Next Article in Journal
Combined Cell Therapy in the Treatment of Neurological Disorders
Next Article in Special Issue
Intratumoural Cytochrome P450 Expression in Breast Cancer: Impact on Standard of Care Treatment and New Efforts to Develop Tumour-Selective Therapies
Previous Article in Journal
Genetic and Acquired Heterotopic Ossification: A Translational Tale of Mice and Men
Previous Article in Special Issue
8-Hydroxydaidzein, an Isoflavone from Fermented Soybean, Induces Autophagy, Apoptosis, Differentiation, and Degradation of Oncoprotein BCR-ABL in K562 Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Anti-c-myc RNAi-Based Onconanotherapeutics

Nano-Gene and Drug Delivery Group, Discipline of Biochemistry, University of KwaZulu-Natal, Private Bag, Durban X54001, South Africa
*
Author to whom correspondence should be addressed.
Biomedicines 2020, 8(12), 612; https://doi.org/10.3390/biomedicines8120612
Submission received: 17 November 2020 / Revised: 3 December 2020 / Accepted: 5 December 2020 / Published: 15 December 2020

Abstract

:
Overexpression of the c-myc proto-oncogene features prominently in most human cancers. Early studies established that inhibiting the expression of oncogenic c-myc, produced potent anti-cancer effects. This gave rise to the notion that an appropriate c-myc silencing agent might provide a broadly applicable and more effective form of cancer treatment than is currently available. The endogenous mechanism of RNA interference (RNAi), through which small RNA molecules induce gene silencing by binding to complementary mRNA transcripts, represents an attractive avenue for c-myc inhibition. However, the development of a clinically viable, anti-c-myc RNAi-based platform is largely dependent upon the design of an appropriate carrier of the effector nucleic acids. To date, organic and inorganic nanoparticles were assessed both in vitro and in vivo, as carriers of small interfering RNA (siRNA), DICER-substrate siRNA (DsiRNA), and short hairpin RNA (shRNA) expression plasmids, directed against the c-myc oncogene. We review here the various anti-c-myc RNAi-based nanosystems that have come to the fore, especially between 2005 and 2020.

1. Introduction

The c-myc gene encodes a nuclear phosphoprotein that is widely recognized for its role as a transcription factor. The c-Myc protein is believed to participate in the regulation of 10–15% of all genes [1]. These include genes involved in cell cycle progression [2,3], metabolism [4], cell growth [5,6,7], differentiation [8], adhesion [9], and apoptosis [10].
Due to its function in regulating essential cellular functions, expression of the c-myc gene and activity of the c-Myc protein is, under normal circumstances, tightly controlled. Control points include the transcriptional regulation of the c-myc gene itself [11,12], the activity of translation initiation factor eIF4E, which ensures that only faithful c-myc mRNA transcripts are exported to the cytoplasm [13]; the short half-life of c-myc mRNA [14], post-translational modifications such as phosphorylation, acetylation, and ubiquitinylation [15]; and proteins which either directly interact with c-Myc [16] or influence dimerization with its obligate partner protein, Max [17]. As shown in Figure 1, c-myc functions in response to signals from several ligand membrane receptor complexes, which cause either positive or negative regulation. When associated with Max, c-Myc binds to DNA E-boxes and this, in turn, regulates the transcription of its target genes [18].
Abnormal c-myc expression can occur due to genetic events that include translocations [19], rearrangements [20], and amplification [21], as well as flaws in the pathways implicated in the regulation of this gene or the protein that it encodes [22]. Research carried out in the 1980s showed an association between the deregulated expression of c-myc and tumorigenesis [23,24]. Further work showed that abnormal c-myc expression causes neoplastic changes, by eliminating check-points in the cell cycle [25,26], prompting genomic instability [27], and through association with other oncogenes [28,29]. In fact, tumor cells often rely on c-myc expression for the maintenance of the cancerous state. This phenomenon, known as the oncogene addiction, was emphasized by studies that showed that c-myc inactivation caused tumor regression in transgenic mice [30], by inhibiting the cellular proliferation and inducing senescence or apoptosis and differentiation [31]. Moreover, the effects of systemic c-myc inhibition were found to be mild in normal tissues, and were well tolerated over time [32]. These findings, together with an estimation that c-myc is deregulated in up to 70% of human cancers [18], making it the most frequently altered oncogene, motivate strongly for the therapeutic value of inhibiting oncogenic c-myc.
In theory, the oncogenic activity of c-myc can be eliminated by inhibiting the expression of the activated gene, inhibiting inter-protein associations that are critical for c-Myc function, or by disrupting pathways that support c-myc deregulation in cancer cells. This provided a basis for the design and evaluation of several potential anti-cancer strategies. The antisense oligonucleotides were featured in some of the earliest reports of c-myc inhibition [33,34,35]. The application of antisense technology to c-Myc inhibition expanded with nucleotide modifications designed to confer greater stability and specificity [36,37]. However, Nobel Prize-winning work that described an endogenous gene silencing mechanism, known as RNA interference (RNAi) [38], presented further possibilities.
Short RNA duplexes of 19–21 base pairs with 2 nucleotide 3′ overhangs, known as small interfering RNA (siRNA), are the key mediators of this pathway. siRNA associates with a network of cytoplasmic proteins to form the RNA-induced silencing complex (RISC), through which it guides the degradation of mRNA, bearing a complementary sequence [39]. Short dsRNA molecules, lacking the dinucleotide overhangs that typify siRNA, termed DICER-substrate siRNA (DsiRNA), can also induce RNAi, with a reportedly higher efficiency. These are processed by the enzyme DICER into siRNA molecules that associate with the RNAi machinery [40]. DNA-directed RNAi, a strategy that generates specific siRNA molecules in vivo, is a useful gene-silencing tool [41]. This involves the construction of a RNA pol-driven plasmid expression vector, into which an antigene sequence of at least 19 nucleotides is inserted, together with appropriate termination signals. When introduced into cells, the antigene sequence is transcribed in the nucleus as a stem-loop structure, which is essentially 2 complementary sequences, 19–22 ribonucleotides in length, linked by a short loop of 4–11 ribonucleotides. This is known as short hairpin RNA (shRNA). The shRNA is exported to the cytoplasm where it is processed into siRNA by DICER [42]. In theory, effective silencing of c-myc, or any oncogene, might be achieved using endogenous cellular machinery, provided that the appropriately designed effector nucleic acid is successfully introduced. However, several factors militate against the success of naked nucleic acids in vivo. Naked nucleic acids are highly susceptible to serum nucleases [43] and are rapidly cleared by the kidneys [44]. However, studies have reported that chemical modifications such 2′-O-methylation of the guide strand [45], and the use of a passenger 3′ 19ntDNA/siRNA construct (previously referred to as “crook siRNA”), endowed the nucleic acid with nuclease resistance [46].
Furthermore, the size and net negative charge prevent passage across biological membranes [47]. Much effort was focused on the design of delivery agents that would mask its negative charge, protect its integrity, prevent its early removal from the body and facilitate cellular entry. In this regard, nano delivery systems received considerable attention, many of which are based on the principle that nucleic acids can electrostatically associate with positively charged agents [48]. This review discusses the potential application of anti-c-myc RNAi nanosystems in cancer treatment.

