Review Open Access
Copyright ©2014 Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Sep 7, 2014; 20(33): 11727-11735
Published online Sep 7, 2014. doi: 10.3748/wjg.v20.i33.11727
MicroRNAs as novel predictive biomarkers and therapeutic targets in colorectal cancer
Verena Stiegelbauer, Samantha Perakis, Armin Gerger, Division of Clinical Oncology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
Alexander Deutsch, Division of Hematology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
Hui Ling, Martin Pichler, Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
Author contributions: Stiegelbauer V designed research and wrote the paper; Perakis S designed research and wrote the paper; Deutsch A designed research and wrote the paper; Ling H designed research and wrote the paper; Gerger A designed research and wrote the paper; Pichler M designed research and wrote the paper.
Supported by Erwin Schroedinger Scholarship of the Austrian Science Funds, No. J3389-B23 (all to Pichler M)
Correspondence to: Martin Pichler, MD, Division of Clinical Oncology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria. MPichler@mdanderson.org
Telephone: +43-316-3853115 Fax: +43-316-3853115
Received: January 17, 2014
Revised: April 4, 2014
Accepted: June 2, 2014
Published online: September 7, 2014

Abstract

Colorectal cancer (CRC) is the third most common cancer in western countries. Despite significant improvement in available treatment options, CRC still remains the second leading cause of cancer-related death. Traditionally, 5-fluorouracil has been used as the main chemotherapy drug for treatment of metastatic CRC (mCRC). However, during the last two decades more effective chemotherapeutic agents such as oxaliplatin, irinotecan and the monoclonal antibodies cetuximab, panitumumab and bevacizumab have been used in clinical practice. More recently, the therapeutic armamentarium has been supplemented by the monoclonal antibodies bevacizumab, cetuximab and panitumumab as well as the protein-trap aflibercept and the small molecule multi-kinase inhibitor regorafenib. One of the major problems for the management of CRC is the inherent or acquired resistance to therapeutic approaches. The discovery of microRNAs (miRNAs), a class of small, endogenous, non-coding, single-stranded RNAs that play a role as post-transcriptional regulators, has added new dimensions to the diagnosis and treatment of cancer. Because miRNAs are important regulators of carcinogenesis, progression, invasion, angiogenesis and metastases in CRC, they might serve as potential predictive and prognostic factors and even as therapeutic targets themselves. Several miRNAs are already known to be dysregulated in CRCs and have been linked to biological processes involved in tumor progression and response to anti-cancer therapies. This review summarizes current therapeutic approaches for treating CRC and highlights the role of miRNAs as novel predictive biomarkers and potential drug targets in CRC patients.

Key Words: Colorectal cancer, MicroRNAs, 5-fluorouracil, Epidermal growth factor receptor, Targeted therapy

Core tip: In this review article, we summarize the status quo of the current literature regarding microRNAs and their role in resistance against anti-cancer drugs in colorectal cancer. This Review Article explains how microRNAs influence colorectal cancer, and how these small molecules might be useful as predictive factors and drug targets by themselves.



INTRODUCTION

Colorectal cancer (CRC) is the third most common cancer in western societies with about 1.2 million new cases and 608700 deaths estimated to have occurred in 2008[1]. Outcomes for patients with mCRC still remain poor, with an average survival of less than 30 mo[2]. The majority of CRC cases arise from dysplastic adenomatous polyps. The process of transformation includes a few essential events characterized by the activation of oncogenes such as KRAS (Kirsten rat sarcoma viral oncogene homolog), c-MYC (v-myc avian myelocytomatosis viral oncogene homolog) and NRAS (neuroblastoma RAS viral oncogene homolog) and by inactivation of tumor suppressor genes [e.g., p53 (tumor protein p53) and APC (adenomatous polyposis coli)] or DNA repair genes such as hMSH2 (human mutS homolog 2) or hMSLH1[3]. Although 5-fluorouracil (5-FU) has proven to be moderately effective in CRC as a monotherapy, its combination with the chemotherapeutic drugs oxaliplatin and irinotecan improved the therapeutic outcome. During the last few years, the combination of chemotherapeutic agents with more effective systemic agents such as bevacizumab, panitumumab or cetuximab has been established in clinical practice and improved survival rates in patients with CRC[4,5].

CURRENT COLORECTAL CANCER TREATMENT REGIMENS
5-fluorouracil

5-FU is one of the main chemotherapeutic drugs used for treatment of CRC[6,7]. It exhibits its cytotoxicity by incorporating fluoronucleotides into RNA and DNA molecules, but its main toxic effects are mediated by inhibiting the nucleotide synthetic enzyme thymidylate synthase (TYMS)[7]. However, one of the major problems in managing progressed colorectal cancer is both the inherent and acquired failure of 5-FU-based therapy. Several analyses of 5-FU resistance in CRC have focused on genes involved in pharmacodynamic and pharmacokinetic pathways. One main point has been the activity of TYMS, a key therapeutic target of 5-FU. CRCs that are resistant to 5-FU-based chemotherapy have been shown to possess greater TYMS enzymatic activity than cancers that are sensitive to this therapy[8]. High levels of TYMS mRNA or protein expression in liver metastases have also been linked to lack of clinical response to 5-FU in vivo[9]. Moreover, a meta-analysis of 24 studies demonstrated that low expression levels of TYMS in metastatic colorectal tumors are associated with greater sensitivity to fluoropyrimidine-based chemotherapy[10]. Additionally, the activity of thymidine phosphorylase (TP), a key enzyme that catalyzes the transformation from 5-FU prodrugs of 5′-deoxy-5-fluorouridine (5′-DFUR) to 5-FU, is closely linked to failure of 5-FU and targeted therapy in CRC cells[11].

