Abstract
Over the past few decades, siRNA and miRNA have attracted a great deal of attention from researchers and clinicians. These molecules have been extensively studied from the standpoint of developing biopharmaceuticals against various diseases, including heart disease, diabetes and cancers. siRNA suppresses only a single target, whereas each miRNA regulates the expression of multiple target genes. More importantly, because miRNA are also secreted from cancer cells, and their aberrant expression is associated with tumor development and progression, they represent not only therapeutic targets but also promising biomarkers for diagnosis and prognosis. Therefore, miRNA may be more effective tools against cancers, in which multiple signal pathways are dysregulated. In this review, we summarize recent progress in the development of miRNA therapeutics for the treatment of cancer patients, and describe delivery systems for oligonucleotide therapeutics.
Keywords: cancer biology, delivery system, exosomes, miRNA, therapeutics
Abbreviations
- 3′UTR
3′‐untranslated region
- BCR‐ABL
breakpoint cluster region‐Abelson murine leukemia viral oncogene homolog 1
- CDS
coding sequences
- ce‐miRNA
cellular miRNA
- CML
chronic myelogenous leukemia
- CSC
cancer stem cells
- cssDNA
circular ssDNA
- EMT
epithelial‐to‐mesenchymal transition
- ENPP1
ectonucleotide pyrophosphatase/phosphodiesterase 1
- EV
extracellular vesicle
- EV
extracellular vesicles
- FDA
US Food and Drug Administration
- HCC
hepatocellular carcinoma
- HCV
hepatitis C virus
- HCV
hepatitis C virus
- HE
hepatic encephalopathy
- LNA
locked nucleic acid
- LSC
leukemia stem cells
- nt
nucleotides
- onco‐miRNA
oncogenic miRNA
- PD‐1
programmed death 1
- PD‐L1
programmed death‐ligand 1
- SCD1
stearoyl desaturase 1
- se‐miRNA
secreted miRNA
- SPRED1
Sprouty‐related EVH1‐domain‐containing 1
- TAM
tumor‐associated macrophages
- TGF
transforming growth factor
- TKI
tyrosine kinase inhibitors
- Treg
regulatory T cells
- TS‐miRNA
tumor suppressive miRNA
- ZO‐1
zonula occludens‐1
1. INTRODUCTION
Several lines of evidence indicate that ncRNA regulate multiple stages of life cycle, including development, differentiation and aging, through the regulation of target gene expression.1, 2, 3 Broadly, ncRNA can be classified into two groups: small ncRNA 18‐200 nt in length and lncRNA.4
miRNA is a type of small ncRNA. The primary function of miRNA is negative regulation of their target genes at the post‐transcriptional level. Specifically, miRNA induce transcript degradation or inhibition of protein translation through sequence‐specific binding with the 3′UTR of their target mRNA (Figure 1). miRNA also inhibit the translation or facilitate the cleavage of their target mRNA by binding to their CDS.5, 6, 7 To date, multiple miRNA have been identified in various types of cancers, and these tumor‐associated miRNA can be classified into two groups: TS‐miRNA and onco‐miRNA.8, 9, 10, 11
Figure 1.

Biogenesis of miRNA. Most miRNA are transcribed by RNA polymerase II (RNA pol II) as primary‐miRNA (pri‐miRNA), and then processed in the nucleus by Drosha‐DGCR8 into precursor miRNA (pre‐miRNA). The pre‐miRNA is exported to the cytoplasm by exportin‐5 and further cleaved by a complex containing Dicer and TRBP. The functional strand of mature miRNA is incorporated into the RNA‐induced silencing complex (RISC), which contains GW182 and Argonaute protein. As a component of this complex, the mature miRNA regulates gene expression by binding to complementary sequences in the 3′ untranslated region (UTR) or coding regions of its target mRNA, leading to mRNA degradation or translational repression. Alternatively, miRNA can induce translational activation by the 5′UTR of target mRNA. In addition, miRNA can be secreted through the exosomal pathway and regulate gene expression in recipient cells
Because miRNA are considerably smaller than proteins, they can be introduced to cells by the same techniques used for siRNA.12, 13, 14 Consequently, the emergence of the miRNA field prompted many cancer researchers and clinicians to develop miRNA delivery strategies for the treatment of cancer patients. In this review, we summarize the major findings regarding miRNA function in tumor development, and describe recent advances in the preclinical and clinical development of miRNA delivery systems for cancer therapeutics.
2. ROLES OF miRNA IN CANCER BIOLOGY
The first two miRNA to be reported, lin‐4 and let‐7, were discovered in the nematode Caenorhabditis elegans in 1993 and 2000, respectively.15, 16, 17 Because let‐7 is conserved across animal species, from flies to mammals, and plays important roles in the various biological processes, including cancer biology,18 it has been extensively studied. Subsequently, multiple studies have reported the roles of miRNA in cancer biology, including tumor initiation, drug resistance and metastasis.
miRNA are also important for cell‐to‐cell communications involved in the inflammatory response, differentiation and tumor progression.19, 20, 21 miRNA are secreted from various types of cells through EV containing membrane proteins such as CD63, CD81 and CD9.22, 23, 24 In the next section, we introduce the functions of TS‐miRNA and onco‐miRNA in relation to malignant cancer phenotypes such as tumor initiation, EMT, drug resistance and metastasis (Table 1).
Table 1.
