Abstract
Colorectal carcinogenesis represents a sequential progression of normal colonic mucosa from adenoma to carcinoma. It has become apparent that miRNA deregulation contributes to the initiation and progression of colorectal cancer (CRC). These oncogenic or tumor-suppressive miRNAs interact with intracellular signaling networks and lead to alteration of cell proliferation, apoptosis, metastasis and even response to chemotherapeutic treatments. This article aims to review the cutting edge progress in the discovery of the role of novel mechanisms for miRNAs in the development of CRC. We will also discuss the potential use of miRNAs as biomarkers for early diagnosis and prognosis of CRC. Furthermore, with advancements in RNA delivery technology, it is anticipated that manipulation of miRNAs may offer an alternative therapy for CRC treatment.
Keywords: carcinogenesis, clinical application, colorectal cancer, diagnosis, miRNA, predictive biomarkers, prognosis
Colorectal cancer (CRC) is one of the leading malignancies in the western countries, and is the second leading cause of cancer-related deaths in the USA. The pathogenesis of CRC usually follows a protracted stepwise sequence from benign polyps to malignant adenocarcinoma and distant metastasis [1]. Considering that it may take up to a decade or longer for this disease to fully develop and it is a truly preventable disease if diagnosed early, there is an urgent need for developing potential biomarkers for early detection and disease prevention, as well as therapeutic targets for treatment, leading to an overall improvement in reducing the mortality burden a ssociated with this disease.
Although the etiology of CRC is multifactorial, genetic and epigenetic alterations remain the central theme and fundamental mechanisms underlying its pathogenesis. The aberrant expression or deregulation of oncogenes and tumor suppressor genes leads to the malignant transformation of normal intestinal epithelial cells by several molecular processes including cell proliferation, a poptosis, invasion, angiogenesis, multidrug resistance and maintenance of genetic and genomic stability [2].
Discovery of miRNAs has opened a new frontier of cancer research and provides an unprecedented potential for the development of diagnostic biomarkers, as well as identification of novel miRNA-based treatment targets that can influence downstream genes and signaling pathways. A growing body of published studies has recently shown that miRNAs are involved in regulation of cancer-related genes through post-transcriptional processing, indicating that miRNAs play an important role during colorectal carcinogenesis. In this review, we will discuss global differences in miRNA expression in colorectal tumors, followed by discussion for the function of specific miRNAs in carcinogenesis and their potential diagnostic, prognostic and therapeutic applications for the management of CRC in the clinic.
miRNA expression is frequently & consistently altered in CRC
Regulation of normal function of intestinal epithelial cells is controlled by miRNAs. Therefore, it is not surprising that dysregulation of miRNA expression has been linked to various human cancers. Several groups have reported that global miRNA expression patterns are frequently altered in CRC [3,4]. Based upon hundreds and thousands of scientific studies, several miRNA-related databases have been curated, such as miRNA body map analysis [5] and Human MicroRNA Disease Database or HMDD [6], which hold expression profiles of miRNAs from several hundred patients with CRC and normal healthy subjects. The differential expression of selected miRNAs in tumor versus healthy, control tissues is clearly highlighted in these databases. In this review, we searched NIH’s PubMed database for literature searches from 2012 to 2015, and have summarized all the key miRNAs that have been reported to be differentially expressed in CRC (Table 1). Based on these expression studies, the aberrant expression of miRNAs and their downstream intracellular gene targets, provide us with the global landscape of molecular events that contribute to colorectal carcinogenesis, as well as highlight the potential of specific miRNAs that can be used as candidates for development of disease biomarkers.
Table 1.
The novel functional role of miRNAs in the colorectal cancer progression.
| miRNA | Target genes | Phenotype(s) affected | Ref. |
|---|---|---|---|
| Upregulated miRNAs | |||
|
| |||
| miR-17 | PTEN | Cell invasiveness | [7] |
|
| |||
| miR-31 | SATB2 | Proliferation, invasion and migration | [8] |
| RASA1 | Tumor growth | [9] | |
| RhoBTB1 | Proliferation | [10] | |
|
| |||
| miR-92a | PTEN | Migration | [11] |
|
| |||
| miR-96 | TP53INP1/FOXO1/ FOXO3a | Proliferation | [12] |
|
| |||
| miR-103/107 | DAPK/ KLF4 | Cell motility, cell–matrix adhesion and cell–cell adhesion | [13] |
|
| |||
| miR-130b | Integrin β1 | Migration and invasion | [14] |
|
| |||
| miR-153 | FOXO3a | Cell invasiveness and response to chemotherapy | [15] |
|
| |||
| miR-181a | WIF-1 | Migration and invasion | [16] |
|
| |||
| miR-221/222 | RelA | Proliferation and colony formation | [17] |
|
| |||
| miR-224 | PHLPP1/2 | Proliferation, tumor growth | [18] |
|
| |||
| miR-301a | SOCS6 | Proliferation, migration and invasion, tumor growth | [19] |
|
| |||
| miR-362 | E2F1/USF2/PTPN1 | Proliferation, cell cycle | [20] |
|
| |||
| miR-429 | SOX2 | Apoptosis | [21] |
|
| |||
| miR-574 | Qki6/7 | Proliferation, migration and invasion, differentiation and cell cycle | [22] |
|
| |||
| miR-708 | CDKN2B | Proliferation and invasion | [23] |
|
| |||
| Downregulated miRNAs | |||
|
| |||
| let-7c | MMP11 | Migration and invasion | [24] |
|
| |||
| miR-7 | EGFR | Response to cetuximab | [25] |
| YY1 | Proliferation, apoptosis, cell cycle | [26] | |
|
| |||
| miR-18a | CDC42 | Migration, cell cycle, apoptosis | [27] |
|
| |||
| miR-27a | SGPP1 | Proliferation, apoptosis and cell migration | [28] |
|
| |||
| miR-27b | VEGFC | Proliferation, colony formation | [29] |
|
| |||
| miR-29c | GNA13 | Migration and invasion | [30] |
|
| |||
| miR-30a | IRS2 | Proliferation, migration and invasion | [31] |
|
| |||
| miR-30b | KRAS/PIK3CD/BCL2 | Proliferation and tumor growth | [32] |
|
| |||
| miR-100 | RAP1B | Cell growth and invasion and induce apoptosis | [33] |
|
| |||
| miR-124 | PRRX1 | Response to radiosensitivity | [34] |
|
| |||
| miR-126 | CXCR4 | Proliferation, migration and invasion and cell cycle | [35,36] |
| IRS-1 | Proliferation, migration, invasion and cell cycle | [37] | |
| VEGF | Migration and invasion, tumor growth | [38] | |
| RhoA/ROCK | Cell cycle, invasion, tumor growth | [39] | |
|
| |||
| miR-133b | CXCR4 | Invasion and apoptosis | [40] |
|
| |||
| miR-139 | AMFR/NOTCH1 | Migration and invasion | [41] |
| NOTCH1 | Tumor growth | [42] | |
|
| |||
| miR-143 | IGF1R | Proliferation | [43] |
| IGF1R | Proliferation, migration, angiogenesis and chemosensitivity | [44] | |
| TLR2 | Invasion and migration | [45] | |
| HK2 | Metabolism | [46] | |
| MACC1 | Invasion and migration | [47] | |
|
| |||
| miR-144 | mTOR | Proliferation, response to rapamycin | [48] |
|
| |||
| miR-145 | fascin-1 | Proliferation, migration and invasion | [49] |
| catenin δ-1 | Tumor growth | [50] | |
| N-RAS, IRS1 | Proliferation, migration and invasion | [51] | |
|
| |||
| miR-146a | MMP16 | Migration and invasion | [52] |
|
| |||
| miR-185 | STIM1 | Migration and invasion | [53] |
|
| |||
| miR-199a | DDR1 | Proliferation, colony formation, cell cycle, invasion and migration | [54] |
|
| |||
| miR-202 | ARL5A | Tumor growth | [55] |
|
| |||
| miR-203 | TYMS | Response to 5-fluorouracil | [56] |
|
| |||
| miR-212 | MnSOD | Migration and invasion | [57] |
|
| |||
| miR-375 | PIK3CA | Tumor growth | [58] |
|
| |||
| miR-378 | VIMENTIN | Tumor growth and invasion | [59] |
|
| |||
| miR-399 | MDM2 | Apoptosis, senescence, migration and invasion | [60] |
|
| |||
| miR-455 | RAF1 | Proliferation and invasion | [61] |
|
| |||
| miR-497 | IGF1-R | Proliferation, invasion and apoptosis | [62] |
Upregulation and downregulation of miRNAs is defined by differential expression between cancer vs matched normal tissues.
The functional role of miRNAs in the initiation of CRC development
Just in a short time span during the last decade, plethora of scientific studies have been gathered that described a mechanistic role for altered miRNAs expression patterns in the initiation of CRC. Notwithstanding the challenge to adequately review each of these studies within the scope of this review article, we will summarize some of recent important findings on specific miRNAs and their critical role in CRC development (Table 1 & Figure 1).
Figure 1. A network of miRNAs involved in the multiple steps of the development of colorectal cancer.
Colorectal carcinogenesis follows a stepwise sequence where normal colonic epithelium transforms into adenomatous polyps followed by progression to malignant carcinoma. Specific miRNAs such as miR-143 involve in the initiation of colorectal cancer. Neoplastic cells may acquire the ability to invade or spread to distant through complex processes including directional activation of proteolytic enzymes, epithelial-to-mesenchymal transition and translocation of cancer cells. miR-146 promote invasion via targeting MMP proteins and miR-200 family contributed to epithelial-to-mesenchymal transition. As a part of metastatic process, colorectal cancer cells must enter the bloodstream. miR-21, miR-29c and miR-22, among others, modulate colorectal cancer cell intravasation. Once tumor cells enter the bloodstream, they utilize specific mechanisms to avoid immune system attack. Tumor-derived exosomes containing miR-146a, miR-29a and miR-21 affect lymphocyte development and function. Metastatic colonization is the final step during cancer progression. It was demonstrated miR-126, miR-26b and miR-146 promote tumor growth at the metastatic sites. Furthermore, angiogenesis, which represents the growth of new blood vessels, is also an essential component for colorectal cancer growth and distant metastasis. miR-27b, miR-143, miR-126 and miR-145 were shown as a critical role in the regulation of angiogenesis and the VEGF pathway.
One of the first miRNAs to be implicated in colon cancer was miR-143. miR-143 is particularly interesting because it possesses tumor suppressive activity and its expression is substantially reduced in several human cancers, especially in CRC [45,63–64]. Since downregulated miR-143 expression can frequently precede APC gene mutations, it is possible that miR-143 downregulation is essential for the initiation of CRC development [65]. Furthermore, reduced expression of miR-143 was also observed in dextran sulfate sodium (DSS)-induced chronic inflammation mouse model [66]. As inflammation is known to provide a procarcinogenic milieu for tumorigenesis, decreased levels of miR-143 indicate an early step in malignant transformation of normal colonic epithelium. It is also noted that constitutively active KRAS mutations, which occurs in half of all CRCs, repress miR-143 expression via its downstream effector RREB1 [67], suggesting that downregulation of miR-143 is indispensable for the subsequent CRC development. In addition, other studies have implicated tumor suppressive role of miR-143 by uncovering its target genes such as IGF-IR [43], TLR2 [45], D44, KLF5, KRAS and BRAF [68], and DNMT3A [69].
