153
Views
101
CrossRef citations to date
0
Altmetric
Review

Research progress on siRNA delivery with nonviral carriers

, &
Pages 1017-1025 | Published online: 11 May 2011

Abstract

RNA interference is a powerful method for the knockdown of pathologically relevant genes. Small interfering RNAs (siRNAs) have been widely demonstrated as effective biomedical genetic-therapy applications for many diseases. Unfortunately, siRNA duplexes are not ideal drug-like molecules. Problems hindering their effective application fundamentally lie in their delivery, stability, and off-target effects. Delivery systems provide solutions to many of the challenges facing siRNA therapeutics. Due to some fatal disadvantages of viral vectors, nonviral carriers have been studied extensively. Aside from liposomes, nanoparticles and cationic polymer carriers have exhibited improved in vivo stability, better biocompatibility, and efficiency for gene silencing with less cellular toxicity. They may represent a promising strategy for siRNA-based therapies, especially as nanomaterials. The present review also summarizes other methods of siRNA delivery and the side effects of the nanoparticles.

Introduction

Since its discovery in 1998 by Fire et al, RNA interference (RNAi) has represented a promising new approach towards the inhibition of gene expression in vitro or in vivo.Citation1Citation3 In 2001, Elbashir et alCitation4 using synthetic small interfering RNA (siRNA), showed for the first time that RNAi also occurs in mammalian cell lines, making successful development of RNAi possible. Rapid progress in our understanding of RNAi-based mechanisms has led to the application of this powerful mechanism in the study of gene function, as well as therapeutic applications for disease treatment.

RNAi is mediated through approximately 21–23 nt, double-stranded siRNAs that trigger sequence-specific cleavage of mRNA molecules, leading to their subsequent degradation.Citation5 These siRNAs are generated intracellularly through the cleavage of longer double-stranded RNAs,Citation6,Citation7 or are introduced into the cell as chemically synthesized siRNA molecules.Citation4

However, the naked siRNA molecule, with negative charges, is susceptible to serum nucleases, renal clearance, and nontargeted biodistribution, making cellular target sites more difficult to access. Thus, it has many limitations, such as poor stability, short half-life, and low efficiency. The major hurdle faced by current RNAi therapeutic strategies is the efficiency of the delivery system. Recently, viral delivery, such as lentivirus and adenovirus delivery, of DNA-based siRNA constructs for RNAi-mediated vascular endothelial growth factor (VEGF) downregulation showed anti-neovascularization effects in various animal models.Citation8Citation10 Viral vectors are highly efficient delivery systems for nucleic acids; however, the potential for mutagenicity, limited loading capacities, high production costs, and most importantly, safety risks caused by their inflammatory and immunogenic effects severely limit the applicability of viruses. These concerns have led to the pursuit of nonviral alternatives. Thus, the direct, systemic, nonviral administration of siRNA molecules that allows for therapeutic use is most desirable.Citation11 Nonviral vectors are capable of delivering nucleic acids, including genes, siRNA, or antisense RNA into cells, thus potentially resulting in their functional expression. These vectors are considered an attractive alternative for virus-based delivery systems.Citation12

Multiple nonviral siRNA delivery systems include chemical modification of siRNA, liposomes, nanoparticles for siRNA delivery, cell-penetrating peptides, and targeted delivery. Positively charged cationic liposomes and polymers, such as polyethyleneimine, are currently the two major carriers used to complex with negatively charged siRNA for systemic delivery.Citation13 Some of these novel vehicles can potentially overcome extracellular and intracellular barriers, and facilitate the site-specific delivery, cellular uptake, and intracellular target interactions of siRNA.Citation14

The current review discusses the progress in the application of nonviral systems for synthetic RNAi molecule delivery, with focus on the characteristics and advantageous properties of siRNA nanoparticle systems.

Chemical modification of siRNA

The half-life of unmodified naked siRNAs in vivo ranges in minutes, but this can be significantly improved to hours by chemical modifications.Citation15,Citation16 In addition, chemical modification of siRNAs may enhance biological stability without adverse effects on the gene-silencing activity. Furthermore, modified siRNAs with superior potency reduces the dose required for gene silencing,Citation10 and specific chemical modifications can minimize siRNA side effects, such as the induction of recipient immune responses and inherent off-targeting effects.Citation17,Citation18 Various chemical modifications to the backbone, nucleobases, terminals, and sugars of siRNAs have been reported so far, which are mainly focused on increasing its stability and enhancing its cellular uptake.

The most widely used siRNA modifications are on sugar moieties, which commonly include replacement of the 2′-fluoro (2′-F), 2′-O-methyl, 2′-halogen, 2′-amine,Citation19 and locked nucleic acid (LNA),Citation20 all of which have shown significant increase in siRNA serum stability. Layzer et alCitation21 have demonstrated that siRNA modified with 2′-fluoro (2′-F) pyrimidines are more functional in cell culture and have greatly increased stability and prolonged half-life (24 hours) in human plasma, compared with 2′-OH (1 minute) containing siRNAs. Jackson et alCitation22 showed that 2′-O-methyl modifications to specific positions within the siRNA seed region reduces the number of off-target transcripts and the magnitude of their regulation, without significantly affecting the silencing of the intended targets.