Classification of miRNA according to their functions
| Phenotype | Cancer | miRNA | Target | References |
|---|---|---|---|---|
| Tumorigenicity | Breast | Let‐7 | H‐RAS and HMGA2 | 35 |
| Breast | miR‐600 | SCD1 | 36 | |
| CML | Ex‐miR‐126 | SPRED1 | 38 | |
| Drug resistance | Breast | miR‐27b | ENPP1 | 9 |
| Liver | miR‐221 | Caspase‐3 | 39 | |
| CML | miR‐377 | BCL‐XL | 40 | |
| EMT | Breast | miR‐200 family and miR‐205 | ZEB1 and ZEB2 | 50 |
| miR‐22 | TET family | 51 | ||
| Metastasis | Breast | miR‐29b | VEGFA, ANGPTL4, PDGF, LOX and MMP9 | 61 |
| Colon | 135b | FIH‐1 | 62 | |
| Breast | Ex‐miR‐105 | ZO‐1 | 64 | |
| Breast | Ex‐miR‐122 | PKM‐2 | 65 |
CML, chronic myelogenous leukemia; EMT, epithelial‐to‐mesenchymal transition; Ex‐miRNA, exosomal miRNA.
Each miRNA is classified according to its role in cancer biology.
3. TUMORIGENICITY
Over the past decade, CSC have been identified in many common cancer types, ranging from leukemia to solid tumors.25‐33 The main properties of CSC, including tumor‐seeding ability and therapy resistance, have been evaluated in immunodeficient mice. Specifically, CSC were isolated by cell sorting for specific markers and then transplanted into immunodeficient mice, where their tumor‐seeding abilities were examined.34
Yu et al35 initially identified let‐7 as a master regulator of the self‐renewal and tumor‐seeding ability of breast cancer cells. They observed a high percentage of CSC with the CD44(high)/CD24(‐/low) antigen phenotype in tumors isolated from the patients who received chemotherapy. In addition, they showed that CSC exhibited an elevated ability to form mammospheres in vitro and tumors in NOD/SCID mice. Importantly, breast CSC‐enriched cells expressed much lower levels of let‐7 than parental cells or in vitro differentiated progeny with the CD44(low/‐)/CD24(high) antigen phenotype.
Recently, El Helou et al36 reported that downregulation of miR‐600 is significantly associated with poor prognosis in breast cancer patients. miR‐600 inhibits WNT signaling, which plays an important role in CSC renewal by directly suppressing SCD1, an enzyme required for the production of active, lipid‐modified WNT proteins. Thus, miR‐600 inhibits CSC renewal, resulting in suppression of tumor‐seeding ability. By contrast, miR‐31 promotes the tumor‐seeding ability of breast cancer cells through activating WNT signaling.37
Exosomal miRNA is also important for regulation of CSC properties. Zhang et al38 showed that in CML, miR‐126 supports the quiescence, self‐renewal and engraftment capacity of LSC. Using a mouse model, they demonstrated that endothelial cells in the bone marrow support the CSC properties of LSC by supplying exosomal miR‐126 to those cells. They also found that a feedback loop between miR‐126 and its target SPRED1 inhibits miRNA maturation. Because SPRED1 is a substrate of BCR‐ABL, TKI targeting BCR‐ABL inhibited the phosphorylation of SPRED1, resulting in an increase in miR‐126. On the basis of these findings, they proposed that the combination of TKI and miRNA inhibitors may prevent recurrence in CML patients.
4. DRUG RESISTANCE
Resistance to chemotherapy is a major obstacle for cancer therapy, and is frequently observed in patients with cancer recurrence. The mechanisms underlying drug resistance can be classified into two main groups: (a) upregulation of drug‐transporters and anti‐apoptotic genes; and (b) activation of survival pathways. Several studies demonstrate that miRNA are also involved in regulating these drug‐resistance pathways.
In breast cancer, miR‐27b regulates resistance to docetaxel, an anti‐microtubule agent, through its direct target ENPP1.9 ENPP1 promotes the cell surface localization of ABCG2, which mediates the efflux of several types of small compounds, resulting in acquisition of docetaxel resistance. Therefore, docetaxel in combination with a miR‐27b mimic effectively inhibits tumor growth in an animal model of breast cancer.
In HCC, miR‐221 induces sorafenib resistance through direct inhibition of caspase 3.39 More importantly, the expression level of circulating miR‐221 in serum is significantly associated with sorafenib resistance in HCC patients. In B‐cell lymphoma, miR‐377 promotes resistance to ABT199 (venetoclax) by directly targeting B‐cell lymphoma‐extra large 37.40 Because venetoclax is a promising small compound for treatment of multiple myeloma and chronic lymphocytic leukemia, it is likely that combination treatment with venetoclax and a miR‐377 inhibitor could improve patient outcomes.
In ovarian cancer, elevated expression of nc886, a non‐coding RNA induced by TGF‐β treatment is associated with a poor prognosis.41 nc886 inhibits miRNA maturation through the physical interaction with Dicer, resulting in acquisition of invasive ability and drug resistance.
Several groups also succeeded in the selective inhibition of miRNA processing by targeting the Drosha processing site of miRNA.42, 43 Childs‐Disney et al43 reported that neomycin B and its derivative 5′‐azido neomycin B bind the Drosha processing site in the miR‐525 precursor. Neomycin B is an FDA‐approved drug and used for the prevention of HE and bacterial infections caused by cirrhosis. Because upregulation of miR‐525 is observed in approximately 60% of liver cancer tissues and induces the migration and invasion of liver cancer cells by directly targeting ZNF395,44 neomycin B or its derivative may be a promising drug for the treatment of liver cancer patients. Velagapudi et al42 also succeeded in the synthesis of a small compound that binds to the Drosha processing site in the miR‐96 precursor. miR‐96 is reported to be an oncogenic miRNA in breast cancer cells.45 Consistent with this, a small compound directed against miR‐96 efficiently inhibits the growth of breast cancer cells in vitro and in vivo.
5. EPITHELIAL‐TO‐MESENCHYMAL TRANSITION
The EMT, a well‐studied process in which epithelial cells trans‐differentiate into motile mesenchymal cells, is integral for stem cell behavior and development, and it is also observed in the context of malignant tumor behaviors such as drug resistance and metastasis.46, 47 Two recent studies showed that EMT is not necessarily required for metastasis in mouse tumor models,48, 49 arguing that EMT is mainly associated with resistance to chemotherapy. Although the role of EMT in metastasis is not fully elucidated, these studies suggest that EMT inhibition may improve resistance to conventional chemotherapy.
