Review Open Access
Copyright ©The Author(s) 2023. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Jan 7, 2023; 29(1): 1-18
Published online Jan 7, 2023. doi: 10.3748/wjg.v29.i1.1
Emerging roles of non-coding RNAs in colorectal cancer oxaliplatin resistance and liquid biopsy potential
Zheng-Dong Luo, Yi-Feng Wang, Yu-Xiao Zhao, Long-Chen Yu, Tian Li, Ying-Jing Fan, Shun-Jie Zeng, Yi Zhang, Xin Zhang, Department of Clinical Laboratory, Qilu Hospital of Shandong University, Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan 250012, Shandong Province, China
Yan-Li Zhang, Department of Clinical Laboratory, Shandong Provincial Third Hospital, Jinan 250012, Shandong Province, China
ORCID number: Zheng-Dong Luo (0000-0002-4304-2695); Yi-Feng Wang (0000-0001-5955-2449); Yu-Xiao Zhao (0000-0003-0503-8633); Long-Chen Yu (0000-0001-6101-3751); Tian Li (0000-0001-7061-6925); Ying-Jing Fan (0000-0002-3457-8121); Shun-Jie Zeng (0000-0002-5975-3280); Yan-Li Zhang (0000-0002-3424-9731); Yi Zhang (0000-0002-0440-1798); Xin Zhang (0000-0003-2138-5600).
Author contributions: Zhang Y and Zhang X designed the manuscript; Luo ZD, Wang YF, and Zeng SJ drafted the manuscript; Zhao YX, Yu LC, Li T, and Fan YJ collected the information; Luo ZD, Zhang YL, and Zhang X revised the manuscript; Zhang YL and Zhang X coordinated the research and provided financial support; All authors read and approved the final manuscript.
Supported by The Natural Science Foundation of Shandong Province, No. ZR2020MH238.
Conflict-of-interest statement: All authors declare that we have no other real or potential conflict of interest for this article.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Xin Zhang, MD, PhD, Additional Professor, Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China. xinzhang@sdu.edu.cn
Received: September 10, 2022
Peer-review started: September 10, 2022
First decision: September 29, 2022
Revised: October 11, 2022
Accepted: November 4, 2022
Article in press: November 4, 2022
Published online: January 7, 2023

Abstract

Colorectal cancer (CRC) is one of the most common malignancies of the digestive tract, with the annual incidence and mortality increasing consistently. Oxaliplatin-based chemotherapy is a preferred therapeutic regimen for patients with advanced CRC. However, most patients will inevitably develop resistance to oxaliplatin. Many studies have reported that non-coding RNAs (ncRNAs), such as microRNAs, long non-coding RNAs, and circular RNAs, are extensively involved in cancer progression. Moreover, emerging evidence has revealed that ncRNAs mediate chemoresistance to oxaliplatin by transcriptional and post-transcriptional regulation, and by epigenetic modification. In this review, we summarize the mechanisms by which ncRNAs regulate the initiation and development of CRC chemoresistance to oxaliplatin. Furthermore, we investigate the clinical application of ncRNAs as promising biomarkers for liquid CRC biopsy. This review provides new insights into overcoming oxaliplatin resistance in CRC by targeting ncRNAs.

Key Words: Colorectal cancer, Non-coding RNAs, Oxaliplatin, Resistance, Liquid biopsy biomarkers

Core Tip: Oxaliplatin has served as a first-line chemotherapy option for colorectal cancer (CRC). However, owing to congenital or acquired resistance, treatment failure is common in some patients with CRC. Abundant evidence has revealed that non-coding RNAs (ncRNAs) are extensively involved in cancer progression, including drug resistance. Specifically, ncRNAs mediate resistance to oxaliplatin by mediating drug carriers, tumor microenvironment, resistance-related signaling pathways, and patterns of cell death. Importantly, we investigated the potential and clinical application values of these ncRNAs as liquid biopsy markers for CRC.



INTRODUCTION

Colorectal cancer (CRC) is considered to be the leading cause of death associated with malignancy of the gastrointestinal tract, with approximately 1932 million new cases and 935000 deaths in 2020[1]. Currently, surgery is the preferred treatment option for early CRC patients; however, it has few clinical benefits for advanced patients due to high rates of postoperative metastasis and recurrence[2,3]. The annual survival rate for stage III CRC patients is about 30%-60%; this figure drops to just 10% for stage IV patients[4,5]. The combination of oxaliplatin, 5-fluorouracil, and leucovorin (FOLFOX) has significantly increased the overall survival of patients and now serves as a first-line standard regimen for metastatic CRC (mCRC)[6]. Oxaliplatin is a widely used third-generation platinum analog that functions by reacting with DNA to form hydration derivatives with intra- and inter-strand crosslinks eventually resulting in cell death[7]. However, oxaliplatin has not been satisfactory in improving the survival rate of some patients due to drug resistance[8]. Moreover, some toxic side effects persist after oxaliplatin treatment; these include peripheral neurotoxicity, which further hinders chemotherapy[9]. Fortunately, oxaliplatin-based chemotherapy retreatment strategies are being optimized to provide further personalized therapy for patients with mCRC[10]. Therefore, it is of great importance to understand the mechanism by which oxaliplatin resistance occurs, and to identify new biomarkers that can aid prediction of the outcomes for CRC patients treated with oxaliplatin-based therapy.

Although the processes of chemoresistance in CRC are intricate and inconclusive[11], ongoing research has revealed multiple oxaliplatin resistance mechanisms. Several studies have confirmed that chemoresistance is associated with the dysregulation of efflux proteins and drug-metabolizing enzymes, such as ABC transporters and GSTP1; these enzymes mediate drug uptake, transport, and toxicity[12,13]. Furthermore, accumulating evidence suggests that epithelial-mesenchymal transition (EMT)[14] and DNA damage repair[15] are driving factors that result in chemoresistance and tumor progression. Many studies have reported that various signaling pathways, such as the TGF-β/Smad[16], JNK/p38 MAPK[17], Wnt/β-catenin[18], and MEK/ERK/ELK1[19] pathways are closely associated with oxaliplatin resistance. As the main regulatory components of the tumor microenvironment (TME), cancer-associated fibroblasts (CAFs)[20] and tumor-associated macrophages[21] can modulate cancer chemotherapy resistance through complex mechanisms of crosstalk. Of note, emerging studies have confirmed that cell death mechanisms play a pivotal role in chemoresistance. It has been reported that apoptosis inhibition[22] and autophagy dysregulation[23] are underlying mechanisms leading to chemoresistance. Furthermore, numerous studies have identified that several additional death mechanisms can mediate CRC chemoresistance, including ferroptosis[24], pyroptosis[25], necroptosis[26], and others. However, the detailed molecular mechanisms by which these cell death modes lead to oxaliplatin resistance are unclear and require further investigation.

Non-coding RNAs (ncRNAs) refer to RNA molecules transcribed from genes that cannot encode proteins, accounting for more than 90% of human gene transcripts. ncRNAs can be divided into either small ncRNAs (< 200 nucleotides) or long non-coding RNAs (lncRNAs) (> 200 nucleotides) based on their length[27-29]. In the last decade, circular RNAs (circRNAs), a type of circular RNA generated in the splicing process of pre-mRNA, have emerged as a focus of research[30] (Figure 1). A growing number of studies have revealed that ncRNAs play critical roles in the occurrence and development of multiple human diseases via various mechanisms, such as epigenetics, transcription, and post-transcription[31,32]. Moreover, microRNA (miRNAs), lncRNAs, and circRNAs are three of the more widely studied types of ncRNAs and have been shown to mediate tumor progression by regulating tumor cell proliferation, aggression, metastasis, apoptosis, and drug resistance, amongst other pathways[33,34]. Drug resistance is one of the key factors in malignancy treatment failure, and whether this is mediated by ncRNAs has attracted increasing attention. In recent years, the rapid development of next-generation sequencing technologies has expanded the understanding of tumor pathogenesis and drug resistance mechanisms, and an increasing number of tumor chemoresistance-associated differential ncRNAs have been identified[35-37]; these developments have provided the expansion of open datasets of candidate ncRNAs for basic research and clinical application. Currently, the targeting of ncRNAs as biomarkers in the clinic is being actively facilitated[38]. As such, a systematic understanding of the underlying roles of ncRNAs in malignancies provides valuable insights into overcoming chemoresistance in CRC patients.

Figure 1
Figure 1 Classification of non-coding RNAs. Non-coding RNAs can be divided into either small ncRNAs (< 200 nucleotides) or long non-coding RNAs (> 200 nucleotides) based on their length. Among these, small ncRNAs consist of ribosomal RNAs, transfer RNAs, small nucleosome/spliced RNAs, microRNAs, small interfering RNAs, PiWI-Interacting RNAs, and small nucleolar RNAs. Furthermore, the circular RNAs generated in the splicing process of pre-mRNA are also considered to be special ncRNAs. ncRNAs: Non-coding RNAs; lncRNAs: Long non-coding RNAs; rRNAs: Ribosomal RNAs; tRNAs: Transfer RNAs; snRNAs: Small nucleosome/spliced RNAs; miRNAs; MicroRNAs; siRNAs: Small interfering RNAs; piRNAs: PiWI-Interacting RNAs; snoRNAs: Small nucleolar RNAs.

Although tissue biopsy is the gold standard for malignancy diagnosis, the availability of clinical samples is often limited, and subsequently, tumor heterogeneity may not be reflected in analysis and is therefore not suitable for longitudinal clinical monitoring[39]. As a noninvasive approach, liquid biopsy has been extensively applied for the real-time monitoring of variations in tumor dynamics in body fluids including blood, ascites, and others. With the development of novel molecular detection technologies, growing evidence suggests that circulating tumor cells, circulating tumor DNA, circulating free nucleic acids, tumor-educated platelet, exosomes, etc[40,41] have gradually become prominent liquid biopsy hallmarks. Owing to their stability and high abundance, circulating ncRNAs can provide substantial details regarding tumor biology and therapeutic efficacy, and have become candidate biomarkers for earlier diagnosis, therapeutic monitoring, and prognosis evaluation of malignancies[42,43].

In this study, we performed a systematic literature review to identify the latent mechanisms of ncRNAs (miRNAs, lncRNAs, and circRNAs) in CRC oxaliplatin resistance. In particular, exosomal ncRNAs, as one of the most promising biomarkers in liquid biopsies, are expected to improve diagnostic, therapeutic, and drug monitoring in CRC patients.

OVERVIEW OF THE REGULATORY ROLES OF NCRNAS IN CRC OXALIPLATIN RESISTANCE
Classification and biological functions of miRNAs

MiRNAs are a class of small ncRNAs of approximately 22 nucleotides in length; they are produced by two RNase III proteins, Drosha and Dicer[44]. In general, miRNAs bind to the 3' untranslated region (UTR) of their target mRNAs resulting in cleavage or translational repression in the cytosol[45]. Intriguingly, numerous recent studies have demonstrated that some miRNAs activate gene transcription unconventionally in the nucleus by targeting enhancers, such miRNAs are known as nuclear activating miRNAs[46]. Moreover, enhancers (including super-enhancers) have been revealed to synergistically promote NamiRNA biogenesis and activate the expression of proximal genes[47,48]. Furthermore, studies have reported that the dysregulation of miRNAs may play an important role in CRC oxaliplatin resistance, as illustrated in Table 1.

