IMR Press / FBS / Volume 11 / Issue 1 / DOI: 10.2741/S526
Review
Delivery of siRNA therapeutics: PLGA nanoparticles approach
Show Less
1 Department of Sciences, Faculty of Natural and Applied Sciences, Notre Dame University - Louaize, Zouk Mosbeh, Lebanon Zouk Mikael
Send correspondence to: Rita Maalouf, Department of Sciences, Faculty of Natural and Applied Sciences, Notre Dame University - Louaize, Zouk Mosbeh, Lebanon P.O.Box: 72, Zouk Mikael, Tel: 961-9 208745, Fax: 961-9 218771, E-mail: rita.maalouf@ndu.edu.lb
Front. Biosci. (Schol Ed) 2019, 11(1), 56–74; https://doi.org/10.2741/S526
Published: 1 March 2019
Abstract

RNA interference is currently one of the most advanced genomic tools providing promising insight to manage countless pathophysiological conditions that seemed unapproachable few years ago. Researchers across the globe have devised numerous methods in which small RNA molecules can be administered into target cells to manipulate cellular functions at the molecular level and eradicate defects that lead to pronounced phenotypes, which might sometimes be as fatal as malignant tumors. The present review provides an overview of the non-viral delivery approaches for siRNA therapeutics with an emphasis on the PLGA polymeric strategy and its use in different biological applications. Due to its versatile surface, PLGA is not only used in tissue engineering such as remodeling of bone and cartilage, but it indeed plays a crucial role as chemical carrier for a broad range of therapeutic agents. It is greatly implicated in gene therapies and inherited genetic defects.

Keywords
RNA interference
siRNA
gene therapy
Non-Viral Delivery
PLGA nanoparticles
Review
2. THE HISTORY OF RNA INTERFERENCE
2.1. Introduction

RNA interference (RNAi) is a conserved mechanism from yeast to humans that uses small regulatory non-coding RNAs to control the expression of genes. Over more than 1000 small RNAs are encoded in the human genome regulating approximately 60% of our genes (1, 2). The first hint to a role for RNA species in gene silencing came from the work of Napoli and Jorgensen who described in 1990 a revolutionary experiment done on Petunia flowers, which laid the foundations for the later discovery of RNAi as a mechanism of gene silencing. Initially, the aim of their study was to overexpress chalcone synthase (CHS) to test whether or not this enzyme is rate-limiting in the biosynthesis of anthocyanin, a purple plant pigment. Unexpectedly, a portion of the transgenic flowers ended up with a white instead of a violet color. Napoli and Jorgensen hypothesized that the dual transcription of the endogenous and introduced CHS genes coordinately suppressed their expression, thus resulting in the white phenotype (3). This phenomenon, known as post-transcriptional gene silencing (PTGS), was reported shortly afterwards in Caenorhabditis elegans. Two simultaneous reports at that time revealed that the gene lin-4 encodes small regulatory RNA which silence post-transcriptionally the lin-14 gene involved in the temporal regulation of postembryonic developmental events (4, 5). These initial studies left a dilemma on the nature of this RNA-RNA interaction phenomenon and the structure of the RNA species involved. It was not until 1998 that Craig C. Mello and Andrew Z. Fire, who shared the 2006 Nobel Prize in Medicine for their discovery, were able to solve this scientific dilemma presented by the earlier studies. They showed that the double-stranded RNA (dsRNA) manifests a potent gene silencing effect that is more pronounced than that of purified single-stranded RNAs (ssRNA) of either sense, by noting a synergistic effect of sense and antisense RNA mixture (6). Later, studies in plants and Drosophila identified the stable RNA intermediate generated from dsRNA that was involved in silencing the target messenger RNA (mRNA). It appeared to be an interfering antisense RNA of small size. Since its discovery, RNAi proved to be a potent tool for the large scale systematic analysis of gene function in many organisms including Arabidopsis (7), C. elegans (8), Drosophila (9, 10), mosquitoes (11, 12), Zebrafish (13), and human cells (14) as well as several others.

RNA interference has been observed in several living organisms, from plants to vertebrates. It has been the most extensively used reverse genetics tool to study gene function since the last decade. Its success is attributed to the evolutionary conserved pathways that regulate gene expression via small non-coding RNAs (15), the relatively simple way to generate siRNAs or dsRNA in vitro and their ease of use to silence genes in several organisms. Despite its success as a gene silencing tool, RNAi has also its limitations including; incomplete phenotypes due to the lack of complete silencing, off-target effects with the 3´ untranslated region of genes which may be even more problematic if the design of small RNAs is based on genomic sequences of low quality (16, 17). Additionally, some non-specific RNAi effects like activation of the interferon system by siRNA have been reported in the literature (18). Despite these drawbacks, RNAi remains the most powerful functional genomic tool recently developed.

2.2. Origins of small non-coding RNAs and mechanism of gene silencing

There are three species of small non-coding RNAs involved in gene regulation which have disparate origins but their pathways converge downstream when they assemble into the RNA-induced silencing complex (RISC). These are the microRNA (miRNA), the short interfering RNA (siRNA) and PIWI-interacting RNA (piRNA). The miRNAs originate from the genome as long primary transcripts (pri-miRNA) with single or clustered double-stranded hairpins (19) that are processed by a protein complex containing the RNase III family enzyme Drosha into short ~ 65-70 nucleotide pre-miRNAs (20-22) that are transported to the cytoplasm for further processing. On the other hand, siRNAs may originate from exogenously introduced dsRNA molecules, from viral genomes or from endogenous genomic loci such as centromeres, transposons or other repetitive sequences (23). The piRNA pathway however silences transposable elements in the gonads of metazoans through PTGS and transcriptional gene silencing (TGS). However the biogenesis and processing of piRNAs remain not fully understood but shows clear differences with the miRNA and siRNA pathways notably in being independent of Dicer (24, 25).

In the cytoplasm, pri-mRNA and long dsRNA, whether endogenous or exogenous, are diced by the endoribonuclease Dicer that contains two RNase III domains each involved in cleaving one strand of the duplex RNA (26). The resulting dsRNAs are 21-25 nucleotides in length containing 2 nucleotides overhangs at each 3´ terminal end, and a phosphate group at each 5´ end (27). Small dsRNAs are then loaded into the RNA-induced silencing complex (RISC) constituted of a tri-molecular assembly of dicer, TAR-RNA binding protein (TRBP), and Argonaute 2 (Ago2). In Drosophila, there are two Ago proteins with clearly distinct functions. Typically, miRNAs are loaded on Ago1 that lacks slicing activity and functions by repressing mRNA translation. While siRNAs are loaded on Ago2 which harbors slicing activity that results in target mRNA cleavage. In humans, however all four Ago proteins seem to participate in miRNA-mediated silencing (24). RISC assembly requires two steps: loading and unwinding (28, 29). Loading to Ago2 necessitates help from Dicer and TRBP in addition to ATP. Strand selection is not random; the strand harboring the thermodynamically less stable base pairing at its 5´ end will constitute the guide strand. In Drosophila, this thermodynamic instability is initially detected by dicer and the dsRNA-binding protein R2D2 (30). In the unwinding step Ago2 cleaves the passenger strand in a similar manner it does a target RNA. The cleaved strand is released from Ago2 to liberate the guide strand (31, 32). In case of miRNA, the strand that is loaded to the RISC is known as the miRNA and the opposing strand is the miRNA*. Finally, silencing of the target RNA may occur by endonucleolytic cleavage or by translation repression depending on the type of Ago present in the RISC and the extent of base-pairing between the guide strand (or miRNA) and the target RNA (33). Figure 1 shows a schematic representation of the siRNA, miRNA and piRNA silencing pathways.

Figure 1

Schematic representation of siRNA, miRNA and piRNA silencing pathways.

