C H A P T E R
13 Circulating miRNAs as Tumor Biomarkers Vikas Ghai, Inyoul Lee and Kai Wang Institute for Systems Biology, Seattle, WA, United States
Early detection, diagnosis, and stratification are vital for the proper management and treatment of cancer patients, and as such the development of sensitive, specific, and informative cancer biomarkers is critical. However, the current diagnostic approach is still based on invasive procedures. For example, tissue biopsy is usually needed to confirm and classify cancer type and grade. The advent of so called “liquid biopsies”—to detect and analyze molecules or cells in circulation that are usually tumor-derived, or produced in response to tumor formation—has shown a lot of promise as a noninvasive method to detect and classify various types of cancer. In this chapter we will look at the use of circulating cell-free microRNAs as tumor biomarkers, and reflect on the challenges and opportunities they face gaining traction in the clinic.
MICRORNAs: BIOSYNTHESIS AND FUNCTION There are different classes of ribonucleic acid (RNA) molecules in cells, but recently
Oncogenomics DOI: https://doi.org/10.1016/B978-0-12-811785-9.00013-2
much attention has been focused on understanding the function of noncoding RNA (ncRNAs). In contrast to the well-studied messenger RNAs (mRNAs), ncRNAs do not encode a protein product but rather function as either structural or critical regulators of cellular processes. Of these, microRNAs (miRNAs) are the best characterized. MiRNAs are small (B22 bp) ncRNAs that have been shown to modulate the proteome by targeting the turnover of specific mRNAs. Currently more than 25,000 individual miRNAs have been identified across more than 200 species, with some of the sequences being highly conserved across species, suggesting the role they play in biology is of critical importance. In the nucleus, miRNAs are initially transcribed as part of a larger primary miRNA (pri-miRNA) product, and through a series of modifications (done by a multienzyme complex including the ribonuclease, Drosha, and the RNA binding protein, DGCR8) the 50 and 30 features are trimmed to produce a shorter (B70 bp) precursor miRNA (pre-miRNA). In the cytoplasm, the pre-miRNA is then further
191
© 2019 Elsevier Inc. All rights reserved.
192
13. CIRCULATING MIRNAS AS TUMOR BIOMARKERS
processed by the ribonuclease, Dicer, into a 1924 bp mature miRNA duplex. One of the strands (either 23p or 25p) of the duplex then joins with other proteins (including the Argonaut protein, Ago2) to form a ribonucleoprotein structure called the RNA Induced Silencing Complex (RISC). The RISC complex interacts with target mRNA through imperfect base-paring usually at their 30 untranslated regions (UTR). This RISC- miRNA-mRNA interaction promotes degradation of the transcript, or inhibits protein translation by blocking the progression or binding of ribosome.
miRNAs DYSREGULATED IN CANCER Initially, miRNAs were discovered as regulators of heterochrony in Caenorhabditis elegans (Lee, Feinbaum, & Ambros, 1993), but now they have been shown to regulate a wide array of cellular processes including signal transduction, cell-cycle, cell proliferation, cell growth, and metabolism, all of which play central roles in cancer (Huang et al., 2011). Some miRNAs have been shown to be dysregulated in various cancers (coined “OncomiRs”), often due to expansion or deletion of miRNA gene clusters that regulate key processes involved in tumorigenesis, or through aberrant changes to their regulation. For example, the loss of the miR-15a16 miRNA cluster on chromosome 13 has been frequently observed in B-cell chronic lymphocytic leukemia (Cimmino et al., 2005), and the duplication of the miR-17-92 gene cluster has been observed in B-cell lymphomas and lung cancer (Hayashita et al., 2005; Mavrakis et al., 2010). Interestingly, the miRNAs in the miR-1792 miRNA cluster are under the transcriptional control of the oncogene, C-Myc (O’Donnell, Wentzel, Zeller, Dang, & Mendell, 2005). C-Myc not only activates the oncogenic miR-17-92 cluster, but also acts to repress the expression of several tumor-suppressive miRNAs, such as
members of the miR-15a, miR-26, miR-29, miR30, and let-7 families (Chang et al., 2008). The p53 transcriptional regulatory network that controls tumorigenesis, and is often mutated in various cancers, also regulates miRNA expression (He et al., 2007). The p53 tumor suppressor guards entry into the cell-cycle through a complex pathway that includes directly regulating the expression of miR-34a, miR-107, miR-605, and miR-1246 (Chang et al., 2007; Hermeking, 2010; Raver-Shapira et al., 2007; Xiao, Lin, Luo, Luo, & Wang, 2011; Yamakuchi et al., 2010; Zhang, Liao, Zeng, & Lu, 2011). Additionally, miR-34a acts to further promote p53 expression by negatively regulating a p53 suppressor, SIRT1 (Yamakuchi & Lowenstein, 2009). Signaling through the transforming growth factor-β (TGFβ) pathway regulates the process of the epithelialmesenchymal transition (EMT), which plays a critical role during metastasis. The miR-200 family acts to repress the expression of ZEB1 and ZEB2 (the downstream transcription targets of TGF-β in the EMT), and is often dysregulated in metastasis in various cancers (Feng, Wang, Fillmore, & Xi, 2014). Epigenetic modifications to miRNAs also lead to their dysregulation in cancer. Hypermethylation of the antitumorigenic miR-148a, and the miR-34b/c cluster results in silencing in cancer cells and increased metastasis in vivo (Lujambio et al., 2008). Similarly, hypermethylation of miR-9-1, miR-124a, and miR-1455p are attributed to decreased expression in breast, lung, and colon carcinomas, respectively (Donzelli et al., 2015; Lehmann et al., 2008; Lujambio, 2007).