EGFR inhibitors

The epidermal growth factor receptor (EGFR) belongs to the ErbB family of receptors that are able to promote tumor cell proliferation in diverse epithelial malignancies[12]. For this reason, EGFR has become an important target in oncology. Monoclonal antibodies against EGFR are used as a standard therapy for some types of solid tumors. The chimeric IgG1 mouse/human antibody cetuximab and the human IgG2 antibody panitumumab are considered to be equally effective in mCRC. However, it has been extensively reported that primary resistance to these agents is mediated by mutations in downstream signaling molecules[13]. Misale et al[14] showed that the development of resistance to anti-EGFR treatment in CRC is associated with molecular alterations of KRAS. Although KRAS mutations confer strong resistance to anti-EGFR antibodies, not all CRC patients with KRAS wildtype (KRASwt) benefit from these agents. Therefore, there is a need for novel biomarkers to better identify which KRASwt patients would benefit from this therapy[15]. A few studies have shown that high EGFR gene copy number could be a potential favorable marker for anti-EGFR therapy in CRC, as patients with low gene copy number are unlikely to respond to anti-EGFR agents[16,17]. Very recently, rare mutations in exons 3 and 4 in the both the KRAS gene and NRAS gene have demonstrated predictive potential in regards to panitumumab treatment[18].

VEGF targeted therapy

Angiogenesis is widely regarded as an important therapeutic target in many different types of cancer, including CRC[19]. It has been suggested that inhibition of angiogenesis in tumors can influence the development of new tumor blood vessels and probably lead to normalization of the existing tumor vasculature. The vascular endothelial growth factor (VEGF) is a pro-angiogenic factor that plays a key role in tumor vascular development[20]. VEGF-neutralizing antibodies can prevent the binding to and activation of VEGFR1 (vascular endothelial growth factor receptor 1), VEGFR2 (vascular endothelial growth factor receptor 2) and the co-receptors NP1 (Neuropilin 1) and NP2 (Neuropilin 2). Bevacizumab, a humanized monoclonal antibody that binds to all isoforms of VEGF, is the first anti-angiogenic therapy approved for treatment of CRC[21,22]. It has also been used in combination with common chemotherapeutic agents, such as 5-FU and capecitabine as well as irinotecan and oxaliplatin[23]. Resistance to combinatorial therapy in CRC treatment may be due to resistance to bevacizumab, resistance to the chemotherapeutic with which bevacizumab was administered or a resistance to both. As observed, several pathways in addition to the VEGF pathway are involved in angiogenesis. Therefore, mechanisms of resistance to anti-angiogenic therapy may also include VEGF-independent anti-angiogenesis pathways[24].

MIRNA BIOGENESIS AND FUNCTION

MiRNAs are a class of small, endogenous, non-coding, single-stranded RNAs that play a role as post-transcriptional regulators by suppressing the translation or inducing the mRNA degradation of their target genes[25]. Dysregulated miRNA expression in human cancers plays a role in carcinogenesis mainly by regulating their mRNA targets, which could be oncogenes or tumor suppressors[26]. Various combinations of miRNAs are expressed in different cell types and regulate cell-specific target genes. MiRNAs regulate about one-third of all human genes and a single miRNA can target around 200 or more transcripts that are key regulators of multiple signaling pathways in the cell[27,28]. Aberrational miRNA expression patterns, commonly seen in human diseases including various types of cancer, can serve as prognostic factors and may have implications for cancer stem cell regulation[29-32]. More than 50% of known human miRNA genes are localized in fragile chromosomal regions that are susceptible to amplification, deletion or translocation during cancer development[33].

MiRNAs are transcribed as long primary transcripts called pri-miRNAs and are processed into precursor miRNAs (pre-miRNAs) in the nucleus by the enzyme Drosha. These short hairpin RNAs of approximately 70 nt are then translocated to the cytoplasm where they are cleaved by the enzyme Dicer to generate the miRNA duplex[34,35]. Afterwards, the miRNA is unwound by a helicase and one of the strands is defined as the mature strand while the other one is quickly degraded. The mature miRNA is incorporated into an RNA-induced silencing complex (RISC) that mediates gene silencing[36].

MiRNAs are important regulators of oncogenesis, progression, invasion, angiogenesis and metastasis in colorectal cancer. Both upregulation and downregulation have been linked to carcinogenesis in CRC[37]. Many proteins that play a role in key signaling pathways of CRC seem to be influenced by miRNA regulation, such as members of the Wnt/beta-catenin and phosphatidylinositol-3-kinase (PI-3-K) pathways, KRAS, p53, extracellular matrix regulators as well as epithelial-mesenchymal transition (EMT) proteins and transcription factors[26,38,39].

New findings suggest that miRNAs could be linked to sensitivity to chemotherapeutic drugs in tumor cells. For this reason, researchers are interested in the potential role of miRNAs in pharmacogenomics[40]. A recent study by Pardini et al[41] provided evidence that a modulation of the expression of base excision repair (BER) genes, such as a post-transcriptional change caused by microRNAs, could influence the efficiency of this repair system. Single-nucleotide polymorphisms (SNP) within the 3′-untranslated regions (UTR) of target genes could lead to an alteration in binding of specific miRNAs that modulate gene expression. Such changes could affect cancer prognosis and therapy outcomes. Hence, characterization of polymorphisms in miRNA-related genes or target sites might afford a basis for miRNA-based therapy approaches.

In the next few pages, we will discuss particular mi-RNAs which have been experimentally proven to play a role in drug resistance in the past few years.