Table 1 Dysregulation of microRNAs and oxaliplatin resistance in colorectal cancer.
MiRNAs
Expression1
Targets and pathways
Ref.
miR-135b-5pMUL1/ULK1[49]
miR-454-3pPTEN[50]
miR-19aPTEN/PI3K/AKT[51]
miR-543PTEN/Akt/mTOR[52]
miR-107CAB39/AMPK/mTOR[53]
miR-503-5pPUMA[54]
miR-744BIN1[55]
miR-5000-3pUSP49[56]
miR-146b-5pWBSCR22[57]
miR-19b-3pSMAD4[58]
miR-1278-[59]
miR-506Wnt/β-catenin[60]
miR-200b-3pTUBB3[61]
miR-122XIAP[62]
miR-193a-5pCXCR4[63]
miR-483-3pFAM171B[64]
miR-325HSPA12B/PI3K/AKT/Bcl-2[65]
miR-195-5p-[66]
miR-497-5p-
miR-34aTGF-β/Smad4[68]
miR-27b-3p-[69]
Oncogenic miRNAs mediate oxaliplatin resistance in CRC

MiR-135b-5p has been confirmed to be upregulated in the serum of CRC patients, mechanistically this has been shown to induce protective autophagy via the MUL1/ULK1 signaling pathway, a process that contributes to oxaliplatin resistance[49]. Another miRNA, miR-454-3p, is highly expressed in oxaliplatin-resistant CRC cells compared to oxaliplatin-sensitive cells and promoted oxaliplatin resistance by inhibiting PTEN expression and activation of the AKT pathway[50]. Similarly, targeting miR-19a with high expression in oxaliplatin-resistant CRC cell lines could promote oxaliplatin sensitization of resistant cells through activation of the PTEN/PI3K/AKT pathway[51]. Compounding these findings, it has been revealed that dichloroacetate, a pyruvate dehydrogenase kinase inhibitor, can enhance the chemosensitivity of oxaliplatin-resistant CRC cells through the miR-543/PTEN/Akt/ mTOR pathway and the miR-107/CAB39/AMPK/mTOR pathway[52,53]. In addition, an unbiased microRNA array demonstrated that miR-503-5p was up-regulated in oxaliplatin-resistant CRC cells; overexpression of miR-503-5p conferred resistance to oxaliplatin-induced apoptosis by inhibiting PUMA expression[54]. Further studies have revealed that by targeting BNI1, up-regulation of miR-744 enhanced the resistance of T84 and HCT116 cells to oxaliplatin[55]. Moreover, the oncogenic miR-5000-3p was upregulated in CRC tissues and oxaliplatin-resistant CRC cells, this miRNA negatively regulated USP9 to facilitate CRC chemoresistance[56]. Additionally, natural killer cells contributed to enhancing the sensitivity of oxaliplatin resistant CRC cells through the microRNA-146b-5p/WBSCR22 axis[57]. Remarkably, miR-19b-3p has been documented to be the most significantly upregulated candidate miRNA in colon cancer tissues, its expression correlates with tumorous histologic grading, staging, and poor prognosis in patients. Importantly, miR-19b-3p facilitated proliferation, curbed apoptosis, and induced oxaliplatin resistance in CRC cells by targeting SMAD4[58].

Tumor-suppressive miRNAs mediate oxaliplatin resistance in CRC

Interestingly, it has been demonstrated by some that tumor-suppressive miRNAs are associated with oxaliplatin resistance. Studies have identified that miR-1278 was significantly downregulated in CRC tissues, and that overexpression of miR-1278 inhibited CRC progression and enhanced oxaliplatin sensitivity through targeting of the KIF5B/BTG2 axis[59]. Similarly, miR-506 was weakly expressed in chemoresistant CRC cells, but its overexpression has been confirmed to impair the resistance of HCT116 cells to oxaliplatin via the Wnt/β-catenin pathway[60]. Moreover, miR-200b-3p was down-regulated in oxaliplatin-resistant CRC tissues and cells (HT29-OR and HCT116-OR), and studies revealed that miR-200b-3p reversed oxaliplatin resistance of CRC cells by targeting TUBB3[61]. Furthermore, miR-122, which was down-regulated in oxaliplatin-resistant SW480 and HT29 cells, has been demonstrated to be overexpressed in CRC cells, this could enhance the chemosensitivity of CRC by inhibiting the expression of XIAP[62]. As a tumor suppressor, miR-193a-5p can act directly upon CXCR4 to mitigate the chemosensitivity of CRC cells to 5-FU and oxaliplatin[63]. Using sequencing, Liang et al[64] discovered that miR-483-3p was negatively correlated with FAM171B expression, and targeting the miR-483-3p/FAM171B regulatory axis could enhance the sensitivity of CRC cells to oxaliplatin. Moreover, miR-325 mimics were observed to prevent the development of oxaliplatin resistance in CRC via interference with the HSPA12B/PI3K/AKT/Bcl-2 axis[65]. Finally, a proteomic analysis study reported that regulation of miR-195-5p and miR-497-5p is a potential strategy for alleviating oxaliplatin resistance in CRC cells[66].

As an additional factor, several groups showed that the interaction between ncRNAs and autophagy exerted significant roles in the therapeutic resistance of CRC[67]. Sun et al[68] demonstrated that miR-34a was expressed at low levels in oxaliplatin-resistant CRC patients and cells. Overexpression of miR-34a contributed to facilitating the sensitivity of CRC cells to oxaliplatin by regulating the TGF-β/Smad4 pathway resulting in the inhibition of macroautophagy. Furthermore, c-Myc has been shown to promote oxaliplatin resistance in CRC by integrating into the miR-27B promoter region and further mediating activation of the miR-27b-3p/ATG10 axis[69].

It is noteworthy that miR-181a could suppress the expression of BIRC6, a protein inhibitor of apoptosis, by directly targeting the 3'-UTR of BIRC6 mRNA[70]. Conversely, BIRC6 was observed to be significantly upregulated in acquired oxaliplatin-resistant CRC cells when compared to parental cells[71]. Considering that knockdown of BIRC6 helped enhance the chemosensitivity of CRC cells to oxaliplatin, targeting BIRC6 using miR-181a mimics may be a potential strategy to reverse oxaliplatin resistance in CRC.

Classification and biological functions of lncRNAs

LncRNAs are defined as a class of ncRNA molecules with transcript lengths of more than 200 nucleotides; they do not encode proteins due to their lack of an open reading frame[72]. LncRNAs can be classified into intergenic, intronic, sense, antisense, and bidirectional lncRNAs based on their genomic localization[73]. Alternatively, there are four categories of lncRNAs based on their functional mechanisms, including signal, decoy, guide, and scaffold lncRNAs[74,75]. It has been reported that lncRNAs can regulate transcription, translation, RNA stability, and alternative splicing[76-79]. It is noteworthy that numerous studies have revealed that aberrant lncRNAs (oncogenic and tumor suppressive lncRNAs) contribute to regulating the mechanism of oxaliplatin resistance through multiple cellular mechanisms, including but not limited to DNA damage repair, interference with drug influx and efflux, regulation of the cell cycle and apoptosis, and activation of signaling pathways[80,81]. Table 2 provides an overview of some aberrant lncRNAs and their target molecules in the context of oxaliplatin resistance in CRC.

Table 2 Dysregulation of long non-coding RNAs and oxaliplatin resistance in colorectal cancer.
LncRNAs
Expression1
Targets and pathways
Ref.
GIHCG-[82]
lncARSR-[83]
HOTAIRmiR-1277-5p/ZEB1[85]
MALAT1MALAT1/miR-218/EZH2[86]
miR-324-3p/ADAM17[88]
OIP5-AS1miR-137[89]
Linc00152miR-193a-3p/ERBB4/AKT[90]
CASC15miR-145/ABCC1[92]
CBR3-AS1miR-145-5p[93]
CRNDEmiR-136/E2F1[94]
LINC00460miR-149-5p/miR-150-5p/p53[95]
KCNQ1OT1miR-34a/Atg4B[96]
MIR155HGmiR-650/Annexin A2[91]
lnc-RP11-536 K7.3SOX2/HIF-1α/USP7[97]
TUG1GATA6/BMP[98]
LUCAT1UBA52/RPL40/MDM2/p53[99]
PiHLEZH2/HMGA2/PI3K/Akt[100]
ELFN1-AS1EZH2/DNMT3a/MEIS1[102]
SNHG5STAU1[103]
CCAT2BOP1/AURKA[104]
NBAT-1WWC3/LATS1/YAP[105]
MEG3-[106]
miR-141/PDCD4[107]
lnc-APpep-AP/TALDO1[108]
lncRNA PVT1-hsa-miR-297/GSTA2[109]
Oncogenic lncRNAs as regulators of CRC oxaliplatin resistance

Studies discovered that high expression of the lncRNA GIHCG promoted the proliferation, migration, and invasion of tumor cells, and enhanced their resistance to 5-FU and oxaliplatin[82]. Similarly, overexpression of the lncRNA ARSR reduced cell apoptosis and induced oxaliplatin resistance[83]. Notably, emerging evidence has revealed that lncRNAs could target mRNAs to promote chemoresistance[84]. For instance, upregulation of the lncRNA HOTAIR has been reported to facilitate EMT in a ZEB1-dependent manner by negatively regulating miR-1277-5p, a process that is involved in hypoxia-induced oxaliplatin resistance[85]. We previously discovered that metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) was overexpressed in CRC tissues compared to paired noncancerous tissues. Correspondingly, the overexpression of MALAT1 suppressed E-cadherin expression and promoted oxaliplatin-induced EMT by interacting with EZH2. Furthermore, upregulated MALAT1 further inhibited miR-218 expression, resulting in poor response to oxaliplatin-based chemotherapy in CRC patients[86]. Numerous studies have demonstrated that some endogenous transcripts (such as endogenous pseudogenes, lncRNAs, and circRNAs) can complement miRNAs in sequences and inhibit their expression, resulting in the upregulation of target gene expression. This phenomenon is called the miRNA “sponge” effect[87]. Analogously, Fan et al[88] demonstrated that MALAT1 could function as a “sponge” for miR-324-3p and increase the expression of ADAM17 to facilitate resistance to oxaliplatin in CRC cells.

Moreover, lncRNA Opa-interacting protein 5 antisense RNA 1 can complement the sites of miR-137 in sequence, sponging miR-137 and inhibiting its expression, thus conferring oxaliplatin resistance in CRC cells[89]. It was reported that Linc00152 increased ERB-B2 receptor tyrosine kinase 4 expression by acting as a “sponge” for miR-193a-3p resulting in the promoted phosphorylation of AKT at Thr308 and Ser47; this process mediated oxaliplatin resistance in CRC cells[90]. In addition, researchers have illustrated that the lncRNA MIR155HG promoted M2 macrophage polarization and enhanced oxaliplatin resistance in CRC cells by regulating the miR-650/Annexin A2 axis[91]. Similarly, other lncRNAs such as CASC15[92], CBR3-AS1[93], CRNDE[94], LINC00460[95], and KCNQ1OT1[96] have been reported to serve as “sponges” for miRNAs, thereby mediating oxaliplatin resistance in CRC.

Several research groups have revealed that lncRNAs could stabilize functional proteins, and mediate the chemoresistance of cancers. For example, lncRNA-RP11-536 K7.3 contributed to oxaliplatin resistance by recruiting SOX2 to activate deubiquitinase USP7 and stabilize the protein HIF-1α in CRC cells[97]. Furthermore, the lncRNA TUG1 induced oxaliplatin resistance in CRC stem cells and inhibited cell apoptosis by interacting with GATA6[98]. Moreover, two additional studies have further revealed that the lncRNAs LUCAT1 and PiHL mediated oxaliplatin resistance in CRC cells by interacting with UBA52 and EZH2 proteins, respectively[99,100].