Recently, miR-31 has been shown to contribute to the initiation of CRC. Necela et al. [70] used quantitative PCR arrays to compare miRNA expression in tumors and normal colonic epithelial cells isolated from distal colons of chronically-inflamed mice and APC(Min/+) mice. miR-31 expression was upregulated in all these tumor types. Furthermore, miR-31 levels were found to be increased during inflammatory bowel disease-associated neoplastic transformation, suggesting that the elevated levels miR-31 is indicative of probably an early event in CRC development.
Colorectal carcinogenesis follows a stepwise sequence where normal colonic epithelium transforms into adenomatous polyps followed by progression to malignant carcinoma. Revealing temporal pattern of miRNAs expression alterations during progression in the well-established normal-adenoma-adenocarcinoma sequence will help us to understand the functional role of miRNAs in the initiation of CRC development. Bartley et al. [71] analyzed 69 matched specimens of adenomas with low and high grade dysplasia, adenocarcinomas and normal mucosa by using miRNA microarrays. These investigators clustered differentially expressed miRNAs into two patterns of early (type 1) and late (type 2) expression pattern. Type 1 pattern showed miRNAs that were differentially expressed between normal mucosa and low-grade dysplasia, while type 2 pattern revealed miRNAs alterations between late stage versus early stage CRCs. Since Wnt signaling is considered one of the driver pathways in colorectal carcinogens, these authors also analyzed the correlation between expression of type 1 miRNAs and β-catenin localization in the cancer cells. Not surprisingly, they discovered that expression alterations in type 1 miRNAs preceded nuclear β-catenin accumulation, and some of these miRNAs targeted various component of Wnt signaling pathway. APC mutations are known to trigger Wnt signaling activation. However, since APC mutations are uncommon in colorectal adenomas, these type 1 pattern miRNAs targeting Wnt signaling probably contributed to the initiation of CRC. Another group used similar strategy to compare miRNA expression in cancer tissues and colon tissues from chronically inflamed mice and APC (Min/+) mice. Eight miRNAs were differentially expressed in tumors due to presence of germline APC mutations, while expression of other seven miRNAs was altered due to chronic inflammation [70]. Among these miRNAs, miR-215 and miR-133a were found to belong to type 1 pattern miRNAs, suggesting these two miRNAs have important role in the initiation of CRC development. In addition, the effect of DNA mismatch repair (MMR) status on miRNA expression pattern has also been of interest considering MMR-deficient cancers constitute approximately 15% of all neoplasms, and these patients undergo different clinical management [72]. One of the studies showed that several miRNAs in early stage of disease are important in both MMR-deficient and MMR-proficient CRC. However, four miRNAs (miR-31, miR-552, miR-592 and miR-224) were differentially expressed in MMR-deficient and proficient CRC [73]. Our group also found that Lynch syndrome CRCs displayed a unique miRNA profile compared with sporadic microsatellite instability tumors, suggesting MMR status affects miRNA expression pattern in the transition from normal epithelia to colonic neoplasia [74].
miRNAs affect the invasion & metastasis in CRC
• Angiogenesis
Angiogenesis, which represents the growth of new blood vessels, is an essential component for CRC growth and distant metastasis [75]. Furthermore, tumor [76] angiogenesis is a multi-step process precisely regulated by a number of pro- and anti-angiogenic molecules. In the past decades, several studies have identified a number of growth factor receptor pathways that promote tumor angiogenesis. One of the major pathways involved in this process is the VEGF family of proteins and receptors. Previous studies demonstrated a role for miRNAs in the regulation of angiogenesis and the VEGF pathway [77]. miR-126 was reported to regulate angiogenesis by interfering with VEGF signaling pathway in different cell types [78–81]. In CRC, lower miR-126 expression was associated with poor survival [38,82]. Reduced miR-126 expression was in part caused by promoter methylation of its host gene, EGFL7. Treatment with 5-aza-CdR restored miR-126 expression in cancer cell lines and thereby led to decreased VEGF expression, suggesting miR-126 may be a potential therapeutic target for CRC [38]. Other studies have reported several important miRNAs such as miR-27b [29], miR-143 [44] and miR-145 [51], all of which interact with components of VEGF signaling pathway in CRC angiogenesis. Collectively, discovery of pro- and antiangiogenic miRNAs may possibly be used as diagnostic markers and potential therapeutic targets in the near future.
• Cell invasion
During CRC development, neoplastic cells may acquire the ability to invade or spread to distant organs through complex processes including directional activation of proteolytic enzymes, epithelial-to-mesenchymal transition (EMT) and translocation of cancer cells. Matrix metalloproteases (MMPs) belong to zinc-dependent protease family, and are responsible for pericellular proteolysis for the regulation of neoplastic invasion. One such miRNA, let-7c was found to directly destabilize the MMP11 mRNA, leading to the suppression of cellular migration and invasion in CRC cells [24]. With forced expression of miR-146a in CaCo-2 and HT-29 colon cancer cells, a decrease in mRNA and protein expression levels of MMP-16, and a lower gelatinase activity in a gelatin zymography was also observed [52]. EMT is a highly conserved cellular process which allows polarized, immotile epithelial cells to transform into motile mesenchymal cells. CRC cells undergoing EMT may represent higher metastatic potential and stem cell characteristics. A reversible biological process called mesenchymal-to-epithelial transition (MET), is also an important step for tumor metastasis since MET allows colon cancers to regain epithelial properties and facilitates spread of metastatic cells to distant organs. miR-200 cluster was reported to be involved in the regulation of EMT-MET plasticity in colon cancer. Our group found liver metastasis tissues showed higher expression of miR-200c compared with primary CRCs. The invasive front in primary CRC tissues revealed low expression of miR-200c, and induced overexpression of miR-200c in colon cancer cells contributed to MET through increased E-cadherin and reduced vimentin expression [83]. Another group showed the expression pattern of miR-200 family in EMT and MET in the metastasis cascade. These investigators found decreased expression of miR-200 family members at the invasive front of adenocarcinomas but increased miR-200 level at regional lymph node metastases and vascular carcinoma. Furthermore, the expression of miR-200 was uniform from the tumor core to the tumor-normal interface. These results revealed miR-200 was downregulated when cancer cell underwent invasion, but its expression was restored at metastatic sites [84]. Ascl2, a downstream target of Wnt signaling, is known to regulate EMT and MET programs in colon cancer cells. However, the mechanisms underlying this process still remain unknown. Recently, Tian et al. found Ascl2 to regulate EMT-MET plasticity through modulation of various miR-200 family members including, miR-200a/b/c, miR-429 and miR-141 [85]. Another key miRNA involved in EMT is miR-34a. It was reported that miR-34a and its target gene snail1, which is an EMT driver gene, forms a forward-feedback loop to promote epithelial–mesenchymal transition in CRC [86]. Likewise, another group revealed an active IL-6R/STAT3/miR-34a loop for EMT in CRC cell lines, and also found upregulation of p-STAT3, IL-6R and Snail in colitis-associated intestinal tumors in miR-34 deficient mice [87].
• Intravasation, circulation & extravasation
As a part of metastatic process, CRC cells must enter the bloodstream, defined as intravasation, followed by their dissemination to other parts of the body. The precise mechanisms underlying this process require collagenases, MMPs, urokinase plasminogen activator (uPA), uPA receptor (uPAR) and EMT [88]. Therefore, miRNAs that can modulate cellular invasion are also involved in regulating CRC cell intravasation. Oncogenic miR-21, for example, plays a role in intra vasation through downregulation of the tumor suppressor Pdcd4 [89]. In cancer cells, CD151 was reported to regulate adhesion-dependent signaling and postadhesion events, including intravasation and extravasation [90]. Several miRNAs including miR-22 [91], miR-124 [92] and miR-506 [93] were reported to target CD151 and subsequently cause inhibition of CD151 expression. Furthermore, other essential enzymes required for more efficient intravasation including collagenases, and extracellular matrix proteins such as collagen and laminin gamma-1 were found to be targeted by miR-29c [94].
Once tumor cells enter the bloodstream, they utilize specific mechanisms to avoid shear stress and immune system attack [95]. Cancer cells can be eliminated by innate immune cells like natural killer cells. Currently, tumor-derived exosomes are shown to be of functional relevance for tumorigenesis, as these evade immune attack. Since these vesicles derived tumor cells contain biological active miRNAs which target components critical for the function or maturation of immune cells. In breast cancer, for example, tumor-derived exosomes containing miR-146a, miR-29a and miR-21 affect lymphocyte development and function [96]. In CRC, exosome-associated miRNAs including let-7a, miR-1229, miR-150, miR-21, miR-23a and miR-1246 were expressed much higher than healthy subjects [97]. Among these miRNAs, miR-23a was found to be a strong repressor of transcription factor BLIMP-1 which promotes cytotoxic T lymphocyte activity [98]. In addition, miR-21 was shown to be a negative mediator of signal transduction downstream of TCR in T-lymphocytes. miR-21 inhibition revealed enhanced IFN-Γ production and strong activation in response to TCR engagement [99]. HLA-G is a ligand for both natural killer cells and cytotoxic T lymphocyte, which promotes the activity of both types of immune cells. Recently, miRNAs including miR-148 family and miR-133a have been identified to target HLA-G, which prevents malignant cells from elimination by immune effector cells [100]. Likewise, miR-155 is required for adaptive and innate immunity and miR-17 cluster for adaptive differentiation of B cells and conventional T cells [101]. Furthermore, loss of miR-155 accelerated tumor growth, as well as simultaneously impaired activation of tumor-associated macrophages [102]. Taken together, miRNA-based immune therapy may provide a promising treatment for CRC patients in the near future.
So far, a complete and comprehensive knowledge with regards to the mechanistic role of miRNAs in extravasation is poorly understood. Recently, miR-214 was found to modulate extravasation in melanoma. In an in vitro assay, miR-214 overexpression in melanoma cells resulted in a two- to three-fold more efficient transendothelial migration. In vivo, CMRA-labeled miR-214 overexpressing or silenced cells were injected via tail vein in nude mice, causing a two- to three-fold increased extravasation for miR-214 overexpressing cells and approximately 50% reduction in extravasation ability in miR-214-silenced cells, compared with controls [103]. Similar results and mechanistic data have been observed for miR-31 in breast cancer [104].
• Metastatic colonization
Metastatic colonization is the final step during cancer progression. In CRC, circulating cancer cells predominantly affect and lodge in the liver, but rarely in the bone or brain. This is essentially due to direct portal seeding from the colon and rectum to the liver, as well as the unique features of hepatic blood flow that pivotally contribute to this metastatic pattern [105,106]. Additionally, several other studies have demonstrated a role for the chemokine receptor CXCR4, CCR6 in the dissemination of CRC to the liver [107–109]. miR-126 was found to be downregulated in metastatic colon cancer cells, and overexpression of miR-126 inhibited colon cancer cell viability and reduced tumor cell migration and invasion capacity through negative regulation of CXCR4 expression [35]. Based on the ‘seed and soil’ theory [110], cancer stem cells may provide the ‘seeds’ for CRC in the liver. The stem cell properties ensure proliferation of cancer cells or dormancy in a new environment. Ma et al. compared miRNA profiles of human embryonic stem cells and CRC cells and found that both types of cells shared the common endogenous miR-26b. miR-26b was downregulated in both ESC and Lovo cells. Over-expression of miR-26b significantly suppressed CRC cell growth and induced apoptosis in vitro and in vivo. Since miR-26b may regulate stem-cell properties of cancer cells, it has been hypothesized to play a central role in CRC metastasis [111]. Another group isolated populations of cancer stem cells that possessed CD133+/CD44+ and CD133−/CD44− surface phenotypes from SW1116 colon cancer cells. It was interesting to observe that compared with parental cells, cancer stem cells SW1116 showed differential expression of various miRNAs, including miR29a and miR29b. Recently, miR-146a was found to regulate asymmetrical cell division of cancer stem cells via a Snail-miR-146a-β-catenin feedback loop, causing disruption of stem cell pool balance and promoting tumor growth at the metastatic sites [112].
