and adult SMA type 2 and 3 patients. J Neurol 1, 183–194. [DOI] [PubMed] [Google Scholar]
  204. Xie H, Ma H, and Zhou D (2013). Plasma HULC as a promising novel biomarker for the detection of hepatocellular carcinoma. Biomed Res Int 2013, 136106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  205. Yamamoto Y, Loriot Y, Beraldi E, Zhang F, Wyatt AW, Al Nakouzi N, Mo F, Zhou T, Kim Y, Monia BP, et al. (2015). Generation 2.5 antisense oligonucleotides targeting the androgen receptor and its splice variants suppress enzalutamide-resistant prostate cancer cell growth. Clin Cancer Res 21, 1675–1687. [DOI] [PubMed] [Google Scholar]
  206. Yan LX, Huang XF, Shao Q, Huang MY, Deng L, Wu QL, Zeng YX, and Shao JY (2008). MicroRNA miR-21 overexpression in human breast cancer is associated with advanced clinical stage, lymph node metastasis and patient poor prognosis. RNA 14, 2348–2360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  207. Yang D, Sun Y, Hu L, Zheng H, Ji P, Pecot CV, Zhao Y, Reynolds S, Cheng H, Rupaimoole R, et al. (2013a). Integrated analyses identify a master microRNA regulatory network for the mesenchymal subtype in serous ovarian cancer. Cancer Cell 23, 186–199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  208. Yang F, Fang E, Mei H, Chen Y, Li H, Li D, Song H, Wang J, Hong M, Xiao W, et al. (2019). Cis-acting circ-CTNNB1 promotes beta-catenin signaling and cancer progression via DDX3-mediated transactivation of YY1. Cancer Res 79, 557–571. [DOI] [PubMed] [Google Scholar]
  209. Yang F, Huo XS, Yuan SX, Zhang L, Zhou WP, Wang F, and Sun SH (2013b). Repression of the long noncoding RNA-LET by histone deacetylase 3 contributes to hypoxia-mediated metastasis. Mol Cell 49, 1083–1096. [DOI] [PubMed] [Google Scholar]
  210. Yap KL, Li S, Munoz-Cabello AM, Raguz S, Zeng L, Mujtaba S, Gil J, Walsh MJ, and Zhou MM (2010). Molecular interplay of the noncoding RNA ANRIL and methylated histone H3 lysine 27 by polycomb CBX7 in transcriptional silencing of INK4a. Mol Cell 38, 662–674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  211. Yoon JH, Abdelmohsen K, Srikantan S, Yang X, Martindale JL, De S, Huarte M, Zhan M, Becker KG, and Gorospe M (2012). LincRNA-p21 suppresses target mRNA translation. Mol Cell 47, 648–655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  212. Yu G, Yao W, Gumireddy K, Li A, Wang J, Xiao W, Chen K, Xiao H, Li H, Tang K, et al. (2014). Pseudogene PTENP1 functions as a competing endogenous RNA to suppress clear-cell renal cell carcinoma progression. Mol Cancer Ther 13, 3086–3097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  213. Zeitels LR, Acharya A, Shi G, Chivukula D, Chivukula RR, Anandam JL, Abdelnaby AA, Balch GC, Mansour JC, Yopp AC, et al. (2014). Tumor suppression by miR-26 overrides potential oncogenic activity in intestinal tumorigenesis. Genes Dev 28, 2585–2590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  214. Zhang X, Gejman R, Mahta A, Zhong Y, Rice KA, Zhou Y, Cheunsuchon P, Louis DN, and Klibanski A (2010). Maternally expressed gene 3, an imprinted noncoding RNA gene, is associated with meningioma pathogenesis and progression. Cancer Res 70, 2350–2358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  215. Zhang Y, Pitchiaya S, Cieslik M, Niknafs YS, Tien JCY, Hosono Y, Iyer MK, Yazdani S, Subramaniam S, Shukla SK, et al. (2018). Analysis of the androgen receptor-regulated lncRNA landscape identifies a role for ARLNC1 in prostate cancer progression. Nat Genet 50, 814–824. [DOI] [PMC free article] [PubMed] [Google Scholar]
  216. Zhang Z, Zhu Z, Watabe K, Zhang X, Bai C, Xu M, Wu F, and Mo YY (2013). Negative regulation of lncRNA GAS5 by miR-21. Cell Death Differ 20, 1558–1568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  217. Zhao J, Du P, Cui P, Qin Y, Hu C, Wu J, Zhou Z, Zhang W, Qin L, and Huang G (2018). LncRNA PVT1 promotes angiogenesis via activating the STAT3/VEGFA axis in gastric cancer. Oncogene 37, 4094–4109. [DOI] [PubMed] [Google Scholar]
  218. Zhao X, Wang P, Liu J, Zheng J, Liu Y, Chen J, and Xue Y (2015). Gas5 exerts tumor-suppressive functions in human glioma cells by targeting miR-222. Mol Ther 23, 1899–1911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  219. Zheng R, Du M, Wang X, Xu W, Liang J, Wang W, Lv Q, Qin C, Chu H, Wang M, et al. (2018). Exosome-transmitted long non-coding RNA PTENP1 suppresses bladder cancer progression. Mol Cancer 17, 143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  220. Zhou Y, Zhong Y, Wang Y, Zhang X, Batista DL, Gejman R, Ansell PJ, Zhao J, Weng C, and Klibanski A (2007). Activation of p53 by MEG3 non-coding RNA. J Biol Chem 282, 24731–24742. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1

RESOURCES