MIRNAS AND THEIR INVOLVEMENT IN RESISTANCE TO ANTI-CANCER THERAPIES
Let-7

Clinical trials have demonstrated that KRAS mutations are negative predictive biomarkers for EGFR-targeted therapy in CRC. Mechanisms of post-transcriptional downregulation of mutated KRAS might therefore be of clinical relevance in patients with mCRC[42,43]. The members of the let-7 family are known to target KRAS and their involvement in response to EGFR-targeted therapy has already been reported[44]. Upregulation of let-7 expression levels might provide a survival advantage by inhibiting mutated KRAS under EGFR-targeted therapy. In addition, let-7 may reveal further favorable effects by regulating other genes such as the cell cycle regulators, Myc, Bcl-2 (B-cell CLL/lymphoma 2), integrins and signal transducers. Ruzzo et al[45] analyzed the expression levels of let-7a in colorectal carcinomas with mutated KRAS and in mCRC patients treated with cetuximab and irinotecan. Their study revealed that intra-tumor expression of let-7a correlates with tumor response and overall survival in mCRC patients treated with anti-EGFR-based therapy in both KRAS mutant and wildtype populations. Moreover, they showed that downregulation of let-7e and let-7b can potentially be used to predict resistance to the monoclonal antibody cetuximab. Cappuzzo et al[46] investigated whether microRNAs can predict sensitivity to EGFR-targeting monoclonal antibodies in patients with mCRC. They identified the cluster Let-7c/miR-99a/miR-125b as a signature linked to an outcome different from that of EGFR targeting therapies. In the first cohort, patients with high-intensity signatures showed a significantly longer progression-free survival and longer overall survival. Moreover, in the KRAS wild-type population, high-intensity signature patients had a significantly longer progression-free survival, as demonstrated in the validation cohort. Therefore, the miR-99a/let-7c/miR-125b signature could improve the selection of patients with KRAS wild-type mCRC for treatment with EGFR targeting therapies.

Salendo et al[47] performed a genome-wide miRNA profiling in 12 colorectal cancer cell lines and established an individual in vitro signature for chemoradiosensitivity. Their study demonstrated that high expression of let-7g was linked with a good prognosis in rectal cancer patients. This finding suggests that let7g expression may serve as potential predictive biomarker.

MiR-126

An increasing number of studies proposed miR-126 as a player in the regulation of angiogenesis, a process that has already been considered as a target for development of novel drugs. High expression of miR-126 has already been associated with increased vascular endothelial growth factor A (VEGF-A) mediated signaling in endothelial cells and a higher blood vessel integrity[48]. Additionally, miR-126 has been identified as a putative tumor suppressor in primary tumors[49-51]. Hansen et al[48] investigated the role of miR-126 as a predictive marker in patients with CRC in relation to first-line treatment with capecitabine, a precursor of 5-FU, and oxaliplatin (XELOX). They demonstrated a significant relationship between expression of miR-126 in the primary tumor and sensitivity to first-line XELOX treatment. Low expression of miR-126 might therefore lead to tumor vessels with decreased integrity followed by an increase in interstitial pressure, which may explain the lower sensitivity in patients treated with XELOX. A recent study by Hansen et al[52] revealed that high expression of miR-126 in mCRC patients was strongly related to a longer progression-free survival. Their results confirm their previous findings on the prognostic value of miR-126 in mCRC. As VEGF-A may be a target of miR-126, the results of their study might provide predictive information in regards to next-generation anti-angiogenic therapy approaches.

MiR-31

Several studies revealed that miR-31 is upregulated in CRC, but there is little known about its role in modulating tumor cell response to chemotherapeutic drugs. Wang et al[53] showed that the treatment of HCT-116 colon cancer cells with an anti-miR-31 inhibitor increased the sensitivity to 5-FU as early as 24 hours after exposure without affecting cell proliferation. Combination of 5-FU treatment with a respective negative control did not lead to a reduction in cell viability. The apoptosis rate of HCT-116 cells treated with both 5-FU and the anti-miR-31 inhibitor was the highest among respective control groups, indicating that these agents inhibited proliferation via the apoptotic mechanism.

MiR-192/miR-215

The expression levels of miR-192 and -215 were shown to be significantly reduced in CRC tissues compared to non-tumor counterparts. Furthermore, Chiang et al[54] demonstrated that low expression levels of miR-192 and -215 are related to increased tumor size in CRC. Thus, miR-129 and miR-215 could be useful biomarkers in the carcinogenesis of CRC. In addition, miR-192 and miR-215 were reported to negatively regulate CRC cell proliferation[55]. Boni et al[40] showed that miR-192 and -215 downregulate TYMS expression and thus increase resistance to 5-FU in CRC cell lines. TYMS plays a role in normal cell function and is a potential target for chemotherapeutic drugs such as 5-FU. Transcriptional and translational regulation of TYMS most likely affect cell chemosensitivity. Additionally, they demonstrated that miR-192 and miR-215 inhibit progression into the S phase, play a role in cell cycle control and prevent sensitivity to 5-FU. Zhang et al[56] identified a set of 6 miRNAs including miR-215 which could serve as an authentic prognostic and predictive tool for determination of disease recurrence in patients with stage II colon cancer. miR-215 could potentially predict which patients would benefit from adjuvant chemotherapy, which can in turn facilitate patient consultation and help individualize management of patients with this disease.