In general, colorectal carcinogenesis results from the cumulative effect of multiple genetic and epigenetic alterations. In past decades, accumulating evidence has shown that miRNAs are involved in carcinogenesis by targeting various onco- or tumor-suppressive genes including CRC. Figure 1 illustrates an overview of CRC pathogenesis, as it relates to the involvement of miRNAs. A better understanding of the biological impact of these key miRNAs on colorectal tumorigenesis, as well as their clinical impact will be crucial for our understanding and management of this malignancy in the future.
Clinical application of miRNAs
• Serum & fecal miRNAs serve as promising biomarkers for CRC diagnosis
As an ideal approach for CRC screening, the method must possess a very high degree of sensitivity and specificity for the early detection of cancer. Since fecal occult blood test (FOBT) is safe and acceptable to patients, it has been widely used as noninvasive screening tool for CRC. However, FOBT has relatively poor sensitivity and specificity for CRC diagnosis and FOBT screen only reduces 16–25% of relative risk associated with CRC-related mortality [113]. In an effort to identify better approaches for CRC screening, miRNAs have emerged as solid contenders for development as noninvasive biomarker substrates. Measurement of expression of miRNAs in circulating blood provides a novel and promising early diagnostic option for CRC screening. Several studies till date have now demonstrated that various human tumors possess unique miRNAs expression profiles in serum or plasma. Furthermore, some circulating miRNAs have been found to be packaged and protected in extracellular vesicles, called ‘exosomes’ or ‘microvesicles’. Therefore, high concentration of miRNAs can be discovered in highly stable, cell-free form in the peripheral blood [114,115].
Recently, several circulating miRNAs have been shown to be potential biomarkers for CRC diagnosis (Table 2). Adenoma is the precancerous lesion in CRC patients, and the early and accurate diagnosis for adenoma may reduce risk of CRC mortality. A panel of multiple plasma miRNAs (miR-532-3p, miR-331, miR-195, miR-17, miR-142-3p, miR-15b, miR-532 and miR-652) was found to distinguish polyps from controls with an AUC value of 0.87 [116]. A meta-analysis study compared diagnostic accuracy of currently published circulating miRNAs in discriminating CRC from controls. This study found miR-21 had a higher diagnostic accuracy for CRC diagnosis (AUC = 0.803) [117]. Our group also revealed serum miR-21 expression robustly distinguished adenoma (AUC = 0.81) and CRC patients (AUC = 0.92) from healthy control individuals [118]. Giraldez et al. used a genome-wide miRNA expression profiling assay to display 13 differential plasma miRNAs in 123 patients (63 with CRC and 60 with advanced adenomas) and 73 healthy individuals. These miRNAs were then used for validation in an independent cohort of 135 CRC patients. Six miRNAs, including miR-18a, miR-19a, miR-19b, miR-15b, miR-29a and miR335, were confirmed to be significantly upregulated in CRC patients compared with normal controls. The AUC value of each miRNAs’ ROC ranged from 0.8 to 0.7 [119]. Although circulating miRNAs are emerging as promising biomarkers for CRC, several technical factors may probably have impact on the accurate measurement of miRNAs including hemolysis, storage temperature and time [120]. Zanutto et al. found miR-378 level was not influenced by hemolysis levels of plasma samples [121]. However, Zanutto and our group found miR-21 level correlated with the degree of hemolysis in serum samples. Another factor that influences the results of circulating miRNAs is endogenous control. So far, there is no consensus control that can normalize the value of miRNA, making the results incomparable. Hu et al. found miR-1228 was the most stable endogenous control across eight cancer types and three independent control cohorts [122].
Table 2.
The clinical implication of miRNAs in colorectal cancer.
| miRNA | Specimen | Dysregulation | n | Remark | Ref. |
|---|---|---|---|---|---|
| Diagnostic biomarkers | |||||
| miR-21 | Serum | ↑ | 49 | CRC vs healthy control | [117] |
| miR-92a | Feces | ↑ | 246 | Higher sensitivity for advanced adenoma than minor polyps | [123] |
| miR-92a | Serum | ↑ | 697 | Early stage of colorectal cancer | [124] |
| miR-145 | Tissue | ↓ | 202 | Lymph node metastasis | [125] |
| miR-199a | Serum | ↑ | 116 | Associated with deep wall invasion | [126] |
| miR-221/18a | Feces | ↑ | 595 | CRC vs healthy control | [127] |
| miR-223/451 | Feces | ↑ | 45 | CRC vs healthy control | [128] |
| miR-378 | Serum | ↓ | 181 | CRC vs healthy control | [121] |
| miR-4487/1295b | Feces | ↓ | 56 | Decreased in early stage (I/II) | [129] |
| miR-21/let-7g | Serum | ↑ | 202 | Diagnosis of CRC, with relatively high sensitivity and specificity compared with CEA and CA-199 | [130] |
| miR-31/92a/181b/203 | ↓ | ||||
| miR-126/141/21 | Serum | ↑ | 224 | Early stage liver-metastatic CRC | [131] |
| miR-409-3p | Plasma | ↑ | 241 | Early stage of CRC | [132] |
| miR-7/93 | ↓ | ||||
| miR-18a/19a/19b/15b/29a/335 | Plasma | ↑ | 392 | Early diagnosis of CRC | [119] |
| Prognostic biomarkers | |||||
| miR-10b | Tissue | ↑ | 88 | Associated with poor prognosis | [133] |
| miR-10b | Serum | ↑ | 169 | Associated with distant metastasis | [134] |
| miR-17 | Tissue | ↑ | 96 | Associated with poor prognosis | [135] |
| miR-21 | Tissue | ↑ | 301,764 | Associated with poor prognosis | [136,137] |
| miR-21 | Serum | ↑ | 342 | Associated with poor prognosis | [118] |
| miR-31 | Tissue | ↑ | 1353 | Associated with BRAF mutation and poor prognosis | [138] |
| miR-29a | Tissue | ↓ | 169 | High risk of recurrence in stage II colon cancer | [139] |
| miR-96 | Tissue | ↓ | 80 | Associated with poor prognosis | [140] |
| miR-126 | Tissue | ↓ | 92 | Associated with poor OS | [82] |
| miR-148a | Tissue | ↓ | 273 | Prognostic/predictive biomarker for stage III and IV CRC | [141] |
| miR-148a | serum | ↓ | 30 | Predictive for the risk of recurrence of early stage colon cancer | [142] |
| miR-150 | Tissue | ↓ | 239 | Associated with poor prognosis | [143] |
| miR-200a | Tissue | ↓ | 328 | Associated with poor prognosis | [144] |
| miR-200c | Serum | ↓ | 446 | Prediction for metastasis | [145] |
| miR-145/31 | Tissue | ↑ | 1141 | Associated with advanced CRC | [146] |
| miR-200b/215/451a | ↓ | ||||
| miR-532-3p/331/195/17/142-3p/15b/532/652 | Serum | ↑ | 128 | Distinguished polyps from controls | [116] |
| miR-431/15b/139-3p | ↑ | Distinguished stage IV CRC from controls | |||
| miR-21/20a/miR-103a-/106b miR-143/215 |
Tissue | ↑ ↓ |
638 | Associated with poor prognosis | [147] |
| Predictive biomarkers for chemotherapy | |||||
| miR-17 | Tissue | ↑ | 425 | Poor response to chemotherapy | [148] |
| miR-31 | Tissue | ↑ | 102,87 | Anti-EGFR therapy, associated with shorter PFS | [149,150] |
| miR-126 | Tissue | ↓ | 89 | Associated with responding to XELOX | [151] |
| miR-200a/200c/141/429 | Tissue | ↓ | 127 | Higher level associated with longer OS and DFS in stage II patients treated with fluoropyrimidines | [152] |
| miR-99a/Let-7c/miR-125b | Tissue | ↓ | 183 | Selection of patients with KRAS wild-type mCRC | [153] |
CRC: Colorectal cancer; DFS: Disease-free survival; mCRC: Metastatic colorectal cancer; OS: Overall survival; PFS: Progression-free survival.
Exosomal miRNAs have recently been attracting a lot of interest as potential biomarkers for CRC diagnosis. Exosomes derived from CRC may carry tumor-specific genetic materials that transmit signaling between tumor and stromal cells in the tumor microenvironment. Therefore, serum exosomal miRNAs levels are probably more robust for CRC diagnosis, compared with free circulating miRNAs. A study of 88 primary CRC patients and 11 healthy controls was performed to identify candidate exosomal miRNAs. Seven miRNAs were found upregulated in exosome-enriched fractions of serum samples; however, the level of these miRNA was not dependent on tumor stage. Two of these miRNAs, miR-23 and miR-1246, showed higher sensitivity for detection of stage I CRCs (95 and 90%), compared with CEA and CA-199 (15,10%), suggesting their promising potential as substrates for the development of noninvasive CRC diagnostics [97].
To date, much attention has been focused on miRNAs in tissue and circulation, and to a lesser extent in stool (Table 2). We recently developed a new method called direct miRNA analysis to analyze the fecal miRNA levels. We also reported that fecal miRNAs can be effectively extracted and analyzed from clinical specimens collected in FOBT kits. Furthermore, we noted that miR-21 and miR-106a had a higher expression in patient with adenomas and CRC than in healthy controls [154]. Several other studies have validated these findings and have also reported that miRNAs can be efficiently extracted from stool samples and can be possibly exploited for distinguishing healthy individuals from patients with colorectal adenomas and cancers [123,155–156]. Wu et al. reported that stool miR-92a level, but not miR-21, was increased in patients with adenomatous polyps compared with healthy subjects. Moreover, fecal miR-92a expression showed higher sensitivity for distal compared with proximal CRC, and possessed higher sensitivity for advanced adenomas than minor polyps. Furthermore, decreased levels of fecal miR-92a expression levels were noted after removal of the tumor or advanced adenomas, supporting the specificity of these miRNAs for the presence of a colorectal polyp or cancer [123]. Recently, this research group analyzed 424 stool samples from patients with colorectal adenomas and cancers, inflammatory bowel disease and healthy controls. These assays were performed in both pre- and postsurgery fecal specimens, following up detection after removal of lesions. These results demonstrate the feasibility of fecal-based miR-135b as a potential biomarker for both advanced adenoma and CRC [157].
• miRNA biomarkers for determining prognosis & predicting treatment response in CRC patients
To date, management of CRC is dependent on clinico-pathologic features including TNM stage, histology and tumor margin involvement [158]. Although pathological tumor staging is an important consideration for treatment selection, treatment response among patients with same stage has varied significantly. One of the tragic realities is that the chemotherapeutic drugs that are currently used to treat cancer patients are highly toxic, and their effectiveness varies unpredictably from patient to patient – mainly due to subtle differences in genomic makeup of each patient and the molecular characteristics of their tumor. In particular, development of resistance to chemotherapeutic drugs presents a major challenge in terms of poor response and survival – and remains a critical hurdle while treating patients with advanced CRC. Many of these patients eventually develop resistance to such treatments, leading to tumor relapse or recurrence. In view of this, there is an urgent clinical need for development of tools or (bio)markers that can help stratify which patients will respond to specific drugs or not, so that others can be spared from being needlessly exposed to such toxic and expensive therapeutic drugs, and triaged for alternative treatment options. Accumulation of studies supports the use of molecular biomarkers to refine estimates of prognosis and tailoring of adjuvant and neo-adjuvant treatment to individual patients [159]. The unique miRNA expression patterns in CRC have the potential to be used as prognostic and predictive factors in determining and predicting their clinical outcomes.
