Open access peer-reviewed chapter

MicroRNAs (miRNAs) in Colorectal Cancer

Written By

Burcin Baran, Nazli-Mert Ozupek, Gizem Calibasi-Kocal and Yasemin Basbinar

Submitted: 05 May 2018 Reviewed: 09 August 2018 Published: 07 November 2018

DOI: 10.5772/intechopen.80828

From the Edited Volume

Oncogenes and Carcinogenesis

Edited by Pinar Erkekoglu

Chapter metrics overview

1,417 Chapter Downloads

View Full Metrics

Abstract

Colorectal cancer (CRC) is the third most common cancer in the world and third leading cause of cancer-related deaths in men and women as well. While early screening procedures and removal of small polyps improve the survival rates among the patients, there is still need for new diagnostic and therapeutic approaches for developing more effective treatments. MicroRNAs (miRNAs) are short noncoding RNA fragments, which involve in posttranscriptional regulation of gene expression, and they are shown to involve in tumorigenesis either targeting oncogenes or tumor suppressor genes. Based on the current studies, miRNAs are now suggested as potential biomarkers for CRC diagnosis, prognosis, and therapeutic responses. In this chapter, the latest findings on the role of miRNA in CRC in many aspects are reviewed: diagnosis (role of circular miRNAs in blood and miRNAs from tissue biopsies and their potential role in pathophysiology and diagnosis of CRC), prognosis (miRNAs related with metastasis, recurrence, and survival rates in CRC), and therapeutic responses (role of miRNAs both in chemotherapies and/or in targeted therapies in CRC). In conclusion, miRNAs are promising molecules for diagnosis, prognosis, and therapeutic responses of CRC.

Keywords

  • colorectal cancer
  • diagnosis
  • miRNA
  • prognosis
  • therapeutic response

1. Introduction

MicroRNAs are a subgroup of small noncoding RNAs containing 18–25 nucleotides, and they do not carry any genetic information for protein expression. They regulate the posttranslational gene expression by binding 3′ untranslated region (UTR) of the target messenger RNA (mRNA). Approximately 30% of protein coding genes are regulated by miRNAs, and they have important roles in cellular functions including proliferation, differentiation, apoptosis, signaling, metabolism, and tumorigenesis. Due to their effect on crucial processes, miRNAs are significant modifiers of transcription and translation of both oncogenes and tumor suppressor proteins. Hence, some of them are classified as oncomiR and tumor suppressor miRNA in the cellular processes of tumor [1].

First miRNA, lin-4, was discovered in Caenorhabditis elegans in 1993, and it had role on the regulation of larval development by the repression of a nuclear protein encoded by lin-14. The second discovered miRNA, let-7, is expressed in late development and complementary to the 3′ UTR of the several genes including lin-14, lin-28, lin-41, lin-42, and daf-12. After the discovery of lin-4 and let-7, miRNAs were shown in other organisms including plants and animals [2, 3], and over 10,000 miRNAs have been identified in various organisms. In humans, over 2500 types of encoded miRNAs have been determined [4].

Advertisement

2. Biogenesis of miRNA

The biogenesis of miRNA is a complicated process, starting in the nucleus, following with posttranslational modifications, and finalized in the cytoplasm. Similar to gene encoding, biogenesis of primary miRNAs (pri-miRNAs) is starting with the transcription by RNA polymerase II or RNA polymerase III enzyme. In the nucleus, pri-miRNA is recognized and cleaved by Drosha enzyme to form precursor miRNA (pre-miRNA). The pre-miRNA is exported to cytoplasm by exportin-5. In the cytoplasm, pre-miRNA is bound to cytoplasmic RNase Dicer and RNA-induced silencing complex (RISC), which is composed of argonaute 2 (AGO2) and transactivation response (TAR) RNA-binding protein (TRBP). Firstly, AGO2 cleaves the pre-miRNA from its 3′ end, and the cleaved pre-miRNA is further cleaved by Dicer into mature miRNA duplex. Mature miRNA duplex is then unwounded; while one strand of the miRNA remains on AGO2 protein, and the other strand (passenger strand) is degraded. Mostly, miRNAs are recognizing the complementary sequence of 3′ UTR of mRNAs, hence directing RISC to cleave mRNAs and translational repression of mRNAs [5, 6] (Figure 1).

Figure 1.

miRNA biogenesis. The pathway starts miRNA transcription by RNA polymerase II or III to generate the primary transcripts (pri-miRNAs). Pri-miRNA is processed by the Drosha-DiGeorge syndrome critical region gene 8 (DGCR8, Pasha Pasha in Drosophila melanogaster and Caenorhabditis elegans) complex (also known as the microprocessor complex) that generates ~70 nucleotide (nt) pre-miRNAs. Pre-miRNA, which is recognized by the nuclear export factor exportin-5, is transferred to the cytoplasm. In the cytoplasm, the cytoplasmic RNase Dicer cleaves the pre-miRNA hairpin to its mature length. Dicer in complex with the transactivation response (TAR) RNA-binding protein (also known as TRBP and TARBP2) and argonaute (AGO) 1–4 mediate the processing of pre-miRNA and the assembly of the RISC (RNA-induced silencing complex). With the formation of this complex structure, one strand of the miRNA duplex is removed and single-stranded miRNA is generated. Interaction between microRNA complex and target mRNA induces post-transcriptional silencing by destabilization of mRNA and suppression of translation [7, 8].

Advertisement

3. Involvement of microRNAs in cancer

microRNA studies were began in C. elegans, as lin-4 and let-7 were identified as noncoding RNAs functioning in larval development. Soon after, the research groups focused on the function of these noncoding RNAs and discovered their homologs in vertebrates as well. The role of miRNAs in tumorigenesis was first reported in chronic lymphocytic leukemia (CLL) by two different groups in 2002. Hemizygous or homozygous loss of 13q14 chromosome was frequently observed among CLL patients [9]. Two different miR-15 and miR-16 expression levels were shown to be downregulated with the deletion of this locus [10]. Both miR-15/16 levels are inversely correlated with antiapoptotic B cell lymphoma-2 (Bcl-2) protein level in the cells. Introduction of miR-15/16 to the leukemic cell lines repressed Bcl-2 expression and induced apoptosis in these cells [11]. It is now very well established that aberrant miRNA expression contributes to cancer [12]. miRNAs are targeting the genes, which involve in cell proliferation, migration, invasion, and metastasis; hence dysregulation of these miRNAs leads to transformation and malignancy of cells [13, 14]. miRNA dysregulation in cancer cells can be result of genomic deletion, mutations, amplification, or epigenetic silencing [14]. A single miRNA can target a variety of mRNAs involved in different cell signaling pathways; interestingly, a single mRNA can be targeted by several miRNAs also [15], such as Let-7, which is one of the initially discovered miRNAs, targets human rat sarcoma (RAS), high-mobility group AT-hook 2 (HMGA2), and MYC mRNAs and downregulates their expression [16]. Phosphotensin homolog (PTEN), which is an important regulator of cell cycle, can be targeted by several different miRNAs including miR-21, miR-22, miR-106b-25, miR-17-92 [17].