MiR-148a

It has been shown that miR-148a is a pro-apoptotic miRNA in CRC which acts by targeting Bcl-2, a regulator of apoptosis. In addition, several in vitro studies have demonstrated that miR-148a functions as a tumor suppressor by targeting several genes such as PXR (nuclear receptor subfamily 1, group I, member 2), TGIF2 (TGFB-induced factor homeobox 2), MSX1 (msh homeobox 1), CDC25B (cell division cycle 25B), DNMT1 [DNA (cytosine-5-methyltransferase 1)] and DNMT3b [DNA (cytosine-5-methyltransferase 3 beta)][57]. Takahashi et al[58] showed that low expression of miR-148a is significantly linked to an unfavorable outcome in treatment of stage III CRC patients with 5-FU. They also demonstrated the link between decreased miR-148a expression and poorer sensitivity and survival rate in patients with stage IV CRC treated with oxaliplatin combined with 5-FU. Stage IV CRC patients that showed a high miR-148a expression level were shown to benefit from chemotherapeutic drugs, indicating that miR-148a may have predictive value in the assessment of response to CRC treatment. In addition, their data suggested that downregulation of miR-148a is mediated by DNA methylation. Takahashi et al[58] showed that there is a significant and independent association between miR-148a methylation and poor survival in stage IV patients. Therefore, the methylation status of miR-148a might be a potential prognostic marker in CRC.

Kjersem et al[59] investigated microRNAs in plasma as potential predictive markers for sensitivity to oxaliplatin-based chemotherapy. Their study revealed that a high expression level of miR-148a is associated with a decrease in progression-free survival. The results of their study suggest that miR-148 could serve as a non-invasive biomarker for predicting outcomes in mCRC patients treated with 5-FU and oxaliplatin-based chemotherapy.

MiR-21

Upregulated miR-21 expression is associated with some types of human cancer, including CRC. It has been demonstrated that miR-21 regulates several tumor suppressors such as p21, TGF-beta receptor II and B-cell leukemia/lymphoma 2-associated X protein. Moreover, overexpression of miR-21 is linked to poor response to 5-FU-based chemotherapy. Valeri et al[60] found an inverse expressional correlation of miR-21 and the tumor suppressor hMSH2. They demonstrated that miR-21 directly targets the 3’ UTR of hMSH2 mRNA and significantly reduces its protein expression. CRC cells that showed high expression levels of miR-21 have decreased hMSH2 protein expression and revealed significantly reduced 5-FU-induced G2/M damage arrest and apoptosis. This indicates a characteristic defect in the core mismatch repair system, suggesting that miR-21 might act as a regulator of genes associated with cell-cycle arrest and/or drug resistance. In addition, Deng et al[61] showed that forced miR-21 expression in HT-29 colon cancer cells significantly inhibited apoptosis, enhanced cell proliferation and invasion and increased resistance to the chemotherapeutic agent 5-FU. Moreover, they demonstrated that silencing of miR-21 inverted these effects on HT-29 cells and restored the sensitivity to 5-FU.

MiR-129

Reduced miR-129 expression levels have been reported in several tumor cell lines and primary tumors including CRC[62]. Karaayvaz et al[63] demonstrated that miR-129 expression was clearly lower in CRC patients. There was no significant difference between adenoma and stage I and II carcinomas. They observed that miR-129 expression was dramatically reduced in stage III and IV cancers. Hence, their results suggested that loss of miR-129 is linked to progression in CRC. Additionally, they identified miR-129 as a novel regulator of Bcl-2 expression. The expression of miR-129 promoted apoptosis, inhibited cell proliferation and caused cell-cycle arrest in CRC cells. miR-129-based therapies could help to achieve a multi-targeted anti-cancer therapeutic strategy. 5-FU-based chemotherapy still remains the main option for advanced mCRC treatment, but the possibility of miR-129 restoration may lead to a new strategy in reducing resistance to chemotherapeutic drugs. Furthermore, Karaayvaz et al[63] showed that miR-129 is a suppressor of the 5-FU target protein TYMS and therefore enhances chemosensitivity to 5-FU. In their in vivo tumor xenografts, they demonstrated that increasing miR-129 expression to normal levels using synthetic miRNAs made tumors more sensitive to 5-FU-based drugs.

MiR-19b

MiR-19b is one of the 6 miRNAs which are encoded by the miR-17-92 cluster. The activation of this cluster is mediated by the transcription factor E2F1 which accumulates early in the G1 phase of the cell cycle, suggesting that miRNAs generated from this cluster might play a role in the G1 phase. Kurokawa and collegues found that miR-19b expression is upregulated in the DLD-1/R colon cancer cell line, but they did not observe any changes in the cell cycle profile after 5-FU treatment. In addition, they demonstrated that miR-19b expression correlated with response to the widely-used anti-cancer drug 5-FU[64]. Likewise, a recent study showed that miR-19a, a paralogue of miR-19b, is upregulated in HCT-119 and HT29 cells in response to 5-FU-based treatment[65].

MiR-34a

MiR-34a is a member of the miR-34 family which also includes miR-34b and miR-34c. Low expression levels of miR-34a have been identified in various types of tumors, including CRC[66]. Moreover, miR-34a was observed as one of the most downregulated miRNAs in the 5-FU-resistant DLD-1 CRC cell line. Akao et al[67] showed that exposure to 5-FU activated the PI3K/Akt signaling pathway in the resistant DLD-1 cell line in comparison to the parental cells and led to an apparent increase in growth. In addition, miR-34a expression in the 5-FU-resistant cell line was prolonged at a low level, whereas it showed an upregulation in the parental cells after treatment with a 5-FU-based drug. They observed an upregulation of Sirt-1, a target gene of miR-34a which is associated with drug resistance, in the 5-FU-resistant cells and also that silencing of Sirt-1 significantly increased the sensitivity to 5-FU in the 5-FU-resistant cells. This suggested that miR-34a is a negative regulator of 5-FU resistance in the CRC cell line DLD-1.

