Abstract
miRNA are critical messengers in the tumor microenvironment (TME) that influence various processes leading to immune suppression, tumor progression, metastasis and resistance. Strategies to modulate miRNAs in the TME have important implications in overcoming these challenges. However, miR delivery to specific cells in the TME has been challenging. This review discusses nanomedicine strategies to achieve cell-specific delivery of miRNAs. The key goal of delivery is to activate the tumor immune landscape as well as to prevent chemotherapy resistance. Specifically, the use of hyaluronic acid-based nanoparticle miRNA delivery to the TME is discussed. The discussion is focused on miRNA-125b for reprogramming tumor-associated macrophages to overcome immunosuppression and miRNA-let-7b to overcome resistance to anticancer chemotherapeutics because both these miRNAs have been extensively evaluated for delivery with hyaluronic acid-based delivery systems.
Keywords: : hyaluronic acid nanoparticles, miRNA-125b, miRNA-let7b, miRNAs, polyethyleneimine, tumor-associated macrophages
Plain language summary
miRNAs are the messenger molecules with the tumor that have significant influence on the cancer growth and progression. Many strategies have been evaluated to modulate these messengers artificially to obstruct cancer growth and destroy cancer cells. This review discusses one such strategy to deliver these messenger miRNAs using hyaluronic acid-based nanoparticles that harness the body's own immune system to fight cancer. The two miRNAs that this review discusses are miRNA-125b and miRNA-let7b.
Graphical abstract
Cancer development involves a series of events in which normal cells first convert to rapidly growing cells through a succession of genetic events [1]. However, these rapidly growing cells need a favorable environment to progress to cancer. This favorable environment for tumor cell progression, growth and metastasis is termed the tumor microenvironment (TME) [2]. The rapid progress of scientific knowledge regarding the mechanisms of cancer progression has led to updating the hallmarks of cancers in the scientific community every decade. With the evidence of the existence of different cell types within the tumors, the TME found its place within the hallmarks of cancer in the past decade [3]. These hallmarks of cancers clearly illustrate the importance of the microenvironment in tumor growth and metastasis and in determining a potential therapeutic's efficacy in inhibiting tumor growth [3]. The TME is a dense environment comprising varied cell types such as tumor cells, cancer stem cells, stromal cells (fibroblasts, endothelial cells, adipocytes) and immune cells such as macrophages, dendritic cells and T cells; and the complex interactions between components [4]. The noncellular components of the microenvironment within the tumor include extracellular matrix such as collagen, fibronectin, hyaluronan and laminin. The communication between the tumor cells and the nonmalignant components of the tumor environment is an important factor in tumor development and progression [5].
TME defines the immune landscape & resistance to cancer therapeutics
The diverse TME is one of the leading causes of heterogenicity observed in tumors of similar origin [6]. Heterogenicity among tumors often leads to variance in treatment efficacy among patients [7]. The most widespread example of TME heterogenicity is the occurrence of cold and hot tumors, which solely depends on immune cell populations in the TME [8]. TMEs with an increased population of infiltrating T cells are termed hot tumors, whereas TMEs with a low numbers of infiltrating T cells and increased population of Tregs and/or immune-suppressive cells are termed cold tumors. Thus, the TME plays a major role in immunomodulation [9] and the development of resistance to cancer therapeutics [10,11].
The nonmalignant cellular elements of the TME are significant contributors to antitumor immunity and therapeutics resistance. The nonmalignant cellular components include immune cells, stromal cells, ECM and other secretory molecules. Among immune cells, macrophages are one of the important cell types that undergo a phenotype switch in the TME (termed M2/anti-inflammatory macrophages), in turn supporting tumor development and progression and cancer metastasis [12]. The tumor-associated macrophages (TAMs) are known to induce resistance to cancer therapeutics through multiple mechanisms. TAMs secrete TGF and TNF-α to activate the epithelial to mesenchymal transition (EMT) pathway, in turn inducing EMT in tumor cells and resistance to therapeutics [13]. Additionally, TAMs secrete angiogenesis factors, such as VEGF, thymidine phosphorylates, urokinase-type plasminogen activator and phosphatidylinositol-glycan biosynthesis class F protein in tumor-promoting tumor progression and metastasis [14]. Furthermore, TAMs have been identified for secreting immunosuppressive factors such as prostaglandin E2, IL-10, CCL18, CCL22 and TGF-β, which play a major role in inhibiting Th1 immune responses [15]. Similar to macrophages, neutrophils are another class of immune cells of the TME that show changes in phenotype to perform immunosuppressive functions by recruiting macrophages and Tregs [16]. Additionally, the myeloid-derived suppressors cells are immune cells of the TME that are also involved in inducing immune suppression and resistance to cancer therapeutics [17].
The next class of nonmalignant cellular components of the TME is the stromal cells. Fibroblasts that are stromal cells in the TME mainly remodel the extracellular matrix to promote tumor development and progression [18]. Fibroblasts are involved in the activation of the Wnt signaling pathway, inducing resistance to cancer therapeutics through activation of p-glycoproteins and reducing intracellular reactive oxygen species [19]. Mesenchymal stem cells (MSCs) are yet another stromal cell type involved in promoting tumor progression by inducing resistance to cancer therapeutics [20–22]. These stem cells are also involved in recruiting anti-inflammatory macrophages in the TME [23], increasing the cancer stem cell population [24] and inducing EMT [25]. However, studies have also demonstrated the positive role of MSCs in the TME in multiple cancers and MSC therapy had been evaluated as an anticancer therapy [26,27]. Thus, further detailed evaluation is essential for the role of MSCs in the TME.
The nonmalignant components of the TME include secretory molecules along with the cellular components. Each of the cell types of the TME communicates with the others through direct interactions or paracrine interactions. The secretory molecules for paracrine interactions include growth factors, interferons and interleukins, among others. Another major class of cell-to-cell communications identified over the past decade are exosomes, which contain messenger molecules such as proteins or nucleic acids such as miRNA and mRNA [28]. This review focuses on modulating miRNAs in the TME to improve antitumor immunity and overcome therapeutic resistance.
Biogenesis & functions of miRNAs
miRNAs are a type of small, noncoding RNAs that have evolved in eukaryotes and regulate multiple biological pathways, such as immune responses, cell cycle progression, differentiation, proliferation, metabolism, apoptosis and stress tolerance [29]. About 2 decades ago, the first miRNA lin-4, which was discovered in Caenorhabditis elegans, was found to have complementary antisense to multiple sites in the 3′ UTR of the gene called lin-14 [30,31]. Since then, there has been much progress in the understanding of the biogenesis, functions and diagnostic potential of miRNA in a variety of diseases. Within the genome, miRNAs are encoded as individual genes or as clusters containing multiple different miRNAs [32]. The biogenesis of miRNA starts with the transcription of miRNA genes by RNA polymerase II/III (Pol II) into pri-miRNAs and is regulated by RNA Pol II-associated transcription factors and epigenetic modulators [33]. Pri-miRNAs are processed to single hairpins termed precursor miRNAs (pre-miRNAs) by a nuclear protein complex called a microprocessor. The microprocessor in a protein complex that contains Drosha (RNase III enzyme), the dsRNA-binding protein DiGeorge critical region 8, and additional components such as the DEAD-box RNA helicases p68 and p72 [34]. These processed pre-miRNAs are then transported to the cytoplasm through the export receptor exportin 5 and are processed by Dicer to 20–25 nucleotides dsRNA [35]. Once in the cytoplasm, the miRNAs exhibit their functional effects on various pathways, which is discussed in the next sections.
miRNAs have been shown to mediate gene regulation through either gene silencing or upregulation of gene expression or translation activation or inducing nuclear post-transcriptional gene regulation. In the case of miRNA-mediated gene silencing, the interaction between miRNA and its target mRNA is facilitated by RISC loading in which the ds miRNA produced by Dicer is transferred to the Argonuate (AGO) protein family. RISC chooses one strand to turn into the mature miRNA (guide strand) and discards the passenger strand [36]. miRNA in the RISC complex drives it to an mRNA target in a complementary-nucleotide-based way, initiating pathways such as mRNA deadenylation, decapping or translation repression, finally inhibiting protein synthesis [37,38]. Generally, the 3′UTR contains the mRNA target site, and the seed sequence is positioned at 2–7 nucleotides on the complementary strand, called the seed sequence [39]. A detailed mechanism of the various regulatory elements involved in the RISC complex and the miR-mediated silencing has been previously reported [34]. Finally, it has been reported that shuttling of miR and AGO by TNRC6A, Argonaute-navigator and their interaction with target mRNA within the nucleus leads to nuclear mRNA degradation, but a detailed mechanism has yet to be established [40,41]. Overall, miRNAs regulate gene expression not only in the cytoplasmic compartment but also in the nuclear compartment through their interaction with the aforementioned RNA–protein complexes.
miRNAs play a pivotal role in the regulation of the TME
Given the role of miRNA in the regulation of gene expression and biological functions of the cell, miRNAs have been shown to have an impact on conventional hallmarks of cancer. Abnormal miRNA expression in cancer cells compared with normal cells has been linked to a variety of mechanisms, such as chromosomal abnormalities, transcriptional control changes, epigenetic changes and defects in the miRNA biogenesis machinery [42,43]. Several studies using miRNA profiling and sequencing of tumor samples have shown that miRNAs can act as either tumor suppressors or oncogenic miRNA depending on whether they are downregulated or upregulated in a variety of cancers.
miRNAs as tumor suppressors
miRNA downregulation in human malignancies causes elevation or aberrant levels of target mRNA expression leading to increased cell growth, apoptosis, metabolism and invasion. In mammalian cells, miRNA-let-7 is the keeper of differentiation, and its unusual regulation and reduced expression of let-7 have been linked to cancer initiation and progression [44]. Let-7 targets oncogenes such as c-Myc, RAS, HMGA2, VEGF, Lin28 MDR family and long noncoding RNA H19 [45]. Recent studies have further shown that let-7 suppresses the expression of PD-L1 through interaction with the 3′-UTR in many human cancer cells, demonstrating that the LIN28/let-7 axis takes part in immune surveillance, driven by tumor cells [46]. miRNA levels can be regulated by tumor suppressor genes, and one such example is miRNA-34a, which is dysregulated in various cancers and has been demonstrated to be directly regulated by tumor suppressor p53 [47]. miRNA-34a overexpression can limit cancer cell growth and tumor progression in non-small-cell lung cancer models [48,49]. miRNA-34a also antagonizes many oncogenic processes by regulating genes that function in various cellular pathways [50]. Similarly, the miRNA-200 family (miRNA-200a, miRNA-200b, miRNA-200c, miRNA-141 and miRNA-429) is one of the widely studied miRNAs and is involved in many diverse functions, such as induction of EMT via downregulation of E-cadherin and consequent increases in ZEB proteins [51]. It has also been demonstrated that miRNA-200 influences angiogenesis indirectly via the downregulation of CXCL1 and IL-8, which are major cytokines in the TME [52].