EXTRACELLULAR VESICLES AND EXTRACELLULAR miRNAs Interestingly, miRNAs are also present in the extracellular environment and have been detected in various biological fluids such as serum, plasma, urine, tears, saliva, and others (Chen et al., 2008; Lawrie et al., 2008; Mitchell et al.,
II. ONCOGENOMICS: CIRCULATING BIOMARKERS IN CLINICAL ONCOLOGY
EXTRACELLULAR VESICLES AND EXTRACELLULAR MIRNAS
2008; Valadi et al., 2007; Weber et al., 2010). The specific role of these extracellular miRNAs remains somewhat speculative; however, there is evidence that a subset of these miRNAs may function as intermediaries of paracrine signaling between cells, tissues, or organs, including between a tumor and its microenvironment. The initial observation of these extracellular miRNAs seemed dubious, as purified RNAs are often degraded in the extracellular environment due to excessive ribonuclease activity. To avoid degradation, miRNAs have been observed in complex with RNA binding proteins including AGO2 (Arroyo et al., 2011; Turchinovich, Weiz, Langheinz, & Burwinkel, 2011), NPM1 (Wang, Zhang, Weber, Baxter, & Galas, 2010), hnRNPA2B1 (VillarroyaBeltri et al., 2013), SYNCRIP (Santangelo et al., 2016), and lipoproteins such as HDL (Vickers, Palmisano, Shoucri, Shamburek, & Remaley, 2011; Wagner et al., 2013). However, the release of many of these miRNA-protein complexes (such as those bound to AGO2 and NPM1) are passive and likely the results of cell-death and autophagy, rather than representing actively exported miRNAs that are the result of signaling. MiRNAs have also been found to be encapsulated in lipid vesicles broadly referred to as extracellular vesicles (EVs). EVs include vesicles of different types that differ in size, derivation, and function, including apoptotic bodies (5004000 nm), microvesicles (502000 nm), and exosomes (30100 nm). Among these, exosomes (including related subtypes like the tumor-derived oncosomes) have drawn significant interest in recent years. Exosomes originate from early-endosomederived multivesicle bodies (MVB) inside the cell, and are released through fusion with the plasma membrane (Denzer, Kleijmeer, Heijnen, Stoorvogel, & Geuze, 2000). In addition to miRNAs, exosomes also contain other nucleic acids such as long noncoding RNA (lncRNA), Piwi-interacting RNAs (piRNAs), small
193
nucleolar RNAs (snoRNAs), protein coding mRNA, and genomic DNA fragments. Exosomes also contain various proteins, including tetraspanins (such as Alix, CD9, CD63, and CD81) embedded on the vesicle surface that have been used extensively as exosomal surface markers. Oncosomes, which are tumor-derived, have been known to carry their own unique molecular cargo that may be tumor-specific. For example, small oncosomes secreted by glioma cells carry a mutated form of EGFR, EGFRvIII (Al-Nedawi et al., 2008), and large oncosomes derived from prostate cancer tumors carry Cav-1, a well-studied serum marker associated with metastatic prostate cancer (Di Vizio et al., 2012; Morello et al., 2013). While not entirely definitive, several different mechanistic pathways may explain how miRNAs are loaded into exosomes. One potential mechanism is a ceramide-dependent pathway for sorting miRNA into exosomes. Interference of the ceramide biosynthesis, through the targeting of the enzyme neutral sphingomyelinase 2 (nSMase2), affects the distribution of miRNAs present in exosomes (Kosaka et al., 2010). In colorectal cancer (CRC) cell lines, mutations in KRAS affect the miRNA content in exosomes (Cha et al., 2015) and attenuates the tumor microenvironment, which in turn affects tumor cell growth and migration (Demory Beckler et al., 2013). As discussed above, RNA-binding proteins play a role in protecting miRNAs from degradation, but they also play a role in miRNA export into exosomes. Sumoylated hnRNPPA2B1 has been shown to recognize a G/A rich motif present in the 30 UTR of miRNAs in primary T lymphoblasts, and can facilitate their export into exosomes (Villarroya-Beltri et al., 2013). Another member of the hnRNP family, SYNCRIP, also interacts with a “GGCU” motif present in the 30 UTR of miRNAs in hepatocytes cell lines sorting them into exosomes (Santangelo et al., 2016). Similarly, miRNAs with nontemplate
II. ONCOGENOMICS: CIRCULATING BIOMARKERS IN CLINICAL ONCOLOGY
194
13. CIRCULATING MIRNAS AS TUMOR BIOMARKERS
additions of differing terminal nucleoside bases (uridines more frequently observed in exosomal miRNAs and adenines in cellular miRNAs) can influence export in exosomes (Koppers-Lalic et al., 2014), though the exact mechanism controlling this editing and sorting is unclear.
CIRCULATING miRNAs AS POTENTIAL BIOMARKERS FOR CANCERS Many of the current tumor biomarkers are proteins or sugars/glycans that circulate in body fluids like whole blood or serum. For example, the use of Alpha-fetoprotein (AFP) to screen for liver cancers, CA15-3 for breast cancer, CA19-9 for pancreatic cancer, CA-125 for ovarian cancer, Carcinoembryonic antigen (CEA) for CRC, and Prostate Specific Antigen (PSA) for prostate cancer. While results from these biomarkers are clinically useful when used in the appropriate manner, many cannot detect cancers at an early stage, lack specificity, and have high false negative/positive rates (Diamandis, 2012; Hanash, 2011). There are also many technical challenges, such as low protein abundancy in samples and difficulties in developing high affinity capture reagents (Fu¨ze´ry, Levin, Chan, & Chan, 2013). MiRNAs offer many advantages over their protein counterparts in many of these regards, as they are stable, abundant in biofluids, and can be easily measured using amplification or hybridization based approaches. Historically, extracellular miRNAs were first observed and characterized in cancer cell lines and the serum of cancer patients (Chen et al., 2008; Mitchell et al., 2008; Valadi et al., 2007), thus significant effort has gone towards identifying (1) circulating miRNAs involved in or associated with various forms of cancer, and (2) their potential influence on microenvironment. Here we provide some updates on the current status and
challenges of identifying circulating miRNAs as biomarkers for several different types of cancers.
Breast Cancer While there has been considerable research focused on genetic risk factors for developing breast cancer (e.g., mutations in BRCA1 or 2) or how breast tumors respond to therapy (presence or absence of ER, PR, and HER2 receptors), work to identify circulating miRNA that may have diagnostic potential is still emerging (Table 13.1). There have been two primary studies looking at the changes of miRNAs in whole blood among breast cancer patients. One group found that the concentration of miR-195, let-7, and miR-155 were elevated in the blood of breast cancer patients compared to controls or other cancer patients using qRT-PCR, and found these three miRNAs to have a discriminative power of 94% in this capacity (Heneghan, Miller, Kelly, Newell, & Kerin, 2010). Another study used microarrays to detect changes in the blood of miRNAs in both breast cancer patients and healthy controls, as well as in a mammary tumorigenesis murine model. In the patient samples, they found 77 miRNAs that exhibited concentration changes, and validated the increase of miR-138 concentration in both human blood and mouse tissue (Waters et al., 2014). Profiling of miRNAs in plasma has also been done in breast cancer patients. Cuk et al. used qRT-PCR and identified several miRNAs (miR-148b, miR-376c, miR-409-3p, and miR801) that were increased in plasma of early stage (I and II) breast cancer patients compared to healthy controls, suggesting these miRNAs should be used in early breast cancer detection (Cuk et al., 2013). The increase of plasma miR148b (along with miR-133a) levels was also confirmed using qRT-PCR by an independent study, which also found that miR-148 is actively secreted by breast cancer cells in
II. ONCOGENOMICS: CIRCULATING BIOMARKERS IN CLINICAL ONCOLOGY
TABLE 13.1 Subtype
Studies Examining Circulating miRNAs in Breast Cancer Affected Circulating miRNA (up)
Affected Circulating miRNA (down)
Assay
Sample
References
miR-195, let-7, -155
qRT-PCR
W.blood
Heneghan et al. (2010)
miR-138
qRT-PCR
W. blood
Waters et al. (2014)
miR-148b, miR-376c, miR-409-3p, miR-801
qRT-PCR
Plasma
Cuk et al. (2013)
miR-148b, miR-133a
qRT-PCR
Plasma
Shen et al. (2014)
miR-10b, miR-373
qRT-PCR
Plasma/ tumor
Chen et al. (2013)
qRT-PCR
Plasma
Madhavan et al. (2016)
miR-155
qRT-PCR
Serum
Zhu et al. (2009)
miR-21, miR-126, miR-155, miR-199a, miR-335
qRT-PCR
Serum/ tumor
Wang et al. (2010)
Nonmetastatic/ metastatic