MiR-143

A few studies showed that miR-143 expression is low in tumors compared to their normal counterparts, both at adenomatous and cancer stages of colorectal neoplasia, as well as in CRC cell lines[68]. In addition, miR-143 has been identified to directly target the KRAS mRNA[69]. Borralho et al[70] investigated the role of miR-143 in the HCT116 CRC cell line. They showed that transient overexpression of miR-143 led to a 60% reduction in cell viability and stable overexpression of miR-143 resulted in decreased viability and increased cell death after treatment with 5-FU. These changes were linked to increased nuclear fragmentation and caspase -3, -8 and -9 activities. Furthermore, they demonstrated that exposure of miR-143-overexpressing cells to 5-FU resulted in downregulation of the extracellular-regulated protein kinase 5 and Bcl-2 protein expression. In addition, miR-143 led to increased sensitivity to 5-FU- based drugs, suggesting that miR-143 is involved in CRC development and plays a role as a chemosensitizer to 5-FU.

MiR-203

Zhou et al[71] recently showed that miR-203 is upregulated in three oxaliplatin-resistant CRC cell lines. They demonstrated that exogenous expression of miR-203 in chemonaïve CRC cells reduced sensitivity of cells to oxaliplatin treatment. Silencing of miR-203 expression led to increased sensitivity of CRC cells to oxaliplatin. Furthermore, they performed an in-silico analysis and identified ataxia telangiectasia mutated (ATM), a primary mediator of the DNA damage response, as a potential target of miR-203. Their study showed that mutation of the putative miR-203 target region in the 3’ untranslated region of ATM mRNA eliminated the inhibitory effect of miR-203 on ATM. In addition, they demonstrated that stable knockdown of ATM led to resistance to oxaliplatin in chemo-naïve CRC cells.

MiR-200c

Hur et al[72] reported that miR-200c expression modulates EMT in colorectal metastasis. In their study, they demonstrated for the first time that the miR-200c/429 cluster was significantly overexpressed in liver metastasis in comparison to primary CRC and that the expression of these miRNAs was specifically regulated by aberrant methylation of their promoter regions. A recent study by Toiyama et al[73] revealed that the serum levels of miR-200c might serve as potential prognostic and metastatic-predictive biomarkers in CRC patients. They showed that miR-200c expression in serum is strongly associated with a metastatic phenotype in CRC; in particular, expression of miR-200c in serum was a good predictive marker for lymph node metastasis. Moreover, they demonstrated that miR-200c expression in serum can be used as a prognostic and predictive marker of tumor recurrence in patients undergoing curative surgery.

CONCLUSION

Currently, therapeutic agents including 5-FU, oxaliplatin, irinotecan, bevacizumab cetuximab, panitumumab, aflibercept and regorafenib are widely used in clinical practice for CRC treatment. However, many patients are resistant or develop secondary resistance to these agents, highlighting the necessity for the development of novel prognostic markers for drug resistance. As important regulators of gene expression, miRNAs possess high potential as predictive markers for therapeutic response to chemotherapeutic drugs. On the other hand, targeting miRNAs that are involved in the resistance mechanism may improve the therapeutic efficacy in chemo-resistant patients. We foresee that in the near future, miRNA-based prognostic tools could be developed to aid in patient selection for certain treatments, and miRNA-based therapeutics may finally reach the clinical stages.