3. GENE THERAPY
3.1. Background information

Gene therapy uses a set of approaches to deliver genetic information to a target cell in order to prevent or cure the incidence or progression of a disease. It has recently gained enormous attention as an option for therapeutic treatment of genetic or acquired diseases. siRNA-based gene therapy showed promising potential applications in many research studies. However, the development of naked siRNA-based treatment is hindered by the difficulty of its delivery into the desired cells, tissues or organs. Due to their negative charge and molecular size, siRNAs are unable to cross the lipophilic cellular membranes. In addition, they are subject to enzymatic degradation and are rapidly eliminated by the renal system. To extend their circulation time, facilitate their transfection efficiency, and enable their selective and efficient delivery, siRNAs can be carried through viral and non-viral carriers. These carriers have been extensively researched in many ex vivo, in vivo and in vitro applications. Both types of carriers have their advantages and disadvantages and hence, the use of one instead of the other depends on the experimental conditions and applications (34, 35). In this review, we will focus on non-viral carriers. For the targeted delivery of siRNAs using viral vectors the readers are referred to the following excellent reviews (34, 36-38).

3.2. Non-viral gene therapy

Non-viral gene delivery was developed to avoid the mentioned serious limitations related to viral gene carriers. It involves the use of physical or chemical techniques to transfer nucleic acids into cells (34, 39, 40). Although non-viral vectors have low transfection efficiency, they are much safer than viral vectors. Physical methods, such as electroporation (41, 42), microinjection (43), sonoporation (44), gene gun (45), magnetofection (46), and electric field-induced molecular vibrations (47) rely on the application of physical forces to facilitate the entrance of the desired nucleic acids into the target cell across the phospholipid bilayer. Chemical methods, on the other hand, include direct modification of the siRNA backbone, its conjugation with various molecules such as lipids, polymers, peptides, and aptamers, and siRNA encapsulation in different nanoparticles (NPs) formulations (48). All these modifications decrease the likelihood of triggering an unwanted innate immune response upon the introduction of the siRNA, lower the incidence of undesirable off-target effects, and hence improve the treatment outcomes.

3.2.1. Direct chemical modifications

Direct chemical and structural modifications conducted on the siRNAs terminals, backbone, nucleobases and sugars were found to be effective at improving their pharmacodynamics and pharmacokinetics properties. Given that the 2’ position of pentose sugars is not required for siRNA recognition as well as for RISC activity, incorporation of 2’-fluoro, 2’-O-methyl, 2’-halogen, 2’-amine or 2’-deoxy at this site showed a remarkable enhanced stability compared to wild-type siRNA (49). Moreover, locked nucleic acids (LNA) and unlocked nucleic acids (UNA) proved to be beneficial for siRNA delivery. LNA are rigid structures characterized by the presence of a methylene bridge between the 2’ oxygen and the 4’ carbon however UNA are highly flexible structures obtained following the cleavage of the C2’-C3’ bond of the sugar ring. Although they have differing physical and chemical properties, they were able to protect the siRNA duplex from degradation, increase its stability and reduce the off-target effects (49, 50). Substitution of the phosphodiester bonds by methylphosphonate, phosphorothioate, phosphorodiothioate, and triazole linkages was also investigated in many research studies and was able to improve siRNAs target affinity and plasma half-life (51, 52).

Furthermore, tagging siRNAs with moieties such as cholesterol, peptides, polymers and aptamers lead to beneficial impact on transfection efficiency and allow targeting specific cells and organs. For example, complexation of epidermal growth factor-type 2 siRNA with polyethylenimine (PEI) increased siRNA stability and reduced subcutaneous tumor growth in a mice model (53). Likewise, Soutschek and coworkers developed chemically modified siRNAs towards apolipoprotein B (apoB) for the treatment of hypercholesterolemia. Their intravenous injection in mice exhibited a silencing effect in liver and jejunum denoted by decreased plasma levels of apoB and total cholesterol (54). Cell penetrating peptides (CPP) are a class of short peptides able to translocate across cell membranes. They are used to deliver DNA, RNA, protein molecules, nanomaterials, or pharmaceutical drugs into cells primarily via endocytosis. The bond between cell penetrating peptides and the cargo might be covalent or non-covalent (55). CPP-mediated siRNA delivery has been used to treat diseases such as cancer, infectious diseases, and some genetic disorders and was proved to have high transfection efficiency and low cytotoxicity (56).

Although chemical modifications and siRNA bioconjugation can somehow avoid some problems related to siRNA delivery, nanoparticles that encapsulate siRNA are better at protecting it from nucleases and immune recognition (57). Thus, delivery materials are needed, among them lipid-based and polymer-based vectors hold promising potentials for siRNA transport.

3.2.2. Lipid-based vectors

Liposomes, globular vesicles with an aqueous core surrounded by a phospholipid layer, are by far the most widely used lipid-based vectors. They are biodegradable, biocompatible, and suitable for the delivery of hydrophilic as well as hydrophobic drugs (56). They can be cationic, anionic, or neutral. However, due to the polyanionic nature of RNAi, cationic and neutral liposomes are usually preferred as carriers. The promising in vitro results obtained by Halder et al. (2006) with focal adhesion kinase (FAK) silencing using neutral liposomes pointed out that in vivo delivery of FAK siRNA might be a potential novel therapeutic approach to ovarian cancer (58). Cationic liposomes, on the other hand, are slightly different. They comprise three basic components: a lipophilic tail group, a connecting linker, and a cationic hydrophilic head with one or more amine groups. The positively charged head interacts with the negative phosphate backbone of nucleic acids allowing their condensation (59-61). The structure and the physicochemical properties of each liposome component, as well as the nature of the head group can affect the transfection efficiency (62, 63). Indeed, it has been shown that variables such as the size of lipoplexes and the lipid-to-DNA ratio alter significantly the level of gene expression, in experimental set-ups as well as in the lung, heart, spleen, liver and kidneys (64, 65).

3.2.3. Polymer-based vectors

Polymer-based carriers, termed polymeric nanoparticles, have been extensively used as drug vesicles. Many are biodegradable, biocompatible and non-toxic which make them good candidates for biomedical applications. They are characterized by their high-drug loading capacity, and their ability to incorporate a broad range of therapeutic agents. The polymeric shell provides protection against enzymatic degradation and rapid renal filtration. The type and molecular weight of the polymer used as well as the drug-to-polymer ratio can affect the drug loading efficiency and release profile. They also exhibit a great potential for surface functionalization that allows improving pharmacokinetic and pharmacodynamics properties and permits targeting drug delivery and accumulation of nanoparticles at the desired site of action (66-68). Cyclodextrin, poly-L-lysine (PLL), PEI, chitosan, dendrimers and poly (D,L-lactide-co-glycolide) (PLGA) have received a lot of attention as delivery carriers.

Dendrimers are spherical, highly symmetrical, and usually monodisperse macromolecules with hyperbranched arrays. They have a central core surrounded by concentric layers called generations. Among the many different available dendrimers, polyamidoamine (PAMAM) has gained a particular interest due to its versatile nature that allows the biological and physiochemical modifications of its end groups. It has unique features such as a nanosized, easily modifiable, cationic surface. The primary amine groups on the surfaces of the PAMAM molecules facilitate their electrostatic interaction with nucleic acids, thus forming nanoscale complexes known as “dendriplexes” (69). A recent study screened a library of surface-engineered dendrimers for structure-activity relationships regarding siRNA delivery. It was demonstrated that dendrimers modified with hydrophobic ligands such as aliphatic chains and aromatic rings, as well as functional ligands such as fluorine, bromine, boronic acid, and guanidine groups improve the efficacy of the knockdown with minimal cytotoxicity (70).

