Hostname: page-component-848d4c4894-pftt2 Total loading time: 0 Render date: 2024-04-30T14:28:22.339Z Has data issue: false hasContentIssue false

MicroRNAs in cancer metastasis: biological and therapeutic implications

Published online by Cambridge University Press:  17 March 2023

Marie C. Sell
Affiliation:
School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton, QLD 4701, Australia
Charmaine A. Ramlogan-Steel
Affiliation:
School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton, QLD 4701, Australia
Jason C. Steel
Affiliation:
School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton, QLD 4701, Australia
Bijay P. Dhungel*
Affiliation:
Gene & Stem Cell Therapy Program Centenary Institute, The University of Sydney, Camperdown, NSW 2050, Australia Faculty of Medicine & Health, The University of Sydney, Camperdown, NSW 2050, Australia
*
Author for correspondence: Bijay P. Dhungel, E-mail: b.dhungel@centenary.org.au; bijay.dhungel@sydney.edu.au
Rights & Permissions [Opens in a new window]

Abstract

Cancer metastasis is the primary cause of cancer-related deaths. The seeding of primary tumours at a secondary site is a highly inefficient process requiring substantial alterations in the genetic architecture of cancer cells. These alterations include significant changes in global gene expression patterns. MicroRNAs are small, non-protein coding RNAs which play a central role in regulating gene expression. Here, we focus on microRNA determinants of cancer metastasis and examine microRNA dysregulation in metastatic cancer cells. We dissect the metastatic process in a step-wise manner and summarise the involvement of microRNAs at each step. We also discuss the advantages and limitations of different microRNA-based strategies that have been used to target metastasis in pre-clinical models. Finally, we highlight current clinical trials that use microRNA-based therapies to target advanced or metastatic tumours.

Type
Review
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
Copyright © The Author(s), 2023. Published by Cambridge University Press

Introduction

In 1993, a revolutionary discovery of the first microRNA (miRNA) while studying nematode Caenorhabditis elegans was made by Ambros et al. (Ref. Reference Rawat1). This finding revealed an essential part of the non-coding genome that plays a key role in post-transcriptional gene regulation (Ref. Reference Rawat1). Fast track 29 years, now over 2600 mature human miRNA sequences have been identified (Ref. Reference Kozomara, Birgaoanu and Griffiths-Jones2). MicroRNAs are small, endogenous, single-stranded, non-protein coding RNA molecules of 19 to 24 nucleotides. MiRNAs account for approximately 3% of the human genome and are evolutionary conserved across mammals (Refs Reference Mishra, Yadav and Rani3, Reference To4). The canonical miRNA biogenesis pathway (Fig. 1) starts with RNA polymerase II-mediated transcription of the primary miRNA gene (pri-miRNA). The pri-miRNA is characterised by a hairpin structure with a 5′ cap and polyadenylation site at the 3′ (Ref. Reference Bautista-Sánchez5). Drosha, an RNase III protein, then cleaves the pri-miRNA, releasing a precursor loop (pre-miRNA) of approximately 70–100 nucleotides (Ref. Reference Iorio and Croce6). Upon release, the pre-miRNA is exported to the cytoplasm via Exportin 5 (Ref. Reference Iqbal7). In the cytoplasm, pre-miRNA is cleaved by an RNase III called Dicer, to produce a double-stranded mature miRNA of approximately 22 nucleotides in length (Ref. Reference Acunzo8). The non-canonical pathways utilise different combination of proteins during the biogenesis steps. These pathways can be further classified as Drosha- and Dicer- independent pathways. For instance, miRNAs produced from introns of messenger RNAs or mirtrons can bypass Drosha-mediated processing (Ref. Reference Ruby, Jan and Bartel9). 7-methylguanosine (m7G)-capped pre-miRNA have been identified which are nascent RNAs directly exported to the cytoplasm through exportin 1 without the need for Drosha (e.g. pre-miR-320) (Ref. Reference Xie10). Another example is the biogenesis of miR-451 which is independent of Dicer processing but requires Drosha and Argonuate 2 (AGO2) protein (Ref. Reference Cheloufi11).

Fig. 1. Biogenesis and mechanism of action of miRNAs: RNA polymerase II-mediated transcription forms the primary miRNA (pri-miRNA) which is cleaved by an RNase III enzyme (DROSHA) to produce a precursor miRNA (pre-miRNA) in the canonical pathway of miRNA biogenesis. The pre-miRNA is exported to the cytoplasm via exportin 5, for further processing by RNase III DICER to form a mature miRNA duplex. Non-canonical pathways are independent of Drosha or Dicer processing. The miRNA duplex is then unwound whereby the guide strand along with Argonaute (AGO) proteins form a miRNA-induced silencing complex (RISC). The RISC complex binds to target sequences of mRNA leading to translation repression or degradation. AGO recruits GW182 which forms a complex with CCR4-NOT making the target mRNA susceptible to cleavage by exonucleases while hindrance to the binding of eukaryotic initiation factor-4A (eIF4A) to the target mRNA leads to translational inhibition.

The double-stranded mature miRNA is unwound, and the opposite strand is degraded. The remaining strand, the mature or the guide strand, is the final single-stranded miRNA molecule which forms a complex with the AGO proteins called the RNA-induced silencing complex (RISC) (Ref. Reference Mishra, Yadav and Rani3). There are four mammalian AGO proteins (AGO1–4) but only AGO2 is able to cleave the target mRNA complementary to the miRNA (Ref. Reference Liu12). The RISC complex binds to target messenger RNAs (mRNAs) which possess sequences complementary with the miRNA (Ref. Reference Gurbuz and Ozpolat13). The binding of the RISC complex to mRNAs is mediated by a 6–8 nucleotide long region within the miRNA called the seed sequence or miRNA binding site (Ref. Reference Hayes, Peruzzi and Lawler14). The resulting miRNA-mRNA duplex leads to an inhibition or in some cases enhancement, of translation (Ref. Reference Hayes, Peruzzi and Lawler14). The miRNA target sites are usually located at the 3′-untranslated region (3′-UTR) of mRNA and the binding of the RISC complex leads to gene silencing by translation repression and mRNA decay (Ref. Reference Jonas and Izaurralde15). Mechanistically, the AGO protein recruits the GW182 which interacts with the polyadenylate-binding protein PABPC to induce mRNA deadenylation (Ref. Reference Braun16). This promotes decapping of the mRNA and makes it susceptible to degradation by 5′-3′ exoribonucleases (Ref. Reference Braun17). For translational repression, GW182 recruits carbon catabolite repressor protein 4 complexes which in turn recruit RNA helicases like DDX6 (Refs Reference Fabian18, Reference Mathys19). MicroRNA-mediated inhibition of mRNA translation initiation results from the interference with the eukaryotic initiation factors eIF4A-I and eIF4A-II (Ref. Reference Jonas and Izaurralde15). Although exact molecular details remain to be uncovered, existing evidences suggest that the RISC complex dissociates initiation factors from target mRNAs inhibiting the assembly of the translation initiation complex (Refs Reference Fukao20, Reference Fukaya, Iwakawa and Tomari21).

Although a perfect complementarity with mRNAs is optimal for the function of miRNAs, some miRNAs can regulate mRNAs with partial complementarity (Ref. Reference Ganju22). Consequently, miRNA-mediated regulation of gene expression affects almost every fundamental cellular process, such as development, differentiation, proliferation, metabolism and apoptosis (Ref. Reference Barger and Nana-Sinkam23). Unsurprisingly, the dysregulation of miRNA profile also significantly correlates with the onset and progression of cancer (Ref. Reference Xue24). The impact of individual miRNAs on cancer often differs between cancer types. A miRNA can be tumour suppressive, oncogenic or a regulator of metastasis (Ref. Reference Kim25). This review will focus on the role of miRNA in cancer metastasis. We will investigate miRNA dysregulation in different stages of the metastatic process and examine molecular drivers affected. We will summarise pre-clinical studies that have successfully employed miRNA-based therapies for metastatic cancer and highlight their limitations. Finally, we will examine miRNA-based therapies for the treatment of advanced and metastatic cancer that are currently in clinical trials.

Metastasis

The development of secondary tumours in distant organs, i.e. metastasis, is a hallmark of cancer (Ref. Reference Rajasegaran26). The spread of cancer cells to secondary sites is the main cause of cancer-related morbidity and mortality (Ref. Reference Dillekås, Rogers and Straume27), yet we are only beginning to unravel the molecular mechanisms that drive metastasis (Ref. Reference Fares28). Several phases are involved in the development of secondary tumours (Ref. Reference Seyfried and Huysentruyt29). First, cancer cells loose adhesion factors and detach from the primary tumour allowing penetration and intravasation into the circulatory and lymphatic systems (Ref. Reference Seyfried and Huysentruyt29). The cells in the vasculature called circulating tumour cells (CTCs), exploit mechanisms like cell cycle arrest, to evade and survive immune surveillance (Ref. Reference Zeeshan and Mutahir30). There is significant research aimed at studying and characterising CTCs which is beyond the scope of this review. Secondly, this process is followed by the extravasation and infiltration of the cells into distant capillary beds (Ref. Reference Welch and Hurst31). Finally, invasion and proliferation of the tumour in distant organs occurs (Ref. Reference Welch and Hurst31). Metastasis and the establishment of secondary tumours is a very inefficient process as the majority of tumour cells in circulation are eliminated (Ref. Reference Labelle and Hynes32). The establishment of a microenvironment for cancer cells to seed, known as a premetastatic niche, is essential for the development of secondary cancer (Ref. Reference Liu and Cao33) (Fig. 2). The most common locations of metastasis in the body are liver, bone, lung, nervous system, pleura and peritoneum (Ref. Reference Riihimäki34). In this review, we will focus on the role of miRNAs in metastasis. We will discuss different stages in the metastatic process and summarise miRNAs that have been reported to be involved in each of these steps.

Fig. 2. MiRNAs and different stages of metastasis: Several miRNAs are dysregulated throughout different stages of the metastatic process including disruption of tight junctions, epithelial to mesenchymal transition (EMT), migration and invasion, angiogenesis, stemness and metastatic growth and tumour secondary microenvironment.

MicroRNA dysregulation in cancer metastasis

MiRNAs are regulators of virtually every cellular process, including the ones that lead to the development of metastasis (Ref. Reference Solé and Lawrie35). MiRNA can target the mRNA of tumour suppressors or oncogenes implicated at different phases of the developing metastatic tumour (Table 1) (Ref. Reference Peng and Croce36). Genetic mutations in cellular pathways resulting from the dysregulation of miRNA have been widely identified in the metastatic pathways (Ref. Reference Iorio and Croce37). Here we discuss miRNA dysregulation and how it impacts key stages of metastasis.

Table 1. A list of microRNAs involved at different stages of cancer metastasis

MiRNAs and cell junctions

MiRNAs regulate the transcription of cellular junction proteins critical for signalling communication, growth and migration (Ref. Reference Zhuang38). For metastasis to occur, the disruption of cellular junctions is essential (Ref. Reference Martin39). Several miRNAs regulate the expression of zonula occluden-1 (ZO-1), a major component of tight junctions (Ref. Reference Zhou40), e.g. miR-105 regulates the metastasis of breast cancer by inhibiting ZO-1 (Ref. Reference Zhou40). Furthermore, overexpression of miR-105 induces the metastasis of cancer cells to distant organs including the liver (Ref. Reference Zhou40). The cell-cell adhesion for the adaption of the premetastatic niche is promoted by oncogenic miR-105 through the tight junction protein ZO-1 (Ref. Reference Zhou40).

