Guidelines For Basic Science Open Access
Copyright ©2012 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Gastroenterol. Jun 14, 2012; 18(22): 2745-2755
Published online Jun 14, 2012. doi: 10.3748/wjg.v18.i22.2745
Interrelationship between microsatellite instability and microRNA in gastrointestinal cancer
Hiroyuki Yamamoto, Yasushi Adachi, Hiroaki Kunimoto, Katsuhiko Nosho, Hiromu Suzuki, Yasuhisa Shinomura, First Department of Internal Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8543, Japan
Hiroaki Taniguchi, Division of Cancer Cell Research, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
Hiromu Suzuki, Department of Molecular Biology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
Author contributions: Yamamoto H conceived the topic, reviewed the literature and prepared the manuscript; Adachi Y, Taniguchi H, Kunimoto H, Nosho K and Suzuki H reviewed and analyzed the literature; and Shinomura Y provided intellectual support.
Supported by Grants-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology of Japan
Correspondence to: Hiroyuki Yamamoto, MD, FJSIM, PhD, First Department of Internal Medicine, Sapporo Medical University School of Medicine, S1W16 Chuo-ku, Sapporo 060-8543, Japan. h-yama@sapmed.ac.jp
Telephone: +81-11-6112111 Fax: +81-11-6112282
Received: September 28, 2011
Revised: March 2, 2012
Accepted: March 9, 2012
Published online: June 14, 2012

Abstract

There is an increasing understanding of the roles that microsatellite instability (MSI) plays in Lynch syndrome (by mutations) and sporadic (by mainly epigenetic changes) gastrointestinal (GI) and other cancers. Deficient DNA mismatch repair (MMR) results in the strong mutator phenotype known as MSI, which is the hallmark of cancers arising within Lynch syndrome. MSI is characterized by length alterations within simple repeated sequences called microsatellites. Lynch syndrome occurs primarily because of germline mutations in one of the MMR genes, mainly MLH1 or MSH2, less frequently MSH6, and rarely PMS2. MSI is also observed in about 15% of sporadic colorectal, gastric, and endometrial cancers and in lower frequencies in a minority of other cancers where it is often associated with the hypermethylation of the MLH1 gene. miRNAs are small noncoding RNAs that regulate gene expression at the posttranscriptional level and are critical in many biological processes and cellular pathways. There is accumulating evidence to support the notion that the interrelationship between MSI and miRNA plays a key role in the pathogenesis of GI cancer. As a possible new mechanism underlying MSI, overexpression of miR-155 has been shown to downregulate expression of MLH1, MSH2, and MSH6. Thus, a subset of MSI-positive (MSI+) cancers without known MMR defects may result from miR-155 overexpression. Target genes of frameshift mutation for MSI are involved in various cellular functions, such as DNA repair, cell signaling, and apoptosis. A novel class of target genes that included not only epigenetic modifier genes, such as HDAC2, but also miRNA processing machinery genes, including TARBP2 and XPO5, were found to be mutated in MSI+ GI cancers. Thus, a subset of MSI+ colorectal cancers (CRCs) has been proposed to exhibit a mutated miRNA machinery phenotype. Genetic, epigenetic, and transcriptomic differences exist between MSI+ and MSI− cancers. Molecular signatures of miRNA expression apparently have the potential to distinguish between MSI+ and MSI− CRCs. In this review, we summarize recent advances in the MSI pathogenesis of GI cancer, with the focus on its relationship with miRNA as well as on the potential to use MSI and related alterations as biomarkers and novel therapeutic targets.

Key Words: Microsatellite instability, MicroRNA, DNA mismatch repair, Frameshift mutation, MicroRNA processing



INTRODUCTION

A type of genetic instability characterized by length alterations within simple repeated microsatellite sequences, termed microsatellite instability (MSI), occurs in a majority of patients with Lynch syndrome (also known as hereditary nonpolyposis colorectal cancer) by mutations and in a subset of sporadic gastrointestinal (GI) and other cancers by mainly epigenetic changes[1-10]. Genetic and epigenetic inactivation of DNA mismatch repair (MMR) genes results in the mutator phenotype, mutations in cancer-related genes, and cancer development (Figure 1). MSI underlies a distinctive carcinogenic pathway because MSI-positive (MSI+) cancers exhibit many differences in clinical, pathological, and molecular characteristics relative to MSI-negative (MSI-) cancers irrespective of their hereditary or sporadic origins. The differences in genotype can be explained because deficient MMR leads to a strong mutator phenotype with a very specific mutation spectrum. MSI accumulates frameshift mutations in repeated sequences located in coding regions of target tumor suppressor genes. The peculiar genotype of MSI+ cancers also includes specific patterns of gene regulation. MSI+ GI cancers often show an aberrant epigenetic pattern such as hypermethylation of various genes, including the key MMR gene, MLH1. The differences in genotype and phenotype between MSI+ and MSI- GI cancers are likely to be causally linked to their differences in biological and clinical features. Diagnostic characterization of the MSI status thus has implications in basic and clinical oncology. MiRNAs are small RNA molecules that regulate gene expression at the posttranscriptional level and are critical for many cellular functions[11-20]. There is accumulating evidence to support the notion that the interrelationship between MSI and miRNA plays a key role in the pathogenesis of GI cancer.

Figure 1
Figure 1 A model of DNA mismatch repair and molecular pathways for microsatellite instability+ colorectal cancers. A: A model of the proposed mechanism of mismatch repair (MMR) proteins, illustrating patterns of relevant heterodimerization; B: The models for colorectal cancer (CRC) carcinogenesis are presented in parallel for Lynch syndrome and sporadic cases. HNPCC: Hereditary nonpolyposis colorectal cancer; MSI: Microsatellite instability; CIMP: CpG island methylator phenotype; MSI: Microsatellite instability.
MSI BY THE OVEREXPRESSION OF MIR-155 OR MIR-21

Various pathogenic events, including germline and somatic mutations, promoter methylation, and reduced histone acetylation[21], lead to inactivation of core MMR proteins. A vast majority of MSI+ cancers can be explained by mutation and/or epigenetic inactivation of the core MMR proteins[22]. The etiologies of the remaining MSI+ cancers remain poorly understood. In an unselected series of 1066 colorectal cancer (CRC) patients, 135 (13%) were MSI+[22]. Of these, 23 (6%) had germline mutations in one of the MMR gene, and 106 (79%) showed methylation of the MLH1 promoter. Approximately 5% of these MSI+ cancers displayed loss of expression of at least one of the core MMR proteins without a well-defined genetic or epigenetic cause[22].