Although this field is relatively young, there are several studies that suggest the key role of miRNAs in determining prognosis and predicting responses to chemotherapeutic treatments. EMT-related miRNAs play an important role in CRC progression and metastasis. As mentioned previously, we found that miR-200c modulated EMT in human CRC metastasis [83]. We have performed another three-phase study in which we analyzed 446 colorectal specimens and found serum miR-200c serves as a good prognostic biomarker for CRC. High serum miR-200c was found in stage IV patients compared with its levels in stage I–III CRC patients. Furthermore, serum miR-200c emerged as an independent predictor for lymph node metastasis and tumor recurrence [145]. Diaz et al. determined the expression of miR-200c in 127 surgically-resected patients with stage I–III CRC. The higher expression of miR-200c correlated with longer overall survival. Furthermore, in the subgroup of patients treated with fluoropyrimidines, the higher levels of miR-200c showed longer overall survival, as well as improved disease free survival, suggesting miR-200c as a potential predictive biomarker for chemotherapeutic response [152].
Likewise, miR-21 has been shown to be a robust prognostic biomarker for CRC. A meta-analysis revealed that higher expression of miR-21 predicts poor overall survival [160]. The pooled hazard ratios were 1.76 (95% CI: 1.34– 2.32) in this report. Another clinically significant biomarker is miR-625, which was found to be decreased, indicating unfavorable prognosis for CRC patients [161]. Table 2 summarizes some of the recently published miRNA biomarkers that are associated with clinical outcomes in CRC patients. As of now, several miRNA biomarkers have been developed for the predicting therapeutic response in CRC patients. For instance, miRNAs including miR-222 [162], miR-297 [163], miR-19b [164] and miR-224 [165] have been demonstrated to confer CRC cells with multidrug resistant ability in cell culture studies. miR-126 has been known to be an angiogenesis-specific miRNA, and the expression levels of miR-126 were noted to be higher in metastatic CRC patients that responded to capecitabine and oxaliplatin (XELOX) treatment, compared with those that failed to show any response. Furthermore, progression-free survival was 11.5 months in patients with higher expression of miR-126, compared with 6 months in those with lower expression of this noncoding RNA [151]. Other studies have showed that increased levels of miR-10b and miR-192/215 predict response to 5-FU-based chemotherapy while low levels of miR-19a indicate resistance to FOLOFOX treatment. Finally, there are data suggesting that miR-200a, miR-200c, miR-141 and miR-429 expression levels may identify CRC patients who stand to benefit from fluoropyrimidine-based treatments and suffering from CRC [166].
• miRNAs as potential therapeutic targets in CRC
A wealth of published data has shown that blocking the expression of specific onco-miRNAs or restoring the levels of suppressive-miRNAs in CRC cells can result in inhibiting tumor growth, angiogenesis and metastasis, as well as enhancement of response to various chemo and radiation therapies – providing a rational for the development of miRNA-based targeted CRC therapeutic strategies. However, the major hurdle in this arena is safe delivery of these therapies to the target tissues with less/ minimal side effects to the surrounding normal tissues. Several viral and nonviral strategies have been reported in previous studies. A novel RNA delivery system, immune-liposomes, is currently being developed for cancer treatment. Immunoliposomes are coupled with antibodies which allow for more efficient and focused targeting of tumor tissues by binding to tumor-specific receptors. The distribution of immune-liposomes in tumor-bearing animals has shown their higher concentration in tumor tissues compared with significantly lower concentrations in normal tissues [167]. Sonoporation is another delivery technology used for gene therapy. During this process, the delivery of the intended therapy is facilitated via creation of temporary pores in the cell membranes induced by ultrasonic waves [168]. The first clinical trial utilizing such a sonoporation and microbubbles approach for local therapeutic delivery was recently published by Kotopoulis and colleagues [169]. Another challenge with miRNA-based therapy is ‘off target’ effects. Since miRNAs target plethora of mRNAs in context of signaling networks, it is possible that undesired toxicity and even immune reactions may manifest, making it more difficult to achieve desired results. Furthermore, rapid degradation and blocked uptake of miRNAs by nucleases or other unknown cellular mechanisms can also potentially affect the clinical impact of such an approach. To be on the safe side, lowest optimal concentrations of miRNAs along with effective target delivery systems need to be developed and validated prior to considering such tools for safe and efficacious treatment offering for patients with colorectal and other cancers.
Thus far, no clinical trials for miRNA replacement therapy in patients with CRC have been reported. However, in vivo studies have shown that miRNAs are promising therapeutic targets for CRC treatment. Liu et al. delivered lentiviral-based vectors expressing miR-221 and miR-222 sponges in an animal model of DSS-induced colitis. Treatment with miR-221 and miR-222 significantly decreased tumor number and size. Furthermore, markedly reduced advanced adenomas were found in DSS-mice treated with miR-221/222 [17]. In another study, investigators developed a robust nonviral delivery method for in vivo miRNA administration. After systemic or local application of polyethylenimine (PEI)-miR-145/-33a complex, the mouse xenograft tumors were significantly inhibited [170]. Another promising therapy is antiangiogenesis-based miRNA replacement. A special modified, dicetyl phosphate-tetraethylenepentamine-based polycation liposomes (TEPA-PCL), were used as delivery system of miR-92a. After transfection of human umbilical vein endothelial cells (HUVECs) with miR-92a-TEPA-PCL complex, the cytoplasm of HUVECs was filled with miR-29a. Furthermore, HUVECs showed impaired capability of forming capillary tubes, suggesting miR-29a-TEPA-PCL transfection system is a candidate antiangiogenesis treatment for CRC [171].
Conclusion & future perspective
Dysregulation of miRNAs is now being regarded as a critical step for the pathogenesis of CRC. There are convincing data that altered expression of miRNAs facilitates tumor initiation, proliferation and angiogenesis, confers resistance to chemotherapy and enhances metastasis in colon cancer cells through interaction with intracellular signaling networks. From a clinical perspective, there is no doubt that miRNAs will be important diagnostic and prognostic biomarkers for CRC (Table 2 & Figure 2). Given the ever-expanding number of miRNAs, it is a challenge to accurately determine which of these miRNAs can be used for the earlier diagnosis, risk assessment, prognosis prediction and determining response to chemotherapeutic treatments in patients suffering with CRC. In addition, there is a growing level of interest for miRNAs as potential therapeutic targets. miR-21, for example, not only serves as a promising biomarker for CRC but also as potential therapeutic target since miR-21 is involved in both cancer development and immune regulation. Although the safety and efficacy of delivering miRNA therapeutics effectively to the target tumor tissues in miRNA-based therapeutic approaches, there is undeniable body of data already gathered indicating that miRNAs do offer a promising substrate for the development of biomarkers aimed for earlier diagnosis, determining prognosis and predicting response to chemotherapeutic treatments in patients suffering from CRC.
Figure 2. The clinical application of miRNAs in colorectal cancer.
Colorectal cancer-related miRNAs can be extracted from tissue, blood or stools samples and analyzed by real-time PCR, microarray or next-generation sequence. These circulating miRNAs, tissue or fecal-based miRNAs provide useful tools of diagnostic and prognostic information.
EXECUTIVE SUMMARY.
miRNA expression is frequently & consistently altered in colorectal cancer
Global miRNA expression patterns are altered in colorectal cancer (CRC), and existing data suggest that these alterations can occur in a cancer-specific manner.
The functional role of miRNAs in the initiation of CRC development has been studied, and there is a growing interest in associating miRNA-mediated transcriptional repression of downstream target genes.
Several miRNAs such as miR-143 and miR-31 contribute to the initiation of CRC.
miRNA expression profiles are altered during progression in the normal adenoma carcinoma sequence.
MMR status affects miRNA expression pattern in the transition from normal epithelia to colonic neoplasia.
miRNAs affect the invasion & metastasis in CRC
Specific miRNAs and families promote tumor angiogenesis by targeting VEGF signaling pathways.
miR-200 clusters mediate epithelial–mesenchymal transition in CRC.
Several miRNAs contribute to the development of CRC during intravasation, circulation extravasation and metastatic colonization.
Clinical application of miRNAs
Serum and fecal miRNAs serve as promising biomarkers for CRC diagnosis.
miRNA biomarkers are being developed for determining prognosis and predicting treatment response in CRC patients.
Several miRNAs have been identified as potential therapeutic targets for CRC.
Footnotes
For reprint orders, please contact: reprints@futuremedicine.com
Financial & competing interests disclosure
This work was supported by grants R01 CA72851, CA181572, CA184792 and U01 CA187956 from the National Cancer Institute, NIH, a pilot grant from Charles A Sammons Cancer Center, and funds from the Baylor Research Institute to A Goel. This work was also supported by grants from the National Natural Science Foundation of China (no. 81372615 and no. 81201884), Shanghai Science and Technology Development Fund (no. 12140902300 and no. 12410707400), Shanghai Health System Outstanding Young Talent Training Plan (no. XYQ2013118) and Shanghai Tenth people’s Hospital CLIMB project for outstanding Young Clinical Investigators (W Weng). The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.
No writing assistance was utilized in the production of this manuscript.
References
Papers of special note have been highlighted as:
• of interest;
•• of considerable interest
- 1.Vogelstein B, Fearon ER, Hamilton SR, et al. Genetic alterations during colorectal-tumor development. N Engl J Med. 1988;319(9):525–532. doi: 10.1056/NEJM198809013190901. [DOI] [PubMed] [Google Scholar]
- 2.Harris TJ, Mccormick F. The molecular pathology of cancer. Nat Rev Clin Oncol. 2010;7(5):251–265. doi: 10.1038/nrclinonc.2010.41. [DOI] [PubMed] [Google Scholar]
- 3.Lu J, Getz G, Miska EA, et al. MicroRNA expression profiles classify human cancers. Nature. 2005;435(7043):834–838. doi: 10.1038/nature03702. [DOI] [PubMed] [Google Scholar]
- 4.Schetter AJ, Leung SY, Sohn JJ, et al. MicroRNA expression profiles associated with prognosis and therapeutic outcome in colon adenocarcinoma. JAMA. 2008;299(4):425–436. doi: 10.1001/jama.299.4.425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.miRNA Body Map. www.mirnabodymap.org.
- 6.HMDD version 2. www.cuilab.cn/hmdd.