A highly elevated expression of miR-486-5p in CD31+ vascular endothelial (VE) cells increases permeability and promotes non-small cell lung cancer (NSCLC) metastasis (Ref. Reference Sun, Han and Shi41). The transfection of human VE cells, with miR-486-5p antagomirs, targets CADM1 and destroys the tight junctions of VE cells. Similarly, the downregulation of adherens junction protein, VE cadherin (CDH5), by miRNA, enhances breast cancer cell migration (Ref. Reference Di Modica42). MDA-MB-231-GFP breast cancer cells transfected with miR-939 mimics, show higher migration through the endothelial barrier which is mediated by a downregulation of CDH5 (Ref. Reference Di Modica42). MiR-145 negatively correlates with the focal junction protein paxillin in colorectal cancer (CRC) (Ref. Reference Qin43). Delivery of miR-145 mimics downregulates paxillin and inhibits cell proliferation, migration and invasion (Ref. Reference Qin43). Similarly, miR-7 targets focal adhesion kinase (FAK) expression to suppress colon cancer proliferation, migration and invasion (Ref. Reference Zeng44). HCT-8 and Caco-2 colon cancer cell lines transfected with miR-7 mimics have been shown to have a negative correlation with colon cancer metastasis (Ref. Reference Zeng44).

MiRNAs in epithelial to mesenchymal transition (EMT)

Epithelial to mesenchymal transition (EMT) of cells is mostly controlled by zinc finger E-box binding protein transcription factors (TFs) ZEB1 and ZEB2 and leads to an upregulation of vimentin and N-cadherin and repression of E-cadherin (Ref. Reference Vu and Datta45). The miR-200 family (miR-200a, miR-200b, miR-200c, miR-124, miR-429) has been extensively investigated as a regulator of ZEB1 and ZEB2 in cancer that mostly metastasises to the liver, including colorectal (Ref. Reference Vu and Datta45), pancreatic (Ref. Reference Gui46), lung (Ref. Reference Liu47), breast (Ref. Reference Ji48) and gastric cancers (Ref. Reference Yu49). ZEB1 and ZEB2 are direct targets of several miRNAs in CRC and NSCLC. Zheng et al. reported a significant downregulation of miR-132 in CRC and furthermore used a luciferase activity assay to demonstrate miR-132-mediated regulation of ZEB2 (Ref. Reference Zheng50). MiR-215-mediated regulation of ZEB2 expression has been demonstrated in CRC while low levels of miR-186-5p in CRC inhibit EMT by targeting ZEB1 (Refs Reference Chen51, Reference Li52). Additionally, it was reported that miR-132 directly targets ZEB2 in NSCLC driving EMT (Ref. Reference You53). There are other miRNAs that promote EMT in breast cancer. MiR-9 repression of E-cadherin, increases EMT in breast cancer (Ref. Reference Ma54) while oncogene miR374a targets negative regulators of Wnt/B-catenin pathway including WIF1, PTEN and WNT5A and activates the Wnt/B-catenin pathway to promote EMT in breast cancer (Ref. Reference Cai55). The downregulation of miR-203 promotes EMT in gastric cancer by releasing the repression of its target gene Annexin A4 (Ref. Reference Li56). In pancreatic ductal adenocarcinoma (PDAC), EMT is driven by an increase in the expression of c-MYC, HMGA2 and KRAS mediated by a reduction in Let-7 miRNA expression (Ref. Reference Wang57).

MiRNAs in cancer cell migration and invasion

MiR-10b was the first miRNA to be associated with metastasis in patients with advanced breast cancer (Ref. Reference Kim58). MiR-10b promotes the migration and invasion in different cancer types including colorectal, breast and gastric cancers by regulating HOXD10 (Refs Reference Kim58Reference Wang60). In breast cancer, migration and invasion is enhanced by an upregulation of miR-96, miR-135a and miR-21 and the downregulation of miR-150. Similarly, in breast cancer, oncogenic miR-96 targets PTPN9 (Ref. Reference Hong61) and miR-135a represses HOXA10 to drive the migration and invasion (Ref. Reference Chen62). Another oncogene miR-21-5p upregulates the expression of programmed cell death protein 4 (PDCD4) to increase breast cancer invasion and migration (Ref. Reference Li63). Tumour suppressor miR-150, which targets HMGA2, is aberrantly expressed in breast cancer and promotes migration and invasion (Ref. Reference Wang64). In NSCLC, a downregulation of miR-373 which regulates TFIIB-related factor 2 (BRF2), promotes cell migration and invasion (Ref. Reference Wang65). Multiple investigations of cancers metastasising to the liver including pancreatic cancer and lung cancer, demonstrate that cell migration and invasion is controlled by miR-29c-mediated regulation of MMP2 (Ref. Reference Wang66). Low levels of miR-124 and miR-203 promote migration and invasion in CRC by targeting ROCK1 and eukaryotic initiation factor 5A2, respectively (Refs Reference Deng67, Reference Zhou68). Similarly, gastric cancer migration and invasion is driven by the dysregulation in miR-520c-mediated regulation of IRF2 (Ref. Reference Li69).

MiRNAs in stemness, angiogenesis and metastatic growth

Key events of metastatic growth and tumour mediated angiogenesis are regulated by miRNAs. Downregulation of miR-200c and overexpression of miR-30c targets stemness-related genes in breast cancer influencing secondary tumour growth (Ref. Reference Rahimi70). Oncogene miR-9-mediated repression of E-Cadherin contributes to an overexpression of vascular endothelial growth factor (VEGF) leading to an increase in angiogenesis which is essential for the growth of secondary tumour (Ref. Reference Ma54). The co-repression of miR-129-2 and miR-335 significantly upregulates oncogenic SOX4 driving metastatic growth in PDAC (Ref. Reference Huang71). Likewise, VEGF overexpression leading to angiogenesis, tumour growth and invasion in uveal melanoma was significantly suppressed by miR-145 mimics directly targeting N-RAS and VEGF signalling pathways (Ref. Reference Yang72). Upregulation of miR-574-5p promotes metastatic growth in NSCLC enhancing tyrosine phosphorylation of B-catenin via the repression of protein tyrosine phosphate receptor type U (PTPRU) (Ref. Reference Zhou73).

MiRNAs in tumour microenvironment

MiRNA is an important mediator of the crosstalk between the tumour microenvironment and tumour cells, playing an important role in metastasis progression. Tumour-associated macrophages (TAM) are key components of the tumour microenvironment, regulated by miRNAs, to exhibit pro-tumour activity in the microenvironment. For example, highly expressed miR-301a in pancreatic cancer cells induces M2 macrophage polarisation via the PTEN/PI13Kℽ signalling pathway to promote pancreatic cancer cell metastasis (Ref. Reference Wang74). Similarly, miR-130-3p upregulated in gastric cancer mediates communication between M2 macrophages and cancer cells in the tumour microenvironment by promoting the expression of mixed lineage leukaemia 3 (MLL3) gene and grainyhead-like 2 (GRHL2) gene (Ref. Reference Zhang75). Tumour-derived exosomal miR-934 promotes liver metastasis of CRC by regulating the interaction between TAMs and the metastatic microenvironment (Ref. Reference Zhao76). Downregulated miR-148b expression negatively correlates with the upregulation of colony-stimulating factor-1 (CSF1), promoting CSF1 signalling and inducing TAM infiltration to promote hepatocellular carcinoma (HCC) metastasis (Ref. Reference Ke77). Furthermore, M2 macrophage-derived exosomal miR-501-3p downregulated TGFRR3 to promote liver and lung metastasis of PDAC in nude mice by activating the TGF-β signalling pathway (Ref. Reference Yin78). Upregulated miR-214 negatively correlates with PTEN in several cancers including breast, HCC, NSCLC and pancreatic cancer. The upregulated miR-214 promotes regulatory T-cells (Tregs) which secret high levels of IL-10 and enhance immune suppression for metastatic progression (Ref. Reference Yin79).

Regulation of metastatic miRNAs

The inter-regulation between miRNAs and different TFs lead to a finely tuned and spatio-temporally regulated transcriptional and post-transcriptional gene regulation system which gets perturbed during metastasis (Ref. Reference Liu80). Evidences suggest that the dysregulation of miRNAs in cancer can occur at the genomic level. An example is the frequently lost genomic locus of miR-146a in acute myeloid leukaemia (Ref. Reference Zhao and Starczynowski81). However, aberrations at the transcription level are widely studied and thought to be more impactful. For instance, tumour suppressive TF p53 regulates the expression of the miR-16, miR-145 and miR-34 family (Ref. Reference He82). While miR-145 is repressed by the oncogenic RAS-responsive element-binding protein 1 (RREB1) (Ref. Reference Kent, Fox-Talbot and Halushka83). Other reported TFs that regulate miR-145 include CCAAT/enhancer-binding protein beta, beta-catenin/T cell factor 4 and forkhead TFs FOXO1 and FOXO3 (Refs Reference Zeinali84, Reference Gan85). The oncogenic c-Myc TF suppresses expression of miRNAs 29, 30 and let-7 family (Refs Reference Chang86Reference Zhang88). ZEB1 and ZEB2, key activators of EMT repress the expression of miR-200 family of genes (Ref. Reference Guan89) including miR-200c (Ref. Reference Chen90). Similarly, studies have demonstrated that nuclear receptors, especially estrogen receptor (ER) and androgen receptor (AR) can directly regulate the transcriptional activity of miRNAs in cancer by binding to promoter or repressor regions. For instance, ER binds to the promoter region of the miR-221 and inhibits its expression in breast cancer (Ref. Reference Di Leva91). Interestingly in prostate cancer, a negative feedback loop that regulates miR-135a and AR protein expression in an androgen-dependent manner was identified. Here, androgen stimulates the expression of miR-135a which inhibits AR expression. In turn, AR binds to the miR-135a locus and controls its expression (Ref. Reference Coarfa92). Some studies have also indicated roles of epigenetic factors like DNA methylation in the regulation of metastatic miRNAs like miR-200c (Refs Reference Davalos93, Reference Ceppi94). Similarly, the promoter of miR-34a is hypermethylated in ovarian cancer (Ref. Reference Schmid95). Other factors that modulate miRNA activity like regulation and post-translational modifications of AGO proteins (Ref. Reference Cheng, Li and Han96), miRNA transport to the cytoplasm and regulation of miRNA–mRNA interactions need to be further explored in the context of metastasis.

Several recent studies have also compared changes in miRNA expression in primary and secondary tumours to identify potential drivers of metastasis. For instance, a study involving 33 CRC patients with metastasis and 14 patients without metastasis revealed differential expression of 17 miRNAs and their 198 predicted targets. There was a strong association of the target genes with cancer progression and metastasis (Ref. Reference Lee97). In another study involving metastatic breast cancer patients, the upregulation of miR-342-3p and miR-187-3p was associated with an increased progression-free survival (PFS) and overall survival (OS); while, the downregulation of miR-301a-3p was associated with a higher PFS and OS (Ref. Reference Martinez-Gutierrez98). In addition to being therapeutic targets, studies in several different cancer types have identified differences in expression levels of several miRNA indicating that they may serve as diagnostic or prognostic markers (Refs Reference McGuire, Brown and Kerin99Reference Song106).

MiRNA-based therapies

Endogenous miRNAs play a crucial role in maintaining cellular homoeostasis (Ref. Reference Rupaimoole107). The genomic and transcriptomic alterations in cancer cells can perturb the global miRNA expression profile causing genome-wide transcriptional changes (Ref. Reference Bartel108). These changes can lead to an upregulation of oncogenes and/or a downregulation of tumour suppressors which is critical for metastasis (Ref. Reference Bartel108). Most miRNA-targeted cancer therapies focus on restoration or inhibition of dysregulated miRNAs (Ref. Reference Raue109) but recently, miRNA-based detargeting strategies have been utilised for cell/tissue-specific targeted therapies (Ref. Reference Dhungel, Ramlogan-Steel and Steel110). Table 2 provides a comprehensive list of miRNA-based therapeutic strategies which will be discussed in this section in detail.