Vareli et al[23] have shown that overexpression of miR-155 significantly downregulates the core MMR proteins; namely, MLH1, MSH2, and, MSH6 in CRC cell lines, thus inducing an MSI. The downregulation of MLH1 and MSH2 proteins by miR-155 lead to destabilization of the respective heterodimeric complex proteins and a mutator phenotype[24]. An inverse correlation between miR-155 overexpression and the expression of MLH1 and MSH2 was further demonstrated in human CRC tissues. Most MSI+ cancers without a known cause of MMR inactivation show miR-155 overexpression. However, not all CRCs with increased miR-155 expression were MSI+. It is also possible that miR-155 affects other related DNA repair proteins, thus enhancing the phenotypic effect of MMR defects. Although further confirmation is required, the results suggest that miR-155 overexpression is an additional mechanism underlying the development of MSI in cancer (Figure 2)[23].

Figure 2
Figure 2 Cancer progression of sporadic microsatellite instability+ colorectal cancers. The model for microsatellite instability (MSI)+ colorectal cancer progression is presented based on levels of MSI and chromosomal instability (CIN), and genetic, epigenetic and miRNA alterations. SSA/P: Sessile serrated adenomas/polyps; CIMP: CpG island methylator phenotype; MMR: Mismatch repair.

The reduced expression of a single allele of the adenomatous polyposis coli and transforming growth factor (TGF)-β receptor I gene has been linked to CRC[25,26]. Thus, incomplete repression of MMR proteins by miR-155 is not unique to tumor suppressor genes in cancer. Recently, miRNAs have been suggested to act as transactivating elements involved in allele and gene expression regulation[27,28]. These results support the notion that miRNAs play a role in the non-Mendelian regulation of MMR genes.

miR-21 is overexpressed in various types of human cancers, including CRC[29]. Valeri et al[30] reported that miR-21 directly targets the 3’ untranslated region (UTR) of MSH2 and MSH6 mRNA, resulting in downregulation of protein expression. The inverse correlation between miR-21 overexpression and MSH2 expression was shown in CRC tissues. Cells that overexpress miR-21 showed significantly reduced 5-fluorouracil (5-FU)-induced G2/M damage arrest and apoptosis that is characteristic of defective MMR. Because miR-21 expression could increase in cell lines continuously exposed to 5-FU[31], cancer cells may develop a secondary resistance to 5-FU through miR-21 overexpression. Thus, miR-21-dependent downregulation of MSH2-MSH6 may be responsible for both primary and secondary resistance to 5-FU.

TARGET CANCER-RELATED GENES OF MSI

The instability in cancer-related genes at coding microsatellites causes frameshift mutations and functional inactivation of affected proteins, thereby providing a selective growth advantage to deficient MMR cells[32]. For instance, TGF-βreceptor II and the pro-apoptotic gene BAX are frequently inactivated by slippage-induced frameshift mutations in mononucleotide tracts present in their gene coding regions[33,34]. These findings have provided proof for the causative link between MSI and mutations in cancer-related genes, and they were also convincing examples of the differences between the mutator and suppressor pathways for cancer. These genes have also been mutated in cancers in the suppressor pathway, but at decreased frequencies and not by slippage-linked frameshifts[35,36].

A number of cancer-related genes mutated in MSI+ cancers have been reported (Figure 3). Mutations that promote cancer cell growth are assumed to be the driving force during MSI+ carcinogenesis and are designated as real common target genes (Figure 2)[37]. Mutations of microsatellite-harboring genes that do not contribute to carcinogenesis are designated as bystander genes. However, it is not always clear which mutations are “driver mutations” and which are “passenger mutations”[9,38]. The Selective Targets database (SelTarbase) (http://www.seltarbase.org) of human mononucleotide-microsatellite mutations and their potential impact to carcinogenesis and immunology has been developed[39]. The database includes a comprehensive database of all human coding, untranslated, non-coding RNA and intronic mononucleotide repeat tracts and is useful for basic and clinical oncology.

Figure 3
Figure 3 Representative target genes in microsatellite instability+ gastrointestinal cancers. A number of cancer-related genes mutated in microsatellite instability+ gastrointestinal cancers have been reported. The relevance of each mutation is not necessarily proven.

Because MSI+ cancers accumulate many mutations, disruption of cell growth and survival regulation can be accomplished in different cancers by mutations in different genes of the same signaling pathways[40]. Therefore, genes with infrequent and/or monoallelic mutations should not be regarded as irrelevant. Thus, the relevance of microsatellite-specific mutations in MSI+ cancers can be proven only when there is supporting evidence for functionality irrespective of mutation incidence[2].

Target cancer-related genes of MSI+ cancers can be functionally categorized as tumor suppressors and genes involved in DNA repair, cell cycle, cell proliferation, apoptosis and others (Figure 3). Interestingly, every human MMR gene except MLH1 contains a mononucleotide repeat of at least A7[41]. Thus, frameshift mutations of MSH3 and MSH6 led to the concept of “the mutator that mutates the other mutator” (Figure 2)[42].

The spectrum of mutations of target genes could affect cancer biology, therapeutic response, and prognosis of patients. Most putative MSI target genes have been proposed mainly on the basis of high mutation frequency detected within their coding regions. However, genes containing microsatellites that are located within noncoding regulatory regions, such as introns, promoters, and 5′ and 3′ UTRs, could be also mutated in MSI+ cancers. Alterations within untranslated mononucleotide repeat tracts can alter transcription level or transcript stability. It has been suggested that some intronic repeat mutations in genes, such as ATM, MYB[43], and MRE11[44], play a role in MSI carcinogenesis[45]. Decreased matrix metalloproteinase (MMP)-3 expression due to insertions and/or deletions in the MMP-3 promoter region led to a decrease in the levels of the active MMP-9 form, which may explain the less invasive potential of MSI+ cancer cells[46] and the propensity for a good prognosis in the case of MSI+ CRCs[47].

Genomic copy number changes are frequently observed in cancers. It is well known that MSI+ cancers show less genomic copy number changes and are mostly diploid[48]. However, genes responsible for chromosomal instability (CIN) could be mutated in MSI+ cancers, and these defects may be selected during the course of cancer progression (Figures 2 and 3)[49]. Furthermore, mutations of RAD50 and MRE11 are reportedly associated with defects in nonhomologous end-joining, leading to chromosomal alterations during cancer progression[45].