2. Anti-c-myc-siRNA

2.1. Lipid-Based Nanosystems

The liposome is arguably the least complicated lipid-based delivery agent—the simplest of which is a self-assembled phospholipid bilayer that encircles an aqueous core in which a variety of molecules might be entrapped [49]. It is this carrying capability that was exploited for the delivery of several therapeutically important molecules, including siRNA. A neutral liposome composed of dioleoylphosphatidylcholine (DOPC), cholesterol (Chol), and distearoylphosphatidylethanolamine- poly(ethylene glycol) (DSPE-PEG) was used to encapsulate and deliver anti-c-myc siRNA in vivo [50]. Pegylation, the introduction of the PEG polymer, served to create a hydration shell around the liposome that sterically inhibits adverse interparticle associations that reduce nanoparticle longevity in the body [51]. Systemic administration of the DOPC/Chol/DSPE-PEG/siRNA complex reduced the growth of ovarian cancer xenograft tumors and did not inhibit the growth of cells with low c-myc expression [41]. Anti-c-myc siRNA delivered via pegylated DOPC liposomes also showed promise in the treatment of cisplatin-resistant ovarian tumors [52]. Figure 2 provides a representation of some lipid-based delivery systems for anti-c-myc-siRNA.
Felgner et al. [53] first reported that the hydration of a mixture containing a synthetic cationic lipid and zwitterionic phospholipid create vesicles that bear a net positive charge, and paved the way for the use of cationic liposomes in nucleic acid delivery. Unlike neutral liposomes in which the siRNA must be encapsulated, cationic liposomes electrostatically associate with siRNA to form nanostructures, known as lipoplexes [54]. Early experiments involved the use of a commercially available cationic liposomal reagent, LipofectamineTM 2000, to demonstrate the therapeutic value of siRNA-mediated c-myc inhibition in human colon cancer [55]. Later, liposomes prepared from equimolar quantities of the cationic lipid N,N-dimethylaminopropylamidosuccinyl- cholesterylformylhydrazide (MS09), and Chol proved to be simple, but effective anti-c-myc agents, which elicited apoptotic cancer cell death and loss of migratory potential in colorectal and breast carcinoma cell lines [56].
Besides being limited to use in cationic liposome formulations, cationic lipids contributed to the development of more elaborate lipid nanoparticles. For example, Chen et al. [57] used a traditional cationic liposome made up of 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP) and Chol, to envelope a core of protamine-bound anti-c-myc siRNA and calf thymus DNA (Figure 2). This is known as a liposome-polycation-DNA (LPD) nanoparticle. Surface modifications included post-inserted PEG chains for steric stabilization and a peptide directed to aminopeptidase N, that is overexpressed by cancer cells. Effective siRNA delivery, c-myc inhibition, and tumor cell apoptosis were noted after these nanoparticles were intravenously administered in a xenograft model. Co-formulation of the anti-cancer drug doxorubicin with siRNA in targeted LPD nanoparticles further improved treatment efficacy [57]. Following the concept of stabilized core/shell lipid nano-assemblies, Zhang et al. [58] used a DOTAP/Chol/PEG formulation as the outer coating of a calcium phosphate core containing anti-c-myc siRNA (Figure 2). The resulting lipid calcium phosphate (LCP) nanoparticle was directed to sigma receptor-positive tumor cells by attachment of anisamide to the distal ends of PEG chains. Similar to the findings of Chen et al. [57], co-encapsulation of anti-c-myc siRNA and a chemotherapeutic agent, in this case, gemcitabine, gave a more pronounced anti-cancer effect.
Physical agents might prove useful in promoting the deposition of systemically introduced liposomal anti-c-myc siRNA nanoparticles in tumors. A tumor-targeted formulation of 3β[N-(N′,N′-dimethylaminoethane)-carbamoyl] cholesterol (DC-Chol), Chol, and DSPE-PEG with a photolabile-caged cell-penetrating peptide was used to deliver anti-c-myc siRNA. The application of near-infrared light at the tumor site, activated the cell-penetrating ability of the peptide [59]. Liposomes were also used as ultrasound cavitation agents for site-specific release of anti-c-myc siRNA conjugated to a cell-penetrating peptide [60]. In both instances, treatment delayed tumor progression in fibrosarcoma xenograft models.
Anti-c-myc siRNA was included in multi-targeted anti-cancer strategies, which involve the combined delivery of siRNAs against several genes implicated in cancer. A mixture of siRNAs against c-myc, MDM2, and VEGF was shown to inhibit tumor growth more effectively than the individual siRNAs [61]. Li et al. [62] co-encapsulated siRNA molecules against the same targets in a pegylated LPD nanocarrier, for systemic administration in a murine model of metastatic lung cancer. This treatment simultaneously silenced all three genes in cancerous tissue, reduced metastasis by approximately 80%, and extended survival time, with minimal toxicity. Similar results were obtained when siRNAs against the aforementioned oncogenes were pooled in pegylated LCP nanoparticles [63]. Later, a mechanistic study showed that this system impaired the growth of tumors in mice, by simultaneously inhibiting cell proliferation and angiogenesis [64].
Besides delivery via synthetic lipid vesicles, it is worth mentioning that siRNA can also be loaded in exosomes. Exosomes are vesicles that are naturally released by cells for the purposes of intercellular communication and represent an emerging nanocarrier system for a variety of medically relevant molecules [65]. The potential for exosome-mediated anti-c-myc siRNA delivery was demonstrated by Lunavat et al. [66] in vitro.

2.2. Miscellaneous Organic Nanosystems

Other organic anti-c-myc nano delivery systems reported are often complex polymer- and peptide-based nanocomposites. Folate-targeted, pegylated chitosan nanoparticles were used to encapsulate anti-c-myc siRNA associated with packaging RNA, to give a dual-targeting anti-tumor system that improved cellular uptake, gene silencing, and cancer cell death [67]. As a further example, Raichur et al. [68] used a layer-by-layer approach to associate anti-c-myc siRNA with poly(lactic-co-glycolic acid) hollow nanoparticles. In vitro experiments showed that the nanoparticles were taken up by aggressive cancer cells and reduced c-myc expression with loss of cell viability. More recently, Misra et al. [69] achieved approximately 90% growth inhibition in human melanoma with a nano assembly of palmitoyl-bioconjugated acetyl coenzyme-A termed “siNozyme”, which co-delivered anti-c-myc siRNA and the chemotherapeutic agent, amonafide.
Anti-c-myc siRNA is associated with cell-penetrating peptides (CPP), the simplest of which is an epidermal growth factor receptor-targeted fusion peptide, SPACE–EGF, for topical application to skin cancers [70]. A more complex peptide assembly that contained cationic peptides for siRNA-binding, pH-sensitive peptides for endosomal escape, and a tumor-targeting motif was used for the simultaneous delivery of siRNA against c-myc and Stat3. This system markedly reduced anchorage-independent growth in recalcitrant breast cancer cells [71]. An elaborate system consisting of a disulfide linked anti-c-myc siRNA-CPP encapsulated by a thermosensitive liposome, decorated with a tumor-targeting peptide motif showed effective c-myc silencing and antitumor activity in a fibrosarcoma xenograft model [72]. In a related study, an anti-c-myc siRNA-CPP conjugate contained within thermal and magnetic dual-responsive liposomes gave encouraging results in a murine breast cancer model [73].

2.3. Inorganic Nanosystems

The use of inorganic nanoparticles in siRNA delivery was explored in recent years. These are often modified with organic components to improve surface properties and reduce toxicity [74,75,76,77,78]. Attachment of siRNA involves either covalent conjugation or electrostatic association with positively charged groups introduced on the surface of the nanoparticle [79,80]. Anti-c-myc siRNA carried by PEG- [81], poly(ethylene imine)- [82], and chitosan- [83] functionalized gold nanoparticles was shown to reduce c-myc expression in human cervical, liver, and breast cancer cell lines, respectively. In separate in vivo experiments, gold nanoparticles modified with cationic [84] and anionic polymer shells [85], glucose residues [86], and an RGD tumor-targeting peptide [87], delivered anti-c-myc siRNA and suppressed the growth of lung tumors.
Nanoparticles based on selenium [77,88] and graphene oxide [82] was also introduced as potential carriers of anti-c-myc siRNA. Huang et al. [88] modified a doxorubicin selenium core with RGD-linked polyamidoamine for cancer cell-specific combination therapy, using anti-c-myc siRNA. The resultant nanostructure was serum-stable, successfully penetrated the blood–brain barrier and inhibited the growth of glioblastoma spheroids in vitro. In the same year, Imani et al. [89] showed that nano-graphene oxide with PEG and octaarginine conjugation effectively delivered anti-c-myc siRNA to human breast cancer cell lines, due to its superior stability and cell-penetrating ability. Some proof of principle studies using nanoparticles such as gold [78], selenium [77], and hydrotalcites [90], showed the potential for the delivery of anti-Luc-siRNA, paving the way for the delivery of other therapeutic siRNA molecules, including anti-c-myc siRNA.