The simplest approach to increase nuclease stability has been to modify the internucleotide phosphate linkage directly.Citation23 Phosphorothioate (P = S) modifications can be placed in the RNA duplex easily at any desired position and will enhance the stability of siRNA in nuclease environments. Overhoff and Sczakiel stated that phosphorothioate (PTO)-derived oligonucleotides stimulate the physical cellular uptake of siRNA in human cells.Citation24 On the other hand, siRNAs with boranophosphonate (P = B) backbone modifications have much higher nuclease resistance than unmodified ones, with less cytotoxicity. In addition, Hall et alCitation25 demonstrated that boranophosphate siRNAs are consistently more effective than siRNAs with the widely used phosphorothioate modification. Furthermore, boranophosphate siRNAs are at least 10 times more nuclease resistant than unmodified siRNAs. Therefore, the biochemical properties of boranophosphate siRNAs make them promising candidates for RNAi-based therapeutic applications. Recently, some groups used the same modified siRNA to treat patients with age-related macular degeneration (AMD). The process has reached Phase II clinical trials, and it was found to have no observable side effects.Citation26

To date, much of the focus has been on modifying the RNA backbone, and some laboratories have modified the siRNA bases that are centrally involved in target recognition. Terrazas and KoolCitation27 explored the effects of methyl and propynyl substitution on siRNA duplex stability and cellular RNAi activity. The results suggested that smaller 5-methyl substitutions do not adversely affect gene-silencing activity; furthermore, this modification contributes positively to siRNA stability in human serum.

Terminal nucleotide modifications of the siRNA (also called siRNA conjugates) have also been reported as an efficient delivery strategy. These include peptide modification, such as transactivating transcriptional activator (TAT) peptide, cholesterol conjugation, folate, and aptamer conjugation, which offer opportunities to enhance pharmacological characteristics of or introduce special features to siRNA.Citation28,Citation29 Soutschek et alCitation30 reported that if both 5′- and 3′-terminal modifications of the sense strand are well modified siRNAs, they could silence the endogenous gene that encodes apolipoprotein B after intravenous injection in mice.

Chemical modification of siRNA can increase the stability of the RNA duplex to nucleases, minimize the possibility of immunostimulatory responses, and decrease the possibility of off-target effects, as well as improve its pharmacodynamic properties and delivery to target cells.Citation31 Chemical modification provides solutions to many of the challenges facing siRNA therapeutics, but the remaining challenge is to find universal chemical modification strategies, or to predict reliably which modifications will be effective for a given sequence.Citation32

Liposome-formulated delivery system

Liposomes are probably the most extensively used materials for the delivery of gene molecules ever since their ability to transport the preproinsulin gene to the liverCitation33 was demonstrated 25 years ago. Hence, their characteristics are outlined in detail. Liposomes, vesicles with an aqueous compartment enclosed in a phospholipid bilayer, can fuse with cell membranes and enhance drug delivery into cells. Polar drugs can be entrapped in their aqueous center. When lipids combine with nucleic acids to form amorphous particles, they are known as lipoplexes.Citation34 siRNAs can either be entrapped within a lipid coreCitation35 or attached to the surface of the lipid materialsCitation36 for delivery. The liposome can protect nucleic acids from enzymatic degradation and deliver nucleic acids into cells by interacting with the negatively charged cell membrane. There are two kinds of liposome: neutral and cationic lipid material. Neutral liposomes confer low toxicity to mammalians with low transfection efficiency due to their surface charge. But cationic liposomes can cross the cell membrane and reach the target genes with good biocompatibility.Citation37,Citation38

There are two kinds of liposome, neutral and cationic, based on their lipid composition. Initially, neutral liposomes were used for in vitro and in vivo delivery of nucleic acids because of their low toxicity, low immunogenicity, and easy production. Halder et alCitation39 examined the therapeutic potential of focal adhesion kinase (FAK)-siRNA in the neutral liposome 1,2-dioleoyl-sn-glycero-3-phosphatidylcholine (DOPC). The results show that treatment with FAK siRNA-DOPC results in decreased microvessel density, decreased expression of VEGF and matrix metalloproteinase-9, and increased apoptosis of tumor-associated endothelial cells and tumor cells.

Given that neutral liposomes confer low transfection efficiency due to their lack of surface charges, this methodology was soon supplanted when cationic liposomes were developed in 1989.Citation40 The cationic charge can electrostatically combine with siRNA to achieve a more robust construct to improve cell entry and protect siRNA against serum degradation, whereas neutral lipids facilitate fusion with the host cell membrane.Citation41 Spagnou et alCitation37 selected a number of cationic liposome/lipid-based systems to investigate the optimum lipid to nucleic acid ratio, mode of delivery, biocompatibility, and dose-response effects. The results demonstrate that the systems mediate a maximal specific gene silencing and knockdown with significantly low toxicity to mammalians. Leal et alCitation42 reported the development of cationic liposome (CL)-siRNA complexes with novel cubic phase nanostructures that exhibit efficient silencing at low toxicity. This finding underscored the importance of understanding the membrane-mediated interactions between the CL-siRNA complex nanostructure and cell components in developing CL-based gene-silencing vectors.

Some cationic liposomes, such as Lipofectamine™ 2000, Oligofectamine™, and Lipofectamine (Invitrogen), are commercially available.Citation43 HuangCitation44 transfected two keratoepithelin (KE)-specific siRNAs into HEK293 cells via Lipofectamine to treat certain types of inherited corneal stromal dystrophy, and observed that KE expression is reduced by approximately 50%–80%. Kim et alCitation45 applied water-soluble lipopolymer (WSLP) for delivering siRNA targeting VEGF in vitro and in vivo, which significantly increased the efficiency of inhibition. WSLP/siRNA complexes can efficiently protect siRNAs from enzymatic degradation in serum-conditioned media.

