REVIEW ON NON-VIRAL DELIVERY SYSTEMS FOR siRNA
HTML Full TextREVIEW ON NON-VIRAL DELIVERY SYSTEMS FOR siRNA
Vikas Verma* and Preeti Sangave
Department of Pharmaceutical Chemistry and Pharmaceutical Biotechnology, Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management (SPPSPTM), SVKM’s NMIMS (Deemed to be University), V.L. Mehta Road, Vile Parle (W), Mumbai – 400 056, Maharashtra, India
ABSTRACT: The emergence of RNAi, has found new horizons in siRNA as drug molecule. This technology offers a great promise in the achieving efficient treatment. There is a demand for designing efficient siRNA delivery systems/ vehicles that can achieve targeted delivery and replicate the invitro success. The siRNA delivery system selection is based on target cell type. The siRNA delivery systems are categorized as viral and non-viral delivery systems. Non-viral delivery system includes liposomes; polymers; peptides etc. are the widely studied delivery systems for siRNA. Non-viral delivery systems have advantages of control over functionality and industrial scalability. Liposomes were one of the first studied siRNA non-viral delivery systems which with time have evolved on account of modifications on certain attributes like surface charge, surface tagging, composition etc. Peptides and Polymers are the recent addition to the non-viral siRNA delivery vehicles on account of biocompatibility factor. This review clubs the non-viral siRNA delivery systems which have attracted research and have shown efficiency in the clinical trials.
Keywords: |
Delivery systems, Lipidoids, Liposomes, Peptides, RNAi, siRNA, SNALP
INTRODUCTION:RNAi (RNA interference) is one of the approaches for the treatment of diseases which occur due protein over expression. The RNAi mechanism acts at the molecular level and hence degrade the mRNA that in future will be translated to protein. The RNAi technology utilizes siRNA (short interfering RNA) and miRNA (micro RNA) as its action molecules. Studies in the past have demonstrated siRNA as a successful drug candidate in inhibiting various target classes like transcription factors, growth factors, factor receptors, ion channels and neurotransmitters 1
siRNA is double stranded RNA (dsRNA) which is about 21-25 nucleotides in length and is highly specific for complimentary mRNA sequence 1. The invitro and/or invivo success of siRNA has not been translated to the clinical trials, one of the reasons being delivery challenges and hence there is a demand for safe and effective siRNA delivery systems 2, 3.
The first therapeutic application siRNA in mammals was demonstrated by down regulation of Fas in a mice model with Fas mediated liver fibrosis. It was observed that 82% of mice treated with siRNA against Fas were able to survive for 10 days as compared to 3 days for the control group 4. One of the first examples for cell type specific delivery was shown when siRNA conjugated with protamine/antibody specifically acted on cells expressing the HIV-1 envelope protein 5.
This paper presents the non-viral siRNA delivery systems that have shown promise and have travelled to clinical trials. The non-viral vector designing and selection needs a careful understanding of the target cell type to have an efficient response. As shown in Fig. 1, the siRNA delivery system/vector should also be non-toxic, biocompatible, non-immunogenic with high efficiency to ensure commercial acceptance.
FIGURE 1: DELIVERY SYSTEM/VECTORS FEATURES FOR IDEAL siRNA FORMULATION
Delivery Systems: The delivery system for siRNA has been grouped broadly into two major categories as: viral vectors and non-viral vectors. Non-viral delivery systems offer advantages of ease of synthesis and industrial scalability. As shown in fig. 2, liposomes, peptides and polymers are the extensively studied non-viral delivery vehicles 6. Different types of reported non-viral delivery systems have been discussed in this section. There are constant efforts to achieve siRNA mediated RNAi by playing with the delivery systems and route of administration to treat diseases like cancer, CNS disorders and CVS disorders 7–14.
FIGURE 2: NON-VIRAL DELIVERY SYSTEMS FOR siRNA
- Liposomes: Interaction of the lipids with the nucleic acid leads to the formation of either coated vesicles having nucleic acid in the core or the aggregates, both of which are studied as lipoplexes 15.
Alnylam Inc., (Cambridge, Massachusetts) and Massachusetts Institute of Technology (MIT) collaborated on a project to synthesize a library of more than thousand different lipid-like molecules and screened them for their efficiency to deliver siRNA. They tested these delivery systems in mice to treat the respiratory ailment and found that some of their molecules are ten times more efficacious in delivering siRNA in comparison to the existing non-encapsulated siRNA delivery 16.