Gregory et al50 reported that the miR‐200 family (miR‐200a, miR‐200b, miR‐200c, miR‐141 and miR‐429) and miR‐205 inhibit the EMT phenotype by directly targeting ZEB1 and ZEB2, which are transcription repressors of E‐cadherin. Subsequently, miR‐22 was also shown to influence the EMT phenotype through epigenetic regulation of the miR‐200 family.51
Transforming growth factor‐β signaling is important for EMT induction, and several miRNA have been identified as novel regulators of TGF‐β‐mediated EMT induction. For example, Subramanyam et al52 reported that miR‐302 and ‐372 promote the reprogramming of human fibroblasts into induced pluripotent stem cells through direct suppression of TGF‐β receptor type II and Ras homolog family member, both of which are associated with the EMT phenotype.53
In colon cancer, EMT and liver metastasis are induced by exosomal secretion of the miR‐200 family, which suppresses the EMT phenotype described above.50, 54 Shelton et al54 reported that the intracellular levels of miR‐200 family members are post‐transcriptionally regulated by PKCζ which is reported to act as a tumor suppressor.55 PKCζ and its substrate ADAR2 play important roles in the accumulation of intracellular miR‐200 family members in cancer cells. When PKCζ is downregulated, miRNA of this family are secreted through the exosomal pathway, resulting in EMT induction and metastasis.
6. METASTASIS
In 2008, Tavazoie et al56 first reported miR‐126 and miR‐335 as suppressors of metastasis in breast cancer. Subsequently, multiple miRNA were identified as metastasis‐associated miRNA.57, 58, 59, 60 For example, in breast cancer, Chou et al61 reported that GATA3 suppresses tumor metastasis by upregulating miR‐29b. GATA3 is a transcription factor involved in maintenance of the luminal epithelial cell phenotype, and its expression is downregulated or lost in cancer patients. miR‐29b directly suppresses expression of pro‐metastatic regulators such as VEGFA, ANGPTL4, PDGF, LOX and MMP9. Therefore, GATA3 functions as a tumor suppressor through miR‐29b‐mediated suppression of pro‐metastatic niche. In colon cancer, Valeri et al62 identified miR‐135b as an oncogenic miRNA in colon cancer. Aberrant expression of miR‐135b is associated with mutation or inactivation of APC. In addition, miR‐135b directly targets factor inhibiting HIF1α, which functions as a tumor suppressor by repressing the HIF1α pathway.63 Therefore, elevated expression of miR‐135b promotes colon cancer metastasis by activating the HIF1α pathway.
In addition to the intracellular miRNA, secreted miRNA are also involved in the regulation of tumor environment and metastasis.64, 65, 66, 67 Zhou et al reported that miR‐105 is expressed and secreted specifically by metastatic breast cancer cells, but not in less invasive cells. Exosomal miR‐105 destroys the tight junctions by directly targeting ZO‐1, resulting in tumor invasiveness and elevated vascular permeability in distant organs.64 Interestingly, secretion of exosomal miR‐105 is induced by the oncoprotein MYC.68 Moreover, exosomal miR‐105 from breast cancer cells activates MYC signaling in cancer‐associated fibroblasts by directly targeting MAX‐interacting protein 1, a repressor of the c‐MYC promoter,69 resulting in c‐MYC‐mediated upregulation of glucose and glutamine metabolism and increased growth of neighboring cancer cells.
Fong et al65 reported that miR‐122 also promotes breast cancer metastasis by activating glucose metabolism in pre‐metastatic sites. In contrast to the intracellular level of miR‐122, which does not significantly differ between non‐metastatic and metastatic cancer cells, the amount of exosomal miR‐122 is associated with the metastatic capacity of breast cancer cells. Exosomal miR‐122 promotes glucose metabolism by directly targeting pyruvate kinase isozymes M2 in pre‐metastatic sites, resulting in formation of a pre‐metastatic niche.
7. THERAPEUTIC APPROACHES BASED ON miRNA REGULATION
As described above, miRNA play important roles in tumor biology and have low toxicity, as exhibited by their endogenous expression in human tissues. Thus, investigations are in progress to develop onco‐miRNA antagonizing or TS‐miRNA replacement systems (Figure 2). The motivation for attempting to modulate miRNA expression in disease tissues is based on the concept that TS‐miRNA are more highly expressed or functional in normal tissues than in tumor tissues, whereas onco‐miRNA are upregulated and activated mainly in tumor tissues. Using LNA‐modified oligonucleotides, a class of high‐affinity bicyclic RNA analogs, miRNA can be easily detected in these tissues, and their functions can be inhibited in in vitro and in vivo studies.70, 71, 72 In addition to LNA‐mediated suppression of miRNA, TS‐miRNA replacement has been attempted using miRNA mimics, viral vectors expressing miRNA, and small compounds that regulate the endogenous expression of miRNA.14, 73, 74
Figure 2.

Therapeutic approaches that target miRNA. Therapeutic approaches targeting miRNA expression are based on the observation that tumor suppressive miRNA (TS‐miRNA) are lost or downregulated in tumor tissues, whereas onco‐miRNA are upregulated and activated. onco‐miRNA can be inhibited by introduction of locked nucleic acid LNA), decoy vectors or sponge vectors. For replacement of TS‐miRNA, miRNA mimics are introduced by viral vectors or nanoparticles, or upregulated by small compounds
Because miR‐199a is the third most highly expressed miRNA in normal liver and its downregulation correlates with poor prognosis,75 Callegari et al76 investigated the tumor suppressive effects of miR‐199a using a transgenic mouse model of HCC. For their in vivo delivery experiments, they used lipid nanoparticles that were composed of 1,2‐dioleoyl‐sn‐glycero‐3‐phosphoethanolamine, 1,2‐dimyristoyl‐sn‐glycerol, methoxypolyethylene glycol, and linoleic acid at a ratio of 50:48:2, and demonstrated that miR‐199a exerted anti‐tumor activity against mouse HCC.