LncRNAs have also been demonstrated to be regulators of gene expression by enhancing the stability of mRNA through epigenetic mechanisms[101]. Studies have elucidated that the lncRNA ELFN1-AS1 suppressed myeloid ecotype virus insertion site 1 transcription and promoted oxaliplatin resistance by interacting with EZH2 and DNA methyltransferase 3 alpha after localization to the promoter region of MEIS1 gene[102]. In addition, overexpression of the lncRNA SNHG 5 contributed to the proliferation of CRC cells and their resistance to oxaliplatin-induced apoptosis by blocking the degradation of SPATS 2 by STAU 1[103]. Of note, it has been identified that in microsatellite stable CRC, overexpression of the lncRNA CCAT2 could induce chromosomal instability, and enhance 5-FU and oxaliplatin-resistance by the upregulation of ribosomal biogenesis factor and the activation of aurora kinase A[104].

Tumor-suppressive lncRNAs as regulators of CRC oxaliplatin resistance

Previously, several tumor suppressor lncRNAs were reported to be associated with CRC oxaliplatin resistance. For instance, the expression levels of NBAT-1 and MEG3 in CRC tissues were low compared with normal adjacent tissues, especially in oxaliplatin-resistant patients[105,106]. Mechanistically, NBAT-1 was revealed to inhibit the growth of oxaliplatin-resistant CRC cells by activating the WWC 3/LATS 1/YAP pathway after acting as a “sponge” for miR-454[105]. Similarly, MEG3 promoted the sensitivity of CRC cells to oxaliplatin by regulating the miR-141/PDCD4 axis[107]. It was further reported that lnc-AP is a lncRNA with coding potential, and is highly associated with oxaliplatin resistance in CRC; lnc-AP encodes the short peptide pep-AP that enhances sensitization of CRC cells to oxaliplatin via the pep-AP/TALDO1 pathway[108]. Intriguingly, an epidemiological investigation demonstrated that an rs2278176 CT/TT mutation in the lncRNA PVT1 increased the chemosensitivity of CRC patients to FOLFOX compared to CRC patients carrying distinct genotypes[109].

Of note, when compared to parental cells, the lncRNA CRNDE was significantly down-regulated in oxaliplatin-resistant cells and can be considered a predictor of oxaliplatin treatment response and tumor prognosis[110]. Interestingly, this lncRNA can be combined with arginine-rich splicing factor 6 to reduce its stability, knockdown of the latter can inhibit autophagy and increase the sensitivity of cancer cells to oxaliplatin by the alternative splicing of PICALM[111]. Given that up-regulated expression of CRNDE aids an increase in the sensitivity of cancer cells to oxaliplatin, we speculate that increasing CRNDE expression and decreasing the stability of SRSF6 may be a promising strategy to ameliorate CRC resistance to oxaliplatin.

Classification and biological functions of circRNAs

Most circRNAs consist of covalently closed loop single-stranded RNAs produced by the back-splicing of exon precursor mRNAs that are more stable than their linear precursor gene[30]. Currently, circRNAs consist of four general categories: Exonic circRNAs, circular intronic RNAs, exon-intron circRNAs, and circRNA from other sources (such as antisense circular RNA or intergenic circular RNA) based on their structural domains and biogenesis features[112,113]. As vital biological regulators, circRNAs have been reported to widely participate in multiple oxaliplatin resistance-related mechanisms, such as apoptosis, autophagy, glycolysis, TME, EMT, DNA damage repair, etc[114]. Studies identified that hsa_circ_0040809 was highly expressed in CRC cells and tissues; hsa_circ_0040809 upregulates DNMT1 expression by competitively combining with miR-515-5p, thus suppressing apoptosis and promoting cancer progression[115]. Conversely, as a tumor suppressor, circTADA2A was found to be down-regulated in cancer cells and tissues. Overexpression of circTADA2A elevated the expression of KLF14 by targeting miR-374a-3p, thereby promoting cell apoptosis and inhibiting tumor growth[116]. Moreover, has_circ_0001666 is a circRNA originating from exons 2, 3, and 4 of the host gene FAM120B. Studies have demonstrated that hsa_circ_0001666 competitively bound to miR-576-5p to increase PCDH10 expression, which in turn restrained cell stemness and EMT, and the Wnt/β-catenin pathway[117]. Zhang et al[118] revealed that the knockdown of circRNA_103948 contributed to enhanced autophagy of CRC cells by targeting the miR-1236-3p/TPT1 signaling pathway, inhibiting tumor growth. In addition, hypoxia facilitated circCCDC66 expression, which boosted the development of CRC by modulating the miR-3140/autophagy axis[119]. Furthermore, studies observed that hsa_circ_101555 was more highly expressed in CRC tissues than in normal tissues and was associated with adverse prognosis of patients. Inhibition of circ101555 resulted in the initiation of cell apoptosis and devastated DNA repair by activating the miR-597-5p/CDK6/RPA3 axis, thereby repressing the progression of CRC[120]. These studies suggested that circRNAs might be closely associated with the process leading to CRC oxaliplatin resistance.

CircRNAs regulate CRC oxaliplatin resistance

The majority of circRNAs containing miRNA response elements (MREs) can regulate gene expression and signaling pathways by functioning as ceRNAs, mediating tumor chemoresistance[121] (Table 3). Studies have identified that circular RNA protein tyrosine kinase 2 was significantly up-regulated in CRC tissues compared to normal tissues, and its high levels of expression promoted CRC progression and oxaliplatin resistance by regulation of the miR-136-5p/YTHDF1 axis[122]. Similarly, circ_0032833 was highly expressed in FOLFOX-resistant CRC cells, and knock-down of circ_0032833 could promote apoptosis and partially enhance the sensitivity of CRC cells to 5-FU and oxaliplatin through the activity of miR-125-5p/MSI1[123]. Lai et al[124] also demonstrated that hsa_circ_0079662 functioned as a “sponge” for hsa-mir-324-5p and activated HOXA9 through activation of the TNF-α pathway; this process induced oxaliplatin-resistance in CRC cells. Furthermore, our previous research revealed that circHIPK3 was highly expressed in chemoresistance CRC patients, and this circRNA was strongly associated with tumor size, regional lymph node metastasis, and distant metastasis. The following functional investigation revealed that circHIPK3 facilitated oxaliplatin resistance, but not 5-FU resistance, by restraining autophagy-related cell death. Mechanistically, circHIPK3 acted as a ceRNA and a “sponge” for miR-637 resulting in activation of the STAT3/Bcl-2/Beclin1 pathway, and subsequent induction of oxaliplatin-resistance in CRC cells[125].

Table 3 Dysregulation of circular RNAs and oxaliplatin resistance in colorectal cancer.
CircRNAs
Expression1
Targets and pathways
Ref.
circPTK2miR-136-5P/YTHDF1[122]
(hsa_circ_0003221)
circ_0032833miR-125-5p/MSI1[123]
hsa_circ_0079662hsa-mir-324-5p/TNF-α/HOXA9[124]
circHIPK3miR-637/STAT3/Bcl-2/Beclin1[125]
Circular CCDC66PI3KK/p-DHX9[126]
hsa_circ_32883/-[127,128]
hsa_circ_0338

Intriguingly, oxaliplatin could directly induce the overexpression of some oncogenes, further exacerbating chemoresistance. Circular CCDC66, derived from exons 6 to 11, was highly expressed in oxaliplatin-resistant CRC tissues and cells, a phenomenon caused by oxaliplatin-triggered cell stress through phosphoinositide 3-kinase related kinases-mediated phosphorylation of DHX9[126]. Despite extensive literature, many circRNAs remain to be explored in the study of oxaliplatin-resistant CRC. Abu et al[127] identified differential expression of circular RNAs when analyzing chemosensitive and chemoresistant CRC cells using microarray; hsa_circ_32883 and hsa_circ_0338 were screened as promising candidates. Subsequently, their study also identified that exosome-derived circRNAs (hsa_circ_0032883, hsa_circ_0002039, and hsa_circ_0000338) may play important roles in the manifestation of chemoresistance[128].

Studies have reported that the expression of exosomal miR-21-5p from oxaliplatin-resistant cells was significantly elevated, and therefore levels of this miRNA could act as a predictor of chemotherapy response in CRC patients[129]. Interestingly, miR-21-5p can be sponged by some tumor-suppressive circRNAs, such as circDDX17[35] and circEPB41L2[130], thus resulting in activation of the downstream PTEN/AKT pathway. Importantly, previous studies have confirmed that targeting the PTEN/PI3K/AKT/mTOR pathway partially reversed oxaliplatin resistance in CRC[51,52]. Given this, we speculate that targeting these suppressive circRNAs might contribute to the sensitization of CRC cells to oxaliplatin via the miR-21-5p/PTEN/AKT axis. Furthermore, hsa_circ_0001955 and hsa_circ_0000977, potential upstream targets for chemoresistance-associated miRNAs[49,93], were demonstrated to be dysfunctional in CRC tissue, a finding that suggests these circRNAs may be involved in the progression of CRC chemoresistance in a ceRNA-dependent manner[131]. Currently, studies regarding the roles of circRNAs in oxaliplatin resistance are less commonly reported, the precise and comprehensive mechanisms of circRNAs in CRC chemoresistance need to be explored by further study.

INTEGRATION, APPLICATION, AND CHALLENGES OF TARGETING EXOSOMAL NCRNAS IN CRC CHEMORESISTANCE
Exosomes biogenesis and characterization

Extracellular vesicles (EVs) are defined as a class of membranous vesicles that are released by cells to the extracellular matrix and play a key role in various physiological and pathological processes. According to the difference in their origin, size, content, and biological function, EVs can be roughly divided into three main subtypes—microbubbles, exosomes, and apoptotic bodies[132]. Exosomes are an emerging hallmark of liquid biopsy and have attracted much scientific attention of late, these small disc-shaped EVs have a diameter of approximately 40–150 nm and are enclosed by a lipid bilayer membrane[133]. Almost all cell types can secrete exosomes, and they also exist broadly within fluids including blood, urine, saliva, milk, ascites, cerebrospinal fluid, and others[40,134]. At present, ultracentrifugation remains the standard method for exosome extraction. However, with the emergence of novel technologies, exosome isolation and purification strategies are being continuously optimized, such as size exclusion chromatography, magnetic bead immune capture, and microfluidic-based chip technology techniques[135], these are aimed at efficiently obtaining exosomes with high abundance and quality. The characterization of exosomes has emerged as a prerequisite for their utilization, and the three basic characterization modes of morphological identification are particle size distribution and protein markers. However, more advanced features are gradually being included, in exosome characterization, such as the identification of the purity of exosomes and their uptake[135,136]. As signaling entities, numerous studies have demonstrated that exosomes exert biological effects in two ways: Firstly, exosomal membrane proteins or lipids act as ligands, directly activating receptors at the surface of target cells, generating cascade signal events and activating intracellular signaling pathways; Secondly, exosomes entrain and transport cellular signal-regulating molecules to target cells; these include DNA, lipids, proteins, and RNAs, which are extensively involved in tumorigenesis, metastasis, recurrence, and chemoresistance of cancers[137,138]. In particular, exosomal ncRNAs have received unprecedented appreciation in biomedical research and clinical application recently[139,140]. Several exosomal ncRNAs involved in CRC oxaliplatin resistance are listed in Table 4.