Table 2. MiRNA-based therapies for cancer metastasis: Dysregulated miRNAs are potential therapeutic targets to treat metastatic cancer

MiRNA replacement

The restoration of miRNAs that are downregulated in cancer is one approach to target metastasis (Ref. Reference To4). MiRNAs in metastatic cells can be restored using miRNA mimics (Ref. Reference Hosseinahli111) which are small synthetic RNA duplexes containing an antisense strand with the same sequence as the endogenous miRNA (Ref. Reference van Rooij and Kauppinen112). To increase stability of the duplex and to enhance cellular uptake, the sense strand can be chemically modified. The sense strand may also contain several mismatches to minimise off-target effects (Ref. Reference van Rooij and Kauppinen112). Like the naturally occurring miRNA, these miRNA mimics are loaded into the RISC complex and inhibit downstream targets (Fig. 3a) (Ref. Reference van Rooij and Kauppinen112). MiRNA mimics have been widely studied for therapeutic purposes in both in vitro and in vivo cancer models (Ref. Reference Zhang113). For instance, miR-149-3p mimics suppress breast cancer growth and metastasis by regulating inhibitory receptors and Foxp1 gene expression in CD8+ T cells in a homograft mouse model (Ref. Reference Zhang113). Treatment with miR-149-3p mimics reduced the apoptosis of CD8+ T cells which mediate the immune surveillance of cancer cells (Ref. Reference Zhang113). The promotion of CD8+ T cells resulted in the death of 4T1 mouse breast cancer cells (Ref. Reference Flamini, Jiang and Cui114). Similarly, a reduction of migration and invasion in A549 and SK-MES1 squamous carcinoma NSCLC cell lines was observed after the transfection of miR-140-5p mimics (Ref. Reference Flamini, Jiang and Cui114). The authors also reported a higher adhesion to an artificial extracellular matrix (ECM), indicating a change in EMT (Ref. Reference Flamini, Jiang and Cui114). A combined delivery of three miRNA mimics, miR-195-5p, miR-101-3p and miR-338-5p, is more effective in reducing tumour growth and the number of metastatic nodules in animal models of lung cancer (Ref. Reference Liu115). Similarly, delivering miR-34a mimic sensitises primary and metastatic derived lung cancer cell lines to radiotherapy, in vitro and in vivo (Ref. Reference Cortez116). Several other miRNA replacement therapies are currently being tested in both clinical and pre-clinical settings.

Fig. 3. MiRNA-based therapies. (a) MiRNA replacement with mimics function like an overexpression of endogenous miRNA and increase the degradation or repression of target mRNAs. (b) The miRNA inhibitor approach minimises the binding of miRNA-induced silencing complex (miRISC) to target mRNAs. Different strategies used for miRNA inhibition includes antisense oligonucleotides (ASOs), antagomir antisense oligonucleotides, locked nucleic acid (LNA), antisense oligonucleotide and small RNA zippers. (c) MiRNA sponge binds to the miRISC complex reducing its binding to the target mRNA. (d) MiRNA mask prevents the miRISC from binding to the mRNA by ‘masking’ the miRNA binding site.

MiRNA inhibition

Another approach to target metastasis is to inhibit upregulated oncogenic miRNAs (Ref. Reference Nguyen and Chang117). The inhibition of oncogenic miRNAs overexpressed during metastasis can restore silenced tumour suppressors (Ref. Reference Nguyen and Chang117). MiRNA inhibitors are single-stranded oligonucleotides complimentary to an endogenous miRNA (Ref. Reference To4). These inhibitors can bind to endogenous miRNAs and inhibit their incorporation into the RISC complex (Fig. 3b) (Ref. Reference Rupaimoole and Slack118). Several types of miRNA inhibitors have shown therapeutic advantages both in vitro and in vivo including antisense oligonucleotides (ASOs) (Ref. Reference Ge119), antagomirs (Ref. Reference Xie120), miRNA sponges (Ref. Reference Tay121), miRNA masks (Ref. Reference Zhang122), locked nucleic acid (LNA) anti-miRNAs (Ref. Reference Nedaeinia123) and small miRNA Zippers (Ref. Reference Meng124).

Synthetic Antisense Oligonucleotides (ASOs)

ASOs are single-stranded, chemically modified DNA molecules, 20-25 nucleotides in length, with a full complementarity to a target miRNA (Ref. Reference Bajan and Hutvagner125). ASOs inhibit the binding of mature miRNA to its target mRNA by producing an ASO-miRNA duplex which can lead to the cleavage of the miRNA and the upregulation of the target mRNA (Ref. Reference Bajan and Hutvagner125). ASOs have already been approved by the Food and Drug Administration for the treatment of Duchenne muscular dystrophy and spinal muscular atrophy whereby exon skipping strategies are utilised to restore the dystrophin expression (Ref. Reference Rinaldi and Wood126). For cancer therapy and metastasis inhibition, some pre-clinical studies have been reported with ASOs. For example, Ge et al. designed an ASO to target miR-21 which is overexpressed in NSCLC. MiR-21 regulates the activity of PTEN, a regulator of invasion and a metastasis promoter (Ref. Reference Ge119). The ASO-based drug was successful in reducing miR-21 expression and induced apoptosis in H1650 NSCLC cell line (Ref. Reference Ge119). Likewise, the transfection of synthetic ASOs targeting miR-21 significantly reduced migration and invasion of HCT116 human colon carcinoma cell line accompanied by a reduction in the expression of VEGF which is critical for colon cancer metastasis (Ref. Reference Tao127). Multiple oncogenic miRNAs can also be targeted simultaneously for additive therapeutic effects (Ref. Reference Devulapally128). ASOs targeting miR-21 and miR-10b have been successfully delivered in cell lines and tumour xenografts for triple-negative breast cancer (TNBC) (Ref. Reference Devulapally128). The simultaneous delivery of these miRNAs induces cancer apoptosis and inhibits tumour growth and metastasis in a mouse model of TNBC (Ref. Reference Devulapally128).

Antagomir antisense oligonucleotides

Antagomirs are artificially synthesised single stranded RNA of 23 nucleotides length complementary to a miRNA. Antagomirs can be chemically modified with a cholesterol moiety for greater stability (Ref. Reference Xie120). In a mouse model of pancreatic cancer, a cholesterol-modified polymetric CXCR4 antagonist was delivered with nanoparticles via an intraperitoneal delivery to localise efficacy and limit systemic side (Ref. Reference Xie120). The co-delivery of antagomirs against miR-210 and siRNA against KRAS to this model demonstrated a reduced metastatic activity (Ref. Reference Xie120). Of particular importance was the complete inhibition of liver metastasis, the primary metastatic site of PDAC (Ref. Reference Xie120).

MiRNA sponges antisense oligonucleotide

MiRNA sponges are short, synthetic transcripts with the same sequence as the 3′UTR of mRNAs targeted by the miRNA. Acting as a decoy, sponges inhibit the ability of miRNAs to regulate their target mRNAs (Fig. 3c) (Ref. Reference Tay121). There are some reports where miRNA sponging has been successfully performed for a single miRNA (Ref. Reference Ebert and Sharp129). Liang, Zhang, Zhou, Wu, Lin and Liu (Ref. Reference Liang130) designed a miRNA sponge plasmid to target miR-10b in metastatic breast cancer cell lines, MDA-MB-231 and MCF-7, demonstrating an inhibition of miR-10b and upregulation of its target HOXD10. This resulted in an inhibition of cancer growth and proliferation as well as a reduction in migration and invasion (Ref. Reference Liang130). Additionally, a multi-potent miRNA sponge that simultaneously inhibits 4 oncogenic miRNAs, miR-155, miR-21, miR-221 and miR-222 was developed (Ref. Reference Jung131). This multi-potent miRNA sponge was successful in inhibiting multiple oncogenic miRNAs, thus promoting anti-tumour effects in human breast cancer and pancreatic cancer cells (Ref. Reference Jung131). Results demonstrated the multi-potent miRNA sponge to be more effective in inhibiting proliferation when compared to single miRNA-targeted sponges and demonstrated a 1.3-2.3-fold change in the protein levels of Foxo3a, PTEN and RhoA which are associated with an increased metastatic potential (Ref. Reference Jung131).

MiRNA-masking antisense oligonucleotide

MiRNA-Masking (miR-Mask) is an inverted approach to protect mRNAs from miRNA-mediated repression (Ref. Reference Zhang122). In this approach, the miR-Masking oligonucleotides shield the miRNA binding sites of the mRNA to be protected (Fig. 3d) (Ref. Reference Zhang122). A full complementarity is required for better specificity (Ref. Reference Zhang122). This approach inhibits miRNA-mediated repression of targeted mRNAs without effecting the expression and potentially important functions of a miRNA (Ref. Reference Zhang122). Zhang et al. studied the effects of a miR-mask designed to complement the miR-522 binding site within DENND2D for the treatment of NSCLC and observed a reduced cell migration and invasion in NSCLC cells (Ref. Reference Zhang122).

Locked nucleic acid (LNA) antisense oligonucleotide

Another alternative oligonucleotide designed to inhibit miRNA oncogenic function are locked nucleic acid anti-miRs (LNA-i-miR) (Ref. Reference Nedaeinia123). LNAs are chemically modified by connecting the 2′ oxygen and 4′ carbon to form an extra methylene bridge locking the ribose ring (Ref. Reference Nedaeinia123). This leads to a higher thermal and in vivo stability and a greater binding affinity with mRNA targets (Ref. Reference Nedaeinia123). LNA against miR-21 was effective in reducing the invasiveness and inhibited the proliferation of human colorectal adenocarcinoma cells (Ref. Reference Nedaeinia123). Likewise, Lima et al. devised a strategy whereby LNA was efficient even when delivered at a low dose. In their study, miR-9 was targeted with LNAs to promote the expression of CDH1 for the reestablishment of E-cadherin in human gastric cancer cells (Ref. Reference Lima132). In another study, the delivery of LNA-i-miRs against miR-663a and miR-4787-5p reduced tumour burden and metastasis in an orthotopic mouse model of pancreatic cancer by decreasing TGFβ1-induced EMT (Ref. Reference Mody133).

Small RNA zippers

In this approach, oligonucleotides complementary to the second and the first half of a miRNA are synthesised and delivered into the cells (Ref. Reference Meng124). Small RNA zippers connect multiple copies of a miRNA end-to-end by forming a duplex of multiple miRNA copies and inhibit the function of the target miRNA (Ref. Reference Meng124). Like chemically modified LNAs, small RNA zippers have increased affinity, specificity and stability (Ref. Reference Meng124). A 70–90% inhibition of miR-221 and miR-17 and rescue of their target genes was observed in breast cancer cell lines using miRNA zippers (Ref. Reference Meng124). Further, the oncogenic effects of miR-221 were reversed by miR-221 zippers as demonstrated by the cell migration assay. However, the in vivo applications of miRNA zippers are yet to be tested (Ref. Reference Meng124).