Many researchers studied immunoliposomes to develop a vehicle that can be systemically administered safely and repeatedly, and will deliver the siRNA specifically and efficiently to the targeted tissues. Pirollo et alCitation46 engineered a nanosized immunoliposome-based delivery complex that, when systemically administered, will preferentially target and deliver siRNA to tumor cells wherever they occur in the body. They enhanced the efficiency of this complex with the inclusion of a pH-sensitive histidine–lysine peptide in the complex and by delivery of a modified anti-HER-2 siRNA molecule. The complex can sensitize human tumor cells to chemotherapeutics, silence the target gene, and inhibit tumor growth in a cancer model.

Successful lipoplex deliveries of siRNA has been reported in several studies, and mechanisms for the release of payload from lipoplexes have been suggested. Continued research into the mechanism of release may lead to the development of more bioresponsive vectors that can achieve higher levels of silencing for a given dose.Citation47

Nanotechnology-based siRNA delivery system

The birth of nanotechnology has allowed people to transform nature at the molecular and atomic level, enabling the manipulation of single atoms. With the progress of material synthesis and the rise of nanotechnology, the synthesis of nanomaterials with specific functions has become possible. The development of nanotechnology in the biological area has made nanodelivery systems popular. Currently, nanospheres can smoothly pass the blood–brain barrier, testicle–blood barrier, and placenta.Citation48,Citation49 Thus, nanospheres will be suitable transfection carriers for improving the effect of gene therapy.

Due to their low toxicity, biodegradability, and biocompatibility, many nanomaterials are used as transfection carriers, such as chitosan, cyclodextrin, polyethyleneimine (PEI), poly(lactic-co-glycolic acid) (PLGA), dendrimers, magnetic nanoparticles, carbon nanotubes, and gold nanorods. Because of its high nuclease resistance and mucoadhesive properties, chitosan, natural polymers extracted from crustaceans, has become popular siRNA vectors. Howard et alCitation50 used a chitosan-based siRNA nanoparticle delivery system for RNA interference in vitro and in vivo. They observed, using fluorescence microscopy, that Cy5-labeled nanoparticles were rapidly uptaken by NIH 3T3 cells within 1 hour and are accumulated over a 24-hour period. In vivo, effective RNAi was achieved in bronchiole epithelial cells of transgenic enhanced green fluorescent protein (EGFP) mice after nasal administration of chitosan/siRNA formulations. Ghosn et alCitation51 reported the use of imidazole-modified chitosan-siRNA nanoparticles to mediate gene silencing via either intravenous or intranasal administration. The results showed that intravenous delivery demonstrated significant knockdown of glyceraldehyde 3-phosphate dehydrogenase (GAPDH) enzymes in both the lungs and the liver at as low as 1 mg/kg siRNA dose. For intranasal delivery, significant silencing of GAPDH protein expression was seen in the lungs with only 0.5 mg/kg/day siRNA delivered over three consecutive days.

As a cationic polymer, PEI with its high-density charge, which can protect DNA from degeneration by nuclease, has been widely studied in gene delivery. Höbel et alCitation11 established the therapeutic efficacy and safety of PEI F25-LMW/siRNA-mediated knockdown of VEGF in tumor cells. The results showed that PEI F25-LMW/siRNA complexes, which can be stored frozen as opposed to many other carriers, represent an efficient, safe, and promising modality in antitumor therapy. Modification of PEI with several functional moieties revealed that PEI succinylation resulted in up to a 10-fold reduction of polymer toxicity compared with unmodified PEI. Moreover, succinylated PEI/siRNA complexes were able to induce remarkable knockdown of the target luciferase gene at the lowest tested siRNA concentration.Citation52

In the study by Wang,Citation53 TAT-conjugated PEGylated magnetic polymeric liposomes (TAT-PEG-MPLs) formed with superparamagnetic nanoparticles and TAT were successfully prepared and evaluated in vitro and in vivo. The results indicated that TAT-PEG-MPLs were spherical and nonaggregated in solution, with significantly small mean diameters (83.2 nm) and high magnetization. In cell penetration tests performed through fluorescein isothiocyanate (FITC) labeling, the uptake of TAT-PEG-MPLs by MCF-7 cells was greater than that of PEG-MPLs. Most importantly, in vivo animal experiment, MRI, histological analysis, and atomic absorption spectrophotometry revealed that TAT-PEG-MPLs nanoparticles significantly accumulated around the target site, and even inside nerve cells.

PLGA is an amorphous polymer, with molecular weights ranging from 5000–300,000. It can be used as medical film and carrier material for sustained-release dosage systems, with lower toxicity, less irritation, and minimal inflammatory responses. Luo et alCitation54 incorporated the siRNA sequence of the methyl-CpG binding domain protein 1 (MBD1) plasmid into a PLGA-poloxamer carrier to test the therapeutic effect of this compound on BxPC-3 human pancreatic cancer cells. They found that the PLGA-poloxamer carriers can successfully transfect the MBD1 siRNA plasmid into tumor cells and that the PLGA-MBD1 nanoparticle compound can inhibit cell growth and induce apoptosis.

Katas et alCitation55 incorporated PEI into PLGA particles to produce spherical, positively charged nanoparticles that are able to protect siRNA against nuclease degradation. Cell culture studies showed that PLGA-PEI nanoparticles with encapsulated siRNA are more efficient in silencing the targeted gene than PEI alone, with certain biocompatibility.

Ladeira et alCitation56 described a novel approach to siRNA delivery by single-walled carbon nanotubes. In the study, the siRNA delivery system showed nonspecific toxicity and transfection efficiency greater than 95%. This approach offers the potential for siRNA delivery into different types of cells, including hard-to-transfect cells such as neuronal cells and cardiomyocytes.