Expression of miR‐21 is elevated in human glioma cells, and LNA against miR‐21 can efficiently inhibit tumor growth in a xenograft‐model of human glioblastoma.77, 78 MRG‐106, an inhibitor of miR‐155, has been studied in phase I clinical trials (http://www.miragen.com). To date, MRG‐106 has mainly been used to treat blood cancer such as T‐cell lymphoma, diffuse large B‐cell lymphoma and chronic lymphocytic leukemia. In addition to cancer, LNA‐mediated‐miRNA inhibition is also effective against other diseases. For example, miR‐122 is a liver‐specific miRNA that stimulates translation of HCV by interacting with the 5′UTR of the HCV genome.79, 80, 81, 82, 83 This interaction also induces “sponge effects” that repress host genes targeted by miR‐122, yielding a fertile environment for long‐term replication of HCV.84 Several studies show that LNA against miR‐122 represents a promising approach for HCV treatment in primates.13, 85 Moreover, a clinical study revealed that LNA against miR‐122 (miravirsen, SPC3649) achieved prolonged reduction in the levels of viral RNA in HCV patients.86
In addition to LNA treatment, miRNA sponges are also being used to inhibit onco‐miRNA. miRNA sponges are artificial transcripts that contain multiple tandem high‐affinity binding sites for one or more miRNA of interest.87 Meng et al88 developed an miRNA sponge using cssDNA. In several types of cancer, miR‐9 commonly inhibits expression of tumor‐suppressor genes such as KLF17, CDH1 and LASS2. A cssDNA sponge containing four miR‐9 binding sites can efficiently block miR‐9 function, restoring endogenous expression of KLF17, CDH1 and LASS2.
Several studies indicate that miRNA modulation is also a promising approach for cancer immunotherapy.89, 90 TAM and regulatory T cells are key components of the tumor microenvironment in various types of cancers.91, 92 TAM develop from monocytes and are categorized into two main functional groups: (a) tumor suppressive type (M1 phenotype); and (b) immunosuppressive type (M2 phenotype). Baer et al89 reported that let‐7 is important for acquisition of the M2 phenotype and promotes tumor‐infiltrating cytotoxic T lymphocytes. On the other hand, they also demonstrated that conditional deletion of the DICER in macrophages induces the acquisition of the M1 phenotype. Xu et al90 reported that miR‐424 regulates the PD‐L1/PD‐1 and CD80/CTLA‐4 pathways in drug‐resistant ovarian cancer. miR‐424 expression was reported to be inversely correlated with PD‐L1 and CD80 expression in ovarian cancer patients through its direct targeting of the 3′UTR of PD‐L1 and CD80. Therefore, these studies suggest that the inhibition of let‐7 or introduction of miR‐424 could represent an effective approach for improving cancer immunotherapy.
8. DELIVERY SYSTEM OF miRNA
Modification of miRNA with LNA protects the nucleotides from degradation by serum ribonucleases, thereby extending the half‐life of oligonucleotides in serum. Accordingly, multiple studies, including clinical trials, have demonstrated that LNA‐modified miRNA inhibitors are functional even in vivo (see previous section). To further develop miRNA replacement therapy or LNA‐mediated miRNA inhibition, a great deal of work has focused on improving the efficacy and accuracy of miRNA delivery systems. Two main strategies, intra‐tumor or systemic delivery, have been considered for miRNA delivery. The advantage of local delivery is that it can selectively deliver miRNA into target tissues without non‐specifically introducing miRNA into non‐tumor tissues. On the other hand, local delivery is not suitable for metastatic cancer, which is often observed in late stages of disease. To address this need, significant efforts have been made to develop systemic miRNA delivery strategies, in particular by improving targeting delivery systems and transduction efficiency.
To improve the transduction efficacy of oligonucleotides, Hossain et al93 developed the carbonate apatite‐based delivery system. Using this system, several groups succeeded in the efficient transduction of TS‐miRNA into colon cancer cells.94, 95 Hiraki et al94 identified miR‐4689 as a TS‐miRNA whose expression is downregulated by the K‐RAS mutant (KRASG12V). The expression level of miR‐4689 is also down‐regulated in KRAS mutant colon cancer patients. Their study revealed that miR‐4869 inhibits tumor cell proliferation by directly targeting KRAS and AKT. Inoue et al95 also reported that miR‐29b‐1‐5p is a TS‐miRNA and a passenger strand of miR‐29b‐3p. Compared with miR‐29b‐3p, miR‐29b‐1‐5p was significantly downregulated in K‐RAS mutant colon cancer patients. In animal experiments, miR‐29b‐1‐5p selectively inhibited the proliferation of K‐RAS mutant colon cancer cells.
For selective delivery of miRNA, Orellana et al96 developed a selective miRNA delivery system without vehicle. Specifically, they directly conjugated a folate ligand, vitamin B9, to the strong TS‐miRNA miR‐34a.12 Folate receptor is more highly expressed in epithelial cancers (breast, lung, ovary and colon) and various hematological malignancies than on normal tissues.97 To stabilize the folate‐conjugated miRNA, they also modified the passive strand of miR‐34a using 2′‐O‐methyl RNA bases. This ligand‐conjugated miRNA selectively inhibited tumor growth in vitro and in vivo without the use of vehicle. More importantly, Orellana et al confirmed the tumor‐suppressive effects of this modified miRNA using an immunocompetent mouse model that mimics aggressive human non‐small‐cell lung carcinoma.