miR-10b, miR-34a, miR-155
qRT-PCR
Serum
Roth et al. (2010)
Nonmetastatic/ metastatic
miR-10b, miR-17, miR-34a, miR-93, miR-155, miR-373
qRT-PCR
Serum
Eichelser et al. (2013)
miR-10b, miR-21, miR-125b, miR-145, miR-155, miR-191, miR-382
qRT-PCR
Serum
Mar-Aguilar et al. (2013)
Early-stage
Metastatic Metastatic
Early-stage
miR-141, miR-144, miR-193b, miR-200a, miR-200b, miR-200c, miR-203, miR-210, miR-215, miR-365, miR-375, miR-429, miR-486-5p, miR-801, miR-1260, miR-1274a
miR-505-5p, miR-96-5p
miR-505-5p (with treatment)
Microarray Serum
Matamala et al. (2015)
miR-1246, miR-1307-3p, miR-6861-5p
miR-4634, miR-6875-5p
Microarray Serum
Hamam et al. (2017)
Microarray Serum
Shimomura et al. (2016)
sRNAseq
Zhu et al. (2014)
miR-4270, miR-1225-5p, miR -188-5p, miR-1202, miR-4281, miR-1207-5p, miR-642b-3p, miR-1290, and miR -3141 miR-184, miR-1246, miR-224-5p
miR-598-3p, miR-323b-3p, miR485-5p, miR-382-3p, miR-34c-5p, miR-125b-1-3p, miR-132-5p
Serum/ tumor
196
13. CIRCULATING MIRNAS AS TUMOR BIOMARKERS
culture (Shen et al., 2014). Looking at the potential correlation between miRNA concentration changes in plasma and in metastatic breast cancer tissue, one study found miR-10b and miR-373 to be overexpressed in breastlymph node metastases, and elevated in plasma from preoperative breast cancer patients with lymph node metastasis compared with patients without metastasis and healthy controls (Chen, Cai, Zhang, Barekati, & Zhong, 2013). Recently, additional circulating plasma miRNAs associated with overall survival of patients with metastatic breast cancer have been identified, including several members of the miR-200 family, which are involved in promoting metastasis and tumor progression (Feng et al., 2014; Madhavan et al., 2016). The majority of the circulating miRNA studies for breast cancer have been done using serum. One of the first groups to look at circulating miRNAs in breast cancer patients used qRT-PCR to measure several miRNAs in the serum of breast cancer patients and found that miR-155 was elevated in cancer patients compared to controls (Zhu, Qin, Atasoy, & Sauter, 2009). Wang et al. followed up with these results and validated miR-155 (Wang, Zheng, Guo, & Ding, 2010). They also found the concentration changes for several additional miRNAs (miR-21, miR-126, miR-199a and miR335) in serum and tumor tissues, and these changes are closely associated with clinical features such as tumor grades and receptor expression status (Wang et al., 2010). Similarly, another study looking at serum miRNAs that might be associated with breast cancer identified miR-10b, miR-34a, and miR-155 to be elevated in breast cancer patients and also associated with tumor stage and metastasis (Roth et al., 2010). An expanded set of these miRNAs (miR-10b, miR-17, miR-34a, miR-93, miR-155, and miR-373) were later used by the same group to show that they can be used to either discriminate breast cancer patients from healthy controls, or further stratify patients
based on different stages of metastasis and HER2 tumor expression status (Eichelser, Flesch-Janys, Chang-Claude, Pantel, & Schwarzenbach, 2013). A similar seven miRNA signature (miR-10b, miR-21, miR-125b, miR145, miR-155, miR-191, and miR-382) was increased in the serum of breast cancer patients, and a panel comprised of miR-145, miR-155, and miR-382 was used to discriminate breast cancer patients from healthy controls with high sensitivity and specificity (Mar-Aguilar et al., 2013). Several recent studies used high-throughput methods to profile circulating miRNAs and identified several previously uncharacterized breast cancer associated miRNAs in serum. As measured by microarrays, the concentrations of miR-505-5p and miR-96-5p were also identified as being significantly increased in the serum of early stage breast cancer patients versus controls, and miR-505-5p levels decreased in early stage breast cancer patients that had undergone treatment (Matamala et al., 2015). Another microarray experiment identified several uncharacterized serum miRNAs that were elevated in breast cancer patients compared to controls, and correlated well with stage and molecular subtype (Hamam et al., 2017). A similar study used microarrays to identify additional serum miRNAs that also performed well in discriminating breast cancer patients from healthy controls, with very high sensitivity (97.3%), specificity of (82.9%), and accuracy (89.7%), even at early stages (Shimomura et al., 2016). Taking a next-generation sequencing (NGS) approach, Zhu et al. used small RNAseq (sRNAseq) to analyze the spectrum of miRNA in breast cancer tumors/healthy tissues and serum samples between breast cancer patients and healthy controls. Initially, they identified many dysregulated miRNAs in tumors (174 between breast cancer tumors and healthy tissue) and in serum (109 between breast cancer patients and healthy controls), but found only 10 miRNAs had common changes between
II. ONCOGENOMICS: CIRCULATING BIOMARKERS IN CLINICAL ONCOLOGY
CIRCULATING MIRNAS AS POTENTIAL BIOMARKERS FOR CANCERS
tumors and serum (Zhu et al., 2014). Several of these had been previously identified as showing concentration changes in serum (miR-125b and miR-382) (Mar-Aguilar et al., 2013) while others were previously uncharacterized (miR323b, miR-598, and miR-184).
Colorectal Cancer Noninvasive blood biomarkers, such as CEA, have been used for CRC diagnosis, however CEA has very little specificity since its levels also changed in patients with other cancers. While miRNAs have primarily been investigated in CRC tumor biopsies (Boisen et al., 2015; Chang, Mestdagh, Vandesompele, Kerin, & Miller, 2010; Eriksen et al., 2016; Peltier & Latham, 2008; Yang et al., 2016) and stool samples (Ahmed et al., 2009; Ahmed et al., 2013; Wu et al., 2012; Zhu et al., 2016), studies have also been examining the changes of circulating miRNAs in CRC patients. In one of the first studies looking at circulating miRNAs in CRC, Ng et al. found elevated levels of miR-135b, miR-95, miR-222, and miR-17-3p in serum, and miR-92 in the plasma of CRC patients compared to healthy controls (Ng et al., 2009). Further work confirmed the elevated levels of miR-92 (along with miR-29a) in a different cohort of CRC patients (Huang et al., 2010; Zheng et al., 2013). Interestingly, elevated miR-29a and miR-141 serum levels can be used to discriminate metastatic CRC patients from controls (Wang & Gu, 2012). A recent study found that whole serum, serum exosome, and corresponding colon and rectal tumor levels of miR-19a-3p, miR-21-5p, and miR-425-5p were elevated in CRC patients compared to controls, and a panel of all three had good discriminatory ability in a separate validation set of CRC patients and controls (AUC of 0.87) (Zhu et al., 2017). In a similar study, Ogata-Kawata et al. used microarrays to measure exosomal miRNAs isolated from both CRC patient serum and CRC cell lines, and identified several miRNAs (let-7a,
197
miR-1229, miR-1246, miR-150, miR-21, miR-223, and miR-23a) that were elevated (Ogata-Kawata et al., 2014). Similar studies have been undertaken for plasma miRNA profiling in CRC patients. As in serum, the level of circulating miR-141 can also be used as a prognostic marker, as elevated plasma miR-141 is associated with poor outcomes in metastatic CRC patients (Cheng et al., 2011). From pooled plasma samples, one group used qRT-PCR profiling to identify nine miRNAs (miR-18a, miR-20a, miR-21, miR-29a, miR-92a, miR-106b, miR-133a, miR-143, and miR-145) that were elevated in CRC plasma samples compared to neoplasm-free controls, and had moderate discriminatory function (AUC of 0.745) in a separate validation set of CRC patients and controls (Luo, Stock, Burwinkel, & Brenner, 2013). Gira´ldez et al. used sRNAseq to profile miRNAs from the plasma of CRC patients and healthy controls, and identified 13 miRNAs exhibiting concentration changes, and were able to validate six (miR-18a, miR-19a, miR-19b, miR-15b, miR-29a, and miR-335) with qRT-PCR. These miRNAs had moderate discriminatory power (AUC of 0.80) to identify CRC patients in an independent validation cohort (Gira´ldez et al., 2013). Some miRNAs have been shown to have prognostic application in CRC. For example, Hansen et al. showed that miR-126 levels in plasma can be used to evaluate responses to treatment in metastatic CRC patients undergoing chemotherapy (Hansen, Carlsen, Heegaard, Sørensen, & Jakobsen, 2015), and Toiyama et al. demonstrated that miR-200c levels in serum are associated with evaluating lymph node metastasis and long term survival in CRC patients (Toiyama et al., 2014) (Table 13.2).