In tumorigenesis, miRNAs either act as tumor suppressor or as an oncogene; interestingly, their expression is repressed or induced by transcription factors such as p53 or MYC via their promoter regions. miR-145 is one of the initial examples of tumor suppressor miRNAs. miR-145 was found to be downregulated in a variety of tumors including colon, breast carcinomas [18, 19]. It is interesting that tumor suppressor protein p53 induces miR-145 expression via p53 response element in its promoter. Later, miR-145 targets c-Myc or insulin receptor substrate I (IGF-R1) protooncogenes and silences their expressions, hence preventing tumor cell proliferation [18, 20] . Furthermore miR-145 inhibits invasion and metastasis by targeting Fli-1 or Mucin-1 [20, 21]. miR-145 also targets estrogen receptor-α (ER-α) via its two complementary sites and downregulates ER-α expression [22]. miR-34 family is another target of p53 tumor suppressor protein [23]. Another important tumor suppressor miRNA is miR-34 family. miR-34 family comprises three members: miR-34a, miR-34b, and miR-34c. While miR-34a is ubiquitously expressed in every tissue, expression of miR-34b and miR-34c is restricted to fallopian tubes, lungs, and brain [24, 25]. miR-34a is a very potential tumor suppressor since it is targeting many mRNAs related with proliferation [such as cyclin-dependent kinase-4 (CDK4) and cyclin-dependent kinase-6 (CDK6)], cellular growth [such as Notch2, platelet-derived growth factor receptor A (PDGFRA)], antiapoptosis [Bcl-2, sirtuin 1 (SIRT1), survivin], invasion, and migration [MET, SNAIL, cluster of differentiation (CD44)] [26, 27, 28]. Downregulation of miR-34 is observed among many malignancies and associated with poor prognosis [29, 30]. As a result of its role as a tumor suppressor, miR-34 has been applied either alone or in combination with conventional therapies on several tumor cell lines and mouse tumor models and showed promising results [31, 32, 33, 34]. miR-34 was first miRNA tested in human Phase I trial (NCT01829971). MRX34, liposomal miR-34 mimic, was tested among patients with solid advanced tumors. While MRX34 treatment showed evidence of antitumor activity in a subset of patients, it exerts some toxicities in patients. Hence, there is need for further studies for improving tolerability among the patients [35, 36].

In addition to tumor suppressor miRNAs, miRNAs behave like oncogenes, called as “oncomiRs.” mir-21 is the first miRNA identified as oncogenic; it is significantly upregulated in many tumors including colon cancer, breast cancer, hepatocellular carcinoma, and glioblastoma [37]. miR-21 overexpression contributes to cell proliferation and antiapoptotic responses by targeting important downstream proteins such as phosphotensin homolog (PTEN), programmed cell death protein 4 (PDC4), and tropomyosin I [38, 39, 40]. Besides this, miR-21 was shown to be bona fide oncogene by causing pre-B-cell lymphoma in mouse models by overexpression. When mir-21 expression was inactivated, tumors regressed completely in few days [41].

As the importance of miRNAs became evident, miRNA expression profiles for each tumor type have been studied with several methodologies including microarray, QRT-PCR, and next-generation sequencing [15, 42]. miRNA expression profiles can reflect embryonic or development origin of the tissue and able to classify the origin of tissue with high accuracy (>90%), even separate cell subtypes (stem cells vs. progenitor cells) in the same tissue [43, 44, 45]. These miRNA profiling studies open the way for biomarker studies. In the biomarker studies, it is aimed to find diagnostic, prognostic, and predictive markers for better characterization of the disease and therapy response as an outcome [46].

Advertisement

4. miRNA and colorectal cancer

Colorectal cancer (CRC) is the second most common cancer among the women and third most common cancer among men. In 2016, more than 1.4 million men and women in the USA have been diagnosed with CRC [47]. Despite the availability of successful treatment options such as surgery, chemotherapy, and radiotherapy, the prognosis of CRC is not promising. Relapse or metastatic spread occurs after surgery in many CRC patients. Colorectal cancer is divided into two phenotypes according to mutational status. In chromosomal instability phenotype (CIN), high rate of inactivating mutations in adenomatous polyposis coli (APC) and tumor protein P53 (TP53) genes are found as well as activating mutations in Kirsten rat sarcoma viral oncogene homolog (KRAS) gene. However mutations in DNA repair genes, transforming growth factor-beta receptor II (TGFBRII) gene, Bcl2- associated C protein (BAX) and BRAF genes are commonly existed in microsatellite instability-high tumors (MSI-H) [48]. Certainly, genomic background affects the miRNA expression in CRC, such as TP53 mutations affect miR-145 expression levels, which is downregulated among many CRC patients [49, 50]. Furthermore, miR-193a-3p expression was found as specifically downregulated in BRAF-mutated CRC cases [51]. The distinction between these phenotypes became more prominent in disease progression and therapy response, which will be discussed in the following sections. In CRC, to date, totally, 1870 original studies were retrieved in PubMed (as of May 2018), in which 38 of them were clinical trials investigating miRNA expression patterns in both CRC tissue specimen and plasma samples and compared them with normal samples. Bunch of miRNAs were found to be dysregulated in CRC samples in these studies [52, 53, 54]. While some of these miRNAs are related with early stages of tumorigenesis and can be used as diagnostic markers, the others are associated with therapeutic response, resistance to chemotherapy, and survival prognosis, hence aiding the physician in making therapeutic decisions as prognostic and predictive biomarkers [55].

4.1 miRNAs in colorectal cancer diagnosis

Early diagnosis is essential for CRC patients since they have more favorable prognosis. Fecal blood test and colonoscopy techniques are being currently used for early screening. However, fecal blood tests are not very efficient for detecting early carcinoma formation. Colonoscopy is a gold standard technique, it reduces cancer risk about 30–75%, yet it is invasive and expensive technique and highly uncomfortable for a patient [56]. Therefore, noninvasive and inexpensive screening and diagnostic methods or biomarkers are needed. miRNAs are promising candidates for noninvasive biomarker diagnosis. Diagnostic miRNAs can be isolated from blood or stool samples as well as tumor tissues [57] (Table 1).

miRNAsExpressionTarget genes
miR-15aUpregulateBcl-2
miR-17-3pUpregulateE2F, CDKN1A
miR-18aUpregulateSMAD4, KRAS
miR-19a/miR-19bUpregulatePTEN
miR-20aUpregulateBECN1, ATG16L1, SQSTM1
miR-21UpregulatePDCD4, PTEN, SPRY2, TPMI
miR-24DownregulateTopoisomerase-1
miR-29aUpregulateDNMT3
miR-31DownregulateWNT, β-catenin
miR-34aDownregulateSMAD4, FRAT1, Bcl-2, c-Met
miR-92aUpregulatePHLPP2, VHL, Bim
Let-7gUpregulateKRAS, Cdk6, Cdc25, HMGA2
miR-106bUpregulateP21, E2F1
miR-133aDownregulateMCL1, BCL2L2
miR-143DownregulateErk5, DNMT3, KRAS
miR-145DownregulateEGFR, IRS-1
miR-181bDownregulateATM
miR-203DownregulateABL1, TP63
miR-223UpregulateSTMN1
miR-302UpregulateGAB2, AKT2
miR-320aDownregulateVDAC, STAT3, SOX4
miR-335UpregulateRASA-1
miR-375DownregulateSLC7A11, IGFR1, SEC23A
miR-422aDownregulateTGF-β, CD73
miR-423-5pDownregulateRFVT3
miR-601DownregulatePTP4A1
miR-760DownregulatePHLPP2

Table 1.