3. DsiRNA

Like conventional siRNA, DsiRNA requires a vehicle for successful entry. Of significance to this discussion is the fact that pharmaceutical company, Dicerna, reported on a DsiRNA specific for the c-myc oncogene, DCR-MYC, delivered using a proprietary EnCore lipid nanoparticle. DCR-MYC in this delivery platform is the first, and only anti-c-myc RNAi system, to date, to have reached clinical trials [91]. Although the outcome of the initial trial was encouraging, a subsequent trial showed an unsatisfactory knockdown efficiency and its development was discontinued [92].

4. Anti-c-myc-shRNA

Most experiments with anti-c-myc shRNA plasmids involved their introduction into cells in culture, with the aid of commercial cationic lipid transfection reagents. In one such study, plasmid-driven anti-c-myc shRNA silenced c-myc expression by as much as 80%, reduced the colony-forming ability, and promoted apoptosis in MCF-7 breast cancer cells [93]. A similar plasmid system impaired proliferation, invasion, and motility in the hepatocellular carcinoma cell line, HepG2 [94]. The transfection of colon cancer cells with anti-c-myc shRNA plasmids not only reduced c-myc expression, but also that of the human telomerase reverse transcriptase gene (hTERT), which is under the transcriptional regulation of c-myc, and also contributes towards carcinogenesis, when abnormally expressed [95].
As with siRNA, the effect of multigene silencing using shRNA expression plasmids was also explored. A single plasmid was engineered to direct the transcription of shRNAs against c-myc, VEGF, hTERT, and BIRC5, which encodes Survivin. This produced a more effective anti-cancer effect than shRNA plasmids targeting individual oncogenes [96]. Similarly, Tai et al. [97] observed a synergistic anti-cancer effect, in colon cancer cells, when the cells were co-transfected with two shRNA plasmids, each separately targeting c-myc and VEGF. The field of anti-c-myc RNAi also benefitted from advances in the design of shRNA-encoding vectors. Recently Cheng et al. [98] used branched PCR technology to introduce a multisite-targeting c-Myc shRNA array into DNA nanovectors that reduced cellular c-myc mRNA levels by approximately 98%.
Thus far, only one in vivo experiment with anti-c-myc shRNA was reported. In this study, a poly(ethylene imine)-grafted polyglycidal methacrylate nanoparticle was used as a carrier of the shRNA expression vector. Anti-c-myc shRNA delivered in this manner suppressed tumor growth in murine models of breast and colon cancer [99].
Table 1 provides a summary of the anti-c-myc RNAi-based systems developed to date. The systems discussed were selectively used for the delivery of anti-c-myc RNAi molecules. Nano-delivery systems such as mesoporous silica nanoparticles [100,101] and magnetic nanoparticles [102,103] are not discussed in this review, but showed potential in drug delivery, which can be considered for as future carriers of siRNA, shRNA, or DsiRNA.

5. Conclusions

Anti-c-myc RNAi-based nanosystems have, in many instances, induced potent anti-cancer effects in vitro and in vivo. To date, only DsiRNA was evaluated as an alternative cancer treatment in clinical trials but did not progress further. Several groups and companies are pursuing the idea of inhibiting c-myc at the level of translation as a means of designing a clinically viable anti-c-myc agent. Hence, RNAi-based strategies are currently significant [93]. Although longer lasting oncogene inhibition can be achieved with DNA-directed RNAi [104], mature siRNA molecules are easily synthesized and pose fewer delivery concerns, as they are of lower molecular weight and do not require genome integration [48,105]. Hence, siRNA is considered more suitable for therapeutic use.
While gene expression might be interrupted by other means such as the restriction enzyme-based system, CRISPR/Cas9, RNAi is most likely the better strategy for c-myc inhibition. Given that the RNAi apparatus is present in all mammalian somatic cells, no prior genetic manipulation of the diseased cell line is needed [106]. This is a massive advantage because simple, transient transfection with anti-c-myc siRNA is sufficient to achieve anticancer activity [106,107,108]. Moreover, since RNAi occurs in the cytoplasm, there are no issues with chromatin accessibility, which can perturb gene-editing attempts with CRISPR/Cas9 technology. It is worth mentioning at this point that RNAi is not without its drawbacks, notably the occurrence of off-target effects. However, these are relatively easily attenuated by careful optimization of the design and dose of anti-c-myc siRNA molecules [106].
Research to date has emphasized that the development of a suitable anti-c-myc agent is largely dependent upon the design of an appropriate nano delivery system. Great strides were made since the first anti-c-myc oligomers were introduced in nanoparticle form [109,110,111]. siRNA, which functions catalytically and non-stoichiometrically, has surpassed the potency of the antisense oligomers. In recent years, nanosystems were used to deliver small molecule inhibitors of c-Myc-Max dimerization in prodrug form. Initially plagued by issues such as poor bioavailability, low solubility, rapid metabolism, and low potency; their incorporation into nanoparticles showed promise [112,113,114]. However, anti-c-myc RNAi nanotechnology is, at present, a more developed field, and presents a large body of knowledge upon which to improve.
Of all nucleic acid carriers explored thus far, the most significant development were made in the field of lipid-based delivery. Inorganic nanoplatforms, an emerging field, served to solidify the notion that anti-c-myc RNAi is a potent anti-cancer instrument. However, with systemic administration, RNAi nanoparticles still have significant challenges to overcome. Their entry into clinical trials might highlight difficulties that include poor retention time in the body and target-site penetration [115]. To this end, polymer and ligand-targeting modifications are common features of several anti-c-myc RNAi nanosystems. Such advances in nanoparticle design and expanding test systems for the newly developed nanoparticles represent additional avenues of research in this field. Hence, the advent of a clinically viable anti-c-myc RNAi-based anti-neoplastic agent is eagerly awaited.

Author Contributions

Conceptualization, S.H.; M.A. and M.S.; software, S.H.; resources, M.S.; writing—original draft preparation, S.H.; writing—review and editing, M.S. and M.A.; supervision, M.S. and M.A.; project administration, M.S.; funding acquisition, M.S. All authors have read and agreed to the published version of the manuscript.

Funding

This review received no external funding, but research in this area was funded by the National Research Foundation of South Africa, grant numbers 113850 and 120455.