Table 4 Aberrant exosomal non-coding RNAs and their targets in the chemoresistance of colorectal cancer.
Exosomal ncRNAs1
Donor cells
Recipient cells
Targets and pathways
Ref.
miR-21↓THLG-293T/LG-293T cellsCRC cells-[156]
miR-208b↑CRC cellsT cellsPDCD4[145]
miR-92a-3p↑CAFs cellsCRC cellsFBXW7/MOAP1[20]
miR-46146↑CRC-R cellsCRC-S cellsPDCD10[146]
miR-1915-3p↓FHC cellsCRC cellsPFKFB3/USP2[147]
lncRNA H19↑CAFs cellsCRC cells-[141]
lncRNA CCAL↑CAFs cellsCRC cellsHuR/β-catenin[142]
cricN4BP2L2CAFs cellsCRC cellsEIF4A3/PI3K/AKT/mTOR[143]
ciRS-122↑CRC-R cellsCRC-S cellsmiR-122/PKM2[148]
circ_0094343↓NCM460 cellsCRC cellsmiR-766-5p/TRIM67[144]
The latent mechanisms of exosomal ncRNAs in CRC oxaliplatin resistance

Increasing numbers of reports have revealed that exosomal ncRNAs can mediate chemoresistance via remodeling of the tumor microenvironment. Studies have identified that lncRNA H19 is highly expressed in CRC tissues compared to normal tissues. Moreover, lncRNA H19 derived from CAFs can act as a direct “sponge” for miR-141 and activate the β-catenin pathway, thus promoting stemness and oxaliplatin resistance[141]. Similarly, exosomal miR-92a-3p was transmitted from CAFs to cancer cells, promoting chemoresistance via the Wnt/β-catenin/apoptosis pathway[20]. Moreover, CAF-derived exosomes were revealed to deliver CRC-associated lncRNA to cancer cells, interacting with the mRNA of the stable protein human antigen R and increasing the expression of β-catenin; these processes promoted oxaliplatin resistance in CRC cells[142]. Qu et al[143] also found that exosomal cricN4BP2L2 secreted by CAFs can interact with EIF4A3 and modulate the PI3K/AKT/mTOR signaling pathway, thus promoting oxaliplatin resistance and the stemness of CRC cells. In addition, exosomal circ_0094343 derived from human colonic epithelial cells (NCM460 cells) was identified to be taken up by CRC cells in which it regulated cell apoptosis and glycolysis via the miR-766-5p/TRIM67 pathway, thereby enhancing the chemosensitivity of tumor cells[144].

Conversely, tumor cells have been revealed to generate exosomes in an autocrine or paracrine manner; they are taken up by themselves or delivered to the local microenvironment, thus exerting functional regulation. Ning et al[145] demonstrated that CRC cell-derived exosomal miR-208b could promote Treg expansion by directly inhibiting the expression of programmed cell death factor 4, resulting in promoted tumor progression and oxaliplatin resistance. Similarly, studies have revealed that exosomal miR-46146 conferred chemoresistance via the targeting of programmed cell death factor 10 in CRC cells[146]. Interestingly, one particular study revealed that miR-1915-3p was down-regulated in oxaliplatin-resistant CRC cells compared to oxaliplatin- sensitive cells and that EVs-derived miR-1915-3p increased the oxaliplatin sensitivity of drug-resistant cells by targeting PFKFB3/USP2[147]. Moreover, it has been reported that hsa_circ_0005963 can be transferred from oxaliplatin-resistant cells to sensitive cells by exosomes, directly acting as a miR-122 “sponge” and increasing the expression of PKM2, which promotes glycolysis and oxaliplatin-resistance in sensitive cells[148].

Exosomal ncRNAs as potential liquid biopsy biomarkers

Liquid biopsy, in which diseases are evaluated via sampling biological fluids, can avoid the influence of tissue heterogeneity on tumor molecular typing to some extent. More and more evidence show that abundant exosomes are enriched in body fluids and participate in many physiological and pathological processes. As an emerging hallmark of liquid biopsy, exosomes are of substantial value in cancer diagnosis, monitoring, and prediction[136,140]. Studies found that the ratio of exosomal miRNAs is an effective predictor of tumor response in peritoneal metastatic patients after repeated intraperitoneal chemotherapy, as patients with high ratios of miR-223-3p/miR-29b-3p or miR-21-5p/miR-29b-3p had inferior survival outcomes compared to patients with low ratios[149]. By analogy, exosomes derived miR-21-5p, miR-1246, miR-1229-5p, and miR-96-5p were highly expressed in CRC patients and cancer cell lines resistant to 5-FU and oxaliplatin compared with those sensitive to chemotherapy. The area under the curve (AUC) of four combinations of exosomal miRNAs was 0.804, which is expected to be an effective predictor of chemotherapy[129]. In addition, researchers found that plasma-derived exosomal miRNA-125b was highly expressed in patients with progressive disease (PD) compared with the healthy control group or patients with stable disease. Importantly, some patients with PD who respond to modified fluorouracil, leucovorin, and oxaliplatin (mFOLFOX6) have obvious differences in the expression of plasma exosomal miRNA-125b before and after chemotherapy, and patients with low expression of exosomal miRNA-125b have higher progression-free survival (PFS)[150]. Furthermore, it was reported that miR-208b was highly expressed in the serum of FOLFOX-resistant CRC patients and the AUC of serum miRNA-208b was 0.771 [95% confidence interval (CI): 0.688-0.855], which is better than that of serum CEA of 0.493 (95%CI: 0.385-0.601). Their research suggests that this miRNA can be regarded as a promising liquid biopsy indicator to predict FOLFOX sensitivity in cancer patients[145]. Encouragingly, a panel of plasma exosomal miR-17-5p and miR-185-5p were successfully used to predict FOLFOX4/FOLFIRI responses in patients with advanced CRC[151].

Exosomal ncRNAs as delivery vehicles for chemosensitization

A large number of reports have revealed the prospects for the application of exosomes to overcome chemoresistance, their strong stability, intrinsic biocompatibility, low immunogenicity, and natural targeting ability making them favorable targets[152,153]. Novel research has exhibited an engineered exosome encapsulating oxaliplatin and PGM5 Antisense RNA 1 that were delivered into CRC cells, and these effectively relieved chemoresistance and inhibited tumor progression[154]. Furthermore, Pi et al[155] constructed engineered EVs enwrapped in folate nanoparticles and survivin siRNA, which significantly inhibited tumor growth in CRC xenograft mice. Similarly, miR-21 inhibitors have been co-incorporated into exosomes, resulting in enhanced chemosensitivity of CRC cells[156]. Taken together, these findings suggest that exogenous delivery of ncRNAs could serve as a novel therapeutic target for CRC.

CONCLUSION

A multitude of emerging studies have confirmed that dysfunctional ncRNAs contribute to the development of malignancies, including chemoresistance. In this review, we systematically summarized the multiple mechanisms by which ncRNAs function in CRC oxaliplatin resistance and discussed the biomedical prospects of exosomal ncRNAs as hallmarks for fluid biopsy and therapeutic targets. Figure 2 schematically illustrates the multiple regulatory roles of ncRNAs in CRC oxaliplatin resistance. Owing to the differential expression patterns of ncRNAs in cancer patients, it is considered they may be used as biomarkers for early detection, tumor staging, and clinical outcome. Although many mechanisms by which ncRNAs exert their biological effects remain to be deciphered, targeting these ncRNAs has emerged as a promising strategy to ameliorate chemoresistance.

Figure 2
Figure 2 Overview of the mechanisms by which several non-coding RNAs exert oxaliplatin resistance in colorectal cancer. Various microRNAs, long non-coding RNAs, and circular RNAs are up-regulated or down-regulated in colorectal cancer and mediate oxaliplatin resistance via a multitude of mechanisms. These mechanisms include acting as competing endogenous RNAs, generating encoded peptides, regulating autophagy, inducing apoptosis, and interfering with DNA damage and repair. In particular, several non-coding RNAs have been demonstrated to confer oxaliplatin resistance in colorectal cancer cells via exosomes. CRC: Colorectal cancer; lncRNAs: Long non-coding RNAs; miRNAs; MicroRNAs; circRNAs: Circular RNAs.

Nevertheless, the integration and application of ncRNAs in clinical practice are far from the existing research fervor and are currently being tested in experimental studies; there are still many challenges to be overcome before ncRNAs can be extensively implemented in clinics. Firstly, the results of existing research are confounding due to differing nomenclature being utilized for circRNAs. The circRNAs labeled using different naming standards make the subsequent genomic localization cumbersome. Secondly, the expression of ncRNAs in biological fluids is relatively low, and therefore accurate detection and quantification of these molecules is a prerequisite for the development of biomarkers. Furthermore, due to differences in the lengths and mechanisms by which ncRNAs function in distinct tumor types, it is a challenge to select appropriate targets for these multitudinous candidates. Exosomes may be a better intermediary for delivering ncRNAs therapies; this could be achieved in an endogenous or exogenous manner. However, it is crucial to increase the loading of ncRNAs delivered by endogenous exosomes and ensure the safety of engineered exosomes with complex structures. Measurements of the efficacy of exosomal ncRNAs in clinical applications cannot be limited to laboratory data alone. A series of large validation cohorts will be required to obtain reliable predictive models and accurate therapeutic doses. In the future, clinical translational trials utilizing target ncRNAs are expected to overcome oxaliplatin resistance and thus improve the clinical outcomes of CRC patients.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country/Territory of origin: China

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): 0

Grade C (Good): C, C, C

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Imai Y, Japan; Jeong KY, South Korea; Manojlovic N, Serbia S-Editor: Liu GL L-Editor: Webster JR P-Editor: Liu GL