Altered histone modifications that affect chromatin structures are also involved in carcinogenesis[49]. Epigenetic modifier genes could also be MSI target genes. Ropero et al[50] detected frameshift mutations in the histone deacetylase (HDAC) 2 gene in MSI+ GI cancers. This HDAC2 mutation made mutation-positive cancer cells more resistant to the antiproliferative and proapoptotic effects of certain HDAC inhibitors such as trichostatin A, but not to others such as butyric acid and valproic acid. Since HDAC inhibitors may serve as therapeutic agents for cancer, these findings support the use of HDAC2 mutation status in future pharmacogenetic treatment[50].

MIRNA PROCESSING MACHINERY GENES AS MSI TARGET GENES

MiRNAs are small noncoding RNAs that regulate gene expression at the posttranscriptional level and are critical in many biological processes and cellular pathways (Figure 4)[11-20]. MiRNA expression profiles of human cancers have been characterized by an overall mature miRNA downregulation[51-53]. The causes of the aberrant miRNA expression patterns in cancer involve DNA copy number amplification or deletion[54], inappropriate transactivation, transcriptional repression by oncogenic and/or other factors[55], failure of miRNA post-transcriptional regulation[56], and genetic mutation[57] or transcriptional silencing associated with hypermethylation of CpG island promoters[58-62].

Figure 4
Figure 4 MiRNA biogenesis and genes mutated in microsatellite instability+ gastrointestinal cancers. A consequence of perfect complementarity between miRNA and mRNA is mRNA cleavage and degradation. Imperfect alignment represses gene translation. ORF: Open reading frame; RISC: RNA-induced silencing complexes.

The control of the miRNA biosynthesis pathway is important in the spatiotemporal pattern of miRNA expression in cells. Thus, impaired miRNA processing pathways may themselves be targets of genetic and/or epigenetic disruption in cancer[63,64]. Recently, it has been reported that mitogenic signaling can be translated into changes in cell viability and proliferation through the miRNA biogenesis pathway. A TAR RNA-binding protein 2 (TARBP2) gene encodes TRBP, an essential functional partner of DICER1 (Figure 4)[65,66]. TARBP2 is phosphorylated under normal growth conditions, which increases the stability of TARBP2 and DICER[67]. Upon growth factor stimulation, MAPK/ERK pathway increases TARBP2 phosphorylation, leading to a coordinated increase in levels of growth-promoting miRNA and a decrease in the expression of let-7 tumor suppressor miRNA. In contrast, pharmacological inhibition of MAPK/ERK resulted in an anti-growth miRNA profile[67]. These results further suggest the important role of miRNA processing mediated by TARBP2 in preservation of a normal, untransformed cell state[17].

Melo et al[68] have found truncating heterozygous mutations in TARBP2 in MSI+ cancer cell lines and in primary sporadic and hereditary MSI+ GI cancers (Figure 4). TARBP2 mutations diminished TRBP protein expression, resulting in impaired miRNA processing and enhanced cellular transformation. The TRBP impairment was associated with a secondary defect in DICER1 activity by destabilization of the DICER1 protein. Thus, TARBP2 mutations may explain overall miRNA downregulation in a subset of MSI+ cancers. Because the restoration of efficient miRNA production by the reintroduction of TRBP can suppress cancer cell growth, these findings are important for the development of new therapeutic strategies for the treatment of cancer[68].

MUTATIONS OF THE EXPORTIN 5 GENE

Because of the nuclear retention of certain precursor miRNAs (pre-miRNAs), mature miRNA expression levels are not consistent with pre-miRNA expression levels in various human cancer cell lines[17]. Thus, defects in the nuclear export of pre-miRNAs may be one of the mechanisms underlying the global impairment of mature miRNAs in human cancer. The exportin 5 (XPO5) mediates nuclear export of pre-miRNA (Figure 4). Melo et al[69,70] have identified inactivating heterozygous mutations of XPO5 in MSI+ cancer cell lines and in primary sporadic and hereditary MSI+ GI cancers. The mutant form of XPO5 does not comprise a C-terminal region that is important for the formation of the pre-miRNA/XPO5/Ran-GTP ternary complex. Thus, the XPO5 defect trapped certain pre-miRNAs in the nucleus, reduced miRNA processing, and impaired miRNA-target inhibition. It is important to note that the restoration of XPO5 functions rescued the disturbed export of critical tumor-suppressive pre-miRNAs, which results in tumor suppression[69].

Interestingly, although the heterozygous XPO5 mutation decreased accumulation of a fraction of detectable miRNAs, many others were not affected. It seems that XPO5 does not bind to pre-miRNAs universally but has certain substrate preferences, which are possibly mediated by sequence or structure[38]. Strategies directed toward stimulating the activity of miRNA processing factors and restoring the production of mature growth inhibitory miRNAs may have therapeutic value[69].

MUTATED MIRNA MACHINERY PHENOTYPE AS A NEW CANCER PHENOTYPE

Recent works have suggested that the other component of the miRNA biogenesis pathway, DICER1, is a haploinsufficient tumor suppressor[71,72]. Therefore, it appears that at least three components of the miRNA biogenesis pathway are haploinsufficient tumor suppressors, with TARBP2 and XPO5, but not DICER, mutations prevalent in MSI+ cancers[38]. In addition, the miRISC components AGO2, TNRC6A, and TNRC6C can also be mutated in MSI+ cancers (Figure 4)[73], although the functional significances remain to be determined[38,70].

From these observations, a new cancer phenotype known as mutated miRNA machinery phenotype (MMMP) has been proposed for MSI+ CRCs having mutations in the miRNA machinery genes and the deregulated miRNAome. Although larger studies are required to fully characterize and validate this feature as a criterion for classification, a broader miRNAome-modifying approach may be effective for cancer patients with MMMP[15].