Simplified list of diagnostic miRNA markers for colorectal cancer (modified from Refs. [58, 59]).

Abbreviations: Bcl-2, B cell lymphoma-2; E2F, E2F transcription factor 1; CDKN1A, cyclin-dependent kinase inhibitor 1A; KRAS, Kirsten rat sarcoma viral oncogene homolog; PTEN, phosphotensin homolog; BECN1, Beclin 1; ATG16L1, autophagy-related 16 like 1; SQSTM1, sequestosome 1; PDCD4, programmed cell death 4; SPRY2, sprouty RTK signaling antagonist 2; DNMT3, DNA methyl transferase 3; FRAT1, WNT signaling pathway regulator; PHLPP2, PH domain leucine-rich repeat protein phosphatase 2; VHL, von Hippel-Lindau tumor suppressor; Cdk6, cyclin-dependent kinase 6; Cdc25, cell division cycle 25A; HMGA2, high-mobility group gene; P21, CDKN1A, cyclin-dependent kinase inhibitor 1A; E2F1, E2F transcription factor 1; MCL1, BCL2 family apoptosis regulator; BCL2L2, BCL2 like 2; EGFR, epidermal growth factor receptor; IRS-1, insulin receptor substrate 1; ATM, ataxia telangiectasia mutated; ABL1, v-abl Abelson murine leukemia viral oncogene homolog 1; TP63, tumor protein p63; STMN1, stathmin 1; GAB2, GRB2-associated binding protein 2; AKT2, v-akt murine thymoma viral oncogene homolog 2; VDAC, voltage-dependent anion channel; SOX4, SRY (sex-determining region Y)-box 4; SLC7A11, solute carrier family 7 member 11; IGFR1, insulin-like growth factor 1 receptor; TGF-β, transforming growth factor-beta; CD73, cluster of differentiation 73; RFVT3, known as SLC52A3 (solute carrier family 52 member 3); PTP4A1, protein tyrosine phosphatase 4a1.

There are different miRNA profiling studies comparing CRC samples with normal healthy tissue samples; however, each study emphasized on different set of miRNAs in CRC diagnosis and progression. According to miRNA profile study, miR-18a, -20a, -21, -29a, -92a, -106b, -133a, -143, and -145 expression levels were found to be significantly changed in CRC patients when compared with normal patients, and these markers can be used for CRC diagnosis [59]. In a systematic review, miR-106a, -30a-3p, -139, -145, -125a, and -133a were proposed as diagnostic biomarkers [60]. In another study, miR-143, -145, -21, -320, -126, -484-5p, -143, -145, -16, -125b, -21, and -106 were found to be candidate for diagnostic biomarkers [57]. While studies share some common miRNAs (such as miR143, miR145, miR106, miR21), they are differing in their list of miRNAs. In fact, the type of miRNAs can be differed due to the type of sample (blood or stool), experimental procedures, and used microRNA platforms. Another handicap of these studies is that they have been conducted with a small number of samples. Larger sample studies and additional meta-analyses are need for better determination of CRC-related diagnostic markers. Still, it can be said that miRNAs are very promising noninvasive markers for tumor diagnosis.

4.2 miRNAs in colorectal cancer prognosis

Taking part in CRC diagnosis, miRNAs are also affecting prognosis and therapeutic response. As mentioned before, the expression and deregulation of miRNAs in CRC patients are affected by chromosomal abnormalities and microsatellite instability [61, 62]. In CRC, miRNA expression dysregulation is shown especially in microsatellite instability (MSI-high) tumors. MSI-high groups are distinct population among CRC patients, which accounts for 15% of all cases, observed in hereditary cases such as Lynch syndrome or in sporadic cases mostly as a result of hypermethylation or inactivation of mismatch repair (MMR) genes [63]. These MSI tumors characterized by distinct behavior are associated with proximal tumor localization and high infiltration of lymphocytes. These phenotypes showed less distant organ metastasis than MSI stable tumors and have better prognosis [64]. Several miRNAs have been shown in participating in inactivation of several DNA mismatch repair genes, such as miR-155 downregulates mutL protein homolog 1 (MLH1), mutS homolog 2 (MSH2), and mutS homolog 6 (MSH6) mRNAs expression, whereas miR-21 targets MSH2 and MSH6 mRNA and inactivates them [6566]. Overall 94 miRNAs are differently expressed in microsatellite stable and in microsatellite instable tumors [67]. Upregulation (miR-17, miR-20, miR-25, miR-31, miR92, miR-93, miR-133b, miR-135a, miR-183, miR-203, and miR-223) and downregulation (miR-16, miR-26b, miR-143, miR-145, miR-191, miR-192, miR-215, and let-7a) are generally observed in MSI-high tumors [68]. miRNA expression is also differed among TP53 and KRAS mutated tumors as well. miR-125p targets 3′ UTR region of p53 and represses p53 expression and accelerates the tumor growth; hence, expression levels of miR-125p are associated with poor survival among CRC patients [69]. However, miR34 expression is a good prognostic marker. miR-34 is one of the targets of p53 protein and it increases miRNA expression. miR-34 then suppresses the expression of WNT pathway and epithelial mesenchymal transition (EMT)-related genes. Increase of miR-34b and miR-34c levels in stromal tissue is leading to poor prognosis in colon cancer [70, 71, 72]. miR-122, miR-214, miR-372, miR-15b, let-7e, and miR-17 are other dysregulated miRNAs found in TP53 mutated tumors [73]. miR-148-b and miR-221 are also important diagnostic markers associated with p53 mutational status, and their overexpression is associated with worse prognosis [74, 75]. miR-143 and miR-145 are frequently downregulated in CRC and their one of the targets is KRAS mRNA; hence, they are important prognostic and predictive biomarkers in CRC [76, 77]. Let-7 role is one of the well-studied tumor suppressor miRNAs, which targets RAS. Let-7a expression is higher in KRAS mutated metastatic samples than normal mucosa or nonmetastatic disease [78]. Decrease Let-7b expression is worse prognostic marker, which is associated with recurrence and low overall survival of patients [79]. Furthermore, decrease in miR-487b levels is associated with liver metastasis in CRC patients [80]. Not only KRAS-associated miRNAs act as tumor suppressor, some of them are acting oncogenic in prognosis. miR-200 and miR-221 are downstream miRNAs of RAS pathway, and high expression of these miRNAs is related with worse prognosis [81].