Gold nanoparticles/nanorods (GNPs/GNRs) have the distinct advantage of being easily modified. Hence, with their positively charged surfaces, GNPs/GNRs can easily attach to negatively charged siRNA. Bonoiu et alCitation57 applied GNRs as siRNA delivery to explore the dopaminergic brain signaling pathway in vitro. Gene silencing in these cells was evident, with no observed cytotoxicity. Moreover, these nanoplexes can transmigrate across the model of the blood–brain barrier. However, reports about GNPs as gene carriers are rare.

Nevertheless, research regarding siRNA delivery was inspired by the molecular machinery within the phi29 bacteriophage DNA packaging motor, which contains six copies of packaging RNA (pRNA) molecules that form a hexameric ring, which is the crucial part of the motor.Citation58 Utilizing the novel properties of this pRNA, we constructed pRNA dimers and trimers with potential to serve as parts in nanotechnology. pRNA-derived nanoparticles have small sizes (20–40 nm), making them particularly suited for in vivo systemic delivery; the optimal size range for cell uptake is 10–100 nm.Citation59 Tarapore et alCitation60 explored the potential of pRNA as a vehicle in carrying siRNA to target metallothionein-IIa (MT-IIA) messenger RNA (mRNA) specifically. They found that pRNA chimeras targeting MT-IIA are localized within the GW/P-bodies, and are more potent than siRNA alone in silencing MT-IIA expression. Therefore, the pRNA system, which can assemble into multivalent nanoparticles, has great potential as a highly potent therapeutic agent.

Targeted siRNA delivery system

Targeted RNAi therapy is a relatively new approach that can be used to silence genes reversibly in vivo by selective targeting. Targeting the diseased cell, organ, or tissue will increase the silencing potency of a given dose of siRNA. Specific cell targeting will also prevent side effects by avoiding nondiseased cells.Citation13 Aptamers, antibodies, small peptides, and other ligands that bind to the signature molecules with high specificity and affinity have been studied extensively for their ability to guide siRNA to the target tissues and cells.Citation47

Dickerson et alCitation61 designed nanoparticles functionalized with peptides that specially target the erythropoietin-producing hepatocellular (Eph) A2 receptor to deliver siRNAs targeting epidermal growth factor receptor (EGFR). The results showed that the nanoparticles decreased EGFR expression levels and significantly increased the sensitivity of this cell line to docetaxel. Agrawal et alCitation62 designed dendrimer-conjugated magnetofluorescent nanoworms called “dendriworms” as a modular platform for siRNA delivery in vivo. Their study showed that dendriworms carrying siRNA against EGFR reduced EGFR protein levels in human glioblastoma cells by 70%–80%; anti-EGFR dendriworms led to specific and significant suppression of EGFR expression.

The folate receptor is also an important target for anticancer drug delivery. Many anticancer drugs, such as prodrugenzymes, toxic proteins, liposome drugs, and nucleic acid molecules, including siRNA, can be combined with folic acid to achieve target specificity. For example, liposome-wrapped siRNA nanoparticles can be modified with folic acid to inhibit the growth of a targeted tumor.Citation63

Aptamers are oligonucleic acid or peptide molecules that are normally selected from a large random sequence pool to bind to a specific target molecule. Chimeric RNA molecules that contain an RNA aptamer directly linked to the passenger strand of siRNA can be transcribed in vitro and readily purified in large quantities. Therefore, aptamers can enhance the ability of siRNAs to target different cells. To date, only a couple of aptamers have been harnessed for targeted siRNA delivery to specific cell populations. Some studies have shown enhancement of target gene silencing activity and specificity using aptamer–siRNA chimeras. Furthermore, anti-tumor activity is further enhanced by appending a polyethylene glycol moiety, which increases the circulating half-life of the chimeras.Citation64

Other delivery systems

Lee et alCitation65 developed pluronic/polyethylenimine shell cross-linked nanocapsules with embedded magnetite nanocrystals (PPMCs) for magnetically triggered delivery of siRNA. PPMC/siRNA–PEG complexes were efficiently taken up by cancer cells upon exposure to a magnet, thereby enhancing intracellular uptake and the silencing effect of siRNA. The study suggests that these novel nanomaterials could be potentially utilized as magnetically triggered delivery of various nucleic acid-based therapeutic agents.

Recently, ultrasound-mediated gene delivery with nano- and microbubbles was developed as a novel nonviral vector system. This system can directly deliver plasmid DNA and siRNA into the cytosol without endocytosis. Therefore, these genes are able to escape lysosomal degradation, resulting in enhanced gene expression efficiency.Citation66,Citation67 Zhou et alCitation68 transfected plasmid DNA of pigment epithelial derived factor (PEDF) into human retinal pigment epithelial (hRPE) cells and choroidal neovascularization (CNV) animal models by ultrasound-mediated microbubbles. The results showed that microbubbles with ultrasound irradiation could significantly enhance PEDF delivery compared with microbubbles or ultrasound alone, and that the CNV of rats was inhibited effectively.