MiRNA-mediated detargeting

Unlike miRNA replacement or inhibition therapies, this approach utilises the binding sites of miRNAs that are downregulated in cancer for detargeting the therapy from the normal cells and thereby reduce off-target effects. This approach is mostly useful for genetic therapies which utilise therapeutic gene transfer (Ref. Reference Kopp134). For instance, Baertsch et al. utilised three miRNAs; miR-122, miR-7, miR-148a, expressed at high levels in the liver, brain and the gastrointestinal tract, respectively, and demonstrated successful detargeting of these organs for a measles virus-mediated oncolytic virotherapy of pancreatic cancer in cell lines and murine xenograft models (Ref. Reference Baertsch135). In another example, the binding sites of miRNAs downregulated in PDAC, miR-148a and miR-216a, were used for detargeting in locally advanced and metastatic pancreatic and liver cancer (Ref. Reference Bofill-De Ros, Gironella and Fillat136). 8-miR148aT demonstrated detargeting effects by repressing all miR-148/152 family members in the pancreas and liver (Ref. Reference Bofill-De Ros, Gironella and Fillat136). This study demonstrated that this method was highly efficient for targeted therapies as a significant decrease in cancer growth and metastasis was observed (Ref. Reference Bofill-De Ros, Gironella and Fillat136). To prevent off-targets effects in the liver after suicide gene therapy, the binding sites of miR-122a and miR-199a, which are significantly downregulated in HCC, were used in multiple studies. Adeno-associated virus-based vectors were used to deliver miRNA122a and/or miRNA199a-regulated the suicide gene therapy system cytosine deaminase (CD)/ 5-fluorocytosine (Refs Reference Dhungel137Reference Dhungel, Ramlogan-Steel and Steel139). Limited killing of normal liver cells with this system demonstrated an efficient liver detargeting using the binding sites of these miRNAs (Refs Reference Dhungel137Reference Dhungel, Ramlogan-Steel and Steel139).

MiRNA-based therapies in clinical trials

MiRNA-based strategies have demonstrated therapeutic potential in a range of conditions including advanced cancers (Ref. Reference Fortunato and Iorio140). In fact, several miRNA-targeted therapeutics are at different phases of clinical development for the treatment of advanced cancers and metastases (Ref. Reference Ivkovic141). For cancer therapy, miRNA therapeutics are injected directly into the site of the tumour which can increase the specificity, efficacy and reduce off-target effects (Ref. Reference van Rooij and Kauppinen112). Below we summarise clinically applied miRNA-targeted therapies (Table 3). Although not exclusive to metastasis, these therapies have shown promise in treating advanced cancers including those with metastasis to secondary organs.

Table 3. A list of cancer therapy clinical trials utilising miRNA-based strategies

Clinical miRNA replacement

Tumour suppressor miR-15/16 family is downregulated in various cancers including lung and colon cancer that metastasise to the liver (Ref. Reference Reid142). Downregulation of miR15/16 can increase drivers of metastasis including tumour growth, angiogenesis, EMT and stemness (Ref. Reference Ivkovic141). In pre-clinical studies, miR-16 mimic replacement safely inhibited growth and metastasis of Malignant Pleural Mesothelioma (MPM) and NSCLC xenograft tumours (Ref. Reference Reid143). TargomiRs are minicells coated with an anti-EGFR-specific antibody carrying miR-16 mimics for a cancer-targeted delivery (Ref. Reference Reid143). A Phase 1 clinical trial of TargomiR initiated in 2014 demonstrated the safety of the approach in 26 patients. Of the 22 patients who were assessed for response, one showed a partial response and 15 had stable disease (Ref. Reference van Zandwijk144).

Tumour suppressor miR-34a downregulates the expression of oncogenes including MET, MYC, PDGFR-α, CDK4/6 and BCL2 (Ref. Reference Hong145). Both in vitro and in vivo studies have reported that miR-34a mimics can reduce tumour growth, migration and invasion and metastasis (Ref. Reference Reda El Sayed146). Anti-tumour activity of co-injecting let-7 and miR-34 was demonstrated in multiple NSCLC cell lines (Ref. Reference Kasinski147). The anti-tumour activity of this combinatorial therapy was also tested in KrasLSD−G12D/+; p53flx, flx mouse model of NSCLC (Ref. Reference Kasinski147). A second in vivo study was then initiated using lipid-based delivery agent (NOV340) to deliver miR-34a mimics (Ref. Reference Kasinski147). A Phase I clinical trial with MRX34, a liposomal formulation of a synthetic, double-stranded miR-34a mimic, was initiated for patients with HCC and unresectable liver metastasis. (Refs Reference Kasinski147, Reference Beg148). Unfortunately, adverse immune-mediated toxicities precluded the trial advancing to phase II (Clinical Trial identifiers: NCT01829971, NCT02862145) (Ref. Reference Beg148).

Tumour suppressor miR-193a-3p is downregulated in a range of cancers including HCC (Ref. Reference Grossi149), NSCLC (Ref. Reference Gao150) and TNBC (Ref. Reference Yu151). The repression of miR-193a-3p in these cancers decreases apoptosis, increases cell proliferation and migration tumour growth and metastasis (Ref. Reference van den Bosch152). Targets of miR-193a-3p play an important role in malignant cell behaviour including KRAS (Ref. Reference Fan153), ERBB (Ref. Reference Liang154), and S6K2 (Ref. Reference Yu155) in lung cancer, PLAU in bladder cancer (Ref. Reference Lv156), MCL-1 in glioma (Ref. Reference Kwon157), CCND1 in prostate cancer (Ref. Reference Liu158), RAB27B in osteosarcoma (Ref. Reference Pu159) and SRSF2 in HCC (Ref. Reference Bader160). Telford et al. reported that miR-193a-3p mimics reduce cancer cell proliferation/survival by inducing cell cycle arrest, apoptosis, increased cell senescence, DNA damage and inhibit migration (Ref. Reference Telford161). INT-1B3, a 193a-3p mimic replacement drug, consists of a lipid nanoparticle-based delivery system (Ref. Reference van den Bosch152). In preclinical studies with tumour bearing mice, systemic injection of INT-1B3 shows significant anti-tumour activity (Ref. Reference van den Bosch152). A phase 1/1b clinical trial to investigate the safety, preliminary efficacy, pharmacokinetics and pharmacodynamics of INT-1B3 is currently ongoing (Clinical Trial identifier: NCT04675996) (Ref. Reference van den Bosch152).

Clinical miRNA inhibition

Oncogenic miR-221-222 cluster located on the X chromosome is highly expressed in several solid tumours such as lung cancer (Ref. Reference Garofalo162), breast cancer (Ref. Reference Miller163), HCC (Ref. Reference Callegari164), and glioblastoma (Ref. Reference Zhang165) as well as haematological malignancies including myeloma (Ref. Reference Di Martino166). In advanced cancers, upregulation of miR-221 interferes with the expression of its targets p27, p57, PUMA and PTEN promoting tumour growth (Ref. Reference van den Bosch152). Di Martino et al. reported anticancer effects of anti-miR221-targeted LNAs both in vitro and in vivo. LNA-i-miR-221 is a 13-mer antisense oligonucleotide that uses LNA technology and phosphorothioate backbone chemistry for increased affinity for miR221 targeting (Ref. Reference Di Martino166). A phase I clinical trial of LNA-i-miR-221 will administer the drug via an intravenous injection to patients with multiple myeloma and advanced solid tumour (Clinical Trial identifier: NCT04811898).

Oncogene miR-155 regulates immune cell function and its overexpression affects multiple genes associated with the promotion of solid tumours including breast cancer, lung cancer, liver cancer as well as haematological malignancies including leukaemia (Ref. Reference Higgs and Slack167). Upregulation of miR-155 has been linked to JAK/STST, NK-KB and PI3K/AKT survival pathways stimulating T-cell receptors (Ref. Reference Seto168). MiR-155 inhibition reduces proliferation and increases apoptosis in T-cell lymphoma cell lines (Ref. Reference Seto168). In xenografts of B-cell lymphoma, miR-155 silencing with a LNA delivered systemically, reduced tumour burden and metastasis (Ref. Reference Zhang169). In another preclinical study, an anti-miR-155 molecule with a peptide nucleic acid backbone was used for greater sensitivity and efficient delivery to treat haematological malignancies (Ref. Reference Cheng170). These preclinical studies lead to the development of Cobomarsen which is a single-stranded, chemically modified miR-155-targeting molecule with chemical modifications for increased stability (Ref. Reference Seto168). A Phase 1 trial of Cobomarsen in patients with cutaneous T-cell lymphoma (CTCL) [mycosis fungoides (MF) subtype] was initiated in 2016 and reported some therapeutic benefit for 95% of all enrolled patients with increased benefits reported for subjects who underwent more than one cycle (Ref. Reference Seto168). Encouraging early data was followed by phase 2 clinical trial which started in 2019 but was terminated in 2020 due to commercial reasons (Clinical Trial identifiers: NCT03603431, NCT03713320, NCT03837457).

Challenges for miRNA-based cancer therapies

There are several challenges in using mi-RNA based therapies including insufficient delivery to the target tissue (cancer), stability in the biological system, immune responses and unwanted off-targeting. Arguably the primary challenge for miRNA-based cancer therapies is their efficient delivery to target tissues. Tumours can have poor blood perfusion and the complexity of ECM often hinder the delivery of miRNA-based therapies. In addition, scavenging cells like TAMs, neutrophils and monocytes can prevent the miRNA carrying vehicle from reaching cancer cells (Ref. Reference Yu, Huang and Li171). To overcome these challenges, different modes of delivery are being investigated, including both viral and non-viral vector systems (Refs Reference Bulcha172, Reference Santana-Armas and Tros de Ilarduya173). The more common viral vectors are derived from adeno-associated viruses, adenoviruses and lentiviruses, while non-viral vectors can include exosomes, polymers and liposomes (Refs Reference Bulcha172, Reference Santana-Armas and Tros de Ilarduya173). Viral vectors have a high efficiency and have been successfully used in several clinical trials. They also form the basis of a number of FDA approved therapies. (Ref. Reference Lundstrom174). Non-viral approaches may also be beneficial. The biocompatibility and biodegradability of polymers and liposomes are some of their advantages (Ref. Reference Karlsen and Brinchmann175). The conjugate vehicles have selective targeting and high stability due to the use of lipid or receptor binding molecules (Ref. Reference Biscans176). Naturally occurring exosomes are an advantage due to their immune compatibility (Ref. Reference Li177). Both viral and non-viral systems have inherent advantages and shortcomings, therefore the choice of the delivery system should be based on the overall design of a study.

Once delivered to the target tissue, the effect of miRNA therapeutics in non-cancer cells needs to be prevented. Similarly, effects on off-target mRNAs within cancer cells is another concern as miRNAs can target multiple transcripts simultaneously (Ref. Reference Bandi and Vassella178). MiRNA imperfect complementarity to the targeted mRNA 3′UTR has the capacity to indiscriminately silence off-target genes (Ref. Reference Kara, Calin and Ozpolat179). Another cause of off-targeting is the possibility of artificial exogenous miRNA competing with endogenous miRNA creating a dysregulation in gene expression (Ref. Reference Kara, Calin and Ozpolat179). Off-target effects can induce the silencing of tumour suppressors or activation of oncogenes in normal cells (Ref. Reference Seok180). For instance, miR-15/16 cluster regulate a large proportion of the whole transcriptome in leukaemia cells (Ref. Reference Pepe181). Thus, these miRNAs would not likely be used as therapeutic targets. The use of cancer targeted viral and non-viral delivery vectors can also reduce unwanted off-target effects (Refs Reference Santana-Armas and Tros de Ilarduya173, Reference Segal and Slack182). Furthermore, cancer cell-specific regulatory elements like tumour specific promoters can be used in the delivery system (Ref. Reference Dhungel183). Vigilant bioinformatic and wet-lab studies need to be performed with proposed inhibitors or mimics to identify any potential off-target effects in pre-clinical studies.