Acknowledgments

The authors acknowledge members of the Nano-Gene and Drug Delivery group for their advice and technical support.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Zeller, K.I.; Zhao, X.; Lee, C.W.H.; Chiu, K.P.; Yao, F.; Yustein, J.T.; Ooi, H.S.; Orlov, Y.L.; Shahab, A.; Yong, H.C.; et al. Global mapping of c-Myc binding sites and target gene networks in human B cells. Proc. Natl. Acad. Sci. USA 2006, 103, 17834–17839. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Berns, K.; Hijmans, E.M.; Bernards, R. Repression of c-Myc responsive genes in cycling cells causes G1 arrest through reduction of cyclin E/CDK2 kinase activity. Oncogene 1997, 15, 1347–1356. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Hermeking, H.; Rago, C.; Schuhmacher, M.; Li, Q.; Barrett, J.F.; Obaya, A.; O’Connell, B.C.; Mateyak, M.K.; Tam, W.; Kohlhuber, F.; et al. Identification of CDK4 as a target of c-MYC. Proc. Natl. Acad. Sci. USA 2000, 97, 2229–2234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Kim, J.-W.; Zeller, K.I.; Wang, Y.; Jegga, A.G.; Aronow, B.J.; O’Donnell, K.A.; Dang, C.V. Evaluation of Myc E-Box Phylogenetic Footprints in Glycolytic Genes by Chromatin Immunoprecipitation Assays. Mol. Cell. Biol. 2004, 24, 5923–5936. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Liu, Y.-C.; Li, F.; Handler, J.; Huang, C.R.L.; Xiang, Y.; Neretti, N.; Sedivy, J.M.; I Zeller, K.; Dang, C.V. Global Regulation of Nucleotide Biosynthetic Genes by c-Myc. PLoS ONE 2008, 3, e2722. [Google Scholar] [CrossRef] [PubMed]
  6. Schuhmacher, M.; Staege, M.S.; Pajic, A.; Polack, A.; Weidle, U.H.; Bornkamm, G.W.; Eick, D.; Kohlhuber, F. Control of cell growth by c-Myc in the absence of cell division. Curr. Biol. 1999, 9, 1255–1258. [Google Scholar] [CrossRef] [Green Version]
  7. Van Riggelen, J.; Alper, S.L.; Felsher, D.W. MYC as a regulator of ribosome biogenesis and protein synthesis. Nat. Rev. Cancer 2010, 10, 301–309. [Google Scholar] [CrossRef] [PubMed]
  8. Wilson, A.; Murphy, M.J.; Oskarsson, T.; Kaloulis, K.; Bettess, M.D.; Oser, G.M.; Pasche, A.-C.; Knabenhans, C.; Macdonald, H.R.; Trumpp, A. c-Myc controls the balance between hematopoietic stem cell self-renewal and differentiation. Genes Dev. 2004, 18, 2747–2763. [Google Scholar] [CrossRef] [Green Version]
  9. Gebhardt, A.; Frye, M.; Herold, S.; Benitah, S.A.; Braun, K.; Samans, B.; Watt, F.M.; Elsässer, H.-P.; Eilers, M. Myc regulates keratinocyte adhesion and differentiation via complex formation with Miz1. J. Cell Biol. 2006, 172, 139–149. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Morrish, F.; Giedt, C.; Hockenbery, D. c-MYC apoptotic function is mediated by NRF-1 target genes. Genes Dev. 2003, 17, 240–255. [Google Scholar] [CrossRef] [Green Version]
  11. Eick, D.; Bornkamm, G.W. Transcriptional arrest within the first exon is a fast control mechanism in c-myc gene expression. Nucleic Acids Res. 1986, 14, 8331–8346. [Google Scholar] [CrossRef] [PubMed]
  12. Bentley, D.L.; Groudine, M. Novel promoter upstream of the human c-myc gene and regulation of c-myc expression in B-cell lymphomas. Mol. Cell. Biol. 1986, 6, 3481–3489. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Culjkovic, B.; Topisirovic, I.; Skrabanek, L.; Ruiz-Gutierrez, M.; Borden, K.L.B. eIF4E is a central node of an RNA regulon that governs cellular proliferation. J. Cell Biol. 2006, 175, 415–426. [Google Scholar] [CrossRef] [PubMed]
  14. Dani, C.; Blanchard, J.M.; Piechaczyk, M.; El Sabouty, S.; Marty, L.; Jeanteur, P. Extreme instability of myc mRNA in normal and transformed human cells. Proc. Natl. Acad. Sci. USA 1984, 81, 7046–7050. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Vervoorts, J.; Lüscher-Firzlaff, J.; Lüscher, B. The Ins and Outs of MYC Regulation by Posttranslational Mechanisms. J. Biol. Chem. 2006, 281, 34725–34729. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Dai, M.-S.; Arnold, H.; Sun, X.-X.; Sears, R.; Lü, H. Inhibition of c-Myc activity by ribosomal protein L11. EMBO J. 2009, 28, 993. [Google Scholar] [CrossRef] [Green Version]
  17. Grandori, C.; Cowley, S.M.; James, L.P.; Eisenman, R.N. The Myc/Max/Mad Network and the Transcriptional Control of Cell Behavior. Annu. Rev. Cell Dev. Biol. 2000, 16, 653–699. [Google Scholar] [CrossRef]
  18. Dang, C.V. MYC on the Path to Cancer. Cell 2012, 149, 22–35. [Google Scholar] [CrossRef] [Green Version]
  19. Battey, J.; Moulding, C.; Taub, R.; Murphy, W.; Stewart, T.; Potter, H.; Lenoir, G.; Leder, P. The human c-myc oncogene: Structural consequences of translocation into the igh locus in Burkitt lymphoma. Cell 1983, 34, 779–787. [Google Scholar] [CrossRef]
  20. Dalla-Favera, R.; Martinotti, S.; Gallo, R.C.; Erikson, J.; Croce, C.M. Translocation and rearrangements of the c-myc oncogene locus in human undifferentiated B-cell lymphomas. Science 1983, 219, 963–967. [Google Scholar] [CrossRef]
  21. Treszl, A.; Ádány, R.; Rákosy, Z.; Kardos, L.; Bégány, Á.; Gilde, K.; Balazs, M. Extra copies of c-mycare more pronounced in nodular melanomas than in superficial spreading melanomas as revealed by fluorescence in situ hybridisation. Cytom. Part B Clin. Cytom. 2004, 60, 37–46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Smith, D.R.; Myint, T.; Goh, H.S. Over-expression of the c-myc proto-oncogene in colorectal carcinoma. Br. J. Cancer 1993, 68, 407–413. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Adams, J.M.; Harris, A.W.; Pinkert, C.A.; Corcoran, L.M.; Alexander, W.S.; Cory, S.; Palmiter, R.D.; Brinster, R.L. The c-myc oncogene driven by immunoglobulin enhancers induces lymphoid malignancy in transgenic mice. Nat. Cell Biol. 1985, 318, 533–538. [Google Scholar] [CrossRef]
  24. Leder, A.; Pattengale, P.K.; Kuo, A.; Stewart, T.A.; Leder, P. Consequences of widespread deregulation of the c-myc gene in transgenic mice: Multiple neoplasms and normal development. Cell 1986, 45, 485–495. [Google Scholar] [CrossRef]
  25. Li, Q.; Dang, C.V. c-Myc Overexpression Uncouples DNA Replication from Mitosis. Mol. Cell. Biol. 1999, 19, 5339–5351. [Google Scholar] [CrossRef] [Green Version]
  26. Gil, J.J.; Kerai, P.P.; Lleonart, M.M.; Bernard, D.; Cigudosa, J.C.J.; Peters, G.G.; Carnero, A.; Beach, D.D. Immortalization of Primary Human Prostate Epithelial Cells by c-Myc. Cancer Res. 2005, 65, 2179–2185. [Google Scholar] [CrossRef] [Green Version]
  27. Kuzyk, A.; Mai, S. c-MYC-Induced Genomic Instability. Cold Spring Harb. Perspect. Med. 2014, 4, a014373. [Google Scholar] [CrossRef] [PubMed]
  28. Vaux, D.L.; Cory, S.; Adams, J.M. Bcl-2 gene promotes haemopoietic cell survival and cooperates with c-myc to immortalize pre-B cells. Nat. Cell Biol. 1988, 335, 440–442. [Google Scholar] [CrossRef]