Furthermore, exosome-containing miRNAs (miR-17/92 cluster and miR-19a cluster) are evaluated as biomarkers for early diagnosis and high recurrence in patients with CRC [82]. miR-21-5p, miR-29-3p, and miR-148-3p levels were studied in CRC samples and show that dysregulation in these miRNAs is associated with high mortality risk [83].

4.3 miRNAs in treatment response prediction of colorectal cancer

A variety of therapeutic advances are existed for CRC treatment such as conventional chemotherapy (5-fluorouracil, capecitabine, irinotecan, oxaliplatin), immunotherapy, radiotherapy, and chemoradiotherapy. miRNAs play an important role in the regulation of effectiveness and resistance to these therapies and prediction of personalized therapy response [84, 85]. Resistance to therapy is still the biggest challenge for defeating cancer. It may be caused by a variety of reasons such as reduction in transportation and intracellular accumulation of drugs by modulating the activity of drug transporters such ATP-binding cassette subfamily B (ABCB)/multidrug resistance (MDR) transporters (which is reviewed in reference [86]), dysregulation in DNA damage repair mechanisms, insufficient or oncogenic immune response, blockage of apoptosis, emergence of inflammation, and altered expression of oncogenes and tumor suppressor genes related with therapy response. miRNAs are actively participating in all of these resistance mechanisms [87, 88].

4.3.1 Chemotherapy

Although there are advances in cytotoxic and targeted therapy in CRC, drug resistance is one of the most important obstacles in front of successful chemotherapy [89]. Fluoropyrimidine-based chemotherapy (5-FU or capecitabine), vascular endothelial growth factor (VEGF)/vascular endothelial growth factor receptor (VEGFR)-targeted, and epidermal growth factor receptor (EGFR)-targeted therapies are the main therapeutic methods for CRC [87]. miRNAs have role in chemotherapy resistance in terms of deregulation of drug metabolism-related enzymes, increased efflux of chemotherapeutics, impairment of chemotherapeutic-induced apoptosis, modulation of DNA damage repair, and autophagy [87].

miR-92b-3p, miR-3156-5p, miR-10a-5p, and miR-125a-5 were found to be related with progression-free survival in meta;static CRC patients treated with 5-FU/oxaliplatin/bevacizumab regime [90]. A negative relationship was found between miR-27b, miR-148a, and miR-326 expression levels and progression-free survival in metastatic colorectal cancer patients receiving first-line oxaliplatin-based treatment [91]. The expression of miR-326 was related with decreased overall survival. These results proposed that plasma miRNAs can be used as noninvasive biomarkers for evaluating drug response in metastatic CRC patients who are treated with 5-FU and oxaliplatin-based chemotherapy [91] (Table 2).

miRNAsTherapyExpressionTarget genes
miR-7EGFR-targetedDownregulateEGFR, RAF-1
miR-10b5-FUUpregulateBIM
miR-20aOxaliplatinUpregulateBNIP2
miR-215-FUUpregulateMSH2
miR-225-FUDownregulateBTG-1
miR-23a5-FUUpregulateAPAF-1, ABCF-1
miR-27a, miR-27b5-FUDownregulateDPYD
miR-133bEGFR-targetedDownregulateEGFR
miR-139-5p5-FUDownregulateBcl-2
miR-143OxaliplatinDownregulateIGF-1R
miR-153OxaliplatinUpregulateFOXO3a
miR-199-5p, miR-375EGFRUpregulatePHLPP1
miR-2035-FUDownregulateTYMS
miR-203OxaliplatinUpregulateATM
miR-2045-FUDownregulateHMGA2
miR-2185-FUDownregulateTYMS, BIRC5
miR-302, miR-369, miR-200c5-FUUpregulateMRP8
miR-409-3pOxaliplatinDownregulateBeclin-1
miR-425-5p5-FUUpregulatePDCDIO
miR-4945-FUDownregulateDPYD
miR-519c5-FUDownregulateABCG2, HuR
miR-520gOxaliplatinUpregulateP21

Table 2.

The expression profile of miRNAs that have role on chemotherapy response in colorectal cancer (modified from Ref. [85]).

Abbreviations: 5-FU, 5-fluorouracil; EGFR, epidermal growth factor; RAF-1, Raf protooncogene; BNIP2, BCL2-interacting protein 2; MSH2, human mutS homolog 2; BTG-1, BTG antiproliferation factor 1; APAF-1, apoptotic peptidase-activating factor 1; ABCF-1, ATP-binding cassette subfamily D member 1; DPYD, dihydropyrimidine dehydrogenase; Bcl-2, B cell lymphoma-2; IGF-1R, insulin-like growth factor 1 receptor; FOXO3a, forkhead box class O3; PHLPP1, Phlpp1 PH domain and leucine-rich repeat protein; TYMS, thymidylate synthase; ATM, ataxia telangiectasia mutated; HMGA2, high mobility group AT-hook 2; BIRC5, baculoviral IAP repeat containing 5; MRP8, myeloid-related protein 8; ABCG2, ATP-binding cassette subfamily G member 2; P21, cyclin-dependent kinase inhibitor 1A.

4.3.2 Immunotherapy

Since chemo/radio therapies and surgery have limitations, immunotherapy is a good alternative to treat CRC patients. Immunotherapy aimed to evoke immune system to eliminate tumors either using immune stimulatory cytokines (vaccines, etc.) or checkpoint inhibitors [such as cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), programmed death 1 (PD-1) receptor, and its ligands (PD-L1/2)] [92]. Interestingly, immune cell filtrates more in MSI-high CRC, and these subtypes are responding better to immunotherapies [93].

miRNAs are essential in regulation of the immune response as well. The role of miR-34 has been mentioned earlier. Upregulation of miR-34a elicits the activation of tumor-infiltrating CD8+ T cells by targeting PD-L1 [94]. miRNAs are also involved in innate immunity by macrophages and NK cells, and adaptive immunity by B cells, T cells, and dendritic cells. miR-124 modulates signal transducer and activator of transcription 3 (STAT3) pathway and enhances the T cell-mediated immune clearance [95]. miR-491 regulates the proliferation and apoptosis of CD8+ T cells [96]. miR-491 inhibits the activation of CD8+ T cells and promotes its apoptosis via targeting B-cell lymphoma-extra-large (Bcl-xL), cyclin-dependent kinase-4 (CDK4), and T cell factor 1 (TCF1), hence aiding tumor cells escaping from immune system. Tumor-derived TGF-β also induces the miR-491 expression. Thus, miR-491 can be evaluated as a new immunotarget for CRC treatment [96].

miR-196b, miR-378a, and miR-486-5p are evaluated as predictive biomarkers for the efficacy of the vaccine treatment in CRC [97]. miRNAs were enrolled in Phase II studies. In 16 patients, high expression of miR-196b-5p and low expression of miR-378a-3p and miR-486-5p are associated with better prognosis after vaccine treatment. Hence, these miRNAs can be determined as novel biomarkers for prediction of outcome responses of patients [97].