- 7.Fang L, Li H, Wang L, et al. MicroRNA-17-5p promotes chemotherapeutic drug resistance and tumour metastasis of colorectal cancer by repressing pten expression. Oncotarget. 2014;5(10):2974–2987. doi: 10.18632/oncotarget.1614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Yang MH, Yu J, Chen N, et al. Elevated microRNA-31 expression regulates colorectal cancer progression by repressing its target gene satb2. PLoS ONE. 2013;8(12):e85353. doi: 10.1371/journal.pone.0085353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Sun D, Yu F, Ma Y, et al. MicroRNA-31 activates the ras pathway and functions as an oncogenic microRNA in human colorectal cancer by repressing RAS p21 GTPase activating protein 1 (RASA1) J Biol Chem. 2013;288(13):9508–9518. doi: 10.1074/jbc.M112.367763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Xu RS, Wu XD, Zhang SQ, et al. The tumor suppressor gene RHOBTB1 is a novel target of miR-31 in human colon cancer. Int J Oncol. 2013;42(2):676–682. doi: 10.3892/ijo.2012.1746. [DOI] [PubMed] [Google Scholar]
- 11.Ke TW, Wei PL, Yeh KT, Chen WT, Cheng YW. miR-92a promotes cell metastasis of colorectal cancer through pten-mediated PI3K/AKT pathway. Ann Surg Oncol. 2014 doi: 10.1245/s10434-014-4305-2. Epub ahead of print. [DOI] [PubMed] [Google Scholar]
- 12.Gao F, Wang W. MicroRNA-96 promotes the proliferation of colorectal cancer cells and targets tumor protein p53 inducible nuclear protein 1, forkhead box protein o1 (FOXO1) and FOXO3a. Mol Med Rep. 2015;11(2):1200–1206. doi: 10.3892/mmr.2014.2854. [DOI] [PubMed] [Google Scholar]
- 13.Chen HY, Lin YM, Chung HC, et al. miR-103/107 promote metastasis of colorectal cancer by targeting the metastasis suppressors dapk and klf4. Cancer Res. 2012;72(14):3631–3641. doi: 10.1158/0008-5472.CAN-12-0667. [DOI] [PubMed] [Google Scholar]
- 14.Zhao Y, Miao G, Li Y, et al. MicroRNA-130b suppresses migration and invasion of colorectal cancer cells through downregulation of integrin beta1 [corrected] PLoS ONE. 2014;9(2):e87938. doi: 10.1371/journal.pone.0087938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Zhang L, Pickard K, Jenei V, et al. miR-153 supports colorectal cancer progression via pleiotropic effects that enhance invasion and chemotherapeutic resistance. Cancer Res. 2013;73(21):6435–6447. doi: 10.1158/0008-5472.CAN-12-3308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Ji D, Chen Z, Li M, et al. MicroRNA-181a promotes tumor growth and liver metastasis in colorectal cancer by targeting the tumor suppressor wif-1. Mol Cancer. 2014;13:86. doi: 10.1186/1476-4598-13-86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Liu S, Sun X, Wang M, et al. A microRNA 221- and 222-mediated feedback loop maintains constitutive activation of nfkappab and stat3 in colorectal cancer cells. Gastroenterology. 2014;147(4):847–859. e811. doi: 10.1053/j.gastro.2014.06.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Liao WT, Li TT, Wang ZG, et al. MicroRNA-224 promotes cell proliferation and tumor growth in human colorectal cancer by repressing PHLPP1 and PHLPP2. Clin Cancer Res. 2013;19(17):4662–4672. doi: 10.1158/1078-0432.CCR-13-0244. [DOI] [PubMed] [Google Scholar]
- 19.Fang Y, Sun B, Xiang J, Chen Z. miR-301a promotes colorectal cancer cell growth and invasion by directly targeting socs6. Cell Physiol Biochem. 2015;35(1):227–236. doi: 10.1159/000369690. [DOI] [PubMed] [Google Scholar]
- 20.Christensen LL, Tobiasen H, Holm A, et al. miRNA-362-3p induces cell cycle arrest through targeting of E2F1, USF2 and PTPN1 and is associated with recurrence of colorectal cancer. Int J Cancer. 2013;133(1):67–78. doi: 10.1002/ijc.28010. [DOI] [PubMed] [Google Scholar]
- 21.Li J, Du L, Yang Y, et al. miR-429 is an independent prognostic factor in colorectal cancer and exerts its anti-apoptotic function by targeting sox2. Cancer letters. 2013;329(1):84–90. doi: 10.1016/j.canlet.2012.10.019. [DOI] [PubMed] [Google Scholar]
- 22.Ji S, Ye G, Zhang J, et al. miR-574-5p negatively regulates QKI6/7 to impact beta-catenin/wnt signalling and the development of colorectal cancer. Gut. 2013;62(5):716–726. doi: 10.1136/gutjnl-2011-301083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Lei SL, Zhao H, Yao HL, et al. Regulatory roles of microRNA-708 and microRNA-31 in proliferation, apoptosis and invasion of colorectal cancer cells. Oncol Lett. 2014;8(4):1768–1774. doi: 10.3892/ol.2014.2328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Han HB, Gu J, Zuo HJ, et al. Let-7c functions as a metastasis suppressor by targeting MMP11 and PBX3 in colorectal cancer. J Pathol. 2012;226(3):544–555. doi: 10.1002/path.3014. [DOI] [PubMed] [Google Scholar]
- 25.Suto T, Yokobori T, Yajima R, et al. MicroRNA-7 expression in colorectal cancer is associated with poor prognosis and regulates cetuximab sensitivity via EGFR regulation. Carcinogenesis. 2014;36(3):338–345. doi: 10.1093/carcin/bgu242. [DOI] [PubMed] [Google Scholar]
- 26.Zhang N, Li X, Wu CW, et al. MicroRNA-7 is a novel inhibitor of YY1 contributing to colorectal tumorigenesis. Oncogene. 2013;32(42):5078–5088. doi: 10.1038/onc.2012.526. [DOI] [PubMed] [Google Scholar]
- 27.Humphreys KJ, Mckinnon RA, Michael MZ. miR-18a inhibits CDC42 and plays a tumour suppressor role in colorectal cancer cells. PLoS ONE. 2014;9(11):e112288. doi: 10.1371/journal.pone.0112288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Bao Y, Chen Z, Guo Y, et al. Tumor suppressor microRNA-27a in colorectal carcinogenesis and progression by targeting SGPP1 and SMAD2. PLoS ONE. 2014;9(8):e105991. doi: 10.1371/journal.pone.0105991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Ye J, Wu X, Wu D, et al. miRNA-27b targets vascular endothelial growth factor c to inhibit tumor progression and angiogenesis in colorectal cancer. PLoS ONE. 2013;8(4):e60687. doi: 10.1371/journal.pone.0060687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Zhang JX, Mai SJ, Huang XX, et al. miR-29c mediates epithelial-to-mesenchymal transition in human colorectal carcinoma metastasis via PTP4A and GNA13 regulation of beta-catenin signaling. Ann Oncol. 2014;25(11):2196–2204. doi: 10.1093/annonc/mdu439. [DOI] [PubMed] [Google Scholar]
- 31.Zhang Q, Tang Q, Qin D, et al. Role of miR-30a targeting insulin receptor substrate 2 in colorectal tumorigenesis. Mol Cell Biol. 2015;35(6):988–1000. doi: 10.1128/MCB.01242-14. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 32.Liao WT, Ye YP, Zhang NJ, et al. MicroRNA-30b functions as a tumour suppressor in human colorectal cancer by targeting KRAS, PIK3CD and BCL2. J Pathol. 2014;232(4):415–427. doi: 10.1002/path.4309. [DOI] [PubMed] [Google Scholar]
- 33.Peng H, Luo J, Hao H, et al. MicroRNA-100 regulates SW620 colorectal cancer cell proliferation and invasion by targeting RAP1B. Oncol Rep. 2014;31(5):2055–2062. doi: 10.3892/or.2014.3075. [DOI] [PubMed] [Google Scholar]
- 34.Zhang Y, Zheng L, Huang J, et al. miR-124 radiosensitizes human colorectal cancer cells by targeting PRRX1. PLoS ONE. 2014;9(4):e93917. doi: 10.1371/journal.pone.0093917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Li Z, Li N, Wu M, Li X, Luo Z, Wang X. Expression of miR-126 suppresses migration and invasion of colon cancer cells by targeting CXCR4. Mol Cell Biochem. 2013;381(1–2):233–242. doi: 10.1007/s11010-013-1707-6. [DOI] [PubMed] [Google Scholar]
- 36.Liu Y, Zhou Y, Feng X, et al. MicroRNA-126 functions as a tumor suppressor in colorectal cancer cells by targeting CXCR4 via the AKT and ERK1/2 signaling pathways. Int J Oncol. 2014;44(1):203–210. doi: 10.3892/ijo.2013.2168. [DOI] [PubMed] [Google Scholar]
- 37.Zhou Y, Feng X, Liu YL, et al. Down-regulation of miR-126 is associated with colorectal cancer cells proliferation, migration and invasion by targeting IRS-1 via the AKT and ERK1/2 signaling pathways. PLoS ONE. 2013;8(11):e81203. doi: 10.1371/journal.pone.0081203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Zhang Y, Wang X, Xu B, et al. Epigenetic silencing of miR-126 contributes to tumor invasion and angiogenesis in colorectal cancer. Oncol Rep. 2013;30(4):1976–1984. doi: 10.3892/or.2013.2633. [DOI] [PubMed] [Google Scholar]
- 39.Li N, Tang A, Huang S, et al. miR-126 suppresses colon cancer cell proliferation and invasion via inhibiting rhoa/rock signaling pathway. Mol Cell Biochem. 2013;380(1–2):107–119. doi: 10.1007/s11010-013-1664-0. [DOI] [PubMed] [Google Scholar]
- 40.Duan FT, Qian F, Fang K, Lin KY, Wang WT, Chen YQ. miR-133b, a muscle-specific microRNA, is a novel prognostic marker that participates in the progression of human colorectal cancer via regulation of CXCR4 expression. Mol Cancer. 2013;12:164. doi: 10.1186/1476-4598-12-164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Song M, Yin Y, Zhang J, et al. miR-139-5p inhibits migration and invasion of colorectal cancer by downregulating amfr and NOTCH1. Protein Cell. 2014;5(11):851–861. doi: 10.1007/s13238-014-0093-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Zhang L, Dong Y, Zhu N, et al. MicroRNA-139-5p exerts tumor suppressor function by targeting NOTCH1 in colorectal cancer. Mol Cancer. 2014;13:124. doi: 10.1186/1476-4598-13-124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Su J, Liang H, Yao W, et al. miR-143 and miR-145 regulate IGF1R to suppress cell proliferation in colorectal cancer. PLoS ONE. 2014;9(12):e114420. doi: 10.1371/journal.pone.0114420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Qian X, Yu J, Yin Y, et al. MicroRNA-143 inhibits tumor growth and angiogenesis and sensitizes chemosensitivity to oxaliplatin in colorectal cancers. Cell Cycle. 2013;12(9):1385–1394. doi: 10.4161/cc.24477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Guo H, Chen Y, Hu X, Qian G, Ge S, Zhang J. The regulation of toll-like receptor 2 by miR-143 suppresses the invasion and migration of a subset of human colorectal carcinoma cells. Mol Cancer. 2013;12:77. doi: 10.1186/1476-4598-12-77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Gregersen LH, Jacobsen A, Frankel LB, Wen J, Krogh A, Lund AH. MicroRNA-143 down-regulates hexokinase 2 in colon cancer cells. BMC Cancer. 2012;12:232. doi: 10.1186/1471-2407-12-232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Zhang Y, Wang Z, Chen M, et al. MicroRNA-143 targets MACC1 to inhibit cell invasion and migration in colorectal cancer. Mol Cancer. 2012;11:23. doi: 10.1186/1476-4598-11-23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Iwaya T, Yokobori T, Nishida N, et al. Downregulation of miR-144 is associated with colorectal cancer progression via activation of mTOR signaling pathway. Carcinogenesis. 2012;33(12):2391–2397. doi: 10.1093/carcin/bgs288. [DOI] [PubMed] [Google Scholar]