  29. Wang, C.; Lisanti, M.P.; Liao, D.J. Reviewing once more the c-myc and Ras collaboration. Cell Cycle 2011, 10, 57–67. [Google Scholar] [CrossRef]
  30. Arvanitis, C.; Felsher, D.W. Conditional transgenic models define how MYC initiates and maintains tumorigenesis. Semin. Cancer Biol. 2006, 16, 313–317. [Google Scholar] [CrossRef]
  31. Felsher, D.W. MYC Inactivation Elicits Oncogene Addiction through Both Tumor Cell-Intrinsic and Host-Dependent Mechanisms. Genes Cancer 2010, 1, 597–604. [Google Scholar] [CrossRef]
  32. Soucek, L.; Whitfield, J.R.; Martins, C.P.; Finch, A.J.; Murphy, D.J.; Sodir, N.M.; Karnezis, A.N.; Swigart, L.B.; Nasi, S.; Evan, G.I. Modelling Myc inhibition as a cancer therapy. Nat. Cell Biol. 2008, 455, 679–683. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Holt, J.T.; Redner, R.L.; Nienhuis, A.W. An oligomer complementary to c-myc mRNA inhibits proliferation of HL-60 promyelocytic cells and induces differentiation. Mol. Cell. Biol. 1988, 8, 963–973. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Loke, S.L.; Stein, C.; Zhang, X.; Avigan, M.; Cohen, J.; Neckers, L.M. Delivery of c-myc Antisense Phosphorothioate Oligodeoxynucleotides to Hematopoietic Cells in Culture by Liposome Fusion: Specific Reduction in c-myc Protein Expression Correlates with Inhibition of Cell Growth and DNA Synthesis. Endogenous ADP-Ribosylation 1988, 141, 282–289. [Google Scholar] [CrossRef]
  35. Wickstrom, E.L.; Bacon, T.A.; Gonzalez, A.; Freeman, D.L.; Lyman, G.H.; Wickstrom, E. Human promyelocytic leukemia HL-60 cell proliferation and c-myc protein expression are inhibited by an antisense pentadecadeoxynucleotide targeted against c-myc mRNA. Proc. Natl. Acad. Sci. USA 1988, 85, 1028–1032. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Cutrona, G.; Carpaneto, E.M.; Ponzanelli, A.; Ulivi, M.; Millo, E.; Scarfì, S.; Ferrarini, M. Inhibition of the translocated c-myc in Burkitt’s lymphoma by a PNA Complementary to the Eμ Enhancer. Cancer Res. 2003, 63, 6144–6148. [Google Scholar] [PubMed]
  37. Hudziak, R.M.; Summerton, J.; Weller, D.D.; Iversen, P.L. Antiproliferative Effects of Steric Blocking Phosphorodiamidate Morpholino Antisense Agents Directed against c-myc. Antisense Nucleic Acid Drug Dev. 2000, 10, 163–176. [Google Scholar] [CrossRef]
  38. Fire, A.; Xu, S.; Montgomery, M.K.; Kostas, S.A.; Driver, S.E.; Mello, C.C. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature 1998, 391, 806–811. [Google Scholar] [CrossRef]
  39. Elbashir, S.M.; Lendeckel, W.; Tuschl, T. RNA interference is mediated by 21- and 22-nucleotide RNAs. Genes Dev. 2001, 15, 188–200. [Google Scholar] [CrossRef] [Green Version]
  40. Kim, D.-H.; Behlke, M.A.; Rose, S.D.; Chang, M.-S.; Choi, S.; Rossi, J.J. Synthetic dsRNA Dicer substrates enhance RNAi potency and efficacy. Nat. Biotechnol. 2005, 23, 222–226. [Google Scholar] [CrossRef] [Green Version]
  41. Ambesajir, A.; Kaushik, A.; Kaushik, J.J.; Petros, S.T. RNA interference: A futuristic tool and its therapeutic applications. Saudi J. Biol. Sci. 2012, 19, 395–403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Taxman, D.J.; Moore, C.B.; Guthrie, E.H.; Huang, M.T.-H. Short Hairpin RNA (shRNA): Design, Delivery, and Assessment of Gene Knockdown. In Structural Genomics and Drug Discovery; Springer Science and Business Media LLC: New York, NY, USA, 2010; Volume 629, pp. 139–156. [Google Scholar]
  43. Cao, B.; Ji, A.-M. Construction of small interfering RNA targeting mouse vascular endothelial growth factor receptor-2: Its serum stability and gene silencing efficiency in vitro. J. South. Med. Univ. 2009, 29, 864–867. [Google Scholar]
  44. Huang, Y.; Hong, J.; Zheng, S.; Ding, Y.; Guo, S.; Zhang, H.; Zhang, X.; Du, Q.; Liang, Z. Elimination Pathways of Systemically Delivered siRNA. Mol. Ther. 2011, 19, 381–385. [Google Scholar] [CrossRef] [PubMed]
  45. Hoerter, J.A.H.; Walter, N.G. Chemical modification resolves the asymmetry of siRNA strand degradation in human blood serum. RNA 2007, 13, 1887–1893. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Allison, S.J.; Milner, J. RNA Interference by Single- and Double-stranded siRNA with a DNA Extension Containing a 3′ Nuclease-resistant Mini-hairpin Structure. Mol. Ther. Nucl. Acids 2014, 2, e141. [Google Scholar] [CrossRef] [Green Version]
  47. Akhtar, S.; Benter, I. Toxicogenomics of non-viral drug delivery systems for RNAi: Potential impact on siRNA-mediated gene silencing activity and specificity. Adv. Drug Deliv. Rev. 2007, 59, 164–182. [Google Scholar] [CrossRef]
  48. Wang, J.; Lu, Z.; Wientjes, M.G.; Au, J.L.-S. Delivery of siRNA Therapeutics: Barriers and Carriers. AAPS J. 2010, 12, 492–503. [Google Scholar] [CrossRef]
  49. Batzri, S.; Korn, E.D. Single bilayer liposomes prepared without sonication. Biochim. Biophys. Acta (BBA) Biomembr. 1973, 298, 1015–1019. [Google Scholar] [CrossRef]
  50. Reyes-González, J.M.; Armaiz-Peña, G.N.; Mangala, L.S.; Valiyeva, F.; Ivan, C.; Pradeep, S.; Echevarría-Vargas, I.M.; Rivera-Reyes, A.; Sood, A.K.; Vivas-Mejía, P.E. Targeting c-MYC in Platinum-Resistant Ovarian Cancer. Mol. Cancer Ther. 2015, 14, 2260–2269. [Google Scholar] [CrossRef] [Green Version]
  51. Suk, J.S.; Xu, Q.; Kim, N.; Hanes, J.; Ensign, L.M. PEGylation as a strategy for improving nanoparticle-based drug and gene delivery. Adv. Drug Deliv. Rev. 2016, 99, 28–51. [Google Scholar] [CrossRef] [Green Version]
  52. Vivas-Mejia, P.E.; Gonzalez, J.M.R.; Sood, A.K. Nanoliposomal c-MYC-siRNA Inhibits In Vivo Tumor Growth of Cisplatin-Resistant Ovarian Cancer. U.S. Patent No. 10,548,842, 4 February 2020. [Google Scholar]
  53. Felgner, P.L.; Gadek, T.R.; Holm, M.; Roman, R.; Chan, H.W.; Wenz, M.; Northrop, J.P.; Ringold, G.M.; Danielsen, M. Lipofection: A highly efficient, lipid-mediated DNA-transfection procedure. Proc. Natl. Acad. Sci. USA 1987, 84, 7413–7417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Khatri, N.; Baradia, D.; Vhora, I.; Rathi, M.; Misra, A. Development and Characterization of siRNA Lipoplexes: Effect of Different Lipids, In Vitro Evaluation in Cancerous Cell Lines and In Vivo Toxicity Study. AAPS PharmSciTech 2014, 15, 1630–1643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Zhang, X.; Ge, Y.; Tian, R.-H. The knockdown of c-myc expression by RNAi inhibits cell proliferation in human colon cancer HT-29 cells in vitro and in vivo. Cell. Mol. Biol. Lett. 2009, 14, 305–318. [Google Scholar] [CrossRef] [PubMed]
  56. Habib, S.; Daniels, A.; Ariatti, M.; Singh, M. Anti-c-myc cholesterol based lipoplexes as onco-nanotherapeutic agents in vitro. F1000Research 2020, 9, 770. [Google Scholar] [CrossRef]