Footnotes

P- Reviewer: Fang Z, Liu XF S- Editor: Ma YJ L- Editor: A E- Editor: Wang CH

References
1.  Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin. 2011;61:69-90.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Scartozzi M, Giampieri R, Del Prete M, Faloppi L, Bianconi M, Vincenzi B, Tonini G, Santini D, Cascinu S. Selected gastrointestinal cancer presentations from the American Society of Clinical Oncology annual meeting 2013 in review: it is not about the destination, it is about the journey. Expert Opin Pharmacother. 2014;15:143-150.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Gout S, Huot J. Role of cancer microenvironment in metastasis: focus on colon cancer. Cancer Microenviron. 2008;1:69-83.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  André T, Boni C, Navarro M, Tabernero J, Hickish T, Topham C, Bonetti A, Clingan P, Bridgewater J, Rivera F. Improved overall survival with oxaliplatin, fluorouracil, and leucovorin as adjuvant treatment in stage II or III colon cancer in the MOSAIC trial. J Clin Oncol. 2009;27:3109-3116.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Papadimitriou CA, Papakostas P, Karina M, Malettou L, Dimopoulos MA, Pentheroudakis G, Samantas E, Bamias A, Miliaras D, Basdanis G. A randomized phase III trial of adjuvant chemotherapy with irinotecan, leucovorin and fluorouracil versus leucovorin and fluorouracil for stage II and III colon cancer: a Hellenic Cooperative Oncology Group study. BMC Med. 2011;9:10.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Dallas NA, Xia L, Fan F, Gray MJ, Gaur P, van Buren G, Samuel S, Kim MP, Lim SJ, Ellis LM. Chemoresistant colorectal cancer cells, the cancer stem cell phenotype, and increased sensitivity to insulin-like growth factor-I receptor inhibition. Cancer Res. 2009;69:1951-1957.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  de la Cueva A, Ramírez de Molina A, Alvarez-Ayerza N, Ramos MA, Cebrián A, Del Pulgar TG, Lacal JC. Combined 5-FU and ChoKα inhibitors as a new alternative therapy of colorectal cancer: evidence in human tumor-derived cell lines and mouse xenografts. PLoS One. 2013;8:e64961.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Etienne MC, Chazal M, Laurent-Puig P, Magné N, Rosty C, Formento JL, Francoual M, Formento P, Renée N, Chamorey E. Prognostic value of tumoral thymidylate synthase and p53 in metastatic colorectal cancer patients receiving fluorouracil-based chemotherapy: phenotypic and genotypic analyses. J Clin Oncol. 2002;20:2832-2843.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Watson RG, Muhale F, Thorne LB, Yu J, O’Neil BH, Hoskins JM, Meyers MO, Deal AM, Ibrahim JG, Hudson ML. Amplification of thymidylate synthetase in metastatic colorectal cancer patients pretreated with 5-fluorouracil-based chemotherapy. Eur J Cancer. 2010;46:3358-3364.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Qiu LX, Tang QY, Bai JL, Qian XP, Li RT, Liu BR, Zheng MH. Predictive value of thymidylate synthase expression in advanced colorectal cancer patients receiving fluoropyrimidine-based chemotherapy: evidence from 24 studies. Int J Cancer. 2008;123:2384-2389.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Ye DJ, Zhang JM. Research development of the relationship between thymidine phosphorylase expression and colorectal carcinoma. Cancer Biol Med. 2013;10:10-15.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Chang DZ, Kumar V, Ma Y, Li K, Kopetz S. Individualized therapies in colorectal cancer: KRAS as a marker for response to EGFR-targeted therapy. J Hematol Oncol. 2009;2:18.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Voigt M, Braig F, Göthel M, Schulte A, Lamszus K, Bokemeyer C, Binder M. Functional dissection of the epidermal growth factor receptor epitopes targeted by panitumumab and cetuximab. Neoplasia. 2012;14:1023-1031.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Misale S, Yaeger R, Hobor S, Scala E, Janakiraman M, Liska D, Valtorta E, Schiavo R, Buscarino M, Siravegna G. Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature. 2012;486:532-536.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Jiang Z, Li C, Li F, Wang X. EGFR gene copy number as a prognostic marker in colorectal cancer patients treated with cetuximab or panitumumab: a systematic review and meta analysis. PLoS One. 2013;8:e56205.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Moroni M, Veronese S, Benvenuti S, Marrapese G, Sartore-Bianchi A, Di Nicolantonio F, Gambacorta M, Siena S, Bardelli A. Gene copy number for epidermal growth factor receptor (EGFR) and clinical response to antiEGFR treatment in colorectal cancer: a cohort study. Lancet Oncol. 2005;6:279-286.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Personeni N, Fieuws S, Piessevaux H, De Hertogh G, De Schutter J, Biesmans B, De Roock W, Capoen A, Debiec-Rychter M, Van Laethem JL. Clinical usefulness of EGFR gene copy number as a predictive marker in colorectal cancer patients treated with cetuximab: a fluorescent in situ hybridization study. Clin Cancer Res. 2008;14:5869-5876.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Douillard JY, Oliner KS, Siena S, Tabernero J, Burkes R, Barugel M, Humblet Y, Bodoky G, Cunningham D, Jassem J. Panitumumab-FOLFOX4 treatment and RAS mutations in colorectal cancer. N Engl J Med. 2013;369:1023-1034.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Grothey A, Allegra C. Antiangiogenesis therapy in the treatment of metastatic colorectal cancer. Ther Adv Med Oncol. 2012;4:301-319.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Kerbel RS. Tumor angiogenesis. N Engl J Med. 2008;358:2039-2049.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Ellis LM, Hicklin DJ. VEGF-targeted therapy: mechanisms of anti-tumour activity. Nat Rev Cancer. 2008;8:579-591.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Saif MW. Anti-VEGF agents in metastatic colorectal cancer (mCRC): are they all alike? Cancer Manag Res. 2013;5:103-115.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Van Cutsem E, Tabernero J, Lakomy R, Prausova J, Ruff P, Van Hazel G. Intravenous (IV) aflibercept versus placebo in combination with irinotecan/5-FU (FOLFIRI) for second-line treatment of metastatic colorectal cancer (mCRC): Results of a multinational phase III trial (EFC10262-VELOUR). Ann Oncol. 