Lung Cancer Nonsmall cell lung cancer (NSCLC) accounts for the majority of cases of lung cancers globally,
II. ONCOGENOMICS: CIRCULATING BIOMARKERS IN CLINICAL ONCOLOGY
198
13. CIRCULATING MIRNAS AS TUMOR BIOMARKERS
TABLE 13.2 Studies Examining Circulating miRNAs in Colorectal Cancer Subtype
Metastatic
Metastatic
Affected Circulating miRNA (up)
Affected Circulating miRNA (down) Assay
Sample
References
miR-135b, miR-95, miR-222, miR-17-3p, miR-92
qRT-PCR
Serum
Ng et al. (2009)
miR-92, miR-29a
qRT-PCR
Serum
Huang et al. (2010)
miR-92
qRT-PCR
Serum
Zheng et al. (2013)
miR-29a, miR-141
qRT-PCR
Serum
Wang and Gu (2012)
miR-19a-3p, miR-21-5p, miR-425-5p
qRT-PCR
Serum/ tumor
Zhu et al. (2017)
let-7a, miR-1229, miR-1246, miR-150, miR-21, miR-223, miR-23a
microarray
Serum exosomes
Ogata-Kawata et al. (2014)
miR-141
qRT-PCR
Plasma
Cheng et al. (2011)
miR-18a, miR-20a, miR-21, miR-29a, miR-92a, miR-106b, miR-133a, miR-143, miR-145
qRT-PCR
Plasma
Luo et al. (2013)
miR-18a, miR-19a, miR-19b, miR-15b, miR-29a, miR-335
sRNAseq/ qRT-PCR
Plasma
Gira´ldez et al. (2013)
qRT-PCR
Plasma
Hansen et al. (2015)
Metastatic
miR-126 (nonresponders to therapy)
miR-126 (responders to therapy)
Metastatic
miR-200c (metastatsis)
miR-200c (long termsurvival)
so major effort has been taken to identify biomarkers for NSCLC. MiRNA signatures present in tumor samples are a valuable tool in predicting survival and relapse in NSCLC (Yu et al., 2008). Accordingly, similar studies looking at their counterparts present in circulation as a noninvasive alternative have been undertaken. In serum, Chen et al. made one of the first observational reports of cell-free miRNAs in circulation, and found the increase of miR-24 and miR-223 levels in samples from NSCLC patients versus healthy controls. Another group also found the increase of miR-24 (along with miR-21, miR-205, and miR-30d) levels in the sera of NSCLC patients compared to controls (Le et al., 2012). Not only did this panel of four
miRNAs (miR-24, miR-21, miR-205 and miR30d) have good performance at discriminating between NSCLC and controls, but it also performed better than the traditional serum biomarker, CAE in the same samples. Interestingly, serum levels of miR-21 and miR-24 decreased in postoperative NSCLC patients in the same study. Hu et al. used sRNAseq and RT-qPCR platforms, and identified several serum miRNAs (miR-1, miR-30d, miR-499, and miR-486) that were associated with either short or long term survival in NSCLC patients (Hu et al., 2010). In a separate study, serum miR-486 was decreased in NSCLC patients compared to controls based on results from microarrays and qRT-PCR (Petriella et al., 2015). Another group used qRT-PCR profiling of
II. ONCOGENOMICS: CIRCULATING BIOMARKERS IN CLINICAL ONCOLOGY
199
CIRCULATING MIRNAS AS POTENTIAL BIOMARKERS FOR CANCERS
serum miRNAs to develop a 34-miRNA diagnostic panel (Table 13.3) for early stage NSCLC that could accurately discriminate NSCLC patients at different stages (Bianchi et al., 2011). Similar studies have been done to profile plasma miRNAs in NSCLC patients. Shen et al. identified a signature of six miRNAs (miR-21, miR-182, miR-210, miR-126, and miR-486-5p) that exhibited similar concentration changes
between plasma and corresponding tumor tissues in NSCLC patients, and was able to discriminate NSCLC patients from healthy controls (Shen et al., 2011). As discussed previously, both miR-21 and miR-486 show the same serum concentration changes in NSCLC patients in other studies (Table 13.3). Interestingly, elevated serum, plasma, and whole blood levels of miR-21 have been shown
TABLE 13.3 Studies Examining Circulating miRNAs in Nonsmall Cell Lung Cancer Subtype
Affected Circulating miRNA (up)
Affected Circulating miRNA (down)
Assay
Sample
References
qRT-PCR
Serum
Chen et al. (2008)
qRT-PCR
Serum
Le et al. (2012)
NGS, qRT-PCR
Serum
Hu et al. (2010)
miR-486-5p
Microarray, qRT-PCR
Serum
Petriella et al. (2015)
miR-32, miR-133b, miR-566, miR-432*, miR-223, miR-29a, miR-148a, miR-142-5p, miR-140-5p
miR-92a, miR-484, miR-328, miR-191, miR-376a, miR-342-3p, miR-331-2p, miR-30c, miR-285p, miR-98, miR-17, miR-26b, miR-374a, miR-30b, miR-26a, miR-134-3p, miR-103, miR-126, let-7a, let-7d, let-7b, miR-22. miR-148, miR-139
qRT-PCR
Serum
Bianchi et al. (2011)
miR-21, miR-182, miR-210
miR-126, miR-486-5p
qRT-PCR
Plasma/ tumor
Shen et al. (2011)
Microarray, qRT-PCR
Plasma/ tumor
Boeri et al. (2011)
miR-218, miR-566, miR-661, miR-485-3p, miR-203, miR-517b, miR-122, miR-182, miR-193a, miR-411, miR-450b-5p
qRT-PCR
plasma
Wozniak et al. (2015)
miR-139-5p, miR-378a, miR-379,
qRT-PCR
Plasma Cazzoli et al. exosomes (2013)
qRT-PCR
W. blood Li et al. (2011)
miR-24, miR-223 miR-21, miR-205, miR-30d, and miR-24
miR-21, miR-24 (postoperative)
miR-1, miR-499 (long survivial); miR-30d, miR-486 (short survivial) Metastatic Early stage
miR-221, miR-660, miR-486-5p, miR-28-3p, miR-197, miR-106a, miR-451, miR-140-5p, miR-16 Metastatic let-7c, let-7b, miR-192, miR-200c, miR-155, miR-519a, miR-641, miR-520f, miR-206, miR-304, miR-1243, miR-720, miR-543, miR-1276 miR-200b-5p miR-21
II. ONCOGENOMICS: CIRCULATING BIOMARKERS IN CLINICAL ONCOLOGY
200
13. CIRCULATING MIRNAS AS TUMOR BIOMARKERS
to be associated with poor prognosis of NSCLC patients in several studies (Gao et al., 2010; Li, Li, Ouyang, Hu, & Tang, 2011; Liu et al., 2012; Wang et al., 2011) and is elevated in the sputum of NSCLC patients (Xie et al., 2010). Like with serum, several groups have used plasma miRNA profiling to develop larger panels of diagnostic miRNAs that can be used to discriminate NSCLC patients from heathy controls, and correlate concentration changes in plasma to expression changes in tumor samples (Boeri et al., 2011; Wozniak et al., 2015). Cazzoli et al. specifically looked at miRNAs from plasma exosomes in lung cancer patients, and identified a six miRNA signature (miR151a-5p, miR-30a-3p, miR-200b-5p, miR-629, miR-100, and miR-154-3p) that discriminates lung cancer patients from healthy controls with an AUC of 0.90, and patients with lung adenocarcinoma from those with granuloma with an AUC of 0.760 (Cazzoli et al., 2013).