4.3.3 Potential candidates

miRNAs are also involving in radiotherapy responses. The expression of miRNA-processing enzymes Drosha and Dicer was found to be upregulated in radioresistant cell lines when compared with radiosensitive cell lines [98]. The role of miRNAs in radiotherapy response was evaluated further in the study cited as reference [87]. In the study, biomarkers for the prediction of chemoradiotherapy response in CRC were identified by using integrative and systematic bioinformatics analysis. The unique target genes of miR-198 and miR-765 were altered significantly upon transfection of specific miRNA mimics in the radiosensitive cell line. Thus, it could be said that miR-198, miR-202, miR-371-5p, miR-513a-5p, miR-575, miR-630, and miR-765 could be used for predicting the response of CRC to preoperative chemoradiotherapy [87]. Still, further studies are needed to understand the miRNA role in radiotherapy/radiochemotherapy prediction.

Advertisement

5. Concluding remarks and limitations

By the discovery of miRNAs, a significant number of studies have been conducted to indicate the utility of miRNAs. According to the highlighted studies, miRNAs in body fluids have potential to be predictive, diagnostic or prognostic biomarkers; and also they can be therapeutic targets due to their inducer ability on tumorigenesis. Basically, miRNAs offer promising practice for screening, diagnosis, prognosis, and treatment of cancer. Therefore, these noncoding RNA fragments may be used alone or combined with other protocols to screen, diagnose, prognose, and treat cancer. However, their clinical importance is still not conclusive, and validation studies are needed for routine-based clinical application.

Evidences showed that inhibition of oncomiRs or replacement of tumor suppressive miRNAs could be used to develop innovative treatment approaches. Further studies are needed to reveal the molecular mechanisms on the regulation of miRNA biogenesis. Determination of miRNA target genes, molecular interactions between target mRNA and miRNAs, and signaling pathways will help to interpret molecular mechanisms of cancer. Besides investigations on miRNA expression patterns and their molecular mechanisms, studies on technological developments for reliable and cost-effective miRNA applications are also extremely important to enhance minimally invasive routine miRNA applications. Methodological variability among different clinical centers is the biggest limitation for the successful combination of miRNAs in cancer management. Standardization and normalization of essential steps of miRNA applications, such as miRNA extraction, processing, biobanking, and quantitation, eliminate the clinical facility-based variations. Using internal controls and enrollment of the laboratory accreditation/validation programs may present benefits for standardization. miRNAs have potential to be therapeutic targets and treatment options. But determination of mRNAs and miRNAs interactions and obtaining the large population-based multicenter cohorts are essential to use miRNAs in therapy. Especially before the implementation of miRNAs in clinics, evaluation of miRNA panels on large patient cohorts must be achieved.