Furthermore, cationic polymers such as PLL, PEI, and chitosan have attracted the attention of many scientists due to their ability to form complexes with nucleic acids by means of stable electrostatic interactions (71, 72). Their properties such as size, surface charge, and structure are dependent on the ratio of the positive charges on cationic polymers to the number of negatively charged phosphate groups in nucleic acids. Polyethyleneimine has been examined as a potential non-viral delivery vehicle for oligonucleotides, siRNA and plasmid DNA in vitro and in vivo (73). PEI exists in different molecular weights and with degrees of branching (74). Its amino groups confer two important properties: a superior buffering capacity over a wide range of pHs and an ability to condense nucleic acids to form positively charged complexes. The toxicity and the transfection efficiency using this compound depend on the properties of the molecule used. When PEI is more branched and has a higher molecular weight, it condenses siRNA more efficiently thus increasing the delivery efficiency but at the expense of being more toxic (75). Hence, the toxicity associated with PEI has been regulated by using low molecular weight molecules, by incorporating them into other polymeric constructs, or by modifying them to either shield the charge or transform the backbone into its biodegradable analogue (71). The complexation of PEI and siRNA demonstrated a reduction in target gene in different cellular models. Kim et al. demonstrated the delivery of siRNA against vascular endothelial growth factor (VEGF) through polyelectrolyte complex micelles formed by the conjugation of PEGylated siRNAs (siRNA-PEG) with PEI. These micelles showed enhanced stability compared to naked VEGF siRNA. Furthermore, they were able to silence VEGF gene expression in prostate carcinoma cells up to 96.5%. They also demonstrated a greater VEGF gene silencing effect than VEGF siRNA/PEI complexes (76).

Among all, PLGA has shown immense potential as a good delivery material for a variety of drugs including anticancer and antihypertensive agents, hormones, vaccines, as well as for macromolecules such as nucleic acids, proteins, peptides, and antibodies. PLGA, is a copolymer of poly(D,L-lactic acid) and poly(glycolic acid). It is biocompatible, biodegradable, and non-toxic. Most significantly, it has been approved for human use by the US Food and Drug administration (77). In the next section, we will mainly focus on the in vitro and in vivo biological applications of PLGA vectors for gene silencing. Some of the selected examples listed below are summarized in table 1.

4. PLGA NANOPARTICLES’ APPROACH
4.1. General overview

Different mechanisms are used to synthesize PLGA. Ajioka et al. prepared high molecular weight PLGA by azeotropic dehydration, using a solution of diphenylether with L-lactic acid and glycolic acid in the presence of tin powder as a catalyst at 130°C. It typically requires 20-40 hours to obtain PLGA using this protocol. However, this reaction is hard to control and the desired product has to be purified from many other by-products (78). To prepare PLGA in a shorter time, the cationic ring-opening polymerization of cyclic dimers, glycolide and lactide, is performed. Since lactide exists in two enantiomeric isomers, the synthesized PLGA polymer can be under the form of D-, L-, or D, L- stereoisomer. This reaction requires 2-6 hours and is commonly done in the presence of stannous octoate as catalyst at 175°C. During its synthesis, fine-tuning of relevant parameters such as molecular weight, lactide to glycolide ratio, and lactide stereoisomeric composition (L- or D, L-lactide) is one of the key components for acquiring the appropriate and specific properties needed for various applications. Therefore, PLGA solubility, crystallinity, thermal stability, mechanical properties, and degradation can be controlled widening its performance spectrum (79, 80). Due to these characteristics, PLGA have found a broad range of applications in tissue engineering including blood vessel replacement, bone, cartilage and ligament restoration as well as skin healing (81-88). Furthermore, PLGA nanoparticles have been demonstrated as a promising drug delivery platform owing to the aforementioned properties, along with their ability to sustain optimal therapeutic doses for a prolonged time. They have been used for diagnosis, vaccine delivery, and for the treatment of many pathophysiological diseases carrying a variety of drugs, as well as, antibiotics, proteins, and nucleic acids (89-91). PLGA nanoparticles’ cells uptake involves an endocytic pathway. However, the major drawback is their low intracellular retention due the fast exocytosis. Surface functionalization and complexation with ligands can reduce this occurrence, enhance cellular uptake and permit targeted drug-delivery (92). For instance, nanoparticles conjugated with transferrin ligands showed increased cellular uptake and accumulation in cell populations filled with transferrin receptors. It was demonstrated that only 50% of transferrin-conjugated nanoparticles exhibit exocytosis vs. 75% of the unconjugated nanoparticles. This is mainly related to different endocytic pathways involved in their internalization (93).

4.2. Biological applications
4.2.1. Cardiovascular diseases

A recent report from the American Heart Association estimated that 92.1 million American adults are living with some form of cardiovascular disease. Coronary heart disease is the leading cause of deaths in the US (43.8%), followed by stroke (16.8%), high blood pressure (9.4%) and heart failure (9%) (94). PLGA have found many applications in cardiology, in particular as scaffolds and coating matrices for drug-eluting stents, which have been proven to be highly effective in reducing stent thrombosis and restenosis (95). Qiu et al. prepared a novel external stent by deposition of hybrid fibrous membranes consisting of tissue factor siRNA loaded in PLGA/chitosan nanoparticles. These siRNA eluting stents were able to attenuate neointima hyperplasia in rat vein grafts (96). In another study, siRNAs against intracellular adhesion molecule-1 (ICAM-1) were incorporated into different PLGA resomers. These coatings proved to have no cytotoxic effect towards human umbilical vein cell line and a good hemocompatibility since no reduction in the number of platelets, leukocytes, lymphocytes, monocytes, and granulocytes was noticed after 1 h incubation. ICAM-1 siRNAs loaded in the PLGA Resomer® RG 752 H (75:25, acid terminated, 4,000–15,000 Da) exhibited optimal results with 95% transfection efficiency and 36% knockdown effect. These findings prove the potential incorporation of siICAM-1 in PLGA coatings for the elaboration of novel drug eluting stents (97). Cohen-Sacks and coworkers showed that platelet-derived growth factor beta-receptor antisense loaded PLGA nanoparticles could serve as effective gene delivery system for the treatment of restenosis. The synthesized nanoparticles displayed a homogenous size, high encapsulation efficiency and a sustained release (98). Dual targeting approach has been greatly reported for enhanced drug delivery and synergistic effect. In a recent study, Zhao et al. prepared dual-targeting bifunctional core-shell nanoparticles to deliver atorvastatin and siRNA against lectin-like oxidized low-density lipoprotein receptor-1 to endothelial cells and macrophages in the atherosclerotic lesions. The drugs were encapsulated within the first PLGA core shell followed by three consecutives layers made up from lipid for cholesterol receiving, apoliprotein A-I for macrophage targeting and hyaluronic acid for endothelial cell targeting. These nanoparticles showed enhanced delivery and improved anti-atherosclerotic efficacy (99).

4.2.2. Cancer treatment

According to the national institute of cancer, cancer is a set of malignant diseases characterized by uncontrolled proliferation of mutated cells in a body tissue that has been exposed to mutagens. In 2012, there were 14.1 million new cases and this number is expected to rise to 23.6 million per year by 2030 (100). Several treatment options such as chemotherapy, radiotherapy, surgery and gene therapy have been used so far but each has its implications on patients. Targeting efficiently cancerous cells, while sparing normal cells from the adverse effects of the delivered chemicals and genes, remain a challenging goal. Numerous targeted nanoparticle approaches are currently being investigated to deliver the appropriate drugs or genes into the malignant cells by mean of ligand-based targeting mechanisms. For that purpose, nanoparticles must undergo some modifications to allow their efficient delivery into the abnormal cells. This can be done using hydrophilic surfaces, coatings with a positive charge carrier and/or with proper markers to improve cellular incorporation, increase their circulation duration, and permit the differentiation between normal and cancerous cells (101). PLGA nanoparticles have attracted extensive interest for applications in cancer therapy. Woodrow et al. reported the efficient and sustained gene silencing of siRNA-spermidine loaded PLGA nanoparticles for the treatment of vaginal mucosa (102). Another study reported the use of siRNA-chitosan complex, encapsulated into a multilayer shell of PLGA and poly (vinyl alcohol) (PVA) for silencing Aquaporin-1 in cancer cells. These nanoparticles demonstrated high stability under physiological conditions. They reduced the expression of Aquaporin-1 by 70%, and showed no cytotoxicity up to 72 hours of incubation (103). Kundu and coworkers reported the use of lipid based NPs with either PLGA or PLGA-PEG to encapsulate p53 siRNA against osteocarcinoma. Incorporation of 10% PLGA showed the highest p53 knockdown efficiency (104).