Limitations and other side-effects of siRNA delivery

siRNA is easily degraded by enzymes in blood, tissues, and cells. Several types of chemically modified siRNA have been produced and investigated to improve stability; however, target site accumulation after administration is still extremely low, even when stability is improved.Citation69 Moreover, the cost and safety of these formulations must also be considered,Citation70 and many groups have found that large numbers of 2′-O-Me modifications (in either strand) decrease siRNA activity.Citation71Citation73

Even though an increasing number of cationic liposomes are used as nonviral-based gene vectors, studies have demonstrated that these liposomes still cause significant toxicity,Citation74,Citation75 such as cell contraction, mitotic inhibition, formation of aggregates in blood, and the tendency to induce inflammatory response. Some researchers have been trying to design new liposomal structures to reduce their cytotoxicity, and found that modifying the cationic liposome structure can be an effective strategy for controlling its toxicity.Citation76 Hundreds of lipids share the common structure of a positively charged hydrophilic head and hydrophobic tail that are connected via a linker structure. Most of the linkages between the hydrophilic and hydrophobic moieties are ether, ester, carbamate, or amide bonds that can affect the rate of biodegradation.Citation77 Moreover, their transfection efficiency needs further improvement for in vivo application.

Aside from studies of its efficacy, some researchers investigated the biosafety and toxicity issues of chitosan. A recent study by Chellat et alCitation78 found that high chitosan/DNA nanoparticle concentrations do not induce macrophage secretion of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, and IL-10, which showed that nanoparticles have no evident inflammatory activity. The toxicity of chitosan can be reduced by modification with the nontoxic and biocompatible acrylic imidazole, and additional modifications of the nanoparticle components helpfully reduce toxicity and increase transfection efficiency.Citation79 Dabold et alCitation80 reported that the in vitro toxicity of liposome–chitosan nanoparticle complexes (LCS-NP) in the conjunctival epithelial cell (CEC) line was very low. LCS-NPs were identified inside CECs after 15 minutes and inside primary cultures of CECs after 30 minutes.

Watari et alCitation81 revealed the micro/nanosizing effect of materials on living organisms. Various sizes of particles, from 500 nm to 150 μm, were used to co-culture with human neutrophils, which play a central role in the initial stage of inflammation against foreign bodies, as probe cells. Particles were also inserted into the subcutaneous connective tissue in the abdominal region of Wistar rats. The results showed that: 1) the toxicity level, shown as an absolute intensity of superoxide production, as well as the expression of cytokines IL-1β and TNF-α by micro/nanoparticles is very low; 2) the reaction is nonspecific and any particle below cell size are regarded by cells as a foreign object, including neutrophils and macrophages; and 3) for the materials as implants, there is no strong rejection and it is, therefore, generally acceptable to any person.

However, other researchers reported different results. Nanoscale gene vectors, after being transfected into the body as exogenous materials, are taken up by the reticuloendothelial mononuclear phagocytic system, which is mainly concentrated in the spleen, liver, lungs, bone marrow, and lymph nodes. The nanoparticles could be retained in the tissues for over 30 days; therefore, if not effectively removed, accumulation of the nanosized vectors could lead to certain organ damage.Citation82Citation84

Low immunogenicity is one of the unique characteristics of nonviral gene vectors. However, studies have shown that the DNA–cationic liposome complex (lipoplex) systemic drugs can activate the innate immune system.Citation85 Sakurai et alCitation86 found that the administration of lipoplex produces a greater amount of inflammatory cytokines, such as IL-6, IL-12, and TNF-α, than adenovirus vector administration. On the other hand, chitosan/DNA nanoparticles and single-walled carbon nanotubes are not available for immune stimulation.Citation87Citation89

Clinical trials

From the first in vivo evidence of RNAi-based therapeutic efficacy in an animal disease model in 2003,Citation90 the pace of its drug development has been rapid. Most of the clinical trials approved by the FDA are nontargeted, designed for intravitreal injection or inhalation to cure age-related macular degeneration (AMD), a leading cause of blindness, or respiratory syncytial virus (RSV), the leading cause of pediatric hospitalizations in the United States today. These trials with naked siRNA have shown certain results without untoward toxicity.Citation14,Citation91

Multiple nanoparticle formulations of siRNA for oncology are currently under clinical development.Citation92 The first targeted-delivery siRNA therapeutic agent approved by the US Food and Drug Administration (FDA) is CALAA-01, a PEGylated, transferrin-targeted nanoparticle that can combine siRNA molecule with the transferrin receptor on the tumor cell surface. This is currently being investigated clinically in patients with solid cancers in the United States.Citation93 The results demonstrate that the siRNA nanoparticles are able to provide transient inhibition of tumor growth.

Atu027 is a liposomal formulation of siRNA against protein kinase N3, developed by Silence Therapeutics AG, and is currently being investigated in Phase I clinical trials in Germany.Citation94 Atu027 comprises neutral fusogenic and PEG-modified lipid components with improved pharmacokinetic properties, cellular uptake, and efficient siRNA release from endosomes after endocytosis.Citation95

ALN-VSP, a nontargeted liposomal formulation of two siRNAs targeting kinesin spindle protein and VEGF, is in clinical development in the United States for the treatment of liver cancers. Due to interests in the potent, promising, and novel siRNAs and the need to protect them from degradation within the circulation, interest and research into nanoparticle formulations of nucleic acids is likely to grow continually.Citation92

Perspective of nonviral vector

siRNA technology, which is attracting much interest in the research community, holds great promise as a therapeutic intervention for targeted gene silencing in cancers and other diseases. Several siRNA-based therapeutic agents are already in clinical trials. Further development of siRNA therapy depends on the development of safe and effective carriers for systemic administration. As described in the present study, cationic nanoparticles have emerged as one of the most attractive carriers because of their ability to form complexes with negatively charged siRNA and their high transfection efficiency. As a whole, the transfection efficiency reported thus far for nonviral approaches is still below that of the highly efficient viral vectors. Further improvements to increase the efficiency and reduce the toxicity of nonviral vectors are needed before their clinical significance can be maximized. Therefore, to achieve more efficient, long-lasting, and nontoxic gene delivery vectors, optimized delivery systems still have many challenges to overcome.