Several studies demonstrate that exosomes are useful carriers for in vivo siRNA and miRNA delivery.98, 99 Kamerkar et al98 reported that CD47‐positive exosomes derived from normal fibroblast‐like mesenchymal cells are more effective than liposomes for in vivo delivery of siRNA against KRAS. CD47 inhibits the phagocytosis of exosomes by monocytes and macrophages. Consequently, CD47‐positive exosomes are retained in the blood longer than liposomes, resulting in improved oligonucleotide transduction efficacy. In addition, Pi et al99 reported that folate ligand‐ or aptamer‐conjugated exosomes are effective for the targeted delivery of oligonucleotides in xenograft models of prostate, breast and colorectal cancer.
9. CONCLUDING REMARKS
Here, we described recent studies of the functional roles of miRNA in cancer biology and discussed the development of clinical applications based on miRNA mimics or inhibitors. Multiple experiments in animal models demonstrate that single miRNA could suppress multiple oncogenic pathways, indicating that miRNA‐based therapeutics represent a generally effective strategy. Therefore, miRNA‐based therapeutics could be used to support the conventional therapeutics such as surgical intervention, chemotherapy and radiotherapy, thereby improving treatment for cancer patients.
CONFLICTS OF INTEREST
Authors declare no conflict of interest for this article.
ACKNOWLEDGMENTS
This work was supported in part by a Grant‐in‐Aid for Scientific Research C (16K08261) and the Japan Agency for Medical Research and Development (AMED).
Takahashi R, Prieto‐Vila M, Kohama I, Ochiya T. Development of miRNA‐based therapeutic approaches for cancer patients. Cancer Sci. 2019;110:1140–1147. 10.1111/cas.13965
REFERENCES
- 1. Chiu HS, Somvanshi S, Patel E, et al. Pan‐cancer analysis of lncRNA regulation supports their targeting of cancer genes in each tumor context. Cell Rep. 2018;23:297‐312 e12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Hu WL, Jin L, Xu A, et al. GUARDIN is a p53‐responsive long non‐coding RNA that is essential for genomic stability. Nat Cell Biol. 2018;20:492‐502. [DOI] [PubMed] [Google Scholar]
- 3. Kim HK, Fuchs G, Wang S, et al. A transfer‐RNA‐derived small RNA regulates ribosome biogenesis. Nature. 2017;552:57‐62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Ambros V. The functions of animal microRNAs. Nature. 2004;431:350‐355. [DOI] [PubMed] [Google Scholar]
- 5. Ito Y, Inoue A, Seers T, et al. Identification of targets of tumor suppressor microRNA‐34a using a reporter library system. Proc Natl Acad Sci USA. 2017;114:3927‐3932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Forman JJ, Legesse‐Miller A, Coller HA. A search for conserved sequences in coding regions reveals that the let‐7 microRNA targets Dicer within its coding sequence. Proc Natl Acad Sci USA. 2008;105:14879‐14884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Hausser J, Syed AP, Bilen B, Zavolan M. Analysis of CDS‐located miRNA target sites suggests that they can effectively inhibit translation. Genome Res. 2013;23:604‐615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Nezu Y, Hagiwara K, Yamamoto Y, et al. miR‐135b, a key regulator of malignancy, is linked to poor prognosis in human myxoid liposarcoma. Oncogene. 2016;35:6177‐6188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Takahashi RU, Miyazaki H, Takeshita F, et al. Loss of microRNA‐27b contributes to breast cancer stem cell generation by activating ENPP1. Nat Commun. 2015;6:7318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Takahashi RU, Miyazaki H, Ochiya T. The role of microRNAs in the regulation of cancer stem cells. Front Genet. 2014;4:295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Gailhouste L, Gomez‐Santos L, Hagiwara K, et al. miR‐148a plays a pivotal role in the liver by promoting the hepatospecific phenotype and suppressing the invasiveness of transformed cells. Hepatology. 2013;58:1153‐1165. [DOI] [PubMed] [Google Scholar]
- 12. Tazawa H, Tsuchiya N, Izumiya M, Nakagama H. Tumor‐suppressive miR‐34a induces senescence‐like growth arrest through modulation of the E2F pathway in human colon cancer cells. Proc Natl Acad Sci USA. 2007;104:15472‐15477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Elmen J, Lindow M, Schutz S, et al. LNA‐mediated microRNA silencing in non‐human primates. Nature. 2008;452:896‐899. [DOI] [PubMed] [Google Scholar]
- 14. Takeshita F, Patrawala L, Osaki M, et al. Systemic delivery of synthetic microRNA‐16 inhibits the growth of metastatic prostate tumors via downregulation of multiple cell‐cycle genes. Mol Ther. 2010;18:181‐187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin‐4 encodes small RNAs with antisense complementarity to lin‐14. Cell. 1993;75:843‐854. [DOI] [PubMed] [Google Scholar]
- 16. Wightman B, Ha I, Ruvkun G. Posttranscriptional regulation of the heterochronic gene lin‐14 by lin‐4 mediates temporal pattern formation in C. elegans . Cell. 1993;75:855‐862. [DOI] [PubMed] [Google Scholar]
- 17. Reinhart BJ, Slack FJ, Basson M, et al. The 21‐nucleotide let‐7 RNA regulates developmental timing in Caenorhabditis elegans . Nature. 2000;403:901‐906. [DOI] [PubMed] [Google Scholar]
- 18. Takamizawa J, Konishi H, Yanagisawa K, et al. Reduced expression of the let‐7 microRNAs in human lung cancers in association with shortened postoperative survival. Cancer Res. 2004;64:3753‐3756. [DOI] [PubMed] [Google Scholar]