Lipofectamine- siRNA formulation achieved 50% knock down of the transgenic mutant huntingtin expression, when it was given by i.v. injection to Huntington’s disease mouse model, R6/2. This formulation decreased the number and size of intra nuclear inclusions in striatal neurons by the end of eighth week 17. Successful delivery of siRNA was observed against Japanese encephalitis and West nile viruses utilizing iFECT and JetSI/DOPE 18.
- pH sensitive Lipoplex: The purpose of designing the pH sensitive liposomes is to ensure an enhanced siRNA release from the endosomal compartment. The lipoplexes can be made pH sensitive in one of the two ways; first, by fusing them with the pH sensitive membrane active polyanions which are termed as ternary complex; secondly, conjugation of pH sensitive liposomes to the polyplexes, which show enhancement of transfection efficiency 15.
Ternary complex prepared with either poly(propyl acrylic acid) or copolymers of MAA/ethyl acrylate (EA)/butyl methacrylate (BMA) have shown enhanced efficiency in comparison to parent, polymer free complex 19–21. These pH sensitive ternary complexes have been reported stable in the presence of serum components 22.
- Cationic lipid based Liposomes: siRNA formulation with a new cationic, amino acid derived lipid, N′,N″-dioleylglutamide (DG), incorporating the fusogenic lipid, 1,2- dioleyl-sn-glycero-3-phosphoethanolamine (DOPE) and cholesterol had high transfection efficiency and low cytotoxicity in comparison with the conventional cationic lipid based liposomes, Lipofectamine® 2000. This formulation achieved a higher viability and delivery of the fluorescent siRNA in the A549, HeLa, and WM266.4 human cancer cell lines in comparison with Lipofectamine® 2000 and cholesterol (DC-Chol) - based liposomes and was found effective in lowering surviving mRNA levels 23.
- Multifunctional Envelope-Type Nano Device (MEND): MENDassembles multiple devices in a single delivery system 24. The lipid envelope of MEND is prepared by condensation with polycation. Modifications of MEND are also reported like; Stearylated octaarginine (STR-R8) modification on the lipid envelope of MEND resulted in enhanced siRNA cellular uptake by macro pinocytosis. MEND modification by addition of dioleoylphosphatidyl ethanolamine (DOPE) and phosphatidic acid (PA) led to an increase in siRNA release. MEND modification by conjugation of pH sensitive fusogenic peptide GALA (WEAALAEALAEALAEHLAEALAEALEALAA) along with cholesterol favored endosomal release of siRNA. STR-R8-MEND modification with a cleavable PEG–peptide–DOPE conjugate (PPD) showed efficiency in in-vivo tumor targeting 25–27.
Nanoparticle in Microparticle Oral System (NiMOS), is a delivery technology for shorter nucleotides, and has been successfully applied to murine IL-10 gene expressing plasmid DNA and later to anti-TNF Alpha siRNA 28.
- Stable Nucleic Acid Lipid Particle (SNALP): Tekmira Pharmaceuticals, in partnership with Alnylam Pharmaceuticals, developed a specialized liposome nanoparticle, Stable Nucleic Acid Lipid Particle (SNALP) which is one of the major achievements in systemic siRNA delivery29. SNALPs are different from the conventional liposomes as their lipid bilayer consists of cationic lipids, neutral lipids and PEG-lipid fusion regulators. SNALP formulations are reported to have a longer half-life in plasma and liver 30. SNALP also protects the siRNA from RNase degradation 31. Anti- hepatitis B virus (HBV) siRNA formulated in SNALP successfully inhibited the HBV replication 32.
Studies on cynomolgus monkeys showed more than 90% silencing of the Apolipoprotein B messenger RNA (ApoBmRNA) in liver 48 hrs after i.v. administration of SNALP formulated siRNA against apolipoprotein B (siApoB). This formulation was shown to have an immediate, potent and lasting biological effect of siRNA treatment 33. SNALP has attracted much of the research as evident from the 6 clinical candidates (ALN-VSP02, TKMApoB, ALN-TTR01, TKM-PLK1, ALN-PCS02, TKM-EBOLA) 34. The phase 1 clinical trials of SNALP-formulated siRNA drugs of Tekmira (TKM-ApoB) targeting ApoB and Alnylam (ALN-PCS) targeting proprotein convertase subtilisn/kexin type 9 (PCSK9) both of which are for hypercholesterolemia were found to be safe and well tolerated without any serious adverse events 35, 36.
- AtuPLEX: Recent modification to liposomal carrier includes 'AtuPLEX', a mixture of cationic and fusogenic lipids developed by Silence Therapeutics plc (formerly Atugen AG) for enhanced transfection efficiency. They reported successful silencing of CD31/Platelet endothelial cell adhesion molecule 1 by i.v. injection of anti-CD31 siRNA-AtuPLEXes. This silencing inhibited tumor growth and suppressed metastases in a PC-3 prostate xenograft model. Repeated systemic administration of the anti-CD31 siRNA-AtuPLEXes did not induced any toxicity 37, 38.
- Lipidoids: Lipidoids are the liposome analogues with different chemical modifications of lipids to meet challenges of degradation, solubility and siRNA release. The siRNA-lipidoid formulation “98N12-5”, which comprises five 12-carbon alkyl-acrylamide chains attached to an amine core, successfully knocked down the blood clotting factor, Factor VII in hepatocytes, mice and rats 16.
Efficient silencing of Hmox1, a liver gene encoding the protein heme oxygenase-1 in mice was achieved using siRNA-lipidoid formulation. The group believes this silencing might also find application in malaria 39. The lipidoid molecule C12-200 is reported as the most efficacious lipidoid to date 40.
- Polymers - Chitosan and Cyclodextrin: Chitosan is a cationic polysaccharide having muco adhesive properties and which is reported to have low cytotoxicity. Chitosan is reported to have low transfection efficiency due to endosomal release challenges 41. It is reported that fully de-N-acetylated chitosan are superior siRNA carriers in comparison to conventional partially N-acetylated chitosan 42.
Chitosan modification by polyethylenimine (PEI) conjugation, Chitosan-graft-polyethylenimine (CHI-g-PEI) made the acid soluble chitosan easily soluble in water at the physiological pH and showed enhancement of transfection efficiency both invitro as well as invivo upon aerosol delivery. The complex CHI-g-PEI and siGFP/ scrsiRNA (scrambled siRNA) showed efficient siRNA condensing and efficiency in silencing of the oncoprotein Akt1.
The group also reported a higher silencing of EGFP expression in A549 using anti-EGFP siRNA 41. siRNA-chitosan formulation was effective in decreasing the population of EGFP expressing epithelial cells in the bronchioles by 43%, upon intranasal administration to EGFP expressing transgenic mice 43.
Glyceraldehyde 3-phosphate dehydrogenase (GAPDH) gene silencing was observed using siRNA- chitosan formulation in HEK293, HeLa and H1299 cell lines. This formulation also showed its efficacy in to primary cells (HUVEC) and MCF-7 cells which were difficult to transfect 42.
Besides chitosan cyclodextrin is another polymer that has also been studied as siRNA delivery system.Cyclodextrin–siRNA nanoparticle formulation of Calando Pharmaceuticals (CALAA-01), against the M2 subunit of ribonucleotide reductase, a critical enzyme in cancer cell proliferation, has entered Phase 1b clinical trial, which is the first-in human study involving systemic siRNA administration to patients (NCT00689065) 44,45.
- Peptides: Ease of synthesis, control over functionalization and stability of the peptide oligonucleotide complex make the low molecular weight peptides as the favorable candidates over lipoplexes as siRNA delivery vehicle. Peptides are viewed as alternative to the cationic polymers for the siRNA delivery due to four major reasons a) Their efficient packaging b) Cell specific delivery c) Efficient membrane transport and d) pH based membrane disruption 46.
The cell penetrating peptides (CPPs) are the widely studied peptides as siRNA delivery system. Table 1 shows some of the extensively studied CPPs. CPPs are less than thirty amino acids and have a positive charge at the physiological pH 6,47. The CPPs are studied under three classes: a) naturally derived peptides (e.g. HIV-1 transactivating protein TAT and penetratin); b) chimeric peptides (e.g. transportan, a chimeric peptide composed of galanin and mastoparan) and c) synthetic ones (e.g. oligoarginine) 48–53.
Suppression of VEGF production in CT-26 cells and in mouse model was observed upon local administration of Chol-R9/siVEGF complexes to subcutaneous tumor 57. Enhanced silencing of p38 MAP kinase mRNA in mouse lung was reported using delivery systems containing cholesterol and cell penetrating peptides (CPPs) TAT and penetratin 58.
TABLE 1: CPPs FOR siRNA DELIVERY
Peptides | Sequence |
HIV TAT | CGRKKRRQRRRPPQC 46 |
JTS | GLFEALLELLESLWELLLEA 46 |
Transportan | GWTLNSAGYLLGKINKALAALAKKIL 52 |
TAT 48-60 | GRKKRRQRRRPPQ 54 |
Penetratin (Antp 43-58) | RQIKIWFQNRRMKWKK 55 |
MPG | GALFLAFLAAALSLMGLWSQPKKKRKV 56 |
Complexing of siRNA with CADY (Ac-GLWRALWRLLRSLWRLLWRA-cysteamide) (1:20 molar ratio) protects siRNA from serum nucleases. siRNA-CADY formulation successfully reduced GAPDH mRNA and protein levels in various cell lines including THP1 and primary HUVEC and 3T3C which are reported as difficult to transfect 59.
The silencing of galanin receptor in human bowes cells was one of the first demonstrations of antisense activity utilizing a 21-mer PNA disulfide having conjugation to the CPPs penetratin or transportan (hybrid of a section of the neuropeptide galanin and the wasp venom peptide mastoparan). This formulation was reported nontoxic at the working concentration and reflected pain modifying activity upon intrathecal injection to rats 60.
The Membrane perturbing peptides (MPPs) are also studied as peptide delivery systems. The MPPs are studied in two categories depending upon the DNA release behavior:
1) Fusogenic peptides
2) Endoosmolytic peptides. The fusogenic peptides act by mediating the DNA release at the endosomal pH.
The endoosmolytic peptides act by endosomal lysis followed by DNA release. Examples of fusogenic peptides include HA-2, N terminal end of influenza virus hemagglutinin and its derivatives like GALA, KALA and synthetic peptides like JTS 61.
- Hybrid Non-viral Delivery Systems: Combination of two or more types of delivery vehicles have led to generation of this category. These combinations may be categorized as Liposome-Peptide, Liposome-Polymer, Peptide -Polymer and any other combination thereof 62, 63. Studies have demonstrated invitro effectiveness of Liposome-siRNA-peptide complexes (LSPCs) in delivering PrP siRNA specifically to AchR-expressing cells, leading to suppressed PrPC expression and eliminated PrPRES formation. This formulation was reported to be stable in serum upon i.v. injection to mice 9.
Liposomal-siRNA- peptide formulation with siRNA against endogenous luciferase and GAPDH successfully silenced these genes in 3 cell lines (1HAEo- human airway epithelial, B104 rat neuroblastoma, Neuro2A-Luc mouse neuroblastoma) with 80% efficiency, similar in efficiency to Lipofectamine® 2000. This formulation had an additional advantage of siRNA protection from RNAse A 64.
Kodama et al (2013) reported binary and ternary complexes of liposomes and polymers for safe and effective siRNA delivery. They utilized cationic polymers and cationic liposomes like polyethylenimine (PEI), polyamidoamine (PAMAM) dendrimer, poly-L-arginine (PLA), trimethyl[2,3-(dioleoxy)-propyl]ammonium chloride (DOTMA), and cholesteryl 3β-N-(dimetylaminnoethyl) carbamate hydrochloride (DC-Chol) for the binary complexation and added γ-polyglutamic acid (γ-PGA) to form the ternary complex.
The ternary complexes showed significant suppression of luciferase activity upon direct injection into the tumors of mice bearing Colon26/Luc cells 65.
PATENTS: In a short span of its existence, RNAi via siRNA have resulted in generation of significant intellectual property. Table 2 shows some of the patents on siRNA delivery vehicles.
TABLE 2: PATENTS ON NON-VIRAL SIRNA DELIVERY SYSTEMS
S. No. | Patent No. | Title | Publication Date |
1 | WO/2004/045582A1 | High-Efficiency Fusogenic Vesicles, Methods of Producing them, and Pharmaceutical Compositions Containing them 66 | 06/03/2004 |
2 | US20050008617A1 | Compositions and methods for delivery of short interfering RNA and short hairpin RNA 67 | 01/13/2005 |
3 | WO/2006/016097A2 | Vector Comprising Polymer Modified siRNA Liposomes 68 | 02/16/2006 |
4 | WO/2009/006905A1 | Dehydrated Chitosan Nanoparticles 69 | 01/15/2009 |
5 | WO/2010/113172A1 | Amphoteric Liposomal Compositions for Cellular Delivery of Small RNA Molecules for use in RNA Interference 70 | 10/07/2010 |
6 | US20130072424A1 | Compounds and Methods for Peptide Ribonucleic Acid Condensate Particles for RNA Therapeutics 71 | 03/21/2013 |
7 | WO/2013/075244A1 | Peptide Sequence Design and Use thereof for Peptide-Mediated siRNA Delivery 72 | 05/30/2013 |
8 | EP2626427A2 | siRNA conjugate and preparation method thereof 73. | 08/14/2013 |
CONCLUSION: siRNA delivery has been attempted by both viral vector and the non-viral vectors. Limitations with the viral vector are a high probability of immune response and random binding of the nucleotide to the non-target sequences, both of which are not desired. The non-viral vectors on the other hand have a lower efficiency in comparison to the viral counterparts but they show very less immune response besides targeted binding. It is these attributes that have made them as the favorable siRNA delivery vehicle.
It is the liposomal delivery systems which appeared in most of the clinical trial formulations of siRNA. The role of proteins and polymers is also under research for its efficient targeting. In the near future it will be the protein/ peptide delivery systems that will challenge the liposomal delivery systems for the efficient silencing. As these proteins/ peptides upon degradation form amino acids which can be taken up by the cell itself. Role of polymers is equally valued although it is more stressed on the polymers from the natural sources like bacteria etc.
One of the sections the paper discusses the hybrid vehicles which are actually the intra non-viral hybrid vehicles. These hybrid siRNA delivery systems will be the future attraction of the siRNA delivery research. These hybrid siRNA delivery systems play with the surface properties of the liposomes which have proved its efficiency. Hybrid non-viral vectors like, liposome –siRNA- peptide are also reported to be efficient and have advantage of serum stability which is very vital for successful application of RNAi.
The selection of the appropriate delivery system is very critical decision. This selection has to be made on account of certain attributes like the surface properties, nucleotide loading capacity, and stability of the delivery systems besides the invivo behavior. In order to achieve targeted delivery various modifications in the non-viral siRNA delivery vehicles have been attempted like, surface charge, peptide tagging or covalent attachment of target cell receptor’s activators for efficient entry into the cell.
The non-viral vectors have advantages of industrial scale up which favor commercialization possibility. It is the industrial scale up property of the non-viral siRNA delivery vehicles which have made them travel from the lab to the clinical trials. It is evident from the literature that the RNAi technology is growing but there is still a missing link that needs to be filled to convert the clinical failure into success and siRNA delivery system designing is a critical feature of that link.
ACKNOWLEDGEMENTS: We gratefully acknowledge Sneha Ranade, Doctoral Fellow, Dr. R.S. Gaud, Dean SPPSPTM and Dr. Bala Prabhakar, Associate Dean, SPPSPTM for their continual support and motivation.
REFERENCES:
- Sah DWY. Therapeutic potential of RNA interference for neurological disorders. Life Sci 2006, 79:1773-80.
- Nguyen T, Menocal EM, Harborth J, Fruehauf JH. RNAi therapeutics: an update on delivery. Curr Opin Mol Ther 2008, 10:158-67.
- Xie FY, Woodle MC, Lu PY. Harnessing in vivo siRNA delivery for drug discovery and therapeutic development. Drug Discov Today 2006, 11:67-73.
- Song E, Lee S-K, Wang J, et al. RNA interference targeting Fas protects mice from fulminant hepatitis. Nat Med 2003,9:347-51.
- Song E, Zhu P, Lee S-K, et al. Antibody mediated in vivo delivery of small interfering RNAs via cell-surface receptors. Nat Biotechnol 2005, 23:709-17.
- Laufer S, Detzer A, Sczakiel G, et al: RNA Technol Their Appl, Selected Strategies for the Delivery of siRNA In vitro and In vivo, Springer-Verlag: Berlin Heidelberg, Germany, 2010: 29-58.
- Gomes-da-Silva LC, Fernández Y, Abasolo I, et al. Efficient intracellular delivery of siRNA with a safe multitargeted lipid-based nanoplatform. Nanomedicine (Lond) 2013, 8:1397-413.
- Kenjo E, Asai T, Yonenaga N, et al. Systemic delivery of small interfering RNA by use of targeted polycation liposomes for cancer therapy. Biol Pharm Bull 2013, 36:287-91.
- Pulford B, Reim N, Bell A, et al. Liposome-siRNA-peptide complexes cross the blood-brain barrier and significantly decrease PrP on neuronal cells and PrP in infected cell cultures. PLoS One 2010, 5:e11085.
- Khatri N, Misra A. Development of siRNA lipoplexes for intracellular delivery in lung cancer cells. J Pharm Bioallied Sci 2012, 4:S1–3.
- Sato Y, Hatakeyama H, Sakurai Y, et al. A pH-sensitive cationic lipid facilitates the delivery of liposomal siRNA and gene silencing activity in vitro and in vivo. J Control Release 2012, 163:267-76.
- Zhang S, Zhi D, Huang L. Lipid-based vectors for siRNA delivery. J Drug Target 2012, 20:724-35.
- Dorasamy S, Narainpersad N, Singh M, et al. Novel targeted liposomes deliver sirna to hepatocellular carcinoma cells in vitro. Chem Biol Drug Des 2012, 80:647-56.
- Krebs MD, Alsberg E. Localized, targeted, and sustained siRNA delivery. Chemistry 2011, 17:3054-62.
- Felber AE, Dufresne MH, Leroux JC. pH-sensitive vesicles, polymeric micelles, and nanospheres prepared with polycarboxylates. Adv Drug Deliv Rev 2012, 64:979-92.
- Akinc A, Zumbuehl A, Goldberg M, et al. A combinatorial library of lipid-like materials for delivery of RNAi therapeutics. Nat Biotechnol 2008, 26:561-9.
- Wang YL, Liu W, Wada E, et al. Clinico-pathological rescue of a model mouse of Huntington’s disease by siRNA. Neurosci Res 2005, 53:241-9.
- Kumar P, Lee SK, Shankar P, et al. A single siRNA suppresses fatal encephalitis induced by two different flaviviruses. PLoS Med 2006, 3:e96.
- Cheung CY, Murthy N, Stayton PS, et al.A pH-sensitive polymer that enhances cationic lipid-mediated gene transfer. Bioconjug Chem 2001, 12:906-10.
- Jones RA, Cheung CY, Black FE, et al. Poly(2-alkylacrylic acid) polymers deliver molecules to the cytosol by pH-sensitive disruption of endosomal vesicles. Biochem J 2003, 372:65-75.
- Yessine MA, Meier C, Petereit HU, et al. On the role of methacrylic acid copolymers in the intracellular delivery of antisense oligonucleotides. Eur J Pharm Biopharm 2006, 63:1-10.
- Cheung CY, Stayton PS, Hoffman AS. Poly(propylacrylic acid)-mediated serum stabilization of cationic lipoplexes. J Biomater Sci Polym Ed 2005, 16:163-79.
- Suh MS, Shim G, Lee HY, et al. Anionic amino acid-derived cationic lipid for siRNA delivery. J Control Release 2009,140:268-76.
- Kogure K, Akita H, Yamada Y, et al. Multifunctional envelope-type nano device (MEND) as a non-viral gene delivery system. Adv Drug Deliv Rev 2008, 60:559–71.
- Li W, Nicol F, Szoka FC. GALA: a designed synthetic pH-responsive amphipathic peptide with applications in drug and gene delivery. Adv Drug Deliv Rev 2004, 56:967-85.
- Nakamura Y, Kogure K, Futaki S, Harashima H. Octaarginine-modified multifunctional envelope-type nano device for siRNA. J Control Release 2007, 119:360-7.
- Sakurai Y, Hatakeyama H, Akita H, et al. Efficient short interference RNA delivery to tumor cells using a combination of octaarginine, GALA and tumor-specific, cleavable polyethylene glycol system. Biol Pharm Bull 2009, 32:928-32.
- Bhavsar MD, Amiji MM. Oral IL-10 gene delivery in a microsphere-based formulation for local transfection and therapeutic efficacy in inflammatory bowel disease. Gene Ther 2008, 15:1200-9.
- Rossi JJ. RNAi therapeutics: SNALPing siRNAs in vivo. Gene Ther 2006, 13:583-4.
- Zhou J, Shum KT, Burnett JC, et al. Nanoparticle-Based Delivery of RNAi Therapeutics: Progress and Challenges. Pharmaceuticals (Basel) 2013, 6:85-107.
- Jones D. Teaming up to tackle RNAi delivery challenge. Nat Rev Drug Discov 2009, 8:525-6.
- Morrissey DV, Lockridge JA, Shaw L, et al. Potent and persistent in vivo anti-HBV activity of chemically modified siRNAs. Nat Biotechnol 2005, 23:1002-7.
- Zimmermann TS, Lee ACH, Akinc A, et al. RNAi-mediated gene silencing in non-human primates. Nature 2006, 441:111-4.
- 34. Haussecker D. The Business of RNAi Therapeutics in 2012. Mol Ther Nucleic Acids 2012, 1:e8.
- Burnett JC, Rossi JJ. RNA-based therapeutics: current progress and future prospects. Chem Biol 2012, 19:60-71.
- 36. Frank-Kamenetsky M, Grefhorst A, Anderson NN, et al. Therapeutic RNAi targeting PCSK9 acutely lowers plasma cholesterol in rodents and LDL cholesterol in nonhuman primates. Proc Natl Acad Sci U S A 2008, 105:11915-20.
- Santel A, Aleku M, Keil O, et al. A novel siRNA-lipoplex technology for RNA interference in the mouse vascular endothelium. Gene Ther 2006, 13:1222-34.
- Santel A, Aleku M, Keil O, et al. RNA interference in the mouse vascular endothelium by systemic administration of siRNA-lipoplexes for cancer therapy. Gene Ther 2006, 13:1360-70.
- Epiphanio S, Mikolajczak SA, Gonçalves LA, et al. Heme oxygenase-1 is an anti-inflammatory host factor that promotes murine plasmodium liver infection. Cell Host Microbe 2008,3:331-8.
- Love KT, Mahon KP, Levins CG, et al. Lipid-like materials for low-dose, in vivo gene silencing. Proc Natl Acad Sci U S A 2010, 107:1864-9.
- Jere D, Jiang HL, Kim YK, et al. Chitosan-graft-polyethylenimine for Akt1 siRNA delivery to lung cancer cells. Int J Pharm 2009, 378:194-200.
- Malmo J, Sørgård H, Vårum KM, et al. siRNA delivery with chitosan nanoparticles: Molecular properties favoring efficient gene silencing. J Control Release 2012, 158:261-8.
- Howard KA, Rahbek UL, Liu X, et al. RNA interference in vitro and in vivo using a novel chitosan/siRNA nanoparticle system. Mol Ther 2006, 14:476–84.
- Davis ME. The first targeted delivery of siRNA in humans via a self-assembling, cyclodextrin polymer-based nanoparticle: from concept to clinic. Mol Pharm 2009, 6:659-68.
- Alabi C, Vegas A, Anderson D. Attacking the genome: emerging siRNA nanocarriers from concept to clinic. Curr Opin Pharmacol 2012, 12:427-33.
- Mann A, Thakur G, Shukla V, et al. Peptides in DNA delivery: current insights and future directions. Drug Discov Today 2008, 13:152-60.
- Hoyer J, Neundorf I. Peptide vectors for the nonviral delivery of nucleic acids. Acc Chem Res 2012, 45:1048-56.
- Frankel AD, Pabo CO. Cellular uptake of the tat protein from human immunodeficiency virus. Cell 1988, 55:1189-93.
- Futaki S. Oligoarginine vectors for intracellular delivery: design and cellular-uptake mechanisms. Biopolymers 2006, 84:241-9.
- Futaki S, Suzuki T, Ohashi W, et al. Arginine-rich peptides. An abundant source of membrane-permeable peptides having potential as carriers for intracellular protein delivery. J Biol Chem 2001, 276:5836-40.
- Green M, Loewenstein PM. Autonomous functional domains of chemically synthesized human immunodeficiency virus tat trans-activator protein. Cell 1988, 55:1179-88.
- Pooga M, Hällbrink M, Zorko M, et al. Cell penetration by transportan. FASEB J 1998, 12:67-77.
- Tréhin R, Merkle HP. Chances and pitfalls of cell penetrating peptides for cellular drug delivery. Eur J Pharm Biopharm 2004, 58:209-23.
- Vivès E, Brodin P, Lebleu B. A truncated HIV-1 Tat protein basic domain rapidly translocates through the plasma membrane and accumulates in the cell nucleus. J Biol Chem 1997, 272:16010-7.
- Derossi D, Calvet S, Trembleau A, et al. Cell internalization of the third helix of the Antennapedia homeodomain is receptor-independent. J Biol Chem 1996, 271:18188-93.
- Deshayes S, Plénat T, Aldrian-Herrada G, Divita G, et al. Primary amphipathic cell-penetrating peptides: structural requirements and interactions with model membranes. Biochemistry 2004, 43:7698-706.
- Kim WJ, Christensen LV, Jo S, et al. Cholesteryl oligoarginine delivering vascular endothelial growth factor siRNA effectively inhibits tumor growth in colon adenocarcinoma. Mol Ther 2006, 14:343-50.
- Moschos SA, Jones SW, Perry MM, et al. Lung delivery studies using siRNA conjugated to TAT(48-60) and penetratin reveal peptide induced reduction in gene expression and induction of innate immunity. Bioconjug Chem 2007,18:1450–9.
- Crombez L, Aldrian-Herrada G, Konate K, et al. A new potent secondary amphipathic cell-penetrating peptide for siRNA delivery into mammalian cells. Mol Ther 2009, 17:95-103.
- Pooga M, Soomets U, Hällbrink M, et al. Cell penetrating PNA constructs regulate galanin receptor levels and modify pain transmission in vivo. Nat Biotechnol 1998, 16:857-61.
- Wagner E. Application of membrane-active peptides for nonviral gene delivery. Adv Drug Deliv Rev 1999,38:279-89.
- Lee SK, Siefert A, Beloor J, et al. Cell-specific siRNA delivery by peptides and antibodies. Methods Enzymol 2012, 502:91-122.
- Schäfer J, Höbel S, Bakowsky U, et al. Liposome-polyethylenimine complexes for enhanced DNA and siRNA delivery. Biomaterials 2010, 31:6892-900.
- Tagalakis AD, He L, Saraiva L, et al. Receptor-targeted liposome-peptide nanocomplexes for siRNA delivery. Biomaterials 2011, 32:6302-15.
- Kodama Y, Harauchi S, Kawanabe S, et al. Safe and effective delivery of small interfering RNA with polymer- and liposomes-based complexes. Biol Pharm Bull 2013, 36:995-1001.
- Jorgensen M, Keller M, Miller AD, et al. Vector comprising polymer modified sirna liposomes. WO2006016097A2, 2006.
- Han BR, Park HO, Shin MS, et al. Sirna conjugate and preparation method thereof. EP2626427A2, 2013.
- Brunner J, Vadrucci S, Zurbriggen R. High-efficiency fusogenic vesicles, methods of producing them, and pharmaceutical compositions containing them. WO2004045582A1, 2004.
- Adami RC, Zhu T, Cui K, et al. Compounds and methods for peptide ribonucleic acid condensate particles for rna therapeutics. US20130072424A1, 2013.
- Chen J, Eisen H, Ge Q. Compositions and methods for delivery of short interfering RNA and short hairpin RNA. US20050008617A1, 2005.
- Bhattacharyya J, Chaudhuri A. Amphoteric liposomal compositions for cellular delivery of small RNA molecules for use in rna interference. WO2010113172A1, 2010.
- Flemming OM, Andersen B, Howard KA, et al. Dehydrated chitosan nanoparticles. WO2009006905A1, 2009.
- Chen P, Jafari M, Xu W, et al. Peptide sequence design and use thereof for peptide-mediated sirna delivery. WO2013075244A1, 2013.
How to cite this article:
Verma V and Sangave P: Review on Non-viral delivery systems for siRNA. Int J Pharm Sci Res 2014; 5(2): 294-01.doi: 10.13040/IJPSR.0975-8232.5(2).294-01
All © 2013 are reserved by International Journal of Pharmaceutical Sciences and Research. This Journal licensed under a Creative Commons Attribution-NonCommercial-ShareAlike 3.0 Unported License.
Article Information
1
294-301
377
1105
English
IJPSR
Vikas Verma* and Preeti Sangave
Department of Pharmaceutical Chemistry and Pharmaceutical Biotechnology, Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management (SPPSPTM), SVKM’s NMIMS (Deemed to be University), V.L. Mehta Road, Vile Parle (W), Mumbai – 400 056, Maharashtra, India
vikasvma@gmail.com
07 September, 2013
30 October, 2013
16 January, 2014
http://dx.doi.org/10.13040/IJPSR.0975-8232.5(2).294-01
01 February, 2014