Abstract
Tumor suppressor genes (TSGs) are frequently downregulated in cancer, leading to dysregulation of the pathways that they control. The continuum model of tumor suppression suggests that even subtle changes in TSG expression, for example, driven by epigenetic modifications or copy number alterations, can lead to a loss of gene function and a phenotypic effect. This approach to exploring tumor suppression provides opportunities for alternative therapies that may be able to restore TSG expression toward normal levels, such as oligonucleotide therapies. Oligonucleotide therapies involve the administration of exogenous nucleic acids to modulate the expression of specific endogenous genes. This review focuses on two types of activating oligonucleotide therapies, small-activating RNAs and synthetic mRNAs, as novel methods to increase the expression of TSGs in cancer.
Keywords: MT: oligonucleotides: therapies and applications, oligonucleotide, cancer, tumor suppressor gene, saRNA, mRNA
Graphical abstract

TSGs are genes that are commonly downregulated in cancer. An emerging classification of drugs, called oligonucleotide therapies, have the ability to upregulate or restore the expression of TSGs. This review summarizes saRNAs and synthetic mRNAs and their capability to modulate TSG expression.
Introduction
What are tumor suppressor genes?
Tumor suppressor genes (TSGs) are a category of genes that serve to keep cell growth tightly regulated, such that a cell will only divide when absolutely necessary and in response to the appropriate external signals, such as growth factors. In addition to controlling proliferation, TSGs are also involved in preventing cells from migrating to, and invading, other tissues, as well as stimulating cells to undergo apoptosis when they encounter a cellular stress, such as DNA damage. If the latter is left unchecked, this could result in the introduction of mutations and dysregulation of the cell cycle. When TSG function is lost, this can result in critical cellular processes becoming dysregulated and cells may proliferate uncontrollably, fail to initiate apoptosis in response to damage, or start to invade through the basement membrane and metastasize to a different part of the body.1 TSGs are, therefore, an important group of genes in the context of cancer pathogenesis and therapy.
Although TSGs are a vast group of genes, they can be further categorized according to their function and the pathways which they control, see below and Table 1. TSGs have traditionally been labeled as being homozygous recessive in terms of their role in promoting carcinogenesis, meaning that both alleles of the TSG have to exhibit loss of function to result in loss of protein activity and promotion of cell proliferation and tumorigenesis. The “two-hit hypothesis” was first defined during the analysis of retinoblastoma (RB), a cancer of the eye in children.38 Knudson38 reported that familial RB was dominantly inherited. Indeed, it was shown that children inherit a mutation in the RB1 gene, which encodes the RB protein (although the actual disease-causing gene was not identified at the time), from one parent, which predisposes them to developing RB. A secondary mutation in the other RB1 allele is acquired somatically as the eye develops, meaning that both alleles of the gene are mutated, leading to a loss of function. This means that there is no functional RB protein in the retinoblast cell; therefore, it cannot exert its function as a repressor of cell cycle progression. The deficient cells are, therefore, able to progress through the cell cycle, even in the absence of the appropriate growth factor signal. This results in the initiation of tumorigenesis.
Table 1.
Summary of studies investigating upregulation of TSGs using saRNAs in cancers
| Classification | Gene targeted for upregulation using saRNAs | Protein encoded by gene | Function of tumor suppressor | Cancer(s) in which upregulation has been investigated | Stage of development | Reference(s) |
|---|---|---|---|---|---|---|
| Cell-cycle arrest | CDKN1A | P21 | Activated by p53 in response to DNA damage, CDKN1A is a cyclin-dependent kinase inhibitor which prevents arrests cells in G1/S-phase of the cell cycle. | Colorectal cancer, HCC, Prostate cancer, Bladder cancer |
In vivo (Colorectal) In vivo (Prostate) In vitro (HCC) In vitro (Bladder) |
Li et al., Wu et al., Kang et al., Place et al., Wang et al., Kosaka et al., Zhang et al., Wu et al., Chen et al. (Refs.2,3,4,5,6,7,8,9,10) |
| TP53 | P53 | Commonly referred to as the guardian of the genome, p53 is activated in response to cellular stress to induce cell-cycle arrest, apoptosis, DNA repair and cell senescence. | Bladder cancer, Pheochromocytoma | In vivo (Bladder)In vivo (Pheochromocytoma) | Wang et al., Lin et al. (Refs.11,12) | |
| VHL | Von Hippel-Lindau protein | Forms part of an E3 ubiquitin ligase complex, involved in regulating angiogenesis, cell cycle progression and apoptosis. | Renal cell carcinoma | In vitro | Kang et al. (Ref.13) | |
| Inhibit pro-proliferative signaling pathways | CEBPA | CCAAT/enhancer-binding protein alpha | Transcription factor which prevents over-proliferation of mature hepatocytes and helps maintain healthy hepatocyte function. | HCC PDAC |
Phase II clinical trials (HCC) In vivo (PDAC) | Huan et al., Sarker et al., Hashimoto et al., Yoon et al., Reebye et al.(Refs.14,15,16,17,18,19) |
| INTS6 | Integrator complex subunit 6 | Able to interact with RNA polymerase II and is involved in the processing of small nuclear RNAs. Can downregulate the pro-proliferative Wnt signaling pathway. | Prostate cancer | In vitro | Chen et al. (Ref.20) | |
| HIC1 | Hypermethylated in cancer 1 | Transcriptional repressor of pro-proliferative genes such as CCND1. | Gastric cancer | In vitro | Pan et al. (Ref.21) | |
| PTEN | Phosphatase and tensin homologue on chromosome 10 | Dephosphorylates PIP3 to PIP2, preventing activation of AKT/PKB and its downstream effects including proliferation and cell survival. | Lung cancer | In vitro | Li et al. (Ref.22) | |
| Maintain cell morphology and motility | CDH1 | E-cadherin | Forms adherens junctions between adjacent cells, preventing epithelial-to-mesenchymal transition. Sequesters β-catenin to the membrane, blocking the pro-proliferative Wnt signaling pathway. | Bladder cancer, Prostate cancer, Renal cell carcinoma, Breast cancer |
In vitro (bladder) In vitro (prostate) In vitro (renal) In vivo (breast) |
Li et al., Dai et al., Wu et al., Li et al., Junxia et al., Mao et al. (Refs.2,23,24,25,26,27) |
| VEZT | Vezatin, adherens junctions transmembrane protein | Transmembrane protein involved in maintaining adherens’ junctions. | Gastric cancer | In vitro | Xie et al. (Ref.28) | |
| DPYSL3 | Dihydropyrimidinase like 3 | Involved in semaphorin/collapsin-induced signaling, which regulate cell morphology and motility. Prevents cancer cell metastasis. | Prostate cancer | In vivo | Li et al. (Ref.29) | |
| NKX3-1 | NK3 Homeobox 1 | Thought to be a prostate-specific tumor suppressor, involved in maintaining normal cellular phenotype. | Prostate cancer | In vivo | Ren et al. (Ref.30) | |
| Promote apoptosis | PAWR | Pro-apoptotic WT1 regulator | Stimulates apoptosis by activating the Fas-mediated apoptotic pathway and inhibiting pro-survival pathways. | Prostate cancer | In vitro | Yang et al. (Ref.31) |
| FHIT | Fragile histidine triad | Plays a role in promoting apoptosis by preventing the degradation of p53. | Endometrial cancer | In vitro | Zhu et al. (Ref. 32) | |
| Function unknown/unclear | DIRAS1 | Di-Ras1 | Member of the guanosine triphosphatase (GTPase) Ras superfamily, specific function is unclear. | Renal cell carcinoma | In vivo | Xu et al. (Ref.33) |
| KLF4 | Kruppel like factor 4 | A zinc-finger transcription factor. Can act as a tumor suppressor or oncogene dependent on the genetic context of the tissue in which it is expressed. | Colorectal cancer, Prostate cancer | In vitro | Zhou et al., Wang et al. (Refs.34,35) | |
| WT1 | Wilms’ tumor protein | Can act as a tumor suppressor or oncogene, depending on the cell type in which it is expressed, and level of expression. | HCC | In vitro | Qin et al. (Ref.36) | |
| MAS1 | Mas receptor 1 | Modulates the renin-angiotensin system to control the tumor microenvironment and inhibit cell migration. | Ovarian cancer, Breast cancer, Pancreatic cancer |
In vivo (ovarian) In vivo (breast) In vitro (pancreatic) |
Xiong et al. (Ref.37) |
CCND1, cyclin D1; CDH1, cadherin-1; CDKN1A, cyclin dependent kinase inhibitor 1A; HCC, hepatocellular carcinoma; PDAC, Pancreatic ductal adenocarcinoma; PIP2, phosphatidylinositol 4,5-bisphosphate; PIP3, phosphatidylinositol 3,4,5-trisphosphate.
However, many sporadic cancers (i.e., cancers where there are no inherited pre-disposing gene mutations) exhibit a loss of function in a single allele of a TSG, whereas the other allele seems to be normal. These instances led to the development of the concept of haploinsufficiency, which suggests that the loss of a single allele may be sufficient for a TSG to have decreased function and, hence, play a role in the development of a tumor.39
The continuum model of tumor suppression takes this concept a step further, and suggests that even subtle changes in the expression of a TSG can impact its function and tumor-suppressive activity.39 This model takes into consideration the fact that genes can be regulated in ways other than by mutation or allele loss, such as by epigenetic modifications, microRNAs and post-translational modifications. These changes can result in altered expression of the gene, but the gene itself is not mutated. This means that the transcription and translation of the gene produces a normal, functional protein, but the levels of the protein are lower in the tumor compared with normal, healthy tissue. The continuum model of tumor suppression, therefore, implies that even a slight upregulation in a TSG may recover some of its tumor-suppressive function, and hence stunt the growth, invasiveness, or malignancy of a tumor, depending on the molecular pathways that the TSG acts on.