Despite all the efforts, drug resistance remains a major challenge in the treatment of cancer. It can occur by different mechanisms, including decreased drug uptake, increased drug efflux, activation of DNA repair mechanisms, suppression of drug-induced apoptosis, etc (105). It has been shown that overexpression of FAK plays a major key role in ovarian cancer and promotes chemoresistance (106). Byeon et al. suggested the development of hyaluronic acid-PLGA nanoparticles encapsulating both paclitaxel and FAK–siRNA to reverse chemoresistance. This combination therapy proved to be effective in inhibiting tumor growth in patient-derived xenograft model compared with paclitaxel alone (107). However, other researchers showed optimal suppression of chemoresistance when siRNAs loaded PLGA nanoparticles are administrated 1 day earlier than drugs. Therefore, they designed PLGA nanoparticles to co-deliver MDR1 and BCL2 siRNA aimed for the simultaneous inhibition of drug reflux and cell death defense pathways and to avoid resistance towards paclitaxel and cisplastin in recurrent or advanced ovarian cancer. To ensure high encapsulation efficiency, siRNAs were complexed with the cationic poly-L-lysine (108). Furthermore, Wu et al. reported the elaboration of programmable PLGA biomaterials for dynamic and consecutive release of siRNA followed by the drug. In this work, o-nitrobenzyl ester derivative caged doxorubicin (DOX) was encapsulated in polymeric NPs constructed by the self-assembly of C16-S-S-PEI and PLGA. siRNA of P-glycoprotein (P-gp) were adsorbed onto its cationic polymer shell and their release were triggered by the reductive cleavage of the disulfide bond in the presence of high levels of glutathione in cancer cells. DOX is afterwards released upon irradiation and its conversion DOX prodrug into hydrophilic active DOX. This sequential release strategy showed enhanced efficacy of DOX following P-gp silencing and can be used against multiple drug resistance (109).

4.2.3. Inflammatory diseases

Inflammation occurs when tissues are injured by bacteria, trauma, toxins, heat, or any other cause. It is a defense mechanism in the body and an essential component of many illnesses including rheumatoid arthritis, sepsis, lung and gastrointestinal diseases, etc. Tumor necrosis factor-α (TNF-α) might be considered as potential therapeutic target due to its high levels in many acute and chronic inflammations. It was shown that PLGA nanoparticles loaded with TNF-α siRNA have a significant effect in decreasing the paw scores and joints effusion in a murine arthritis model (110). Furthermore, kelly et al. used anti-TNF-α siRNA encapsulated in PLGA microparticles for the treatment of inflammatory lung disease. This work revealed a sustained siRNA knockdown of TNF-α in primary monocytes for up to 72 hours (111). In another study, siRNA against TNF-α, keratinocide-derived cytokine or interferon gamma-induced protein were adsorbed to the surface of a calcium phosphate core, followed by their encapsulation into PLGA nanoparticles coated with an outer layer of PEI to enhance cellular uptake and subsequent endosomal escape. Intrarectal application of these nanoparticles demonstrated a significant decrease of the target gene with a subsequent reduction of intestinal inflammation in mice (112). Aldayel and coworkers reported the elaboration of TNF-α siRNA loaded PLGA NPs by the double-emulsion method using a special emulsifying agent which renders the nanoparticles PEGylated and the PEGylation readily sheddable in low pH environment such as that in chronic inflammation sites. This technique showed a higher accumulation of nanoparticles in inflamed mouse foot compared to nanoparticles prepared by an acid-insensitive emulsifying agent (113).

4.2.4. Other miscellaneous applications

siRNA loaded PLGA nanoparticles have attracted a lot of interest in many other biomedical applications. They have been used to enhance drug delivery to human brain endothelial cells for future medical therapeutic prospects aiming to surpass the blood-brain-barrier and overcome central nervous system disorders. PLGA nanoparticles were loaded with siRNA against P-gp, a drug efflux transporter, and were functionalized with a peptide-binding transferrin receptor. They demonstrated no cytotoxicity and a 4-fold increased efficiency compared to non-functionalized NPs (114). In another study, Chen et al. reported the delivery of transferrin-siRNA loaded PLGA NPs to injured brain microvascular endothelial cells. These nanoparticles showed very low cytotoxic effect and good transfection efficiency mainly due to their functionalization with enhanced green fluorescent protein – epidermal growth factor -1 that targets the overexpressed tissue factor in the injured cells (115). Moreover, mTOR siRNA was loaded into hybrid NPs made from PEI and PLGA. They inhibited proliferation and activated apoptosis of hypoxic pulmonary arterial smooth muscle cells (116).

5. CONCLUSION

Due to the rapid progress in the applications of RNAi, it is considered as one of the most powerful tools for gene therapy. First described in 1990, it now opens endless possibilities and provides a promising insight to manage countless pathophysiological conditions that seemed inaccessible few years ago. Scientists across the globe have devised numerous methods in which small RNA molecules can be administered into target cells to manipulate cellular functions at the molecular level and eradicate defects that lead to pronounced phenotypes, which might sometimes be as fatal as malignant tumors. Targeted delivery of these RNA molecules, the action of which relies on the RNAi and subsequent gene silencing, can be done using viral or non-viral vectors. PLGA is one of the chemicals that have been approved by the FDA for use in humans. This huge medical leap is extended beyond its initial aim. Hence, it is no longer only used in tissue engineering such as remodeling of bone and cartilage, but it indeed plays a crucial role now as chemical carrier for a broad range of therapeutic agents. It is greatly implicated in gene therapies and inherited genetic defects. This field, as interesting and promising as it seems, is still in its neonatal stages. Investigations are being done in collaboration with physiologists, biomedical engineers, medical doctors, biochemists, and specialists from all scientific disciplines to adopt this modern approach and improve it, invest it optimally to yield the best results, and subsequently achieve what sounded once far-fetched.

6. ACKNOWLEDGEMENT

This work is supported by the National Council for Scientific Research (CNRS) of Lebanon.

Abbreviations: apoB: apolipoprotein B, Ago2: Argonaute 2, CHS: chalcone synthase, CPP: cell-penetrating peptides, dsRNA: double-stranded RNA, DOX: doxorubicin, FAK: focal adhesion kinase, ICAM-1: intracellular adhesion molecule 1, LNA: locked nucleic acids, mRNA: messenger RNA, miRNA: micro RNA, NPs: nanoparticles, piRNA: PIWI-interacting RNA, pre-miRNA: precursor-microRNA, pri-miRNA: primary-microRNA, PTGS: post-transcriptional gene silencing, RISC: RNA-induced silencing, RNAi: RNA interference, siRNA: small interfering RNA, ssRNA: single-stranded RNA, TRBP: TAR-RNA binding protein, PAMAM: polyamidoamine, PEG: polyethylene glycol, PEI: complex polyethyleneimine, P-gp: P-glycoprotein, PLGA: poly(lactide-co-glycolide), PLL: poly-L-lysine, PVA: poly (vinyl alcohol), TGS: transcriptional gene silencing, UNA: unlocked nucleic acids, TNF-α: Tumor necrosis factor-α, VEGF: vascular endothelial growth factor.