- 19. Tang X, Lu H, Dooner M, Chapman S, Quesenberry PJ, Ramratnam B. Exosomal Tat protein activates latent HIV‐1 in primary, resting CD4+ T lymphocytes. JCI Insight. 2018;3:pii: 95676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Cooks T, Pateras IS, Jenkins LM, et al. Mutant p53 cancers reprogram macrophages to tumor supporting macrophages via exosomal miR‐1246. Nat Commun. 2018;9:771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Fry CS, Kirby TJ, Kosmac K, McCarthy JJ, Peterson CA. Myogenic progenitor cells control extracellular matrix production by fibroblasts during skeletal muscle hypertrophy. Cell Stem Cell. 2017;20:56‐69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Valadi H, Ekstrom K, Bossios A, Sjostrand M, Lee JJ, Lotvall JO. Exosome‐mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007;9:654‐659. [DOI] [PubMed] [Google Scholar]
- 23. Escola JM, Kleijmeer MJ, Stoorvogel W, Griffith JM, Yoshie O, Geuze HJ. Selective enrichment of tetraspan proteins on the internal vesicles of multivesicular endosomes and on exosomes secreted by human B‐lymphocytes. J Biol Chem. 1998;273:20121‐20127. [DOI] [PubMed] [Google Scholar]
- 24. Thery C, Regnault A, Garin J, et al. Molecular characterization of dendritic cell‐derived exosomes. Selective accumulation of the heat shock protein hsc73. J Cell Biol. 1999;147:599‐610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Lapidot T, Sirard C, Vormoor J, et al. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature. 1994;367:645‐648. [DOI] [PubMed] [Google Scholar]
- 26. Boumahdi S, Driessens G, Lapouge G, et al. SOX2 controls tumour initiation and cancer stem‐cell functions in squamous‐cell carcinoma. Nature. 2014;511:246‐250. [DOI] [PubMed] [Google Scholar]
- 27. Ishiguro T, Sato A, Ohata H, et al. Establishment and characterization of an in vitro model of ovarian cancer stem‐like cells with an enhanced proliferative capacity. Cancer Res. 2016;76:150‐160. [DOI] [PubMed] [Google Scholar]
- 28. Lupia M, Angiolini F, Bertalot G, et al. CD73 regulates stemness and epithelial‐mesenchymal transition in ovarian cancer‐initiating cells. Stem Cell Reports. 2018;10:1412‐1425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Szotek PP, Pieretti‐Vanmarcke R, Masiakos PT, et al. Ovarian cancer side population defines cells with stem cell‐like characteristics and Mullerian Inhibiting Substance responsiveness. Proc Natl Acad Sci USA. 2006;103:11154‐11159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Al‐Hajj M, Wicha MS, Benito‐Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci USA. 2003;100:3983‐3988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. O'Brien CA, Pollett A, Gallinger S, Dick JE. A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature. 2007;445:106‐110. [DOI] [PubMed] [Google Scholar]
- 32. Ricci‐Vitiani L, Lombardi DG, Pilozzi E, et al. Identification and expansion of human colon‐cancer‐initiating cells. Nature. 2007;445:111‐115. [DOI] [PubMed] [Google Scholar]
- 33. Boiko AD, Razorenova OV, van de Rijn M, et al. Human melanoma‐initiating cells express neural crest nerve growth factor receptor CD271. Nature. 2010;466:133‐137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Kreso A, Dick JE. Evolution of the cancer stem cell model. Cell Stem Cell. 2014;14:275‐291. [DOI] [PubMed] [Google Scholar]
- 35. Yu F, Yao H, Zhu P, et al. let‐7 regulates self renewal and tumorigenicity of breast cancer cells. Cell. 2007;131:1109‐1123. [DOI] [PubMed] [Google Scholar]
- 36. El Helou R, Pinna G, Cabaud O, et al. miR‐600 acts as a bimodal switch that regulates breast cancer stem cell fate through WNT signaling. Cell Rep. 2017;18:2256‐2268. [DOI] [PubMed] [Google Scholar]
- 37. Lv C, Li F, Li X, et al. MiR‐31 promotes mammary stem cell expansion and breast tumorigenesis by suppressing Wnt signaling antagonists. Nat Commun. 2017;8:1036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Zhang B, Nguyen LXT, Li L, et al. Bone marrow niche trafficking of miR‐126 controls the self‐renewal of leukemia stem cells in chronic myelogenous leukemia. Nat Med. 2018;24:450‐462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Fornari F, Pollutri D, Patrizi C, et al. In hepatocellular carcinoma miR‐221 modulates sorafenib resistance through inhibition of caspase‐3‐mediated apoptosis. Clin Cancer Res. 2017;23:3953‐3965. [DOI] [PubMed] [Google Scholar]
- 40. Al‐Harbi S, Choudhary GS, Ebron JS, et al. miR‐377‐dependent BCL‐xL regulation drives chemotherapeutic resistance in B‐cell lymphoid malignancies. Mol Cancer. 2015;14:185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Ahn JH, Lee HS, Lee JS, et al. nc886 is induced by TGF‐beta and suppresses the microRNA pathway in ovarian cancer. Nat Commun. 2018;9:1166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Velagapudi SP, Cameron MD, Haga CL, et al. Design of a small molecule against an oncogenic noncoding RNA. Proc Natl Acad Sci USA. 2016;113:5898‐5903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Childs‐Disney JL, Disney MD. Small molecule targeting of a MicroRNA associated with hepatocellular carcinoma. ACS Chem Biol. 2016;11:375‐380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Pang F, Zha R, Zhao Y, et al. MiR‐525‐3p enhances the migration and invasion of liver cancer cells by downregulating ZNF395. PLoS ONE. 2014;9:e90867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Guttilla IK, White BA. Coordinate regulation of FOXO1 by miR‐27a, miR‐96, and miR‐182 in breast cancer cells. J Biol Chem. 2009;284:23204‐23216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Onder TT, Gupta PB, Mani SA, Yang J, Lander ES, Weinberg RA. Loss of E‐cadherin promotes metastasis via multiple downstream transcriptional pathways. Cancer Res. 2008;68:3645‐3654. [DOI] [PubMed] [Google Scholar]