TRANSCRIPTOMIC DIFFERENCES BETWEEN MSI+ AND MSI- CRCS

As molecular markers, gene expression profiles are being developed for many cancers. Array technology has identified a number of genes that are expressed differentially between MSI+ and MSI- CRCs[74-76]. By using supervised analysis of cDNA microarray data, Giacomini et al[77] identified a robust expression signature distinguishing MSI+ and MSI- CRC cell lines. By using high-density oligonucleotide microarrays, Kruhoffer et al[78] constructed a gene signature that distinguished MSI+ and MSI- CRCs. The authors further constructed a signature that distinguished sporadic and hereditary cases of MSI+ CRCs. Identification of a signature for MMR deficiency would be relevant, both biologically and clinically[78].

As for miRNAs, Lanza et al[79] analyzed 16 MSI+ and 23 MSI- CRCs for genome-wide expression of miRNA and mRNA. On the basis of combined miRNA and mRNA expression, a molecular signature comprising 27 differentially expressed genes, including 8 miRNAs, could correctly distinguish MSI+ and MSI- CRCs. Among the differentially expressed miRNAs, various members of the oncogenic miR-17-92 family were significantly upregulated in MSI- cancers. Among these, miR-17-5p, miR-20, miR-25, miR-92-1, miR-92-2, miR-93-1, and miR-106a were significantly upregulated in MSI- when compared with MSI+ CRCs. Because members of the miR-17-92 family act as oncogenes, these results may explain, at least in part, the less aggressive behavior of MSI+ CRCs when compared with their MSI- counterparts.

Earle et al[80] analyzed 22 MSI-H, including 6 Lynch syndrome, 8 MSI-L, and 25 MSS CRCs for a selected panel of 24 miRNAs. Relative expression of miR-26b, miR-31, miR-92, miR-155, miR-196a, and miR-223 were significantly different among MSI subgroups, and miR-31 and miR-223 were overexpressed in CRCs of patients with Lynch syndrome. These findings indicate that miRNA expression in CRC is associated with MSI status, including Lynch syndrome and MSI-L, and that miRNAs may play significant roles in these MSI subgroups in addition to having possible effects on cancer characteristics.

Slattery et al[81] analyzed 70 CRCs for 866 miRNAs using microarrays. At the 1.5-fold level, 143 miRNAs were differentially expressed in MSI+ CRCs. MiR-139-3p, miR-223, and miR-370 were upregulated and miR-24-2, miR-424, miR-552, and let-7g were downregulated at a level of 1.5-fold or greater in MSI+ CRCs when compared with MSI- CRCs.

Thus, differentially expressed miRNAs are likely to be relevant, both biologically and clinically, although their functional significances remain to be determined. High levels of miR-21 in the stroma of CRCs reportedly predict short disease-free survival in stage II CRC patients; however, the levels are not associated with the MSI status[82].

DEFECTIVE MMR AS A NOVEL THERAPEUTIC TARGET

MSI+ cancers may be managed more effectively with novel targeted therapies based on molecular alterations (Figure 5). A combination of treatments that target both primary alterations of DNA MMR gene and secondary alterations, such as frameshift mutations of target genes, may be also effective. A synthetic lethal relationship, where the simultaneous inhibition of two different regulatory pathways leads to cell death, is a recent therapeutic strategy[83]. Therefore, identification of synthetic lethal interactions with MMR deficiency could potentially lead to the identification of specific therapeutic targets[84]. The inhibition of poly (adenosine diphosphate-ribose) polymerase (PARP) is a potential synthetic lethal therapeutic strategy for the treatment of cancers with specific DNA repair defects, such as a BRCA1 or BRCA2 mutation[85].

Figure 5
Figure 5 Targeted therapies based on molecular alterations in microsatellite instability+ colorectal cancers. Microsatellite instability+ cancers may be managed more effectively with novel targeted therapies based on molecular alterations. MSI: Microsatellite instability; HDAC: Histone deacetylase; PI3K/mTOR: Phosphoinositide 3-kinase/mammalian target of rapamycin; XPO5: Exportin 5.

A subset of MSI+ CRCs may also be suitable for this strategy. A novel PARP inhibitor, ABT-888, showed preferential activity on MSI+ CRC cell lines harboring mutations in both MRE11 and RAD50 genes when compared with MSI- cell lines that were wild type for both genes[86]. Recently, Vilar et al[87] reported that MSI+ CRCs deficient in double strand break (DSB) repair due to MRE11 mutations show a higher sensitivity to PARP-1 inhibition. A phase II study assessing the efficacy of a PARP-1 inhibitor, olaparib, in CRCs stratified by MSI status is ongoing. Further clinical studies regarding combinations of a PARP-1 inhibitor with other DSB-inducing therapies, such as radiation or irinotecan, are warranted in MSI+ CRCs with MRE11 mutations. Although these results need to be confirmed in other settings, they suggest that specific mutations such as MRE11 can be used to exploit the concept of synthetic lethality in MSI+ cancers, which has been successful in BRCA1-mutant breast cancers[88].

Methotrexate reportedly induces oxidative DNA damage and is selectively lethal to cancer cells with MSH2 defects[84,89]. Thus, a synthetic lethal relationship between deficient MSH2 and treatment with methotrexate led to a phase II trial, incorporating measurement of 8-oxoG DNA lesions as a biomarker, in metastatic CRC patients with germline mutation or loss of MSH2.

Because MSI+ CRCs often harbor a near diploid stable karyotype, these cancers may be sensitive to mitotic inhibitors, such as taxanes and kinesin-5 inhibitors[90]. To determine the effect of CIN and MSI on the efficacy of the microtubule-stabilizing agent patupilone/EPO960, a phase II study called CIN and Anti-Tubulin Response Assessment in CRC is ongoing. It is assumed that MSI+ CRC patients will benefit more than MSI- CRC patients.

Gene expression signatures can also be used for new MSI+ cancer therapies. Fourteen of the 164 compounds were shown to target MSI+ cancer cell lines using combined gene expression data sets and a connectivity map[91]. Rapamycin, LY-294002, 17-(allylamino)-17-demethoxygeldanamycin, and trichostatin A were the most convincing candidate compounds. MSI+ cell lines with MLH1 hypermethylation were preferentially targeted by rapamycin and LY-294002 when compared with MSI- cells. These results underscore the relevant role of the PI3K/AKT/mTOR pathway and its therapeutic application in MSI+ cancer, although its clinical significance needs confirmation.