References

  1. 1. Vannini I, Fanini F, Fabbri M. Emerging roles of microRNAs in cancer. Current Opinion in Genetics & Development. 2018;48:128-133. DOI: 10.1016/j.gde.2018.01.001
  2. 2. Lee RC, Feinabum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 1993;75:843-854. DOI: 10.1016/0092-8674(93)90529-Y
  3. 3. Harfe BD. MicroRNAs in vertebrate development. Current Opinion in Genetics & Development. 2005;15:410-415. DOI: 10.1016/j.gde.2005.06.012
  4. 4. Huang Y, Shen XJ, Zou Q , Wang SP, Tang SM, Zhang GZ. Biological functions of microRNAs: A review. Journal of Physiology and Biochemistry. 2011;67(1):129-139. DOI: 10.1007/s13105-010-0050-6
  5. 5. Wahid F, Shehzad A, Khan T, Kim YY. MicroRNAs: Synthesis, mechanism, function and recent clinical trials. Biochimica et Biophysica Acta. 2010;1803:1231-1243. DOI: 10.1016/j.bbamcr.2010.06.013
  6. 6. Garzon R, Fabbri M, Cimmino A, et al. MicroRNA expression and function in cancer. Trends in Molecular Medicine. 2006;12:580-587. DOI: 10.1016/j.molmed.2006.10.006
  7. 7. Winter J, Jung S, Keller S, Gregory RI, Diederichs S. Many roads to maturity: microRNA biogenesis pathways and their regulation. Nature Cell Biology. 2009;11(3):228-234. DOI: 10.1038/ncb0309-228
  8. 8. He L, Hannon GJ. MicroRNAs: Small RNAs with a big role in gene regulation. Nature Reviews. Genetics. 2004;5(7):522-531. Review. Erratum in: Nat Rev Genet. 2004 Aug;5(8):631. DOI: 10.1038/nrg1379
  9. 9. Dohner H, Stilgenbauer S, Benner A, et al. Genomic aberrations and survival in chronic lymphocytic leukemia. The New England Journal of Medicine. 2000;343:1910-1916. DOI: 10.1056/NEJM200012283432602
  10. 10. Calin GA, Dumitru CD, Shimizu M, et al. Frequent deletions and down-regulation of micro-RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. PNAS. 2002;99:15524-15529. DOI: 10.1073/pnas.242606799
  11. 11. Cimmino A, Calin GA, Fabbri M, et al. miR-15 and miR-16 induce apoptosis by targeting BCL2. PNAS. 2005;102(39):13944-13949. DOI: 10.1073/pnas.0506654102
  12. 12. Kent OA, Mendell JT. A small piece in the cancer puzzle: microRNAs as tumor suppressors. Oncogene. 2006;25:6188-6196. DOI: 10.1038/sj.onc.1209913
  13. 13. Price C, Chen J. MicroRNAs in cancer biology and therapy: Current status and perspectives. Genes & Diseases. 2014;1(1):53-63. DOI: 10.1016/j.gendis.2014.06.004
  14. 14. Garzon R, Calin G, Croce CA. MicroRNAs in cancer. Annual Review of Medicine. 2009;60:167-179. DOI: 10.1146/annurev.med.59.053006.104707
  15. 15. Hayes J, Peruzzi P, Lawler S. MicroRNAs in cancer: Biomarkers, functions and therapy. Trends in Molecular Medicine. 2014;20(8):460-469. DOI: 10.1016/j.molmed.2014.06.005
  16. 16. Lee H, Han S, Kwon CS, Lee D. Biogenesis and regulation of the let-7 miRNAs and their functional implications. Protein & Cell. 2016;7(2):100-113. DOI: 10.1007/s13238-015-0212-y
  17. 17. Bermúdez Brito M, Goulielmaki E, Papakonstanti EA. Focus on PTEN regulation. Frontiers in Oncology. 2015;5:166. DOI: 10.3389/fonc.2015.00166
  18. 18. Shi B, Sepp-Lorenzino L, Prisco M, Linsley P, de Angelis T, Baserga R. Micro RNA 145 targets the insulin receptor substrate-1 and inhibits the growth of colon cancer cells. The Journal of Biological Chemistry. 2007;282:32582-32590. DOI: 10.1074/jbc.M702806200
  19. 19. Wang S, Bian C, Yang Z, Bo Y, Li J, Zeng L, et al. miR-145 inhibits breast cancer cell growth through RTKN. International Journal of Oncology. 2009;34:1461-1466. DOI: 10.3892/ijo_00000275
  20. 20. Sachdeva M, Yuan MY. MicroRNA-145 suppresses cell invasion and metastasis by directly targeting mucin 1. Cancer Research. 2010;70(1):378-387. DOI: 10.1158/0008-5472.CAN-09-2021
  21. 21. Larsson E, Fuchs PF, Heldin J, et al. Discovery of microvascular miRNAs using public gene expression data: miR-145 is expressed in pericytes and is a regulator of Fli1. Genome Medicine. 2009;1(11):108. DOI: 10.1186/gm108
  22. 22. Spizzo R, Nicoloso MS, Lupini L, et al. miR-145 participates with TP53 in a death-promoting regulatory loop and targets estrogen receptor-α in human breast cancer cells. Cell Death and Differentiation. 2010;17(2):246-254. DOI: 10.1038/cdd.2009.117
  23. 23. Chang TC, Wentzel EA, Kent OA, et al. Transactivation of miR-34a by p53 broadly influences gene expression and promotes apoptosis. Molecular Cell. 2007;26(5):745-752. DOI: 10.1016/j.molcel.2007.05.010
  24. 24. Slabáková E, Culig Z, Remšík J, Souček K. Alternative mechanisms of miR-34a regulation in cancer. Cell Death & Disease. 2017;8:e3100. DOI: 10.1038/cddis.2017.495
  25. 25. Liang Y, Ridzon D, Wong L, Chen C. Characterization of microRNA expression profiles in normal human tissues. BMC Genomics. 2007;8:166. DOI: 10.1186/1471-2164-8-166
  26. 26. Agostini M, Knight RA. miR-34: From bench to bedside. Oncotarget. 2014;5(4):872-881. DOI: 10.18632/oncotarget.1825
  27. 27. Bommer GT, Gerin I, Feng Y, et al. p53-mediated activation of miRNA34 candidate tumor-suppressor genes. Current Biology. 2007;17:1298-1307. DOI: 10.1016/j.cub.2007.06.068
  28. 28. Rokavec M, Li H, Jiang L, Hermeking H. The p53/miR-34 axis in development and disease. Journal of Molecular Cell Biology. 2014;6(3):214-230. DOI: 10.1093/jmcb/mju003
  29. 29. Vogt M, Munding J, Grüner M, et al. Frequent concomitant inactivation of miR-34a and miR-34b/c by CpG methylation in colorectal, pancreatic, mammary, ovarian, urothelial, and renal cell carcinomas and soft tissue sarcomas. Virchows Archiv. 2011;458(3):313-322. DOI: 10.1007/s00428-010-1030-5
  30. 30. Lodygin D, Tarasov V, Epanchintsev A, et al. Inactivation of miR-34a by aberrant CpG methylation in multiple types of cancer. Cell Cycle. 2008;7(16):2591-2600. DOI: 10.416/cc.7.16.6533
  31. 31. Welch C, Chen Y, Stallings RL. MicroRNA-34a functions as a potential tumor suppressor by inducing apoptosis in neuroblastoma cells. Oncogene. 2007;26(34):5017-5022. DOI: 10.1038/sj.onc.1210293
  32. 32. Ji Q , Hao X, Zhang M, et al. MicroRNA miR-34 inhibits human pancreatic cancer tumor-initiating cells. PLoS One. 2009;4:e6816. DOI: 10.1371/journal.pone.000681
  33. 33. Wiggins JF, Ruffino L, Kelnar K, et al. Development of a lung cancer therapeutic based on the tumor suppressor microRNA-34. Cancer Research. 2010;70(14):5923-5930. DOI: 10.1158/0008-5472.CAN-10-0655
  34. 34. Zarone MR, Misso G, Grimaldi A, et al. Evidence of novel miR-34a-based therapeutic approaches for multiple myeloma treatment. Scientific Reports. 2017;7:17949. DOI: 10.1038/s41598-017-18186-0
  35. 35. Bader AG. miR-34—A microRNA replacement therapy is headed to the clinic. Frontiers in Genetics. 2012;3:120. DOI: 10.3389/fgene.2012.00120
  36. 36. Beg MS, Brenner AJ, Sachdev J, et al. Phase I study of MRX34, a liposomal miR-34a mimic, administered twice weekly in patients with advanced solid tumors. Investigational New Drugs. 2017;35(2):180-188. DOI: 10.1007/s10637-016-0407-y