- 47. Gupta PB, Onder TT, Jiang G, et al. Identification of selective inhibitors of cancer stem cells by high‐throughput screening. Cell. 2009;138:645‐659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Fischer KR, Durrans A, Lee S, et al. Epithelial‐to‐mesenchymal transition is not required for lung metastasis but contributes to chemoresistance. Nature. 2015;527:472‐476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Zheng X, Carstens JL, Kim J, et al. Epithelial‐to‐mesenchymal transition is dispensable for metastasis but induces chemoresistance in pancreatic cancer. Nature. 2015;527:525‐530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Gregory PA, Bert AG, Paterson EL, et al. The miR‐200 family and miR‐205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat Cell Biol. 2008;10:593‐601. [DOI] [PubMed] [Google Scholar]
- 51. Song SJ, Poliseno L, Song MS, et al. MicroRNA‐antagonism regulates breast cancer stemness and metastasis via TET‐family‐dependent chromatin remodeling. Cell. 2013;154:311‐324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Subramanyam D, Lamouille S, Judson RL, et al. Multiple targets of miR‐302 and miR‐372 promote reprogramming of human fibroblasts to induced pluripotent stem cells. Nat Biotechnol. 2011;29:443‐448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Mani SA, Guo W, Liao MJ, et al. The epithelial‐mesenchymal transition generates cells with properties of stem cells. Cell. 2008;133:704‐715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Shelton PM, Duran A, Nakanishi Y, et al. The secretion of miR‐200s by a PKCzeta/ADAR2 signaling axis promotes liver metastasis in colorectal cancer. Cell Rep. 2018;23:1178‐1191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Ma L, Tao Y, Duran A, et al. Control of nutrient stress‐induced metabolic reprogramming by PKCzeta in tumorigenesis. Cell. 2013;152:599‐611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Tavazoie SF, Alarcon C, Oskarsson T, et al. Endogenous human microRNAs that suppress breast cancer metastasis. Nature. 2008;451:147‐152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Huang Q, Gumireddy K, Schrier M, et al. The microRNAs miR‐373 and miR‐520c promote tumour invasion and metastasis. Nat Cell Biol. 2008;10:202‐210. [DOI] [PubMed] [Google Scholar]
- 58. Valastyan S, Reinhardt F, Benaich N, et al. A pleiotropically acting microRNA, miR‐31, inhibits breast cancer metastasis. Cell. 2009;137:1032‐1046. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 59. Lin CW, Chang YL, Chang YC, et al. MicroRNA‐135b promotes lung cancer metastasis by regulating multiple targets in the Hippo pathway and LZTS1. Nat Commun. 2013;4:1877. [DOI] [PubMed] [Google Scholar]
- 60. Okamoto K, Ishiguro T, Midorikawa Y, et al. miR‐493 induction during carcinogenesis blocks metastatic settlement of colon cancer cells in liver. EMBO J. 2012;31:1752‐1763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Chou J, Lin JH, Brenot A, Kim JW, Provot S, Werb Z. GATA3 suppresses metastasis and modulates the tumour microenvironment by regulating microRNA‐29b expression. Nat Cell Biol. 2013;15:201‐213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Valeri N, Braconi C, Gasparini P, et al. MicroRNA‐135b promotes cancer progression by acting as a downstream effector of oncogenic pathways in colon cancer. Cancer Cell. 2014;25:469‐483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Mahon PC, Hirota K, Semenza GL. FIH‐1: a novel protein that interacts with HIF‐1alpha and VHL to mediate repression of HIF‐1 transcriptional activity. Genes Dev. 2001;15:2675‐2686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Zhou W, Fong MY, Min Y, et al. Cancer‐secreted miR‐105 destroys vascular endothelial barriers to promote metastasis. Cancer Cell. 2014;25:501‐515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Fong MY, Zhou W, Liu L, et al. Breast‐cancer‐secreted miR‐122 reprograms glucose metabolism in premetastatic niche to promote metastasis. Nat Cell Biol. 2015;17:183‐194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Zhang L, Zhang S, Yao J, et al. Microenvironment‐induced PTEN loss by exosomal microRNA primes brain metastasis outgrowth. Nature. 2015;527:100‐104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Tominaga N, Kosaka N, Ono M, et al. Brain metastatic cancer cells release microRNA‐181c‐containing extracellular vesicles capable of destructing blood‐brain barrier. Nat Commun. 2015;6:6716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Yan W, Wu X, Zhou W, et al. Cancer‐cell‐secreted exosomal miR‐105 promotes tumour growth through the MYC‐dependent metabolic reprogramming of stromal cells. Nat Cell Biol. 2018;20:597‐609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Lee TC, Ziff EB. Mxi1 is a repressor of the c‐Myc promoter and reverses activation by USF. J Biol Chem. 1999;274:595‐606. [DOI] [PubMed] [Google Scholar]
- 70. Valoczi A, Hornyik C, Varga N, Burgyan J, Kauppinen S, Havelda Z. Sensitive and specific detection of microRNAs by northern blot analysis using LNA‐modified oligonucleotide probes. Nucleic Acids Res. 2004;32:e175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Kloosterman WP, Wienholds E, de Bruijn E, Kauppinen S, Plasterk RH. In situ detection of miRNAs in animal embryos using LNA‐modified oligonucleotide probes. Nat Methods. 2006;3:27‐29. [DOI] [PubMed] [Google Scholar]
- 72. Neely LA, Patel S, Garver J, et al. A single‐molecule method for the quantitation of microRNA gene expression. Nat Methods. 2006;3:41‐46. [DOI] [PubMed] [Google Scholar]