An example of a TSG that does not follow Knudson’s two-hit hypothesis of tumor suppression is phosphatase and tensin homolog on chromosome 10 (PTEN). Studies have shown that subtle changes in the expression level of PTEN can result in the loss of its function and an increase in carcinogenesis in certain tissues. For example, a 20% decrease in the normal level of PTEN expression is sufficient to cause cancer in the breast,39,40 but is not sufficiently low enough to cause carcinogenesis in the liver, small intestine, pancreas, adrenal glands, and prostate.40 Furthermore, haploinsufficiency of PTEN is sufficient to accelerate prostate tumor progression in mice.41
TSGs in the clinic
It is now well recognized that cancer is a genetic disease, with the two major categories of genes involved in the initiation of cancer being oncogenes and TSGs. There are many drugs that have been developed that target overactive oncogenes, such as kinase inhibitors like sorafenib in hepatocellular carcinoma, and imatinib in chronic myeloid leukemia.42,43 However, small molecule drugs targeting underactive or mutated TSGs have been somewhat lacking.
There are some examples where this has been the case, such as the identification of thioemicarbazone family molecules in targeting mutant p53, allowing for zinc chelation and the restoration of the DNA-binding properties of the important transcription factor.44 Furthermore, compounds such as PhiKan083 allow for the restoration of normal function in p53 mutant cells harboring a Y220C or Y220S mutation by stabilizing the protein structure and preventing denaturation.45,46 However, no drugs directly targeting p53 have yet to make it through clinical trials to approval, indicating the difficulty in targeting TSGs, even after years of extensive research into the gene and protein’s structure and function.
A p53 activator that has entered clinical trials is APR-246, a quinuclidinone derivative that is able to rescue the ability of mutant p53 to interact with DNA by interacting with the cysteine residues within the protein to restore the wild-type conformation.47 APR-246 has completed phase I and II trials for multiple different cancer types and in combination with other anti-cancer therapies.48,49,50 The first-in-human trial showed that the drug does have some promising anti-tumour activity, indicated by increased apoptosis of circulating malignant cells in selected patients, regardless of TP53 mutation status.48 However, the active form of the drug, methylene quinuclidinone, to which APR-246 is converted once inside the body, is rapidly degraded under physiological conditions, which limits the effectiveness of APR-246 as a monotherapy.51
Another approach that has been used to circumvent underactive TSGs is the targeting of the downstream consequences of such downregulation. For example, the downregulation or loss of PTEN leads to overactivity of the AKT pathway, which provides an ideal target for small molecule drugs via inhibition of the kinase AKT.52 However, oligonucleotide therapies offer an advantage as they allow for the root of the issue (i.e., the underactive TSG) to be targeted directly.
In light of the critical role of TSG modulation in various forms of cancer, there is much interest in the potential to target specific TSGs as a novel therapeutic approach in patients. This review explores the potential of using oligonucleotide therapies to restore the function of TSGs in cancer (Figure 1), and the benefits and challenges of such an approach.
Figure 1.
An overview of the use of activating oligonucleotides to target TSGs in cancer
Synthetic mRNAs and saRNAs are two types of activating oligonucleotide which have the potential to upregulate TSGs and restore their function. There are challenges with the delivery of such oligonucleotides in vivo, and their mechanisms of action differ once inside their target cell. GalNAc, N-acetylgalactosamine.
What are oligonucleotide therapies?
Oligonucleotide therapies include inhibitory antisense oligonucleotides (ASOs) and short interfering RNAs (siRNAs), along with stimulatory small-activating RNAs (saRNAs) and synthetic, nucleoside-modified mRNAs. Another example of an activating oligonucleotide is plasmid DNA, which is an approach used in gene therapy. Plasmid DNA can be delivered to a host cell via both viral and non-viral methods, with viral-based delivery systems accounting for the majority of gene therapy approaches in clinical trials.53 However, such methods of delivery are associated with immunogenicity and the risk of insertional mutagenesis54,55 and are not discussed further in the current review. Broadly, oligonucleotide therapies involve the administration of exogenous nucleic acids to a cell to modulate expression of specific target genes. Oligonucleotides can interact with complementary sequences on RNA or DNA, depending on their mechanism of action, and they offer an advantage over small molecule drugs, which tend to work on protein targets and can lack specificity. Oligonucleotide therapies offer a more specific approach to modulating gene expression, which can minimize off-target effects and toxicity. Given that the exact sequence of the oligonucleotide is known, and the fact that these therapies interact with their target via complementary Watson-Crick base pairing, any off-target interactions can be predicted bioinformatically, and adverse effects that these interactions may cause can be forecasted and potentially circumvented.
An increased understanding of the roles of genes and other oligonucleotides in disease is leading to an expansion of opportunities for developing oligonucleotide therapies directed to previously undruggable targets. A particular focus of oligonucleotide therapy is oncology, with approximately one-quarter of oligonucleotide therapy candidates being developed to modulate targets in cancer.56 Given that TSGs are commonly downregulated in cancer, and small molecules have traditionally been used to inhibit, rather than stimulate, a target, there is the potential to restore TSG expression and function using stimulatory oligonucleotides, such as saRNAs and synthetic nucleoside-modified mRNAs. The remainder of this review describes these forms of oligonucleotide therapy and summarizes the current evidence that they can be used to modulate TSG expression in the context of cancer.
Different approaches to restoring tumour suppressor function
saRNAs
saRNAs are an emerging sub-class of oligonucleotides that have the potential to stimulate the transcription, and hence increase the expression, of a target gene. Unlike siRNAs, which work by interacting with mRNA to block its translation or mediate its degradation, saRNAs instead mediate transcriptional activation by binding to the complementary sequence in or near the promoter region of the target gene.57 saRNAs are short, 21-nucleotide, double-stranded oligomers that are typically delivered to the target organ encapsulated in a lipid nanoparticle. There are many research groups interested in using saRNAs to upregulate TSGs which, as discussed above, are commonly downregulated in various forms of cancer. To date, there is a single saRNA candidate that has progressed into clinical trials, targeting the CEBPA gene in hepatocellular carcinoma (HCC). However, there are many other TSGs being explored as potential saRNA targets preclinically (Table 1).
Mechanism of action
Once an saRNA has entered the cell, the guide strand of the double-stranded molecule is loaded into argonaut protein 2 (Ago2).2 The guide strand of the saRNA duplex is determined by the thermodynamic stability of the 5′ end of the molecule and is usually the antisense strand of the complex.58,59,60 Chu et al.58 (2010) showed that siRNA repression of Ago2 expression significantly decreased saRNA activity, but did not diminish it completely, suggesting there may be a secondary, currently unknown mechanism that also facilitates saRNA delivery to the nucleus.
saRNA-loaded Ago2 protein is shuttled to the nucleus via importin-8,61 and the saRNA guide strand interacts with the complementary sequence in the promoter region of the target gene. This interaction facilitates the recruitment of other proteins required for transcriptional activation, including proteins involved in RNA splicing and binding such as heterogeneous nuclear ribonucleoproteins.60 Others include RNA helicase A (RHA) and RNA polymerase II complex component 9 (CTR9), both of which are known transcriptional activators. RHA is a helicase that catalyzes the unwinding of double-stranded DNA and acts as a bridging factor linking β-actin and RNA polymerase II to form part of the pre-initiation complex.62 CTR9 forms part of the polymerase-associated factor 1 complex (PAF1C), which can interact with histone-modifying enzymes and RNA polymerase II directly to activate gene transcription.60 PAF1C can induce mono-ubiquitination of histone 2B and recruit methyltransferase enzymes to methylate histone 3 lysine 4 (H3K4). H3K4 methylation results in the DNA becoming less condensed around the histone nucleosome, thus making the promoter more accessible for the transcriptional machinery and leading to an increase in gene expression (Figure 2).3
Figure 2.
saRNA structure and mechanism
saRNAs are small, 21 oligonucleotide double-stranded molecules. Once the saRNA has entered the target cell, the double-strand molecule is loaded onto Ago2 (1), and the passenger strand (usually the sense strand of the complex) is released and degraded (2). The Ago2/guide strand complex translocates into the nucleus, facilitated by transporters such as importin-8 and the guide strand interacts with the complementary sequence within the promoter region of the target gene (3). This interaction facilitates recruitment of transcriptional machinery including RHA, PAF1C, and RNA polymerase II (RNAPII) to stimulate transcription of the target gene (4), resulting in an increase in target gene mRNA (5) and functional protein (6).
Targeting TSGs using saRNAs
saRNAs were discovered by two groups in tandem. Janowski et al.63 (2007) discovered them during the development of anti-gene oligonucleotides (agRNAs) designed to downregulate the expression of the progesterone receptor (PR) in breast cancer. Unexpectedly, the group found that some of their synthesized oligonucleotides actually increased the expression of PR by up to 2-fold in T47D and MCF7 cells. The authors also showed that selected agRNAs targeting major vault protein could upregulate its protein expression by up to 4-fold.63 At a similar time, Li et al.2 (2006) successfully developed saRNAs targeting CDH1, CDKN1A, and VEGF in prostate cancer cell lines. Since their initial discovery, there has been a number of groups interested in using saRNAs to target and upregulate genes in specific diseases, in particular, targeting TSGs in cancer. TSGs that function in different capacities to avert cancer, such as by inducing cell-cycle arrest, preventing pro-proliferative signaling, maintaining cell morphology, and inducing apoptosis have been targeted using saRNAs, and examples from each classification are discussed below. Further examples can be found in Table 1.