References
1.  Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71:209-249.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Dekker E, Tanis PJ, Vleugels JLA, Kasi PM, Wallace MB. Colorectal cancer. Lancet. 2019;394:1467-1480.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Biller LH, Schrag D. Diagnosis and Treatment of Metastatic Colorectal Cancer: A Review. JAMA. 2021;325:669-685.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Kanemitsu Y, Shitara K, Mizusawa J, Hamaguchi T, Shida D, Komori K, Ikeda S, Ojima H, Ike H, Shiomi A, Watanabe J, Takii Y, Yamaguchi T, Katsumata K, Ito M, Okuda J, Hyakudomi R, Shimada Y, Katayama H, Fukuda H; JCOG Colorectal Cancer Study Group. Primary Tumor Resection Plus Chemotherapy Versus Chemotherapy Alone for Colorectal Cancer Patients With Asymptomatic, Synchronous Unresectable Metastases (JCOG1007; iPACS): A Randomized Clinical Trial. J Clin Oncol. 2021;39:1098-1107.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Grothey A, Sobrero AF, Shields AF, Yoshino T, Paul J, Taieb J, Souglakos J, Shi Q, Kerr R, Labianca R, Meyerhardt JA, Vernerey D, Yamanaka T, Boukovinas I, Meyers JP, Renfro LA, Niedzwiecki D, Watanabe T, Torri V, Saunders M, Sargent DJ, Andre T, Iveson T. Duration of Adjuvant Chemotherapy for Stage III Colon Cancer. N Engl J Med. 2018;378:1177-1188.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Hecht JR, Lonardi S, Bendell J, Sim HW, Macarulla T, Lopez CD, Van Cutsem E, Muñoz Martin AJ, Park JO, Greil R, Wang H, Hozak RR, Gueorguieva I, Lin Y, Rao S, Ryoo BY. Randomized Phase III Study of FOLFOX Alone or With Pegilodecakin as Second-Line Therapy in Patients With Metastatic Pancreatic Cancer That Progressed After Gemcitabine (SEQUOIA). J Clin Oncol. 2021;39:1108-1118.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Martinez-Balibrea E, Martínez-Cardús A, Ginés A, Ruiz de Porras V, Moutinho C, Layos L, Manzano JL, Bugés C, Bystrup S, Esteller M, Abad A. Tumor-Related Molecular Mechanisms of Oxaliplatin Resistance. Mol Cancer Ther. 2015;14:1767-1776.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Van der Jeught K, Xu HC, Li YJ, Lu XB, Ji G. Drug resistance and new therapies in colorectal cancer. World J Gastroenterol. 2018;24:3834-3848.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Kang L, Tian Y, Xu S, Chen H. Oxaliplatin-induced peripheral neuropathy: clinical features, mechanisms, prevention and treatment. J Neurol. 2021;268:3269-3282.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Mauri G, Gori V, Bonazzina E, Amatu A, Tosi F, Bencardino K, Ruggieri L, Patelli G, Arena S, Bardelli A, Siena S, Sartore-Bianchi A. Oxaliplatin retreatment in metastatic colorectal cancer: Systematic review and future research opportunities. Cancer Treat Rev. 2020;91:102112.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Wei L, Wang X, Lv L, Zheng Y, Zhang N, Yang M. The emerging role of noncoding RNAs in colorectal cancer chemoresistance. Cell Oncol (Dordr). 2019;42:757-768.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Ghanbarian M, Afgar A, Yadegarazari R, Najafi R, Teimoori-Toolabi L. Through oxaliplatin resistance induction in colorectal cancer cells, increasing ABCB1 level accompanies decreasing level of miR-302c-5p, miR-3664-5p and miR-129-5p. Biomed Pharmacother. 2018;108:1070-1080.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Cui J, Li G, Yin J, Li L, Tan Y, Wei H, Liu B, Deng L, Tang J, Chen Y, Yi L. GSTP1 and cancer: Expression, methylation, polymorphisms and signaling (Review). Int J Oncol. 2020;56:867-878.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Sreekumar R, Al-Saihati H, Emaduddin M, Moutasim K, Mellone M, Patel A, Kilic S, Cetin M, Erdemir S, Navio MS, Lopez MA, Curtis N, Yagci T, Primrose JN, Price BD, Berx G, Thomas GJ, Tulchinsky E, Mirnezami A, Sayan AE. The ZEB2-dependent EMT transcriptional programme drives therapy resistance by activating nucleotide excision repair genes ERCC1 and ERCC4 in colorectal cancer. Mol Oncol. 2021;15:2065-2083.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Xiao Y, Lin FT, Lin WC. ACTL6A promotes repair of cisplatin-induced DNA damage, a new mechanism of platinum resistance in cancer. Proc Natl Acad Sci U S A. 2021;118.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Liu J, Miao X, Xiao B, Huang J, Tao X, Zhang J, Zhao H, Pan Y, Wang H, Gao G, Xiao GG. Obg-Like ATPase 1 Enhances Chemoresistance of Breast Cancer via Activation of TGF-β/Smad Axis Cascades. Front Pharmacol. 2020;11:666.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Kwak AW, Park JW, Lee SO, Lee JY, Seo JH, Yoon G, Lee MH, Choi JS, Shim JH. Isolinderalactone sensitizes oxaliplatin-resistance colorectal cancer cells through JNK/p38 MAPK signaling pathways. Phytomedicine. 2022;105:154383.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Liu X, Su K, Sun X, Jiang Y, Wang L, Hu C, Zhang C, Lu M, Du X, Xing B. Sec62 promotes stemness and chemoresistance of human colorectal cancer through activating Wnt/β-catenin pathway. J Exp Clin Cancer Res. 2021;40:132.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Ma J, Zeng S, Zhang Y, Deng G, Qu Y, Guo C, Yin L, Han Y, Cai C, Li Y, Wang G, Bonkovsky HL, Shen H. BMP4 promotes oxaliplatin resistance by an induction of epithelial-mesenchymal transition via MEK1/ERK/ELK1 signaling in hepatocellular carcinoma. Cancer Lett. 2017;411:117-129.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Hu JL, Wang W, Lan XL, Zeng ZC, Liang YS, Yan YR, Song FY, Wang FF, Zhu XH, Liao WJ, Liao WT, Ding YQ, Liang L. CAFs secreted exosomes promote metastasis and chemotherapy resistance by enhancing cell stemness and epithelial-mesenchymal transition in colorectal cancer. Mol Cancer. 2019;18:91.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Yin Y, Yao S, Hu Y, Feng Y, Li M, Bian Z, Zhang J, Qin Y, Qi X, Zhou L, Fei B, Zou J, Hua D, Huang Z. The Immune-microenvironment Confers Chemoresistance of Colorectal Cancer through Macrophage-Derived IL6. Clin Cancer Res. 2017;23:7375-7387.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Rubio MF, Lira MC, Rosa FD, Sambresqui AD, Salazar Güemes MC, Costas MA. RAC3 influences the chemoresistance of colon cancer cells through autophagy and apoptosis inhibition. Cancer Cell Int. 2017;17:111.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Zhang H, Lu B. The Roles of ceRNAs-Mediated Autophagy in Cancer Chemoresistance and Metastasis. Cancers (Basel). 2020;12.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Yang C, Zhang Y, Lin S, Liu Y, Li W. Suppressing the KIF20A/NUAK1/Nrf2/GPX4 signaling pathway induces ferroptosis and enhances the sensitivity of colorectal cancer to oxaliplatin. Aging (Albany NY). 2021;13:13515-13534.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Hou X, Xia J, Feng Y, Cui L, Yang Y, Yang P, Xu X. USP47-Mediated Deubiquitination and Stabilization of TCEA3 Attenuates Pyroptosis and Apoptosis of Colorectal Cancer Cells Induced by Chemotherapeutic Doxorubicin. Front Pharmacol. 2021;12:713322.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Lan H, Liu Y, Liu J, Wang X, Guan Z, Du J, Jin K. Tumor-Associated Macrophages Promote Oxaliplatin Resistance via METTL3-Mediated m6A of TRAF5 and Necroptosis in Colorectal Cancer. Mol Pharm. 2021;18:1026-1037.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Djebali S, Davis CA, Merkel A, Dobin A, Lassmann T, Mortazavi A, Tanzer A, Lagarde J, Lin W, Schlesinger F, Xue C, Marinov GK, Khatun J, Williams BA, Zaleski C, Rozowsky J, Röder M, Kokocinski F, Abdelhamid RF, Alioto T, Antoshechkin I, Baer MT, Bar NS, Batut P, Bell K, Bell I, Chakrabortty S, Chen X, Chrast J, Curado J, Derrien T, Drenkow J, Dumais E, Dumais J, Duttagupta R, Falconnet E, Fastuca M, Fejes-Toth K, Ferreira P, Foissac S, Fullwood MJ, Gao H, Gonzalez D, Gordon A, Gunawardena H, Howald C, Jha S, Johnson R, Kapranov P, King B, Kingswood C, Luo OJ, Park E, Persaud K, Preall JB, Ribeca P, Risk B, Robyr D, Sammeth M, Schaffer L, See LH, Shahab A, Skancke J, Suzuki AM, Takahashi H, Tilgner H, Trout D, Walters N, Wang H, Wrobel J, Yu Y, Ruan X, Hayashizaki Y, Harrow J, Gerstein M, Hubbard T, Reymond A, Antonarakis SE, Hannon G, Giddings MC, Ruan Y, Wold B, Carninci P, Guigó R, Gingeras TR. Landscape of transcription in human cells. Nature. 2012;489:101-108.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature. 2012;489:57-74.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Ali SA, Peffers MJ, Ormseth MJ, Jurisica I, Kapoor M. The non-coding RNA interactome in joint health and disease. Nat Rev Rheumatol. 2021;17:692-705.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Liu CX, Chen LL. Circular RNAs: Characterization, cellular roles, and applications. Cell. 2022;185:2016-2034.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Kaikkonen MU, Lam MT, Glass CK. Non-coding RNAs as regulators of gene expression and epigenetics. Cardiovasc Res. 2011;90:430-440.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Eidem TM, Kugel JF, Goodrich JA. Noncoding RNAs: Regulators of the Mammalian Transcription Machinery. J Mol Biol. 2016;428:2652-2659.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Slack FJ, Chinnaiyan AM. The Role of Non-coding RNAs in Oncology. Cell. 2019;179:1033-1055.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Yan H, Bu P. Non-coding RNA in cancer. Essays Biochem. 2021;65:625-639.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Li XN, Wang ZJ, Ye CX, Zhao BC, Li ZL, Yang Y. RNA sequencing reveals the expression profiles of circRNA and indicates that circDDX17 acts as a tumor suppressor in colorectal cancer. J Exp Clin Cancer Res. 2018;37:325.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Zhu KP, Zhang CL, Ma XL, Hu JP, Cai T, Zhang L. Analyzing the Interactions of mRNAs and ncRNAs to Predict Competing Endogenous RNA Networks in Osteosarcoma Chemo-Resistance. Mol Ther. 2019;27:518-530.