- 73. Kota J, Chivukula RR, O'Donnell KA, et al. Therapeutic microRNA delivery suppresses tumorigenesis in a murine liver cancer model. Cell. 2009;137:1005‐1017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Hagiwara K, Kosaka N, Yoshioka Y, Takahashi RU, Takeshita F, Ochiya T. Stilbene derivatives promote Ago2‐dependent tumour‐suppressive microRNA activity. Sci Rep. 2012;2:314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Hou J, Lin L, Zhou W, et al. Identification of miRNomes in human liver and hepatocellular carcinoma reveals miR‐199a/b‐3p as therapeutic target for hepatocellular carcinoma. Cancer Cell. 2011;19:232‐243. [DOI] [PubMed] [Google Scholar]
- 76. Callegari E, D'Abundo L, Guerriero P, et al. miR‐199a‐3p modulates MTOR and PAK4 pathways and inhibits tumor growth in a hepatocellular carcinoma transgenic mouse model. Mol Ther Nucleic Acids. 2018;11:485‐493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Corsten MF, Miranda R, Kasmieh R, Krichevsky AM, Weissleder R, Shah K. MicroRNA‐21 knockdown disrupts glioma growth in vivo and displays synergistic cytotoxicity with neural precursor cell delivered S‐TRAIL in human gliomas. Cancer Res. 2007;67:8994‐9000. [DOI] [PubMed] [Google Scholar]
- 78. Lee TJ, Yoo JY, Shu D, et al. RNA nanoparticle‐based targeted therapy for glioblastoma through inhibition of oncogenic miR‐21. Mol Ther. 2017;25:1544‐1555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Henke JI, Goergen D, Zheng J, et al. microRNA‐122 stimulates translation of hepatitis C virus RNA. EMBO J. 2008;27:3300‐3310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Jopling CL, Norman KL, Sarnow P. Positive and negative modulation of viral and cellular mRNAs by liver‐specific microRNA miR‐122. Cold Spring Harb Symp Quant Biol. 2006;71:369‐376. [DOI] [PubMed] [Google Scholar]
- 81. Randall G, Panis M, Cooper JD, et al. Cellular cofactors affecting hepatitis C virus infection and replication. Proc Natl Acad Sci USA. 2007;104:12884‐12889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Fabani MM, Gait MJ. miR‐122 targeting with LNA/2′‐O‐methyl oligonucleotide mixmers, peptide nucleic acids (PNA), and PNA‐peptide conjugates. RNA. 2008;14:336‐346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Jopling CL, Yi M, Lancaster AM, Lemon SM, Sarnow P. Modulation of hepatitis C virus RNA abundance by a liver‐specific MicroRNA. Science. 2005;309:1577‐1581. [DOI] [PubMed] [Google Scholar]
- 84. Luna JM, Scheel TK, Danino T, et al. Hepatitis C virus RNA functionally sequesters miR‐122. Cell. 2015;160:1099‐1110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Lanford RE, Hildebrandt‐Eriksen ES, Petri A, et al. Therapeutic silencing of microRNA‐122 in primates with chronic hepatitis C virus infection. Science. 2010;327:198‐201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Janssen HL, Reesink HW, Lawitz EJ, et al. Treatment of HCV infection by targeting microRNA. N Engl J Med. 2013;368:1685‐1694. [DOI] [PubMed] [Google Scholar]
- 87. Ebert MS, Neilson JR, Sharp PA. MicroRNA sponges: competitive inhibitors of small RNAs in mammalian cells. Nat Methods. 2007;4:721‐726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Meng J, Chen S, Han JX, et al. Derepression of co‐silenced tumor suppressor genes by nanoparticle‐loaded circular ssDNA reduces tumor malignancy. Sci Transl Med. 2018;10:eaa06321. [DOI] [PubMed] [Google Scholar]
- 89. Baer C, Squadrito ML, Laoui D, et al. Suppression of microRNA activity amplifies IFN‐gamma‐induced macrophage activation and promotes anti‐tumour immunity. Nat Cell Biol. 2016;18:790‐802. [DOI] [PubMed] [Google Scholar]
- 90. Xu S, Tao Z, Hai B, et al. miR‐424(322) reverses chemoresistance via T‐cell immune response activation by blocking the PD‐L1 immune checkpoint. Nat Commun. 2016;7:11406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Muliaditan T, Caron J, Okesola M, et al. Macrophages are exploited from an innate wound healing response to facilitate cancer metastasis. Nat Commun. 2018;9:2951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Saito T, Nishikawa H, Wada H, et al. Two FOXP3(+)CD4(+) T cell subpopulations distinctly control the prognosis of colorectal cancers. Nat Med. 2016;22:679‐684. [DOI] [PubMed] [Google Scholar]
- 93. Hossain S, Stanislaus A, Chua MJ, et al. Carbonate apatite‐facilitated intracellularly delivered siRNA for efficient knockdown of functional genes. J Control Release. 2010;147:101‐108. [DOI] [PubMed] [Google Scholar]
- 94. Hiraki M, Nishimura J, Takahashi H, et al. Concurrent targeting of KRAS and AKT by MiR‐4689 is a novel treatment against mutant KRAS colorectal cancer. Mol Ther Nucleic Acids. 2015;4:e231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Inoue A, Mizushima T, Wu X, et al. A miR‐29b byproduct sequence exhibits potent tumor‐suppressive activities via inhibition of NF‐kappaB signaling in KRAS‐mutant colon cancer cells. Mol Cancer Ther. 2018;17:977‐987. [DOI] [PubMed] [Google Scholar]
- 96. Orellana EA, Tenneti S, Rangasamy L, Lyle LT, Low PS, Kasinski AL. FolamiRs: Ligand‐targeted, vehicle‐free delivery of microRNAs for the treatment of cancer. Sci Transl Med. 2017;9:eaam9327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Fernandez M, Javaid F, Chudasama V. Advances in targeting the folate receptor in the treatment/imaging of cancers. Chem Sci. 2018;9:790‐810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Kamerkar S, LeBleu VS, Sugimoto H, et al. Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer. Nature. 2017;546:498‐503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Pi F, Binzel DW, Lee TJ, et al. Nanoparticle orientation to control RNA loading and ligand display on extracellular vesicles for cancer regression. Nat Nanotechnol. 2018;13:82‐89. [DOI] [PMC free article] [PubMed] [Google Scholar]