Cell-cycle arrest: CDKN1A
A TSG that is commonly downregulated in cancer is cyclin-dependent kinase inhibitor 1A (CDKN1A), which encodes p21. p21 is involved in controlling the cell cycle and is able to induce cell-cycle arrest by preventing cyclin-dependent kinase 4 and cyclin D1 from forming an active kinase complex. This prevents the RB protein from becoming phosphorylated and, thus, prevents transcription factor E2F from being active and driving the transcription of genes involved in the S-phase of the cell cycle, such as DNA polymerase and cyclin A.4 p21 can also induce apoptosis by suppressing the pro-apoptotic protein B-cell lymphoma-extra-large and activating caspase-3 and poly-ADP ribose polymerase.4 Therefore, because of the anti-proliferative and pro-apoptotic roles of p21, there has been interest in targeting this protein in oncology.
Since the initial discovery of CDKN1A saRNAs by Li et al.2 (2006), Place et al.5 (2012) showed that addition of 2′ fluoro modifications to the pyrimidine bases in the guide strand increases the stability of the saRNA while retaining RNA activation activity. The saRNA developed by Place et al.5 (2012) also successfully upregulated p21 in a mouse prostate cancer tumor model leading to significant anti-tumour activity. Another study has tested the efficacy of p21 saRNA in vitro in many different cancer cell lines, including PC3 (prostate), T24 (bladder), ACHN (renal), U2-OS (osteosarcoma), and 293T (embryonic kidney).3 The saRNA was able to upregulate transcriptional activity at the p21 promoter by increasing methylation at H3K4.3
A number of saRNAs targeting CDKN1A have been developed and shown to successfully upregulate gene expression at both the mRNA and protein level. Furthermore, the upregulation of p21 leads to changes in cell behavior; in colorectal cancer, HCC, prostate cancer, and renal cell carcinoma cell lines, p21 upregulation results in more cells residing in G0 or G1 phase of the cell cycle,6,7,8,64 as well as a decrease in migrative and invasive capabilities in prostate cancer cell lines.8
Inhibition of pro-proliferative signaling: CEBPA
The first saRNA candidate to enter human clinical trials targets the CEBPA gene, which encodes CCAAT/enhancer-binding protein alpha (C/EBP-α), one of six members of the C/EBP transcription factor family. C/EBP-α upregulates expression of albumin (ALB), adipose triglyceride lipase (ATGL), colony stimulating factor 3 receptor (CSF3R), CDKN1A, and glycogen synthase 1 (GYS1), among other genes.57 These targets encode proteins that are involved in preventing the over-proliferation of mature hepatocytes and maintaining healthy hepatocyte metabolic function. C/EBP-α also downregulates expression of pro-proliferative and pro-inflammatory genes such as c-MYC and interferon-gamma (INFG).57 C/EBP-α expression is decreased in 60% of HCC tumors in comparison with non-tumour tissue from the same patients, and its deficiency is associated with increased hepatic proliferation in mice, thus resulting in worsened tumor phenotype.65
In vitro studies showed that increased CEBPA expression in HCC cell lines led to decreased expression of mesenchymal markers such as N-cadherin, Slug, and vimentin and upregulated epithelial markers such as E-cadherin, implying that the upregulation of C/EBP-α helps to prevent epithelial-to-mesenchymal transition.14 The upregulation of C/EBP-α also resulted in the downregulation of other pro-proliferative proteins such as β-catenin and epidermal growth factor receptor (EGFR), c-MYC, axis inhibition protein 2 (AXIN2), and cyclin D1 (CCND1).14
Huan et al.14 (2016) investigated the effects of saRNA-targeting CEBPA in a mouse orthotopic HCC tumor model. Over 40 days, the group saw a decrease in the tumor size and intrahepatic metastasis in CEBPA saRNA-treated mice compared with mice treated with a scrambled saRNA. Furthermore, the downregulation of pro-proliferative genes EGFR and catenin beta 1 (CTNNB1) was observed in CEBPA-saRNA-treated mice using immunohistochemistry analysis.14
In another study, CEBPA-51, the preclinical candidate saRNA targeting CEBPA, was encapsulated into liposomal nanoparticles (SMARTICLES) for targeted delivery to the liver, and injected into rats exhibiting cirrhotic HCC, induced by injection of diethylnitrosamine over a 9-week period.66 Animals treated with saRNA targeting CEBPA had an 80% decrease in hepatic tumor size compared with animals treated with a non-specific oligonucleotide.
Recently, MTL-CEBPA completed first-in-human clinical trials to assess tolerability in patients. The trial consisted of 34 patients with advanced HCC with underlying cirrhosis, metastasis or resulting from non-alcoholic steatohepatitis. Despite the drug not causing a significant improvement in liver function test results compared with baseline after two cycles of treatment, the relative expression of CEBPA mRNA in white blood cells increased 1.5-fold consistently across all treatment groups.15 Furthermore, one patient achieved a confirmed partial tumor response, as seen both by computed tomography scan and by a rapid decrease in the alpha-fetoprotein level, which was maintained for 24 months. The mean progression-free survival of the entire patient cohort (different doses of MTL-CEBPA) was 4.6 months, suggesting that the drug has some anti-tumour activity.15
A phase Ib clinical trial of MTL-CEBPA in combination with sorafenib in patients with advanced HCC showed that the treatment can successfully decrease the tumor burden. One-quarter of patients who were naive to tyrosine-kinase inhibitor treatment and had a viral etiology showed a complete or partial objective response to treatment after 12 months, with three patients exhibiting a complete response, as demonstrated by a complete eradication of target lesions at month 12.16 Furthermore, the study demonstrated that the upregulation of CEBPA expression caused by the treatment leads to the downregulation of immune suppressive genes in myeloid-derived suppressor cells and the upregulation of genes associated with monocyte and neutrophil function.16 Although no upregulation in monocyte and neutrophil levels was seen in the patients’ blood 24 h and 7 days after MTL-CEBPA administration, a marked decrease in the levels of monocytic MDSCs was observed. This suggests that MTL-CEBPA is able to exert its tumor suppressive effects by abrogating the immune suppressive activity of monocytic MDSCs in the tumor microenvironment.16
Given the success of MTL-CEBPA in clinical trials for advanced HCC, the drug has attracted interest as a novel therapeutic agent in other cancers, such as pancreatic ductal adenocarcinoma, for which saRNA targeting CEBPA has been reported to have anti-tumour effects both in vitro and in vivo.17,18
Maintaining cell morphology: CDH1
CDH1 is a TSG that has gained attention for its potential to be upregulated using saRNAs and has been investigated in this context across many cancer types.2,23,24,25,26,27 CDH1 encodes the transmembrane glycoprotein E-cadherin, which is involved in maintaining epithelial cell morphology via adherens junction connections.67 Furthermore, E-cadherin is anchored to the cell’s cytoskeleton via an interaction with β-catenin, a key protein involved in the pro-proliferative Wnt signaling pathway.68,69 saRNAs targeting CDH1 were initially discovered in 2006 in the context of prostate cancer.2 Wu et al. (2016) further investigated the effects of saRNAs targeting CDH1 in prostate cancer, using different saRNAs to those designed by Li et al. (2006). The group found that one of their candidates, an saRNA that binds to the −661 region of the CDH1 promoter, could successfully upregulate CDH1 expression by up to 5-fold at the mRNA level, and that the anti-sense strand of the saRNA duplex was responsible for the RNA activating activity. The same group tested this saRNA in HCC cell lines and observed a similar increase in E-cadherin protein, as observed by western blot.
The −215 region of the CDH1 promoter has also been commonly targeted, with saRNAs designed around this region being shown to successfully upregulate CDH1 in renal carcinoma and breast cancer, and produce beneficial downstream phenotypic effects.23,26 Dai et al.23 (2018) found that renal carcinoma cell lines transfected with CDH1 saRNA had a reduced migration capability compared with non-transfected cells, as measured using a Matrigel invasion chamber assay. When CDH1 was knocked down again using RNA interference, the renal carcinoma cells regained their migration capability.23 Similarly, saRNAs targeting the −215 region of the CDH1 promoter in breast cancer led to cells having a decreased proliferation rate, increased apoptosis and decreased migration capabilities, compared with mock-transfected and control sequence transfected cells, as well as a potent growth inhibitory effect in vivo.26
Promoting apoptosis: PAWR
An example of a TSG which is involved in promoting apoptosis is PRKC apoptosis WT1 regulator (PAWR). This TSG has been implicated in the apoptotic response of prostate cancer cells to several exogenous agents. One group has investigated the impact of saRNAs targeting PAWR in vitro and has found that transfection with the oligonucleotide results in a decrease in cell growth, as well as cell shrinkage and apoptosis.31 This study provides proof-of-concept that PAWR upregulation in prostate cancer is a potential therapeutic avenue to explore, and provides opportunities for further in vivo work to build upon the preclinical profile of such a molecule.