Both the miRNA and the delivery vector can elicit an immune response (Ref. Reference Ceppi184). MiRNA duplexes can trigger toll like receptor response (Refs Reference Yu, Huang and Li171, Reference Ceppi184) leading to an interferon response against miRNA therapeutics (Ref. Reference Yu, Huang and Li171). MiRNA therapeutics designed with certain chemical modifications can mitigate these immune responses (Ref. Reference Yu, Huang and Li171). Chemical modifications can enhance the miRNA stability in vivo. For instance, miRNAs without chemical modification of the ribose 2′-OH are prone to nuclease-mediated degradation and have a short half-life when injected systemically. Three generations of ASOs modification techniques developed (Ref. Reference Rawat1) First-generation modifications substitute phosphodiester backbone with phosphothiorate to increase in vivo stability; (Ref. Reference Kozomara, Birgaoanu and Griffiths-Jones2) Second-generation modifications substitute the 2′-O-alkyl group of the sugar moieties with 2′-OMe, 2′-O-methoxyethyl (2′MOE) or 2′-Fluoro to enhance efficacy and bioavailability and to reduce the immune stimulation and toxicity; (Ref. Reference Mishra, Yadav and Rani3) Third-generation modifications are the chemical alteration to the furanose ring with 2′4′-methylene producing LNAs to reduce nuclease degradation and increase membrane penetration (Ref. Reference Rinaldi and Wood126).

Concluding remarks

The inability of current therapies to effectively treat advanced or metastatic cancers stems from an incomplete understanding of the molecular mechanisms governing metastasis. Understanding molecular drivers of cancer metastasis can provide opportunities to develop novel therapeutic approaches. MiRNAs play a central role in regulating gene expression, thus, the dysregulation of miRNAs in metastasising cells warrants special attention. The dysregulation of several miRNAs is observed at every step of the metastatic process and restoring their levels is an attractive therapeutic avenue. Depending on the cancer type, dysregulated miRNAs can function as an oncogene or tumour suppressor. These dysregulations can lead to significant alterations in the expression of downstream target genes. Therapeutic approaches that utilise miRNAs aim to restore the normal levels of dysregulated miRNAs using miRNA mimics and inhibitors. There are several studies which report a successful use of miRNA mimics and inhibitors in pre-clinical in vitro and in vivo studies for targeting both primary tumours and metastasis for different cancer types. This has led to the initiation of human clinical trials using miRNA-based therapies for both solid and blood cancers. However, there are both technical and practical limitations for delivering miRNA-based therapies to patients. The in vivo stability and delivery of these miRNAs at therapeutic levels to the target tissue is a major issue for clinical applications. Similarly, therapy-induced toxicity and potential off-target effects are major concerns. There are several ongoing developments in this area to increase the stability of miRNAs mostly involving chemical modifications. Similarly, developments in the field of both viral and non-viral vector-based delivery can make the therapy cancer-specific and reduce off-target effects. Further research needs to be performed in order to identify novel miRNAs which control metastasis and potential therapeutic target; only then will the full potential of miRNA-based therapies for cancer metastasis be realised.

Abbreviations: BRF2, TFIIB-related factor 2, CADM1, Cell Adhesion Molecule 1, CDH1, Cadherin-1 or Epithelial cadherin, CDH5, Cadherin 5 or VE-Cadherin, CSF1, Colony-stimulating factor-1, EIF5A2, Eukaryotic Translation Initiation Factor 5A2, FAK, Focal adhesion kinase, HMGA2, High-mobility group AT-hook 2, HOXA10, Homeobox A10, HOXD10, Homeobox D10, IRF2, Interferon regulatory factor 2, IL-10, Interleukin 10, KLF4, Krüppel-like factor 4, KRAS, Kirsten rat sarcoma viral oncogene homologue, MMP2, Matrix metalloproteinase-2, MYC, MYC proto-oncogene, bHLH transcription factor, NANOG, Nanog Homeobox, N-RAS, Neuroblastoma RAS viral oncogene homologue, Oct-4, Octamer-binding transcription factor 4, PDCD4, Programmed Cell Death 4, PTEN, Phosphatase and tensin homologue, PTPN9, Tyrosine-protein phosphatase non-receptor type 9, PTPRU, Protein Tyrosine Phosphatase Receptor Type U, ROCK1, Rho Associated Coiled-Coil Containing Protein Kinase 1, SOX2, SRY-Box Transcription Factor 2, SOX4, SRY-Box Transcription Factor 4, TGF-β, Transforming growth factor beta, TGFBR3, Transforming growth factor beta receptor 3, VEGF, Vascular Endothelial Growth Factor, WIF1, WNT Inhibitory Factor 1, Wnt5a, Wnt Family Member 5A, ZEB1, Zinc finger E-box binding homeobox 1, ZEB2, Zinc finger E-box-binding homeobox 2, ZO-1, Zonula occludens-1. Cancers: Breast, Colon, CRC, Colorectal cancer, Gastric, HCC, Hepatocellular carcinoma, NSCLC, non-small cell lung cancer, pancreatic, PDAC, Pancreatic ductal adenocarcinoma, uveal melanoma.

Acknowledgements

The figures in this manuscript were drawn with Biorender.

Conflict of interest

The authors declare no competing interests.