- 49.Feng Y, Zhu J, Ou C, et al. MicroRNA-145 inhibits tumour growth and metastasis in colorectal cancer by targeting fascin-1. Br J Cancer. 2014;110(9):2300–2309. doi: 10.1038/bjc.2014.122. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 50.Yamada N, Noguchi S, Mori T, Naoe T, Maruo K, Akao Y. Tumor-suppressive microRNA-145 targets catenin DELTA-1 to regulate wnt/beta-catenin signaling in human colon cancer cells. Cancer Lett. 2013;335(2):332–342. doi: 10.1016/j.canlet.2013.02.060. [DOI] [PubMed] [Google Scholar]
- 51.Yin Y, Yan ZP, Lu NN, et al. Downregulation of miR-145 associated with cancer progression and vegf transcriptional activation by targeting N-RAS and IRS1. Biochim Biophys Acta. 2013;1829(2):239–247. doi: 10.1016/j.bbagrm.2012.11.006. [DOI] [PubMed] [Google Scholar]
- 52.Astarci E, Erson-Bensan AE, Banerjee S. Matrix metalloprotease 16 expression is downregulated by microRNA-146a in spontaneously differentiating caco-2 cells. Dev Growth Differ. 2012;54(2):216–226. doi: 10.1111/j.1440-169X.2011.01324.x. [DOI] [PubMed] [Google Scholar]
- 53.Zhang Z, Liu X, Feng B, et al. STIM1, a direct target of microRNA-185, promotes tumor metastasis and is associated with poor prognosis in colorectal cancer. Oncogene. 2014 doi: 10.1038/onc.2014.404. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Hu Y, Liu J, Jiang B, et al. miR-199a-5p loss up-regulated DDR1 aggravated colorectal cancer by activating epithelial-tomesenchymal transition related signaling. Dig Dis Sci. 2014;59(9):2163–2172. doi: 10.1007/s10620-014-3136-0. [DOI] [PubMed] [Google Scholar]
- 55.Wang Q, Huang Z, Guo W, et al. MicroRNA-202-3p inhibits cell proliferation by targeting adp-ribosylation factor-like 5a in human colorectal carcinoma. Clin Cancer Res. 2014;20(5):1146–1157. doi: 10.1158/1078-0432.CCR-13-1023. [DOI] [PubMed] [Google Scholar]
- 56.Li T, Gao F, Zhang XP. miR-203 enhances chemosensitivity to 5-fluorouracil by targeting thymidylate synthase in colorectal cancer. Oncol Rep. 2015;33(2):607–614. doi: 10.3892/or.2014.3646. [DOI] [PubMed] [Google Scholar]
- 57.Meng X, Wu J, Pan C, et al. Genetic and epigenetic down-regulation of microRNA-212 promotes colorectal tumor metastasis via dysregulation of MnSOD. Gastroenterology. 2013;145(2):426–436. e421–e426. doi: 10.1053/j.gastro.2013.04.004. [DOI] [PubMed] [Google Scholar]
- 58.Wang Y, Tang Q, Li M, Jiang S, Wang X. MicroRNA-375 inhibits colorectal cancer growth by targeting pik3ca. Biochem Biophys Res Commun. 2014;444(2):199–204. doi: 10.1016/j.bbrc.2014.01.028. [DOI] [PubMed] [Google Scholar]
- 59.Zhang GJ, Zhou H, Xiao HX, Li Y, Zhou T. miR-378 is an independent prognostic factor and inhibits cell growth and invasion in colorectal cancer. BMC Cancer. 2014;14:109. doi: 10.1186/1471-2407-14-109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Zhang C, Liu J, Wang X, et al. MicroRNA-339-5p inhibits colorectal tumorigenesis through regulation of the MDM2/p53 signaling. Oncotarget. 2014;5(19):9106–9117. doi: 10.18632/oncotarget.2379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Chai J, Wang S, Han D, Dong W, Xie C, Guo H. MicroRNA-455 inhibits proliferation and invasion of colorectal cancer by targeting raf proto-oncogene serine/threonine-protein kinase. Tumour Biol. 2014;36(2):1313–1321. doi: 10.1007/s13277-014-2766-3. [DOI] [PubMed] [Google Scholar]
- 62.Guo ST, Jiang CC, Wang GP, et al. MicroRNA-497 targets insulin-like growth factor 1 receptor and has a tumour suppressive role in human colorectal cancer. doi: 10.1038/onc.2012.214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Schee K, Lorenz S, Worren MM, et al. Deep sequencing the microRNA transcriptome in colorectal cancer. PLoS ONE. 2013;8(6):e66165. doi: 10.1371/journal.pone.0066165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Ak S, Tunca B, Tezcan G, et al. MicroRNA expression patterns of tumors in early-onset colorectal cancer patients. J Surg Res. 2014;191(1):113–122. doi: 10.1016/j.jss.2014.03.057. [DOI] [PubMed] [Google Scholar]
- 65.Kamatani A, Nakagawa Y, Akao Y, et al. Downregulation of anti-oncomirs miR-143/145 cluster occurs before APC gene aberration in the development of colorectal tumors. Med Mol Morphol. 2013;46(3):166–171. doi: 10.1007/s00795-013-0020-5. [DOI] [PubMed] [Google Scholar]
- 66.Josse C, Bouznad N, Geurts P, et al. Identification of a microRNA landscape targeting the PI3K/AKT signaling pathway in inflammation-induced colorectal carcinogenesis. Am J Physiol Gastrointest Liver Physiol. 2014;306(3):G229–G243. doi: 10.1152/ajpgi.00484.2012. [DOI] [PubMed] [Google Scholar]
- 67.Kent OA, Fox-Talbot K, Halushka MK. Rreb1 repressed miR-143/145 modulates KRAS signaling through downregulation of multiple targets. Oncogene. 2013;32(20):2576–2585. doi: 10.1038/onc.2012.266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Pagliuca A, Valvo C, Fabrizi E, et al. Analysis of the combined action of miR-143 and miR-145 on oncogenic pathways in colorectal cancer cells reveals a coordinate program of gene repression. Oncogene. 2013;32(40):4806–4813. doi: 10.1038/onc.2012.495. [DOI] [PubMed] [Google Scholar]
- 69.Ng EK, Tsang WP, Ng SS, et al. MicroRNA-143 targets DNA methyltransferases 3a in colorectal cancer. Br J Cancer. 2009;101(4):699–706. doi: 10.1038/sj.bjc.6605195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Necela BM, Carr JM, Asmann YW, Thompson EA. Differential expression of microRNAs in tumors from chronically inflamed or genetic (APC(min/+)) models of colon cancer. PLoS ONE. 2011;6(4):e18501. doi: 10.1371/journal.pone.0018501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71•.Bartley AN, Yao H, Barkoh BA, et al. Complex patterns of altered microRNA expression during the adenoma-adenocarcinoma sequence for microsatellite-stable colorectal cancer. Clin Cancer Res. 2011;17(23):7283–7293. doi: 10.1158/1078-0432.CCR-11-1452. miRNAs expression pattern was altered during the adenoma–adenocarcinoma sequence. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Boland CR, Goel A. Microsatellite instability in colorectal cancer. Gastroenterology. 2010;138(6):2073–2087. e2073. doi: 10.1053/j.gastro.2009.12.064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Oberg AL, French AJ, Sarver AL, et al. miRNA expression in colon polyps provides evidence for a multihit model of colon cancer. PLoS ONE. 2011;6(6):e20465. doi: 10.1371/journal.pone.0020465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Balaguer F, Moreira L, Lozano JJ, et al. Colorectal cancers with microsatellite instability display unique miRNA profiles. Clin Cancer Res. 2011;17(19):6239–6249. doi: 10.1158/1078-0432.CCR-11-1424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Hicklin DJ, Ellis LM. Role of the vascular endothelial growth factor pathway in tumor growth and angiogenesis. J Clin Oncol. 2005;23(5):1011–1027. doi: 10.1200/JCO.2005.06.081. [DOI] [PubMed] [Google Scholar]
- 76.Chen H, Li L, Wang S, et al. Reduced miR-126 expression facilitates angiogenesis of gastric cancer through its regulation on vegf-a. Oncotarget. 2014;5(23):11873–11885. doi: 10.18632/oncotarget.2662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Hua Z, Lv Q, Ye W, et al. miRNA-directed regulation of VEGF and other angiogenic factors under hypoxia. PLoS ONE. 2006;1:e116. doi: 10.1371/journal.pone.0000116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Fish JE, Santoro MM, Morton SU, et al. miR-126 regulates angiogenic signaling and vascular integrity. Dev Cell. 2008;15(2):272–284. doi: 10.1016/j.devcel.2008.07.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Wang S, Aurora AB, Johnson BA, et al. The endothelial-specific microRNA miR-126 governs vascular integrity and angiogenesis. Dev Cell. 2008;15(2):261–271. doi: 10.1016/j.devcel.2008.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Nicoli S, Standley C, Walker P, Hurlstone A, Fogarty KE, Lawson ND. MicroRNA-mediated integration of haemodynamics and vegf signalling during angiogenesis. Nature. 2010;464(7292):1196–1200. doi: 10.1038/nature08889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Guo C, Sah JF, Beard L, Willson JK, Markowitz SD, Guda K. The noncoding rna, miR-126, suppresses the growth of neoplastic cells by targeting phosphatidylinositol 3-kinase signaling and is frequently lost in colon cancers. Genes Chromosomes Cancer. 2008;47(11):939–946. doi: 10.1002/gcc.20596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Liu Y, Zhou Y, Feng X, et al. Low expression of microRNA-126 is associated with poor prognosis in colorectal cancer. Genes Chromosomes Cancer. 2014;53(4):358–365. doi: 10.1002/gcc.22146. [DOI] [PubMed] [Google Scholar]
- 83••.Hur K, Toiyama Y, Takahashi M, et al. MicroRNA-200c modulates epithelial-to-mesenchymal transition (EMT) in human colorectal cancer metastasis. Gut. 2013;62(9):1315–1326. doi: 10.1136/gutjnl-2011-301846. miR-200c modulate epithelial-to-mesenchymal transition in colorectal cancer (CRC) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Paterson EL, Kazenwadel J, Bert AG, Khew-Goodall Y, Ruszkiewicz A, Goodall GJ. Down-regulation of the miRNA-200 family at the invasive front of colorectal cancers with degraded basement membrane indicates emt is involved in cancer progression. Neoplasia. 2013;15(2):180–191. doi: 10.1593/neo.121828. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Tian Y, Pan Q, Shang Y, et al. MicroRNA-200 (miR-200) cluster regulation by achaete scute-like 2 (ASCL2): impact on the epithelial-mesenchymal transition in colon cancer cells. J Biol Chem. 2014;289(52):36101–36115. doi: 10.1074/jbc.M114.598383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Hahn S, Jackstadt R, Siemens H, Hunten S, Hermeking H. SNAIL and miR-34a feed-forward regulation of ZNF281/ZBP99 promotes epithelial-mesenchymal transition. EMBO J. 2013;32(23):3079–3095. doi: 10.1038/emboj.2013.236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Rokavec M, Oner MG, Li H, et al. Il-6R/ STAT3/miR-34a feedback loop promotes emt-mediated colorectal cancer invasion and metastasis. J Clin Invest. 2014;124(4):1853–1867. doi: 10.1172/JCI73531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Weiss L, Orr FW, Honn KV. Interactions of cancer cells with the microvasculature during metastasis. FASEB J. 1988;2(1):12–21. doi: 10.1096/fasebj.2.1.3275560. [DOI] [PubMed] [Google Scholar]