Synthetic mRNAs
Although saRNAs are a promising novel approach for upregulating TSGs in cancer, their ability to increase gene expression is limited to the upregulation of genes in which at least one allele retains normal activity. When both alleles of a gene are deleted or mutated, saRNAs are unable to exert any beneficial effect. An alternative oligonucleotide therapy that can be used to restore the expression of TSGs is synthetic, nucleoside-modified mRNA. Synthetic mRNAs have been explored for many years for their potential in cancer immunotherapy and have recently made headlines for their use in the development of sever acute respiratory syndrome coronavirus 2 vaccines.70,71,72 Synthetic mRNAs are synthesized to mimic natural mRNAs and are translated in the cytoplasm to produce protein, which is indistinguishable to that resulting from the translation of endogenous mRNA.
The in vitro mRNA synthesis process involves ligating the open reading frame of the gene of interest, along with any other desired sequences such as 5′ and 3′ untranslated regions into a plasmid with a viral promoter, usually T7, upstream of the transcription start site. The plasmid must be linearized, such that RNA transcripts of defined length are produced. The viral promoter is recognized by phage RNA polymerases to initiate RNA synthesis. These sequences are then transcribed in vitro to produce mRNA.73 The use of synthetic mRNAs to increase translation of a target protein has been under investigation since the 1980s. Malone et al.73 (1989) successfully transfected in vitro transcribed mRNA encoding Photinus pyralis luciferase into NIH 3T3 mouse cells. The same group showed that surrounding the open reading frame (ORF) of the target gene with 5′ and 3′ untranslated regions (UTRs) helped to increase translational efficiency.73
Given their size in comparison with saRNAs, synthetic mRNAs are more difficult to deliver to a cell; however, once inside the cell they can exert their effects rapidly, as the transcription stage is bypassed. However, the rapid degradation of mRNAs and the ability to activate toll-like receptors (TLRs) and stimulate the immune system led to a period of decline in interest in their use as a therapeutic agent.74
However, interest in the use of synthetic mRNAs was reignited in the mid-2000s, when Karikó et al. (2005)75 discovered that the addition of naturally modified nucleosides, such as pseudouridine and 5-methylcytidine, to the ORF resulted in a decreased secretion of cytokines and interleukin-8 in response to their administration in vitro. The modified mRNAs led to less stimulation of immune response mediators such as interferon-gamma, interleukin-2 (IL-12), interferon-alpha, retinoic acid-inducible gene 1, and TLRs 3, 7, and 8.76
Alongside the addition of 5′ and 3′ UTRs and nucleoside modifications to the ORF, synthetic mRNAs can be further modified with the addition of a 5′ cap and poly-A tail to mimic endogenous mRNAs (Figure 3). Endogenous mRNAs contain a single 7-methylguanosine residue at their 5′ end, attached to the next residue by a 5′-5′ triphosphate linkage.77 The 5′ cap confers resistance to enzymatic cleavage and allows for the translational machinery to recognise and bind to the mRNAs to allow efficient translation into protein.73
Figure 3.
Synthetic mRNA structure and examples of their use in TSG upregulation
Synthetic mRNAs are designed to mimic endogenous mRNAs by the addition of a 5′ cap, which contains a 5′-5′ triphosphate bridge to improve stability, 5′ and 3′ UTRs and a poly-A tail. The ORF of the structure can be further modified to include naturally modified bases such as 5-methylcytidine and N6-methyladenosine, which decrease immunogenicity and facilitate in vivo delivery. Examples of the use of synthetic mRNAs to restore TSG expression include PTEN in prostate cancer and melanoma and BAX in malignant melanoma.
The 3′ poly(A) tail plays a role in improving mRNA stability. Optimization studies have shown that an increased length of poly(A) tail from 20 nucleotides to 120 nucleotides is associated with improved translational efficiency and therefore protein expression.78 However, other studies have used even longer poly(A) tails (e.g., ≤200 nucleotides in length) with similar success.79
Using synthetic mRNAs to upregulate TSGs in cancer
Given the recent improvements in the stability and immunogenicity of synthetic mRNAs and the emergence of delivery systems such as lipid nanoparticles to facilitate the entry of large synthetic mRNAs into the cell, research into the use of such nucleoside-modified, in vitro transcribed mRNAs as therapeutic agents has become more established. mRNA therapy provides an advantage over small molecule drugs because of its ability to increase or restore protein expression, and therefore activity, whereas small molecules target proteins that are already present, often in an inhibitory manner. A particular area that has gained much traction in recent years is cancer immunotherapy.80,81,82 Alongside this, some groups have started to investigate the potential of using synthetic mRNAs to upregulate or restore the function of the TSGs that are commonly downregulated or lost in various cancers. Although synthetic mRNAs are a promising area of research and restoring gene function in other disease contexts has been explored extensively, such as CFTR in cystic fibrosis,83 research into the use of synthetic mRNAs to restore TSGs is currently somewhat lacking. This review discusses the literature demonstrating the potential of using synthetic mRNAs as a means of restoring TSG function in the context of cancer.
PTEN in prostate cancer and melanoma
Islam et al.84 (2018) have delivered synthetic mRNA encoding PTEN encapsulated in hybrid lipid nanoparticles into the prostate cancer cell line PC3. The hybrid nanoparticles were prepared using cationic lipid-like compound G0-C14 (for mRNA complexation) and poly(lactic-co-glycolic acid) polymer coated with a lipid-polyethyl glycol (PEG) shell to make a stable nanoparticle core.84 The group showed that transfection of the encapsulated mRNA was more effective than transfection of naked mRNA using a commercial cationic lipid transfection reagent. An increase in PTEN protein expression was seen 48 h after transfection, alongside a decrease in cell viability. Furthermore, the synthetic PTEN mRNA provoked a downregulation of the PI3K-AKT signaling pathway, as demonstrated by decreased phosphorylation of eukaryotic translation initiation factor 4E-binding protein 1, proline-rich AKT substrate of 40 kDa, and Foxo3a.84 Decreased phosphorylation of these proteins indicates the inhibition of AKT activity and its downstream pro-proliferative and pro-survival effects. In vivo injection of the PTEN mRNA encapsulated in nanoparticles caused a suppression of growth of PC3 xenograft tumors compared with control injections of PBS or mRNA encoding EGFR.84
The same group went on to develop a nanoparticle containing PTEN mRNA for the delivery of the oligonucleotide into Pten-null or mutated tumor cells and explored the effects of such therapy on the tumor microenvironment.85 The treatment of Pten-deficient murine cells with nanoparticles containing PTEN mRNA led to the induction of apoptosis and autophagy pathways in vitro, as well as calreticulin exposure on the cell membrane and ATP release into the extracellular environment, indicating that PTEN upregulation may influence the tumor microenvironment. The hypothesis was explored further in vivo in a melanoma tumor model, and the group saw that injection of PTEN mRNA-containing nanoparticles led to an increase in CD3+CD8+ T cells and decrease in regulatory T cells and myeloid-derived suppressive cells, which helps to reverse the immunosuppressive tumor microenvironment.85 Furthermore, the upregulation of PTEN in melanoma and prostate cancer tumor models led to an increase in efficacy of anti-programmed cell death protein 1 therapy, highlighting the potential benefits that activating oligonucleotides can have in the clinical oncology field.85
Bax in malignant melanoma
Okumura et al. (2008) used cationic liposomes to deliver mRNA encoding Bax, a pro-apoptotic protein into HMG malignant melanoma cells. Bax stimulates the release of cytochrome C from the mitochondria, which in turn leads to the initiation of apoptosis.86,87 The group found that encapsulation of the mRNA into cationic liposomes was sufficient to complex the mRNA and deliver it to the cells. Furthermore, an increase in the levels of Bax protein and caspase-3 activity were observed 24 h after transfection, alongside a significant decrease in cell survival.88 The only modifications added to the synthetic mRNA were anti-reverse cap analogs (ARCA) 5′ cap and poly(A) tail. ARCAs are commonly used in the synthesis of commercial mRNAs to increase translational efficiency.89 They prevent incorporation of the 5′ cap in the reverse orientation, which would stop the mRNA from being recognized by the ribosome and translated.
Benefits and challenges of using different oligonucleotide therapies
saRNAs and synthetic mRNAs are two types of oligonucleotide therapy that can be used to upregulate the expression of TSGs in cancer. However, they differ in terms of size and mechanism of action, as well as their potential for gene upregulation (Table 2). Given that saRNA activity requires entry into the nucleus and interaction with the promoter region of the target gene, saRNAs have the potential to upregulate only functional genes. In contrast, synthetic mRNAs operate within the cytoplasm and provide the cell with the correct mRNA sequence, which is then directly translated into functional protein. This allows for synthetic mRNAs to be used to restore otherwise absent proteins in disease, as evidenced by the use of this approach in protein replacement therapies, such as mRNA encoding the cystic fibrosis transmembrane conductance regulator protein in cystic fibrosis.83 The requirement for increased transcription of endogenous DNA in the saRNA mechanism means that protein production is slow, taking up to 72 h in many in vitro models.19 However, protein production can be seen within 24 h of administration of synthetic mRNAs.90,91,92
Table 2.