Acknowledgements

This subject was supported by fund of Key Project of Tianjin Medical University (2009XK20).

Disclosure

The authors report no conflicts of interest in this work.

References

  • FireAXuSMontgomeryMKKostasSADriverSEMelloCCPotent and specific genetic interference by doublestranded RNA in Caenorhabditis elegansNature199839166698068119486653
  • TurchinovichAZoidlGDermietzelRNon-viral siRNA delivery into the mouse retina in vivoBMC Ophthalmology2010102514711475
  • LeungRKWhittakerPARNA interference: from gene silencing to gene-specific therapeuticsPharmacol Ther2005107222223915908010
  • ElbashirSMHarborthJLendeckelWYalcinAWeberKTuschlTDuplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cellsNature2001411683649449811373684
  • BumcrotDManoharanMKotelianskyVSahDWRNAi therapeutics: a potential new class of pharmaceutical drugsNat Chem Biol200621271171917108989
  • ZamorePDTuschlTSharpPABartelDPRNAi: doublestranded RNA directs the ATP-dependent cleavage of mRNA at 21–23 nucleotide intervalsCell20001011253310778853
  • ParrishSFleenorJXuSMelloCFireAFunctional anatomy of a dsRNA trigger: differential requirement for the two trigger strands in RNA interferenceMol Cell2000651077108711106747
  • GuoBZhangYLuoGLiLZhangJLentivirus-mediated small interfering RNA targeting VEGF-C inhibited tumor lymphangiogenesis and growth in breast carcinomaAnat Rec (Hoboken)20095292563363919382240
  • ChenCSunJLiuGChenJEffect of small interference RNA targeting HIF-1alpha mediated by rAAV combined L: -ascorbate on pancreatic tumors in athymic micePathol Oncol Res200915110911418509748
  • GaoYSMeiJTongTLHuMXueHMCaiXSInhibitory effects of VEGF-siRNA mediated by adenovirus on osteosarcoma-bearing nude miceCancer Biother Radiopharm200924224324719409047
  • HöbelSKoburgerIJohnMPolyethylenimine/small interfering RNA-mediated knockdown of vascular endothelial growth factor in vivo exerts anti-tumor effects synergistically with BevacizumabJ Gene Med201012328730020052738
  • ZuhornISEngbertsJBHoekstraDGene delivery by cationic lipid vectors: overcoming cellular barriersEur Biophys J20073634936217019592
  • PirolloKFChangEHTargeted delivery of small interfering RNA: approaching effective cancer therapiesCancer Res2008681247125018316585
  • WangYLiZHanYLiangLHJiANanoparticle-based delivery system for application of siRNA in vivoCurr Drug Metab201011218219620359287
  • ChoungSKimYJKimSParkHOChoiYCChemical modification of siRNAs to improve serum stability without loss of efficacyBiochem Biophys Res Commun2006342391992716598842
  • MookORBaasFde WisselMBFluiterKEvaluation of locked nucleic acid-modified small interfering RNA in vitro and in vivoMol Cancer Ther20076383384317363479
  • Ui-TeiKNaitoYZennoSFunctional dissection of siRNA sequence by systematic DNA substitution: modified siRNA with a DNA seed arm is a powerful tool for mammalian gene silencing with significantly reduced off-target effectNucleic Acids Res20083672136215118267968
  • BramsenJBLaursenMBNielsenAFA large-scale chemical modification screen identifies design rules to generate siRNAs with high activity, high stability and low toxicityNucleic Acids Res20093792867288119282453
  • ChiuYLRanaTMsiRNA function in RNAi: a chemical modification analysisRNA2003991034104812923253
  • PiekenWAOlsenDBBenselerFAurupHEcksteinFKinetic characterization of ribonucleaseresistant 2′-modified hammerhead ribozymesScience19912533143171857967
  • LayzerJMMcCaffreyAPTannerAKHuangZKayMASullengerBAIn vivo activity of nuclease-resistant siRNAsRNA20041076677115100431
  • JacksonALBurchardJLeakeDPosition-specific chemical modification of siRNAs reduces “off-target” transcript silencingRNA2006121197120516682562
  • GaglioneMMessereARecent progress in chemically modified siRNAsMini Rev Med Chem20101057859520500149
  • OverhoffMSczakielGPhosphorothioate-stimulated uptake of short interfering RNA by human cellsEMBO Rep20056121176118116170302
  • HallAHWanJShaughnessyEERamsay ShawBAlexanderKARNA interference using boranophosphate siRNAs: structure-activity relationshipsNucleic Acids Res200432205991600015545637
  • BehlkeMAChemical modification of siRNAs for in vivo useOligonucleotides200818430531919025401
  • TerrazasMKoolETRNA major groove modifications improve siRNA stability and biological activityNucleic Acids Res200937234635319042976
  • ChiuYLAliAChuCYCaoHRanaTMVisualizing a correlation between siRNA localization, cellular uptake, and RNAi in living cellsChem Biol20041181165117515324818
  • LorenzCHadwigerPJohnMVornlocherHPUnverzagtCSteroid and lipid conjugates of siRNAs to enhance cellular uptake and gene silencing in liver cellsBioorg Med Chem Lett2004144975497715341962
  • SoutschekJAkincABramlageBTherapeutic silencing of an endogenous gene by systemic administration of modified siRNAsNature200443217317815538359
  • BehlkeMAChemical modification of siRNAs for in vivo useOligonucleotides200818430531919025401
  • WattsJKDeleaveyGFDamhaMJChemically modified siRNA: tools and applicationsDrug Discov Today20081384285618614389
  • SorianoPDijkstraJLegrandATargeted and nontargeted liposomes for in vivo transfer to rat liver cells of a plasmid containing the preproinsulin I geneProc Natl Acad Sci U S A19838023712871316580630
  • De FougerollesAVornlocherHPMaraganoreJLiebermanJInterfering with disease: a progress report on siRNA-based therapeuticsNat Rev Drug Discov20076644345317541417
  • LiJChenYCTsengYCMozumdarSHuangLBiodegradable calcium phosphate nanoparticle with lipid coating for systemic siRNA deliveryJ Control Release2010142341642119919845
  • BarichelloJMIshidaTKiwadaHComplexation of siRNA and pDNA with cationic liposomes: the important aspects in lipoplex preparationMethods Mol Biol201060546147220072901
  • SpagnouSMillerADKellerMLipidic carriers of siRNA: differences in the formulation, cellular uptake, and delivery with plasmid DNABiochemistry20044342133481335615491141
  • PeerDParkEJMorishitaYCarmanCVShimaokaMSystemic leukocyte-directed siRNA delivery revealing cyclin D1 as an antiinflammatory targetScience2008319586362763018239128
  • HalderJKamatAALandenCNJrFocal adhesion kinase targeting using in vivo short interfering RNA delivery in neutral liposomes for ovarian carcinoma therapyClin Cancer Res200612164916492416914580
  • FelgnerPLRingoldGMCationic liposome-mediated transfectionNature198933762053873882463491
  • ZhangCTangNLiuXLiangWXuWTorchilinVPsiRNA-containing liposomes modified with polyarginine effectively silence the targeted geneJ Control Release200611222923916545478
  • LealCBouxseinNFEwertKKSafinyaCRHighly efficient gene silencing activity of siRNA embedded in a nanostructured gyroid cubic lipid matrixJ Am Chem Soc201013247168411684721028803
  • KimWJKimSWEfficient siRNA delivery with non-viral polymeric vehiclesPharm Res200926365766619015957
  • HuangAJSuppression of keratoepithelin and myocilin by small interfering RNA (an American Ophthalmological Society thesis)Trans Am Ophthalmol Soc200710536537818427622
  • KimWJChangCWLeeMKimSWEfficient siRNA delivery using water soluble lipopolymer for anti-angiogenic gene therapyJ Control Release2007118335736317313987
  • PirolloKFRaitAZhouQMaterializing the potential of small interfering RNA via a tumor-targeting nanodelivery systemCancer Res20076772938294317409398
  • GuoPCobanOSneadNMEngineering RNA for targeted siRNA delivery and medical applicationAdv Drug Deliv Rev201062665066620230868
  • KimDHKangJWKimTRKimEJImJSKimJA polyol-mediated synthesis of titania-based nanoparticles and their electrochemical propertiesJ Nanosci Nanotechnol20077113954395818047095
  • KimBHChoMSKimSYDry sol-gel polycondensation of hydrosilanes to organosilicas catalyzed by colloidal nickel nanoparticlesJ Nanosci Nanotechnol20077113964396818047097
  • HowardKARahbekULLiuXRNA interference in vitro and in vivo using a novel chitosan/siRNA nanoparticle systemMol Ther200614447648416829204
  • GhosnBSinghALiMEfficient gene silencing in lungs and liver using imidazole-modified chitosan as a nanocarrier for small interfering RNAOligonucleotides201020316317220565242
  • ZintchenkoAPhilippADehshahriAWagnerESimple modifications of branched PEI lead to highly efficient siRNA carriers with low toxicityBioconjug Chem20081971448145518553894
  • WangHJZhangSGLiaoZYPEGlated magnetic polymeric liposome anchored with TAT for delivery of drugs across the blood-spinal cord barrierBiomaterials201031256589659620553983
  • LuoGJinCLongJRNA interference of MBD1 in BxPC-3 human pancreatic cancer cells delivered by PLGA-poloxamer nanoparticlesCancer Biol Ther20098759459819276660
  • KatasHCevherEAlparHOPreparation of polyethyleneimine incorporated poly (D,L-lactide-co-glycolide) nanoparticles by spontaneous emulsion diffusion method for small interfering RNA deliveryInt J Pharm20093691–214415419010405
  • LadeiraMSAndradeVAGomesERHighly efficient siRNA delivery system into human and murine cells using single-wall carbon nanotubesNanotechnology2010213838510120798464
  • BonoiuACMahajanSDDingHNanotechnology approach for drug addiction therapy: gene silencing using delivery of gold nanorod-siRNA nanoplex in dopaminergic neuronsProc Natl Acad Sci U S A2009106145546555019307583
  • GuoSTschammerNMohammedSGuoPSpecific delivery of therapeutic RNAs to cancer cells via the dimerization mechanism of phi29 motor pRNAHum Gene Ther20051691097110916149908
  • ShuDMollWDDengZMaoCGuoPBottom-up assembly of RNA arrays and superstructures as potential parts in nanotechnologyNano Lett2004491717172321171616
  • TaraporePShuYGuoPHoSMApplication of Phi29 motor pRNA for targeted therapeutic delivery of siRNA silencing metallothionein-IIA and survivin in ovarian cancersMol Ther201119238639421063391
  • DickersonEBBlackburnWHSmithMHKapaLBLyonLAMcDonaldJFChemosensitization of cancer cells by siRNA using targeted nanogel deliveryBMC Cancer20101011120047689
  • AgrawalAMinDHSinghNFunctional delivery of siRNA in mice using dendriwormsACS Nano2009932495250419673534