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Li Y, Zhao J, Yu S, Wang Z, He X, Su Y, Guo T, Sheng H, Chen J, Zheng Q, Li Y, Guo W, Cai X, Shi G, Wu J, Wang L, Wang P, Huang S. Extracellular Vesicles Long RNA Sequencing Reveals Abundant mRNA, circRNA, and lncRNA in Human Blood as Potential Biomarkers for Cancer Diagnosis. Clin Chem. 2019;65:798-808.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Chen B, Dragomir MP, Yang C, Li Q, Horst D, Calin GA. Targeting non-coding RNAs to overcome cancer therapy resistance. Signal Transduct Target Ther. 2022;7:121.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Overman MJ, Modak J, Kopetz S, Murthy R, Yao JC, Hicks ME, Abbruzzese JL, Tam AL. Use of research biopsies in clinical trials: are risks and benefits adequately discussed? J Clin Oncol. 2013;31:17-22.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Yu W, Hurley J, Roberts D, Chakrabortty SK, Enderle D, Noerholm M, Breakefield XO, Skog JK. Exosome-based liquid biopsies in cancer: opportunities and challenges. Ann Oncol. 2021;32:466-477.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Zhou H, Zhu L, Song J, Wang G, Li P, Li W, Luo P, Sun X, Wu J, Liu Y, Zhu S, Zhang Y. Liquid biopsy at the frontier of detection, prognosis and progression monitoring in colorectal cancer. Mol Cancer. 2022;21:86.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Anfossi S, Babayan A, Pantel K, Calin GA. Clinical utility of circulating non-coding RNAs - an update. Nat Rev Clin Oncol. 2018;15:541-563.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Winkle M, El-Daly SM, Fabbri M, Calin GA. Noncoding RNA therapeutics - challenges and potential solutions. Nat Rev Drug Discov. 2021;20:629-651.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Ha M, Kim VN. Regulation of microRNA biogenesis. Nat Rev Mol Cell Biol. 2014;15:509-524.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Lu TX, Rothenberg ME. MicroRNA. J Allergy Clin Immunol. 2018;141:1202-1207.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Liang Y, Zou Q, Yu W. Steering Against Wind: A New Network of NamiRNAs and Enhancers. Genomics Proteomics Bioinformatics. 2017;15:331-337.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Suzuki HI, Young RA, Sharp PA. Super-Enhancer-Mediated RNA Processing Revealed by Integrative MicroRNA Network Analysis. Cell. 2017;168:1000-1014.e15.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Odame E, Chen Y, Zheng S, Dai D, Kyei B, Zhan S, Cao J, Guo J, Zhong T, Wang L, Li L, Zhang H. Enhancer RNAs: transcriptional regulators and workmates of NamiRNAs in myogenesis. Cell Mol Biol Lett. 2021;26:4.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Wang H, Wang X, Zhang H, Deng T, Liu R, Liu Y, Li H, Bai M, Ning T, Wang J, Ge S, Ba Y. The HSF1/miR-135b-5p axis induces protective autophagy to promote oxaliplatin resistance through the MUL1/ULK1 pathway in colorectal cancer. Oncogene. 2021;40:4695-4708.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Qian XL, Zhou F, Xu S, Jiang J, Chen ZP, Wang SK, Zuo Y, Ni C. MiR-454-3p Promotes Oxaliplatin Resistance by Targeting PTEN in Colorectal Cancer. Front Oncol. 2021;11:638537.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Zhang Y, Liu X, Zhang J, Xu Y, Shao J, Hu Y, Shu P, Cheng H. Inhibition of miR-19a partially reversed the resistance of colorectal cancer to oxaliplatin via PTEN/PI3K/AKT pathway. Aging (Albany NY). 2020;12:5640-5650.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Liang Y, Zhu D, Zhu L, Hou Y, Hou L, Huang X, Li L, Wang Y, Zou H, Wu T, Yao M, Wang J, Meng X. Dichloroacetate Overcomes Oxaliplatin Chemoresistance in Colorectal Cancer through the miR-543/PTEN/Akt/mTOR Pathway. J Cancer. 2019;10:6037-6047.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Liang Y, Zhu D, Hou L, Wang Y, Huang X, Zhou C, Zhu L, Li L, Gu Y, Luo M, Wang J, Meng X. MiR-107 confers chemoresistance to colorectal cancer by targeting calcium-binding protein 39. Br J Cancer. 2020;122:705-714.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Xu K, Chen G, Qiu Y, Yuan Z, Li H, Yuan X, Sun J, Xu J, Liang X, Yin P. miR-503-5p confers drug resistance by targeting PUMA in colorectal carcinoma. Oncotarget. 2017;8:21719-21732.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Zhou Y, He A, Zhang L, Yi G. MiR-744 mediates the Oxaliplatin chemoresistance in colorectal cancer through inhibiting BIN1. Neoplasma. 2020;67:296-303.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Zhuang YY, Zhong W, Xia ZS, Lin SZ, Chan MC, Jiang K, Li WF, Xu XY. miR-5000-3p confers oxaliplatin resistance by targeting ubiquitin-specific peptidase 49 in colorectal cancer. Cell Death Discov. 2021;7:129.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Zhao H, Su W, Kang Q, Xing Z, Lin X, Wu Z. Natural killer cells inhibit oxaliplatin-resistant colorectal cancer by repressing WBSCR22 via upregulating microRNA-146b-5p. Am J Cancer Res. 2018;8:824-834.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Jiang T, Ye L, Han Z, Liu Y, Yang Y, Peng Z, Fan J. miR-19b-3p promotes colon cancer proliferation and oxaliplatin-based chemoresistance by targeting SMAD4: validation by bioinformatics and experimental analyses. J Exp Clin Cancer Res. 2017;36:131.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Lin W, Zou H, Mo J, Jin C, Jiang H, Yu C, Jiang Z, Yang Y, He B, Wang K. Micro1278 Leads to Tumor Growth Arrest, Enhanced Sensitivity to Oxaliplatin and Vitamin D and Inhibits Metastasis via KIF5B, CYP24A1, and BTG2, Respectively. Front Oncol. 2021;11:637878.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Zhou H, Lin C, Zhang Y, Zhang X, Zhang C, Zhang P, Xie X, Ren Z. miR-506 enhances the sensitivity of human colorectal cancer cells to oxaliplatin by suppressing MDR1/P-gp expression. Cell Prolif. 2017;50.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Wu YZ, Lin HY, Zhang Y, Chen WF. miR-200b-3p mitigates oxaliplatin resistance via targeting TUBB3 in colorectal cancer. J Gene Med. 2020;22:e3178.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Hua Y, Zhu Y, Zhang J, Zhu Z, Ning Z, Chen H, Liu L, Chen Z, Meng Z. miR-122 Targets X-Linked Inhibitor of Apoptosis Protein to Sensitize Oxaliplatin-Resistant Colorectal Cancer Cells to Oxaliplatin-Mediated Cytotoxicity. Cell Physiol Biochem. 2018;51:2148-2159.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Azar MRMH, Aghazadeh H, Mohammed HN, Sara MRS, Hosseini A, Shomali N, Tamjidifar R, Tarzi S, Mansouri M, Sarand SP, Marofi F, Akbari M, Xu H, Shotorbani SS. miR-193a-5p as a promising therapeutic candidate in colorectal cancer by reducing 5-FU and Oxaliplatin chemoresistance by targeting CXCR4. Int Immunopharmacol. 2021;92:107355.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Liang H, Xu Y, Zhang Q, Yang Y, Mou Y, Gao Y, Chen R, Chen C, Dai P. MiR-483-3p regulates oxaliplatin resistance by targeting FAM171B in human colorectal cancer cells. Artif Cells Nanomed Biotechnol. 2019;47:725-736.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Zhang L, Chen H, Song Y, Gu Q, Zhang L, Xie Q, Xu J, Zhang M. MiR-325 Promotes Oxaliplatin-Induced Cytotoxicity Against Colorectal Cancer Through the HSPA12B/PI3K/AKT/Bcl-2 Pathway. Dig Dis Sci. 2021;66:2651-2660.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Poel D, Boyd LNC, Beekhof R, Schelfhorst T, Pham TV, Piersma SR, Knol JC, Jimenez CR, Verheul HMW, Buffart TE. Proteomic Analysis of miR-195 and miR-497 Replacement Reveals Potential Candidates that Increase Sensitivity to Oxaliplatin in MSI/P53wt Colorectal Cancer Cells. Cells. 2019;8.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Chen L, He M, Zhang M, Sun Q, Zeng S, Zhao H, Yang H, Liu M, Ren S, Meng X, Xu H. The Role of non-coding RNAs in colorectal cancer, with a focus on its autophagy. Pharmacol Ther. 2021;226:107868.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Sun C, Wang FJ, Zhang HG, Xu XZ, Jia RC, Yao L, Qiao PF. miR-34a mediates oxaliplatin resistance of colorectal cancer cells by inhibiting macroautophagy via transforming growth factor-β/Smad4 pathway. World J Gastroenterol. 2017;23:1816-1827.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Sun W, Li J, Zhou L, Han J, Liu R, Zhang H, Ning T, Gao Z, Liu B, Chen X, Ba Y. The c-Myc/miR-27b-3p/ATG10 regulatory axis regulates chemoresistance in colorectal cancer. Theranostics. 2020;10:1981-1996.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Liu XY, Zhang FR, Shang JY, Liu YY, Lv XF, Yuan JN, Zhang TT, Li K, Lin XC, Liu X, Lei Q, Fu XD, Zhou JG, Liang SJ. Renal inhibition of miR-181a ameliorates 5-fluorouracil-induced mesangial cell apoptosis and nephrotoxicity. Cell Death Dis. 2018;9:610.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Van Houdt WJ, Emmink BL, Pham TV, Piersma SR, Verheem A, Vries RG, Fratantoni SA, Pronk A, Clevers H, Borel Rinkes IH, Jimenez CR, Kranenburg O. Comparative proteomics of colon cancer stem cells and differentiated tumor cells identifies BIRC6 as a potential therapeutic target. Mol Cell Proteomics. 2011;10:M111.011353.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Chen LL. Linking Long Noncoding RNA Localization and Function. Trends Biochem Sci. 2016;41:761-772.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Connerty P, Lock RB, de Bock CE. Long Non-coding RNAs: Major Regulators of Cell Stress in Cancer. Front Oncol. 2020;10:285.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Gao N, Li Y, Li J, Gao Z, Yang Z, Liu H, Fan T. Long Non-Coding RNAs: The Regulatory Mechanisms, Research Strategies, and Future Directions in Cancers. Front Oncol. 2020;10:598817.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Rai MI, Alam M, Lightfoot DA, Gurha P, Afzal AJ. Classification and experimental identification of plant long non-coding RNAs. Genomics. 2019;111:997-1005.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Sun Q, Hao Q, Prasanth KV. Nuclear Long Noncoding RNAs: Key Regulators of Gene Expression. Trends Genet. 2018;34:142-157.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Postepska-Igielska A, Giwojna A, Gasri-Plotnitsky L, Schmitt N, Dold A, Ginsberg D, Grummt I. LncRNA Khps1 Regulates Expression of the Proto-oncogene SPHK1 via Triplex-Mediated Changes in Chromatin Structure. Mol Cell. 2015;60:626-636.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Xu M, Chen X, Lin K, Zeng K, Liu X, Pan B, Xu X, Xu T, Hu X, Sun L, He B, Pan Y, Sun H, Wang S. The long noncoding RNA SNHG1 regulates colorectal cancer cell growth through interactions with EZH2 and miR-154-5p. Mol Cancer. 2018;17:141.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Lan Z, Yao X, Sun K, Li A, Liu S, Wang X. The Interaction Between lncRNA SNHG6 and hnRNPA1 Contributes to the Growth of Colorectal Cancer by Enhancing Aerobic Glycolysis Through the Regulation of Alternative Splicing of PKM. Front Oncol. 2020;10:363.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Chen QN, Wei CC, Wang ZX, Sun M. Long non-coding RNAs in anti-cancer drug resistance. Oncotarget. 2017;8:1925-1936.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Hu Y, Zhu QN, Deng JL, Li ZX, Wang G, Zhu YS. Emerging role of long non-coding RNAs in cisplatin resistance. Onco Targets Ther. 2018;11:3185-3194.  [PubMed]  [DOI]  [Cited in This Article: ]