References

Rawat, M et al. (2019) MicroRNA in pancreatic cancer: from biology to therapeutic potential. Genes (Basel) 10, 752.CrossRefGoogle ScholarPubMed
Kozomara, A, Birgaoanu, M and Griffiths-Jones, S (2019) miRBase: from microRNA sequences to function. Nucleic Acids Research 47, D155–D162.CrossRefGoogle ScholarPubMed
Mishra, S, Yadav, T and Rani, V (2016) Exploring miRNA based approaches in cancer diagnostics and therapeutics. Critical Reviews in Oncology/Hematology 98, 1223.CrossRefGoogle ScholarPubMed
To, KKW et al. (2020) Advances in the discovery of microRNA-based anticancer therapeutics: latest tools and developments. Expert Opinion On Drug Discovery 15, 6383.CrossRefGoogle ScholarPubMed
Bautista-Sánchez, D et al. (2020) The promising role of miR-21 as a cancer biomarker and Its importance in RNA-based therapeutics. Molecular Therapy. Nucleic Acids 20, 409420.CrossRefGoogle ScholarPubMed
Iorio, MV and Croce, CM (2012) MicroRNA dysregulation in cancer: diagnostics, monitoring and therapeutics. A comprehensive review. EMBO Molecular Medicine 4, 143159.CrossRefGoogle ScholarPubMed
Iqbal, MA et al. (2019) MicroRNA in lung cancer: role, mechanisms, pathways and therapeutic relevance. Molecular Aspects of Medicine 70, 320.CrossRefGoogle ScholarPubMed
Acunzo, M et al. (2015) MicroRNA and cancer – a brief overview. Advances in Biological Regulation 57, 19.CrossRefGoogle ScholarPubMed
Ruby, JG, Jan, CH and Bartel, DP (2007) Intronic microRNA precursors that bypass Drosha processing. Nature 448, 8386.CrossRefGoogle ScholarPubMed
Xie, M et al. (2013) Mammalian 5'-capped microRNA precursors that generate a single microRNA. Cell 155, 15681580.CrossRefGoogle ScholarPubMed
Cheloufi, S et al. (2010) A dicer-independent miRNA biogenesis pathway that requires Ago catalysis. Nature 465, 584589.CrossRefGoogle ScholarPubMed
Liu, J et al. (2004) Argonaute2 is the catalytic engine of mammalian RNAi. Science (New York, N.Y.) 305, 14371441.CrossRefGoogle ScholarPubMed
Gurbuz, N and Ozpolat, B (2019) MicroRNA-based targeted therapeutics in pancreatic cancer. Anticancer Research 39, 529532.CrossRefGoogle ScholarPubMed
Hayes, J, Peruzzi, PP and Lawler, S (2014) MicroRNAs in cancer: biomarkers, functions and therapy. Trends in Molecular Medicine 20, 460469.CrossRefGoogle ScholarPubMed
Jonas, S and Izaurralde, E (2015) Towards a molecular understanding of microRNA-mediated gene silencing. Nature Reviews Genetics 16, 421433.CrossRefGoogle ScholarPubMed
Braun, JE et al. (2011) GW182 proteins directly recruit cytoplasmic deadenylase complexes to miRNA targets. Molecular Cell 44, 120133.CrossRefGoogle ScholarPubMed
Braun, JE et al. (2012) A direct interaction between DCP1 and XRN1 couples mRNA decapping to 5' exonucleolytic degradation. Nature Structural & Molecular Biology 19, 13241331.CrossRefGoogle ScholarPubMed
Fabian, MR et al. (2011) miRNA-mediated deadenylation is orchestrated by GW182 through two conserved motifs that interact with CCR4-NOT. Nature Structural & Molecular Biology 18, 12111217.CrossRefGoogle ScholarPubMed
Mathys, H et al. (2014) Structural and biochemical insights to the role of the CCR4-NOT complex and DDX6 ATPase in microRNA repression. Molecular Cell 54, 751765.CrossRefGoogle Scholar
Fukao, A et al. (2014) MicroRNAs trigger dissociation of eIF4AI and eIF4AII from target mRNAs in humans. Molecular Cell 56, 7989.CrossRefGoogle ScholarPubMed
Fukaya, T, Iwakawa, HO and Tomari, Y (2014) MicroRNAs block assembly of eIF4F translation initiation complex in Drosophila. Molecular Cell 56, 6778.CrossRefGoogle ScholarPubMed
Ganju, A et al. (2017) miRNA nanotherapeutics for cancer. Drug Discovery Today 22, 424432.CrossRefGoogle ScholarPubMed
Barger, JF and Nana-Sinkam, SP (2015) MicroRNA as tools and therapeutics in lung cancer. Respiratory Medicine 109, 803812.CrossRefGoogle ScholarPubMed
Xue, J et al. (2017) MicroRNA-targeted therapeutics for lung cancer treatment. Expert Opinion on Drug Discovery 12, 141157.CrossRefGoogle ScholarPubMed
Kim, J et al. (2018) MicroRNAs and metastasis: small RNAs play big roles. Cancer and Metastasis Reviews 37, 515.CrossRefGoogle ScholarPubMed
Rajasegaran, Y et al. (2021) Footprints of microRNAs in cancer biology. Biomedicines 9, 14941536CrossRefGoogle ScholarPubMed
Dillekås, H, Rogers, MS and Straume, O (2019) Are 90% of deaths from cancer caused by metastases? Cancer Medicine 8, 55745576.CrossRefGoogle ScholarPubMed
Fares, J et al. (2020) Molecular principles of metastasis: a hallmark of cancer revisited. Signal Transduction and Targeted Therapy 5, 28.CrossRefGoogle ScholarPubMed
Seyfried, TN and Huysentruyt, LC (2013) On the origin of cancer metastasis. Critical Reviews in Oncogenesis 18, 4373.CrossRefGoogle ScholarPubMed
Zeeshan, R and Mutahir, Z (2017) Cancer metastasis – tricks of the trade. Bosnian Journal of Basic Medical Sciences 17, 172182.Google ScholarPubMed
Welch, DR and Hurst, DR (2019) Defining the hallmarks of metastasis. Cancer Research 79, 30113027.CrossRefGoogle ScholarPubMed
Labelle, M and Hynes, RO (2012) The initial hours of metastasis: the importance of cooperative host-tumor cell interactions during hematogenous dissemination. Cancer Discovery 2, 10911099.CrossRefGoogle ScholarPubMed
Liu, Y and Cao, X (2016) Characteristics and significance of the pre-metastatic niche. Cancer Cell 30, 668681.CrossRefGoogle ScholarPubMed
Riihimäki, M et al. (2018) Clinical landscape of cancer metastases. Cancer Medicine 7, 55345542.CrossRefGoogle ScholarPubMed
Solé, C and Lawrie, CH (2019) MicroRNAs and metastasis. Cancers (Basel) 12, 96117CrossRefGoogle ScholarPubMed
Peng, Y and Croce, CM (2016) The role of MicroRNAs in human cancer. Signal Transduction and Targeted Therapy 1, 15004.CrossRefGoogle ScholarPubMed
Iorio, MV and Croce, CM (2012) Causes and consequences of microRNA dysregulation. Cancer Journal 18, 215222.CrossRefGoogle ScholarPubMed
Zhuang, Y et al. (2016) MicroRNA regulation of endothelial junction proteins and clinical consequence. Mediators of Inflammation 2016, 5078627.CrossRefGoogle ScholarPubMed
Martin, TA (2014) The role of tight junctions in cancer metastasis. Seminars in Cell & Developmental Biology 36, 224231.CrossRefGoogle ScholarPubMed
Zhou, W et al. (2014) Cancer-secreted miR-105 destroys vascular endothelial barriers to promote metastasis. Cancer Cell 25, 501515.CrossRefGoogle ScholarPubMed
Sun, B, Han, Y and Shi, M (2021) Stromal-derived miR-486-5p promotes metastasis of non-small-cell lung cancer cells by targeting the CADM1/tight junctions axis in vascular endothelial cells. Cell Biology International 45, 849857.CrossRefGoogle ScholarPubMed
Di Modica, M et al. (2017) Breast cancer-secreted miR-939 downregulates VE-cadherin and destroys the barrier function of endothelial monolayers. Cancer Letters 384, 94100.CrossRefGoogle ScholarPubMed
Qin, J et al. (2015) MicroRNA-145 suppresses cell migration and invasion by targeting paxillin in human colorectal cancer cells. International Journal of Clinical and Experimental Pathology 8, 13281340.Google ScholarPubMed
Zeng, CY et al. (2016) MicroRNA-7 suppresses human colon cancer invasion and proliferation by targeting the expression of focal adhesion kinase. Molecular Medicine Reports 13, 12971303.CrossRefGoogle ScholarPubMed
Vu, T and Datta, PK (2017) Regulation of EMT in colorectal cancer: a culprit in metastasis. Cancers (Basel) 9, 171193CrossRefGoogle ScholarPubMed
Gui, Z et al. (2017) Oridonin inhibition and miR-200b-3p/ZEB1 axis in human pancreatic cancer. International Journal of Oncology 50, 111120.CrossRefGoogle ScholarPubMed
Liu, C et al. (2018) Roles of miR-200 family members in lung cancer: more than tumor suppressors. Future Oncology (London, England) 14, 28752886.CrossRefGoogle ScholarPubMed
Ji, H et al. (2019) miR-124 regulates EMT based on ZEB2 target to inhibit invasion and metastasis in triple-negative breast cancer. Pathology Research and Practice 215, 697704.CrossRefGoogle ScholarPubMed
Yu, L et al. (2022) Complete loss of miR-200 family induces EMT associated cellular senescence in gastric cancer. Oncogene 41, 2636.CrossRefGoogle ScholarPubMed
Zheng, YB et al. (2014) miR-132 inhibits colorectal cancer invasion and metastasis via directly targeting ZEB2. World Journal of Gastroenterology 20, 65156522.CrossRefGoogle ScholarPubMed
Chen, DL et al. (2017) Long non-coding RNA UICLM promotes colorectal cancer liver metastasis by acting as a ceRNA for microRNA-215 to regulate ZEB2 expression. Theranostics 7, 48364849.CrossRefGoogle Scholar
Li, J et al. (2018) MiR-186-5p upregulation inhibits proliferation, metastasis and epithelial-to-mesenchymal transition of colorectal cancer cell by targeting ZEB1. Archives of Biochemistry and Biophysics 640, 5360.CrossRefGoogle ScholarPubMed
You, J et al. (2014) MiR-132 suppresses the migration and invasion of lung cancer cells via targeting the EMT regulator ZEB2. PLoS One 9, e91827.CrossRefGoogle ScholarPubMed
Ma, L et al. (2010) miR-9, a MYC/MYCN-activated microRNA, regulates E-cadherin and cancer metastasis. Nature Cell Biology 12, 247256.CrossRefGoogle ScholarPubMed
Cai, J et al. (2013) MicroRNA-374a activates Wnt/β-catenin signaling to promote breast cancer metastasis. Journal of Clinical Investigation 123, 566579.Google ScholarPubMed
Li, J et al. (2019) miR-203 inhibits the invasion and EMT of gastric cancer cells by directly targeting annexin A4. Oncology Research 27, 789799.CrossRefGoogle ScholarPubMed
Wang, Y et al. (2017) Lin28B facilitates the progression and metastasis of pancreatic ductal adenocarcinoma. Oncotarget 8, 6041460428.CrossRefGoogle ScholarPubMed
Kim, J et al. (2016) Ablation of miR-10b suppresses oncogene-induced mammary tumorigenesis and metastasis and reactivates tumor-suppressive pathways. Cancer Research 76, 64246435.CrossRefGoogle ScholarPubMed
Wang, Y et al. (2016) miR-10b promotes invasion by targeting HOXD10 in colorectal cancer. Oncology Letters 12, 488494.CrossRefGoogle ScholarPubMed
Wang, YY et al. (2015) MicroRNA-10b promotes migration and invasion through Hoxd10 in human gastric cancer. World Journal of Surgical Oncology 13, 259.CrossRefGoogle ScholarPubMed
Hong, Y et al. (2016) miR-96 promotes cell proliferation, migration and invasion by targeting PTPN9 in breast cancer. Scientific Reports 6, 37421.CrossRefGoogle ScholarPubMed
Chen, Y et al. (2012) miRNA-135a promotes breast cancer cell migration and invasion by targeting HOXA10. BMC Cancer 12, 111.CrossRefGoogle ScholarPubMed
Li, J et al. (2021) Andrographolide suppresses the growth and metastasis of luminal-like breast cancer by inhibiting the NF-κB/miR-21-5p/PDCD4 signaling pathway. Frontiers in Cell and Developmental Biology 9, 643525.CrossRefGoogle ScholarPubMed
Wang, Z et al. (2019) Long intergenic Non-coding RNA 01121 promotes breast cancer cell proliferation, migration, and invasion via the miR-150–5p/HMGA2 axis. Cancer Management and Research 11, 1085910870.CrossRefGoogle ScholarPubMed
Wang, L et al. (2018) MicroRNA-373 inhibits cell proliferation and invasion via targeting BRF2 in human non-small cell lung cancer A549 cell line. Cancer Research and Treatment: Official Journal of Korean Cancer Association 50, 936949.CrossRefGoogle ScholarPubMed
Wang, H et al. (2013) miRNA-29c suppresses lung cancer cell adhesion to extracellular matrix and metastasis by targeting integrin β1 and matrix metalloproteinase2 (MMP2). PLoS One 8, e70192.CrossRefGoogle ScholarPubMed
Deng, B et al. (2016) MiRNA-203 suppresses cell proliferation, migration and invasion in colorectal cancer via targeting of EIF5A2. Scientific Reports 6, 28301.CrossRefGoogle ScholarPubMed
Zhou, L et al. (2016) MicroRNA-124 (MiR-124) inhibits cell proliferation, metastasis and invasion in colorectal cancer by downregulating rho-associated protein kinase 1(ROCK1). Cellular Physiology and Biochemistry 38, 17851795.CrossRefGoogle ScholarPubMed
Li, YR et al. (2016) MicroRNA-520c enhances cell proliferation, migration, and invasion by suppressing IRF2 in gastric cancer. FEBS Open Bio 6, 12571266.CrossRefGoogle ScholarPubMed
Rahimi, M et al. (2020) Down-Regulation of miR-200c and Up-regulation of miR-30c target both stemness and metastasis genes in breast cancer. Cell Journal 21, 467478.Google ScholarPubMed
Huang, HY et al. (2012) SOX4 Transcriptionally regulates multiple SEMA3/plexin family members and promotes tumor growth in pancreatic cancer. PLoS One 7, e48637.CrossRefGoogle ScholarPubMed
Yang, JY et al. (2020) MicroRNA-145 suppresses uveal melanoma angiogenesis and growth by targeting neuroblastoma RAS viral oncogene homolog and vascular endothelial growth factor. Chinese Medical Journal (English 133, 19221929.CrossRefGoogle ScholarPubMed
Zhou, R et al. (2016) MicroRNA-574-5p promotes metastasis of non-small cell lung cancer by targeting PTPRU. Scientific Reports 6, 35714.CrossRefGoogle ScholarPubMed
Wang, X et al. (2020) Correction: hypoxic tumor-derived exosomal miR-301a mediates M2 macrophage polarization via PTEN/PI3Kγ to promote pancreatic cancer metastasis. Cancer Research 80, 922.CrossRefGoogle ScholarPubMed
Zhang, Y et al. (2020) M2 macrophage-derived extracellular vesicles promote gastric cancer progression via a microRNA-130b-3p/MLL3/GRHL2 signaling cascade. Journal of Experimental & Clinical Cancer Research: CR 39, 134.CrossRefGoogle Scholar
Zhao, S et al. (2020) Tumor-derived exosomal miR-934 induces macrophage M2 polarization to promote liver metastasis of colorectal cancer. Journal of Hematology & Oncology 13, 156.CrossRefGoogle ScholarPubMed
Ke, M et al. (2019) MicroRNA-148b-colony-stimulating factor-1 signaling-induced tumor-associated macrophage infiltration promotes hepatocellular carcinoma metastasis. Biomedicine & Pharmacotherapy 120, 109523.CrossRefGoogle ScholarPubMed
Yin, Z et al. (2019) Macrophage-derived exosomal microRNA-501-3p promotes progression of pancreatic ductal adenocarcinoma through the TGFBR3-mediated TGF-β signaling pathway. Journal of Experimental & Clinical Cancer Research: CR 38, 310.CrossRefGoogle ScholarPubMed
Yin, Y et al. (2014) Tumor-secreted miR-214 induces regulatory T cells: a major link between immune evasion and tumor growth. Cell Research 24, 11641180.CrossRefGoogle Scholar
Liu, F (2017) Genomic regulation of MicroRNA expression in disease development. Methods in Molecular Biology 1617, 159167.CrossRefGoogle ScholarPubMed
Zhao, JL and Starczynowski, DT (2014) Role of microRNA-146a in normal and malignant hematopoietic stem cell function. Frontiers in Genetics 5, 219.CrossRefGoogle ScholarPubMed
He, L et al. (2007) A microRNA component of the p53 tumour suppressor network. Nature 447, 11301134.CrossRefGoogle ScholarPubMed
Kent, OA, Fox-Talbot, K and Halushka, MK (2013) RREB1 repressed miR-143/145 modulates KRAS signaling through downregulation of multiple targets. Oncogene 32, 25762585.CrossRefGoogle ScholarPubMed
Zeinali, T et al. (2019) Regulatory mechanisms of miR-145 expression and the importance of its function in cancer metastasis. Biomedicine & Pharmacotherapy 109, 195207.CrossRefGoogle ScholarPubMed
Gan, B et al. (2010) FoxOs enforce a progression checkpoint to constrain mTORC1-activated renal tumorigenesis. Cancer Cell 18, 472484.CrossRefGoogle ScholarPubMed
Chang, TC et al. (2008) Widespread microRNA repression by Myc contributes to tumorigenesis. Nature Genetics 40, 4350.CrossRefGoogle ScholarPubMed
Molenaar, JJ et al. (2012) LIN28B induces neuroblastoma and enhances MYCN levels via let-7 suppression. Nature Genetics 44, 11991206.CrossRefGoogle ScholarPubMed
Zhang, X et al. (2012) Coordinated silencing of MYC-mediated miR-29 by HDAC3 and EZH2 as a therapeutic target of histone modification in aggressive B-cell lymphomas. Cancer Cell 22, 506523.CrossRefGoogle ScholarPubMed
Guan, T et al. (2018) ZEB1, ZEB2, and the miR-200 family form a counterregulatory network to regulate CD8. Journal of Experimental Medicine 215, 11531168.CrossRefGoogle Scholar
Chen, P et al. (2017) MiR-200c is a cMyc-activated miRNA that promotes nasopharyngeal carcinoma by downregulating PTEN. Oncotarget 8, 52065218.CrossRefGoogle ScholarPubMed
Di Leva, G et al. (2010) MicroRNA cluster 221–222 and estrogen receptor alpha interactions in breast cancer. Journal of the National Cancer Institute 102, 706721.CrossRefGoogle ScholarPubMed
Coarfa, C et al. (2016) Comprehensive proteomic profiling identifies the androgen receptor axis and other signaling pathways as targets of microRNAs suppressed in metastatic prostate cancer. Oncogene 35, 23452356.CrossRefGoogle ScholarPubMed
Davalos, V et al. (2012) Dynamic epigenetic regulation of the microRNA-200 family mediates epithelial and mesenchymal transitions in human tumorigenesis. Oncogene 31, 20622074.CrossRefGoogle ScholarPubMed
Ceppi, P et al. (2010) Loss of miR-200c expression induces an aggressive, invasive, and chemoresistant phenotype in non-small cell lung cancer. Molecular Cancer Research 8, 12071216.CrossRefGoogle ScholarPubMed
Schmid, G et al. (2016) Expression and promotor hypermethylation of miR-34a in the various histological subtypes of ovarian cancer. BMC Cancer 16, 102.CrossRefGoogle ScholarPubMed
Cheng, N, Li, Y and Han, ZG (2013) Argonaute2 promotes tumor metastasis by way of up-regulating focal adhesion kinase expression in hepatocellular carcinoma. Hepatology 57, 19061918.CrossRefGoogle ScholarPubMed