References
[1]
R. C Friedman K. K Farh C. B Burge D. P BarTel Most mammalian mRNAs are conserved targets of microRNAs Genome Res 2009 19 1 92 105 DOI:10.1101/gr.082701.108
[2]
A Kozomara S Griffiths-Jones miRBase: annotating high confidence microRNAs using deep sequencing data Nucleic Acids Res 2014 42 Database issue D68 73 DOI:10.1093/nar/gkt1181
[3]
C Napoli C Lemieux R Jorgensen Introduction of a Chimeric Chalcone Synthase Gene into Petunia Results in Reversible Co-Suppression of Homologous Genes in trans Plant Cell 1990 2 4 279 289 DOI:10.1105/tpc.2.4.2.79
[4]
R. C Lee R. L Feinbaum V Ambros The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14 Cell 1993 75 5 843 54 DOI:10.1016/0092-8674(93)90529-Y
[5]
B Wightman I Ha G Ruvkun Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C. elegans Cell 1993 75 5 855 62 DOI:10.1016/0092-8674(93)90530-4
[6]
A Fire S Xu M. K Montgomery S. A Kostas S. E Driver C. C Mello Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans Nature 1998 391 6669 806 11 DOI:10.1038/35888
[7]
P Hilson J Allemeersch T Altmann S Aubourg A Avon J Beynon R. P Bhalerao F Bitton M Caboche B Cannoot V Chardakov C Cognet-Holliger V Colot M Crowe C Darimont S Durinck H Eickhoff A. F de Longevialle E. E Farmer M Grant M. T Kuiper H Lehrach C Leon A Leyva J Lundeberg C Lurin Y Moreau W Nietfeld J Paz-Ares P Reymond P Rouze G Sandberg M. D Segura C Serizet A Tabrett L Taconnat V Thareau P Van Hummelen S Vercruysse M Vuylsteke M Weingartner P. J Weisbeek V Wirta F. R Wittink M Zabeau I Small Versatile gene-specific sequence tags for Arabidopsis functional genomics: transcript profiling and reverse genetics applications Genome Res 2004 14 10B 2176 89 DOI:10.1101/gr.2544504
[8]
A. G Fraser R. S Kamath P Zipperlen M Martinez-Campos M Sohrmann J Ahringer Functional genomic analysis of C. elegans chromosome I by systematic RNA interference Nature 2000 408 6810 325 30 DOI:10.1038/35042517
[9]
M Boutros A. A Kiger S Armknecht K Kerr M Hild B Koch S. A Haas R Paro N Perrimon C Heidelberg Fly Array Genome-wide RNAi analysis of growth and viability in Drosophila cells Science 2004 303 5659 832 5 DOI:10.1126/science.1091266
[10]
L Lum S Yao B Mozer A Rovescalli D Von Kessler M Nirenberg P. A Beachy Identification of Hedgehog pathway components by RNAi in Drosophila cultured cells Science 2003 299 5615 2039 45 DOI:10.1126/science.1081403
[11]
S Blandin L. F Moita T Kocher M Wilm F. C Kafatos E. A Levashina Reverse genetics in the mosquito Anopheles gambiae: targeted disruption of the Defensin gene EMBO Rep 2002 3 9 852 6 DOI:10.1093/embo-reports/kvf180
[12]
L. F Moita R Wang-Sattler K Michel T Zimmermann S Blandin E. A Levashina F. C Kafatos In vivo identification of novel regulators and conserved pathways of phagocytosis in A. gambiae Immunity 2005 23 1 65 73 DOI:10.1016/j.immuni.2005.05.006
[13]
A Dodd S. P Chambers D. R Love Short interfering RNA-mediated gene targeting in the zebrafish FEBS Lett 2004 561 1-3 89 93 DOI:10.1016/S0014-5793(04)00129-2
[14]
K Berns E. M Hijmans J Mullenders T. R Brummelkamp A Velds M Heimerikx R. M Kerkhoven M Madiredjo W Nijkamp B Weigelt R Agami W Ge G Cavet P. S Linsley R. L Beijersbergen R Bernards A large-scale RNAi screen in human cells identifies new components of the p53 pathway Nature 2004 428 6981 431 7 DOI:10.1038/nature02371
[15]
R. C Wilson J. A Doudna Molecular mechanisms of RNA interference Annu Rev Biophys 2013 42 217 39 DOI:10.1146/annurev-biophys-083012-130404
[16]
M Boettcher M. T McManus Choosing the Right Tool for the Job. RNAi, TALEN, or CRISPR Mol Cell 2015 58 4 575 85 DOI:10.1016/j.molcel.2015.04.028
[17]
S Sharma A Rao RNAi screening: tips and techniques Nat Immunol 2009 10 8 799 804 DOI:10.1038/ni0809-799
[18]
C. A Sledz M Holko M. J de Veer R. H Silverman B. R Williams Activation of the interferon system by short-interfering RNAs Nat Cell Biol 2003 5 9 834 9 DOI:10.1038/ncb1038
[19]
B. R Cullen Transcription and processing of human microRNA precursors Mol Cell 2004 16 6 861 5 DOI:10.1016/j.molcel.2004.12.002
[20]
R. I Gregory K. P Yan G Amuthan T Chendrimada B Doratotaj N Cooch R Shiekhattar The Microprocessor complex mediates the genesis of microRNAs Nature 2004 432 7014 235 40 DOI:10.1038/nature03120
[21]
J Han Y Lee K. H Yeom Y. K Kim H Jin V. N Kim The Drosha-DGCR8 complex in primary microRNA processing Genes Dev 2004 18 24 3016 27 DOI:10.1101/gad.1262504
[22]
Y Lee C Ahn J Han H Choi J Kim J Yim J Lee P Provost O Radmark S Kim V. N Kim The nuclear RNase III Drosha initiates microRNA processing Nature 2003 425 6956 415 9 DOI:10.1038/nature01957
[23]
R. W Carthew E. J Sontheimer Origins and Mechanisms of miRNAs and siRNAs Cell 2009 136 4 642 55 DOI:10.1016/j.cell.2009.01.035
[24]
J. J Ipsaro L Joshua-Tor From guide to target: molecular insights into eukaryotic RNA-interference machinery Nat Struct Mol Biol 2015 22 1 20 8 DOI:10.1038/nsmb.2931
[25]
A Le Thomas K. F Toth A. A Aravin To be or not to be a piRNA: genomic origin and processing of piRNAs Genome Biol 2014 15 1 204 DOI:10.1186/gb4154
[26]
H Zhang F. A Kolb L Jaskiewicz E Westhof W Filipowicz Single processing center models for human Dicer and bacterial RNase III Cell 2004 118 1 57 68 DOI:10.1016/j.cell.2004.06.017
[27]
D. S Schwarz G Hutvagner T Du Z Xu N Aronin P. D Zamore Asymmetry in the assembly of the RNAi enzyme complex Cell 2003 115 2 199 208 DOI:10.1016/S0092-8674(03)00759-1
[28]
T Kawamata Y Tomari Making RISC Trends Biochem Sci 2010 35 7 368 76 DOI:10.1016/j.tibs.2010.03.009
[29]
H Kobayashi Y Tomari RISC assembly Coordination between small RNAs and Argonaute proteins Biochim Biophys Acta 2016 1859 1 71 81 DOI:10.1016/j.bbagrm.2015.08.007
[30]
Y Tomari C Matranga B Haley N Martinez P. D Zamore A protein sensor for siRNA asymmetry Science 2004 306 5700 1377 80 DOI:10.1126/science.1102755
[31]
P. J Leuschner S. L Ameres S Kueng J Martinez Cleavage of the siRNA passenger strand during RISC assembly in human cells EMBO Rep 2006 7 3 314 20 DOI:10.1038/sj.embor.7400637
[32]
T. A Rand S Petersen F Du X Wang Argonaute2 cleaves the anti-guide strand of siRNA during RISC activation Cell 2005 123 4 621 9 DOI:10.1016/j.cell.2005.10.020
[33]