Pancreatic Cancer Pancreatic cancer is one of the deadliest cancers due to the lack of early stage clinical symptoms and diagnostic tools. The 5-year survival rate for the most common type of pancreatic cancer, pancreatic ductal adenocarcinoma (PDAC) is only about 5%. MiRNA studies for PDAC have not only been done in circulation (plasma, serum, whole blood), but also in pancreatic biofluids, such as bile and pancreatic juice. In plasma, the levels of four miRNAs (miR-21, miR-155, miR-210, and miR-196a) were shown to be increased in the plasma of PDAC patients versus healthy controls when measured by qRT-PCR (Wang et al., 2009). Another group validated the increase of plasma miR-21 levels in PDAC patients, and found that when combined with miR-483-3p, it performs better than CA19-9 in discriminating PDAC patients
from heathy controls (Abue et al., 2015). Liu et al. also validated the elevated levels of miR196a in plasma, and found when combined with miR-16 (also elevated in PDAC patient plasma) and CA 19-9, results provided the most effective diagnosis of PDAC. Moreover, an elevated miR-196a concentration in plasma correlates with a lower survival rate in advanced stage PDAC patients (Kong et al., 2011). Other studies have highlighted additional plasma miRNAs that could have prognostic application. For example, the miR-18a level in plasma is elevated in preoperative PDAC patients, but decreased after surgery (whereas CA 19-9 level did not change) (Morimura et al., 2011). The concentration of miR-182 is increased in PDAC patient plasma samples and the level inversely correlates with overall survival and disease-free survival (Chen, Yang, Xiao, Zhu, & Li, 2014). In serum and whole blood, several similar diagnostic cell-free miRNAs for PDAC have been developed, for example, the miR-21 level is also increased in serum and whole blood in PDAC patients. Liu et al. identified a seven miRNA panel (miR-20a, miR-21, miR-24, miR25, miR-99a, miR-185, and miR-191) in serum from PDAC patients that can be used to discriminate PDAC from chronic pancreatitis (CP) patients (Liu et al., 2012), where CA 19-9 lacks this capability. Like in plasma, combining the level of miR-21 in serum with other miRNAs such as miR-34a can enhance its diagnostic performance (Alemar et al., 2016). Looking at whole blood, Schultz et al. identified a miRNA biomarker panel (see Table 13.4) that outperformed CA 19-9 in discriminating PDAC patients from healthy controls, but performed better when used in combination with CA 19-9 measurement (Schultz, Dehlendorff, & Jensen, 2014). The pancreas secretes proximal biofluid, including pancreatic juice and bile, which offer an opportunity to examine the local tumor microenvironment more closely (though often
II. ONCOGENOMICS: CIRCULATING BIOMARKERS IN CLINICAL ONCOLOGY
201
CHALLENGES AND OPPORTUNITIES FOR MIRNA BASED BIOMARKERS
TABLE 13.4 Studies Examining Circulating miRNAs in Pancreatic Ductal Adenocarcinoma Subtype Affected Circulating miRNA (up)
Affected Circulating miRNA (down)
Assay
Sample
References
miR-21, miR-210, miR-155, miR-196a
qRT-PCR
Plasma
Wang et al. (2009)
miR-21, miR-483-3p
qRT-PCR
Plasma
Abue et al. (2015)
miR-16, miR-196a
qRT-PCR
Plasma
Liu et al. (2012), Kong et al. (2011)
miR-18a
qRT-PCR
Plasma
Morimura et al. (2011)
miR-182
qRT-PCR
Plasma
Chen et al. (2014)
miR-20a, miR-21, miR-24, miR-25, miR-99a, miR-185, miR-191
NGS, qRTPCR
Serum
Liu et al. (2012)
miR-21, miR-34a
qRT-PCR
Serum
Alemar et al. (2016)
miR-145, miR-150, miR-223, miR-636, miR-26b, miR-34a, miR-122, miR-126-5p, miR-145, miR-150, miR-223, miR-505, miR-636, miR-885.5p
qRT-PCR
W. blood
Schultz et al. (2014)
miR-10b, miR-155, miR-106b, miR-30c, miR-212
qRT-PCR
Bile
Cote et al. (2014)
miR-205, miR-210, miR-492, miR-1247
microarray, qRT-PCR
Pancreatic juice
Wang et al. (2014)
at the expense of increased invasiveness). One study used microarrays and qRT-PCR to measure miRNAs in pancreatic juice, and found the concentrations of miR-205, miR-210, miR492, and miR-1247 were not only increased in PDAC patients, but were also able to discriminate between PDAC and non-PDAC controls alone, or with increased sensitivity when combined with CA 19-9 results (Wang et al., 2014). Using the bile of PDAC patients, Cote et al. examined several miRNAs known to be involved in PDAC and found five (miR-10b, miR-155, miR-106b, miR-30c, and miR-212) that were elevated and the five miRNAs showed better diagnostic performance in bile compared to plasma, suggesting that miRNA in bile may offer more precise diagnosis in detecting PDAC.
CHALLENGES AND OPPORTUNITIES FOR miRNA BASED BIOMARKERS Challenges: There are number of factors that could affect the spectrum of circulating miRNA. While some of these factors are associated with samples such as sample types, sample collection time, and gender or state of the subject, other causes are experimental procedure related including sample processing, storage condition, RNA purification methods, or measurement platform (Lee, Baxter, Lee, Scherler, & Wang, 2017; Weber et al., 2010) To ensure sample consistency, several steps can be taken during sample collection and preparation, including limiting contamination from other elements (such as platelets or RBCs when
II. ONCOGENOMICS: CIRCULATING BIOMARKERS IN CLINICAL ONCOLOGY
202
13. CIRCULATING MIRNAS AS TUMOR BIOMARKERS
preparing plasma or serum), and collecting samples under the similar conditions (controlling for collection time and diet). The measurement platform greatly impacts the miRNA spectra and can contribute to bias in what miRNAs are prominently detected (Git et al., 2010; Wang et al., 2012). This is even true for intraplatform-specific differences (that is the same platform, but using reagents from different vendors) (Mestdagh et al., 2014). Some of the common platforms discussed in this chapter include qRT-PCR, microarrays, and NGS. qRTpCR is the most common measurement platform used in literature (see Tables 13.113.4) and has been considered the “gold standard” of assaying miRNAs as it is sensitive, quantifiable, and relatively low per assay cost. However, limitations include that it is low throughput, cannot detect novel miRNAs, and is affected by primer design. Microarrays have an advantage over qRT-PCR, as they can assay many miRNAs simultaneously, however they require larger amounts of RNA input, and also cannot identify novel miRNAs. Microarray probe design also affects its signal. NGS-based miRNA measurement (sRNAseq) allows for the identification of novel miRNAs, and avoids the sequencedependent primer and probe issues of qRT-PCR and microarrays. However, the sequencing of miRNAs from biofluids can be challenging, as inconsistencies exist between sequencing library construction kits, and computational support is needed to analyze the data (Yuan et al., 2016). Opportunities: Although we do not fully understand all the factors that could impact the level of miRNA in circulation, there is compelling evidence indicating that circulating RNAs play important roles in cancer diagnosis. Some cancer associated miRNAs along with other molecules—large RNAs, proteins, lipids— encapsulated in EVs may serve as biomarkers to reflect the status of cells in the disease site. While this chapter has primarily discussed miRNAs as potential biomarkers, other RNA species, like mRNAs, might also provide
insights to diseases, as there is a large diversity of cell-free mRNAs present in circulation, some of which have been proven to be useful in the detection and monitoring of various cancers (Garcı´a et al., 2008; Garcia et al., 2009; MarchVillalba et al., 2012). Additionally, while the majority of studies discussed here do not specifically look at EVs like exosomes or oncosomes, there has been considerable interest in using EVs as a source for circulating RNA biomarkers, as some of these vesicles are derived from tumors/CTCs. Further work detailing the intricate molecular cargo of these EVs is essential to understand the true source of these EVs, what their final targets may be, and how this may influence the local tumor microenvironment.