Lee, J et al. (2020) Identifying metastasis-initiating miRNA-target regulations of colorectal cancer from expressional changes in primary tumors. Scientific Reports 10, 14919.CrossRefGoogle ScholarPubMed
Martinez-Gutierrez, AD et al. (2019) miRNA profile obtained by next-generation sequencing in metastatic breast cancer patients is able to predict the response to systemic treatments. International Journal of Molecular Medicine 44, 12671280.Google ScholarPubMed
McGuire, A, Brown, JA and Kerin, MJ (2015) Metastatic breast cancer: the potential of miRNA for diagnosis and treatment monitoring. Cancer and Metastasis Reviews 34, 145155.CrossRefGoogle ScholarPubMed
Zografos, E et al. (2019) Prognostic role of microRNAs in breast cancer: a systematic review. Oncotarget 10, 71567178.CrossRefGoogle ScholarPubMed
Sasaki, R, Osaki, M and Okada, F (2019) MicroRNA-based diagnosis and treatment of metastatic human osteosarcoma. Cancers (Basel) 11, 553–567CrossRefGoogle ScholarPubMed
Gajos-Michniewicz, A and Czyz, M (2019) Role of miRNAs in melanoma metastasis. Cancers (Basel) 11, 326–349CrossRefGoogle ScholarPubMed
Santos, JMO et al. (2018) The role of MicroRNAs in the metastatic process of high-risk HPV-induced cancers. Cancers (Basel) 10, 493–508CrossRefGoogle ScholarPubMed
Wu, SG et al. (2019) MicroRNA in lung cancer metastasis. Cancers (Basel) 11, 553–567CrossRefGoogle ScholarPubMed
Zhu, Z et al. (2020) Identifying the key genes and microRNAs in prostate cancer bone metastasis by bioinformatics analysis. FEBS Open Bio 10, 674688.CrossRefGoogle ScholarPubMed
Song, Z et al. (2018) Elementary screening of lymph node metastatic-related genes in gastric cancer based on the co-expression network of messenger RNA, microRNA and long non-coding RNA. Brazilian Journal of Medical and Biological Research 51, e6685.CrossRefGoogle ScholarPubMed
Rupaimoole, R et al. (2016) miRNA deregulation in cancer cells and the tumor microenvironment. Cancer Discovery 6, 235246.CrossRefGoogle ScholarPubMed
Bartel, DP (2009) MicroRNAs: target recognition and regulatory functions. Cell 136, 215233.CrossRefGoogle ScholarPubMed
Raue, R et al. (2021) Therapeutic targeting of MicroRNAs in the tumor microenvironment. International Journal of Molecular Sciences 22, 2210–2247CrossRefGoogle ScholarPubMed
Dhungel, B, Ramlogan-Steel, CA and Steel, JC (2018) MicroRNA-regulated gene delivery systems for research and therapeutic purposes. Molecules 23.CrossRefGoogle ScholarPubMed
Hosseinahli, N et al. (2018) Treating cancer with microRNA replacement therapy: a literature review. Journal of Cellular Physiology 233, 55745588.CrossRefGoogle ScholarPubMed
van Rooij, E and Kauppinen, S (2014) Development of microRNA therapeutics is coming of age. EMBO Molecular Medicine 6, 851864.CrossRefGoogle ScholarPubMed
Zhang, M et al. (2019) miR-149-3p reverses CD8(+) T-cell exhaustion by reducing inhibitory receptors and promoting cytokine secretion in breast cancer cells. Open Biology 9, 190061.CrossRefGoogle ScholarPubMed
Flamini, V, Jiang, WG and Cui, Y (2017) Therapeutic role of MiR-140-5p for the treatment of non-small cell lung cancer. Anticancer Research 37, 43194327.Google ScholarPubMed
Liu, SH et al. (2021) Systematic identification of clinically relevant miRNAs for potential miRNA-based therapy in lung adenocarcinoma. Molecular Therapy. Nucleic Acids 25, 110.CrossRefGoogle ScholarPubMed
Cortez, MA et al. (2015) In vivo delivery of miR-34a sensitizes lung tumors to radiation through RAD51 regulation. Molecular Therapy. Nucleic Acids 4, e270.CrossRefGoogle ScholarPubMed
Nguyen, DD and Chang, S (2017) Development of novel therapeutic agents by inhibition of oncogenic MicroRNAs. International Journal of Molecular Sciences 19.CrossRefGoogle ScholarPubMed
Rupaimoole, R and Slack, FJ (2017) MicroRNA therapeutics: towards a new era for the management of cancer and other diseases. Nature Reviews. Drug Discovery 16, 203222.CrossRefGoogle ScholarPubMed
Ge, JH et al. (2019) An antisense oligonucleotide drug targeting miR-21 induces H1650 apoptosis and caspase activation. Technology in Cancer Research & Treatment 18, 1533033819892263.CrossRefGoogle ScholarPubMed
Xie, Y et al. (2020) Stromal modulation and treatment of metastatic pancreatic cancer with local intraperitoneal triple miRNA/siRNA nanotherapy. ACS Nano 14, 255271.CrossRefGoogle ScholarPubMed
Tay, FC et al. (2015) Using artificial microRNA sponges to achieve microRNA loss-of-function in cancer cells. Advanced Drug Delivery Reviews 81, 117127.CrossRefGoogle ScholarPubMed
Zhang, T et al. (2016) Downregulation of miR-522 suppresses proliferation and metastasis of non-small cell lung cancer cells by directly targeting DENN/MADD domain containing 2D. Scientific Reports 6, 19346.CrossRefGoogle ScholarPubMed
Nedaeinia, R et al. (2016) Locked nucleic acid anti-miR-21 inhibits cell growth and invasive behaviors of a colorectal adenocarcinoma cell line: LNA-anti-miR as a novel approach. Cancer Gene Therapy 23, 246253.CrossRefGoogle ScholarPubMed
Meng, L et al. (2017) Small RNA zippers lock miRNA molecules and block miRNA function in mammalian cells. Nature Communications 8, 13964.CrossRefGoogle ScholarPubMed
Bajan, S and Hutvagner, G (2020) RNA-Based Therapeutics: from antisense oligonucleotides to miRNAs. Cells 9.CrossRefGoogle ScholarPubMed
Rinaldi, C and Wood, MJA (2018) Antisense oligonucleotides: the next frontier for treatment of neurological disorders. Nature Reviews. Neurology 14, 921.CrossRefGoogle ScholarPubMed
Tao, YJ et al. (2015) Antisense oligonucleotides against microRNA-21 reduced the proliferation and migration of human colon carcinoma cells. Cancer Cell International 15, 77.CrossRefGoogle ScholarPubMed
Devulapally, R et al. (2015) Polymer nanoparticles mediated codelivery of antimiR-10b and antimiR-21 for achieving triple negative breast cancer therapy. ACS Nano 9, 22902302.CrossRefGoogle ScholarPubMed
Ebert, MS and Sharp, PA (2010) MicroRNA sponges: progress and possibilities. RNA 16, 20432050.CrossRefGoogle ScholarPubMed
Liang, AL et al. (2016) MiRNA-10b sponge: an anti-breast cancer study in vitro. Oncology Reports 35, 19501958.CrossRefGoogle ScholarPubMed
Jung, J et al. (2015) Simultaneous inhibition of multiple oncogenic miRNAs by a multi-potent microRNA sponge. Oncotarget 6, 2037020387.CrossRefGoogle ScholarPubMed
Lima, JF et al. (2018) Targeting miR-9 in gastric cancer cells using locked nucleic acid oligonucleotides. BMC Molecular Biology 19, 6.CrossRefGoogle ScholarPubMed
Mody, HR et al. (2016) Inhibition of S-adenosylmethionine-dependent methyltransferase attenuates TGFβ1-induced EMT and metastasis in pancreatic cancer: putative roles of miR-663a and miR-4787-5p. Molecular Cancer Research 14, 11241135.CrossRefGoogle ScholarPubMed
Kopp, F et al. (2013) De-targeting by miR-143 decreases unwanted transgene expression in non-tumorigenic cells. Gene Therapy 20, 11041109.CrossRefGoogle ScholarPubMed
Baertsch, MA et al. (2014) MicroRNA-mediated multi-tissue detargeting of oncolytic measles virus. Cancer Gene Therapy 21, 373380.CrossRefGoogle ScholarPubMed
Bofill-De Ros, X, Gironella, M and Fillat, C (2014) miR-148a- and miR-216a-regulated oncolytic adenoviruses targeting pancreatic tumors attenuate tissue damage without perturbation of miRNA activity. Molecular Therapy 22, 16651677.CrossRefGoogle ScholarPubMed
Dhungel, B et al. (2018) miRNA122a regulation of gene therapy vectors targeting hepatocellular cancer stem cells. Oncotarget 9, 2357723588.CrossRefGoogle ScholarPubMed
Dhungel, B et al. (2018) MicroRNA199a-based post-transcriptional detargeting of gene vectors for hepatocellular carcinoma. Molecular Therapy. Nucleic Acids 13, 7888.CrossRefGoogle ScholarPubMed
Dhungel, B, Ramlogan-Steel, CA and Steel, JC (2018) Synergistic and independent action of endogenous microRNAs 122a and 199a for post-transcriptional liver detargeting of gene vectors. Scientific Reports 8, 15539.CrossRefGoogle ScholarPubMed
Fortunato, O and Iorio, MV (2020) The therapeutic potential of MicroRNAs in cancer: illusion or opportunity? Pharmaceuticals (Basel) 13.CrossRefGoogle ScholarPubMed
Ivkovic, C, et al. T (2017) microRNAs as cancer therapeutics: a step closer to clinical application. Cancer Letters 407, 113122.CrossRefGoogle Scholar
Reid, G et al. (2013) Restoring expression of miR-16: a novel approach to therapy for malignant pleural mesothelioma. Annals of Oncology: Official Journal of the European Society for Medical Oncology 24, 31283135.CrossRefGoogle ScholarPubMed
Reid, G et al. (2016) Clinical development of TargomiRs, a miRNA mimic-based treatment for patients with recurrent thoracic cancer. Epigenomics 8, 10791085.CrossRefGoogle ScholarPubMed
van Zandwijk, N et al. (2017) Safety and activity of microRNA-loaded minicells in patients with recurrent malignant pleural mesothelioma: a first-in-man, phase 1, open-label, dose-escalation study. The Lancet. Oncology 18, 13861396.CrossRefGoogle ScholarPubMed
Hong, DS et al. (2020) Phase 1 study of MRX34, a liposomal miR-34a mimic, in patients with advanced solid tumours. British Journal of Cancer 122, 16301637.CrossRefGoogle ScholarPubMed
Reda El Sayed, S et al. (2021) MicroRNA therapeutics in cancer: current advances and challenges. Cancers (Basel) 13.CrossRefGoogle ScholarPubMed
Kasinski, AL et al. (2015) A combinatorial microRNA therapeutics approach to suppressing non-small cell lung cancer. Oncogene 34, 35473555.CrossRefGoogle ScholarPubMed
Beg, MS et al. (2017) Phase I study of MRX34, a liposomal miR-34a mimic, administered twice weekly in patients with advanced solid tumors. Investigational New Drugs 35, 180188.CrossRefGoogle ScholarPubMed
Grossi, I et al. (2017) Biological function of MicroRNA193a-3p in health and disease. International Journal of Genomics 2017, 5913195.CrossRefGoogle ScholarPubMed
Gao, X et al. (2018) The clinical value of miR-193a-3p in non-small cell lung cancer and its potential molecular mechanism explored in silico using RNA-sequencing and microarray data. FEBS Open Bio 8, 94109.CrossRefGoogle ScholarPubMed
Yu, M et al. (2019) PTP1B markedly promotes breast cancer progression and is regulated by miR-193a-3p. FEBS Journal 286, 11361153.CrossRefGoogle ScholarPubMed
van den Bosch, MTJ et al. (2021) Transcriptome-wide analysis reveals insight into tumor suppressor functions of 1B3, a novel synthetic miR-193a-3p mimic. Molecular Therapy. Nucleic Acids 23, 11611171.CrossRefGoogle ScholarPubMed
Fan, Q et al. (2017) MiR-193a-3p is an important tumour suppressor in lung cancer and directly targets KRAS. Cellular Physiology and Biochemistry 44, 13111324.CrossRefGoogle ScholarPubMed
Liang, H et al. (2015) miR-193a-3p functions as a tumor suppressor in lung cancer by down-regulating ERBB4. Journal of Biological Chemistry 290, 926940.CrossRefGoogle ScholarPubMed
Yu, T et al. (2015) MicroRNA-193a-3p and -5p suppress the metastasis of human non-small-cell lung cancer by downregulating the ERBB4/PIK3R3/mTOR/S6K2 signaling pathway. Oncogene 34, 413423.CrossRefGoogle ScholarPubMed
Lv, L et al. (2014) The DNA methylation-regulated miR-193a-3p dictates the multi-chemoresistance of bladder cancer via repression of SRSF2/PLAU/HIC2 expression. Cell death & disease 5, e1402.CrossRefGoogle ScholarPubMed
Kwon, JE et al. (2013) Ionizing radiation-inducible microRNA miR-193a-3p induces apoptosis by directly targeting Mcl-1. Apoptosis 18, 896909.CrossRefGoogle ScholarPubMed
Liu, Y et al. (2017) MicroRNA-193a-3p inhibits cell proliferation in prostate cancer by targeting cyclin D1. Oncology Letters 14, 51215128.Google ScholarPubMed
Pu, Y et al. (2016) MiR-193a-3p and miR-193a-5p suppress the metastasis of human osteosarcoma cells by down-regulating Rab27B and SRR, respectively. Clinical & Experimental Metastasis 33, 359372.CrossRefGoogle ScholarPubMed
Bader, AG (2012) miR-34 - a microRNA replacement therapy is headed to the clinic. Frontiers in Genetics 3, 120.CrossRefGoogle ScholarPubMed
Telford, BJ et al. (2021) Multi-modal effects of 1B3, a novel synthetic miR-193a-3p mimic, support strong potential for therapeutic intervention in oncology. Oncotarget 12, 422439.CrossRefGoogle ScholarPubMed
Garofalo, M et al. (2021) Correction: MicroRNA signatures of TRAIL resistance in human non-small cell lung cancer. Oncogene 40, 1204.CrossRefGoogle ScholarPubMed
Miller, TE et al. (2008) MicroRNA-221/222 confers tamoxifen resistance in breast cancer by targeting p27Kip1. Journal of Biological Chemistry 283, 2989729903.CrossRefGoogle ScholarPubMed
Callegari, E et al. (2012) Liver tumorigenicity promoted by microRNA-221 in a mouse transgenic model. Hepatology 56, 10251033.CrossRefGoogle Scholar
Zhang, CZ et al. (2010) MiR-221 and miR-222 target PUMA to induce cell survival in glioblastoma. Molecular Cancer 9, 229.CrossRefGoogle ScholarPubMed
Di Martino, MT et al. (2013) In vitro and in vivo anti-tumor activity of miR-221/222 inhibitors in multiple myeloma. Oncotarget 4, 242255.CrossRefGoogle ScholarPubMed
Higgs, G and Slack, F (2013) The multiple roles of microRNA-155 in oncogenesis. Journal of Clinical Bioinformatics 3, 17.CrossRefGoogle ScholarPubMed
Seto, AG et al. (2018) Cobomarsen, an oligonucleotide inhibitor of miR-155, co-ordinately regulates multiple survival pathways to reduce cellular proliferation and survival in cutaneous T-cell lymphoma. British Journal of Haematology 183, 428444.CrossRefGoogle ScholarPubMed
Zhang, Y et al. (2012) LNA-mediated anti-miR-155 silencing in low-grade B-cell lymphomas. Blood 120, 16781686.CrossRefGoogle ScholarPubMed
Cheng, CJ et al. (2015) MicroRNA silencing for cancer therapy targeted to the tumour microenvironment. Nature 518, 107110.CrossRefGoogle ScholarPubMed
Yu, HR, Huang, LH and Li, SC (2018) Roles of microRNA in the immature immune system of neonates. Cancer Letters 433, 99106.CrossRefGoogle ScholarPubMed
Bulcha, JT et al. (2021) Viral vector platforms within the gene therapy landscape. Signal Transduction and Targeted Therapy 6, 53.CrossRefGoogle ScholarPubMed
Santana-Armas, ML and Tros de Ilarduya, C (2021) Strategies for cancer gene-delivery improvement by non-viral vectors. International Journal of Pharmaceutics 596, 120291.CrossRefGoogle ScholarPubMed
Lundstrom, K (2018) Viral vectors in gene therapy. Diseases (basel, Switzerland) 6.Google ScholarPubMed
Karlsen, TA and Brinchmann, JE (2013) Liposome delivery of microRNA-145 to mesenchymal stem cells leads to immunological off-target effects mediated by RIG-I. Molecular Therapy 21, 11691181.CrossRefGoogle ScholarPubMed
Biscans, A et al. (2019) Diverse lipid conjugates for functional extra-hepatic siRNA delivery in vivo. Nucleic Acids Research 47, 10821096.CrossRefGoogle ScholarPubMed
Li, X et al. (2019) Challenges and opportunities in exosome research-perspectives from biology, engineering, and cancer therapy. APL Bioengineering 3, 011503.CrossRefGoogle ScholarPubMed
Bandi, N and Vassella, E (2011) miR-34a and miR-15a/16 are co-regulated in non-small cell lung cancer and control cell cycle progression in a synergistic and Rb-dependent manner. Molecular Cancer 10, 55.CrossRefGoogle Scholar
Kara, G, Calin, GA and Ozpolat, B (2022) RNAi-based therapeutics and tumor targeted delivery in cancer. Advanced Drug Delivery Reviews 182, 114113.CrossRefGoogle ScholarPubMed
Seok, H et al. (2018) Evaluation and control of miRNA-like off-target repression for RNA interference. Cellular and Molecular Life Sciences 75, 797814.CrossRefGoogle ScholarPubMed
Pepe, F et al. (2022) A large fraction of trisomy 12, 17p(-), and 11q(-) CLL cases carry unidentified microdeletions of miR-15a/16-1. Proceedings of the National Academy of Sciences of the USA 119.CrossRefGoogle ScholarPubMed
Segal, M and Slack, FJ (2020) Challenges identifying efficacious miRNA therapeutics for cancer. Expert Opinion on Drug Discovery 15, 987992.CrossRefGoogle ScholarPubMed
Dhungel, B et al. (2018) Evaluation of the Glypican 3 promoter for transcriptional targeting of hepatocellular carcinoma. Gene Therapy 25, 115128.CrossRefGoogle ScholarPubMed
Ceppi, M et al. (2009) MicroRNA-155 modulates the interleukin-1 signaling pathway in activated human monocyte-derived dendritic cells. Proceedings of the National Academy of Sciences of the USA 106, 27352740.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1. Biogenesis and mechanism of action of miRNAs: RNA polymerase II-mediated transcription forms the primary miRNA (pri-miRNA) which is cleaved by an RNase III enzyme (DROSHA) to produce a precursor miRNA (pre-miRNA) in the canonical pathway of miRNA biogenesis. The pre-miRNA is exported to the cytoplasm via exportin 5, for further processing by RNase III DICER to form a mature miRNA duplex. Non-canonical pathways are independent of Drosha or Dicer processing. The miRNA duplex is then unwound whereby the guide strand along with Argonaute (AGO) proteins form a miRNA-induced silencing complex (RISC). The RISC complex binds to target sequences of mRNA leading to translation repression or degradation. AGO recruits GW182 which forms a complex with CCR4-NOT making the target mRNA susceptible to cleavage by exonucleases while hindrance to the binding of eukaryotic initiation factor-4A (eIF4A) to the target mRNA leads to translational inhibition.