A van den Berg J Mols J Han RISC-target interaction: cleavage and translational suppression Biochim Biophys Acta 2008 1779 11 668 77 DOI:10.1016/j.bbagrm.2008.07.005
[34]
N Nayerossadat T Maedeh P. A Ali Viral and nonviral delivery systems for gene delivery Adv Biomed Res 2012 1 27 DOI:10.4103/2277-9175.98152
[35]
K Gavrilov W. M Saltzman Therapeutic siRNA: principles, challenges, and strategies Yale J Biol Med 2012 85 2 187 200
[36]
M. A Kay J. C Glorioso L Naldini Viral vectors for gene therapy: the art of turning infectious agents into vehicles of therapeutics Nat Med 2001 7 1 33 40 DOI:10.1038/83324
[37]
L. B Couto K. A High Viral vector-mediated RNA interference Curr Opin Pharmacol 2010 10 5 534 42 DOI:10.1016/j.coph.2010.06.007
[38]
J. W Schott M Morgan M Galla A Schambach Viral and Synthetic RNA Vector Technologies and Applications Mol Ther 2016 24 9 1513 27 DOI:10.1038/mt.2016.143
[39]
M. S Al-Dosari X Gao Nonviral gene delivery: principle, limitations, and recent progress AAPS J 2009 11 4 671 81 DOI:10.1208/s12248-009-9143-y
[40]
A. J Mellott M. L Forrest M. S Detamore Physical non-viral gene delivery methods for tissue engineering Ann Biomed Eng 2013 41 3 446 68 DOI:10.1007/s10439-012-0678-1
[41]
J. W Henshaw F Yuan Field distribution and DNA transport in solid tumors during electric field-mediated gene delivery J Pharm Sci 2008 97 2 691 711 DOI:10.1002/jps.21000
[42]
E Neumann M Schaefer-Ridder Y Wang P. H Hofschneider Gene transfer into mouse lyoma cells by electroporation in high electric fields EMBO J 1982 1 7 841 5 DOI:10.1002/j.1460-2075.1982atb01257.x
[43]
J. K Cinkornpumin R. L Hong RNAi mediated gene knockdown and transgenesis by microinjection in the necromenic Nematode Pristionchus pacificus J Vis Exp 2011 56 e3270 DOI:10.3791/3270
[44]
M Postema O. H Gilja Ultrasound-directed drug delivery Curr Pharm Biotechnol 2007 8 6 355 61 DOI:10.2174/138920107783018453
[45]
N. S Yang J Burkholder B Roberts B Martinell D McCabe In vivo and in vitro gene transfer to mammalian somatic cells by particle bombardment Proc Natl Acad Sci U S A 1990 87 24 9568 72 DOI:10.1073/pnas.87.24.9568
[46]
F Scherer M Anton U Schillinger J Henke C Bergemann A Kruger B Gansbacher C Plank Magnetofection: enhancing and targeting gene delivery by magnetic force in vitro and in vivo Gene Ther 2002 9 2 102 9 DOI:10.1038/sj.gt.3301624
[47]
L Song L Chau Y Sakamoto J Nakashima M Koide R. S Tuan Electric field-induced molecular vibration for noninvasive, high-efficiency DNA transfection Mol Ther 2004 9 4 607 16 DOI:10.1016/j.ymthe.2004.01.017
[48]
Z ur Rehman D Hoekstra I. S Zuhorn Mechanism of polyplex- and lipoplex-mediated delivery of nucleic acids: real-time visualization of transient membrane destabilization without endosomal lysis ACS Nano 2013 7 5 3767 77 DOI:10.1021/nn3049494
[49]
Y. L Chiu T. M Rana siRNA function in RNAi: a chemical modification analysis RNA 2003 9 9 1034 48 DOI:10.1261/rna.5103703
[50]
M. A Campbell J Wengel Locked vs. unlocked nucleic acids (LNA vs. UNA): contrasting structures work towards common therapeutic goals Chem Soc Rev 2011 40 12 5680 9 DOI:10.1039/c1cs15048k
[51]
H Peacock A Kannan P. A Beal C. J Burrows Chemical modification of siRNA bases to probe and enhance RNA interference J Org Chem 2011 76 18 7295 300 DOI:10.1021/jo2012225
[52]
M Warminski P. J Sikorski J Kowalska J Jemielity Applications of Phosphate Modification and Labeling to Study (m)RNA Caps Top Curr Chem (Cham) 2017 375 1 16 DOI:10.1007/s41061-017-0106-y
[53]
B Urban-Klein S Werth S Abuharbeid F Czubayko A Aigner RNAi-mediated gene-targeting through systemic application of polyethylenimine (PEI)-complexed siRNA in vivo Gene Ther 2005 12 5 461 6 DOI:10.1038/sj.gt.3302425
[54]
J Soutschek A Akinc B Bramlage K Charisse R Constien M Donoghue S Elbashir A Geick P Hadwiger J Harborth M John V Kesavan G Lavine R. K Pandey T Racie K. G Rajeev I Rohl I Toudjarska G Wang S Wuschko D Bumcrot V Koteliansky S Limmer M Manoharan H. P Vornlocher Therapeutic silencing of an endogenous gene by systemic administration of modified siRNAs Nature 2004 432 7014 173 8 DOI:10.1038/nature03121
[55]
D. M Copolovici K Langel E Eriste U Langel Cell-penetrating peptides: design, synthesis, and applications ACS Nano 2014 8 3 1972 94 DOI:10.1021/nn4057269
[56]
Z Guo H Peng J Kang D Sun Cell-penetrating peptides Possible transduction mechanisms and therapeutic applications Biomed Rep 2016 4 5 528 534 DOI:10.3892/br.2016.639
[57]
N Shajari B Mansoori S Davudian A Mohammadi B Baradaran Overcoming the Challenges of siRNA Delivery. Nanoparticle Strategies Curr Drug Deliv 2017 14 1 36 46 DOI:10.2174/1567201813666160816105408
[58]
J Halder A. A Kamat C. N Landen Jr L. Y Han S. K Lutgendorf Y. G Lin W. M Merritt N. B Jennings A Chavez-Reyes R. L Coleman D. M Gershenson R Schmandt S. W Cole G Lopez-Berestein A. K Sood Focal adhesion kinase targeting using in vivo short interfering RNA delivery in neutral liposomes for ovarian carcinoma therapy Clin Cancer Res 2006 12 16 4916 24 DOI:10.1158/1078-0432CCR-06-0021
[59]
M Saffari H. R Moghimi C. R Dass Barriers to liposomal gene delivery: from application site to the target Iran J Pharm Res 2016 15 Suppl 3 17
[60]
L Zhu R. I Mahato Lipid and polymeric carrier-mediated nucleic acid delivery Expert Opin Drug Deliv 2010 7 10 1209 26 DOI:10.1517/17425247.2010.513969
[61]
M.-G. K Gayong Shim Joo Yeon Park Yu-Kyoung Oh Application of cationic liposomes for delivery of nucleic acids Asian Journal of Pharmaceutical Sciences 2013 8 78 80 DOI:10.1016/j.ajps.2013.07.009
[62]
A Bajaj S. K Mishra P Kondaiah S Bhattacharya Effect of the headgroup variation on the gene transfer properties of cholesterol based cationic lipids possessing ether linkage Biochim Biophys Acta 2008 1778 5 1222 36 DOI:10.1016/j.bbamem.2007.12.010
[63]
O Paecharoenchai N Niyomtham A Apirakaramwong T Ngawhirunpat T Rojanarata B. E Yingyongnarongkul P Opanasopit Structure relationship of cationic lipids on gene transfection mediated by cationic liposomes AAPS PharmSciTech 2012 13 4 1302 8 DOI:10.1208/s12249-012-y9857-5
[64]
D Liu Y. K Song Cationic liposome-mediated transfection in vivo Gene Therapy and Molecular Biology 1998 2 59 68
[65]