2011;22 suppl 5:18.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Tejpar S, Prenen H, Mazzone M. Overcoming resistance to antiangiogenic therapies. Oncologist. 2012;17:1039-1050.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116:281-297.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Slaby O, Svoboda M, Michalek J, Vyzula R. MicroRNAs in colorectal cancer: translation of molecular biology into clinical application. Mol Cancer. 2009;8:102.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Erson AE, Petty EM. MicroRNAs in development and disease. Clin Genet. 2008;74:296-306.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Krek A, Grün D, Poy MN, Wolf R, Rosenberg L, Epstein EJ, MacMenamin P, da Piedade I, Gunsalus KC, Stoffel M. Combinatorial microRNA target predictions. Nat Genet. 2005;37:495-500.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Iorio MV, Ferracin M, Liu CG, Veronese A, Spizzo R, Sabbioni S, Magri E, Pedriali M, Fabbri M, Campiglio M. MicroRNA gene expression deregulation in human breast cancer. Cancer Res. 2005;65:7065-7070.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Schwarzenbacher D, Balic M, Pichler M. The role of microRNAs in breast cancer stem cells. Int J Mol Sci. 2013;14:14712-14723.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Bach D, Fuereder J, Karbiener M, Scheideler M, Ress AL, Neureiter D, Kemmerling R, Dietze O, Wiederstein M, Berr F. Comprehensive analysis of alterations in the miRNome in response to photodynamic treatment. J Photochem Photobiol B. 2013;120:74-81.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Al-Ali BM, Ress AL, Gerger A, Pichler M. MicroRNAs in renal cell carcinoma: implications for pathogenesis, diagnosis, prognosis and therapy. Anticancer Res. 2012;32:3727-3732.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Ma L, Teruya-Feldstein J, Weinberg RA. Tumour invasion and metastasis initiated by microRNA-10b in breast cancer. Nature. 2007;449:682-688.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Chen L, Yan HX, Yang W, Hu L, Yu LX, Liu Q, Li L, Huang DD, Ding J, Shen F. The role of microRNA expression pattern in human intrahepatic cholangiocarcinoma. J Hepatol. 2009;50:358-369.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Denli AM, Tops BB, Plasterk RH, Ketting RF, Hannon GJ. Processing of primary microRNAs by the Microprocessor complex. Nature. 2004;432:231-235.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Sun W, Julie Li YS, Huang HD, Shyy JY, Chien S. microRNA: a master regulator of cellular processes for bioengineering systems. Annu Rev Biomed Eng. 2010;12:1-27.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Rossi S, Kopetz S, Davuluri R, Hamilton SR, Calin GA. MicroRNAs, ultraconserved genes and colorectal cancers. Int J Biochem Cell Biol. 2010;42:1291-1297.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Pichler M, Winter E, Stotz M, Eberhard K, Samonigg H, Lax S, Hoefler G. Down-regulation of KRAS-interacting miRNA-143 predicts poor prognosis but not response to EGFR-targeted agents in colorectal cancer. Br J Cancer. 2012;106:1826-1832.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Pichler M, Winter E, Ress AL, Bauernhofer T, Gerger A, Kiesslich T, Lax S, Samonigg H, Hoefler G. miR-181a is associated with poor clinical outcome in patients with colorectal cancer treated with EGFR inhibitor. J Clin Pathol. 2014;67:198-203.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Boni V, Bitarte N, Cristobal I, Zarate R, Rodriguez J, Maiello E, Garcia-Foncillas J, Bandres E. miR-192/miR-215 influence 5-fluorouracil resistance through cell cycle-mediated mechanisms complementary to its post-transcriptional thymidilate synthase regulation. Mol Cancer Ther. 2010;9:2265-2275.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Pardini B, Rosa F, Barone E, Di Gaetano C, Slyskova J, Novotny J, Levy M, Garritano S, Vodickova L, Buchler T. Variation within 3’-UTRs of base excision repair genes and response to therapy in colorectal cancer patients: A potential modulation of microRNAs binding. Clin Cancer Res. 2013;19:6044-6056.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Amado RG, Wolf M, Peeters M, Van Cutsem E, Siena S, Freeman DJ, Juan T, Sikorski R, Suggs S, Radinsky R. Wild-type KRAS is required for panitumumab efficacy in patients with metastatic colorectal cancer. J Clin Oncol. 2008;26:1626-1634.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Lièvre A, Bachet JB, Boige V, Cayre A, Le Corre D, Buc E, Ychou M, Bouché O, Landi B, Louvet C. KRAS mutations as an independent prognostic factor in patients with advanced colorectal cancer treated with cetuximab. J Clin Oncol. 2008;26:374-379.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Ragusa M, Majorana A, Statello L, Maugeri M, Salito L, Barbagallo D, Guglielmino MR, Duro LR, Angelica R, Caltabiano R. Specific alterations of microRNA transcriptome and global network structure in colorectal carcinoma after cetuximab treatment. Mol Cancer Ther. 2010;9:3396-3409.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Ruzzo A, Graziano F, Vincenzi B, Canestrari E, Perrone G, Galluccio N, Catalano V, Loupakis F, Rabitti C, Santini D. High let-7a microRNA levels in KRAS-mutated colorectal carcinomas may rescue anti-EGFR therapy effects in patients with chemotherapy-refractory metastatic disease. Oncologist. 2012;17:823-829.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Cappuzzo F, Sacconi A, Landi L, Ludovini V, Biagioni F, D’Incecco A, Capodanno A, Salvini J, Corgna E, Cupini S. MicroRNA signature in metastatic colorectal cancer patients treated with anti-EGFR monoclonal antibodies. Clin Colorectal Cancer. 2014;13:37-45.e4.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Salendo J, Spitzner M, Kramer F, Zhang X, Jo P, Wolff HA, Kitz J, Kaulfuß S, Beißbarth T, Dobbelstein M. Identification of a microRNA expression signature for chemoradiosensitivity of colorectal cancer cells, involving miRNAs-320a, -224, -132 and let7g. Radiother Oncol. 2013;108:451-457.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Hansen TF, Sørensen 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.