  37. 37. Li C, Sun J, Xiang Q , et al. Prognostic role of miRNA21 in expression in gliomas: A meta analysis. Journal of Neuro-Oncology. 2016;130(1):11-17. DOI: 10.1007/s11060-016-2233-7
  38. 38. Meng F, Henson R, Wehbe-Janek H, et al. MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer. Gastroenterology. 2007;133(2):647-658. DOI: 10.1053/j.gastro.2007.05.022
  39. 39. Asangani IA, Rasheed SAK, Nikolova DA. 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
  40. 40. Zhu S, Si ML, Wu H, Mo YY. MicroRNA-21 targets the tumor suppressor gene tropomyosin 1. The Journal of Biological Chemistry. 2007;282:14328-14336. DOI: 10.1074/jbc.M611393200
  41. 41. Medina PP, Nolde M, Slack FJ. OncomiR addiction in an in vivo model of microRNA-21-induced pre-B-cell lymphoma. Nature. 2010;467(7311):86-90. DOI: 10.1038/nature09284
  42. 42. Berindan-Neagoe I, Monroig Pdel C, Pasculli B, Calin GA. MicroRNAome genome: A treasure for cancer diagnosis and therapy. CA: A Cancer Journal for Clinicians. 2014;64(5):311-336. DOI: 10.3322/caac.21244
  43. 43. Lu J, Getz G, Miska EA. MicroRNA expression profiles classify human cancers. Nature. 2005;435:834-838. DOI: 10.1038/nature03702
  44. 44. Rosenfeld N, Aharonov R, Meiri E. MicroRNAs accurately identify cancer tissue origin. Nature Biotechnology. 2008;26(4):462-469. DOI: 10.1038/nbt1392
  45. 45. Liu C, Kelnar K, Vlassov AV. Distinct microRNA expression profiles in prostate cancer stem/progenitor cells and tumor-suppressive functions of let-7. Cancer Research. 2012;72(13):3393-3404. DOI: 10.1158/0008-5472.CAN-11-3864
  46. 46. Pritchard CC, Cheng HH, Tewari M. MicroRNA profiling: Approaches and considerations. Nature Reviews. Genetics. 2012;13(5):358-369. DOI: 10.1038/nrg3198
  47. 47. Miller KD, Siegel RS, Lin CC, et al. Cancer treatment and survivorship statistics, 2016. CA: A Cancer Journal for Clinicians. 2016;66:271-289. DOI: 10.3322/caac.21349
  48. 48. Müller MF, Ibrahim AE, Arends MJ. Molecular pathological classification of colorectal cancer. Virchows Archiv. 2016;469(2):125-134. DOI: 10.1007/s00428-016-1956-3
  49. 49. Yang L, Belaguli N, Berger DH. MicroRNA and colorectal cancer. World Journal of Surgery. 2009;33:638-646. DOI: 10.1007/s00268-008-9865-5
  50. 50. Michael MZ, O’Connor SM, van Holst Pellekaan NG, Young GP, James RJ. Reduced accumulation of specific microRNAs in colorectal neoplasia. Molecular Cancer Research. 2003;1:882-891. Online ISSN 1557-3125
  51. 51. Takahashi H, Takahashi M, Ohnuma S, et al. microRNA-193a-3p is specifically down-regulated and acts as a tumor suppressor in BRAF-mutated colorectal cancer. BMC Cancer. 2017;17:723. DOI: 10.1186/s12885-017-3739-x
  52. 52. Cekaite L, Eide PW, Lind GE, et al. MicroRNAs as growth regulators, their function and biomarker status in colorectal cancer. Oncotarget. 2016;7:6476-6505. DOI: 10.18632/oncotarget.6390
  53. 53. Danese E, Minicozzi AM, Benati M, et al. Reference miRNAs for colorectal cancer: Analysis and verification of current data. Scientific Reports. 2017;7:8413. DOI: 10.1038/s41598-017-08784-3
  54. 54. Luo X, Burwinkel B, Tao S, Brenner H. MicroRNA signatures: Novel biomarker for colorectal cancer? Cancer Epidemiology, Biomarkers & Prevention. 2011;20(7):1272-1286. DOI: 10.1158/1055-9965.EPI-11-0035
  55. 55. Schetter AJ, Okayama H, Harris CC. The role of microRNAs in colorectal cancer. Cancer Journal. 2012;18(3):244-252. DOI: 10.1097/PPO.0b013e318258b78f
  56. 56. Sovich JL, Sartor Z, Misra S. Developments in Screening Tests and Strategies for Colorectal Cancer. Biomedical Research International. 2015;2015:326728. DOI: 10.1155/2015/326728
  57. 57. Masuda T, Hayashi N, Kuroda Y, et al. MicroRNAs as biomarkers in colorectal cancer. Cancers (Basel). 2017;9(9):124. DOI: 10.3390/cancers9090124
  58. 58. Luo X, Stock C, Burwinkel B, Brenner H. Identification and evaluation of plasma microRNAs for early detection of colorectal cancer. PLoS One. 2013;8(5):e62880. DOI: 10.1371/journal.pone.0062880
  59. 59. Giráldez MD, Lozano JJ, Ramírez G, et al. Circulating microRNAs as biomarkers of colorectal cancer: Results from a genome-wide profiling and validation study. Clinical Gastroenterology and Hepatology. 2013;11(6):681-8.e3. DOI: 10.1016/j.cgh.2012.12.009
  60. 60. Ma Y, Zhang P, Yang J, et al. Candidate microRNA biomarkers in human colorectal cancer: Systematic review profiling studies and experimental validation. International Journal of Cancer. 2012;130:2077-2087. DOI: 10.1002/ijc.26232
  61. 61. Lindblom A. Different mechanisms in the tumorigenesis of proximal and distal colon cancers. Current Opinion in Oncology. 2001;13(1):63-69. PMID: 11148689
  62. 62. Earle JS, Luthra R, Romans A, Abraham R, Ensor J, Yao H, et al. Association of microRNA expression with microsatellite instability status in colorectal adenocarcinoma. The Journal of Molecular Diagnostics. 2010;12(4):433-440. DOI: 10.2353/jmoldx.2010.090154
  63. 63. Gatalica Z, Vranic S, Xiu J, Swensen J, Reddy S. High microsatellite instability (MSI-H) colorectal carcinoma: A brief review of predictive biomarkers in the era of personalized medicine. Familial Cancer. 2016;15(3):405-412. DOI: 10.1007/s10689-016-9884-6
  64. 64. Kloor M, Staffa L, Ahadova A, von Knebel Doeberitz M. Clinical significance of microsatellite instability in colorectal cancer. Langenbeck's Archives of Surgery. 2014;399(1):23-31. DOI: 0.1007/s00423-013-1112-3
  65. 65. Valeri N, Gasparini P, Fabbri M, et al. Modulation of mismatch repair and genomic stability by miR-155. Proceedings of the National Academy of Sciences of the United States of America. 2010;107(15):6982-6987. DOI: 10.1073/pnas.1002472107
  66. 66. Volinia S, Calin GA, Liu CG, et al. A microRNA expression signature of human solid tumors defines cancer gene targets. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(7):2257-2261. DOI: 10.1073/pnas.0510565103
  67. 67. Slattery ML, Herrick JS, Mullany LE, et al. Colorectal tumor molecular phenotype and miRNA: Expression profiles and prognosis. Mod Pathol. 2016;29(8):915-927. DOI: 10.1038/modpathol.2016.73
  68. 68. Moridikia A, Mirzaei H, Sahebkar A, Salimian J. MicroRNAs: Potential candidates for diagnosis and treatment of colorectal cancer. Journal of Cellular Physiology. 2018;233(2):901-913. DOI: 10.1002/jcp.25801
  69. 69. Nishida N, Yokobori T, Mimori K, Sudo T, Tanaka F, Shibata K, et al. MicroRNA miR-125b is a prognostic marker in human colorectal cancer. International Journal of Oncology. 2011;38:1437-1443. DOI: 10.3892/ijo.2011.969
  70. 70. Kim NH, Kim HS, Kim NG, Lee I, Choi HS, Li XY, et al. p53 and microRNA-34 are suppressors of canonical Wnt signaling. Science Signaling. 2011;4:ra71. DOI: 10.1126/scisignal.2001744