MiRNA-based cancer therapy is limited mainly to targeting a single miRNA[92,93]. However, if most cancers are characterized by a defect in miRNA production and global mature miRNA downregulation[51-53], restoration of the global miRNAome may be an attractive approach in cancer therapy. Melo et al[94] have found that the small molecule enoxacin, a fluoroquinolone used as an antibacterial compound, enhances the miRNA-processing machinery by binding to TRBP. Enoxacin was shown to inhibit the growth of a variety of cancer cells. The enhanced miRNA-processing activity by enoxacin did not depend on general fluoroquinolone activity but on the unique chemical structure of enoxacin. These results highlight the key role of disturbed miRNA expression in carcinogenesis, and suggest the potential of novel miRNA-based cancer therapy to restore the disrupted miRNAome of cancer cells.

Finally, it remains to be determined whether XPO5 mutations can be exploited therapeutically. Since it is difficult to directly restore XPO5 activity, restoring miRNA accumulation by alternative methods may be a more realistic strategy. Given that only a few deregulated tumor suppressor miRNAs appear to be critical for the tumor-promoting effect of XPO5 mutations, it may be possible to supply those miRNAs exogenously as miRNA duplexes that would not need to undergo nuclear export. It may also be possible to find a subset of important target genes of deregulated tumor suppressor miRNAs, which may be responsive to inactivation by conventional pharmacological methodologies or novel biologics.

CONCLUSION

The biological and clinical implications of MSI in GI cancers continue to develop. Recent findings, such as overexpression of miR-21 and miR-155 and mutations of TARBP2 and XPO5 in MSI+ GI cancers, further suggest the important interrelationship between MSI and miRNA in MSI carcinogenesis. The clinicopathological, genetic, epigenetic, prognostic, and therapeutic characteristics of MSI+ cancers are becoming clear, but remain to be fully determined. Analysis of MSI status in cancer patients is warranted as a screening for Lynch syndrome; it could be a potential predictive marker of response to chemotherapy. Since molecular targeting therapeutics are being used in clinical settings and trials, it seems important to clarify if molecular target genes are differentially regulated between MSI+ and MSI- cancers and if the MSI status has the prognostic or predictive significance in metastatic CRC. Further analysis is required to gain insight into MSI carcinogenesis, for a better understanding of disease pathogenesis, and for the development of new diagnostic and/or therapeutic approaches targeting essential pathogenetic alterations.

Footnotes

Peer reviewers: Dr. John Souglakos, Department of Medical Oncology, University Hospital of Heraklion and Laboratory of Cancer Biology, 71110 Heraklion, Greece; Dr. Jose Perea, Department of Surgery, 12 De Octubre University Hospital, Rosas De Aravaca 82A, 28023 Madrid, Spain