N. S Templeton D. D Lasic P. M Frederik H. H Strey D. D Roberts G. N Pavlakis Improved DNA: liposome complexes for increased systemic delivery and gene expression Nat Biotechnol 1997 15 7 647 52 DOI:10.1038/nbt0797-647
[66]
M Ramamoorth A Narvekar Non viral vectors in gene therapy- an overview J Clin Diagn Res 2015 9 1 GE01 6 DOI:10.7860/JCDR/2015/10443.5394
[67]
K Tatiparti S Sau S. K Kashaw A. K Iyer siRNA Delivery Strategies A Comprehensive Review of Recent Developments Nanomaterials (Basel) 2017 7 4 DOI:10.3390/nano7040077
[68]
H Yin R. L Kanasty A. A Eltoukhy A. J Vegas J. R Dorkin D. G Anderson Non-viral vectors for gene-based therapy Nat Rev Genet 2014 15 8 541 55 DOI:10.1038/nrg3763
[69]
S Biswas V. P Torchilin Dendrimers for siRNA Delivery Pharmaceuticals (Basel) 2013 6 2 161 83 DOI:10.3390/ph6020161
[70]
H Liu H Chang J Lv C Jiang Z Li F Wang H Wang M Wang C Liu X Wang N Shao B He W Shen Q Zhang Y Cheng Screening of efficient siRNA carriers in a library of surface-engineered dendrimers Sci Rep 2016 6 25069 DOI:10.1038/srep25069
[71]
LBJ Dongmei Cun Hanne Mørck Nielsen Moein Moghimi Camilla Foged Polymeric Nanocarriers for siRNA Delivery Challenges and Future Prospects Journal of Biomedical Nanotechnology 2008 4 1 18 DOI:10.1166/jbn.2008.328
[72]
F. M van de Water O. C Boerman A. C Wouterse J. G Peters F. G Russel R Masereeuw Intravenously administered short interfering RNA accumulates in the kidney and selectively suppresses gene function in renal proximal tubules Drug Metab Dispos 2006 34 8 1393 7 DOI:10.1124/dmd.106.009555
[73]
M Neu D Fischer T Kissel Recent advances in rational gene transfer vector design based on poly(ethylene imine) and its derivatives J Gene Med 2005 7 8 992 1009 DOI:10.1002/jgm.773
[74]
L Aagaard J. J Rossi RNAi therapeutics: principles, prospects and challenges Adv Drug Deliv Rev 2007 59 2-3 75 86 DOI:10.1016/j.addr.2007.03.005
[75]
D Fischer Y Li B Ahlemeyer J Krieglstein T Kissel In vitro cytotoxicity testing of polycations: influence of polymer structure on cell viability and hemolysis Biomaterials 2003 24 7 1121 31 DOI:10.1016/S0142-9612(02)00445-3
[76]
S. H Kim J. H Jeong S. H Lee S. W Kim T. G Park PEG conjugated VEGF siRNA for anti-angiogenic gene therapy J Control Release 2006 116 2 123 9 DOI:10.1016/j.jconrel.2006.05.023
[77]
H. K Makadia S. J Siegel Poly Lactic-co-Glycolic Acid (PLGA) as Biodegradable Controlled Drug Delivery Carrier Polymers (Basel) 2011 3 3 1377 1397 DOI:10.3390/polym3031377
[78]
HSM Ajioka C Higuchi T Kashima Aliphatic polyesters and their copolymers synthesized through direct condensation polymerization Polymer Degradation and Stability 1998 39 137 143 DOI:10.1016/S0141-3910(97)00165-1
[79]
K Avgoustakis GLB Gary E Wnek Polylactic-co-Glycolic Acid (PLGA) In Encyclopedia of Biomedical Polymers and Polymeric Biomaterials 2008
[80]
T. K Gurpreet Singh Ravinder Kaur Anudeep Kaur Recent biomedical applications and patents on biodegradable polymer-PLGA International Journal of Pharmacology and Pharmaceutical Sciences 2014 1 2 30 42
[81]
M Bouyer R Guillot J Lavaud C Plettinx C Olivier V Curry J Boutonnat J. L Coll F Peyrin V Josserand G Bettega C Picart Surface delivery of tunable doses of BMP-2 from an adaptable polymeric scaffold induces volumetric bone regeneration Biomaterials 2016 104 168 81 DOI:10.1016/j.biomaterials.2016.06.001
[82]
T. M De Witte L. E Fratila-Apachitei A. A Zadpoor N. A Peppas Bone tissue engineering via growth factor delivery: from scaffolds to complex matrices Regen Biomater 2018 5 4 197 211 DOI:10.1093/rb/rby013
[83]
C. M Hwang A Khademhosseini Y Park K Sun S. H Lee Microfluidic chip-based fabrication of PLGA microfiber scaffolds for tissue engineering Langmuir 2008 24 13 6845 51 DOI:10.1021/la800253b
[84]
D. H Kempen L Lu A Heijink T. E Hefferan L. B Creemers A Maran M. J Yaszemski W. J Dhert Effect of local sequential VEGF and BMP-2 delivery on ectopic and orthotopic bone regeneration Biomaterials 2009 30 14 2816 25 DOI:10.1016/j.biomaterials.2009.01.031
[85]
T. H Kim Y. P Yun Y. E Park S. H Lee W Yong J Kundu J. W Jung J. H Shim D. W Cho S. E Kim H. R Song In vitro and in vivo evaluation of bone formation using solid freeform fabrication-based bone morphogenic protein-2 releasing PCL/PLGA scaffolds Biomed Mater 2014 9 2 025008 DOI:10.1088/1748-6041/9/2/025008
[86]
I Ortega-Oller M Padial-Molina P Galindo-Moreno F O'Valle A. B Jodar-Reyes J. M Peula-Garcia Bone Regeneration from PLGA Micro-Nanoparticles Biomed Res Int 2015 2015 415289 DOI:10.1155/2015/415289
[87]
W. J Zhang W Liu L Cui Y Cao Tissue engineering of blood vessel J Cell Mol Med 2007 11 5 945 57 DOI:10.1111/j.1582-4934.2007.00099.x
[88]
J. M Lu X Wang C Marin-Muller H Wang P. H Lin Q Yao C Chen Current advances in research and clinical applications of PLGA-based nanotechnology Expert Rev Mol Diagn 2009 9 4 325 41 DOI:10.1586/erm.09.15
[89]
N Iverson N Plourde E Chnari G. B Nackman P. V Moghe Convergence of nanotechnology and cardiovascular medicine : progress and emerging prospects BioDrugs 2008 22 1 1 10 DOI:10.2165/00063030-200822010-00001
[90]
M. A Wheatley F Forsberg K Oum R Ro D El-Sherif Comparison of in vitro and in vivo acoustic response of a novel 50:50 PLGA contrast agent Ultrasonics 2006 44 4 360 7 DOI:10.1016/j.ultras.2006.04.003
[91]
U. S Toti B. R Guru M Hali C. M McPharlin S. M Wykes J Panyam J. A Whittum-Hudson Targeted delivery of antibiotics to intracellular chlamydial infections using PLGA nanoparticles Biomaterials 2011 32 27 6606 13 DOI:10.1016/j.biomaterials.2011.05.038
[92]
J. K Vasir V Labhasetwar Biodegradable nanoparticles for cytosolic delivery of therapeutics Adv Drug Deliv Rev 2007 59 8 718 28 DOI:10.1016/j.addr.2007.06.003
[93]
S. K Sahoo V Labhasetwar Enhanced antiproliferative activity of transferrin-conjugated paclitaxel-loaded nanoparticles is mediated via sustained intracellular drug retention Mol Pharm 2005 2 5 373 83 DOI:10.1021/mp050032z
[94]