  71. 71. Siemens H, Jackstadt R, Hünten S, Kaller M, Menssen A, Götz U, et al. miR-34 and SNAIL form a double-negative feedback loop to regulate epithelial-mesenchymal transitions. Cell Cycle. 2011;10:4256-4271. DOI: 10.4161/cc.10.24.18552
  72. 72. Hiyoshi Y, Schetter AJ, Okayama H, Inamura K, Anami K, Nguyen GH, et al. Increased microRNA-34b and-34c predominantly expressed in stromal tissues is associated with poor prognosis in human colon cancer. PLoS One. 2015;10(4):e0124899. DOI: 10.1371/journal.pone.0124899
  73. 73. Kanaan Z, Rai SN, Eichenberger MR, Barnes C, Dworkin AM, Weller C, et al. Differential microRNA expression tracks neoplastic progression in inflammatory bowel disease-associated colorectal cancer. Human Mutation. 2012;33:551-560. DOI: 10.1002/humu.22021
  74. 74. Pu XX, Huang GL, Guo HQ , et al. Circulating miR-221 directly amplified from plasma is a potential diagnostic and prognostic marker of colorectal cancer and is correlated with p53 expression. Journal of Gastroenterology and Hepatology. 2010;25(10):1674-1680. DOI: 10.1111/j.1440-1746.2010.06417.x
  75. 75. Wang G, Cao X, Lai S, Luo X, Feng Y, Wu J, et al. Altered p53 regulation of miR-148b and p55PIK contributes to tumor progression in colorectal cancer. Oncogene. 2015;34:912-921. DOI: 10.1038/onc.2014.30
  76. 76. Meng WJ, Yang L, Ma Q, Zhang H, Adell G, Arbman G, et al. MicroRNA Expression Profile Reveals miR-17-92 and miR-143-145 Cluster in Synchronous Colorectal Cancer. Medicine (Baltimore). 2015;94(32):e1297. DOI: 10.1097/MD.0000000000001297
  77. 77. Pichler M, Winter E, Stotz M, et al. Down-regulation of KRAS-interacting miRNA-143 predicts poor prognosis but not response to EGFR-targeted agents in colorectal cancer. British Journal of Cancer. 2012;106:1826-1832. DOI: 10.1038/bjc.2012.175
  78. 78. Vickers MM, Bar J, Gorn-Hondermann I, Yarom N, Daneshmand M, Hanson JE, et al. Stage-dependent differential expression of microRNAs in colorectal cancer: Potential role as markers of metastatic disease. Clinical & Experimental Metastasis. 2012;29:123-132. DOI: 10.1007/s10585-011-9435-3
  79. 79. Kahlert C, Klupp F, Brand K, Lasitschka F, Diederichs S, Kirchberg J, et al. Invasion front-specific expression and prognostic significance of microRNA in colorectal liver metastases. Cancer Science. 2011;102:1799-1807. DOI: 10.1111/j.1349-7006.2011.02023.x
  80. 80. Hata T, Mokutani Y, Takahashi H, Inoue A, Munakata K, Nagata K, et al. Identification of microRNA-487b as a negative regulator of liver metastasis by regulation of KRAS in colorectal cancer. International Journal of Oncology. 2017;50(2):487-496. DOI: 10.3892/ijo.2016.3813
  81. 81. Cheng H, Zhang L, Cogdell DE, Zheng H, Schetter AJ, Nykter M, et al. Circulating plasma MiR-141 is a novel biomarker for metastatic colon cancer and predicts poor prognosis. PLoS One. 2011;6(3):e17745. DOI: 10.1371/journal.pone.0017745
  82. 82. Matsumura T, Sugimachi K, Iinuma H, Takahashi Y, Kurashige J, Sawada G, et al. Exosomal microRNA in serum is a novel biomarker of recurrence in human colorectal cancer. British Journal of Cancer. 2015;113(2):275-281. DOI: 10.1038/bjc.2015.201
  83. 83. Dong Y, Yu J, Ng SS. MicroRNA dysregulation as a prognostic biomarker in colorectal cancer. Cancer Management and Research. 2014;6:405-422. DOI: 10.2147/CMAR.S35164
  84. 84. Xie T, Huang M, Wang Y, Wang L, Chen C, Chu X. MicroRNAs as regulators, biomarkers and therapeutic targets in the drug resistance of colorectal cancer. Cellular Physiology and Biochemistry. 2016;40(1-2):62-76. DOI: 10.1159/000452525
  85. 85. Heneghan HM, Miller N, Kerin MJ. MiRNAs as biomarkers and therapeutic targets in cancer. Current Opinion in Pharmacology. 2010;10(5):543-550. DOI: 10.1016/j.coph.2010.05.010
  86. 86. An X, Sarmiento C, Tan T, Zhu H. Regulation of multidrug resistance by microRNAs in anti-cancer therapy. Acta Pharmaceutica Sinica B. 2017;7(1):38-51. DOI: 10.3389/fgene.2012.00180
  87. 87. Zhu Y, Peng Q , Lin Y, Zou L, Shen P, Chen F, et al. Identification of biomarker microRNAs for predicting the response of colorectal cancer to neoadjuvant chemoradiotherapy based on microRNA regulatory network. Oncotarget. 2017;8(2):2233. DOI: 10.18632/oncotarget.13659
  88. 88. Ramos P, Bentires-Alj M. Mechanism-based cancer therapy: Resistance to therapy, therapy for resistance. Oncogene. 2015;34(8):3617-3626. DOI: 10.1038/onc.2014.314
  89. 89. Hammond WA, Swaika A, Mody K. Pharmacologic resistance in colorectal cancer: A review. Therapeutic Advances in Medical Oncology. 2016;8(1):57-84. DOI: 10.1177/1758834015614530
  90. 90. Kiss I, Mlčochová J, Součková K, Fabian P, Poprach A, Halamkova J, et al. MicroRNAs as outcome predictors in patients with metastatic colorectal cancer treated with bevacizumab in combination with FOLFOX. Oncology Letters. 2017;14(1):743-750. DOI: 10.3892/ol.2017.6255
  91. 91. Kjersem JB, Ikdahl T, Lingjaerde OC, Guren T, Tveit KM, Kure EH. Plasma microRNAs predicting clinical outcome in metastatic colorectal cancer patients receiving first-line oxaliplatin-based treatment. Molecular Oncology. 2014;8(1):59-67. DOI: 10.1016/j.molonc.2013.09.001
  92. 92. Markman JL, Shiao SL. Impact of the immune system and immunotherapy in colorectal cancer. Journal of Gastrointestinal Oncology. 2015;6(2):208-223. DOI: 10.3978/j.issn.2078-6891.2014.077
  93. 93. Giannakis M, Mu XJ, Shukla SA, et al. Genomic correlates of immune-cell infiltrates in colorectal carcinoma. Cell Reports. 2016;15(4):857-865. DOI: 10.1016/j.celrep.2016.03.075
  94. 94. Cortez MA, Ivan C, Valdecanas D, Wang X, Peltier HJ, Ye Y, et al. PDL1 regulation by p53 via miR-34. Journal of National Cancer Institute. 2016;108:djv303. DOI: 10.1093/jnci/djv303
  95. 95. Wei J, Wang F, Kong LY, Xu S, Doucette T, Ferguson SD, et al. miR-124 inhibits STAT3 signaling to enhance T cell-mediated immune clearance of glioma. Cancer Research. 2013;73:3913-3926. DOI: 10.1158/0008-5472.CAN-12-4318
  96. 96. Yu T, Zuo QF, Gong L, Wang LN, Zou QM, Xiao B. MicroRNA-491 regulates the proliferation and apoptosis of CD8+ T cells. Scientific Reports. 2016;6:30923. DOI: 10.1038/srep30923
  97. 97. Ozasa H, Hazama S, Shimizu R, Etoh R, Inoue Y, Kanekiyo S, et al. miR-196b and miR-486 as predictive biomarkers for the efficacy of the vaccine treatment: From the results of phase I and II studies for metastatic colorectal cancer. Oncology Letters. 2017;14(2):1355-1362. DOI: 10.3892/ol.2017.6303
  98. 98. Kraemer A, Barjaktarovic Z, Sarioglu H, et al. Cell survival following radiation exposure requires miR-525-3p mediated suppression of ARRB1 and TXN1. PLoS One. 2013;8(10):e77484. DOI: 10.1371/journal.pone.0077484

Written By

Burcin Baran, Nazli-Mert Ozupek, Gizem Calibasi-Kocal and Yasemin Basbinar

Submitted: 05 May 2018 Reviewed: 09 August 2018 Published: 07 November 2018