miRNAs as oncogenes
miRNAs act as oncogenes by targeting tumor suppressor genes and have been reported to be overexpressed in tumor cells compared with normal cells. Two of the major oncogenic miRNAs reported are the miRNA-17-92 cluster and miRNA-21 [53]. Noncoding RNA C13orf25 encodes the miRNA-17-92 cluster and is known to be upregulated in several cancers. Expression levels of several miRNAs are elevated due to the amplification of the 13q31-32 locus in the chromosome in a variety of cancers [54]. Phosphate and tensin homolog (PTEN) and the transcription factor E2 family are among the earliest validated targets of the miRNA-17-92 cluster [55]. More recently miRNA-17-92 cluster has also been shown to regulate a variety of immune cells in the TME, such as B cells and subsets of T cells, such as Th1, Th2, T follicular helper cells, Tregs, monocytes/macrophages, NK cells and dendritic cells [56]. miRNA-21 is one of the trivial biomarkers in many diseases, its overexpression is strongly associated with a variety of hematological and solid malignancies [57]. miRNA-21 exerts its oncogenic function by binding to the 3′UTR of the multiple tumor suppressor genes such as PTEN, TPM1 and PDCD4 [58–60]. It induces inhibition of cellular apoptosis by negatively regulating p53 and NF-κB signaling [61]. miRNA-21 has also been reported to be one of the most abundant miRNAs in TAMs and was shown to drive tumor growth by modulation of cytokine production [62]. Multiple studies have reported that miR-155 promotes growth in several types of cancer, including breast and lung cancers [63]. miRNA-155 directly targets and inhibits many genes, including SHIP1, WEE1, VHL, TP53INP1, BCL2 and SOX family genes, which are involved in many critical cellular functions including immune response, DNA damage response, hypoxia, inflammation and tumorigenesis [64].
Overall, these miRNAs, in addition to cancer cells, also regulate various stromal cells and exert their impact to recruit or promote the differentiation of suppressive immune cells, such as Treg, cancer-associated fibroblasts, tumor-associated MSCs, TAMs and others involved in the TME [65]. These are some of the most investigated miRNAs, but there are certainly many more examples of specific miRNAs in each type of cancer that have been reviewed extensively by others [66,67].
miRs in macrophage phenotype modulation
The percentage of TAMs in the TME is associated with a poor prognosis [68–70]. Given the role of miR in TME, several studies performed over the last decade suggest a prominent role for miRNAs as key regulators of macrophage differentiation, infiltration and activation. Certain miRNAs such as miRNA-155, miRNA-125b, let-7b and miRNA-29, are known to promote repolarization of macrophages from M2 toward M1 phenotype and play a role in the regulation of immune responses in cancer and inflammation [71–73]. It has been reported that the expression of miRNA-21 in TAMs in particular, were responsible for promoting tumor growth [62]. Another study in prostate cancer showed the let-7b upregulation is characteristic of prostatic TAMs and the downregulation of let-7b in TAMs leads to changes in expression profiles of inflammatory cytokines such as IL-12, IL-23, IL-10 and TNF-α [74]. TAMs treated with let-7b inhibitors reduce angiogenesis and prostate carcinoma cell mobility. Although these studies contradict the previously described role of let-7b as a tumor suppressor miRNA in lung and ovarian cancer, it is important to realize that the impact of miRNAs on tumorigenesis has specific functions depending on the type of cancer, cell type and the baseline levels compared with healthy tissues. Table 1 lists miRNAs involved in immunomodulation for various types of cancers; however, the focus of this review is on miRNA-125b and its role in overcoming immunosuppression.
Table 1. . miRNAs involved in immunomodulation in for various types of cancers.
| Tumor type | miRNA | Mode of overcoming immune suppression | Refs. |
|---|---|---|---|
| Melanoma | miRNA-23a miRNA-155 |
Increases CTL-mediated cytotoxicity Induces effector T-cell responses |
[75] [76] |
| Lung | miRNA-34a miRNA-200a,b,c miRNA-760 miRNA-125b |
Inhibits PD-L1 expression Inhibits PD-L1 expression Downregulating IDO1 and eliminating Tregs Repolarization of TAMs |
[77] [78] [79] [80] |
| Lymphocytic leukemia | miRNA-93 miRNA-106b |
Decreases CXCL12 AND PD-L1 expression Decreases CXCL12 AND PD-L1 expression |
[81] [81] |
| Hepatocellular carcinoma | miRNA-182 | Activates NK cells | [82] |
| Ovarian cancer | miRNA-424 miRNA-125b |
Activation of monocytic differentiation, antitumor immune response Repolarization of TAMs |
[83] [84] |
| Colorectal carcinoma | miRNA-138-5p miRNA-let7a |
Inhibits PD-L1 expression Decreases immunocompetence by T cells |
[85] [86] |
| Acute myeloid leukemia | miRNA-17-5p, miRNA-20a, miRNA-106a | Inhibition of monocytic differentiation and maturation | [87] |
| Breast cancer | miRNA-101-3p | Repolarization of TAMs | [88] |
| B-cell lymphoma | miRNA-152-3p | Promotes cytotoxic T-lymphocyte proliferation via the PD-1/PD-L1 checkpoint | [89] |
TAMs: Tumor-associated macrophages.
Changes in miRNA profiles with tumor relapse & chemoresistance
Each tumor type has a distinct miRNA signature that distinguishes it from normal tissues and other cancer types. Advances in various analytical tools such as miRNA profiling using microarray-based hybridization-based methods (e.g., DNA microarrays, quantitative reverse transcription PCR, Nanostring) and high-throughput sequencing (e.g., RNA sequencing and next-generation sequencing) have enabled the profiling of tumor samples from various cancer types. One of the first studies performed in epithelial ovarian cancer patient samples using genome-wide microarray analysis showed that the most significantly overexpressed miRNAs were miRNA-200a, miRNA-141, miRNA-200c and miRNA-200b, whereas miRNA-199a, miRNA-140, miRNA-145 and miRNA-125b were among the most down-modulated miRNAs. miRNA expression levels within the same type of cancer have been found to have differential expression profiles depending on various factors, such as cancer subgroup and stage, response to chemotherapy, disease prognosis and progression-free survival of disease, among others. miRNA profiling revealed differences in miRNA expression between molecular subtypes of cancer such as high-grade serous ovarian carcinoma, clear cell ovarian carcinoma and ovarian surface epithelium. miRNA-200 family members and miR-182-5p were the most overexpressed in high-grade serous ovarian carcinoma and clear cell ovarian carcinoma compared with ovarian surface epithelium, whereas miRNA-383 was the most underexpressed [90]. The same study in ovarian cancer also identified miRNAs expression that was correlated with specific ovarian cancer bio-pathologic features, such as histotype, lymphovascular and organ invasion and involvement of ovarian surface. Similarly, studies performed in surgically resected EGFR-mutated lung adenocarcinoma samples found that miRNA-21 had a significantly higher expression in lung cancer compared with normal lung tissues [91]. A study designed to identify miRNA-based circulating biomarkers to predict clinical outcomes of colorectal cancer shows that a four miRNA signature (miRNA-652-3p, miRNA-342-3p, miRNA-501-3p and miRNA-328-3p) could help in the prediction of tumor relapse and the overall survival of patients [92].
Changes in miRNA expression are associated with the development of resistance to chemotherapy in patients [93,94]. More evidence linking the miRNA and resistance was established when the NCI-60 panel of various cancer cell types was profiled for miRNA expression in the response to a panel of anticancer drugs. The miRNA expression profile revealed a strong correlation between the expression patterns of miRNAs and the potency patterns of anticancer compounds [95]. In particular, the change in cellular levels of the let-7 family, miRNA-16 and miRNA-21 affected the potencies of several anticancer agents by up to 4-fold depending on the compound class and cancer type. miRNAs have been demonstrated to alter cellular response to anticancer agents via modulation of survival pathways and/or apoptotic response or to affect mechanisms other than survival and apoptotic signaling, such as drug targets and DNA repair systems [93]. miRNAs may also function in drug resistance, including miRNA-130a, which targets the prometastatic and chemoresistance associated M-CSF and miRNA-27a and miRNA-451, which regulates the expression of the drug transporter MDR1 (P-glycoprotein), a protein implicated in paclitaxel-resistant ovarian cancer [96]. Exosome-mediated miRNA transfer between host–tumor cells play a pivotal role in tumor progression and resistance to treatment [97]. These studies indicate the role of miRNA profiling in the identification of novel biomarkers, prognostic indicators and therapy for precision medicine in cancer. Table 2 lists miRNAs that have been involved in drug resistance for various types of cancers; however, the focus of this review is on miRNA-let-7b and its role in overcoming drug resistance.