  57. Chen, Y.; Wu, J.J.; Huang, L. Nanoparticles Targeted With NGR Motif Deliver c-myc siRNA and Doxorubicin for Anticancer Therapy. Mol. Ther. 2010, 18, 828–834. [Google Scholar] [CrossRef] [PubMed]
  58. Zhang, Y.; Peng, L.; Mumper, R.J.; Huang, L. Combinational delivery of c-myc siRNA and nucleoside analogs in a single, synthetic nanocarrier for targeted cancer therapy. Biomaterials 2013, 34, 8459–8468. [Google Scholar] [CrossRef] [Green Version]
  59. Yang, Y.; Yang, Y.; Xie, X.; Wang, Z.; Gong, W.; Zhang, H.; Li, Y.; Yu, F.; Li, Z.; Mei, X. Dual-modified liposomes with a two-photon-sensitive cell penetrating peptide and NGR ligand for siRNA targeting delivery. Biomaterials 2015, 48, 84–96. [Google Scholar] [CrossRef]
  60. Xie, X.; Lin, W.; Li, M.; Yang, Y.; Deng, J.; Liu, H.; Chen, Y.; Fu, X.; Liu, H.; Yang, Y. Efficient siRNA Delivery Using Novel Cell-Penetrating Peptide-siRNA Conjugate-Loaded Nanobubbles and Ultrasound. Ultrasound Med. Biol. 2016, 42, 1362–1374. [Google Scholar] [CrossRef]
  61. Song, E.; Zhu, P.; Lee, S.-K.; Chowdhury, D.; Kussman, S.; Dykxhoorn, D.M.; Feng, Y.; Palliser, D.; Weiner, D.B.; Shankar, P.; et al. Antibody mediated in vivo delivery of small interfering RNAs via cell-surface receptors. Nat. Biotechnol. 2005, 23, 709–717. [Google Scholar] [CrossRef]
  62. Li, S.-D.; Chono, S.; Huang, L. Efficient Oncogene Silencing and Metastasis Inhibition via Systemic Delivery of siRNA. Mol. Ther. 2008, 16, 942–946. [Google Scholar] [CrossRef]
  63. Yang, Y.; Li, J.; Liu, F.; Huang, L. Systemic Delivery of siRNA via LCP Nanoparticle Efficiently Inhibits Lung Metastasis. Mol. Ther. 2012, 20, 609–615. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Yang, Y.; Hu, Y.; Wang, Y.; Li, J.; Liu, F.; Huang, L. Nanoparticle Delivery of Pooled siRNA for Effective Treatment of Non-Small Cell Lung Caner. Mol. Pharm. 2012, 9, 2280–2289. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Ha, D.; Yang, N.; Nadithe, V. Exosomes as therapeutic drug carriers and delivery vehicles across biological membranes: Current perspectives and future challenges. Acta Pharm. Sin. B 2016, 6, 287–296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Lunavat, T.R.; Jang, S.C.; Nilsson, L.; Park, H.T.; Repiska, G.; Lässer, C.; Nilsson, J.A.; Gho, Y.S.; Lötvall, J. RNAi delivery by exosome-mimetic nanovesicles–Implications for targeting c-Myc in cancer. Biomaterials 2016, 102, 231–238. [Google Scholar] [CrossRef] [PubMed]
  67. Li, L.; Hu, X.; Zhang, M.; Ma, S.; Yu, F.; Zhao, S.; Liu, N.; Wang, Z.; Wang, Y.; Guan, H.; et al. Dual Tumor-Targeting Nanocarrier System for siRNA Delivery Based on pRNA and Modified Chitosan. Mol. Ther. Nucleic Acids 2017, 8, 169–183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Raichur, A.; Nakajima, Y.; Nagaoka, Y.; Matsumoto, K.; Mizuki, T.; Kato, K.; Maekawa, T.; Kumar, D.S. Strategist PLGA Nano-capsules to Deliver siRNA for Inhibition of Carcinoma and Neuroblastoma Cell Lines by Knockdown of MYC Proto-oncogene using CPPs and PNA. NanoWorld J. 2015, 1, 32–45. [Google Scholar] [CrossRef] [Green Version]
  69. Misra, S.K.; Kampert, T.L.; Pan, D. Nano-Assembly of Pamitoyl-Bioconjugated Coenzyme-A for Combinatorial Chemo-Biologics in Transcriptional Therapy. Bioconjugate Chem. 2018, 29, 1419–1427. [Google Scholar] [CrossRef]
  70. Ruan, R.; Chen, M.; Sun, S.; Wei, P.; Zou, L.; Liu, J.; Gao, D.; Wen, L.; Ding, W. Topical and Targeted Delivery of siRNAs to Melanoma Cells Using a Fusion Peptide Carrier. Sci. Rep. 2016, 6, 29159. [Google Scholar] [CrossRef] [Green Version]
  71. Bjorge, J.D.; Pang, A.; Fujita, D.J. Delivery of gene targeting siRNAs to breast cancer cells using a multifunctional peptide complex that promotes both targeted delivery and endosomal release. PLoS ONE 2017, 12, e0180578. [Google Scholar] [CrossRef]
  72. Yang, Y.; Yang, Y.; Xie, X.; Xu, X.; Xia, X.; Wang, H.; Li, L.; Dong, W.; Ma, P.; Liu, Y. Dual stimulus of hyperthermia and intracellular redox environment triggered release of siRNA for tumor-specific therapy. Int. J. Pharm. 2016, 506, 158–173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Yang, Y.; Xie, X.; Xu, X.; Xia, X.; Wang, H.; Li, L.; Dong, W.; Ma, P.; Yang, Y.; Liu, Y.; et al. Thermal and magnetic dual-responsive liposomes with a cell-penetrating peptide-siRNA conjugate for enhanced and targeted cancer therapy. Colloids Surf. B Biointerfaces 2016, 146, 607–615. [Google Scholar] [CrossRef] [PubMed]
  74. Chaudhary, S.; Umar, A.; Mehta, S.K. Surface functionalized selenium nanoparticles for biomedical applications. J. Biomed. Nanotechnol. 2014, 10, 3004–3042. [Google Scholar] [CrossRef] [PubMed]
  75. Fraga, S.; Brandão, A.; Soares, M.E.; Morais, T.; Duarte, J.A.; Pereira, L.; Soares, L.; Neves, C.S.; Pereira, E.; Bastos, M.D.L.; et al. Short- and long-term distribution and toxicity of gold nanoparticles in the rat after a single-dose intravenous administration. Nanomed. Nanotechnol. Biol. Med. 2014, 10, 1757–1766. [Google Scholar] [CrossRef] [PubMed]
  76. Zhou, J.; Ralston, J.; Sedev, R.; Beattie, D.A. Functionalized gold nanoparticles: Synthesis, structure and colloid stability. J. Colloid Interface Sci. 2009, 331, 251–262. [Google Scholar] [CrossRef] [PubMed]
  77. Maiyo, F.; Singh, M. Polymerized Selenium Nanoparticles for Folate-Receptor-Targeted Delivery of Anti-Luc-siRNA: Potential for Gene Silencing. Biomaterials 2020, 8, 76. [Google Scholar] [CrossRef] [Green Version]
  78. Mbatha, L.S.; Maiyo, F.C.; Singh, M. Dendrimer functionalized folate-targeted gold nanoparticles for luciferase gene silencing in vitro: A proof of principle study. Acta Pharm. 2019, 69, 49–61. [Google Scholar] [CrossRef] [Green Version]
  79. Conde, J.; Ambrosone, A.; Sanz, V.; Hernandez, Y.; Marchesano, V.; Tian, F.; Child, B.H.; Berry, C.C.; Ibarra, M.R.; Baptista, P.V.; et al. Design of Multifunctional Gold Nanoparticles for In Vitro and In Vivo Gene Silencing. ACS Nano 2012, 6, 8316–8324. [Google Scholar] [CrossRef]
  80. Xia, Y.; Guo, M.; Xu, T.; Li, Y.; Wang, C.; Lin, Z.; Zhao, M.; Zhu, B. siRNA-loaded selenium nanoparticle modified with hyaluronic acid for enhanced hepatocellular carcinoma therapy. Int. J. Nanomed. 2018, 13, 1539–1552. [Google Scholar] [CrossRef] [Green Version]
  81. McCully, M.; Hernandez, Y.; Conde, J.; Baptista, P.V.; De La Fuente, J.M.; Hursthouse, A.; Stirling, D.; Berry, C.C. Significance of the balance between intracellular glutathione and polyethylene glycol for successful release of small interfering RNA from gold nanoparticles. Nano Res. 2015, 8, 3281–3292. [Google Scholar] [CrossRef] [Green Version]
  82. Shaat, H.; Mostafa, A.; Moustafa, M.; Gamal-Eldeen, A.; Emam, A.; El-Hussieny, E.; Elhefnawi, M. Modified gold nanoparticles for intracellular delivery of anti-liver cancer siRNA. Int. J. Pharm. 2016, 504, 125–133. [Google Scholar] [CrossRef] [PubMed]