- 89.Asangani IA, Rasheed SA, Nikolova DA, et al. MicroRNA-21 (miR-21) post-transcriptionally downregulates tumor suppressor pdcd4 and stimulates invasion, intravasation and metastasis in colorectal cancer. Oncogene. 2008;27(15):2128–2136. doi: 10.1038/sj.onc.1210856. [DOI] [PubMed] [Google Scholar]
- 90.Zijlstra A, Lewis J, Degryse B, Stuhlmann H, Quigley JP. The inhibition of tumor cell intravasation and subsequent metastasis via regulation of in vivo tumor cell motility by the tetraspanin cd151. Cancer Cell. 2008;13(3):221–234. doi: 10.1016/j.ccr.2008.01.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Wang X, Yu H, Lu X, Zhang P, Wang M, Hu Y. miR-22 suppresses the proliferation and invasion of gastric cancer cells by inhibiting cd151. Biochem Biophys Res Commun. 2014;445(1):175–179. doi: 10.1016/j.bbrc.2014.01.160. [DOI] [PubMed] [Google Scholar]
- 92.Han ZB, Yang Z, Chi Y, et al. MicroRNA-124 suppresses breast cancer cell growth and motility by targeting cd151. Cell Physiol Biochem. 2013;31(6):823–832. doi: 10.1159/000350100. [DOI] [PubMed] [Google Scholar]
- 93.Arora H, Qureshi R, Park WY. miR-506 regulates epithelial mesenchymal transition in breast cancer cell lines. PLoS ONE. 2013;8(5):e64273. doi: 10.1371/journal.pone.0064273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Sengupta S, Den Boon JA, Chen IH, et al. MicroRNA 29c is down-regulated in nasopharyngeal carcinomas, up-regulating mrnas encoding extracellular matrix proteins. Proc Natl Acad Sci, USA. 2008;105(15):5874–5878. doi: 10.1073/pnas.0801130105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Bockhorn M, Jain RK, Munn LL. Active versus passive mechanisms in metastasis: do cancer cells crawl into vessels, or are they pushed? Lancet Oncol. 2007;8(5):444–448. doi: 10.1016/S1470-2045(07)70140-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Cereghetti DM, Lee PP. Tumor-derived exosomes contain microRNAs with immunological function: implications for a novel immunosuppression mechanism. Micro RNA. 2014;2(3):194–204. doi: 10.2174/22115366113029990012. [DOI] [PubMed] [Google Scholar]
- 97.Ogata-Kawata H, Izumiya M, Kurioka D, et al. Circulating exosomal microRNAs as biomarkers of colon cancer. PLoS ONE. 2014;9(4):e92921. doi: 10.1371/journal.pone.0092921. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Lin R, Chen L, Chen G, et al. Targeting miR-23a in CD8+ cytotoxic t lymphocytes prevents tumor-dependent immunosuppression. J Clin Invest. 2014;124(12):5352–5367. doi: 10.1172/JCI76561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Carissimi C, Carucci N, Colombo T, et al. miR-21 is a negative modulator of t-cell activation. Biochimie. 2014;107(Pt B):319–326. doi: 10.1016/j.biochi.2014.09.021. [DOI] [PubMed] [Google Scholar]
- 100••.Jasinski-Bergner S, Mandelboim O, Seliger B. The role of microRNAs in the control of innate immune response in cancer. J Natl Cancer Inst. 2014;106(10) doi: 10.1093/jnci/dju257. pii: dju257. miRNAs regulate immune cells. [DOI] [PubMed] [Google Scholar]
- 101.Lu LF, Liston A. MicroRNA in the immune system, microRNA as an immune system. Immunology. 2009;127(3):291–298. doi: 10.1111/j.1365-2567.2009.03092.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Mao CP, He L, Tsai YC, et al. In vivo microRNA-155 expression influences antigen-specific t cell-mediated immune responses generated by DNA vaccination. Cell Biosci. 2011;1(1):3. doi: 10.1186/2045-3701-1-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Penna E, Orso F, Cimino D, et al. MicroRNA-214 contributes to melanoma tumour progression through suppression of TFAP2c. EMBO J. 2011;30(10):1990–2007. doi: 10.1038/emboj.2011.102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Valastyan S, Reinhardt F, Benaich N, et al. A pleiotropically acting microRNA, miR-31, inhibits breast cancer metastasis. Cell. 2009;137(6):1032–1046. doi: 10.1016/j.cell.2009.03.047. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 105.Fisher ER, Turnbull RB., Jr The cytologic demonstration and significance of tumor cells in the mesenteric venous blood in patients with colorectal carcinoma. Surg Gynecol Obstet. 1955;100(1):102–108. [PubMed] [Google Scholar]
- 106.Fisher ER, Fisher B. Experimental studies of factors influencing the development of hepatic metastases. XIII. Effect of hepatic trauma in parabiotic pairs. Cancer Res. 1963;23:896–900. [PubMed] [Google Scholar]
- 107.Kim J, Takeuchi H, Lam ST, et al. Chemokine receptor CXCR4 expression in colorectal cancer patients increases the risk for recurrence and for poor survival. J Clin Oncol. 2005;23(12):2744–2753. doi: 10.1200/JCO.2005.07.078. [DOI] [PubMed] [Google Scholar]
- 108.Kim J, Mori T, Chen SL, et al. Chemokine receptor cxcr4 expression in patients with melanoma and colorectal cancer liver metastases and the association with disease outcome. Ann Surg. 2006;244(1):113–120. doi: 10.1097/01.sla.0000217690.65909.9c. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Ghadjar P, Coupland SE, Na IK, et al. Chemokine receptor ccr6 expression level and liver metastases in colorectal cancer. J Clin Oncol. 2006;24(12):1910–1916. doi: 10.1200/JCO.2005.04.1822. [DOI] [PubMed] [Google Scholar]
- 110.Wicha MS, Liu S, Dontu G. Cancer stem cells: an old idea – a paradigm shift. Cancer Res. 2006;66(4):1883–1890. doi: 10.1158/0008-5472.CAN-05-3153. discussion 1895–1886. [DOI] [PubMed] [Google Scholar]
- 111•.Ma YL, Zhang P, Wang F, et al. Human embryonic stem cells and metastatic colorectal cancer cells shared the common endogenous human microRNA-26b. J Cell Mol Med. 2011;15(9):1941–1954. doi: 10.1111/j.1582-4934.2010.01170.x. Stemness-associated miRNAs contribute to CRC development. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Hwang WL, Jiang JK, Yang SH, et al. MicroRNA-146a directs the symmetric division of snail-dominant colorectal cancer stem cells. Nat Cell Biol. 2014;16(3):268–280. doi: 10.1038/ncb2910. [DOI] [PubMed] [Google Scholar]
- 113.Hewitson P, Glasziou P, Irwig L, Towler B, Watson E. Screening for colorectal cancer using the faecal occult blood test, hemoccult. Cochrane Database Syst Rev. 2007;(1):CD001216. doi: 10.1002/14651858.CD001216.pub2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Mitchell PS, Parkin RK, Kroh EM, et al. Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci, USA. 2008;105(30):10513–10518. doi: 10.1073/pnas.0804549105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Kosaka N, Iguchi H, Ochiya T. Circulating microRNA in body fluid: a new potential biomarker for cancer diagnosis and prognosis. Cancer Sci. 2010;101(10):2087–2092. doi: 10.1111/j.1349-7006.2010.01650.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Kanaan Z, Roberts H, Eichenberger MR, et al. A plasma microRNA panel for detection of colorectal adenomas: a step toward more precise screening for colorectal cancer. Ann Surg. 2013;258(3):400–408. doi: 10.1097/SLA.0b013e3182a15bcc. [DOI] [PubMed] [Google Scholar]
- 117.Du M, Liu S, Gu D, et al. Clinical potential role of circulating microRNAs in early diagnosis of colorectal cancer patients. Carcinogenesis. 2014;35(12):2723–2730. doi: 10.1093/carcin/bgu189. [DOI] [PubMed] [Google Scholar]
- 118••.Toiyama Y, Takahashi M, Hur K, et al. Serum miR-21 as a diagnostic and prognostic biomarker in colorectal cancer. J Natl Cancer Inst. 2013;105(12):849–859. doi: 10.1093/jnci/djt101. Serum miR-21 as a promising noninvasive tool for CRC diagnosis. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Giraldez MD, Lozano JJ, Ramirez G, et al. Circulating microRNAs as biomarkers of colorectal cancer: results from a genome-wide profiling and validation study. Clin Gastroenterol Hepatol. 2013;11(6):681–688. e683. doi: 10.1016/j.cgh.2012.12.009. [DOI] [PubMed] [Google Scholar]
- 120.Yamada A, Cox MA, Gaffney KA, Moreland A, Boland CR, Goel A. Technical factors involved in the measurement of circulating microRNA biomarkers for the detection of colorectal neoplasia. PLoS ONE. 2014;9(11):e112481. doi: 10.1371/journal.pone.0112481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Zanutto S, Pizzamiglio S, Ghilotti M, et al. Circulating miR-378 in plasma: a reliable, haemolysis-independent biomarker for colorectal cancer. Br J Cancer. 2014;110(4):1001–1007. doi: 10.1038/bjc.2013.819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Hu J, Wang Z, Liao BY, et al. Human miR-1228 as a stable endogenous control for the quantification of circulating microRNAs in cancer patients. Int J Cancer. 2014;135(5):1187–1194. doi: 10.1002/ijc.28757. [DOI] [PubMed] [Google Scholar]
- 123••.Wu CW, Ng SS, Dong YJ, et al. Detection of miR-92a and miR-21 in stool samples as potential screening biomarkers for colorectal cancer and polyps. Gut. 2012;61(5):739–745. doi: 10.1136/gut.2011.239236. Fecal miR-21 as promsing screening biomarker for CRC. [DOI] [PubMed] [Google Scholar]
- 124.Zheng G, Du L, Yang X, et al. Serum microRNA panel as biomarkers for early diagnosis of colorectal adenocarcinoma. Br J Cancer. 2014;111(10):1985–1992. doi: 10.1038/bjc.2014.489. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Yuan W, Sui C, Liu Q, Tang W, An H, Ma J. Up-regulation of microRNA-145 associates with lymph node metastasis in colorectal cancer. PLoS ONE. 2014;9(7):e102017. doi: 10.1371/journal.pone.0102017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Nonaka R, Nishimura J, Kagawa Y, et al. Circulating miR-199a-3p as a novel serum biomarker for colorectal cancer. Oncol Rep. 2014;32(6):2354–2358. doi: 10.3892/or.2014.3515. [DOI] [PubMed] [Google Scholar]
- 127.Yau TO, Wu CW, Dong Y, et al. MicroRNA-221 and microRNA-18a identification in stool as potential biomarkers for the non-invasive diagnosis of colorectal carcinoma. Br J Cancer. 2014;111(9):1765–1771. doi: 10.1038/bjc.2014.484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Phua LC, Chue XP, Koh PK, Cheah PY, Chan EC, Ho HK. Global fecal microRNA profiling in the identification of biomarkers for colorectal cancer screening among asians. Oncol Rep. 2014;32(1):97–104. doi: 10.3892/or.2014.3193. [DOI] [PubMed] [Google Scholar]
- 129.Ghanbari R, Mosakhani N, Asadi J, et al. Decreased expression of fecal miR-4478 and miR-1295b-3p in early-stage colorectal cancer. Cancer Biomark. 2014;15(2):195–201. doi: 10.3233/CBM-140453. [DOI] [PubMed] [Google Scholar]