Figure 1

Fig. 2. MiRNAs and different stages of metastasis: Several miRNAs are dysregulated throughout different stages of the metastatic process including disruption of tight junctions, epithelial to mesenchymal transition (EMT), migration and invasion, angiogenesis, stemness and metastatic growth and tumour secondary microenvironment.

Figure 2

Table 1. A list of microRNAs involved at different stages of cancer metastasis

Figure 3

Table 2. MiRNA-based therapies for cancer metastasis: Dysregulated miRNAs are potential therapeutic targets to treat metastatic cancer

Figure 4

Fig. 3. MiRNA-based therapies. (a) MiRNA replacement with mimics function like an overexpression of endogenous miRNA and increase the degradation or repression of target mRNAs. (b) The miRNA inhibitor approach minimises the binding of miRNA-induced silencing complex (miRISC) to target mRNAs. Different strategies used for miRNA inhibition includes antisense oligonucleotides (ASOs), antagomir antisense oligonucleotides, locked nucleic acid (LNA), antisense oligonucleotide and small RNA zippers. (c) MiRNA sponge binds to the miRISC complex reducing its binding to the target mRNA. (d) MiRNA mask prevents the miRISC from binding to the mRNA by ‘masking’ the miRNA binding site.

Figure 5

Table 3. A list of cancer therapy clinical trials utilising miRNA-based strategies