Acknowledgments We appreciate critical comments from Shannon Fallen. This work is supported by research contracts from DOD (W81XWH-16-1-0301 and W911NF-17-2-0086) and DTRA (HDTRA1-13-C-0055), and NIH grants (U01HL126496-02, R56HL133887, U01CA213330 and R01 DA040395).
References Abue, M., et al. (2015). Circulating miR-483-3p and miR-21 is highly expressed in plasma of pancreatic cancer. International Journal of Oncology, 46, 539547. Ahmed, F. E., et al. (2009). Diagnostic microRNA markers for screening sporadic human colon cancer and active ulcerative colitis in stool and tissue. Cancer Genomics Proteomics, 6, 281295. Ahmed, F. E., et al. (2013). Diagnostic microRNA markers to screen for sporadic human colon cancer in stool: I. Proof of principle. Cancer Genomics Proteomics, 10, 93113. Alemar, B., et al. (2016). miRNA-21 and miRNA-34a are potential minimally invasive biomarkers for the diagnosis of pancreatic ductal adenocarcinoma. Pancreas, 45, 8492. Al-Nedawi, K., et al. (2008). Intercellular transfer of the oncogenic receptor EGFRvIII by microvesicles derived from tumour cells. Nature Cell Biology, 10, 619624. Arroyo, J. D., et al. (2011). Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma. Proceedings of the National Academy of Sciences of the United States of America, 108, 50035008.
II. ONCOGENOMICS: CIRCULATING BIOMARKERS IN CLINICAL ONCOLOGY
REFERENCES
Bianchi, F., et al. (2011). A serum circulating miRNA diagnostic test to identify asymptomatic high-risk individuals with early stage lung cancer: Serum miRNAs to diagnose asymptomatic NSCLC. EMBO Molecular Medicine, 3, 495503. Boeri, M., et al. (2011). MicroRNA signatures in tissues and plasma predict development and prognosis of computed tomography detected lung cancer. Proceedings of the National Academy of Sciences of the United States of America, 108, 37133718. Boisen, M. K., et al. (2015). MicroRNA expression in formalin-fixed paraffin-embedded cancer tissue: Identifying reference microRNAs and variability. BMC Cancer, 15, 1024. Cazzoli, R., et al. (2013). microRNAs derived from circulating exosomes as noninvasive biomarkers for screening and diagnosing lung cancer. International Association for Study of Lung Cancer, 8, 11561162. Cha, D. J., et al. (2015). KRAS-dependent sorting of miRNA to exosomes. eLife, 4, e07197. Chang, K. H., Mestdagh, P., Vandesompele, J., Kerin, M. J., & Miller, N. (2010). MicroRNA expression profiling to identify and validate reference genes for relative quantification in colorectal cancer. BMC Cancer, 10, 173. Chang, T.-C., et al. (2007). Transactivation of miR-34a by p53 Broadly Influences Gene Expression and Promotes Apoptosis. Molecular Cell, 26, 745752. Chang, T.-C., et al. (2008). Widespread microRNA repression by Myc contributes to tumorigenesis. Nature Genetics, 40, 43. Chen, Q., Yang, L., Xiao, Y., Zhu, J., & Li, Z. (2014). Circulating microRNA-182 in plasma and its potential diagnostic and prognostic value for pancreatic cancer. Medical Oncology (Northwood; London; England), 31, 225. Chen, W., Cai, F., Zhang, B., Barekati, Z., & Zhong, X. Y. (2013). The level of circulating miRNA-10b and miRNA-373 in detecting lymph node metastasis of breast cancer: Potential biomarkers. International Society for Oncodevelopmental Biology and Medicine, 34, 455462. Chen, X., et al. (2008). Characterization of microRNAs in serum: A novel class of biomarkers for diagnosis of cancer and other diseases. Cell Research, 18, 9971006. Cheng, H., et al. (2011). Circulating plasma MiR-141 is a novel biomarker for metastatic colon cancer and predicts poor prognosis. PLoS One, 6, e17745. Cimmino, A., et al. (2005). miR-15 and miR-16 induce apoptosis by targeting BCL2. Proceedings of the National Academy of Sciences of the United States of America, 102, 1394413949. Cote, G. A., et al. (2014). A pilot study to develop a diagnostic test for pancreatic ductal adenocarcinoma based on differential expression of select miRNA in plasma and bile. American Journal of Gastroenterology, 109, 19421952.
203
Cuk, K., et al. (2013). Circulating microRNAs in plasma as early detection markers for breast cancer. International Journal of Cancer, 132, 16021612. Demory Beckler, M., et al. (2013). Proteomic analysis of exosomes from mutant KRAS colon cancer cells identifies intercellular transfer of mutant KRAS. Molecular & Cellular Proteomics, 12, 343355. Denzer, K., Kleijmeer, M. J., Heijnen, H. F., Stoorvogel, W., & Geuze, H. J. (2000). Exosome: From internal vesicle of the multivesicular body to intercellular signaling device. Journal of Cell Science, 113(Pt 19), 33653374. Diamandis, E. P. (2012). The failure of protein cancer biomarkers to reach the clinic: Why, and what can be done to address the problem? BMC Medicine, 10, 87. Di Vizio, D., et al. (2012). Large oncosomes in human prostate cancer tissues and in the circulation of mice with metastatic disease. American Journal of Pathology, 181, 15731584. Donzelli, S., et al. (2015). Epigenetic silencing of miR-1455p contributes to brain metastasis. Oncotarget, 6, 3518335201. Eichelser, C., Flesch-Janys, D., Chang-Claude, J., Pantel, K., & Schwarzenbach, H. (2013). Deregulated serum concentrations of circulating cell-free microRNAs miR-17, miR-34a, miR-155, and miR-373 in human breast cancer development and progression. Clinical Chemist, 59, 14891496. Eriksen, A. H. M., et al. (2016). MicroRNA expression profiling to identify and validate reference genes for the relative quantification of microRNA in rectal cancer. PLoS One, 11, e0150593. Feng, X., Wang, Z., Fillmore, R., & Xi, Y. (2014). MiR-200, a new star miRNA in human cancer. Cancer Letters (New York NY United States), 344, 166173. Fu¨ze´ry, A. K., Levin, J., Chan, M. M., & Chan, D. W. (2013). Translation of proteomic biomarkers into FDA approved cancer diagnostics: Issues and challenges. Clinical Proteomics, 10, 13. Gao, W., et al. (2010). Deregulated expression of miR-21, miR-143 and miR-181a in non small cell lung cancer is related to clinicopathologic characteristics or patient prognosis. Biomed. Pharmacother. Biome´d. Pharmacothe´rapie, 64, 399408. Garcı´a, V., et al. (2008). Free circulating mRNA in plasma from breast cancer patients and clinical outcome. Cancer Letters (New York NY United States), 263, 312320. Garcia, V., et al. (2009). Extracellular tumor-related mRNA in plasma of lymphoma patients and survival implications. PLoS One, 4, e8173. Gira´ldez, M. D., et al. (2013). Circulating microRNAs as biomarkers of colorectal cancer: Results from a genome-wide profiling and validation study. Clinical Gastroenterology and Hepatology, 11, 681688.e3.