- 130.Wang J, Huang SK, Zhao M, et al. Identification of a circulating microRNA signature for colorectal cancer detection. PLoS ONE. 2014;9(4):e87451. doi: 10.1371/journal.pone.0087451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Yin J, Bai Z, Song J, et al. Differential expression of serum miR-126, miR-141 and miR-21 as novel biomarkers for early detection of liver metastasis in colorectal cancer. Chin J Cancer Res. 2014;26(1):95–103. doi: 10.3978/j.issn.1000-9604.2014.02.07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Wang S, Xiang J, Li Z, et al. A plasma microRNA panel for early detection of colorectal cancer. Int J Cancer. 2015;136(1):152–161. doi: 10.1002/ijc.28136. [DOI] [PubMed] [Google Scholar]
- 133.Nishida N, Yamashita S, Mimori K, et al. MicroRNA-10b is a prognostic indicator in colorectal cancer and confers resistance to the chemotherapeutic agent 5-fluorouracil in colorectal cancer cells. Ann Surg Oncol. 2012;19(9):3065–3071. doi: 10.1245/s10434-012-2246-1. [DOI] [PubMed] [Google Scholar]
- 134.Hur K, Toiyama Y, Schetter AJ, et al. Identification of a metastasis-specific microRNA signature in human colorectal cancer. J Natl Cancer Inst. 2015 doi: 10.1093/jnci/dju492. Epub ahead of print. [DOI] [PMC free article] [PubMed]
- 135.Yu G, Tang JQ, Tian ML, et al. Prognostic values of the miR-17-92 cluster and its paralogs in colon cancer. J Surg Oncol. 2012;106(3):232–237. doi: 10.1002/jso.22138. [DOI] [PubMed] [Google Scholar]
- 136.Oue N, Anami K, Schetter AJ, et al. High miR-21 expression from FFPE tissues is associated with poor survival and response to adjuvant chemotherapy in colon cancer. Int J Cancer. 2014;134(8):1926–1934. doi: 10.1002/ijc.28522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Kjaer-Frifeldt S, Hansen TF, Nielsen BS, et al. The prognostic importance of miR-21 in stage ii colon cancer: a population-based study. Br J Cancer. 2012;107(7):1169–1174. doi: 10.1038/bjc.2012.365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Nosho K, Igarashi H, Nojima M, et al. Association of microRNA-31 with braf mutation, colorectal cancer survival and serrated pathway. Carcinogenesis. 2014;35(4):776–783. doi: 10.1093/carcin/bgt374. [DOI] [PubMed] [Google Scholar]
- 139.Weissmann-Brenner A, Kushnir M, Lithwick Yanai G, et al. Tumor microRNA-29a expression and the risk of recurrence in stage II colon cancer. Int J Oncol. 2012;40(6):2097–2103. doi: 10.3892/ijo.2012.1403. [DOI] [PubMed] [Google Scholar]
- 140.Ress AL, Stiegelbauer V, Winter E, et al. miR-96-5p influences cellular growth and is associated with poor survival in colorectal cancer patients. Mol Carcinog. 2014 doi: 10.1002/mc.22218. (Epub ahead of print) [DOI] [PubMed] [Google Scholar]
- 141.Takahashi M, Cuatrecasas M, Balaguer F, et al. The clinical significance of miR-148a as a predictive biomarker in patients with advanced colorectal cancer. PLoS ONE. 2012;7(10):e46684. doi: 10.1371/journal.pone.0046684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Shivapurkar N, Weiner LM, Marshall JL, et al. Recurrence of early stage colon cancer predicted by expression pattern of circulating microRNAs. PLoS ONE. 2014;9(1):e84686. doi: 10.1371/journal.pone.0084686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Ma Y, Zhang P, Wang F, et al. miR-150 as a potential biomarker associated with prognosis and therapeutic outcome in colorectal cancer. Gut. 2012;61(10):1447–1453. doi: 10.1136/gutjnl-2011-301122. [DOI] [PubMed] [Google Scholar]
- 144.Pichler M, Ress AL, Winter E, et al. miR-200a regulates epithelial to mesenchymal transition-related gene expression and determines prognosis in colorectal cancer patients. Br J Cancer. 2014;110(6):1614–1621. doi: 10.1038/bjc.2014.51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Toiyama Y, Hur K, Tanaka K, et al. Serum miR-200c is a novel prognostic and metastasis-predictive biomarker in patients with colorectal cancer. Ann Surg. 2014;259(4):735–743. doi: 10.1097/SLA.0b013e3182a6909d. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Slattery ML, Herrick JS, Mullany LE, et al. An evaluation and replication of miRNAs with disease stage and colorectal cancer-specific mortality. Int J Cancer. 2014 doi: 10.1002/ijc.29384. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Zhang JX, Song W, Chen ZH, et al. Prognostic and predictive value of a microRNA signature in stage II colon cancer: a microRNA expression analysis. Lancet Oncol. 2013;14(13):1295–1306. doi: 10.1016/S1470-2045(13)70491-1. [DOI] [PubMed] [Google Scholar]
- 148.Ma Y, Zhang P, Wang F, et al. Elevated oncofoetal miR-17-5p expression regulates colorectal cancer progression by repressing its target gene p130. Nat Commun. 2012;3:1291. doi: 10.1038/ncomms2276. [DOI] [PubMed] [Google Scholar]
- 149.Igarashi H, Kurihara H, Mitsuhashi K, et al. Association of microRNA-31-5p with clinical efficacy of anti-EGFR therapy in patients with metastatic colorectal cancer. Ann Surg Oncol. 2014 doi: 10.1245/s10434-014-4264-7. (Epub ahead of print) [DOI] [PubMed] [Google Scholar]
- 150.Manceau G, Imbeaud S, Thiebaut R, et al. Hsa-miR-31-3p expression is linked to progression-free survival in patients with KRAS wild-type metastatic colorectal cancer treated with anti-EGFR therapy. Clin Cancer Res. 2014;20(12):3338–3347. doi: 10.1158/1078-0432.CCR-13-2750. [DOI] [PubMed] [Google Scholar]
- 151.Hansen TF, Sorensen FB, Lindebjerg J, Jakobsen A. The predictive value of microRNA-126 in relation to first line treatment with capecitabine and oxaliplatin in patients with metastatic colorectal cancer. BMC Cancer. 2012;12:83. doi: 10.1186/1471-2407-12-83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Diaz T, Tejero R, Moreno I, et al. Role of miR-200 family members in survival of colorectal cancer patients treated with fluoropyrimidines. J Surg Oncol. 2014;109(7):676–683. doi: 10.1002/jso.23572. [DOI] [PubMed] [Google Scholar]
- 153.Cappuzzo F, Sacconi A, Landi L, et al. MicroRNA signature in metastatic colorectal cancer patients treated with anti-egfr monoclonal antibodies. Clin Colorectal Cancer. 2014;13(1):37–45.e34. doi: 10.1016/j.clcc.2013.11.006. [DOI] [PubMed] [Google Scholar]
- 154.Link A, Becker V, Goel A, Wex T, Malfertheiner P. Feasibility of fecal microRNAs as novel biomarkers for pancreatic cancer. PLoS ONE. 2012;7(8):e42933. doi: 10.1371/journal.pone.0042933. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Ahmed FE, Ahmed NC, Vos PW, et al. Diagnostic microRNA markers to screen for sporadic human colon cancer in stool: I. Proof of principle. Cancer Genomics Proteomics. 2013;10(3):93–113. [PubMed] [Google Scholar]
- 156.Kalimutho M, Del Vecchio Blanco G, Di Cecilia S, et al. Differential expression of miR-144* as a novel fecal-based diagnostic marker for colorectal cancer. J Gastroenterol. 2011;46(12):1391–1402. doi: 10.1007/s00535-011-0456-0. [DOI] [PubMed] [Google Scholar]
- 157.Wu CW, Ng SC, Dong Y, et al. Identification of microRNA-135b in stool as a potential noninvasive biomarker for colorectal cancer and adenoma. Clin Cancer Res. 2014;20(11):2994–3002. doi: 10.1158/1078-0432.CCR-13-1750. [DOI] [PubMed] [Google Scholar]
- 158.Van Schaeybroeck S, Allen WL, Turkington RC, Johnston PG. Implementing prognostic and predictive biomarkers in CRC clinical trials. Nat Rev Clin Oncol. 2011;8(4):222–232. doi: 10.1038/nrclinonc.2011.15. [DOI] [PubMed] [Google Scholar]
- 159.Lombardi L, Morelli F, Cinieri S, et al. Adjuvant colon cancer chemotherapy: where we are and where we’ll go. Cancer Treat Rev. 2010;36(Suppl 3):S34–S41. doi: 10.1016/S0305-7372(10)70018-9. [DOI] [PubMed] [Google Scholar]
- 160.Xia X, Yang B, Zhai X, et al. Prognostic role of microRNA-21 in colorectal cancer: a meta-analysis. PLoS ONE. 2013;8(11):e80426. doi: 10.1371/journal.pone.0080426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Lou X, Qi X, Zhang Y, Long H, Yang J. Decreased expression of microRNA-625 is associated with tumor metastasis and poor prognosis in patients with colorectal cancer. J Surg Oncol. 2013;108(4):230–235. doi: 10.1002/jso.23380. [DOI] [PubMed] [Google Scholar]
- 162.Xu K, Liang X, Shen K, et al. miR-222 modulates multidrug resistance in human colorectal carcinoma by down-regulating ADAM-17. Exp Cell Res. 2012;318(17):2168–2177. doi: 10.1016/j.yexcr.2012.04.014. [DOI] [PubMed] [Google Scholar]
- 163.Xu K, Liang X, Shen K, et al. miR-297 modulates multidrug resistance in human colorectal carcinoma by down-regulating MRP-2. Biochem J. 2012;446(2):291–300. doi: 10.1042/BJ20120386. [DOI] [PubMed] [Google Scholar]
- 164.Kurokawa K, Tanahashi T, Iima T, et al. Role of miR-19b and its target mRNAs in 5-fluorouracil resistance in colon cancer cells. J Gastroenterol. 2012;47(8):883–895. doi: 10.1007/s00535-012-0547-6. [DOI] [PubMed] [Google Scholar]
- 165.Mencia N, Selga E, Noe V, Ciudad CJ. Underexpression of miR-224 in methotrexate resistant human colon cancer cells. Biochem Pharmacol. 2011;82(11):1572–1582. doi: 10.1016/j.bcp.2011.08.009. [DOI] [PubMed] [Google Scholar]
- 166.Orang AV, Barzegari A. MicroRNAs in colorectal cancer: from diagnosis to targeted therapy. Asian Pac J Cancer Prev. 2014;15(17):6989–6999. doi: 10.7314/apjcp.2014.15.17.6989. [DOI] [PubMed] [Google Scholar]
- 167.Paszko E, Senge MO. Immunoliposomes. Curr Med Chem. 2012;19(31):5239–5277. doi: 10.2174/092986712803833362. [DOI] [PubMed] [Google Scholar]
- 168.Pan H, Zhou Y, Sieling F, Shi J, Cui J, Deng C. Sonoporation of cells for drug and gene delivery. Conf Proc IEEE Eng Med Biol Soc. 2004;5:3531–3534. doi: 10.1109/IEMBS.2004.1403993. [DOI] [PubMed] [Google Scholar]
- 169.Kotopoulis S, Dimcevski G, Gilja OH, Hoem D, Postema M. Treatment of human pancreatic cancer using combined ultrasound, microbubbles, and gemcitabine: a clinical case study. Med Phys. 2013;40(7):072902. doi: 10.1118/1.4808149. [DOI] [PubMed] [Google Scholar]
- 170.Ibrahim AF, Weirauch U, Thomas M, Grunweller A, Hartmann RK, Aigner A. MicroRNA replacement therapy for miR-145 and miR-33a is efficacious in a model of colon carcinoma. Cancer Res. 2011;71(15):5214–5224. doi: 10.1158/0008-5472.CAN-10-4645. [DOI] [PubMed] [Google Scholar]
- 171.Ando H, Okamoto A, Yokota M, et al. Development of a miR-92a delivery system for anti-angiogenesis-based cancer therapy. J Gene Med. 2013;15(1):20–27. doi: 10.1002/jgm.2690. [DOI] [PubMed] [Google Scholar]