Table 2. . miRNAs involved in drug resistance for various types of cancers.
| Tumor type | miRNA | Targets/pathway | Refs. |
|---|---|---|---|
| Ovarian cancer | miRNA-let-7 (b, c, d, e, f, i, g) miRNA-27a miRNA-451 miRNA-200 (a, b, c) |
MDR1, Myc, RAS, E2F1, E2F5, LIN28, PBX3, HMGA2, BRCA1, RAD51, PARP, IGF1 HIPK2, AMPK, MDR1, AKT, K-RAS/B-RAF c-Myc, MDR1/P-glycoprotein, RAB14 TUBB3, ZEB1, ZEB2, E-Cadherin, DNMT3A/DNMT3B, AP-2α |
[98–102] [103–105] [106–108] [109–112] |
| Lung cancer | miRNA-34 (a, b, c) miRNA17-92 Cluster (miRNA 17, 20a, 20b) miRNA-21 miRNA-221/222 |
Wnt1, Notch1, Wnt3, MTA2, CD44, c-MYC, among others (PEBP4, TCF1/LEF1) p53, TGFβR, PTEN, CDKN1A, BCL-2, BECN1 TRAIL, SIDT1, PI3K/AKT APAF-1 |
[113–116] [117–119] [120,121] [122] |
| Breast cancer | miRNA-30 b, c miRNA-141 miRNA-17/20 miRNA-155 |
TWF1, YWHAZ, MAPK pathway, CCNE2 CTNNB1 P53 and AKT, cyclin D1 RAD51, FOXO3a |
[123,124] [125] [126,127] [128,129] |
| Pancreatic cancer | miRNA-320 a, c miRNA-1246 miRNA-210 miRNA-455 |
PDCD4, SMARCC1 CCNG2 ABCC5 TAZ |
[130,131] [132] [133] [134] |
Challenges associated with miRNA delivery & transfection
The major challenge with miRNAs as therapeutic modalities has been their effective functional delivery to the cell type of interest. Nanocarriers have been at the forefront as delivery systems for miRNAs [90]. The use of positively charged polymers and lipids have been extensively exploited to interact electrostatically with negatively charged miRNAs, forming a complex capable of protecting miRNAs from degradation activity of the nucleases and delivering them intact to the cells [135]. However, for the miRNAs to demonstrate their desired activity, the nanocarriers need to deliver the miRNAs effectively [136]. The nanocarriers must overcome extracellular barriers such as passive delivery to the liver, and spleen because the permeability of vasculature results in leakage of nanocarriers primarily to these organs. Other extracellular barriers include the endo- and exonucleases in the circulation and stimulation of the immune system for the breakdown of nanocarriers before delivering the nucleic acid to the site of action [137].
As the extracellular barriers are surpassed, there is an array of intracellular barriers inhibiting the effective functionality of miRNAs. Unlike DNA delivery wherein the plasmid has to be delivered to the nucleus, miRNA has an added advantage to be delivered to the cytoplasm [138]. However, there are numerous barriers to effective miRNA delivery. The first step is crossing the cell membrane to enter the cytoplasmic space of the cell [139]. For nucleic acid delivery, various physical methods such as electroporation, photoporation and sonoporation have been extensively evaluated and are successful in achieving intracellular delivery. However, these mechanisms result in a reduction of cell viability and, most importantly, cannot be used as an in vivo delivery strategy [140]. Nanoformulations generally rely on endocytosis as a mechanism to enter cells. Nevertheless, once the nanoformulations are engulfed through endocytosis, they mainly localize to early endosomes, which are then converted to late endosomes with a reduced pH and activation of degrading enzymes to eventually form lysosomes [141]. Thus, the nanoformulations that enter the cell through endocytosis need to escape the endosomes before lysosomal degradation. Endosomal escape is a major challenge in the field, and there is an extensive body of research to design polymers and lipids capable of achieving endosomal escape [141].
Many strategies have been evaluated to enable nanoformulations to undergo endosomal escape such as use of lysosomotropic agents [142], photosensitizers [143] and cell-penetrating peptides [144,145]. Additionally, multiple pore-forming and fusion peptides have been evaluated for their ability to promote the endosomal escape of the nanocarriers [146,147]. Studies have demonstrated the use of VIPER (virus-inspired polymer for endosomal release) to enhance siRNA and plasmid delivery [148]. Amphipathic polycations have also been evaluated as a strategy for endosomal escape [149,150]. Apart from all these strategies, we and many others have evaluated is the use of are polymers such as polyethyleneimine (PEI) or poly(amidoamine)s, which demonstrate osmotic swelling and electrostatic interaction with endosomal membrane for efficient gene delivery [151,152].
Hyaluronic acid-based nanoparticles for miRNA delivery to tumor microenvironment
We have improved the PEI with hyaluronic acid (HA), a natural polysaccharide formed of two disaccharide units of N-acetyl D glucosamine and D-glucuronic acid with β(1–4) interglycosidic linkage and is thus biodegradable [153]. Because HA is naturally present in the body, it does not elicit immune/inflammatory response and binds specifically to cell surface receptors such as CD44; thus, it can promote active cell targeting ability in the nanoformulation. Additionally, HA can be conjugated to polymers such as PEI by simple amide linkage using click chemistry. We have coupled HA to PEI in the presence of carbodiimide and N-hydroxysuccinimide to facilitate the amide linkage. The unreacted components are then removed by dialysis of the reaction solution using a 12- to 14-kDa MWCO dialysis membrane. The purified HA–PEI polymer is then lyophilized and stored at −20°C. The conjugation of HA with PEI is confirmed using 1H-NMR spectroscopy (Figure 1A & B) [80]. Thus, HA is a widely used biomaterial in various delivery, therapeutic and theranostic applications [154–157]. HA conjugated with PEG has been used with other HA derivatives because PEG increases the circulation time and escapes the reticuloendothelial system [153,158]. Furthermore, these HA polymers have the ability to self-assemble when suspended in deionized water in the presence of nucleic acids (Figure 1C). PEI is positively charged and interacts with the negatively charged nucleic acids (miRNAs, in this case) through electrostatic interactions to form the core of the nanoparticles. The encapsulation of miRNAs with HA–PEI nanoparticles results in nanoparticles with hydrodynamic size ~100 nm as measured using a zetasizer based on dynamic light scattering (DLS) (Figure 1D). We further discuss these nanoformulations for delivery of miRNA-125b and miRNA-let-7b to the TME for overcoming immune suppression and for overcoming resistance to anticancer chemotherapeutics, respectively.
Figure 1. . Synthesis of hyaluronic acid derivatives and the formulation of microRNA-125b encapsulated nanoparticles.

(A) The 1H NMR spectrum of the HA (20 kDa) and PEI shows the characteristic COCH3 peak at ∽2 ppm for HA and peak for PEI between 2.5 and 3.0 ppm. (B) The 1H NMR spectrum of the HA–PEI with carbodiimide and N-hydroxysuccinimide coupling does not show the PEI peak but shows additional peaks between 2 and 3 ppm due to PEI conjugation on the HA backbone. (C) The HA–PEI/miRNA particles made at 27:1 ratio in deionized water were visualized by transmission electron microscopy. Scale bar indicates 100 nm on both of the images. (D) Hydrodynamic diameters, PDI and ζ-potentials of HA–PEI and HA–PEI–miRNA-125b nanoparticle formulations. n = 4 identically prepared samples; data are expressed as mean ± standard deviation.
HA: Hyaluronic acid; NMR: Nuclear magnetic resonance; PDI: Polydispersity index; PEI: Polyethyleneimine.
Reproduced with permission from Parayath et al. [80].
Intricate mechanism of miRNA-125b in overcoming immune suppression
The implications of miRNA-125b in the TME have been widespread, affecting the phenotype of endothelial cells, immune cells as well as the growth and proliferation of cancer cells [159]. Within different cancer types, there is an extensive disparity related to expression levels of miRNA-125b. Overexpression of miRNA-125b is observed in cancers such as glioblastoma, retinoblastoma and acute myeloid leukemia. In contrast, in cancers such as hepatocellular cancer, melanoma, ovarian cancer, breast cancercinoma, colorectal carcinoma, osteosarcoma and endometrial cancer, there is clinical evidence of downregulation of miRNA-125b [159]. However, most cancers show dysregulation of miRNA-125b. This has necessitated a need for a deeper dive within cancer types to evaluate the mechanism of action of miRNA-125b in the TME. The role of miRNA-125b in inhibiting angiogenesis [160,161], programming tumor macrophages to proinflammatory phenotype [135] and activating immune responses [162,163] has been demonstrated. However, in contrast to the role of miRNA-125b in suppressing tumor growth and metastasis, studies have shown that expression of miR-125b in fibroblasts converts them to cancer-associated fibroblasts promoting tumor growth [164]. This illustrates the contrasting role of miRNA-125b in the TME and emphasizes the fact that miR-125b would play different roles in different cell types within the TME. Thus, it becomes essential to classify the role of miRNA-125b within different cell types of the TME.
Delivery of miRNA-125b to the TME in multiple cancer models
Reprogramming of TAMs to an antitumoral phenotype has been demonstrated as an effective strategy in cancer therapeutics. With the promising role of miRNA-125b in reprogramming TAMs, we and others have evaluated miRNA-125b as a cancer treatment strategy [80,84,165–167]. We evaluated the use of miRNA-125b for reprogramming TAMs to proinflammatory M1 phenotype in multiple cancer models. First, we evaluated the delivery of miR-125b in non-small-cell lung cancers because there is evidence of a positive correlation between M1 macrophage densities in the lung tissue and patient survival time [80]. We demonstrated macrophage-specific transfection using HA–PEI nanoparticles’ affinity for CD44 receptors on the macrophage cell surface. Furthermore, exploiting the ability of peritoneal macrophages to migrate toward the site of injury and/or inflammation, subsequently led to a >sixfold increase in the M1-to-M2 macrophage ratio in lung tissues from a non-small-cell lung cancer mouse model (Figure 2) [80].
Figure 2. . Repolarization of tumor-associated macrophages following intraperitoneal administration of miRNA-125b in hyaluronic acid–polyethyleneimine (HA–PEI) nanoparticle formulations in the KRAS/p53 genetically engineered non-small-cell lung tumor model.

(A) Lungs were isolated from mice on day 12, and immunohistochemistry was performed on lungs sections using CD11b, CD206 and CD80 antibodies. Representative images of random areas on each section were imaged using confocal microscopy. Scale bar = 20 μm. The ratio of relative mean fluorescence intensity of CD80/CD206 in the HA–PEI–miRNA-125b group was compared with the control and HA–PEI-scrambled miRNA group (n = 6); data are expressed as mean ± standard deviation; *p < 0.05 compared with control. (B) Cells from homogenized lungs were stained with F4/80 antibody for isolation using MACS magnetic beads, and RNA was extracted from these cells for expression of inducible nitric oxide synthase (iNOS) and Arg-1. The ratio of iNOS to Arg-1 in IL-4 stimulated peritoneal macrophages treated with HA–PEI nanoformulations. Quantitative PCR was used for quantification of the gene expression level with β-actin as the internal control.
HA-PEI: Hyaluronic acid–polyethyleneimine; miR = miRNA.
Reproduced with permission from Parayath et al. [80].
In the ovarian TME, CSF-1 stimulates macrophages to reprogram to the protumoral phenotype, further facilitating tumor growth [168]. Furthermore, in epithelial-derived ovarian cancers, there is a correlation observed between the macrophage number and the amount of CSF-1 increases with cancer growth [169]. Thus, our strategy was to reprogram TAMs from a protumoral to a proinflammatory phenotype using miRNA-125b as an anticancer therapy for ovarian cancers [84]. We have demonstrated the use of HA-based nanoparticles in the selective targeting and uptake by TAMs in the peritoneal cavity of a syngeneic ovarian cancer mouse model. Additionally, miRNA-125b encapsulated in HA–PEI nanoparticles could reprogram TAMs to an immune-promoting phenotype and can act as an adjuvant therapy along with paclitaxel, which is a standard of care for ovarian cancers (Figure 3) [84].
Figure 3. . Tumor-associated macrophage repolarization using miRNA-125b in HA–PEI nanoparticles.

Female C57BL/6 mice were injected with ID8 cells on day 1. On day 10, the mice were injected intraperitoneally with either HA–PEI-scrambled miRNA or HA–PEI–miRNA-125 b at 1 mg/kg dose on alternate day for 3 days, and the peritoneal cavity was lavaged with 1× phosphate-buffered saline (PBS) on day 16, for peritoneal cells collection and analysis by FACS for (A) ratio of CD206 to CD80. Expression of (B) inducible nitric oxide synthase (iNOS)/Arg-1 ratio tumor-associated macrophages treated with HA–PEI nanoparticle formulations. Quantitative PCR was used for quantification of gene expression level with β-actin as the internal control. (C) Female C57BL/6 mice were injected with ID8 cells on day 1. On day 8, mice were injected intraperitoneally with paclitaxel (2 mg/kg), and on day, 10 mice were injected intraperitoneally with either HA–PEI-scrambled miRNA or HA–PEI–miRNA-125 b at 1 mg/kg dose on alternate days for 3 days, and peritoneal cavity was lavaged with 1× PBS on days 16 and 24. VEGF levels were determined by ELISA in the peritoneal lavage from mice. N = 4; data are analyzed by one-way analysis of variance.
HA: Hyaluronic acid; miR: miRNA; ns: No significance; PEI: Polyethyleneimine.
Reproduced with permission from Parayath et al. [84].
TAMs play an important role in tumor growth in pancreatic ductal adenocarcinoma (PDAC). The complicated TME has been an obstacle to effective PDAC therapy. PDAC progression shows an inverse correlation with numbers of TAMs in pancreatic cancer tissue [170,171], which validates reprogramming of TAMs as an effective therapeutic strategy for fast-growing cancers such as PDAC. In a genetically engineered rodent model of PDAC, we demonstrated preferential accumulation of miRNA-125b encapsulated HA-based nanoparticles encapsulating miRNA-125b in the pancreas of PDAC mice [165]. Moreover, we employed an additional mechanism to target M2 TAMs specifically by conjugating the M2 peptide to HA–PEI nanoparticles [165]. These miRNA-125b encapsulating nanoparticles effectively reprogrammed the TAMs in PDAC to M1 phenotype (proinflammatory) and thus can be evaluated further to understand its potential as an anticancer therapeutic in the PADC model.
miRNA-let-7b therapy to overcome chemoresistance
The aberrant expression of miRNA-let-7b has been correlated with cancer growth and development [172]. The miRNA-let-7 family members have sequence similarity and thus have similar functions [173]. The targets of let-7 targets are oncogenes and other genes critical for cancer growth such as Myc, RAS, HMGA2, VEGF, Lin28 MDR family and long noncoding RNA H19 [174]. The decreased expression of let-7b expression is correlated with ovarian cancer growth and poor prognosis. let-7b levels were shown to be influenced by the two major subtypes: the let-7b high- and let-7b low-expressing tumor cells. Monitoring the levels of let-7 after surgery and chemotherapy has demonstrated its utility as a biomarker. Reduced expression of let-7 is linked to fast-growing cancer with poor prognosis. Similarly, increased let-7 levels are associated with increased lifespan and better prognosis [175–177]. The expression of levels of let-7 have also been shown to differ based on the stage and site of the tumor; for example, miRNA-let-7 shows low expression at metastatic sites compared with the primary cancers [178–180]. Overall, all the aforementioned studies highlight the role of let-7 family members as a biomarker for advanced-stage cancer.
The multidrug resistance (MDR1) protein has been associated with developing resistance to therapeutic drugs [181]. let-7d has been shown to modulate MDR1 in patients treated with chemotherapy after surgical treatment, and loss of let-7d provides a suitable microenvironment for the development of resistance to chemotherapeutics [182]. Thus, there is a good correlation between lowered levels of let-7d and IMP-1 and MDR1 proteins in patients with relapsed ovarian cancer after chemotherapy. Several studies have shown that induction of let-7 expression modulates chemosensitivity in cancer cells and cancer mice models due to inhibition of chemoresistance genes and LIN28A/B, STAT3, E2F1 and IMP1 [174]. Let-7 overexpression in mice model studies show tumor regression and improved survival [183–185]. All these studies led us to hypothesize that delivery of let-7b to the tumor tissue would help protect against recurring chemoresistant ovarian carcinoma.
Delivery of miRNA-let-7b in a relapsed or resistant ovarian cancer model
Because delivery of miRNAs to tumor cells in vivo is challenging, we and others have evaluated the potential of HA–PEI polymers to deliver miRNAs [145]. The cotreatment of HA–PEI–let-7b nanoparticles and paclitaxel has resulted in reduced IC50 values for paclitaxel in resistant ID8 ovarian cancer cells. Furthermore, we observed 3.5-fold reduced expression levels of miRNA-let7b levels in chemoresistant mice model of ovarian cancer compared to expression levels of miR-let7b levels in naïve mice. Additionally, expression levels of miRNA-let-7b are back to baseline in mice treated with HA–PEI encapsulating miR-Let7b nanoparticles (Figure 4A) in a chemoresistant mice model of ovarian carcinoma. Furthermore, we observed a 30–70% reduction in mRNA levels of eight oncogenes in tumors of ovarian cancer mice compared with control (Figure 4B). Additionally, our study demonstrated lowered VEGF levels (20- and 10-fold) in dual paclitaxel and HA–PEI–miRNA-let-7b treated group compared with the control and paclitaxel or HA–PEI–let-7b individual treatment groups respectively [98]. Furthermore, delivering additional miRNAs that show lower expression in tumors would potentially help further improve efficacy. This needs to be validated in appropriate cancer models.
Figure 4. . Treatment of relapsed and refractory ID8-VEGF ovarian cancer bearing mice with microRNA Let7b.

(A) Quantification of absolute copies of let-7b in tumor nodules 48 h after three doses of let-7b dosing (intraperitoneal) (n = 4 per group). (B) Relative quantification of let-7b mediated target gene expression in changes 48 h after three doses of HA–PEI–let-7b/scr (n = 4 per group). (C) Measurement of antitumor efficacy of combination therapy using VEGF ELISA. VEGF levels (pg/ml) were measured from supernatants of ascitic fluid on day 55 (n = 6–8 per group). (D) Representative images of ID8-VEGF resistant tumor-bearing mice on day 55 after tumor inoculation and untreated control (resistant/relapse) showing ascitic fluid buildup, leading to abdominal bloating, paclitaxel (PTX) only with ascites in the peritoneal cavity and PTX + HA–PEI–let-7b group with no ascites formation (p-values for A, B and C = ****p < 0.0001; ***p < 0.001; **p < 0.01, respectively, and were obtained by Student t-test comparing PTX + HA–PEI–let-7b with the other groups).
HA-PEI: Hyaluronic acid–polyethyleneimine; PTX: Paclitaxel.
Conclusion
miRNAs are important players in cell-to-cell communications in the tumor microenvironment and are known to modulate the tumor immune landscape. Inducing favorable miRNAs into the tumor microenvironment to hinder tumor development by activating intrinsic immune cells of the tumor microenvironment is an encouraging anticancer strategy. Delivering miRNAs to the tumor microenvironment to reprogram the TAMs to perform antitumor functions has been an efficient strategy to stimulate the immune system and can be exploited as a combination therapy with the current standard of care cancer treatment options. Furthermore, another class of miRNAs which are the tumor suppressors miRNAs have shown promising results in overcoming chemoresistance. With the increasing acceptance of the nonviral delivery systems for delivering nucleic acids and the advancement of tools for miR profiling of tumors, it is essential to extensively investigate the therapeutic potential of miRNAs as a cancer treatment strategy as monotherapy or adjuvant therapy.
Future perspective
Numerous miRNAs have been identified to overcome immunosuppression and chemoresistance in various cancer models. For effective delivery of these miRNAs to the TME, a competent delivery system is the key criterion for the success. With the rapid advancement of the field of delivery systems and the success of lipid nanoparticles for the COVID vaccine, the delivery of tumor-targeted delivery miRNAs could be the next step in the clinic. However, an important factor to consider with miRNAs is the target specificity. Because these small oligos could potentially have multiple targets, it could be important to consider the off-target effects of these miRNAs in the TME. Additionally, the kinetics of action of these miRNAs as the adjuvant therapy to other anticancer therapies needs to be evaluated in depth. Nonetheless, there will be a surge in RNA therapeutics in clinical settings, and miRNAs could certainly be an important part of these therapeutic modalities.
Executive summary.
The tumor microenvironment is a complex environment comprised of varied cellular and non-cellular components that interact with one another through messenger molecules to facilitate tumor growth and progression.
MiRs are a class of small non-coding RNAs that play a vital role in cell-to-cell communication within the tumor microenvironment.
MiR profiling of the tumor microenvironment has revealed the importance of miRs in determining the immune landscape of the tumors and the probability of tumors developing chemoresistance.
Self-assembled nanoparticles of hyaluronic acid (HA) modified with polyethyleneimine (PEI) are an efficient tool to deliver the miRs to the specific cell type within the tumor microenvironment.
HA–PEI nanoparticles can deliver miR-125b to tumor-associated macrophages and reprogram these macrophages to antitumoral phenotype and can act as an adjuvant therapy along with paclitaxel for ovarian cancers.
Combination therapy of paclitaxel and HA–PEI encapsulating miR-let7b shows a significant reduction in tumor growth in a relapsed mice model of ovarian cancer.
The ability to modulate the miRs of the tumor microenvironment in order to overcome immunosuppression and therapeutic resistance is a promising anticancer treatment strategy.
Acknowledgments
The authors appreciate all the former and current lab members who have contributed immensely to the studies described in this review article.
Footnotes
Financial & competing interests disclosure
Our research on miR-based modulation of the tumor microenvironment has been supported by US National Cancer Institute grants R56-CA198492 and R21-CA213114 and by the Northeastern University–Dana Farber Joint Program on Cancer Drug Development. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.
No writing assistance was utilized in the production of this manuscript.
References
Papers of special note have been highlighted as: • of interest
- 1.Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 100(1), 57–70 (2000). [DOI] [PubMed] [Google Scholar]
- 2.Arneth B. Tumor microenvironment. Medicina. 56(1), 15 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 144(5), 646–674 (2011). [DOI] [PubMed] [Google Scholar]
- 4.Whiteside TL. The tumor microenvironment and its role in promoting tumor growth. Oncogene. 27(45), 5904–5912 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Bejarano L, Jordāo MJ, Joyce JA. Therapeutic targeting of the tumor microenvironment. Cancer Discov. 11(4), 933–959 (2021). [DOI] [PubMed] [Google Scholar]
- 6.Baghban R Roshangar L, Jahanban-Esfahlan R et al. Tumor microenvironment complexity and therapeutic implications at a glance. Cell Commun. Signal. 18(1), 1–19 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Dagogo-Jack I, Shaw AT. Tumour heterogeneity and resistance to cancer therapies. Nat. Rev. Clin. Oncol. 15(2), 81–94 (2018). [DOI] [PubMed] [Google Scholar]
- 8.Duan Q, Zhang H, Zheng J, Zhang L. Turning cold into hot: firing up the tumor microenvironment. Trends Cancer. 6(7), 605–618 (2020). [DOI] [PubMed] [Google Scholar]
- 9.Giustarini G, Pavesi A, Adriani G. Nanoparticle-based therapies for turning cold tumors hot: how to treat an immunosuppressive tumor microenvironment. Front. Bioeng. Biotechnol. 9, 458 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Kumari S, Advani D, Sharma S et al. Combinatorial therapy in the tumor microenvironment: where do we stand? Biochim. Biophys. Acta BBA Rev. Cancer. 1876(2), 188585 (2021). [DOI] [PubMed] [Google Scholar]
- 11.Gonzalez-Molina J, Moyano-Galceran L, Single A et al. Chemotherapy as a regulator of extracellular matrix-cell communication: implications in therapy resistance. Semin. Cancer. Biol. (2022). [DOI] [PubMed] [Google Scholar]
- 12.Tan Y, Wang M, Zhang Y et al. Tumor-associated macrophages: a potential target for cancer therapy. Front. Oncol. 11, 2201 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Chen Y, Tan W, Wang C. Tumor-associated macrophage-derived cytokines enhance cancer stem-like characteristics through EMT. Onco Targets Ther. 11, 3817 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Hwang I, Kim JW, Ylaya K et al. Tumor-associated macrophage, angiogenesis, and lymphangiogenesis markers predict prognosis of non-small-cell lung cancer patients. J. Transl. Med. 18(1), 1–15 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Goswami KK, Bose A, Baral R. Macrophages in the tumor: an inflammatory perspective. Clin. Immunol. 232, 108875 (2021). [DOI] [PubMed] [Google Scholar]
- 16.Mishalian I, Bayuh R, Eruslanov E et al. Neutrophils recruit regulatory T-cells into tumors via secretion of CCL17 – a new mechanism of impaired antitumor immunity. Int. J. Cancer 135(5), 1178–1186 (2014). [DOI] [PubMed] [Google Scholar]
- 17.Kumar V, Patel S, Tcyganov E, Gabrilovich DI. The nature of myeloid-derived suppressor cells in the tumor microenvironment. Trends Immunol. 37(3), 208–220 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Yamauchi M, Gibbons DL, Zong C et al. Fibroblast heterogeneity and its impact on extracellular matrix and immune landscape remodeling in cancer. Matrix. Biol. 91, 8–18 (2020). [DOI] [PubMed] [Google Scholar]
- 19.Wu F, Yang J, Liu J et al. Signaling pathways in cancer-associated fibroblasts and targeted therapy for cancer. Signal Transduct. Target. Ther. 6(1), 1–35 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Vakhshiteh F, Atyabi F, Ostad SN. Mesenchymal stem cell exosomes: a two-edged sword in cancer therapy. Int. J. Nanomed. 14, 2847 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Yang H-Y, Qu R-M, Lin X-S et al. IGF-1 from adipose-derived mesenchymal stem cells promote radioresistance of breast cancer cells. Asian Pac. J. Cancer. Prev. 15(23), 10115–10119 (2015). [DOI] [PubMed] [Google Scholar]
- 22.Houthuijzen JM, Daenen LGM, Roodhart JML, Voest EE. The role of mesenchymal stem cells in anti-cancer drug resistance and tumour progression. Br J. Cancer. 106(12), 1901–1906 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Rivera-Cruz CM, Shearer JJ, Figueiredo Neto M, Figueiredo ML. The immunomodulatory effects of mesenchymal stem cell polarization within the tumor microenvironment niche. Stem Cell. Int. 2017, (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Luo J, Ok Lee S, Liang L et al. Infiltrating bone marrow mesenchymal stem cells increase prostate cancer stem cell population and metastatic ability via secreting cytokines to suppress androgen receptor signaling. Oncogene. 33(21), 2768–2778 (2014). [DOI] [PubMed] [Google Scholar]
- 25.Wu S, Wang Y, Yuan Z et al. Human adipose-derived mesenchymal stem cells promote breast cancer MCF7 cell epithelial-mesenchymal transition by cross interacting with the TGF-β/Smad and PI3K/AKT signaling pathways. Mol. Med. Rept. 19(1), 177–186 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Shah K. Mesenchymal stem cells engineered for cancer therapy. Adv. Drug Deliv. Rev. 64(8), 739–748 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Takayama Y, Tsukimori C, Shimizu Y et al. Anticancer drug-loaded mesenchymal stem cells for targeted cancer therapy. J. Control. Release. 329, 1090–1101 (2021). [DOI] [PubMed] [Google Scholar]
- 28.Conti I, Varano G, Simioni C et al. miRNAs as influencers of cell–cell communication in the tumor microenvironment. Cells. 9(1), 220 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Ghildiyal M, Zamore PD. Small silencing RNAs: an expanding universe. Nat. Rev. Genet. 10(2), 94–108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Ambros V, Lee RC, Lavanaway A et al. MiRs and other tiny endogenous RNAs in C. elegans. Curr. Biol. 13(10), 807–818 (2003). [DOI] [PubMed] [Google Scholar]
- 31.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. 75(5), 855–862 (1993). [DOI] [PubMed] [Google Scholar]
- 32.Lim LP, Glaser ME, Yekta S et al. Vertebrate miR genes. Science. 299(5612), 1540 (2003). [DOI] [PubMed] [Google Scholar]
- 33.Lee Y, Kim M, Han J et al. MiR genes are transcribed by RNA polymerase II. Embo. J. 23(20), 4051–4060 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Treiber T, Treiber N, Meister G. Regulation of miRNA biogenesis and its crosstalk with other cellular pathways. Nat. Rev. Mol. Cell Biol. 20(1), 5–20 (2019). [DOI] [PubMed] [Google Scholar]
- 35.Kim YK, Kim B, Kim VN. Re-evaluation of the roles of DROSHA, Export in 5, and DICER in miR biogenesis. Proc. Natl. Acad. Sci. USA. 113(13), E1881–1889 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Kobayashi H, Tomari Y. RISC assembly: coordination between small RNAs and argonaute proteins. Biochim. Biophys. Acta. 1859(1), 71–81 (2016). [DOI] [PubMed] [Google Scholar]
- 37.Nilsen TW. Mechanisms of miR-mediated gene regulation in animal cells. Trends Genet. 23(5), 243–249 (2007). [DOI] [PubMed] [Google Scholar]
- 38.Huntzinger E, Izaurralde E. Gene silencing by miRs: contributions of translational repression and mRNA decay. Nat. Rev. Genet. 12(2), 99–110 (2011). [DOI] [PubMed] [Google Scholar]
- 39.Seok H, Ham J, Jang E-S et al. MiR target recognition: insights from transcriptome-wide non-canonical interactions. Mol. Cells. 39(5), 375–381 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Bottini S, Hamouda-Tekaya N, Mategot R et al. Post-transcriptional gene silencing mediated by miRNAs is controlled by nucleoplasmic Sfpq. Nat. Commun. 8(1), 1189 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Nishi K, Nishi A, Nagasawa T et al. Human TNRC6A is an Argonaute-navigator protein for miR-mediated gene silencing in the nucleus. RNA 19(1), 17–35 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Peng Y, Croce CM. The role of miRs in human cancer. Signal. Transduct. Targeted Ther. 1(1), 15004 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Calin GA, Dumitru CD, Shimizu M et al. Frequent deletions and down-regulation of micro-RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc. Natl. Acad. Sci. USA 99(24), 15524–9 (2002). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Shell S, Park S-M, Radjabi AR et al. Let-7 expression defines two differentiation stages of cancer. Proc. Natl. Acad. Sci. USA 104(27), 11400–5 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Balzeau J, Menzes MR, Cao S et al. The LIN28/let-7 pathway in cancer. Front. Genet. 8, 31 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Chen Y, Xie C, Zheng X et al. LIN28/let-7/PD-L1 pathway as a target for cancer immunotherapy. Cancer Immunol. Res. 7(3), 487–497 (2019). [DOI] [PubMed] [Google Scholar]
- 47.He L, He X, Lim LP et al. A miRNA component of the p53 tumor suppressor network. Nature. 447(7148), 1130–113 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Raver-Shapira N, Marciano E, Neuru E et al. Transcriptional activation of miR-34a contributes to p53-mediated apoptosis. Mol Cell. 26(5), 731–43 (2007). [DOI] [PubMed] [Google Scholar]
- 49.Li XJ, Ren ZJ, Tang JH. MiR-34a: a potential therapeutic target in human cancer. Cell Death Dis. 5(7), e1327 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Hermeking H. The miR-34 family in cancer and apoptosis. Cell Death Differ. 17(2), 193–9 (2010). [DOI] [PubMed] [Google Scholar]
- 51.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. 10(5), 593–601 (2008). [DOI] [PubMed] [Google Scholar]
- 52.Pecot CV, Rupaimoole R, Yang D et al. Tumour angiogenesis regulation by the miR-200 family. Nat. Commun. 4, 2427 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.He L, Thomson JM, Hermann MT et al. A miR polycistron as a potential human oncogene. Nature. 435(7043), 828–33 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Mogilyansky E, Rigoutsos I. The miR-17/92 cluster: a comprehensive update on its genomics, genetics, functions and increasingly important and numerous roles in health and disease. Cell Death Differ. 20(12), 1603–1614 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Fuziwara CS, Kimura ET. Insights into regulation of the miR-17-92 cluster of miRNAs in cancer. Front. Med. 2, 64 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Kuo G, Wu C-Y, Yang H-Y. MiR-17-92 cluster and immunity. J. Formos. Med. Assoc. 118(1, Part 1), 2–6 (2019). [DOI] [PubMed] [Google Scholar]
- 57.Feng Y-H, Tsao C-J. Emerging role of miR-21 in cancer (review). Biomed. Rep. 5(4), 395–402 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Asangani IA, Rasheed SAK, Nikolova DA et al. MiR-21 (miR-21) post-transcriptionally downregulates tumor suppressor Pdcd4 and stimulates invasion, intravasation, and metastasis in colorectal cancer. Oncogene. 27(15), 2128–36 (2008). [DOI] [PubMed] [Google Scholar]
- 59.Shen Z, Xu X, Lv L et al. miR-21 overexpression promotes esophageal squamous cell carcinoma invasion and migration by repressing tropomyosin 1. Gastroenterol. Res. Pract. 2020, 6478653 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Meng F, Henson R, Wehbe-Janek H et al. MiR-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer. Gastroenterology. 133(2), 647–658 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Papagiannakopoulos T, Shapiro A, Kosik KS. MiR-21 targets a network of key tumor-suppressive pathways in glioblastoma cells. Cancer Res. 68(19), 8164–72 (2008). [DOI] [PubMed] [Google Scholar]
- 62.Sahraei M, Chaube B, Liu Y et al. Suppressing miR-21 activity in TAMs promotes an antitumor immune response. J. Clin. Invest. 129(12), 5518–5536 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Mattiske S, Suetani RJ, Neilsen PM, Callen DF. The oncogenic role of miR-155 in breast cancer. Cancer Epidemiol. Biomarkers Prev. 21(8), 1236–43 (2012). [DOI] [PubMed] [Google Scholar]
- 64.Bayraktar R, Van Roosbroeck K. miR-155 in cancer drug resistance and as target for miR-based therapeutics. Cancer Metastasis Rev. 37(1), 33–44 (2018). [DOI] [PubMed] [Google Scholar]
- 65.Pan Z, Tian Y, Niu G et al. Role of miRNAs in remodeling the tumor microenvironment (review). Int. J. Oncol. 56(2), 407–416 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Azizi M, Othman I, Naidu R. The role of microRNAs in lung cancer metabolism. Cancers (Basel). 13(7), 1716 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Daoud AZ, Mullholland EJ, Cole G, McCarthy HO. MiRs in pancreatic cancer: biomarkers, prognostic, and therapeutic modulators. BMC Cancer. 19(1), 1130 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Morrison C. Immuno-oncologists eye up macrophage targets. Nat. Rev. Drug Discov. 15(6), 373–374 (2016). [DOI] [PubMed] [Google Scholar]
- 69.Azizi E, Carr AJ, Plitas G et al. Single-cell map of diverse immune phenotypes in the breast tumor microenvironment. Cell. 174(5), 1293–1308.e36 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Mills CD, Kincaid K, Alt JM et al. M-1/M-2 macrophages and the Th1/Th2 paradigm. J. Immunol. 164(12), 616 (2000). [DOI] [PubMed] [Google Scholar]
- 71.Cai X, Yin Y, Li N et al. Repolarization of TAMs to proinflammatory M1 macrophages by miR-155. J. Mol. Cell. Biol. 4(5), 341–343 (2012). [DOI] [PubMed] [Google Scholar]
- 72.Martinez-Nunez RT, Louafi F, Sanchez-Elsner T. The interleukin 13 (IL-13) pathway in human macrophages is modulated by miR-155 via direct targeting of interleukin 13 receptor. J. Biol Chem. 286(3), 1786–1794 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Van Roosbroeck K, Fanini F, Setoyama T et al. Combining anti-miR-155 with chemotherapy for the treatment of lung cancers. Clin. Cancer Res. 23(11), 2891–2904 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Wang Z, Xu L, Hu Y et al. miRNA let-7b modulates macrophage polarization and enhances TAMs to promote angiogenesis and mobility in prostate cancer. Sci. Rep. 6(1), 25602 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Lin R, Chen L, Chen G et al. Targeting miR-23a in CD8+ cytotoxic T lymphocytes prevents tumor-dependent immunosuppression. J. Clin. Invest. 124(12), 5352–5367 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Dudda JC, Salaun B, Ji Y et al. MicroRNA-155 is required for effector CD8+ T cell responses to virus infection and cancer. Immunity 38(4), 742–753 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Cortez MA, Ivan C, Valdecanas D et al. PDL1 regulation by p53 via miR-34. J. Natl. Cancer Inst. 108(1), (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Chen L, Gibbons DL, Goswami S et al. Metastasis is regulated via microRNA-200/ZEB1 axis control of tumour cell PD-L1 expression and intratumoral immunosuppression. Nat. Commun. 5(1), 1–12 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Ge H, Wang L, Chen W, Wang L. Mechanism of miR-760 reversing lung cancer immune escape by downregulating IDO1 and eliminating regulatory T cells based on mathematical biology. Comput. Math. Methods Med. (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Parayath NN, Parikh A, Amiji MM. Repolarization of TAMs in a genetically engineered non-small-cell lung cancer model by intraperitoneal administration of HA-based nanoparticles encapsulating miR-125b. Nano Lett. 18(6), 3571–3579 (2019). [DOI] [PubMed] [Google Scholar]; • This study highlights the use of miRNA-125b to reprogram tumor-associated macrophages in a lung cancer mice model.
- 81.Cioffi M, Trabulo SM, Vallespinos M et al. The miR-25-93-106b cluster regulates tumor metastasis and immune evasion via modulation of CXCL12 and PD-L1. Oncotarget 8(13), 21609 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Abdelrahman MM, Fawzy IO, Bassiouni AA et al. Enhancing NK cell cytotoxicity by miR-182 in hepatocellular carcinoma. Hum. Immunol. 77(8), 667–673 (2016). [DOI] [PubMed] [Google Scholar]
- 83.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. 7(1), 1–13 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Parayath NN, Gandham SK, Leslie F, Amiji MM. Improved antitumor efficacy of paclitaxel in combination with microRNA-125b-based tumor-associated macrophage repolarization in epithelial ovarian cancer. Cancer Lett. 461, 1–9 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]; • This study highlights the use of miRNA-125b to reprogram tumor associated macrophages in an ovarian cancer mice model.
- 85.Zhao L, Yu H, Yi S et al. The tumor suppressor miR-138-5p targets PD-L1 in colorectal cancer. Oncotarget 7(29), 45370 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Dou R, Nishihara R, Cao Y et al. MicroRNA let-7, T cells, and patient survival in colorectal cancer microRNA let-7, T cells, and colorectal cancer. Cancer Immunol. Res. 4(11), 927–935 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Fontana L, Pelosi E, Greco P et al. MicroRNAs 17-5p-20a-106a control monocytopoiesis through AML1 targeting and M-CSF receptor upregulation. Nat. Cell Biol. 9(7), 775–787 (2007). [DOI] [PubMed] [Google Scholar]
- 88.Zhao Y, Yu Z, Ma R et al. lncRNA-Xist/miR-101-3p/KLF6/C/EBPα axis promotes TAM polarization to regulate cancer cell proliferation and migration. Mol. Ther. Nucl. Acids 23, 536–551 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Tian Y, Li L, Lin G et al. lncRNA SNHG14 promotes oncogenesis and immune evasion in diffuse large-B-cell lymphoma by sequestering miR-152-3p. Leuk. Lymphoma 62(7), 1574–1584 (2021). [DOI] [PubMed] [Google Scholar]
- 90.Vilming Elgaaen B, Olstad OK, Haug KBF et al. Global miR expression analysis of serous and clear cell ovarian carcinomas identifies differentially expressed miRNAs including miR-200c-3p as a prognostic marker. BMC Cancer. 14(1), 80 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Bjaanaes MM, Halvorsen AR, Solberg S et al. Unique miR-profiles in EGFR-mutated lung adenocarcinomas. Int. J. Cancer 135(8), 1812–21 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Ji D, Qiao M, Yao Y et al. Serum-based miRNA signature predicts relapse and therapeutic outcome of adjuvant chemotherapy in colorectal cancer patients. EBio. Med. 35, 189–197 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.van Jaarsveld MTM, Helleman J, Berns EMJJ et al. MiRs in ovarian cancer biology and therapy resistance. Int. J. Biochem. Cell Biol. 42(8), 1282–1290 (2010). [DOI] [PubMed] [Google Scholar]
- 94.Biamonte F, Santamaria G, Sacco A et al. MicroRNA let-7g acts as tumor suppressor and predictive biomarker for chemoresistance in human epithelial ovarian cancer. Sci. Rep. 9(1), 5668 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Blower PE, Verducci JS, Lin S et al. MicroRNA expression profiles for the NCI-60 cancer cell panel. Mol. Cancer Ther. 6(5), 1483–1491 (2007). [DOI] [PubMed] [Google Scholar]
- 96.Giovannetti E, Erozenci A, Smit J et al. Molecular mechanisms underlying the role of microRNAs (miRNAs) in anticancer drug resistance and implications for clinical practice. Criti. Rev. Oncol. Hematol. 81(2), 103–122 (2012). [DOI] [PubMed] [Google Scholar]
- 97.Au Yeung CL, Co NN, Tsuruga T et al. Exosomal transfer of stroma-derived miR21 confers PTX resistance in ovarian cancer cells through targeting APAF1. Nat. Commun. 7, 11150 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Gandham SK, Rao M, Shah A et al. Combination of clinically relevant microRNA-based cellular reprogramming with paclitaxel enhances therapeutic efficacy in a relapsed and multidrug-resistant model of epithelial ovarian cancer. Mol. Ther. Oncolytics 25, 57–68 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]; • This study highlights the use of miRNA-let7b in combination with paclitaxel in mice model of relapsed epithelial ovarian cancer.
- 99.Chirshev E, Oberg KC, Ioffe YJ, Unternaehrer JJ. Let-7 as biomarker, prognostic indicator, and therapy for precision medicine in cancer. Clin. Transl. Med. 8, 24 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Peng J, Mo R, Ma J, Fan J. let-7b and let-7c are determinants of intrinsic chemoresistance in renal cell carcinoma. World J. Surg. Oncol. 13, 175 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Biamonte F, Santamaria G, Sacco A et al. MicroRNA let-7g acts as tumor suppressor and predictive biomarker for chemoresistance in human epithelial ovarian cancer. Sci. Rep. 9, 5668 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Ferneza S, Fetsych M, Shuliak R et al. Clinical significance of microRNA-200 and let-7 families expression assessment in patients with ovarian cancer. Ecancermedicalscience 15 1249 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Acunzo M, Romano G, Palmieri D et al. Cross-talk between MET and EGFR in non-small-cell lung cancer involves miR-27a and Sprouty2. Proc. Natl. Acad. Sci. USA 110, 8573–8578 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Wu J, Sun Z, Sun H, Li Y. MicroRNA-27a promotes tumorigenesis via targeting AKT in triple negative breast cancer. Mol. Med. Rep. 17, 562–570 (2018). [DOI] [PubMed] [Google Scholar]
- 105.Li Z, Hu S, Wang J et al. MiR-27a modulates MDR1/P-glycoprotein expression by targeting HIPK2 in human ovarian cancer cells. Gynecol. Oncol. 119, 125–130 (2010). [DOI] [PubMed] [Google Scholar]
- 106.Ling S, Ruiqin M, Guohong Z, Ying W. Expression and prognostic significance of microRNA-451 in human epithelial ovarian cancer. Eur. J. Gynaecol. Oncol. 36, 463–468 (2015). [PubMed] [Google Scholar]
- 107.Zhu H, Wu H, Liu X et al. Role of microRNA miR-27a and miR-451 in the regulation of MDR1/P-glycoprotein expression in human cancer cells. Biochem. Pharmacol. 76, 582–588 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Bai H, Wu S. miR-451: a novel biomarker and potential therapeutic target for cancer. Onco Targets Ther. 12, 11069–11082 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Leskelä S, Leandro-García LJ, Mendiola M et al. The miR-200 family controls beta-tubulin III expression and is associated with paclitaxel-based treatment response and progression-free survival in ovarian cancer patients. Endocr. Relat. Cancer 18, 85–95 (2011). [DOI] [PubMed] [Google Scholar]
- 110.Liu J, Zhang X, Huang Y et al. miR-200b and miR-200c co-contribute to the cisplatin sensitivity of ovarian cancer cells by targeting DNA methyltransferases. Oncol. Lett. 17, 1453–1460 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 111.Wu Y, Xiao Y, Ding X et al. A miR-200b/200c/429-binding site polymorphism in the 3′ untranslated region of the AP-2α gene is associated with cisplatin resistance. PLOS ONE 6, e29043 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Koutsaki M, Libra M, Spandidos DA, Zaravinos A. The miR-200 family in ovarian cancer. Oncotarget 8, 66629–66640 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Mizuno K, Mataki H, Arai T et al. The microRNA expression signature of small-cell lung cancer: tumor suppressors of miR-27a-5p and miR-34b-3p and their targeted oncogenes. J. Hum. Genet. 62, 671–678 (2017). [DOI] [PubMed] [Google Scholar]
- 114.Tafsiri E, Darbouy M, Shadmehr MB et al. Expression of miRNAs in non-small-cell lung carcinomas and their association with clinicopathological features. Tumor Biol. 36, 1603–1612 (2015). [DOI] [PubMed] [Google Scholar]
- 115.Stahlhut C, Slack FJ. Combinatorial action of microRNAs let-7 and miR-34 effectively synergizes with erlotinib to suppress non-small-cell lung cancer cell proliferation. Cell Cycle 14, 2171–2180 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Trivedi M, Singh A, Talekar M et al. MicroRNA-34a encapsulated in hyaluronic acid nanoparticles induces epigenetic changes with altered mitochondrial bioenergetics and apoptosis in non-small-cell lung cancer cells. Sci. Rep. 7, 3636 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Jiang Z, Yin J, Fu W et al. MiRNA 17 family regulates cisplatin-resistant and metastasis by targeting TGFbetaR2 in NSCLC. PLOS ONE 9, e94639 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Zhang X, Li Y, Qi P, Ma Z. Biology of miR-17-92 cluster and its progress in lung cancer. Int. J. Med. Sci. 15, 1443–1448 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Chatterjee A, Chattopadhyay D, Chakrabarti G. miR-17-5p downregulation contributes to paclitaxel resistance of lung cancer cells through altering beclin1 expression. PLOS ONE 9, e95716 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Jeon YJ, Middleton J, Kim T et al. A set of NF-κB-regulated microRNAs induces acquired TRAIL resistance in lung cancer. Proc. Natl. Acad. Sci. USA 112, E3355–3364 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Elhassan MO, Christie J, Duxbury MS. Homo sapiens systemic RNA interference-defective-1 transmembrane family member 1 (SIDT1) protein mediates contact-dependent small RNA transfer and microRNA-21-driven chemoresistance. J. Biol. Chem. 287, 5267–5277 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Garofalo M, Romano G, Di Leva G et al. EGFR and MET receptor tyrosine kinase-altered microRNA expression induces tumorigenesis and gefitinib resistance in lung cancers. Nat. Med. 18, 74–82 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 123.Bockhorn J, Dalton R, Nwachukwu C et al. MicroRNA-30c inhibits human breast tumour chemotherapy resistance by regulating TWF1 and IL-11. Nat. Commun. 4, 1393 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Yang SJ, Yang SY, Wang DD et al. The miR-30 family: versatile players in breast cancer. Tumor Biol. 39, 1010428317692204 (2017). [DOI] [PubMed] [Google Scholar]
- 125.Abedi N, Mohammadi-Yeganeh S, Koochaki A et al. miR-141 as potential suppressor of β-catenin in breast cancer. Tumor Biol. 36, 9895–9901 (2015). [DOI] [PubMed] [Google Scholar]
- 126.Yu Z, Xu Z, Disante G et al. miR-17/20 sensitization of breast cancer cells to chemotherapy-induced apoptosis requires Akt1. Oncotarget 5, 1083–1090 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Ao X, Nie P, Wu B et al. Decreased expression of microRNA-17 and microRNA-20b promotes breast cancer resistance to taxol therapy by upregulation of NCOA3. Cell Death Dis. 7, e2463–e2463 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Kong W, He L, Coppola M et al. MicroRNA-155 regulates cell survival, growth, and chemosensitivity by targeting FOXO3a in breast cancer. J. Biol. Chem. 285, 17869–17879 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 129.Gasparini P, Lovat F, Fassan M et al. Protective role of miR-155 in breast cancer through RAD51 targeting impairs homologous recombination after irradiation. Proc. Natl. Acad. Sci. USA 111, 4536–4541 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Wang W, Zhao L, Wei X et al. MicroRNA-320a promotes 5-FU resistance in human pancreatic cancer cells. Sci. Rep. 6, 27641 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Iwagami Y, Eguchi H, Nagano H et al. miR-320c regulates gemcitabine-resistance in pancreatic cancer via SMARCC1. Br. J. Cancer 109, 502–511 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Hasegawa S, Eguchi H, Nagano H et al. MicroRNA-1246 expression associated with CCNG2-mediated chemoresistance and stemness in pancreatic cancer. Br. J. Cancer 111, 1572–1580 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Amponsah PS, Fan P, Bauer N et al. microRNA-210 overexpression inhibits tumor growth and potentially reverses gemcitabine resistance in pancreatic cancer. Cancer Lett. 388, 107–117 (2017). [DOI] [PubMed] [Google Scholar]
- 134.Zhan T, Huang X, Tian X et al. Downregulation of microRNA-455-3p links to proliferation and drug resistance of pancreatic cancer cells via targeting TAZ. Mol. Ther. Nucl. Acids 10, 215–226 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Fernandez-Piñeiro I, Badiola I, Sanchez A. Nanocarriers for miR delivery in cancer medicine. Biotech. Adv. 35(3), 350–360 (2017). [DOI] [PubMed] [Google Scholar]
- 136.Gupta A, Andresen JL, Manan RS, Langer R. Nucleic acid delivery for therapeutic applications. Adv. Drug Deliv. Rev. 178, 113834 (2021). [DOI] [PubMed] [Google Scholar]
- 137.Elsabahy M, Nazarali A, Foldvari M. Non-viral nucleic acid delivery: key challenges and future directions. Curr. Drug Deliv. 8(3), 235–244 (2011). [DOI] [PubMed] [Google Scholar]
- 138.Sinegra AJ, Evangelopoulos M, Park J et al. Lipid nanoparticle spherical nucleic acids for intracellular DNA and RNA delivery. Nano Lett. 21(15), 6584–6591 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Durymanov M, Reineke J. Non-viral delivery of nucleic acids: insight into mechanisms of overcoming intracellular barriers. Front. Pharm. 9, 971 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Shi J, Ma Y, Zhu J et al. A review on electroporation-based intracellular delivery. Molecules. 23(11), 3044 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Smith SA, Selby LI, Johnston AP, Such GK. The endosomal escape of nanoparticles: toward more efficient cellular delivery. Bioconjug. Chem. 30(2), 263–272 (2018). [DOI] [PubMed] [Google Scholar]
- 142.Bhattarai SR, Muthuswamy E, Wani A et al. Enhanced gene and siRNA delivery by polycation-modified mesoporous silica nanoparticles loaded with chloroquine. Pharm. Res. 27(12), 2556–2568 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Nomoto T, Fukushima S, Kumagai M et al. Three-layered polyplex micelle as a multifunctional nanocarrier platform for light-induced systemic gene transfer. Nat. Commun. 5(1), 1–10 (2014). [DOI] [PubMed] [Google Scholar]
- 144.Torchilin VP. Tat peptide-mediated intracellular delivery of pharmaceutical nanocarriers. Adv. Drug Deliv. Rev. 60(4–5), 548–558 (2008). [DOI] [PubMed] [Google Scholar]
- 145.De la Fuente JM, Berry CC. Tat peptide as an efficient molecule to translocate gold nanoparticles into the cell nucleus. Bioconjug. Chem. 16(5), 1176–1180 (2005). [DOI] [PubMed] [Google Scholar]
- 146.Plaza-Ga I, Manzaneda-González V, Kisovec M et al. pH-triggered endosomal escape of pore-forming Listeriolysin O toxin-coated gold nanoparticles. J. Nanobiotech. 17(1), 1–10 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Rausch JM, Marks JR, Rathinakumar R, Wimley WC. β-sheet pore-forming peptides selected from a rational combinatorial library: mechanism of pore formation in lipid vesicles and activity in biological membranes. Biochem. 46(43), 12124–12139 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Cheng Y, Yumul RC, Pun SH. Virus-inspired polymer for efficient in vitro and in vivo gene delivery. Angewandte Chemie Int. Edition. 55(39), 12013–12017 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Cho YW, Kim J-D, Park K. Polycation gene delivery systems: escape from endosomes to cytosol. J. Pharm. Pharmacol. 55(6), 721–734 (2003). [DOI] [PubMed] [Google Scholar]
- 150.Wang S. pH-responsive amphiphilic carboxylate polymers: design and potential for endosomal escape. Front. Chem. 9, 145 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Sun X, Zhang N. Cationic polymer optimization for efficient gene delivery. Mini Rev. Med. Chem. 10(2), 108–125 (2010). [DOI] [PubMed] [Google Scholar]
- 152.Vermeulen LM, De Smedt SC, Remaut K, Braeckmans K. The proton sponge hypothesis: fable or fact? Eur. J. Pharm. Biopharm. 129, 184–190 (2018). [DOI] [PubMed] [Google Scholar]
- 153.Ganesh S, Iyer AK, Morrissey DV, Amiji MM. HA based self-assembling nanosystems for CD44 target mediated siRNA delivery to solid tumors. Biomaterials. 34(13), 3489–3502 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Rao NV, Rho JG, Um W et al. Hyaluronic acid nanoparticles as nanomedicine for treatment of inflammatory diseases. Pharmaceutics. 12(10), 931 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Lee GY, Kim JH, Choi KY et al. Hyaluronic acid nanoparticles for active targeting atherosclerosis. Biomaterials. 53, 341–348 (2015). [DOI] [PubMed] [Google Scholar]
- 156.Choi KY, Chung H, Min KH et al. Self-assembled hyaluronic acid nanoparticles for active tumor targeting. Biomaterials. 31(1), 106–114 (2010). [DOI] [PubMed] [Google Scholar]
- 157.Yu M, Jambhrunkar S, Thorn P et al. HA modified mesoporous silica nanoparticles for targeted drug delivery to CD44-overexpressing cancer cells. Nanoscale 5(1), 178–183 (2013). [DOI] [PubMed] [Google Scholar]
- 158.Su M-J, Aldawsari H, Amiji M. Pancreatic cancer cell exosome-mediated macrophage reprogramming and the role of miRNAs 155 and 125b2 transfection using nanoparticle delivery systems. Sci. Rep. 6(1), 1–15 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Wang Y, Zeng G, Jiang Y. The emerging roles of miR-125b in cancers. Cancer Mang. Res. 12, 1079 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Muramatsu F, Kidoya H, Naito H et al. microRNA-125b inhibits tube formation of blood vessels through translational suppression of VE-cadherin. Oncogene. 32(4), 414–421 (2013). [DOI] [PubMed] [Google Scholar]
- 161.Smits M, Wurdinger T, van het Hof B et al. Myc-associated zinc finger protein (MAZ) is regulated by miR-125b and mediates VEGF-induced angiogenesis in glioblastoma. FASEB J. 26(6), 2639–2647 (2012). [DOI] [PubMed] [Google Scholar]
- 162.Chaudhuri AA, So AY-L, Sinha N et al. MiR-125b potentiates macrophage activation. J. Immunol. 187(10), 5062–5068 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.El Chartouni C, Schwarzfischer L, Rehli M. Interleukin-4 induced interferon regulatory factor (Irf) 4 participates in the regulation of alternative macrophage priming. Immunobiology 215(9–10), 821–825 (2010). [DOI] [PubMed] [Google Scholar]
- 164.Vu LT, Peng B, Zhang DX et al. Tumor-secreted extracellular vesicles promote the activation of cancer-associated fibroblasts via the transfer of microRNA-125b. J. Extracell. Vesicles. 8(1), 1599680 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Parayath NN, Hong BV, Mackenzie GG, Amiji MM. Hyaluronic acid nanoparticle-encapsulated microRNA-125b repolarizes tumor-associated macrophages in pancreatic cancer. Nanomedicine. 16(25), 2291–2303 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Baldari S, Di Rocco G, Magenta A et al. Extracellular vesicles-encapsulated microRNA-125b produced in genetically modified mesenchymal stromal cells inhibits hepatocellular carcinoma cell proliferation. Cells. 8(12), 1560 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Talekar M, Trivedi M, Shah P et al. Combination wt-p53 and microRNA-125b transfection in a genetically engineered lung cancer model using dual CD44/EGFR-targeting nanoparticles. Mol. Ther. 24(4), 759–769 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Kawamura K, Komohara Y, Takaishi K et al. Detection of M2 macrophages and colony-stimulating factor 1 expression in serous and mucinous ovarian epithelial tumors. Pathol. Int. 59(5), 300–305 (2009). [DOI] [PubMed] [Google Scholar]
- 169.Heusinkveld M, van Der Burg SH. Identification and manipulation of tumor associated macrophages in human cancers. J. Transl. Med. 9(1), 1–14 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Mielgo A, Schmid MC. Impact of tumour-associated macrophages in pancreatic cancer. BMB Rep. 46(3), 131 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Liu C-Y, Xu J-Y, Shi X-Y et al. M2-polarized tumor-associated macrophages promoted epithelial-mesenchymal transition in pancreatic cancer cells, partially through TLR4/IL-10 signaling pathway. Lab. Invest. 93(7), 844–854 (2013). [DOI] [PubMed] [Google Scholar]
- 172.Büssing I, Slack FJ, Grosshans H. Let-7 microRNAs in development, stem cells and cancer. Trends Mol. Med. 14(9), 400–9 (2008). [DOI] [PubMed] [Google Scholar]
- 173.Lee H, Han S, Kwon CS et al. Biogenesis, and regulation of the let-7 miRNAs and their functional implications. Protein Cell. 7(2), 100–13 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Chirshev E, Oberg KC, Ioffe YJ et al. Let-7 as biomarker, prognostic indicator, and therapy for precision medicine in cancer. Clin. Transl. Med. 8(1), 24 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Calatayud D, Dehlendorff C, Boisen MK et al. Tissue microRNA profiles as diagnostic and prognostic biomarkers in patients with resectable pancreatic ductal adenocarcinoma and periampullary cancers. Biomark. Res. 5, 8 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Chen KJ, Hou Y, Wang K et al. Reexpression of Let-7g microRNA inhibits the proliferation and migration via K-Ras/HMGA2/snail axis in hepatocellular carcinoma. Biomed. Res. Int. 2014, 742417 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Tang Z, Ow GS, Thiery JP et al. Meta-analysis of the transcriptome reveals let-7b as an unfavorable prognostic biomarker and predicts molecular and clinical subclasses in high-grade serous ovarian carcinoma. Int. J. Cancer. 134(2), 306–318 (2014). [DOI] [PubMed] [Google Scholar]
- 178.Tang R, Yang C, Ma X et al. MiR-let-7a inhibits cell proliferation, migration, and invasion by down-regulating PKM2 in gastric cancer. Oncotarget. 7(5), 5972–5984 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Peng F, Li T-T, Wan K-L et al. H19/let-7/LIN28 reciprocal negative regulatory circuit promotes breast cancer stem cell maintenance. Cell Death Dis. 8(1), e2569 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.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. 64(11), 3753–3756 (2004). [DOI] [PubMed] [Google Scholar]
- 181.Türk D, Szakács G. Relevance of multidrug resistance in the age of targeted therapy. Curr. Opin. Drug Discov. Devel. 12(2), 246–252 (2009). [PubMed] [Google Scholar]
- 182.Boyerinas B, Park S-M, Murmann AE et al. Let-7 modulates acquired resistance of ovarian cancer to Taxanes via IMP-1-mediated stabilization of multidrug resistance 1. Int. J. Cancer 130(8), 1787–1797 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Dai X, Fan W, Wang Y et al. Combined delivery of let-7b microRNA and PTX via biodegradable nanoassemblies for the treatment of KRAS mutant cancer. Mol. Pharm. 13(2), 520–533 (2016). [DOI] [PubMed] [Google Scholar]
- 184.Wu L, Nguyen LH, Zhou K et al. Precise let-7 expression levels balance organ regeneration against tumor suppression. eLife. 4, e09431 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Esquela-Kerscher A, Trang P, Wiggins JF et al. The let-7 microRNA reduces tumor growth in mouse models of lung cancer. Cell Cycle. 7(6), 759–764 (2008). [DOI] [PubMed] [Google Scholar]