  • YoshizawaTHattoriYHakoshimaMKogaKMaitaniYFolate-linked lipid-based nanoparticles for synthetic siRNA delivery in KB tumor xenograftsEur J Pharm Biopharm200870371872518647651
  • DassieJPLiuXYThomasGSSystemic administration of optimized aptamer-siRNA chimeras promotes regression of PSMA-expressing tumorsNat Biotechnol200927983984919701187
  • LeeKBaeKHLeeYLeeSHAhnCHParkTGPluronic/polyethylenimine shell crosslinked nanocapsules with embedded magnetite nanocrystals for magnetically triggered delivery of siRNAMacromol Biosci201010323924519924685
  • SuzukiROdaYUtoguchiNMaruyamaKProgress in the development of ultrasound-mediated gene delivery systems utilizing nano- and microbubblesJ Control Release20111491364120470839
  • HernotSKlibanovALMicrobubbles in ultrasound-triggered drug and gene deliveryAdv Drug Deliv Rev200860101153116618486268
  • ZhouXYLiaoQPuYMUltrasound-mediated microbubble delivery of pigment epithelium-derived factor gene into retina inhibits choroidal neovascularizationChin Med J (Engl)2009122222711271719951601
  • HiguchiYKawakamiSHashidaMStrategies for in vivo delivery of siRNAs: recent progressBioDrugs201024319520520462284
  • ManoharanMRNA interference and chemically modified siRNAsNucleic Acids Res Suppl2003311511614510407
  • ChiuYLRanaTMsiRNA function in RNAi: a chemical modification analysisRNA200391034104812923253
  • BraaschDAJensenSLiuYRNA interference in mammalian cells by chemically modified RNABiochemistry2003427967797512834349
  • CzaudernaFFechtnerMDamesSStructural variations and stabilising modifications of synthetic siRNAs in mammalian cellsNucl Acids Res2003312705271612771196
  • StewartMJPlautzGEDel BuonoLGene transfer in vivo with DNA-liposome complexes: safety and acute toxicity in miceHum Gene Ther1992332672751643147
  • FilionMCPhilipsNCMajor limitations in the use of cationic liposomes for DNA deliveryInt J Pharm19981621159170
  • AberleAMTablinFZhuJWalkerNJGruenertDCNantzMHA novel tetraester construct that reduces cationic lipid-associated cytotoxicity. Implications for the onset of cytotoxicityBiochemistry19983718653365409572871
  • Al-DosariMSGaoXNonviral gene delivery: principle, limitations, and recent progressAAPS J200911467168119834816
  • ChellatFGrandjean-LaquerriereALe NaourRMetalloproteinase and cytokine production by THP-1 macrophages following exposure to chitosan-DNA nanoparticlesBiomaterials200526996197015369684
  • KimTHIhmJEChoiYJNahJWChoCSEfficient gene delivery by urocanic acid-modified chitosanJ Control Release200393338940214644588
  • DieboldYJarrínMSáezVOcular drug delivery by liposome-chitosan nanoparticle complexes (LCS-NP)Biomaterials20072881553156417169422
  • WatariFTakashiNYokoyamaAMaterial nanosizing effect on living organisms: non-specific, biointeractive, physical size effectsJ R Soc Interface20096Suppl 3S371S38819364724
  • ChoMChoWSChoiMThe impact of size on tissue distribution and elimination by single intravenous injection of silica nanoparticlesToxicol Lett2009189317718319397964
  • TangJXiongLWangSDistribution, translocation and accumulation of silver nanoparticles in ratsJ Nanosci Nanotechnol2009984924493219928170
  • XieGSunJZhongGShiLZhangDBiodistribution and toxicity of intravenously administered silica nanoparticles in miceArch Toxicol20108418319019936708
  • UshitoraMSakuraiFYamaguchiTPrevention of hepatic ischemia–reperfusion injury by pre-administration of catalase-expressing adenovirus vectorsJ Control Release2010142343143719951728
  • SakuraiHKawabataKSakuraiFNakagawaSMizuguchiHInnate immune response induced by gene delivery vectorsInt J Pharm20083541–291517640834
  • ChellatFGrandjean-LaquerriereALe NaourRMetalloproteinase and cytokine production by THP-1 macrophages following exposure to chitosan-DNA nanoparticlesBiomaterials200526996197015369684
  • LamCWJamesJTMcCluskeyRHunterRLPulmonary toxicity of single-wall carbon nanotubes in mice 7 and 90 days after intratracheal instillationToxicol Sci200477112613414514958
  • DittoAJShahPNYunYHNon-viral gene delivery using nanoparticlesExpert Opin Drug Deliv20096111149116019780712
  • SongELeeSKWangJRNA interference targeting Fas protects mice from fulminant hepatitisNature Med2003934735112579197
  • DeVincenzoJLambkin-WilliamsRWilkinsonTA randomized, double-blind, placebo-controlled study of an RNAi-based therapy directed against respiratory syncytial virusPNAS2010107198800880520421463
  • HeidelJDDavisMEClinical developments in nanotechnology for cancer therapyPharm Res20112818719920549313
  • DavisMEThe first targeted delivery of siRNA in humans via a self-assembling, cyclodextrin polymer-based nanoparticle: from concept to clinicMol Pharm2009665966819267452
  • AlekuMSchulzPKeilOAtu027, a liposomal small interfering RNA formulation targeting protein kinase N3, inhibits cancer progressionCancer Res200868239788979819047158
  • SantelAAlekuMKeilOA novel siRNA-lipoplex technology for RNA interference in the mouse vascular endotheliumGene Ther200613161222123416625243