82.  Jiang X, Li Q, Zhang S, Song C, Zheng P. Long noncoding RNA GIHCG induces cancer progression and chemoresistance and indicates poor prognosis in colorectal cancer. Onco Targets Ther. 2019;12:1059-1070.  [PubMed]  [DOI]  [Cited in This Article: ]
83.  Li T, Jin X, Dong J, Deng H. Long noncoding RNA ARSR is associated with a poor prognosis in patients with colorectal cancer. J Gene Med. 2020;22:e3241.  [PubMed]  [DOI]  [Cited in This Article: ]
84.  Yamamura S, Imai-Sumida M, Tanaka Y, Dahiya R. Interaction and cross-talk between non-coding RNAs. Cell Mol Life Sci. 2018;75:467-484.  [PubMed]  [DOI]  [Cited in This Article: ]
85.  Weng X, Liu H, Ruan J, Du M, Wang L, Mao J, Cai Y, Lu X, Chen W, Huang Y, Zhi X, Shan J. HOTAIR/miR-1277-5p/ZEB1 axis mediates hypoxia-induced oxaliplatin resistance via regulating epithelial-mesenchymal transition in colorectal cancer. Cell Death Discov. 2022;8:310.  [PubMed]  [DOI]  [Cited in This Article: ]
86.  Li P, Zhang X, Wang H, Wang L, Liu T, Du L, Yang Y, Wang C. MALAT1 Is Associated with Poor Response to Oxaliplatin-Based Chemotherapy in Colorectal Cancer Patients and Promotes Chemoresistance through EZH2. Mol Cancer Ther. 2017;16:739-751.  [PubMed]  [DOI]  [Cited in This Article: ]
87.  Thomson DW, Dinger ME. Endogenous microRNA sponges: evidence and controversy. Nat Rev Genet. 2016;17:272-283.  [PubMed]  [DOI]  [Cited in This Article: ]
88.  Fan C, Yuan Q, Liu G, Zhang Y, Yan M, Sun Q, Zhu C. Long non-coding RNA MALAT1 regulates oxaliplatin-resistance via miR-324-3p/ADAM17 axis in colorectal cancer cells. Cancer Cell Int. 2020;20:473.  [PubMed]  [DOI]  [Cited in This Article: ]
89.  Liang J, Tian XF, Yang W. Effects of long non-coding RNA Opa-interacting protein 5 antisense RNA 1 on colon cancer cell resistance to oxaliplatin and its regulation of microRNA-137. World J Gastroenterol. 2020;26:1474-1489.  [PubMed]  [DOI]  [Cited in This Article: ]
90.  Yue B, Cai D, Liu C, Fang C, Yan D. Linc00152 Functions as a Competing Endogenous RNA to Confer Oxaliplatin Resistance and Holds Prognostic Values in Colon Cancer. Mol Ther. 2016;24:2064-2077.  [PubMed]  [DOI]  [Cited in This Article: ]
91.  Zhou L, Li J, Liao M, Zhang Q, Yang M. LncRNA MIR155HG induces M2 macrophage polarization and drug resistance of colorectal cancer cells by regulating ANXA2. Cancer Immunol Immunother. 2022;71:1075-1091.  [PubMed]  [DOI]  [Cited in This Article: ]
92.  Gao R, Fang C, Xu J, Tan H, Li P, Ma L. LncRNA CACS15 contributes to oxaliplatin resistance in colorectal cancer by positively regulating ABCC1 through sponging miR-145. Arch Biochem Biophys. 2019;663:183-191.  [PubMed]  [DOI]  [Cited in This Article: ]
93.  Xie L, Cui G, Li T. Long Noncoding RNA CBR3-AS1 Promotes Stem-like Properties and Oxaliplatin Resistance of Colorectal Cancer by Sponging miR-145-5p. J Oncol. 2022;2022:2260211.  [PubMed]  [DOI]  [Cited in This Article: ]
94.  Gao H, Song X, Kang T, Yan B, Feng L, Gao L, Ai L, Liu X, Yu J, Li H. Long noncoding RNA CRNDE functions as a competing endogenous RNA to promote metastasis and oxaliplatin resistance by sponging miR-136 in colorectal cancer. Onco Targets Ther. 2017;10:205-216.  [PubMed]  [DOI]  [Cited in This Article: ]
95.  Meng X, Sun W, Yu J, Zhou Y, Gu Y, Han J, Zhou L, Jiang X, Wang C. LINC00460-miR-149-5p/miR-150-5p-Mutant p53 Feedback Loop Promotes Oxaliplatin Resistance in Colorectal Cancer. Mol Ther Nucleic Acids. 2020;22:1004-1015.  [PubMed]  [DOI]  [Cited in This Article: ]
96.  Li Y, Li C, Li D, Yang L, Jin J, Zhang B. lncRNA KCNQ1OT1 enhances the chemoresistance of oxaliplatin in colon cancer by targeting the miR-34a/ATG4B pathway. Onco Targets Ther. 2019;12:2649-2660.  [PubMed]  [DOI]  [Cited in This Article: ]
97.  Li Q, Sun H, Luo D, Gan L, Mo S, Dai W, Liang L, Yang Y, Xu M, Li J, Zheng P, Li X, Li Y, Wang Z. Lnc-RP11-536 K7.3/SOX2/HIF-1α signaling axis regulates oxaliplatin resistance in patient-derived colorectal cancer organoids. J Exp Clin Cancer Res. 2021;40:348.  [PubMed]  [DOI]  [Cited in This Article: ]
98.  Sun J, Zhou H, Bao X, Wu Y, Jia H, Zhao H, Liu G. lncRNA TUG1 Facilitates Colorectal Cancer Stem Cell Characteristics and Chemoresistance by Enhancing GATA6 Protein Stability. Stem Cells Int. 2021;2021:1075481.  [PubMed]  [DOI]  [Cited in This Article: ]
99.  Zhou Q, Hou Z, Zuo S, Zhou X, Feng Y, Sun Y, Yuan X. LUCAT1 promotes colorectal cancer tumorigenesis by targeting the ribosomal protein L40-MDM2-p53 pathway through binding with UBA52. Cancer Sci. 2019;110:1194-1207.  [PubMed]  [DOI]  [Cited in This Article: ]
100.  Deng X, Kong F, Li S, Jiang H, Dong L, Xu X, Zhang X, Yuan H, Xu Y, Chu Y, Peng H, Guan M. A KLF4/PiHL/EZH2/HMGA2 regulatory axis and its function in promoting oxaliplatin-resistance of colorectal cancer. Cell Death Dis. 2021;12:485.  [PubMed]  [DOI]  [Cited in This Article: ]
101.  Quinodoz S, Guttman M. Long noncoding RNAs: an emerging link between gene regulation and nuclear organization. Trends Cell Biol. 2014;24:651-663.  [PubMed]  [DOI]  [Cited in This Article: ]
102.  Li Y, Gan Y, Liu J, Li J, Zhou Z, Tian R, Sun R, Xiao Q, Li Y, Lu P, Peng Y, Shu G, Yin G. Downregulation of MEIS1 mediated by ELFN1-AS1/EZH2/DNMT3a axis promotes tumorigenesis and oxaliplatin resistance in colorectal cancer. Signal Transduct Target Ther. 2022;7:87.  [PubMed]  [DOI]  [Cited in This Article: ]
103.  Damas ND, Marcatti M, Côme C, Christensen LL, Nielsen MM, Baumgartner R, Gylling HM, Maglieri G, Rundsten CF, Seemann SE, Rapin N, Thézenas S, Vang S, Ørntoft T, Andersen CL, Pedersen JS, Lund AH. SNHG5 promotes colorectal cancer cell survival by counteracting STAU1-mediated mRNA destabilization. Nat Commun. 2016;7:13875.  [PubMed]  [DOI]  [Cited in This Article: ]
104.  Chen B, Dragomir MP, Fabris L, Bayraktar R, Knutsen E, Liu X, Tang C, Li Y, Shimura T, Ivkovic TC, De Los Santos MC, Anfossi S, Shimizu M, Shah MY, Ling H, Shen P, Multani AS, Pardini B, Burks JK, Katayama H, Reineke LC, Huo L, Syed M, Song S, Ferracin M, Oki E, Fromm B, Ivan C, Bhuvaneshwar K, Gusev Y, Mimori K, Menter D, Sen S, Matsuyama T, Uetake H, Vasilescu C, Kopetz S, Parker-Thornburg J, Taguchi A, Hanash SM, Girnita L, Slaby O, Goel A, Varani G, Gagea M, Li C, Ajani JA, Calin GA. The Long Noncoding RNA CCAT2 Induces Chromosomal Instability Through BOP1-AURKB Signaling. Gastroenterology. 2020;159:2146-2162.e33.  [PubMed]  [DOI]  [Cited in This Article: ]
105.  Li C, Li X. Antitumor Activity of lncRNA NBAT-1 via Inhibition of miR-4504 to Target to WWC3 in Oxaliplatin-Resistant Colorectal Carcinoma. J Healthc Eng. 2022;2022:9121554.  [PubMed]  [DOI]  [Cited in This Article: ]
106.  Li L, Shang J, Zhang Y, Liu S, Peng Y, Zhou Z, Pan H, Wang X, Chen L, Zhao Q. MEG3 is a prognostic factor for CRC and promotes chemosensitivity by enhancing oxaliplatin-induced cell apoptosis. Oncol Rep. 2017;38:1383-1392.  [PubMed]  [DOI]  [Cited in This Article: ]
107.  Wang H, Li H, Zhang L, Yang D. Overexpression of MEG3 sensitizes colorectal cancer cells to oxaliplatin through regulation of miR-141/PDCD4 axis. Biomed Pharmacother. 2018;106:1607-1615.  [PubMed]  [DOI]  [Cited in This Article: ]
108.  Wang X, Zhang H, Yin S, Yang Y, Yang H, Yang J, Zhou Z, Li S, Ying G, Ba Y. lncRNA-encoded pep-AP attenuates the pentose phosphate pathway and sensitizes colorectal cancer cells to Oxaliplatin. EMBO Rep. 2022;23:e53140.  [PubMed]  [DOI]  [Cited in This Article: ]
109.  Wu S, Yang X, Tang W, Familiari G, Relucenti M, Aschner M, Li X, Chen R. Chemotherapeutic Risk lncRNA-PVT1 SNP Sensitizes Metastatic Colorectal Cancer to FOLFOX Regimen. Front Oncol. 2022;12:808889.  [PubMed]  [DOI]  [Cited in This Article: ]
110.  Sun F, Liang W, Qian J. The identification of CRNDE, H19, UCA1 and HOTAIR as the key lncRNAs involved in oxaliplatin or irinotecan resistance in the chemotherapy of colorectal cancer based on integrative bioinformatics analysis. Mol Med Rep. 2019;20:3583-3596.  [PubMed]  [DOI]  [Cited in This Article: ]
111.  Zhang F, Wang H, Yu J, Yao X, Yang S, Li W, Xu L, Zhao L. LncRNA CRNDE attenuates chemoresistance in gastric cancer via SRSF6-regulated alternative splicing of PICALM. Mol Cancer. 2021;20:6.  [PubMed]  [DOI]  [Cited in This Article: ]
112.  Liu J, Liu T, Wang X, He A. Circles reshaping the RNA world: from waste to treasure. Mol Cancer. 2017;16:58.  [PubMed]  [DOI]  [Cited in This Article: ]
113.  Huang A, Zheng H, Wu Z, Chen M, Huang Y. Circular RNA-protein interactions: functions, mechanisms, and identification. Theranostics. 2020;10:3503-3517.  [PubMed]  [DOI]  [Cited in This Article: ]
114.  Wang J, Zhang Y, Liu L, Yang T, Song J. Circular RNAs: new biomarkers of chemoresistance in cancer. Cancer Biol Med. 2021;.  [PubMed]  [DOI]  [Cited in This Article: ]
115.  Mao G, Zhou B, Xu W, Jiao N, Wu Z, Li J, Liu Y. Hsa_circ_0040809 regulates colorectal cancer development by upregulating methyltransferase DNMT1 via targeting miR-515-5p. J Gene Med. 2021;23:e3388.  [PubMed]  [DOI]  [Cited in This Article: ]