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  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;47:939-946.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Tavazoie SF, Alarcón C, Oskarsson T, Padua D, Wang Q, Bos PD, Gerald WL, Massagué J. Endogenous human microRNAs that suppress breast cancer metastasis. Nature. 2008;451:147-152.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Crawford M, Brawner E, Batte K, Yu L, Hunter MG, Otterson GA, Nuovo G, Marsh CB, Nana-Sinkam SP. MicroRNA-126 inhibits invasion in non-small cell lung carcinoma cell lines. Biochem Biophys Res Commun. 2008;373:607-612.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Hansen TF, Christensen Rd, Andersen RF, Sørensen FB, Johnsson A, Jakobsen A. MicroRNA-126 and epidermal growth factor-like domain 7-an angiogenic couple of importance in metastatic colorectal cancer. Results from the Nordic ACT trial. Br J Cancer. 2013;109:1243-1251.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Wang CJ, Stratmann J, Zhou ZG, Sun XF. Suppression of microRNA-31 increases sensitivity to 5-FU at an early stage, and affects cell migration and invasion in HCT-116 colon cancer cells. BMC Cancer. 2010;10:616.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Chiang Y, Song Y, Wang Z, Liu Z, Gao P, Liang J, Zhu J, Xing C, Xu H. microRNA-192, -194 and -215 are frequently downregulated in colorectal cancer. Exp Ther Med. 2012;3:560-566.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Georges SA, Biery MC, Kim SY, Schelter JM, Guo J, Chang AN, Jackson AL, Carleton MO, Linsley PS, Cleary MA. Coordinated regulation of cell cycle transcripts by p53-Inducible microRNAs, miR-192 and miR-215. Cancer Res. 2008;68:10105-10112.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Zhang JX, Song W, Chen ZH, Wei JH, Liao YJ, Lei J, Hu M, Chen GZ, Liao B, Lu J. Prognostic and predictive value of a microRNA signature in stage II colon cancer: a microRNA expression analysis. Lancet Oncol. 2013;14:1295-1306.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Zhang H, Li Y, Huang Q, Ren X, Hu H, Sheng H, Lai M. MiR-148a promotes apoptosis by targeting Bcl-2 in colorectal cancer. Cell Death Differ. 2011;18:1702-1710.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Takahashi M, Cuatrecasas M, Balaguer F, Hur K, Toiyama Y, Castells A, Boland CR, Goel A. The clinical significance of MiR-148a as a predictive biomarker in patients with advanced colorectal cancer. PLoS One. 2012;7:e46684.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  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. Mol Oncol. 2014;8:59-67.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Valeri N, Gasparini P, Braconi C, Paone A, Lovat F, Fabbri M, Sumani KM, Alder H, Amadori D, Patel T. MicroRNA-21 induces resistance to 5-fluorouracil by down-regulating human DNA MutS homolog 2 (hMSH2). Proc Natl Acad Sci USA. 2010;107:21098-21103.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Deng J, Lei W, Fu JC, Zhang L, Li JH, Xiong JP. Targeting miR-21 enhances the sensitivity of human colon cancer HT-29 cells to chemoradiotherapy in vitro. Biochem Biophys Res Commun. 2014;443:789-795.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Wu J, Qian J, Li C, Kwok L, Cheng F, Liu P, Perdomo C, Kotton D, Vaziri C, Anderlind C. miR-129 regulates cell proliferation by downregulating Cdk6 expression. Cell Cycle. 2010;9:1809-1818.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Karaayvaz M, Zhai H, Ju J. miR-129 promotes apoptosis and enhances chemosensitivity to 5-fluorouracil in colorectal cancer. Cell Death Dis. 2013;4:e659.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Kurokawa K, Tanahashi T, Iima T, Yamamoto Y, Akaike Y, Nishida K, Masuda K, Kuwano Y, Murakami Y, Fukushima M. Role of miR-19b and its target mRNAs in 5-fluorouracil resistance in colon cancer cells. J Gastroenterol. 2012;47:883-895.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Rossi L, Bonmassar E, Faraoni I. Modification of miR gene expression pattern in human colon cancer cells following exposure to 5-fluorouracil in vitro. Pharmacol Res. 2007;56:248-253.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Tazawa H, Tsuchiya N, Izumiya M, Nakagama H. Tumor-suppressive miR-34a induces senescence-like growth arrest through modulation of the E2F pathway in human colon cancer cells. Proc Natl Acad Sci USA. 2007;104:15472-15477.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Akao Y, Noguchi S, Iio A, Kojima K, Takagi T, Naoe T. Dysregulation of microRNA-34a expression causes drug-resistance to 5-FU in human colon cancer DLD-1 cells. Cancer Lett. 2011;300:197-204.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Michael MZ, O’ Connor SM, van Holst Pellekaan NG, Young GP, James RJ. Reduced accumulation of specific microRNAs in colorectal neoplasia. Mol Cancer Res. 2003;1:882-891.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Chen X, Guo X, Zhang H, Xiang Y, Chen J, Yin Y, Cai X, Wang K, Wang G, Ba Y. Role of miR-143 targeting KRAS in colorectal tumorigenesis. Oncogene. 2009;28:1385-1392.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Borralho PM, Kren BT, Castro RE, da Silva IB, Steer CJ, Rodrigues CM. MicroRNA-143 reduces viability and increases sensitivity to 5-fluorouracil in HCT116 human colorectal cancer cells. FEBS J. 2009;276:6689-6700.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Zhou Y, Wan G, Spizzo R, Ivan C, Mathur R, Hu X, Ye X, Lu J, Fan F, Xia L. miR-203 induces oxaliplatin resistance in colorectal cancer cells by negatively regulating ATM kinase. Mol Oncol. 2014;8:83-92.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Hur K, Toiyama Y, Takahashi M, Balaguer F, Nagasaka T, Koike J, Hemmi H, Koi M, Boland CR, Goel A. MicroRNA-200c modulates epithelial-to-mesenchymal transition (EMT) in human colorectal cancer metastasis. Gut. 2013;62:1315-1326.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Toiyama Y, Hur K, Tanaka K, Inoue Y, Kusunoki M, Boland CR, Goel A. Serum miR-200c is a novel prognostic and metastasis-predictive biomarker in patients with colorectal cancer. Ann Surg. 2014;259:735-743.  [PubMed]  [DOI]  [Cited in This Article: ]