S- Editor Gou SX L- Editor A E- Editor Zhang DN

References
1.  Yamamoto H, Imai K, Perucho M. Gastrointestinal cancer of the microsatellite mutator phenotype pathway. J Gastroenterol. 2002;37:153-163.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Perucho M. Tumors with microsatellite instability: many mutations, targets and paradoxes. Oncogene. 2003;22:2223-2225.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Imai K, Yamamoto H. Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis. 2008;29:673-680.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Sinicrope FA, Sargent DJ. Clinical implications of microsatellite instability in sporadic colon cancers. Curr Opin Oncol. 2009;21:369-373.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Poulogiannis G, Frayling IM, Arends MJ. DNA mismatch repair deficiency in sporadic colorectal cancer and Lynch syndrome. Histopathology. 2010;56:167-179.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Goel A, Boland CR. Recent insights into the pathogenesis of colorectal cancer. Curr Opin Gastroenterol. 2010;26:47-52.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Vilar E, Gruber SB. Microsatellite instability in colorectal cancer-the stable evidence. Nat Rev Clin Oncol. 2010;7:153-162.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Hewish M, Lord CJ, Martin SA, Cunningham D, Ashworth A. Mismatch repair deficient colorectal cancer in the era of personalized treatment. Nat Rev Clin Oncol. 2010;7:197-208.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Boland CR, Goel A. Microsatellite instability in colorectal cancer. Gastroenterology. 2010;138:2073-2087.e3.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Iacopetta B, Grieu F, Amanuel B. Microsatellite instability in colorectal cancer. Asia Pac J Clin Oncol. 2010;6:260-269.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Saito Y, Suzuki H, Hibi T. The role of microRNAs in gastrointestinal cancers. J Gastroenterol. 2009;44 Suppl 19:18-22.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Davalos V, Esteller M. MicroRNAs and cancer epigenetics: a macrorevolution. Curr Opin Oncol. 2010;22:35-45.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Krol J, Loedige I, Filipowicz W. The widespread regulation of microRNA biogenesis, function and decay. Nat Rev Genet. 2010;11:597-610.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Song B, Ju J. Impact of miRNAs in gastrointestinal cancer diagnosis and prognosis. Expert Rev Mol Med. 2010;12:e33.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Davis-Dusenbery BN, Hata A. MicroRNA in Cancer: The Involvement of Aberrant MicroRNA Biogenesis Regulatory Pathways. Genes Cancer. 2010;1:1100-1114.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Dong Y, Wu WK, Wu CW, Sung JJ, Yu J, Ng SS. MicroRNA dysregulation in colorectal cancer: a clinical perspective. Br J Cancer. 2011;104:893-898.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Melo SA, Esteller M. Dysregulation of microRNAs in cancer: playing with fire. FEBS Lett. 2011;585:2087-2099.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Cortez MA, Bueso-Ramos C, Ferdin J, Lopez-Berestein G, Sood AK, Calin GA. MicroRNAs in body fluids--the mix of hormones and biomarkers. Nat Rev Clin Oncol. 2011;8:467-477.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  van Kouwenhove M, Kedde M, Agami R. MicroRNA regulation by RNA-binding proteins and its implications for cancer. Nat Rev Cancer. 2011;11:644-656.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Lopez-Serra P, Esteller M. DNA methylation-associated silencing of tumor-suppressor microRNAs in cancer. Oncogene. 2012;31:1609-1622.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 251]  [Cited by in F6Publishing: 262]  [Article Influence: 20.2]  [Reference Citation Analysis (0)]
21.  Edwards RA, Witherspoon M, Wang K, Afrasiabi K, Pham T, Birnbaumer L, Lipkin SM. Epigenetic repression of DNA mismatch repair by inflammation and hypoxia in inflammatory bowel disease-associated colorectal cancer. Cancer Res. 2009;69:6423-6429.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Hampel H, Frankel WL, Martin E, Arnold M, Khanduja K, Kuebler P, Nakagawa H, Sotamaa K, Prior TW, Westman J. Screening for the Lynch syndrome (hereditary nonpolyposis colorectal cancer). N Engl J Med. 2005;352:1851-1860.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Valeri N, Gasparini P, Fabbri M, Braconi C, Veronese A, Lovat F, Adair B, Vannini I, Fanini F, Bottoni A. Modulation of mismatch repair and genomic stability by miR-155. Proc Natl Acad Sci USA. 2010;107:6982-6987.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Marsischky GT, Filosi N, Kane MF, Kolodner R. Redundancy of Saccharomyces cerevisiae MSH3 and MSH6 in MSH2-dependent mismatch repair. Genes Dev. 1996;10:407-420.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Yan H, Dobbie Z, Gruber SB, Markowitz S, Romans K, Giardiello FM, Kinzler KW, Vogelstein B. Small changes in expression affect predisposition to tumorigenesis. Nat Genet. 2002;30:25-26.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Valle L, Serena-Acedo T, Liyanarachchi S, Hampel H, Comeras I, Li Z, Zeng Q, Zhang HT, Pennison MJ, Sadim M. Germline allele-specific expression of TGFBR1 confers an increased risk of colorectal cancer. Science. 2008;321:1361-1365.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Ahluwalia JK, Hariharan M, Bargaje R, Pillai B, Brahmachari V. Incomplete penetrance and variable expressivity: is there a microRNA connection? Bioessays. 2009;31:981-992.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  de la Chapelle A. Genetic predisposition to human disease: allele-specific expression and low-penetrance regulatory loci. Oncogene. 2009;28:3345-3348.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Volinia S, Calin GA, Liu CG, Ambs S, Cimmino A, Petrocca F, Visone R, Iorio M, Roldo C, Ferracin M. A microRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci USA. 2006;103:2257-2261.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  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: ]
31.  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: ]
32.  Woerner SM, Kloor M, Mueller A, Rueschoff J, Friedrichs N, Buettner R, Buzello M, Kienle P, Knaebel HP, Kunstmann E. Microsatellite instability of selective target genes in HNPCC-associated colon adenomas. Oncogene. 2005;24:2525-2535.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Markowitz S, Wang J, Myeroff L, Parsons R, Sun L, Lutterbaugh J, Fan RS, Zborowska E, Kinzler KW, Vogelstein B. Inactivation of the type II TGF-beta receptor in colon cancer cells with microsatellite instability. Science. 1995;268:1336-1338.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Rampino N, Yamamoto H, Ionov Y, Li Y, Sawai H, Reed JC, Perucho M. Somatic frameshift mutations in the BAX gene in colon cancers of the microsatellite mutator phenotype. Science. 1997;275:967-969.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Yamamoto H, Sawai H, Perucho M. Frameshift somatic mutations in gastrointestinal cancer of the microsatellite mutator phenotype. Cancer Res. 1997;57:4420-4426.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Grady WM, Myeroff LL, Swinler SE, Rajput A, Thiagalingam S, Lutterbaugh JD, Neumann A, Brattain MG, Chang J, Kim SJ. Mutational inactivation of transforming growth factor beta receptor type II in microsatellite stable colon cancers. Cancer Res. 1999;59:320-324.