116.  Li Z, Yao H, Wang S, Li G, Gu X. CircTADA2A suppresses the progression of colorectal cancer via miR-374a-3p/KLF14 axis. J Exp Clin Cancer Res. 2020;39:160.  [PubMed]  [DOI]  [Cited in This Article: ]
117.  Zhou J, Wang L, Sun Q, Chen R, Zhang C, Yang P, Tan Y, Peng C, Wang T, Jin C, Ji J, Jin K, Sun Y. Hsa_circ_0001666 suppresses the progression of colorectal cancer through the miR-576-5p/PCDH10 axis. Clin Transl Med. 2021;11:e565.  [PubMed]  [DOI]  [Cited in This Article: ]
118.  Zhang N, Zhang X, Xu W, Mu Z. CircRNA_103948 inhibits autophagy in colorectal cancer in a ceRNA manner. Ann N Y Acad Sci. 2021;1503:88-101.  [PubMed]  [DOI]  [Cited in This Article: ]
119.  Feng J, Li Z, Li L, Xie H, Lu Q, He X. Hypoxiainduced circCCDC66 promotes the tumorigenesis of colorectal cancer via the miR3140/autophagy pathway. Int J Mol Med. 2020;46:1973-1982.  [PubMed]  [DOI]  [Cited in This Article: ]
120.  Chen Z, Ren R, Wan D, Wang Y, Xue X, Jiang M, Shen J, Han Y, Liu F, Shi J, Kuang Y, Li W, Zhi Q. Hsa_circ_101555 functions as a competing endogenous RNA of miR-597-5p to promote colorectal cancer progression. Oncogene. 2019;38:6017-6034.  [PubMed]  [DOI]  [Cited in This Article: ]
121.  Hansen TB, Jensen TI, Clausen BH, Bramsen JB, Finsen B, Damgaard CK, Kjems J. Natural RNA circles function as efficient microRNA sponges. Nature. 2013;495:384-388.  [PubMed]  [DOI]  [Cited in This Article: ]
122.  Jiang Z, Hou Z, Liu W, Yu Z, Liang Z, Chen S. Circular RNA protein tyrosine kinase 2 (circPTK2) promotes colorectal cancer proliferation, migration, invasion and chemoresistance. Bioengineered. 2022;13:810-823.  [PubMed]  [DOI]  [Cited in This Article: ]
123.  Li S, Zheng S. Down-Regulation of Circ_0032833 Sensitizes Colorectal Cancer to 5-Fluorouracil and Oxaliplatin Partly Depending on the Regulation of miR-125-5p and MSI1. Cancer Manag Res. 2020;12:11257-11269.  [PubMed]  [DOI]  [Cited in This Article: ]
124.  Lai M, Liu G, Li R, Bai H, Zhao J, Xiao P, Mei J. Hsa_circ_0079662 induces the resistance mechanism of the chemotherapy drug oxaliplatin through the TNF-α pathway in human colon cancer. J Cell Mol Med. 2020;24:5021-5027.  [PubMed]  [DOI]  [Cited in This Article: ]
125.  Zhang Y, Li C, Liu X, Wang Y, Zhao R, Yang Y, Zheng X, Zhang Y, Zhang X. circHIPK3 promotes oxaliplatin-resistance in colorectal cancer through autophagy by sponging miR-637. EBioMedicine. 2019;48:277-288.  [PubMed]  [DOI]  [Cited in This Article: ]
126.  Lin YC, Yu YS, Lin HH, Hsiao KY. Oxaliplatin-Induced DHX9 Phosphorylation Promotes Oncogenic Circular RNA CCDC66 Expression and Development of Chemoresistance. Cancers (Basel). 2020;12.  [PubMed]  [DOI]  [Cited in This Article: ]
127.  Abu N, Hon KW, Jeyaraman S, Yahaya A, Abdullah NM, Mustangin M, Sulaiman SA, Jamal R, Ab-Mutalib NS. Identification of differentially expressed circular RNAs in chemoresistant colorectal cancer. Epigenomics. 2019;11:875-884.  [PubMed]  [DOI]  [Cited in This Article: ]
128.  Hon KW, Ab-Mutalib NS, Abdullah NMA, Jamal R, Abu N. Extracellular Vesicle-derived circular RNAs confers chemoresistance in Colorectal cancer. Sci Rep. 2019;9:16497.  [PubMed]  [DOI]  [Cited in This Article: ]
129.  Jin G, Liu Y, Zhang J, Bian Z, Yao S, Fei B, Zhou L, Yin Y, Huang Z. A panel of serum exosomal microRNAs as predictive markers for chemoresistance in advanced colorectal cancer. Cancer Chemother Pharmacol. 2019;84:315-325.  [PubMed]  [DOI]  [Cited in This Article: ]
130.  Jiang Z, Hou Z, Li L, Liu W, Yu Z, Chen S. Exosomal circEPB41L2 serves as a sponge for miR-21-5p and miR-942-5p to suppress colorectal cancer progression by regulating the PTEN/AKT signalling pathway. Eur J Clin Invest. 2021;51:e13581.  [PubMed]  [DOI]  [Cited in This Article: ]
131.  Ding B, Yao M, Fan W, Lou W. Whole-transcriptome analysis reveals a potential hsa_circ_0001955/hsa_circ_0000977-mediated miRNA-mRNA regulatory sub-network in colorectal cancer. Aging (Albany NY). 2020;12:5259-5279.  [PubMed]  [DOI]  [Cited in This Article: ]
132.  Van Niel G, D'Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol. 2018;19:213-228.  [PubMed]  [DOI]  [Cited in This Article: ]
133.  Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol. 2013;200:373-383.  [PubMed]  [DOI]  [Cited in This Article: ]
134.  Kalluri R. The biology and function of exosomes in cancer. J Clin Invest. 2016;126:1208-1215.  [PubMed]  [DOI]  [Cited in This Article: ]
135.  Zhu L, Sun HT, Wang S, Huang SL, Zheng Y, Wang CQ, Hu BY, Qin W, Zou TT, Fu Y, Shen XT, Zhu WW, Geng Y, Lu L, Jia HL, Qin LX, Dong QZ. Isolation and characterization of exosomes for cancer research. J Hematol Oncol. 2020;13:152.  [PubMed]  [DOI]  [Cited in This Article: ]
136.  Yu D, Li Y, Wang M, Gu J, Xu W, Cai H, Fang X, Zhang X. Exosomes as a new frontier of cancer liquid biopsy. Mol Cancer. 2022;21:56.  [PubMed]  [DOI]  [Cited in This Article: ]
137.  Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science. 2020;367.  [PubMed]  [DOI]  [Cited in This Article: ]
138.  Yokoi A, Ochiya T. Exosomes and extracellular vesicles: Rethinking the essential values in cancer biology. Semin Cancer Biol. 2021;74:79-91.  [PubMed]  [DOI]  [Cited in This Article: ]
139.  He C, Zheng S, Luo Y, Wang B. Exosome Theranostics: Biology and Translational Medicine. Theranostics. 2018;8:237-255.  [PubMed]  [DOI]  [Cited in This Article: ]
140.  Baassiri A, Nassar F, Mukherji D, Shamseddine A, Nasr R, Temraz S. Exosomal Non Coding RNA in LIQUID Biopsies as a Promising Biomarker for Colorectal Cancer. Int J Mol Sci. 2020;21.  [PubMed]  [DOI]  [Cited in This Article: ]
141.  Ren J, Ding L, Zhang D, Shi G, Xu Q, Shen S, Wang Y, Wang T, Hou Y. Carcinoma-associated fibroblasts promote the stemness and chemoresistance of colorectal cancer by transferring exosomal lncRNA H19. Theranostics. 2018;8:3932-3948.  [PubMed]  [DOI]  [Cited in This Article: ]
142.  Deng X, Ruan H, Zhang X, Xu X, Zhu Y, Peng H, Kong F, Guan M. Long noncoding RNA CCAL transferred from fibroblasts by exosomes promotes chemoresistance of colorectal cancer cells. Int J Cancer. 2020;146:1700-1716.  [PubMed]  [DOI]  [Cited in This Article: ]
143.  Qu Z, Yang KD, Luo BH, Zhang F. CAFs-secreted exosomal cricN4BP2L2 promoted colorectal cancer stemness and chemoresistance by interacting with EIF4A3. Exp Cell Res. 2022;418:113266.  [PubMed]  [DOI]  [Cited in This Article: ]
144.  Li C, Li X. Exosome-Derived Circ_0094343 Promotes Chemosensitivity of Colorectal Cancer Cells by Regulating Glycolysis via the miR-766-5p/TRIM67 Axis. Contrast Media Mol Imaging. 2022;2022:2878557.  [PubMed]  [DOI]  [Cited in This Article: ]
145.  Ning T, Li J, He Y, Zhang H, Wang X, Deng T, Liu R, Li H, Bai M, Fan Q, Zhu K, Ying G, Ba Y. Exosomal miR-208b related with oxaliplatin resistance promotes Treg expansion in colorectal cancer. Mol Ther. 2021;29:2723-2736.  [PubMed]  [DOI]  [Cited in This Article: ]
146.  Xu Y, Zhu M. Novel exosomal miR-46146 transfer oxaliplatin chemoresistance in colorectal cancer. Clin Transl Oncol. 2020;22:1105-1116.  [PubMed]  [DOI]  [Cited in This Article: ]
147.  Xiao Z, Liu Y, Li Q, Liu Q, Luo Y, Wei S. EVs delivery of miR-1915-3p improves the chemotherapeutic efficacy of oxaliplatin in colorectal cancer. Cancer Chemother Pharmacol. 2021;88:1021-1031.  [PubMed]  [DOI]  [Cited in This Article: ]
148.  Wang X, Zhang H, Yang H, Bai M, Ning T, Deng T, Liu R, Fan Q, Zhu K, Li J, Zhan Y, Ying G, Ba Y. Exosome-delivered circRNA promotes glycolysis to induce chemoresistance through the miR-122-PKM2 axis in colorectal cancer. Mol Oncol. 2020;14:539-555.  [PubMed]  [DOI]  [Cited in This Article: ]
149.  Ohzawa H, Kimura Y, Saito A, Yamaguchi H, Miyato H, Sakuma Y, Horie H, Hosoya Y, Lefor AK, Sata N, Kitayama J. Ratios of miRNAs in Peritoneal Exosomes are Useful Biomarkers to Predict Tumor Response to Intraperitoneal Chemotherapy in Patients with Peritoneal Metastases from Gastric Cancer. Ann Surg Oncol. 2020;27:5057-5064.  [PubMed]  [DOI]  [Cited in This Article: ]
150.  Yagi T, Iinuma H, Hayama T, Matsuda K, Nozawa K, Tsukamoto M, Shimada R, Akahane T, Tsuchiya T, Ozawa T, Hashiguchi Y. Plasma exosomal microRNA-125b as a monitoring biomarker of resistance to mFOLFOX6-based chemotherapy in advanced and recurrent colorectal cancer patients. Mol Clin Oncol. 2019;11:416-424.  [PubMed]  [DOI]  [Cited in This Article: ]
151.  Sur D, Balacescu L, Cainap SS, Visan S, Pop L, Burz C, Havasi A, Buiga R, Cainap C, Irimie A, Balacescu O. Predictive Efficacy of MiR-125b-5p, MiR-17-5p, and MiR-185-5p in Liver Metastasis and Chemotherapy Response Among Advanced Stage Colorectal Cancer Patients. Front Oncol. 2021;11:651380.  [PubMed]  [DOI]  [Cited in This Article: ]
152.  Kim H, Kim EH, Kwak G, Chi SG, Kim SH, Yang Y. Exosomes: Cell-Derived Nanoplatforms for the Delivery of Cancer Therapeutics. Int J Mol Sci. 2020;22.  [PubMed]  [DOI]  [Cited in This Article: ]
153.  Zhang Y, Liu Q, Zhang X, Huang H, Tang S, Chai Y, Xu Z, Li M, Chen X, Liu J, Yang C. Recent advances in exosome-mediated nucleic acid delivery for cancer therapy. J Nanobiotechnology. 2022;20:279.  [PubMed]  [DOI]  [Cited in This Article: ]
154.  Hui B, Lu C, Wang J, Xu Y, Yang Y, Ji H, Li X, Xu L, Tang W, Wang K, Gu Y. Engineered exosomes for co-delivery of PGM5-AS1 and oxaliplatin to reverse drug resistance in colon cancer. J Cell Physiol. 2022;237:911-933.  [PubMed]  [DOI]  [Cited in This Article: ]
155.  Pi F, Binzel DW, Lee TJ, Li Z, Sun M, Rychahou P, Li H, Haque F, Wang S, Croce CM, Guo B, Evers BM, Guo P. Nanoparticle orientation to control RNA loading and ligand display on extracellular vesicles for cancer regression. Nat Nanotechnol. 2018;13:82-89.  [PubMed]  [DOI]  [Cited in This Article: ]
156.  Liang G, Zhu Y, Ali DJ, Tian T, Xu H, Si K, Sun B, Chen B, Xiao Z. Engineered exosomes for targeted co-delivery of miR-21 inhibitor and chemotherapeutics to reverse drug resistance in colon cancer. J Nanobiotechnology. 2020;18:10.  [PubMed]  [DOI]  [Cited in This Article: ]