Comparison of saRNAs and synthetic mRNAs as agents for modulation of tumor suppressor activity in cancer
| saRNAs | Synthetic mRNAs | |
|---|---|---|
| Size | Small, 21 oligonucleotides | Long, 1,000s of oligonucleotides |
| Active site | Nucleus | Cytoplasm |
| Delivery |
In vitro – standard transfection reagents In vivo – lipid nanoparticles |
In vitro and in vivo – lipid nanoparticles |
| Onset of effect | Slower (within 72 h of administration) due to requirement for transcription Li et al., Reebye et al. (Refs.2,19) | Faster (within 24 h of administration) due to rapid translation Pardi et al., Rizvi et al. (Refs.90,91,92) |
| Duration of effect | Weeks to months with stabilized saRNA, requiring infrequent dosing for therapeutic effects Hashimoto et al. (Ref.16) | Days, requiring frequent dosing for prolonged therapeutic effects (except vaccines). |
| Safety | mRNA upregulated within normal range effect restricted to cells that normally express target gene |
Risk of overexpression. Effect not restricted to cells that normally express target gene. Potential mitochondrial toxicities (due to addition of unnatural nucleoside analogues). Sahin et al. (Ref.93) |
A major hurdle in translating the use of these classes of oligonucleotide therapy into the clinic is the delivery of the oligonucleotides in vivo. Some of the challenges faced by oligonucleotides include degradation by nucleases in the extracellular space, renal clearance, crossing the capillary endothelium once at the target organ, crossing the cell membrane, and lysosomal degradation.94 Oligonucleotides are hydrophilic and so are unable to penetrate the plasma membrane without assistance. Furthermore, the introduction of any foreign oligonucleotide can result in stimulation of the innate immune system and TLRs. Therefore, a suitable delivery system must be used to ensure that the oligonucleotide reaches its target site, and enters the target cell before it is degraded by extracellular nucleases or stimulates the innate immune response.
The most common non-viral delivery system used for oligonucleotide therapies is lipid nanoparticles. This process involves complexing the anionic oligonucleotide with cationic lipids to produce a nanoparticle. For in vivo delivery, more complex structures may be used to prevent the nanoparticle from being cleared from the circulation by reticuloendothelial system phagocytes. For example, the surface can be coated with a neutral polymer such as PEG to prevent protein adherence to the nanoparticle and clearance.95 saRNAs are small structures and can be delivered in vitro using simple transfection reagents. However, synthetic mRNAs are much larger and require encapsulation within a nanoparticle for both in vitro and in vivo delivery (Table 2).
Another method for oligonucleotide delivery that can be used as an alternative to nanoparticles, or in conjunction with them, is bioconjugation. This is where the oligonucleotide or carrier (i.e., nanoparticle) is conjugated to a ligand that promotes interaction of the molecule with the target cell. For example, a commonly used ligand for selective delivery of oligonucleotides to the liver is N-acetylgalactosamine (GalNAc). The GalNAc ligand is recognized by the asiaglycoprotein receptor, which is highly expressed on the surface of hepatocytes and internalizes the ligand via Ca2+-dependent endocytosis.96,97 The GalNAc ligand can be directly attached to small oligonucleotides, such as saRNAs and siRNAs, but this is not possible for larger molecules such as synthetic mRNAs, which require encapsulation within a nanoparticle for both in vitro and in vivo delivery. Some oligonucleotide therapies approved by the U.S. Food and Drug Administration use the GalNAc conjugation system, such as givosiran, an siRNA to treat acute hepatic porphyria.98
Conclusions and future perspectives
TSGs are commonly downregulated in cancer and, despite the idea gaining much attention within the scientific community, efforts to upregulate or reactivate TSGs thus far have remained relatively fruitless. However, emerging classes of therapy, such as oligonucleotide therapies, offer an advantage over traditional small molecule drugs in their capability to work at the gene level, rather than by targeting a protein. Oligonucleotide therapies have the potential to be combined with small molecules or other oligonucleotides to produce a synergistic effect. saRNAs and nucleoside-modified RNAs are two classes of oligonucleotide therapy with the potential to upregulate expression of target genes, or in the case of synthetic mRNAs, replace a lost or faulty protein in cancer. saRNAs offer a benefit over synthetic mRNAs in terms of their size and ease of delivery; however, they are an inappropriate treatment option for patients with a mutated allele of the target, as the upregulation of a mutated target will not have a beneficial effect and may even be detrimental. Future work should aim to address challenges relating to delivery and immunogenicity to improve the potential of oligonucleotide therapies to progress through clinical trials and to meet the need for TSG upregulation in cancer. Furthermore, as targeted oligonucleotide therapies start to emerge into the clinic, the use of tumor genome sequencing could help to match a patient with an ideal therapy based on the genetic profile of their tumor. The field of research is still in its infancy and provides exciting opportunities for development and clinical translation in the coming years.
Acknowledgments
G.G.’s PhD studentship is funded by North West Cancer Research. The authors thank Dr David Blakey for reading the manuscript and sharing his expert opinions on the oligonucleotide therapy landscape.
Author contributions
G.G. wrote the manuscript. I.C. wrote the manuscript.
Declaration of interests
G.G.’s PhD studentship is supported through in-kind contributions from MiNA Therapeutics. The company has not been involved in the preparation of this manuscript.
References
- 1.Hanahan D., Weinberg R.A. Hallmarks of cancer: the next generation. Cell. 2011;144:646–674. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
- 2.Li L.C., Okino S.T., Zhao H., Pookot D., Place R.F., Urakami S., Enokida H., Dahiya R. Small dsRNAs induce transcriptional activation in human cells. Proc. Natl. Acad. Sci. USA. 2006;103:17337–17342. doi: 10.1073/pnas.0607015103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Wu H.L., Li S.M., Hu J., Yu X., Xu H., Chen Z., Ye Z.Q. Demystifying the mechanistic and functional aspects of p21 gene activation with double-stranded RNAs in human cancer cells. J. Exp. Clin. Cancer Res. 2016;35:145. doi: 10.1186/s13046-016-0423-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Kang M.R., Li G., Pan T., Xing J.-C., Li L.-C. Advances in Experimental Medicine and Biology. Springer New York LLC; 2017. Development of therapeutic dsP21-322 for cancer treatment; pp. 217–229. [DOI] [PubMed] [Google Scholar]
- 5.Place R.F., Wang J., Noonan E.J., Meyers R., Manoharan M., Charisse K., Duncan R., Huang V., Wang X., Li L.C. Formulation of small activating RNA into lipidoid nanoparticles inhibits xenograft prostate tumor growth by inducing p21 expression. Mol. Ther. Nucleic Acids. 2012;1:e15. doi: 10.1038/mtna.2012.5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Wang L.L., Guo H.H., Zhan Y., Feng C.L., Huang S., Han Y.X., Zheng W.S., Jiang J.D. Specific up-regulation of p21 by a small active RNA sequence suppresses human colorectal cancer growth. Oncotarget. 2017;8:25055–25065. doi: 10.18632/oncotarget.15918. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Kosaka M., Kang M.R., Yang G., Li L.C. Targeted p21WAF1/CIP1 activation by RNAa inhibits hepatocellular carcinoma cells. Nucleic Acid Ther. 2012;22:335–343. doi: 10.1089/nat.2012.0354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Zhang Q., Yang X., Luo L., Ma X., Jiao W., Li B., Zhang M., Zhao K., Niu H. Targeted p21 activation by a new double stranded RNA suppresses human prostate cancer cells growth and metastasis. Am. J. Transl. Res. 2020;12:4175–4188. [PMC free article] [PubMed] [Google Scholar]
- 9.Wu Z.M., Dai C., Huang Y., Zheng C.F., Dong Q.Z., Wang G., Li X.W., Zhang X.F., Li B., Chen G. Anti-cancer effects of p21WAF1/CIP1 transcriptional activation induced by dsRNAs in human hepatocellular carcinoma cell lines. Acta Pharmacol. Sin. 2011;32:939–946. doi: 10.1038/aps.2011.28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Chen Z., Place R.F., Jia Z.-J., Pookot D., Dahiya R., Li L.-C. Antitumor effect of dsRNA-induced p21 WAF1/CIP1 gene activation in human bladder cancer cells. Mol. Cancer Ther. 2008;7:698–703. doi: 10.1158/1535-7163.MCT-07-2312. [DOI] [PubMed] [Google Scholar]
- 11.Wang C., Ge Q., Zhang Q., Chen Z., Hu J., Li F., Ye Z. Targeted p53 activation by saRNA suppresses human bladder cancer cells growth and metastasis. J. Exp. Clin. Cancer Res. 2016;35:53. doi: 10.1186/s13046-016-0329-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Lin D., Meng L., Xu F., Lian J., Xu Y., Xie X., Wang X., He H., Wang C., Zhu Y. Enhanced wild-type p53 expression by small activating RNA dsP53-285 induces cell cycle arrest and apoptosis in pheochromocytoma cell line PC12. Oncol. Rep. 2017;38:3160–3166. doi: 10.3892/or.2017.5993. [DOI] [PubMed] [Google Scholar]
- 13.Kang M.R., Park K.H., Lee C.W., Lee M.Y., Han S.B., Li L.C., Kang J.S. Small activating RNA induced expression of VHL gene in renal cell carcinoma. Int. J. Biochem. Cell Biol. 2018;97:36–42. doi: 10.1016/j.biocel.2018.02.002. [DOI] [PubMed] [Google Scholar]
- 14.Huan H., Wen X., Chen X., Wu L., Liu W., Habib N.A., Bie P., Xia F. C/EBPα short-activating RNA suppresses metastasis of hepatocellular carcinoma through inhibiting EGFR/β-Catenin signaling mediated EMT. PLoS One. 2016;11:e0153117. doi: 10.1371/journal.pone.0153117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Sarker D., Plummer R., Meyer T., Sodergren M.H., Basu B., Chee C.E., Huang K.W., Palmer D.H., Ma Y.T., Evans T.R.J., et al. MTL-CEBPA, a small activating RNA therapeutic upregulating C/EBP-α, in patients with advanced liver cancer: a first-in-human, multicenter, open-label, phase I trial. Clin. Cancer Res. 2020;26:3936–3946. doi: 10.1158/1078-0432.CCR-20-0414. [DOI] [PubMed] [Google Scholar]
- 16.Hashimoto A., Sarker D., Reebye V., Jarvis S., Sodergren M.H., Kossenkov A., Sanseviero E., Raulf N., Vasara J., Andrikakou P., et al. Up-regulation of C/EBPα inhibits suppressive activity of myeloid cells and potentiates antitumor response in mice and cancer patients. Clin. Cancer Res. 2021;27:5961–5978. doi: 10.1158/1078-0432.CCR-21-0986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Yoon S., Rossi J.J. Advances in Experimental Medicine and Biology. Springer New York LLC; 2017. Treatment of pancreatic cancer by aptamer conjugated C/EBPα-saRNA; pp. 173–188. [DOI] [PubMed] [Google Scholar]
- 18.Yoon S., Huang K.W., Andrikakou P., Vasconcelos D., Swiderski P., Reebye V., Sodergren M., Habib N., Rossi J.J. Targeted delivery of C/EBPα-saRNA by RNA aptamers shows anti-tumor effects in a mouse model of advanced PDAC. Mol. Ther. Nucleic Acids. 2019;18:142–154. doi: 10.1016/j.omtn.2019.08.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Reebye V., Sætrom P., Mintz P.J., Huang K.W., Swiderski P., Peng L., Liu C., Liu X., Lindkær-Jensen S., Zacharoulis D., et al. Novel RNA oligonucleotide improves liver function and inhibits liver carcinogenesis in vivo. Hepatology. 2014;59:216–227. doi: 10.1002/hep.26669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Chen H., Shen H.X., Lin Y.W., Mao Y.Q., Liu B., Xie L.P. Small RNA-induced INTS6 gene up-regulation suppresses castration-resistant prostate cancer cells by regulating β-catenin signaling. Cell Cycle. 2018;17:1602–1613. doi: 10.1080/15384101.2018.1475825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Pan S., Wang Z., Chen Y., Chen X., Ji J., Zhang J., Li J., Cai Q., Liu B., Zhu Z., Yu Y. Inactivation of tumor suppressor gene HIC1 in gastric cancer is reversed via small activating RNAs. Gene. 2013;527:102–108. doi: 10.1016/j.gene.2013.05.034. [DOI] [PubMed] [Google Scholar]
- 22.Li M., Peng Z., Ren W., Wang Z. Small activating ribonucleic acid reverses tyrosine kinase inhibitor resistance in epidermal growth factor receptor-mutant lung cancer by increasing the expression of phosphatase and tensin homolog. Thorac. Cancer. 2016;7:481–485. doi: 10.1111/1759-7714.12356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Dai J., He H., Lin D., Wang C., Zhu Y., Xu D. Up-regulation of E-cadherin by saRNA inhibits the migration and invasion of renal carcinoma cells. Int. J. Clin. Exp. Pathol. 2018;11:5792–5800. [PMC free article] [PubMed] [Google Scholar]
- 24.Wu Z., Li Y., Li Z., Liu Z., Qin Z., Li X., Ye Y., Bu L., Lin B., Wang Z., et al. Transcriptional regulation of E-cadherin by small activating RNA: a new double-stranded RNA. Int. J. Oncol. 2016;49:1620–1628. doi: 10.3892/ijo.2016.3643. [DOI] [PubMed] [Google Scholar]
- 25.Li C., Liu J., Zhang Q., Cui K., Ge Q., Wang C., Chen Z. Upregulation of E-cadherin expression mediated by a novel dsRNA suppresses the growth and metastasis of bladder cancer cells by inhibiting β-catenin/TCF target genes. Int. J. Oncol. 2018;52:1815–1826. doi: 10.3892/ijo.2018.4346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Junxia W., Ping G., Yuan H., Lijun Z., Jihong R., Fang L., Min L., Xi W., Ting H., Ke D., Huizhong Z. Double strand RNA-guided endogeneous E-cadherin up-regulation induces the apoptosis and inhibits proliferation of breast carcinoma cells in vitro and in vivo. Cancer Sci. 2010;101:1790–1796. doi: 10.1111/j.1349-7006.2010.01594.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Mao Q., Li Y., Zheng X., Yang K., Shen H., Qin J., Bai Y., Kong D., Jia X., Xie L. Up-regulation of E-cadherin by small activating RNA inhibits cell invasion and migration in 5637 human bladder cancer cells. Biochem. Biophys. Res. Commun. 2008;375:566–570. doi: 10.1016/j.bbrc.2008.08.059. [DOI] [PubMed] [Google Scholar]
- 28.Xie D., Shang L., Peng L., Li L. Up-regulation of VEZT by small activating RNA inhibits the proliferation, invasion and migration of gastric cancer cells. Biochem. Biophys. Res. Commun. 2017;482:542–548. doi: 10.1016/j.bbrc.2016.11.071. [DOI] [PubMed] [Google Scholar]
- 29.Li C., Jiang W., Hu Q., Li L.C., Dong L., Chen R., Zhang Y., Tang Y., Thrasher J.B., Liu C.B., Li B. Enhancing DPYSL3 gene expression via a promoter-targeted small activating RNA approach suppresses cancer cell motility and metastasis. Oncotarget. 2016;7:22893–22910. doi: 10.18632/oncotarget.8290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Ren S., Kang M.R., Wang J., Huang V., Place R.F., Sun Y., Li L.C. Targeted induction of endogenous NKX3-1 by small activating RNA inhibits prostate tumor growth. Prostate. 2013;73:1591–1601. doi: 10.1002/pros.22709. [DOI] [PubMed] [Google Scholar]
- 31.Yang K., Shen J., Chen S.W., Qin J., Zheng X.Y., Xie L.P. Upregulation of PAWR by small activating RNAs induces cell apoptosis in human prostate cancer cells. Oncol. Rep. 2016;35:2487–2493. doi: 10.3892/or.2016.4582. [DOI] [PubMed] [Google Scholar]
- 32.Zhu Q., Wu X., Huang Y., Tang M., Wu L. Upregulation of FHIT gene expression in endometrial carcinoma by RNA activation. Int. J. Clin. Exp. Pathol. 2020;13:1372–1380. [PMC free article] [PubMed] [Google Scholar]
- 33.Xu X., Li J., Wang S., Zheng X., Xie L. RNAa and vector-mediated overexpression of DIRAS1 suppresses tumor growth and migration in renal cell carcinoma. Mol. Ther. Nucleic Acids. 2018;12:845–853. doi: 10.1016/j.omtn.2018.07.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Zhou Q., Fan D., Huang K., Chen X., Chen Y., Mai Q. Activation of KLF4 expression by small activating RNA promotes migration and invasion in colorectal epithelial cells. Cell Biol. Int. 2018;42:495–503. doi: 10.1002/cbin.10926. [DOI] [PubMed] [Google Scholar]
- 35.Wang J., Place R.F., Huang V., Wang X., Noonan E.J., Magyar C.E., Huang J., Li L.C. Prognostic value and function of KLF4 in prostate cancer: RNAa and vector-mediated overexpression identify KLF4 as an inhibitor of tumor cell growth and migration. Cancer Res. 2010;70:10182–10191. doi: 10.1158/0008-5472.CAN-10-2414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Qin Q., Lin Y.W., Zheng X.Y., Chen H., Mao Q.Q., Yang K., Huang S.J., Zhao Z.Y. RNAa-mediated overexpression of WT1 induces apoptosis in HepG2 cells. World J. Surg. Oncol. 2012;10:11. doi: 10.1186/1477-7819-10-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Xiong Y., Ke R., Zhang Q., Lan W., Yuan W., Chan K.N.I., Roussel T., Jiang Y., Wu J., Liu S., et al. Small activating RNA modulation of the G protein-coupled receptor for cancer treatment. Adv. Sci. 2022;9:2200562. doi: 10.1002/advs.202200562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Knudson A.G. Mutation and cancer: statistical study of retinoblastoma. Proc. Natl. Acad. Sci. USA. 1971;68:820–823. doi: 10.1073/pnas.68.4.820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Berger A.H., Knudson A.G., Pandolfi P.P. A continuum model for tumour suppression. Nature. 2011;476:163–169. doi: 10.1038/nature10275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Alimonti A., Carracedo A., Clohessy J.G., Trotman L.C., Nardella C., Egia A., Salmena L., Sampieri K., Haveman W.J., Brogi E., et al. Subtle variations in Pten dose determine cancer susceptibility. Nat. Genet. 2010;42:454–458. doi: 10.1038/ng.556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Trotman L.C., Niki M., Dotan Z.A., Koutcher J.A., Di Cristofano A., Xiao A., Khoo A.S., Roy-Burman P., Greenberg N.M., Van Dyke T., et al. Pten dose dictates cancer progression in the prostate. PLoS Biol. 2003;1:E59. doi: 10.1371/journal.pbio.0000059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.O’brien S.G., Guilhot F., Larson R.A., Gathmann I., Baccarani M., Cervantes F., Cornelissen J.J., Fischer T., Hochhaus A., Hughes T., et al. Imatinib compared with interferon and low-dose cytarabine for newly diagnosed chronic-phase chronic myeloid leukemia. N. Engl. J. Med. 2003;348:994–1004. doi: 10.1056/NEJMoa022457. [DOI] [PubMed] [Google Scholar]
- 43.Llovet J.M., Ricci S., Mazzaferro V., Hilgard P., Gane E., Blanc J.-F., de Oliveira A.C., Santoro A., Raoul J.-L., Forner A., et al. Sorafenib in advanced hepatocellular carcinoma. N. Engl. J. Med. 2008;359:378–390. doi: 10.1056/NEJMoa0708857. [DOI] [PubMed] [Google Scholar]
- 44.Yu X., Vazquez A., Levine A.J., Carpizo D.R. Allele specific p53 mutant reactivation. Cancer Cell. 2012;21:614–625. doi: 10.1016/j.ccr.2012.03.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Boeckler F.M., Joerger A.C., Jaggi G., Rutherford T.J., Veprintsev D.B., Fersht A.R. Targeted rescue of a destabilized mutant of p53 by an in silico screened drug. Proc. Natl. Acad. Sci. USA. 2008;105:10360–10365. doi: 10.1073/pnas.0805326105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Raghavan V., Agrahari M., Gowda D.K. Virtual screening of p53 mutants reveals Y220S as an additional rescue drug target for PhiKan083 with higher binding characteristics. Comput. Biol. Chem. 2019;80:398–408. doi: 10.1016/j.compbiolchem.2019.05.005. [DOI] [PubMed] [Google Scholar]
- 47.Zhang Q., Bykov V.J.N., Wiman K.G., Zawacka-Pankau J. APR-246 reactivates mutant p53 by targeting cysteines 124 and 277. Cell Death Dis. 2018;9:439–512. doi: 10.1038/s41419-018-0463-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Lehmann S., Bykov V.J.N., Ali D., Andrén O., Cherif H., Tidefelt U., Uggla B., Yachnin J., Juliusson G., Moshfegh A., et al. Targeting p53 in vivo: a first-in-human study with p53-targeting compound APR-246 in refractory hematologic malignancies and prostate cancer. J. Clin. Oncol. 2012;30:3633–3639. doi: 10.1200/JCO.2011.40.7783. [DOI] [PubMed] [Google Scholar]
- 49.Deneberg S., Cherif H., Lazarevic V., Andersson P.O., von Euler M., Juliusson G., Lehmann S. An open-label phase I dose-finding study of APR-246 in hematological malignancies. Blood Cancer J. 2016;6:447. doi: 10.1038/bcj.2016.60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Sallman D.A., DeZern A.E., Garcia-Manero G., Steensma D.P., Roboz G.J., Sekeres M.A., Cluzeau T., Sweet K.L., McLemore A., McGraw K.L., et al. Eprenetapopt (APR-246) and azacitidine in TP53-mutant myelodysplastic syndromes. J. Clin. Oncol. 2021;39:1584–1594. doi: 10.1200/JCO.20.02341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Lambert J.M.R., Gorzov P., Veprintsev D.B., Söderqvist M., Segerbäck D., Bergman J., Fersht A.R., Hainaut P., Wiman K.G., Bykov V.J.N. PRIMA-1 reactivates mutant p53 by covalent binding to the core domain. Cancer Cell. 2009;15:376–388. doi: 10.1016/j.ccr.2009.03.003. [DOI] [PubMed] [Google Scholar]
- 52.Nitulescu G.M., Margina D., Juzenas P., Peng Q., Olaru O.T., Saloustros E., Fenga C., Spandidos D.Α., Libra M., Tsatsakis A.M. Akt inhibitors in cancer treatment: the long journey from drug discovery to clinical use (Review) Int. J. Oncol. 2016;48:869–885. doi: 10.3892/ijo.2015.3306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Bulcha J.T., Wang Y., Ma H., Tai P.W.L., Gao G. Viral vector platforms within the gene therapy landscape. Signal Transduct. Target. Ther. 2021;6:24–53. doi: 10.1038/s41392-021-00487-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Baum C., Kustikova O., Modlich U., Li Z., Fehse B. Mutagenesis and oncogenesis by chromosomal insertion of gene transfer vectors. Hum. Gene Ther. 2006;17:253–263. doi: 10.1089/hum.2006.17.253. [DOI] [PubMed] [Google Scholar]
- 55.Shirley J.L., de Jong Y.P., Terhorst C., Herzog R.W. Immune responses to viral gene therapy vectors. Mol. Ther. 2020;28:709–722. doi: 10.1016/j.ymthe.2020.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Wang F., Zuroske T., Watts J.K. RNA therapeutics on the rise. Nat. Rev. Drug Discov. 2020;19:441–442. doi: 10.1038/d41573-020-00078-0. [DOI] [PubMed] [Google Scholar]
- 57.Setten R.L., Lightfoot H.L., Habib N.A., Rossi J.J. Development of MTL-CEBPA: small activating RNA drug for hepatocellular carcinoma. Curr. Pharm. Biotechnol. 2018;19:611–621. doi: 10.2174/1389201019666180611093428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Chu Y., Yue X., Younger S.T., Janowski B.A., Corey D.R. Involvement of argonaute proteins in gene silencing and activation by RNAs complementary to a non-coding transcript at the progesterone receptor promoter. Nucleic Acids Res. 2010;38:7736–7748. doi: 10.1093/nar/gkq648. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Meng X., Jiang Q., Chang N., Wang X., Liu C., Xiong J., Cao H., Liang Z. Small activating RNA binds to the genomic target site in a seed-region-dependent manner. Nucleic Acids Res. 2016;44:2274–2282. doi: 10.1093/nar/gkw076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Portnoy V., Lin S.H.S., Li K.H., Burlingame A., Hu Z.H., Li H., Li L.C. SaRNA-guided Ago2 targets the RITA complex to promoters to stimulate transcription. Cell Res. 2016;26:320–335. doi: 10.1038/cr.2016.22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Weinmann L., Höck J., Ivacevic T., Ohrt T., Mütze J., Schwille P., Kremmer E., Benes V., Urlaub H., Meister G. Importin 8 is a gene silencing factor that targets argonaute proteins to distinct mRNAs. Cell. 2009;136:496–507. doi: 10.1016/j.cell.2008.12.023. [DOI] [PubMed] [Google Scholar]
- 62.Tang W., You W., Shi F., Qi T., Wang L., Djouder Z., Liu W., Zeng X. RNA helicase A acts as a bridging factor linking nuclear β-actin with RNA polymerase II. Biochem. J. 2009;420:421–428. doi: 10.1042/BJ20090402. [DOI] [PubMed] [Google Scholar]
- 63.Janowski B.A., Younger S.T., Hardy D.B., Ram R., Huffman K.E., Corey D.R. Activating gene expression in mammalian cells with promoter-targeted duplex RNAs. Nat. Chem. Biol. 2007;3:166–173. doi: 10.1038/nchembio860. [DOI] [PubMed] [Google Scholar]
- 64.Whitson J.M., Noonan E.J., Pookot D., Place R.F., Dahiya R. Double stranded-RNA-mediated activation of P21 gene induced apoptosis and cell cycle arrest in renal cell carcinoma. Int. J. Cancer. 2009;125:446–452. doi: 10.1002/ijc.24370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Tseng H.H., Hwang Y.H., Yeh K.T., Chang J.G., Chen Y.L., Yu H.S. Reduced expression of C/EBPα protein in hepatocellular carcinoma is associated with advanced tumor stage and shortened patient survival. J. Cancer Res. Clin. Oncol. 2009;135:241–247. doi: 10.1007/s00432-008-0448-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Reebye V., Huang K.W., Lin V., Jarvis S., Cutilas P., Dorman S., Ciriello S., Andrikakou P., Voutila J., Saetrom P., et al. Gene activation of CEBPA using saRNA: preclinical studies of the first in human saRNA drug candidate for liver cancer. Oncogene. 2018;37:3216–3228. doi: 10.1038/s41388-018-0126-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Nagafuchi A., Shirayoshi Y., Okazaki K., Yasuda K., Takeichi M. Transformation of cell adhesion properties by exogenously introduced E-cadherin cDNA. Nature. 1987;329:341–343. doi: 10.1038/329341a0. [DOI] [PubMed] [Google Scholar]