E. J Benjamin S. S Virani C. W Callaway A. M Chamberlain A. R Chang S Cheng S. E Chiuve M Cushman F. N Delling R Deo S. D de Ferranti J. F Ferguson M Fornage C Gillespie C. R Isasi M. C Jimenez L. C Jordan S. E Judd D Lackland J. H Lichtman L Lisabeth S Liu C. T Longenecker P. L Lutsey J. S Mackey D. B Matchar K Matsushita M. E Mussolino K Nasir M O'Flaherty L. P Palaniappan A Pandey D. K Pandey M. J Reeves M. D Ritchey C. J Rodriguez G. A Roth W. D Rosamond UKA Sampson G. M Satou S. H Shah N. L Spartano D. L Tirschwell C. W Tsao J. H Voeks J. Z Willey J. T Wilkins J. H Wu H. M Alger S. S Wong P Muntner E American Heart Association Council on C Prevention Statistics S Stroke Statistics Heart Disease and Stroke Statistics-2018 Update A Report From the American Heart Association Circulation 2018 137 12 e67 e492 DOI:10.1161/CIR.0000000000000558
[95]
R. A Byrne M Joner A Kastrati Stent thrombosis and restenosis: what have we learned and where are we going?The Andreas Gruntzig Lecture ESC 2014 Eur Heart J 2015 36 47 3320 31 DOI:10.1093/eurheartj/ehv511
[96]
X. F Qiu N. G Dong Z. Q Sun P Hu W Su J. W Shi Controlled release of siRNA nanoparticles loaded in a novel external stent prepared by emulsion electrospinning attenuates neointima hyperplasia in vein grafts Zhonghua Yi Xue Za Zhi 2009 89 41 2938 42 DOI:10.3760/cma.j.issn.0376-2491.2009.41.015
[97]
O Koenig D Zengerle N Perle S Hossfeld B Neumann A Behring M Avci-Adali T Walker C Schlensak H. P Wendel A Nolte RNA-Eluting Surfaces for the Modulation of Gene Expression as A Novel Stent Concept Pharmaceuticals (Basel) 2017 10 1 DOI:10.3390/ph10010023
[98]
H Cohen-Sacks Y Najajreh V Tchaikovski G Gao V Elazer R Dahan I Gati M Kanaan J Waltenberger G Golomb Novel PDGFbetaR antisense encapsulated in polymeric nanospheres for the treatment of restenosis Gene Ther 2002 9 23 1607 16 DOI:10.1038/sj.gt.3301830
[99]
Y Zhao H Gao J He C Jiang J Lu W Zhang H Yang J Liu Co-delivery of LOX-1 siRNA and statin to endothelial cells and macrophages in the atherosclerotic lesions by a dual-targeting core-shell nanoplatform A dual cell therapy to regress plaques J Control Release 2018 283 241 260 DOI:10.1016/j.jconrel.2018.05.041
[100]
R. L Siegel K. D Miller A Jemal Cancer statistics 2018 CA Cancer J Clin 2018 68 1 7 30 DOI:10.3322/caac.21442
[101]
A Singh P Trivedi N. K Jain Advances in siRNAi delivery in cancer therapy Artif Cells Nanomed Biotechnol 2018 46 2 274 283 DOI:10.1080/21691401.2017.1307210
[102]
K. A Woodrow Y Cu C. J Booth J. K Saucier-Sawyer M. J Wood W. M Saltzman Intravaginal gene silencing using biodegradable polymer nanoparticles densely loaded with small-interfering RNA Nat Mater 2009 8 6 526 533 DOI:10.1038/nmat2444
[103]
C Stigliano S Aryal MD De Tullio G. P Nichhia G Pascazio M Svelto P Decuzzi siRNA-chitosan complexes in poly(lactic-co-glycolic acid) nanoparticles for the silencing of aquaporin-1 in cancer cells Mol Pharm 2013 10 8 3186 3194 DOI:10.1021/mp400224u
[104]
A. K Kundu S. V Iyer S Chandra A. S Adhikari T Iwakuma T. K Mandal Novel siRNA formulation to effectively knockdown mutant p53 in osteosarcoma PLoS One 2017 12 6 e0179168 DOI:10.1371/journal.pone.0179168
[105]
B Mansoori A Mohammadi S Davudian S Shirjang B Baradaran The Different Mechanisms of Cancer Drug Resistance vA Brief Review Adv Pharm Bull 2017 7 3 339 348 DOI:10.15171/apb.2017.041
[106]
A Levy K Alhazzani P Dondapati A Alaseem K Cheema K Thallapureddy P Kaur S Alobid A Rathinavelu Focal Adhesion Kinase in Ovarian Cancer A Potential Therapeutic Target for Platinum and Taxane-Resistant Tumors Curr Cancer Drug Targets 2018 DOI:10.2174/1568009618666180706165222
[107]
Y Byeon J. W Lee W. S Choi J. E Won G. H Kim M. G Kim T. I Wi J. M Lee T. H Kang I. D Jung Y. J Cho H. J Ahn B. C Shin Y. J Lee A. K Sood H. D Han Y. M Park CD44-targeted PLGA nanoparticles incorporating paclitaxel and FAK siRNA overcome chemoresistance in epithelial ovarian cancer Cancer Res 2018 DOI:10.1158/0008-5472.aCAN-17-3871
[108]
C Risnayanti Y. S Jang J Lee H. J Ahn PLGA nanoparticles co-delivering MDR1 and BCL2 siRNA for overcoming resistance of paclitaxel and cisplatin in recurrent or advanced ovarian cancer Sci Rep 2018 8 1 7498 DOI:10.1038/s41598-018-25930-7
[109]
M Wu J Li X Lin Z Wei D Zhang B Zhao X Liu J Liu Reduction/photo dual-responsive polymeric prodrug nanoparticles for programmed siRNA and doxorubicin delivery Biomater Sci 2018 6 6 1457 1468 DOI:10.1039/c8bm00226f
[110]
B. C te Boekhorst L. B Jensen S Colombo A. K Varkouhi R. M Schiffelers T Lammers G Storm H. M Nielsen G. J Strijkers C Foged K Nicolay MRI-assessed therapeutic effects of locally administered PLGA nanoparticles loaded with anti-inflammatory siRNA in a murine arthritis model J Control Release 2012 161 3 772 80 DOI:10.1016/j.jconrel.2012.05.004
[111]
C Kelly A. B Yadav C Lawlor K Nolan J O'Dwyer C. M Greene N. G McElvaney N Sivadas J. M Ramsey S. A Cryan Therapeutic aerosol bioengineering of siRNA for the treatment of inflammatory lung disease by TNFalpha gene silencing in macrophages Mol Pharm 2014 11 11 4270 9 DOI:10.1021/mp500473d
[112]
A Frede B Neuhaus R Klopfleisch C Walker J Buer W Muller M Epple A. M Westendorf Colonic gene silencing using siRNA-loaded calcium phosphate/PLGA nanoparticles ameliorates intestinal inflammation in vivo J Control Release 2016 222 86 96 DOI:10.1016/j.jconrel.2015.12.021
[113]
A. M Aldayel Y. W Naguib H. L O'Mary X Li M Niu T. B Ruwona Z Cui Acid-Sensitive Sheddable PEGylated PLGA Nanoparticles Increase the Delivery of TNF-alpha siRNA in Chronic Inflammation Sites Mol Ther Nucleic Acids 2016 5 7 e340 DOI:10.1038/mtna.2016.39
[114]
M. J Gomes C Fernandes S Martins F Borges B Sarmento Tailoring Lipid and Polymeric Nanoparticles as siRNA Carriers towards the Blood-Brain Barrier - from Targeting to Safe Administration J Neuroimmune Pharmacol 2017 12 1 107 119 DOI:10.1007/s11481-016-9685-6
[115]
C Chen H Mei W Shi J Deng B Zhang T Guo H Wang Y Hu EGFP-EGF1-conjugated PLGA nanoparticles for targeted delivery of siRNA into injured brain microvascular endothelial cells for efficient RNA interference PLoS One 2013 8 4 e60860 DOI:10.1371/journal.pone.0060860
[116]
X Liu G Wang Z You P Qian H Chen Y Dou Z Wei Y Chen C Mao J Zhang Inhibition of hypoxia-induced proliferation of pulmonary arterial smooth muscle cells by a mTOR siRNA-loaded cyclodextrin nanovector Biomaterials 2014 35 14 4401 16 DOI:10.1016/j.biomaterials.2014.02.009
Share
Back to top