II. ONCOGENOMICS: CIRCULATING BIOMARKERS IN CLINICAL ONCOLOGY
204
13. CIRCULATING MIRNAS AS TUMOR BIOMARKERS
Git, A., et al. (2010). Systematic comparison of microarray profiling, real-time PCR, and next-generation sequencing technologies for measuring differential microRNA expression. RNA, 16, 9911006. Hamam, R., et al. (2017). Circulating microRNAs in breast cancer: Novel diagnostic and prognostic biomarkers. Cell Death & Disease, 8, e3045. Hanash, S. M. (2011). Why have protein biomarkers not reached the clinic? Genome Medicine, 3, 66. Hansen, T. F., Carlsen, A. L., Heegaard, N. H. H., Sørensen, F. B., & Jakobsen, A. (2015). Changes in circulating microRNA-126 during treatment with chemotherapy and bevacizumab predicts treatment response in patients with metastatic colorectal cancer. British Journal of Cancer, 112, 624629. Hayashita, Y., et al. (2005). A polycistronic microRNA cluster, miR-17-92, is overexpressed in human lung cancers and enhances cell proliferation. Cancer Research, 65, 96289632. He, L., et al. (2007). A microRNA component of the p53 tumour suppressor network. Nature, 447, 11301134. Heneghan, H. M., Miller, N., Kelly, R., Newell, J., & Kerin, M. J. (2010). Systemic miRNA-195 differentiates breast cancer from other malignancies and is a potential biomarker for detecting noninvasive and early stage disease. The Oncologist, 15, 673682. Hermeking, H. (2010). The miR-34 family in cancer and apoptosis. Cell Death & Differentiation, 17, 193199. Hu, Z., et al. (2010). Serum microRNA signatures identified in a genome-wide serum microRNA expression profiling predict survival of non-small-cell lung cancer. Journal of Clinical Oncology, 28, 17211726. Huang, Y., et al. (2011). Biological functions of microRNAs: A review. Journal of Physiology and Biochemistry, 67, 129139. Huang, Z., et al. (2010). Plasma microRNAs are promising novel biomarkers for early detection of colorectal cancer. International Journal of Cancer, 127, 118126. Kong, X., et al. (2011). Detection of differentially expressed microRNAs in serum of pancreatic ductal adenocarcinoma patients: MiR-196a could be a potential marker for poor prognosis. Digestive Diseases and Sciences, 56, 602609. Koppers-Lalic, D., et al. (2014). Nontemplated nucleotide additions distinguish the small RNA composition in cells from exosomes. Cell Reports, 8, 16491658. Kosaka, N., et al. (2010). Secretory mechanisms and intercellular transfer of microRNAs in living cells. Journal of Biological Chemistry, 285, 1744217452. Lawrie, C. H., et al. (2008). Detection of elevated levels of tumour-associated microRNAs in serum of patients with diffuse large B-cell lymphoma. British Journal of Haematology, 141, 672675.
Le, H.-B., et al. (2012). Evaluation of dynamic change of serum miR-21 and miR-24 in pre- and post-operative lung carcinoma patients. Medical Oncology (Northwood, London, England), 29, 31903197. Lee, I., Baxter, D., Lee, M. Y., Scherler, K., & Wang, K. (2017). The Importance of Standardization on Analyzing Circulating RNA. Molecular Diagnosis & Therapy, 21, 259268. Lee, R. C., Feinbaum, R. L., & Ambros, V. (1993). The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell, 75, 843854. Lehmann, U., et al. (2008). Epigenetic inactivation of microRNA gene hsa-mir-9-1 in human breast cancer. Journal of Pathology, 214, 1724. Li, Y., Li, W., Ouyang, Q., Hu, S., & Tang, J. (2011). Detection of lung cancer with blood microRNA-21 expression levels in Chinese population. Oncology Letters, 2, 991994. Liu, R., et al. (2012). Serum MicroRNA Expression Profile as a Biomarker in the Diagnosis and Prognosis of Pancreatic Cancer. Clinical Chemist, 58, 610618. Liu, X.-G., et al. (2012). High expression of serum miR-21 and tumor miR-200c associated with poor prognosis in patients with lung cancer. Medical Oncology (Northwood, London, England), 29, 618626. Lujambio, A. (2007). CpG island hypermethylation of tumor suppressor microRNAs in human cancer. Cell Cycle, 6, 14541458. Lujambio, A., et al. (2008). A microRNA DNA methylation signature for human cancer metastasis. Proceedings of the National Academy of Sciences of the United States of America, 105, 1355613561. Luo, X., Stock, C., Burwinkel, B., & Brenner, H. (2013). Identification and evaluation of plasma microRNAs for early detection of colorectal cancer. PLoS One, 8, e62880. Madhavan, D., et al. (2016). Circulating miRNAs with prognostic value in metastatic breast cancer and for early detection of metastasis. Carcinogenesis. Available from https://doi.org/10.1093/carcin/bgw008. Mar-Aguilar, F., et al. (2013). Serum circulating microRNA profiling for identification of potential breast cancer biomarkers. Disease Markers, 34, 163169. March-Villalba, J. A., et al. (2012). Cell-free circulating plasma hTERT mRNA is a useful marker for prostate cancer diagnosis and is associated with poor prognosis tumor characteristics. PLoS One, 7, e43470. Matamala, N., et al. (2015). Tumor microRNA expression profiling identifies circulating microRNAs for early breast cancer detection. Clinical Chemist, 61, 10981106. Mavrakis, K. J., et al. (2010). Genome-wide RNA-mediated interference screen identifies miR-19 targets in Notchinduced T-cell acute lymphoblastic leukaemia. Nature Cell Biology, 12, 372379.
II. ONCOGENOMICS: CIRCULATING BIOMARKERS IN CLINICAL ONCOLOGY
REFERENCES
Mestdagh, P., et al. (2014). Evaluation of quantitative miRNA expression platforms in the microRNA quality control (miRQC) study. Nature Methods, 11, 809815. Mitchell, P. S., et al. (2008). Circulating microRNAs as stable blood-based markers for cancer detection. Proceedings of the National Academy of Sciences of the United States of America, 105, 1051310518. Morello, M., et al. (2013). Large oncosomes mediate intercellular transfer of functional microRNA. Cell Cycle Georgetown, Tex, 12, 35263536. Morimura, R., et al. (2011). Novel diagnostic value of circulating miR-18a in plasma of patients with pancreatic cancer. British Journal of Cancer, 105, 17331740. Ng, E. K. O., et al. (2009). Differential expression of microRNAs in plasma of patients with colorectal cancer: A potential marker for colorectal cancer screening. Gut, 58, 13751381. O’Donnell, K. A., Wentzel, E. A., Zeller, K. I., Dang, C. V., & Mendell, J. T. (2005). c-Myc-regulated microRNAs modulate E2F1 expression. Nature, 435, 839843. Ogata-Kawata, H., et al. (2014). Circulating exosomal microRNAs as biomarkers of colon cancer. PLoS One, 9, e92921. Peltier, H. J., & Latham, G. J. (2008). Normalization of microRNA expression levels in quantitative RT-PCR assays: Identification of suitable reference RNA targets in normal and cancerous human solid tissues. RNA NYN, 14, 844852. Petriella, D., et al. (2015). miRNA profiling in serum and tissue samples to assess noninvasive biomarkers for NSCLC clinical outcome. Tumour Biology: The Journal of the International Society for Oncodevelopmental Biology and Medicine. Available from https://doi.org/10.1007/ s13277-015-4391-1. Raver-Shapira, N., et al. (2007). Transcriptional activation of miR-34a contributes to p53-mediated apoptosis. Molecular Cell, 26, 731743. Roth, C., et al. (2010). Circulating microRNAs as bloodbased markers for patients with primary and metastatic breast cancer. Breast Cancer Research BCR, 12, R90. Santangelo, L., et al. (2016). The RNA-Binding Protein SYNCRIP Is a Component of the Hepatocyte Exosomal Machinery Controlling MicroRNA Sorting. Cell Reports, 17, 799808. Schultz, N. A., Dehlendorff, C., Jensen, B. V., et al. (2014). MIcrorna biomarkers in whole blood for detection of pancreatic cancer. JAMA, 311, 392404. Shen, J., et al. (2011). Plasma microRNAs as potential biomarkers for non-small-cell lung cancer. Laboratory Investigation; a Journal of Technical Methods and Pathology, 91, 579587.
205
Shen, J., et al. (2014). Circulating miR-148b and miR-133a as biomarkers for breast cancer detection. Oncotarget, 5, 52845294. Shimomura, A., et al. (2016). Novel combination of serum microRNA for detecting breast cancer in the early stage. Cancer Prevention Research, 107, 326334. Toiyama, Y., et al. (2014). Serum miR-200c is a novel prognostic and metastasis-predictive biomarker in patients with colorectal cancer. Annals of Surgery, 259, 735743. Turchinovich, A., Weiz, L., Langheinz, A., & Burwinkel, B. (2011). Characterization of extracellular circulating microRNA. Nucleic Acids Research, 39, 72237233. Valadi, H., et al. (2007). Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nature Cell Biology, 9, 654659. Vickers, K. C., Palmisano, B. T., Shoucri, B. M., Shamburek, R. D., & Remaley, A. T. (2011). MicroRNAs are transported in plasma and delivered to recipient cells by high-density lipoproteins. Nature Cell Biology, 13, 423433. Villarroya-Beltri, C., et al. (2013). Sumoylated hnRNPA2B1 controls the sorting of miRNAs into exosomes through binding to specific motifs. Nature Communications, 4, 2980. Wagner, J., et al. (2013). Characterization of levels and cellular transfer of circulating lipoprotein-bound microRNAs. Arteriosclerosis, Thrombosis, and Vascular Biology, 33, 13921400. Wang, F., Zheng, Z., Guo, J., & Ding, X. (2010). Correlation and quantitation of microRNA aberrant expression in tissues and sera from patients with breast tumor. Gynecologic Oncology, 119, 586593. Wang, J., et al. (2009). MicroRNAs in plasma of pancreatic ductal adenocarcinoma patients as novel blood-based biomarkers of disease. Cancer Prevention Research, 2, 807813. Wang, J., et al. (2014). Circulating microRNAs in Pancreatic Juice as Candidate Biomarkers of Pancreatic Cancer. Journal of Cancer, 5, 696705. Wang, K., Zhang, S., Weber, J., Baxter, D., & Galas, D. J. (2010). Export of microRNAs and microRNA-protective protein by mammalian cells. Nucleic Acids Research, 38, 72487259. Wang, K., et al. (2012). Comparing the MicroRNA spectrum between serum and plasma. PLoS One, 7, e41561. Wang, L.-G., & Gu, J. (2012). Serum microRNA-29a is a promising novel marker for early detection of colorectal liver metastasis. Cancer Epidemiology, 36, e61e67. Wang, Z.-X., et al. (2011). Prognostic significance of serum miRNA-21 expression in human non-small cell lung cancer. Journal of Surgical Oncology, 104, 847851.
II. ONCOGENOMICS: CIRCULATING BIOMARKERS IN CLINICAL ONCOLOGY
206
13. CIRCULATING MIRNAS AS TUMOR BIOMARKERS
Waters, P. S., et al. (2014). Impact of Tumour Epithelial Subtype on Circulating microRNAs in Breast Cancer Patients. PLoS One, 9, e90605. Weber, J. A., et al. (2010). The microRNA spectrum in 12 body fluids. Clinical Chemist, 56, 17331741. Wozniak, M. B., et al. (2015). Circulating microRNAs as non-invasive biomarkers for early detection of nonsmall-cell lung cancer. PLoS One, 10, e0125026. Wu, C. W., et al. (2012). Detection of miR-92a and miR-21 in stool samples as potential screening biomarkers for colorectal cancer and polyps. Gut, 61, 739745. Xiao, J., Lin, H., Luo, X., Luo, X., & Wang, Z. (2011). miR605 joins p53 network to form a p53:miR-605:Mdm2 positive feedback loop in response to stress. EMBO Journal, 30, 524532. Xie, Y., et al. (2010). Altered miRNA expression in sputum for diagnosis of non-small cell lung cancer. Lung Cancer Amsterdam, Netherlands, 67, 170176. Yamakuchi, M., & Lowenstein, C. J. (2009). MiR-34, SIRT1, and p53: The feedback loop. Cell Cycle, 8, 712715. Yamakuchi, M., et al. (2010). P53-induced microRNA-107 inhibits HIF-1 and tumor angiogenesis. Proceedings of the National Academy of Sciences of the United States of America, 107, 63346339.
Yang, J., et al. (2016). Expression analysis of microRNA as prognostic biomarkers in colorectal cancer. Oncotarget, 8, 5240352412. Yu, S.-L., et al. (2008). MicroRNA signature predicts survival and relapse in lung cancer. Cancer Cell, 13, 4857. Yuan, T., et al. (2016). Plasma extracellular RNA profiles in healthy and cancer patients. Scientific Reports, 6, 19413. Zhang, Y., Liao, J.-M., Zeng, S. X., & Lu, H. (2011). p53 downregulates Down syndrome-associated DYRK1A through miR-1246. EMBO Reports, 12, 811817. Zheng, G., et al. (2013). Identification and validation of reference genes for qPCR detection of serum microRNAs in colorectal adenocarcinoma patients. PLoS One, 8. Zhu, J., et al. (2014). Different miRNA expression profiles between human breast cancer tumors and serum. Frontiers in Genetics, 5. Zhu, M., et al. (2017). A panel of microRNA signature in serum for colorectal cancer diagnosis. Oncotarget, 8, 1708117091. Zhu, W., Qin, W., Atasoy, U., & Sauter, E. R. (2009). Circulating microRNAs in breast cancer and healthy subjects. BMC Research Notes, 2, 89. Zhu, Y., et al. (2016). Fecal miR-29a and miR-224 as the noninvasive biomarkers for colorectal cancer. Cancer Biomarkers: Section A of Disease Markers, 16, 259264.
II. ONCOGENOMICS: CIRCULATING BIOMARKERS IN CLINICAL ONCOLOGY