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Woerner SM, Benner A, Sutter C, Schiller M, Yuan YP, Keller G, Bork P, Doeberitz MK, Gebert JF. Pathogenesis of DNA repair-deficient cancers: a statistical meta-analysis of putative Real Common Target genes. Oncogene. 2003;22:2226-2235.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Grosshans H, Büssing I. MicroRNA biogenesis takes another single hit from microsatellite instability. Cancer Cell. 2010;18:295-297.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Woerner SM, Yuan YP, Benner A, Korff S, von Knebel Doeberitz M, Bork P. SelTarbase, a database of human mononucleotide-microsatellite mutations and their potential impact to tumorigenesis and immunology. Nucleic Acids Res. 2010;38:D682-D689.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Yamamoto H, Gil J, Schwartz S, Perucho M. Frameshift mutations in Fas, Apaf-1, and Bcl-10 in gastro-intestinal cancer of the microsatellite mutator phenotype. Cell Death Differ. 2000;7:238-239.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Chang DK, Metzgar D, Wills C, Boland CR. Microsatellites in the eukaryotic DNA mismatch repair genes as modulators of evolutionary mutation rate. Genome Res. 2001;11:1145-1146.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Malkhosyan S, Rampino N, Yamamoto H, Perucho M. Frameshift mutator mutations. Nature. 1996;382:499-500.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Hugo H, Cures A, Suraweera N, Drabsch Y, Purcell D, Mantamadiotis T, Phillips W, Dobrovic A, Zupi G, Gonda TJ. Mutations in the MYB intron I regulatory sequence increase transcription in colon cancers. Genes Chromosomes Cancer. 2006;45:1143-1154.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Giannini G, Rinaldi C, Ristori E, Ambrosini MI, Cerignoli F, Viel A, Bidoli E, Berni S, D'Amati G, Scambia G. Mutations of an intronic repeat induce impaired MRE11 expression in primary human cancer with microsatellite instability. Oncogene. 2004;23:2640-2647.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Koh KH, Kang HJ, Li LS, Kim NG, You KT, Yang E, Kim H, Kim HJ, Yun CO, Kim KS. Impaired nonhomologous end-joining in mismatch repair-deficient colon carcinomas. Lab Invest. 2005;85:1130-1138.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Morán A, Iniesta P, de Juan C, González-Quevedo R, Sánchez-Pernaute A, Díaz-Rubio E, Ramón y Cajal S, Torres A, Balibrea JL, Benito M. Stromelysin-1 promoter mutations impair gelatinase B activation in high microsatellite instability sporadic colorectal tumors. Cancer Res. 2002;62:3855-3860.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Morán A, Iniesta P, de Juan C, García-Aranda C, Díaz-López A, Benito M. Impairment of stromelysin-1 transcriptional activity by promoter mutations in high microsatellite instability colorectal tumors. Cancer Res. 2005;65:3811-3814.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Camps J, Armengol G, del Rey J, Lozano JJ, Vauhkonen H, Prat E, Egozcue J, Sumoy L, Knuutila S, Miró R. Genome-wide differences between microsatellite stable and unstable colorectal tumors. Carcinogenesis. 2006;27:419-428.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Konishi K, Issa JP. Targeting aberrant chromatin structure in colorectal carcinomas. Cancer J. 2007;13:49-55.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Ropero S, Fraga MF, Ballestar E, Hamelin R, Yamamoto H, Boix-Chornet M, Caballero R, Alaminos M, Setien F, Paz MF. A truncating mutation of HDAC2 in human cancers confers resistance to histone deacetylase inhibition. Nat Genet. 2006;38:566-569.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, Sweet-Cordero A, Ebert BL, Mak RH, Ferrando AA. MicroRNA expression profiles classify human cancers. Nature. 2005;435:834-838.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Calin GA, Croce CM. MicroRNA signatures in human cancers. Nat Rev Cancer. 2006;6:857-866.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Gaur A, Jewell DA, Liang Y, Ridzon D, Moore JH, Chen C, Ambros VR, Israel MA. Characterization of microRNA expression levels and their biological correlates in human cancer cell lines. Cancer Res. 2007;67:2456-2468.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Calin GA, Croce CM. MicroRNA-cancer connection: the beginning of a new tale. Cancer Res. 2006;66:7390-7394.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 787]  [Cited by in F6Publishing: 844]  [Article Influence: 46.9]  [Reference Citation Analysis (0)]
55.  Chang TC, Yu D, Lee YS, Wentzel EA, Arking DE, West KM, Dang CV, Thomas-Tikhonenko A, Mendell JT. Widespread microRNA repression by Myc contributes to tumorigenesis. Nat Genet. 2008;40:43-50.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Thomson JM, Newman M, Parker JS, Morin-Kensicki EM, Wright T, Hammond SM. Extensive post-transcriptional regulation of microRNAs and its implications for cancer. Genes Dev. 2006;20:2202-2207.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Jazdzewski K, Murray EL, Franssila K, Jarzab B, Schoenberg DR, de la Chapelle A. Common SNP in pre-miR-146a decreases mature miR expression and predisposes to papillary thyroid carcinoma. Proc Natl Acad Sci USA. 2008;105:7269-7274.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Saito Y, Liang G, Egger G, Friedman JM, Chuang JC, Coetzee GA, Jones PA. Specific activation of microRNA-127 with downregulation of the proto-oncogene BCL6 by chromatin-modifying drugs in human cancer cells. Cancer Cell. 2006;9:435-443.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Lujambio A, Ropero S, Ballestar E, Fraga MF, Cerrato C, Setién F, Casado S, Suarez-Gauthier A, Sanchez-Cespedes M, Git A. Genetic unmasking of an epigenetically silenced microRNA in human cancer cells. Cancer Res. 2007;67:1424-1429.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Lujambio A, Calin GA, Villanueva A, Ropero S, Sánchez-Céspedes M, Blanco D, Montuenga LM, Rossi S, Nicoloso MS, Faller WJ. A microRNA DNA methylation signature for human cancer metastasis. Proc Natl Acad Sci USA. 2008;105:13556-13561.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Toyota M, Suzuki H, Sasaki Y, Maruyama R, Imai K, Shinomura Y, Tokino T. Epigenetic silencing of microRNA-34b/c and B-cell translocation gene 4 is associated with CpG island methylation in colorectal cancer. Cancer Res. 2008;68:4123-4132.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Suzuki H, Yamamoto E, Nojima M, Kai M, Yamano HO, Yoshikawa K, Kimura T, Kudo T, Harada E, Sugai T. Methylation-associated silencing of microRNA-34b/c in gastric cancer and its involvement in an epigenetic field defect. Carcinogenesis. 2010;31:2066-2073.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Kumar MS, Lu J, Mercer KL, Golub TR, Jacks T. Impaired microRNA processing enhances cellular transformation and tumorigenesis. Nat Genet. 2007;39:673-677.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Viswanathan SR, Daley GQ. Lin28: A microRNA regulator with a macro role. Cell. 2010;140:445-449.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Chendrimada TP, Gregory RI, Kumaraswamy E, Norman J, Cooch N, Nishikura K, Shiekhattar R. TRBP recruits the Dicer complex to Ago2 for microRNA processing and gene silencing. Nature. 2005;436:740-744.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Haase AD, Jaskiewicz L, Zhang H, Lainé S, Sack R, Gatignol A, Filipowicz W. TRBP, a regulator of cellular PKR and HIV-1 virus expression, interacts with Dicer and functions in RNA silencing. EMBO Rep. 2005;6:961-967.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Paroo Z, Ye X, Chen S, Liu Q. Phosphorylation of the human microRNA-generating complex mediates MAPK/Erk signaling. Cell. 2009;139:112-122.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 279]  [Cited by in F6Publishing: 273]  [Article Influence: 18.2]  [Reference Citation Analysis (0)]