  83. Daniels, A.N.; Singh, M. Sterically stabilized siRNA:gold nanocomplexes enhance c-MYC silencing in a breast cancer cell model. Nanomedicine 2019, 14, 1387–1401. [Google Scholar] [CrossRef] [PubMed]
  84. Kim, H.S.; Son, Y.J.; Mao, W.; Leong, K.W.; Yoo, H.S. Atom Transfer Radical Polymerization of Multishelled Cationic Corona for the Systemic Delivery of siRNA. Nano Lett. 2017, 18, 314–325. [Google Scholar] [CrossRef] [PubMed]
  85. Mao, W.; Lee, S.; Shin, J.U.; Yoo, H.S. Surface-Initiated Atom Transfer Polymerized Anionic Corona on Gold Nanoparticles for Anti-Cancer Therapy. Pharmaceutics 2020, 12, 261. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Conde, J.; Tian, F.; Hernandez, Y.; Bao, C.; Baptista, P.V.; Cui, D.; Stoeger, T.; De La Fuente, J.M. RNAi-based glyconanoparticles trigger apoptotic pathways for in vitro and in vivo enhanced cancer-cell killing. Nanoscale 2015, 7, 9083–9091. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Conde, J.; Tian, F.; Hernández, Y.; Bao, C.; Cui, D.; Janssen, K.-P.; Ibarra, M.R.; Baptista, P.V.; Stoeger, T.; De La Fuente, J.M. In vivo tumor targeting via nanoparticle-mediated therapeutic siRNA coupled to inflammatory response in lung cancer mouse models. Biomaterials 2013, 34, 7744–7753. [Google Scholar] [CrossRef]
  88. Huang, W.; Liang, Y.; Sang, C.; Mei, C.; Li, X.; Chen, T. Therapeutic nanosystems co-deliver anticancer drugs and oncogene SiRNA to achieve synergetic precise cancer chemo-gene therapy. J. Mater. Chem. B 2018, 6, 3013–3022. [Google Scholar] [CrossRef] [PubMed]
  89. Imani, R.; Prakash, S.; Vali, H.; Faghihi, S. Polyethylene glycol and octa-arginine dual-functionalized nanographene oxide: An optimization for efficient nucleic acid delivery. Biomater. Sci. 2018, 6, 1636–1650. [Google Scholar] [CrossRef]
  90. Nundkumar, N.; Singh, S.; Singh, M. Amino Acid Functionalized Hydrotalcites for Gene Silencing. J. Nanosci. Nanotechnol. 2020, 20, 3387–3397. [Google Scholar] [CrossRef] [PubMed]
  91. Tolcher, A.W.; Papadopoulos, K.P.; Patnaik, A.; Rasco, D.W.; Martinez, D.; Wood, D.L.; Fielman, B.; Sharma, M.; Janisch, L.A.; Brown, B.D.; et al. Safety and activity of DCR-MYC, a first-in-class Dicer-substrate small interfering RNA (DsiRNA) targeting MYC, in a phase I study in patients with advanced solid tumors. J. Clin. Oncol. 2015, 33, 11006. [Google Scholar] [CrossRef]
  92. Whitfield, J.R.; Beaulieu, M.-E.; Soucek, L. Strategies to Inhibit Myc and Their Clinical Applicability. Front. Cell Dev. Biol. 2017, 5, 10. [Google Scholar] [CrossRef] [Green Version]
  93. Wang, Y.; Liu, S.; Zhang, G.; Zhou, C.-Q.; Zhu, H.-X.; Zhou, X.-B.; Quan, L.-P.; Bai, J.-F.; Xu, N. Knockdown of c-Myc expression by RNAi inhibits MCF-7 breast tumor cells growth in vitro and in vivo. Breast Cancer Res. 2004, 7, R220–R228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Zhao, Y.; Wang, J.; Gao, W.; Zheng, Y.-X.; Wang, Y.-K.; Zhou, Z.-Q.; Zhang, H.; Wang, C.-J. RNAi silencing of c-Myc inhibits cell migration, invasion, and proliferation in HepG2 human hepatocellular carcinoma cell line: C-Myc silencing in hepatocellular carcinoma cell. Cancer Cell Int. 2013, 13, 23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Huang, H.; Nancai, Y.; Lei, F.; Xiong, W.; Wen, S.; Guofu, H.; Yanxia, W.; Hanju, H.; Qian, L.; Hong, X. siRNA directed against c-Myc inhibits proliferation and downregulates human telomerase reverse transcriptase in human colon cancer Colo 320 cells. J. Exp. Clin. Cancer Res. 2008, 27, 27. [Google Scholar] [CrossRef] [Green Version]
  96. Song, Y.; Dong, M.-M.; Yang, H.-F. Effects of RNA interference targeting four different genes on the growth and proliferation of nasopharyngeal carcinoma CNE-2Z cells. Cancer Gene Ther. 2011, 18, 297–304. [Google Scholar] [CrossRef] [PubMed]
  97. Tai, J.; Wang, G.; Liu, T.; Wang, L.; Lin, C.; Li, F. Effects of siRNA Targeting c-Myc and VEGF on Human Colorectal Cancer Volo Cells. J. Biochem. Mol. Toxicol. 2012, 26, 499–505. [Google Scholar] [CrossRef] [PubMed]
  98. Cheng, L.; Ma, D.; Lu, L.; Ouyang, D.; Xi, Z. Building Customizable Multisite-Targeting c-Myc shRNA Array into Branch-PCR-Constructed DNA Nanovectors for Enhanced Tumor Cell Suppression. ChemistrySelect 2020, 5, 10250–10255. [Google Scholar] [CrossRef]
  99. Tangudu, N.K.; Verma, V.K.; Clemons, T.D.; Beevi, S.S.; Hay, T.; Mahidhara, G.; Raja, M.; Nair, R.A.; Alexander, L.E.; Patel, A.B.; et al. RNA Interference Using c-Myc-Conjugated Nanoparticles Suppresses Breast and Colorectal Cancer Models. Mol. Cancer Ther. 2015, 14, 1259–1269. [Google Scholar] [CrossRef] [Green Version]
  100. Moodley, T.; Singh, M. Polymeric Mesoporous Silica Nanoparticles for Enhanced Delivery of 5-Fluorouracil In Vitro. Pharmaceutics 2019, 11, 288. [Google Scholar] [CrossRef] [Green Version]
  101. Moodley, T.; Singh, M. Sterically Stabilised Polymeric Mesoporous Silica Nanoparticles Improve Doxorubicin Efficiency: Tailored Cancer Therapy. Molecules 2020, 25, 742. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Mngadi, S.; Mokhosi, S.; Singh, M.; Mdlalose, W. Chitosan-Functionalized Mg0.5Co0.5Fe2O4 Magnetic Nanoparticles Enhance Delivery of 5-Fluorouracil In Vitro. Coatings 2020, 10, 446. [Google Scholar] [CrossRef]
  103. Synthesis and characterization of magnetic nanoparticles of cobalt ferrite coated with silica. Biointerface Res. Appl. Chem. 2019, 10, 4908–4913. [CrossRef]
  104. Takahashi, Y.; Yamaoka, K.; Nishikawa, M.; Takakura, Y. Quantitative and Temporal Analysis of Gene Silencing in Tumor Cells Induced by Small Interfering RNA or Short Hairpin RNA Expressed from Plasmid Vectors. J. Pharm. Sci. 2009, 98, 74–80. [Google Scholar] [CrossRef] [PubMed]
  105. Xu, C.-F.; Wang, J. Delivery systems for siRNA drug development in cancer therapy. Asian J. Pharm. Sci. 2015, 10, 1–12. [Google Scholar] [CrossRef] [Green Version]
  106. Boettcher, M.; McManus, M.T. Choosing the Right Tool for the Job: RNAi, TALEN, or CRISPR. Mol. Cell 2015, 58, 575–585. [Google Scholar] [CrossRef] [Green Version]
  107. Unniyampurath, U.; Pilankatta, R.; Krishnan, M.N. RNA Interference in the Age of CRISPR: Will CRISPR Interfere with RNAi? Int. J. Mol. Sci. 2016, 17, 291. [Google Scholar] [CrossRef] [PubMed]
  108. Gabay, M.; Li, Y.; Felsher, D.W. MYC Activation Is a Hallmark of Cancer Initiation and Maintenance. Cold Spring Harb. Perspect. Med. 2014, 4, a014241. [Google Scholar] [CrossRef] [Green Version]
  109. Kanamaru, T.; Takagi, T.; Takakura, Y.; Hashida, M. Biological Effects and Cellular Uptake of c-mycAntisense Oligonucleotides and Their Cationic Liposome Complexes. J. Drug Target. 1998, 5, 235–245. [Google Scholar] [CrossRef]