- 68.Ozawa M., Baribault H., Kemler R. The cytoplasmic domain of the cell adhesion molecule uvomorulin associates with three independent proteins structurally related in different species. EMBO J. 1989;8:1711–1717. doi: 10.1002/j.1460-2075.1989.tb03563.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.MacDonald B.T., Tamai K., He X. Wnt/β-Catenin signaling: components, mechanisms, and diseases. Dev. Cell. 2009;17:9–26. doi: 10.1016/j.devcel.2009.06.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Jackson L.A., Anderson E.J., Rouphael N.G., Roberts P.C., Makhene M., Coler R.N., McCullough M.P., Chappell J.D., Denison M.R., Stevens L.J., et al. An mRNA vaccine against SARS-CoV-2 — preliminary report. N. Engl. J. Med. 2020;383:1920–1931. doi: 10.1056/NEJMoa2022483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Corbett K.S., Flynn B., Foulds K.E., Francica J.R., Boyoglu-Barnum S., Werner A.P., Flach B., O’Connell S., Bock K.W., Minai M., et al. Evaluation of the mRNA-1273 vaccine against SARS-CoV-2 in nonhuman primates. N. Engl. J. Med. 2020;383:1544–1555. doi: 10.1056/NEJMoa2024671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Polack F.P., Thomas S.J., Kitchin N., Absalon J., Gurtman A., Lockhart S., Perez J.L., Pérez Marc G., Moreira E.D., Zerbini C., et al. Safety and efficacy of the BNT162b2 mRNA covid-19 vaccine. N. Engl. J. Med. 2020;383:2603–2615. doi: 10.1056/NEJMoa2034577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Malone R.W., Felgner P.L., Verma I.M. Cationic liposome-mediated RNA transfection. Proc. Natl. Acad. Sci. USA. 1989;86:6077–6081. doi: 10.1073/pnas.86.16.6077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Karikó K., Ni H., Capodici J., Lamphier M., Weissman D. mRNA is an endogenous ligand for toll-like receptor 3. J. Biol. Chem. 2004;279:12542–12550. doi: 10.1074/jbc.M310175200. [DOI] [PubMed] [Google Scholar]
- 75.Karikó K., Buckstein M., Ni H., Weissman D. Suppression of RNA recognition by Toll-like receptors: the impact of nucleoside modification and the evolutionary origin of RNA. Immunity. 2005;23:165–175. doi: 10.1016/j.immuni.2005.06.008. [DOI] [PubMed] [Google Scholar]
- 76.Kormann M.S.D., Hasenpusch G., Aneja M.K., Nica G., Flemmer A.W., Herber-Jonat S., Huppmann M., Mays L.E., Illenyi M., Schams A., et al. Expression of therapeutic proteins after delivery of chemically modified mRNA in mice. Nat. Biotechnol. 2011;29:154–157. doi: 10.1038/nbt.1733. [DOI] [PubMed] [Google Scholar]
- 77.Shatkin A.J. Capping of eucaryotic mRNAs. Cell. 1976;9:645–653. doi: 10.1016/0092-8674(76)90128-8. [DOI] [PubMed] [Google Scholar]
- 78.Bangel-Ruland N., Tomczak K., Fernández Fernández E., Leier G., Leciejewski B., Rudolph C., Rosenecker J., Weber W.-M. Cystic fibrosis transmembrane conductance regulator-mRNA delivery: a novel alternative for cystic fibrosis gene therapy. J. Gene Med. 2013;15:414–426. doi: 10.1002/jgm.2748. [DOI] [PubMed] [Google Scholar]
- 79.Karikó K., Muramatsu H., Welsh F.A., Ludwig J., Kato H., Akira S., Weissman D. Incorporation of pseudouridine into mRNA yields superior nonimmunogenic vector with increased translational capacity and biological stability. Mol. Ther. 2008;16:1833–1840. doi: 10.1038/mt.2008.200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Van Tendeloo V.F., Van de Velde A., Van Driessche A., Cools N., Anguille S., Ladell K., Gostick E., Vermeulen K., Pieters K., Nijs G., et al. Induction of complete and molecular remissions in acute myeloid leukemia by Wilms’ tumor 1 antigen-targeted dendritic cell vaccination. Proc. Natl. Acad. Sci. USA. 2010;107:13824–13829. doi: 10.1073/pnas.1008051107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Hewitt S.L., Bailey D., Zielinski J., Apte A., Musenge F., Karp R., Burke S., Garcon F., Mishra A., Gurumurthy S., et al. Intratumoral IL12 mRNA therapy promotes TH1 transformation of the tumor microenvironment. Clin. Cancer Res. 2020;26:6284–6298. doi: 10.1158/1078-0432.CCR-20-0472. [DOI] [PubMed] [Google Scholar]
- 82.Hewitt S.L., Bai A., Bailey D., Ichikawa K., Zielinski J., Karp R., Apte A., Arnold K., Zacharek S.J., Iliou M.S., et al. Durable anticancer immunity from intratumoral administration of IL-23, IL-36γ, and OX40L mRNAs. Sci. Transl. Med. 2019;11:9143. doi: 10.1126/scitranslmed.aat9143. [DOI] [PubMed] [Google Scholar]
- 83.Robinson E., MacDonald K.D., Slaughter K., McKinney M., Patel S., Sun C., Sahay G. Lipid nanoparticle-delivered chemically modified mRNA restores chloride secretion in cystic fibrosis. Mol. Ther. 2018;26:2034–2046. doi: 10.1016/j.ymthe.2018.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Islam M.A., Xu Y., Tao W., Ubellacker J.M., Lim M., Aum D., Lee G.Y., Zhou K., Zope H., Yu M., et al. Restoration of tumour-growth suppression in vivo via systemic nanoparticle-mediated delivery of PTEN mRNA. Nat. Biomed. Eng. 2018;2:850–864. doi: 10.1038/s41551-018-0284-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Lin Y.X., Wang Y., Ding J., Jiang A., Wang J., Yu M., Blake S., Liu S., Bieberich C.J., Farokhzad O.C., et al. Reactivation of the tumor suppressor PTEN by mRNA nanoparticles enhances antitumor immunity in preclinical models. Sci. Transl. Med. 2021;13:eaba9772. doi: 10.1126/scitranslmed.aba9772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Liu X., Kim C.N., Yang J., Jemmerson R., Wang X. Induction of apoptotic program in cell-free extracts: requirement for dATP and cytochrome c. Cell. 1996;86:147–157. doi: 10.1016/s0092-8674(00)80085-9. [DOI] [PubMed] [Google Scholar]
- 87.Khaled A.R., Kim K., Hofmeister R., Muegge K., Durum S.K. Withdrawal of IL-7 induces Bax translocation from cytosol to mitochondria through a rise in intracellular pH. Proc. Natl. Acad. Sci. USA. 1999;96:14476–14481. doi: 10.1073/pnas.96.25.14476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Okumura K., Nakase M., Inui M., Nakamura S., Watanabe Y., Tagawa T. Bax mRNA therapy using cationic liposomes for human malignant melanoma. J. Gene Med. 2008;10:910–917. doi: 10.1002/jgm.1214. [DOI] [PubMed] [Google Scholar]
- 89.Trepotec Z., Lichtenegger E., Plank C., Aneja M.K., Rudolph C. Delivery of mRNA therapeutics for the treatment of hepatic diseases. Mol. Ther. 2019;27:794–802. doi: 10.1016/j.ymthe.2018.12.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Pardi N., Tuyishime S., Muramatsu H., Kariko K., Mui B.L., Tam Y.K., Madden T.D., Hope M.J., Weissman D. Expression kinetics of nucleoside-modified mRNA delivered in lipid nanoparticles to mice by various routes. J. Control. Release. 2015;217:345–351. doi: 10.1016/j.jconrel.2015.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Pardi N., Secreto A.J., Shan X., Debonera F., Glover J., Yi Y., Muramatsu H., Ni H., Mui B.L., Tam Y.K., et al. Administration of nucleoside-modified mRNA encoding broadly neutralizing antibody protects humanized mice from HIV-1 challenge. Nat. Commun. 2017;8:14630. doi: 10.1038/ncomms14630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Rizvi F., Everton E., Smith A.R., Liu H., Osota E., Beattie M., Tam Y., Pardi N., Weissman D., Gouon-Evans V. Murine liver repair via transient activation of regenerative pathways in hepatocytes using lipid nanoparticle-complexed nucleoside-modified mRNA. Nat. Commun. 2021;12:613. doi: 10.1038/s41467-021-20903-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Sahin U., Karikó K., Türeci Ö. MRNA-based therapeutics-developing a new class of drugs. Nat. Rev. Drug Discov. 2014;13:759–780. doi: 10.1038/nrd4278. [DOI] [PubMed] [Google Scholar]
- 94.Juliano R.L. The delivery of therapeutic oligonucleotides. Nucleic Acids Res. 2016;44:6518–6548. doi: 10.1093/nar/gkw236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Roberts T.C., Langer R., Wood M.J.A. Advances in oligonucleotide drug delivery. Nat. Rev. Drug Discov. 2020;19:673–694. doi: 10.1038/s41573-020-0075-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Tanowitz M., Hettrick L., Revenko A., Kinberger G.A., Prakash T.P., Seth P.P. Asialoglycoprotein receptor 1 mediates productive uptake of N-Acetylgalactosamine-conjugated and unconjugated phosphorothioate antisense oligonucleotides into liver hepatocytes. Nucleic Acids Res. 2017;45:12388–12400. doi: 10.1093/nar/gkx960. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Nair J.K., Willoughby J.L.S., Chan A., Charisse K., Alam M.R., Wang Q., Hoekstra M., Kandasamy P., Kelin A.V., Milstein S., et al. Multivalent N -acetylgalactosamine-conjugated siRNA localizes in hepatocytes and elicits robust RNAi-mediated gene silencing. J. Am. Chem. Soc. 2014;136:16958–16961. doi: 10.1021/ja505986a. [DOI] [PubMed] [Google Scholar]
- 98.Balwani M., Sardh E., Ventura P., Peiró P.A., Rees D.C., Stölzel U., Bissell D.M., Bonkovsky H.L., Windyga J., Anderson K.E., et al. Phase 3 trial of RNAi therapeutic givosiran for acute intermittent porphyria. N. Engl. J. Med. 2020;382:2289–2301. doi: 10.1056/NEJMoa1913147. [DOI] [PubMed] [Google Scholar]