68.  Melo SA, Ropero S, Moutinho C, Aaltonen LA, Yamamoto H, Calin GA, Rossi S, Fernandez AF, Carneiro F, Oliveira C. A TARBP2 mutation in human cancer impairs microRNA processing and DICER1 function. Nat Genet. 2009;41:365-370.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Melo SA, Moutinho C, Ropero S, Calin GA, Rossi S, Spizzo R, Fernandez AF, Davalos V, Villanueva A, Montoya G. A genetic defect in exportin-5 traps precursor microRNAs in the nucleus of cancer cells. Cancer Cell. 2010;18:303-315.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Melo SA, Esteller M. A precursor microRNA in a cancer cell nucleus: get me out of here! Cell Cycle. 2011;10:922-925.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Kumar MS, Pester RE, Chen CY, Lane K, Chin C, Lu J, Kirsch DG, Golub TR, Jacks T. Dicer1 functions as a haploinsufficient tumor suppressor. Genes Dev. 2009;23:2700-2704.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Hill DA, Ivanovich J, Priest JR, Gurnett CA, Dehner LP, Desruisseau D, Jarzembowski JA, Wikenheiser-Brokamp KA, Suarez BK, Whelan AJ. DICER1 mutations in familial pleuropulmonary blastoma. Science. 2009;325:965.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Kim MS, Oh JE, Kim YR, Park SW, Kang MR, Kim SS, Ahn CH, Yoo NJ, Lee SH. Somatic mutations and losses of expression of microRNA regulation-related genes AGO2 and TNRC6A in gastric and colorectal cancers. J Pathol. 2010;221:139-146.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Mori Y, Yin J, Sato F, Sterian A, Simms LA, Selaru FM, Schulmann K, Xu Y, Olaru A, Wang S. Identification of genes uniquely involved in frequent microsatellite instability colon carcinogenesis by expression profiling combined with epigenetic scanning. Cancer Res. 2004;64:2434-2438.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Banerjea A, Ahmed S, Hands RE, Huang F, Han X, Shaw PM, Feakins R, Bustin SA, Dorudi S. Colorectal cancers with microsatellite instability display mRNA expression signatures characteristic of increased immunogenicity. Mol Cancer. 2004;3:21.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Kim H, Nam SW, Rhee H, Shan Li L, Ju Kang H, Hye Koh K, Kyu Kim N, Song J, Tak-Bun Liu E, Kim H. Different gene expression profiles between microsatellite instability-high and microsatellite stable colorectal carcinomas. Oncogene. 2004;23:6218-6225.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Giacomini CP, Leung SY, Chen X, Yuen ST, Kim YH, Bair E, Pollack JR. A gene expression signature of genetic instability in colon cancer. Cancer Res. 2005;65:9200-9205.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Kruhøffer M, Jensen JL, Laiho P, Dyrskjøt L, Salovaara R, Arango D, Birkenkamp-Demtroder K, Sørensen FB, Christensen LL, Buhl L. Gene expression signatures for colorectal cancer microsatellite status and HNPCC. Br J Cancer. 2005;92:2240-2248.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Lanza G, Ferracin M, Gafà R, Veronese A, Spizzo R, Pichiorri F, Liu CG, Calin GA, Croce CM, Negrini M. mRNA/microRNA gene expression profile in microsatellite unstable colorectal cancer. Mol Cancer. 2007;6:54.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Earle JS, Luthra R, Romans A, Abraham R, Ensor J, Yao H, Hamilton SR. Association of microRNA expression with microsatellite instability status in colorectal adenocarcinoma. J Mol Diagn. 2010;12:433-440.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Slattery ML, Wolff E, Hoffman MD, Pellatt DF, Milash B, Wolff RK. MicroRNAs and colon and rectal cancer: differential expression by tumor location and subtype. Genes Chromosomes Cancer. 2011;50:196-206.  [PubMed]  [DOI]  [Cited in This Article: ]
82.  Nielsen BS, Jørgensen S, Fog JU, Søkilde R, Christensen IJ, Hansen U, Brünner N, Baker A, Møller S, Nielsen HJ. High levels of microRNA-21 in the stroma of colorectal cancers predict short disease-free survival in stage II colon cancer patients. Clin Exp Metastasis. 2011;28:27-38.  [PubMed]  [DOI]  [Cited in This Article: ]
83.  Kaelin WG. The concept of synthetic lethality in the context of anticancer therapy. Nat Rev Cancer. 2005;5:689-698.  [PubMed]  [DOI]  [Cited in This Article: ]
84.  Martin SA, Lord CJ, Ashworth A. Therapeutic targeting of the DNA mismatch repair pathway. Clin Cancer Res. 2010;16:5107-5113.  [PubMed]  [DOI]  [Cited in This Article: ]
85.  Fong PC, Boss DS, Yap TA, Tutt A, Wu P, Mergui-Roelvink M, Mortimer P, Swaisland H, Lau A, O'Connor MJ. Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. N Engl J Med. 2009;361:123-134.  [PubMed]  [DOI]  [Cited in This Article: ]
86.  Vilar E, Chow A, Raskin L, Iniesta MD, Mukherjee B, Gruber SB. Preclinical testing of the PARP inhibitor ABT-888 in microsatellite instable colorectal cancer. J Clin Oncol. 2009;27:11028 (abstract).  [PubMed]  [DOI]  [Cited in This Article: ]
87.  Vilar E, Bartnik CM, Stenzel SL, Raskin L, Ahn J, Moreno V, Mukherjee B, Iniesta MD, Morgan MA, Rennert G. MRE11 deficiency increases sensitivity to poly(ADP-ribose) polymerase inhibition in microsatellite unstable colorectal cancers. Cancer Res. 2011;71:2632-2642.  [PubMed]  [DOI]  [Cited in This Article: ]
88.  Farmer H, McCabe N, Lord CJ, Tutt AN, Johnson DA, Richardson TB, Santarosa M, Dillon KJ, Hickson I, Knights C. Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature. 2005;434:917-921.  [PubMed]  [DOI]  [Cited in This Article: ]
89.  Martin SA, McCarthy A, Barber LJ, Burgess DJ, Parry S, Lord CJ, Ashworth A. Methotrexate induces oxidative DNA damage and is selectively lethal to tumour cells with defects in the DNA mismatch repair gene MSH2. EMBO Mol Med. 2009;1:323-337.  [PubMed]  [DOI]  [Cited in This Article: ]
90.  Swanton C, Caldas C. Molecular classification of solid tumours: towards pathway-driven therapeutics. Br J Cancer. 2009;100:1517-1522.  [PubMed]  [DOI]  [Cited in This Article: ]
91.  Vilar E, Mukherjee B, Kuick R, Raskin L, Misek DE, Taylor JM, Giordano TJ, Hanash SM, Fearon ER, Rennert G. Gene expression patterns in mismatch repair-deficient colorectal cancers highlight the potential therapeutic role of inhibitors of the phosphatidylinositol 3-kinase-AKT-mammalian target of rapamycin pathway. Clin Cancer Res. 2009;15:2829-2839.  [PubMed]  [DOI]  [Cited in This Article: ]
92.  Duchaine TF, Slack FJ. RNA interference and micro RNA-oriented therapy in cancer: rationales, promises, and challenges. Curr Oncol. 2009;16:61-66.  [PubMed]  [DOI]  [Cited in This Article: ]
93.  Bader AG, Brown D, Winkler M. The promise of microRNA replacement therapy. Cancer Res. 2010;70:7027-7030.  [PubMed]  [DOI]  [Cited in This Article: ]
94.  Melo S, Villanueva A, Moutinho C, Davalos V, Spizzo R, Ivan C, Rossi S, Setien F, Casanovas O, Simo-Riudalbas L. Small molecule enoxacin is a cancer-specific growth inhibitor that acts by enhancing TAR RNA-binding protein 2-mediated microRNA processing. Proc Natl Acad Sci USA. 2011;108:4394-4399.  [PubMed]  [DOI]  [Cited in This Article: ]