  110. Putney, S.D.; Brown, J.; Cucco, C.; Lee, R.; Skorski, T.; Leonetti, C.; Geiser, T.; Calabretta, B.; Zupi, G.; Zon, G. Enhanced Anti-Tumor Effects with Microencapsulated c-mycAntisense Oligonucleotide. Antisense Nucleic Acid Drug Dev. 1999, 9, 451–458. [Google Scholar] [CrossRef]
  111. Carroll, J.S.; Swarbrick, A.; Musgrove, E.A.; Sutherland, R.L. Mechanisms of growth arrest by c-myc antisense oligonucleotides in MCF-7 breast cancer cells: Implications for the antiproliferative effects of antiestrogens. Cancer Res. 2002, 62, 3126–3131. [Google Scholar]
  112. Pan, D.; Kim, B.; Hu, G.; Gupta, D.S.; Senpan, A.; Yang, X.; Schmieder, A.; Swain, C.; A Wickline, S.; Tomasson, M.H.; et al. A strategy for combating melanoma with oncogenic c-Myc inhibitors and targeted nanotherapy. Nanomedicine 2015, 10, 241–251. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Soodgupta, D.; Pan, D.; Cui, G.; Senpan, A.; Yang, X.; Lu, L.; Weilbaecher, K.N.; Prochownik, E.V.; Lanza, G.M.; Tomasson, M.H. Small Molecule MYC Inhibitor Conjugated to Integrin-Targeted Nanoparticles Extends Survival in a Mouse Model of Disseminated Multiple Myeloma. Mol. Cancer Ther. 2015, 14, 1286–1294. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Esser, A.K.; Ross, M.H.; Fontana, F.; Su, X.; Gabay, A.; Fox, G.C.; Xu, Y.; Xiang, J.; Schmieder, A.H.; Yang, X.; et al. Nanotherapy delivery of c-myc inhibitor targets Protumor Macrophages and preserves Antitumor Macrophages in Breast Cancer. Theranostics 2020, 10, 7510–7526. [Google Scholar] [CrossRef] [PubMed]
  115. Sajid, M.I.; Moazzam, M.; Kato, S.; Cho, K.Y.; Tiwari, R.K. Overcoming Barriers for siRNA Therapeutics: From Bench to Bedside. Pharmaceutics 2020, 13, 294. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Transcription of the c-myc gene in normal cells occurs in response to signals from membrane-anchored receptors. White arrows show the direction of transcription (adapted from [18]).
Figure 1. Transcription of the c-myc gene in normal cells occurs in response to signals from membrane-anchored receptors. White arrows show the direction of transcription (adapted from [18]).
Biomedicines 08 00612 g001
Figure 2. Lipid-based delivery agents for anti-c-myc siRNA (a) pegylated, neutral liposome, (b) cationic liposome, (c) liposome-polycation-DNA (LPD) nanoparticle, and (d) lipid calcium phosphate (LCP) nanoparticle. Images were created using DesignSpark Mechanical 2.0 software.
Figure 2. Lipid-based delivery agents for anti-c-myc siRNA (a) pegylated, neutral liposome, (b) cationic liposome, (c) liposome-polycation-DNA (LPD) nanoparticle, and (d) lipid calcium phosphate (LCP) nanoparticle. Images were created using DesignSpark Mechanical 2.0 software.
Biomedicines 08 00612 g002
Table 1. Summary of anti-c-myc RNAi-based nanosystems developed to date. Symbols = present/yes; = absent/no.
Table 1. Summary of anti-c-myc RNAi-based nanosystems developed to date. Symbols = present/yes; = absent/no.
Nucleic AcidCarrierPolymer Modification (✓/✗)Ligand-Targeting Modification (✓/✗)Disease StateTest SystemAdvantagesDisadvantagesClinical Trial (✓/✗)Reference
siRNAneutral liposomeOvarian cancerXenograft tumorsHigh encapsulation efficiency
Good biocompatibility
Low toxicity
Weak immunogenicity
Low transfection efficiency
High production cost
[50,52]
cationic liposomeColon cancer
Breast
cancer
HT-29 cells
MCF-7 cells
High encapsulation efficiency
Good biocompatibility
Low toxicity
Weak immunogenicity
Low transfection efficiency
High production cost
[55,56]
liposome-polycation-DNA nanoparticleFibrosarcomaHT-1080 cells
Xenograft tumors
High transfection efficiency
Good biocompatibility
Complex structure and synthesis[57]
lipid calcium phosphate nanoparticleNon-small cell lung cancerXenograft tumorsHigh transfection efficiency
Good biocompatibility
Low toxicity
Complex structure and synthesis[58]
exosomes-Mouse λ820 cellsHigh encapsulation efficiency
Natural carriers
Good biocompatibility
Steady release profile
Lack of standardized techniques for isolation and purification[66]
chitosan nanoparticlesBreast cancerMCF-7 cells
Xenograft tumors
Small particle size
Good biocompatibility
Low transfection efficiency[67]
poly(lactic-co-glycolic acid) nanocapsuleNeuroblastoma-High stability
Biodegradability
FDA-approved material
Sustained release
Low transfection efficiency[68]
siNozymemelanoma-Biocompatibility
Good bioavailability
Complex structure and synthesis[69]
cell-penetrating peptidemelanomaB16 cells
Xenograft tumors
High transfection efficiency
Low toxicity
The possible need for covalent conjugation
Low cell specificity
[70]
multi-peptide complexBreast cancerMDA-MB-231High transfection efficiency
Low toxicity
Complex structure and synthesis[71]
gold nanoparticlesCervical cancer
Breast cancer
Lung cancer
HeLa cells
MCF-7 cells
Xenograft tumors
A549 cells
CMT/167 cells
Large surface area-high loading capacity
Amenable to chemical manipulation
Toxicity[81,82,83,84,85,86,87]
selenium nanoparticlesGlioblastomaU251 tumor spheroidsLarge surface area-high loading capacity
Amenable to chemical manipulation
Toxicity[88]
nano-graphene oxideBreast cancerMCF-7 cells
MDA-MB-231 cells
High surface area to volume ratio
Flexibility for cargo loading
Amenable to functionalization
Adverse interactions with proteins
Toxicity
Immunogenicity
[89]
DsiRNAEnCore™ lipid nanoparticleAdvanced solid tumors
Multiple myeloma
Lymphoma
-PatientsHigh carrying capacity
Good biocompatibility
Poor tumor penetration
Unsatisfactory knockdown efficiency
Expensive
Labor intensive
[91]
shRNA expression plasmidcationic liposomeBreast cancer
Liver cancer
Colon cancer
Nasopharyngeal cancer
MCF-7 cells
Xenograft tumors
HepG2 cells
Colo320 cells
CNE-2Z
High encapsulation efficiency
Good biocompatibility
Low toxicity
Weak immunogenicity
Low transfection efficiency
High production cost
[93,94,95,96]
polyglycidal methacrylate nanoparticleBreast cancer
Colorectal cancer
-Low toxicity at high concentrationsLow transfection efficiency
Complex structure and synthesis
[99]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Habib, S.; Ariatti, M.; Singh, M. Anti-c-myc RNAi-Based Onconanotherapeutics. Biomedicines 2020, 8, 612. https://doi.org/10.3390/biomedicines8120612

AMA Style

Habib S, Ariatti M, Singh M. Anti-c-myc RNAi-Based Onconanotherapeutics. Biomedicines. 2020; 8(12):612. https://doi.org/10.3390/biomedicines8120612

Chicago/Turabian Style

Habib, Saffiya, Mario Ariatti, and Moganavelli Singh. 2020. "Anti-c-myc RNAi-Based Onconanotherapeutics" Biomedicines 8, no. 12: 612. https://doi.org/10.3390/biomedicines8120612

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop