Skip to main content
Top
Published in: BioDrugs 3/2010

01-06-2010 | Review Article

Strategies for In Vivo Delivery of siRNAs

Recent Progress

Authors: Dr Yuriko Higuchi, Shigeru Kawakami, Mitsuru Hashida

Published in: BioDrugs | Issue 3/2010

Login to get access

Abstract

RNA interference (RNAi) is a post-transcriptional gene-silencing mechanism that involves the degradation of messenger RNA in a highly sequence-specific manner. Double-stranded small interfering RNA (siRNA), consisting of 21–25 nucleotides, can induce RNAi and inhibit the expression of target proteins. Therefore, siRNA is considered a promising therapeutic for treatment of a variety of diseases, including genetic and viral diseases, and cancer. Clinical trials of siRNA are ongoing or have been planned, although some issues need to be addressed. For example, cellular uptake of naked siRNA is extremely low due to its polyanionic nature. Furthermore, 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; these have involved modification of the siRNA backbone, the sugar moiety, and the nucleotide bases of antisense and/or sense strands. Because the accumulation at the target site after administration is extremely low, even if stability is improved, an effective delivery system is required to induce RNAi at the site of action. Delivery strategies can be categorized into physical methods, conjugation methods, and drug delivery system carrier-mediated methods. Physical techniques can enhance siRNA uptake at a specific tissue site using electroporation, pressure, mechanical massage, etc. Terminal modification of siRNAs can enhance their resistance to degradation by exonucleases in serum and tissue. Moreover, modification with a suitable ligand can achieve targeted delivery. Several types of carrier for drug delivery have been developed for siRNA in addition to traditional cationic liposome and cationic polymer systems. Ultrasound and microbubbles or liposomal bubbles have also been used in combination with a carrier for siRNA delivery. New materials with unique characteristics such as carbon nanotubes, gold nanoparticles, and gold nanorods have attracted attention as innovative carriers for siRNA.
Literature
1.
go back to reference Fire A, Xu S, Montgomery MK, et al. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature 1998 Feb 19; 391(6669): 806–11PubMedCrossRef Fire A, Xu S, Montgomery MK, et al. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature 1998 Feb 19; 391(6669): 806–11PubMedCrossRef
2.
go back to reference Elbashir SM, Harborth J, Lendeckel W, et al. Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature 2001 May 24; 411(6836): 494–8PubMedCrossRef Elbashir SM, Harborth J, Lendeckel W, et al. Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature 2001 May 24; 411(6836): 494–8PubMedCrossRef
3.
go back to reference McManus MT, Sharp PA. Gene silencing in mammals by small interfering RNAs. Nat Rev Genet 2002 Oct; 3(10): 737–47PubMedCrossRef McManus MT, Sharp PA. Gene silencing in mammals by small interfering RNAs. Nat Rev Genet 2002 Oct; 3(10): 737–47PubMedCrossRef
4.
go back to reference Hannon GJ, Rossi JJ. Unlocking the potential of the human genome with RNA interference. Nature 2004 Sep 16; 431(7006): 371–8PubMedCrossRef Hannon GJ, Rossi JJ. Unlocking the potential of the human genome with RNA interference. Nature 2004 Sep 16; 431(7006): 371–8PubMedCrossRef
5.
go back to reference Caplen NJ, Parrish S, Imani F, et al. Specific inhibition of gene expression by small double-stranded RNAs in invertebrate and vertebrate systems. Proc Natl Acad Sci U S A 2001 Aug 14; 98(17): 9742–7PubMedCrossRef Caplen NJ, Parrish S, Imani F, et al. Specific inhibition of gene expression by small double-stranded RNAs in invertebrate and vertebrate systems. Proc Natl Acad Sci U S A 2001 Aug 14; 98(17): 9742–7PubMedCrossRef
6.
go back to reference Takakura Y, Hashida M. Macromolecular carrier systems for targeted drug delivery: pharmacokinetic considerations on biodistribution. Pharm Res 1996 Jun; 13(6): 820–31PubMedCrossRef Takakura Y, Hashida M. Macromolecular carrier systems for targeted drug delivery: pharmacokinetic considerations on biodistribution. Pharm Res 1996 Jun; 13(6): 820–31PubMedCrossRef
7.
go back to reference Higuchi Y, Kawakami S, Yamashita F, et al. The potential role of fucosylated cationic liposome/NFκB decoy complexes in the treatment of cytokine-related liver disease. Biomaterials 2007 Jan; 28(3): 532–9PubMedCrossRef Higuchi Y, Kawakami S, Yamashita F, et al. The potential role of fucosylated cationic liposome/NFκB decoy complexes in the treatment of cytokine-related liver disease. Biomaterials 2007 Jan; 28(3): 532–9PubMedCrossRef
8.
go back to reference Higuchi Y, Kawakami S, Oka M, et al. Intravenous administration of man-nosylated cationic liposome/NFκB decoy complexes effectively prevent LPS-induced cytokine production in a murine liver failure model. FEBS Lett 2006 Jun 26; 580(15): 3706–14PubMedCrossRef Higuchi Y, Kawakami S, Oka M, et al. Intravenous administration of man-nosylated cationic liposome/NFκB decoy complexes effectively prevent LPS-induced cytokine production in a murine liver failure model. FEBS Lett 2006 Jun 26; 580(15): 3706–14PubMedCrossRef
9.
go back to reference Miyao T, Takakura Y, Akiyama T, et al. Stability and pharmacokinetic characteristics of oligonucleotides modified at terminal linkages in mice. Antisense Res Dev 1995 Summer; 5(2): 115–21PubMed Miyao T, Takakura Y, Akiyama T, et al. Stability and pharmacokinetic characteristics of oligonucleotides modified at terminal linkages in mice. Antisense Res Dev 1995 Summer; 5(2): 115–21PubMed
10.
go back to reference Takakura Y, Mahato RI, Yoshida M, et al. Uptake characteristics of oligonucleotides in the isolated rat liver perfusion system. Antisense Nucleic Acid Drug Dev 1996 Fall; 6(3): 177–83PubMedCrossRef Takakura Y, Mahato RI, Yoshida M, et al. Uptake characteristics of oligonucleotides in the isolated rat liver perfusion system. Antisense Nucleic Acid Drug Dev 1996 Fall; 6(3): 177–83PubMedCrossRef
11.
go back to reference Sawai K, Miyao T, Takakura Y, et al. Renal disposition characteristics of oligonucleotides modified at terminal linkages in the perfused rat kidney. Antisense Res Dev 1995 Winter; 5(4): 279–87PubMed Sawai K, Miyao T, Takakura Y, et al. Renal disposition characteristics of oligonucleotides modified at terminal linkages in the perfused rat kidney. Antisense Res Dev 1995 Winter; 5(4): 279–87PubMed
12.
go back to reference Sawai K, Mahato RI, Oka Y, et al. Disposition of oligonucleotides in isolated perfused rat kidney: involvement of scavenger receptors in their renal uptake. J Pharmacol Exp Ther 1996 Oct; 279(1): 284–90PubMed Sawai K, Mahato RI, Oka Y, et al. Disposition of oligonucleotides in isolated perfused rat kidney: involvement of scavenger receptors in their renal uptake. J Pharmacol Exp Ther 1996 Oct; 279(1): 284–90PubMed
13.
go back to reference Medarova Z, Pharm W, Farrar C,et al. In vivo imaging ofsiRNA delivery and silencing in tumors. Nat Med 2007 Mar; 13(3): 372–7PubMedCrossRef Medarova Z, Pharm W, Farrar C,et al. In vivo imaging ofsiRNA delivery and silencing in tumors. Nat Med 2007 Mar; 13(3): 372–7PubMedCrossRef
14.
go back to reference Lee JH, Lee K, Moon SH, et al. All-in-one target-cell-specific magnetic nanoparticles for simultaneous molecular imaging and siRNA delivery. Angew Chem Int Ed Engl 2009; 48(23): 4174–9PubMedCrossRef Lee JH, Lee K, Moon SH, et al. All-in-one target-cell-specific magnetic nanoparticles for simultaneous molecular imaging and siRNA delivery. Angew Chem Int Ed Engl 2009; 48(23): 4174–9PubMedCrossRef
15.
go back to reference Yezhelyev MV, Qi L, O'Regan RM, et al. Proton-sponge coated quantum dots for siRNA delivery and intracellular imaging. J Am Chem Soc 2008 Jul 16; 130(28): 9006–12PubMedCrossRef Yezhelyev MV, Qi L, O'Regan RM, et al. Proton-sponge coated quantum dots for siRNA delivery and intracellular imaging. J Am Chem Soc 2008 Jul 16; 130(28): 9006–12PubMedCrossRef
16.
go back to reference Qi L, Gao X. Quantum dot-amphipol nanocomplex for intracellular delivery and real-time imaging of siRNA. ACS Nano 2008 Jul; 2(7): 1403–10PubMedCrossRef Qi L, Gao X. Quantum dot-amphipol nanocomplex for intracellular delivery and real-time imaging of siRNA. ACS Nano 2008 Jul; 2(7): 1403–10PubMedCrossRef
17.
go back to reference Klein S, Zolk O, Fromm MF, et al. Functionalized silicon quantum dots tailored for targeted siRNA delivery. Biochem Biophys Res Commun 2009 Sep 11; 387(1): 164–8PubMedCrossRef Klein S, Zolk O, Fromm MF, et al. Functionalized silicon quantum dots tailored for targeted siRNA delivery. Biochem Biophys Res Commun 2009 Sep 11; 387(1): 164–8PubMedCrossRef
18.
go back to reference Eder PS, DeVine RJ, Dagle JM, et al. Substrate specificity and kinetics of degradation of antisense oligonucleotides by a 3′ exonuclease in plasma. Antisense Res Dev 1991 Summer; 1(2): 141–51PubMed Eder PS, DeVine RJ, Dagle JM, et al. Substrate specificity and kinetics of degradation of antisense oligonucleotides by a 3′ exonuclease in plasma. Antisense Res Dev 1991 Summer; 1(2): 141–51PubMed
19.
go back to reference Kennedy S, Wang D, Ruvkun G. A conserved siRNA-degrading RNase negatively regulates RNA interference in C. elegans. Nature 2004 Feb 12; 427(6975): 645–9PubMedCrossRef Kennedy S, Wang D, Ruvkun G. A conserved siRNA-degrading RNase negatively regulates RNA interference in C. elegans. Nature 2004 Feb 12; 427(6975): 645–9PubMedCrossRef
20.
go back to reference Amarzguioui M, Holen T, Babaie E, et al. Tolerance for mutations and chemical modifications in a siRNA. Nucleic Acids Res 2003 Jan 15; 31(2): 589–95PubMedCrossRef Amarzguioui M, Holen T, Babaie E, et al. Tolerance for mutations and chemical modifications in a siRNA. Nucleic Acids Res 2003 Jan 15; 31(2): 589–95PubMedCrossRef
21.
go back to reference Bramsen JB, Laursen MB, Damgaard CK, et al. Improved silencing properties using small internally segmented interfering RNAs. Nucleic Acids Res 2007; 35(17): 5886–97PubMedCrossRef Bramsen JB, Laursen MB, Damgaard CK, et al. Improved silencing properties using small internally segmented interfering RNAs. Nucleic Acids Res 2007; 35(17): 5886–97PubMedCrossRef
22.
go back to reference Chiu YL, Rana TM. RNAi in human cells: basic structural and functional features of small interfering RNA. Mol Cell 2002 Sep; 10(3): 549–61CrossRef Chiu YL, Rana TM. RNAi in human cells: basic structural and functional features of small interfering RNA. Mol Cell 2002 Sep; 10(3): 549–61CrossRef
23.
go back to reference Jackson AL, Burchard J, Leake D, et al. Position-specific chemical modification of siRNAs reduces “off-target” transcript silencing. RNA 2006 Jul; 12(7): 1197–205PubMedCrossRef Jackson AL, Burchard J, Leake D, et al. Position-specific chemical modification of siRNAs reduces “off-target” transcript silencing. RNA 2006 Jul; 12(7): 1197–205PubMedCrossRef
24.
go back to reference Bramsen JB, Laursen MB, Nielsen AF, et al. A large-scale chemical modification screen identifies design rules to generate siRNAs with high activity, high stability and low toxicity. Nucleic Acids Res 2009 May; 37(9): 2867–81PubMedCrossRef Bramsen JB, Laursen MB, Nielsen AF, et al. A large-scale chemical modification screen identifies design rules to generate siRNAs with high activity, high stability and low toxicity. Nucleic Acids Res 2009 May; 37(9): 2867–81PubMedCrossRef
25.
go back to reference Behlke MA. Chemical modification of siRNAs for in vivo use. Oligonucleotides 2008 Dec; 18(4): 305–19PubMedCrossRef Behlke MA. Chemical modification of siRNAs for in vivo use. Oligonucleotides 2008 Dec; 18(4): 305–19PubMedCrossRef
26.
go back to reference Braasch DA, Jensen S, Liu Y, et al. RNA interference in mammalian cells by chemically-modified RNA. Biochemistry 2003 Jul 8; 42(26): 7967–75PubMedCrossRef Braasch DA, Jensen S, Liu Y, et al. RNA interference in mammalian cells by chemically-modified RNA. Biochemistry 2003 Jul 8; 42(26): 7967–75PubMedCrossRef
27.
go back to reference Chiu YL, Rana TM. siRNA function in RNAi: a chemical modification analysis. RNA 2003 Sep; 9(9): 1034–48PubMedCrossRef Chiu YL, Rana TM. siRNA function in RNAi: a chemical modification analysis. RNA 2003 Sep; 9(9): 1034–48PubMedCrossRef
28.
go back to reference Li ZY, Mao H, Kallick DA, et al. The effects of thiophosphate substitutions on native siRNA gene silencing. Biochem Biophys Res Commun 2005 Apr 15; 329(3): 1026–30PubMedCrossRef Li ZY, Mao H, Kallick DA, et al. The effects of thiophosphate substitutions on native siRNA gene silencing. Biochem Biophys Res Commun 2005 Apr 15; 329(3): 1026–30PubMedCrossRef
29.
go back to reference Choung S, Kim YJ, Kim S, et al. Chemical modification of siRNAs to improve serum stability without loss of efficacy. Biochem Biophys Res Commun 2006 Apr 14; 342(3): 919–27PubMedCrossRef Choung S, Kim YJ, Kim S, et al. Chemical modification of siRNAs to improve serum stability without loss of efficacy. Biochem Biophys Res Commun 2006 Apr 14; 342(3): 919–27PubMedCrossRef
30.
go back to reference Hall AH, Wan J, Shaughnessy EE, et al. RNA interference using boranophosphate siRNAs: structure-activity relationships. Nucleic Acids Res 2004 Nov 15; 32(20): 5991–6000PubMedCrossRef Hall AH, Wan J, Shaughnessy EE, et al. RNA interference using boranophosphate siRNAs: structure-activity relationships. Nucleic Acids Res 2004 Nov 15; 32(20): 5991–6000PubMedCrossRef
31.
go back to reference Hall AH, Wan J, Spesock A, et al. High potency silencing by single-stranded boranophosphate siRNA. Nucleic Acids Res 2006 May 22; 34(9): 2773–81PubMedCrossRef Hall AH, Wan J, Spesock A, et al. High potency silencing by single-stranded boranophosphate siRNA. Nucleic Acids Res 2006 May 22; 34(9): 2773–81PubMedCrossRef
32.
go back to reference Levin AA. A review of the issues in the pharmacokinetics and toxicology of phosphorothioate antisense oligonucleotides. Biochim Biophys Acta 1999 Dec 10; 1489(1): 69–84PubMedCrossRef Levin AA. A review of the issues in the pharmacokinetics and toxicology of phosphorothioate antisense oligonucleotides. Biochim Biophys Acta 1999 Dec 10; 1489(1): 69–84PubMedCrossRef
33.
go back to reference Braasch DA, Paroo Z, Constantinescu A, et al. Biodistribution of phosphodiester and phosphorothioate siRNA. Bioorg Med Chem Lett 2004 Mar 8; 14(5): 1139–43PubMedCrossRef Braasch DA, Paroo Z, Constantinescu A, et al. Biodistribution of phosphodiester and phosphorothioate siRNA. Bioorg Med Chem Lett 2004 Mar 8; 14(5): 1139–43PubMedCrossRef
34.
go back to reference Hoerter JA, Walter NG. Chemical modification resolves the asymmetry of siRNA strand degradation in human blood serum. RNA 2007 Nov; 13(11): 1887–93PubMedCrossRef Hoerter JA, Walter NG. Chemical modification resolves the asymmetry of siRNA strand degradation in human blood serum. RNA 2007 Nov; 13(11): 1887–93PubMedCrossRef
35.
go back to reference Czauderna F, Fechtner M, Dames S, et al. Structural variations and stabilising modifications of synthetic siRNAs in mammalian cells. Nucleic Acids Res 2003 Jun 1; 31(11): 2705–16PubMedCrossRef Czauderna F, Fechtner M, Dames S, et al. Structural variations and stabilising modifications of synthetic siRNAs in mammalian cells. Nucleic Acids Res 2003 Jun 1; 31(11): 2705–16PubMedCrossRef
36.
go back to reference Allerson CR, Sioufi N, Jarres R, et al. Fully 2′-modified oligonucleotide duplexes with improved in vitro potency and stability compared to unmodified small interfering RNA. J Med Chem 2005 Feb 24; 48(4): 901–4PubMedCrossRef Allerson CR, Sioufi N, Jarres R, et al. Fully 2′-modified oligonucleotide duplexes with improved in vitro potency and stability compared to unmodified small interfering RNA. J Med Chem 2005 Feb 24; 48(4): 901–4PubMedCrossRef
37.
go back to reference Prakash TP, Allerson CR, Dande P, et al. Positional effect of chemical modifications on short interference RNA activity in mammalian cells. J Med Chem 2005 Jun 30; 48(13): 4247–53PubMedCrossRef Prakash TP, Allerson CR, Dande P, et al. Positional effect of chemical modifications on short interference RNA activity in mammalian cells. J Med Chem 2005 Jun 30; 48(13): 4247–53PubMedCrossRef
38.
go back to reference Layzer JM, McCaffrey AP, Tanner AK, et al. In vivo activity of nuclease-resistant siRNAs. RNA 2004 May; 10(5): 766–71PubMedCrossRef Layzer JM, McCaffrey AP, Tanner AK, et al. In vivo activity of nuclease-resistant siRNAs. RNA 2004 May; 10(5): 766–71PubMedCrossRef
39.
go back to reference Morrissey DV, Blanchard K, Shaw L, et al. Activity of stabilized short interfering RNA in a mouse model of hepatitis B virus replication. Hepatology 2005 Jun; 41(6): 1349–56PubMedCrossRef Morrissey DV, Blanchard K, Shaw L, et al. Activity of stabilized short interfering RNA in a mouse model of hepatitis B virus replication. Hepatology 2005 Jun; 41(6): 1349–56PubMedCrossRef
40.
go back to reference Grünweller A, Wyszko E, Bieber B, et al. Comparison of different antisense strategies in mammalian cells using locked nucleic acids, 2′-O-methyl RNA, phosphorothioates and small interfering RNA. Nucleic Acids Res 2003 Jun 15; 31(12): 3185–93PubMedCrossRef Grünweller A, Wyszko E, Bieber B, et al. Comparison of different antisense strategies in mammalian cells using locked nucleic acids, 2′-O-methyl RNA, phosphorothioates and small interfering RNA. Nucleic Acids Res 2003 Jun 15; 31(12): 3185–93PubMedCrossRef
41.
go back to reference Elmén J, Thonberg H, Ljungberg K, et al. Locked nucleic acid (LNA) mediated improvements in siRNA stability and functionality. Nucleic Acids Res 2005 Jan 14; 33(1): 439–47PubMedCrossRef Elmén J, Thonberg H, Ljungberg K, et al. Locked nucleic acid (LNA) mediated improvements in siRNA stability and functionality. Nucleic Acids Res 2005 Jan 14; 33(1): 439–47PubMedCrossRef
42.
go back to reference Mook OR, Baas F, de Wissel MB, et al. Evaluation of locked nucleic acid-modified small interfering RNA in vitro and in vivo. Mol Cancer Ther 2007 Mar; 6(3): 833–43PubMedCrossRef Mook OR, Baas F, de Wissel MB, et al. Evaluation of locked nucleic acid-modified small interfering RNA in vitro and in vivo. Mol Cancer Ther 2007 Mar; 6(3): 833–43PubMedCrossRef
43.
go back to reference Dowler T, Bergeron D, Tedeschi AL, et al. Improvements in siRNA properties mediated by 20-deoxy-2′-fluoro-beta-D-arabinonucleic acid (FANA). Nucleic Acids Res 2006 Mar 22; 34(6): 1669–75PubMedCrossRef Dowler T, Bergeron D, Tedeschi AL, et al. Improvements in siRNA properties mediated by 20-deoxy-2′-fluoro-beta-D-arabinonucleic acid (FANA). Nucleic Acids Res 2006 Mar 22; 34(6): 1669–75PubMedCrossRef
44.
go back to reference Gao S, Dagnaes-Hansen F, Nielsen EJ, et al. The effect of chemical modification and nanoparticle formulation on stability and biodistribution of siRNA in mice. Mol Ther 2009 Jul; 17(7): 1225–33PubMedCrossRef Gao S, Dagnaes-Hansen F, Nielsen EJ, et al. The effect of chemical modification and nanoparticle formulation on stability and biodistribution of siRNA in mice. Mol Ther 2009 Jul; 17(7): 1225–33PubMedCrossRef
45.
go back to reference Liu F, Song Y, Liu D. Hydrodynamics-based transfection in animals by systemic administration of plasmid DNA. Gene Ther 1999 Jul; 6(7): 1258–66PubMedCrossRef Liu F, Song Y, Liu D. Hydrodynamics-based transfection in animals by systemic administration of plasmid DNA. Gene Ther 1999 Jul; 6(7): 1258–66PubMedCrossRef
46.
go back to reference Zhang G, Budker V, Wolff J. High levels of foreign gene expression in hepatocytes after tail vein injections of naked plasmid DNA. Hum Gene Ther 1999 Jul 1; 10(10): 1735–7PubMedCrossRef Zhang G, Budker V, Wolff J. High levels of foreign gene expression in hepatocytes after tail vein injections of naked plasmid DNA. Hum Gene Ther 1999 Jul 1; 10(10): 1735–7PubMedCrossRef
47.
go back to reference Suda T, Suda K, Liu D. Computer-assisted hydrodynamic gene delivery. Mol Ther 2008 Jun; 16(6): 1098–104PubMedCrossRef Suda T, Suda K, Liu D. Computer-assisted hydrodynamic gene delivery. Mol Ther 2008 Jun; 16(6): 1098–104PubMedCrossRef
48.
go back to reference Liu F, Huang L. Noninvasive gene delivery to the liver by mechanical massage. Hepatology 2002 Jun; 35(6): 1314–9PubMedCrossRef Liu F, Huang L. Noninvasive gene delivery to the liver by mechanical massage. Hepatology 2002 Jun; 35(6): 1314–9PubMedCrossRef
49.
go back to reference Liu F, Lei J, Vollmer R, et al. Mechanism of liver gene transfer by mechanical massage. Mol Ther 2004 Mar; 9(3): 452–7PubMedCrossRef Liu F, Lei J, Vollmer R, et al. Mechanism of liver gene transfer by mechanical massage. Mol Ther 2004 Mar; 9(3): 452–7PubMedCrossRef
50.
go back to reference Mukai H, Kawakami S, Hashida M. Renal press-mediated transfection method for plasmid DNA and siRNA to the kidney. Biochem Biophys Res Commun 2008 Aug 1; 372(3): 383–7PubMedCrossRef Mukai H, Kawakami S, Hashida M. Renal press-mediated transfection method for plasmid DNA and siRNA to the kidney. Biochem Biophys Res Commun 2008 Aug 1; 372(3): 383–7PubMedCrossRef
51.
go back to reference Mukai H, Kawakami S, Kamiya Y, et al. Tissue press-mediated transfection method: transfection to the spleen as a novel target, controlling the tissue press and evaluation of the pro-inflammatory cytokine production. Hum Gene Ther 2009 Oct; 20(10): 1157–67PubMedCrossRef Mukai H, Kawakami S, Kamiya Y, et al. Tissue press-mediated transfection method: transfection to the spleen as a novel target, controlling the tissue press and evaluation of the pro-inflammatory cytokine production. Hum Gene Ther 2009 Oct; 20(10): 1157–67PubMedCrossRef
52.
go back to reference Coster HG. A quantitative analysis of the voltage-current relationships of fixed charge membranes and the associated property of “punch-through”. Biophys J 1965 Sep; 5(5): 669–86PubMedCrossRef Coster HG. A quantitative analysis of the voltage-current relationships of fixed charge membranes and the associated property of “punch-through”. Biophys J 1965 Sep; 5(5): 669–86PubMedCrossRef
53.
go back to reference Kong XC, Barzaghi P, Ruegg MA. Inhibition of synapse assembly in mam-malian muscle in vivo by RNA interference. EMBO Rep 2004 Feb; 5(2): 183–8PubMedCrossRef Kong XC, Barzaghi P, Ruegg MA. Inhibition of synapse assembly in mam-malian muscle in vivo by RNA interference. EMBO Rep 2004 Feb; 5(2): 183–8PubMedCrossRef
54.
go back to reference Golzio M, Mazzolini L, Moller P, et al. Inhibition of gene expression in mice muscle by in vivo electrically mediated siRNA delivery. Gene Ther 2005 Feb; 12(3): 246–51PubMedCrossRef Golzio M, Mazzolini L, Moller P, et al. Inhibition of gene expression in mice muscle by in vivo electrically mediated siRNA delivery. Gene Ther 2005 Feb; 12(3): 246–51PubMedCrossRef
55.
go back to reference Matsuda T, Cepko CL. Electroporation and RNA interference in the rodent retina in vivo and in vitro. Proc Natl Acad Sci U S A 2004 Jan 6; 101(1): 16–22PubMedCrossRef Matsuda T, Cepko CL. Electroporation and RNA interference in the rodent retina in vivo and in vitro. Proc Natl Acad Sci U S A 2004 Jan 6; 101(1): 16–22PubMedCrossRef
56.
go back to reference Schiffelers RM, Xu J, Storm G, et al. Effects of treatment with small interfering RNA on joint inflammation in mice with collagen-induced arthritis. Arthritis Rheum 2005 Apr; 52(4): 1314–8PubMedCrossRef Schiffelers RM, Xu J, Storm G, et al. Effects of treatment with small interfering RNA on joint inflammation in mice with collagen-induced arthritis. Arthritis Rheum 2005 Apr; 52(4): 1314–8PubMedCrossRef
57.
go back to reference Takahashi Y, Nishikawa M, Kobayashi N, et al. Gene silencing in primary and metastatic tumors by small interfering RNA delivery in mice: quantitative analysis using melanoma cells expressing firefly and sea pansy luciferases. J Control Release 2005 Jul 20; 105(3): 332–43PubMedCrossRef Takahashi Y, Nishikawa M, Kobayashi N, et al. Gene silencing in primary and metastatic tumors by small interfering RNA delivery in mice: quantitative analysis using melanoma cells expressing firefly and sea pansy luciferases. J Control Release 2005 Jul 20; 105(3): 332–43PubMedCrossRef
58.
go back to reference Takei Y, Nemoto T, Mu P, et al. In vivo silencing of a molecular target by short interfering RNA electroporation: tumor vascularization correlates to delivery efficiency. Mol Cancer Ther 2008 Jan; 7(1): 211–21PubMedCrossRef Takei Y, Nemoto T, Mu P, et al. In vivo silencing of a molecular target by short interfering RNA electroporation: tumor vascularization correlates to delivery efficiency. Mol Cancer Ther 2008 Jan; 7(1): 211–21PubMedCrossRef
59.
go back to reference Guignet EG, Meyer T. Suspended-drop electroporation for high-throughput delivery of biomolecules into cells. Nat Methods 2008 May; 5(5): 393–5PubMedCrossRef Guignet EG, Meyer T. Suspended-drop electroporation for high-throughput delivery of biomolecules into cells. Nat Methods 2008 May; 5(5): 393–5PubMedCrossRef
60.
go back to reference Khvorova A, Reynolds A, Jayasena SD. Functional siRNAs and miRNAs exhibit strand bias. Cell 2003 Oct 17; 115(2): 209–16PubMedCrossRef Khvorova A, Reynolds A, Jayasena SD. Functional siRNAs and miRNAs exhibit strand bias. Cell 2003 Oct 17; 115(2): 209–16PubMedCrossRef
61.
go back to reference Schwarz DS, Hutvágner G, Du T, et al. Asymmetry in the assembly of the RNAi enzyme complex. Cell 2003 Oct 17; 115(2): 199–208PubMedCrossRef Schwarz DS, Hutvágner G, Du T, et al. Asymmetry in the assembly of the RNAi enzyme complex. Cell 2003 Oct 17; 115(2): 199–208PubMedCrossRef
62.
go back to reference Soutschek J, Akinc A, Bramlage B, et al. Therapeutic silencing of an endogenous gene by systemic administration of modified siRNAs. Nature 2004 Nov 11; 432(7014): 173–8PubMedCrossRef Soutschek J, Akinc A, Bramlage B, et al. Therapeutic silencing of an endogenous gene by systemic administration of modified siRNAs. Nature 2004 Nov 11; 432(7014): 173–8PubMedCrossRef
63.
go back to reference McNamara 2nd JO, Andrechek ER, Wang Y, et al. Cell type-specific delivery of siRNAs with aptamer-siRNA chimeras. Nat Biotechnol 2006 Aug; 24(8): 1005–15PubMedCrossRef McNamara 2nd JO, Andrechek ER, Wang Y, et al. Cell type-specific delivery of siRNAs with aptamer-siRNA chimeras. Nat Biotechnol 2006 Aug; 24(8): 1005–15PubMedCrossRef
64.
go back to reference Zhou J, Li H, Li S, et al. Novel dual inhibitory function aptamer-siRNA delivery system for HIV-1 therapy. Mol Ther 2008 Aug; 16(8): 1481–9PubMedCrossRef Zhou J, Li H, Li S, et al. Novel dual inhibitory function aptamer-siRNA delivery system for HIV-1 therapy. Mol Ther 2008 Aug; 16(8): 1481–9PubMedCrossRef
65.
go back to reference Muratovska A, Eccles MR. Conjugate for efficient delivery of short interfering RNA (siRNA) into mammalian cells. FEBS Lett 2004 Jan 30; 558(1–3): 63–8PubMedCrossRef Muratovska A, Eccles MR. Conjugate for efficient delivery of short interfering RNA (siRNA) into mammalian cells. FEBS Lett 2004 Jan 30; 558(1–3): 63–8PubMedCrossRef
66.
go back to reference Davidson TJ, Harel S, Arboleda VA, et al. Highly efficient small interfering RNA delivery to primary mammalian neurons induces MicroRNA-like effects before mRNA degradation. J Neurosci 2004 Nov 10; 24(45): 10040–6PubMedCrossRef Davidson TJ, Harel S, Arboleda VA, et al. Highly efficient small interfering RNA delivery to primary mammalian neurons induces MicroRNA-like effects before mRNA degradation. J Neurosci 2004 Nov 10; 24(45): 10040–6PubMedCrossRef
67.
go back to reference 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 Sep–Oct; 18(5): 1450–9PubMedCrossRef 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 Sep–Oct; 18(5): 1450–9PubMedCrossRef
68.
go back to reference Matsumura Y, Maeda H. A new concept for macromolecular therapeutics in cancer chemotherapy: mechanism of tumoritropic accumulation of proteins and the antitumor agent smancs. Cancer Res 1986 Dec; 46(12 Pt 1): 6387–92PubMed Matsumura Y, Maeda H. A new concept for macromolecular therapeutics in cancer chemotherapy: mechanism of tumoritropic accumulation of proteins and the antitumor agent smancs. Cancer Res 1986 Dec; 46(12 Pt 1): 6387–92PubMed
69.
go back to reference Sonoke S, Ueda T, Fujiwara K, et al. Tumor regression in mice by delivery of Bcl-2 small interfering RNA with pegylated cationic liposomes. Cancer Res 2008 Nov 1; 68(21): 8843–51PubMedCrossRef Sonoke S, Ueda T, Fujiwara K, et al. Tumor regression in mice by delivery of Bcl-2 small interfering RNA with pegylated cationic liposomes. Cancer Res 2008 Nov 1; 68(21): 8843–51PubMedCrossRef
70.
go back to reference Kawakami S, Higuchi Y, Hashida M. Nonviral approaches for targeted delivery of plasmid DNA and oligonucleotide. J Pharm Sci 2008 Feb; 97(2): 726–45PubMedCrossRef Kawakami S, Higuchi Y, Hashida M. Nonviral approaches for targeted delivery of plasmid DNA and oligonucleotide. J Pharm Sci 2008 Feb; 97(2): 726–45PubMedCrossRef
71.
go back to reference Peer D, Park EJ, Morishita Y, et al. Systemic leukocyte-directed siRNA delivery revealing cyclin D1 as an anti-inflammatory target. Science 2008 Feb 1; 319(5863): 627–30PubMedCrossRef Peer D, Park EJ, Morishita Y, et al. Systemic leukocyte-directed siRNA delivery revealing cyclin D1 as an anti-inflammatory target. Science 2008 Feb 1; 319(5863): 627–30PubMedCrossRef
72.
go back to reference Zheng X, Vladau C, Zhang X, et al. A novel in vivo siRNA delivery system specifically targeting dendritic cells and silencing CD40 genes for immunomodulation. Blood 2009 Mar 19; 113(12): 2646–54PubMedCrossRef Zheng X, Vladau C, Zhang X, et al. A novel in vivo siRNA delivery system specifically targeting dendritic cells and silencing CD40 genes for immunomodulation. Blood 2009 Mar 19; 113(12): 2646–54PubMedCrossRef
73.
go back to reference Sato Y, Murase K, Kato J, et al. Resolution of liver cirrhosis using vitamin A-coupled liposomes to deliver siRNA against a collagen-specific chaperone. Nat Biotechnol 2008 Apr; 26(4): 431–42PubMedCrossRef Sato Y, Murase K, Kato J, et al. Resolution of liver cirrhosis using vitamin A-coupled liposomes to deliver siRNA against a collagen-specific chaperone. Nat Biotechnol 2008 Apr; 26(4): 431–42PubMedCrossRef
74.
go back to reference Kim SI, Shin D, Choi TH, et al. Systemic and specific delivery of small interfering RNAs to the liver mediated by apolipoprotein A-I. Mol Ther 2007 Jun; 15(6): 1145–52PubMed Kim SI, Shin D, Choi TH, et al. Systemic and specific delivery of small interfering RNAs to the liver mediated by apolipoprotein A-I. Mol Ther 2007 Jun; 15(6): 1145–52PubMed
75.
go back to reference Sato A, Takagi M, Shimamoto A, et al. Small interfering RNA delivery to the liver by intravenous administration of galactosylated cationic liposomes in mice. Biomaterials 2007 Mar; 28(7): 1434–42PubMedCrossRef Sato A, Takagi M, Shimamoto A, et al. Small interfering RNA delivery to the liver by intravenous administration of galactosylated cationic liposomes in mice. Biomaterials 2007 Mar; 28(7): 1434–42PubMedCrossRef
76.
go back to reference Akinc A, Zumbuehl A, Goldberg M, et al. A combinatorial library of lipid-like materials for delivery of RNAi therapeutics. Nat Biotechnol 2008 May; 26(5): 561–9PubMedCrossRef Akinc A, Zumbuehl A, Goldberg M, et al. A combinatorial library of lipid-like materials for delivery of RNAi therapeutics. Nat Biotechnol 2008 May; 26(5): 561–9PubMedCrossRef
77.
go back to reference Akinc A, Goldberg M, Qin J, et al. Development of lipidoid-siRNA formulations for systemic delivery to the liver. Mol Ther 2009 May; 17(5): 872–9PubMedCrossRef Akinc A, Goldberg M, Qin J, et al. Development of lipidoid-siRNA formulations for systemic delivery to the liver. Mol Ther 2009 May; 17(5): 872–9PubMedCrossRef
78.
go back to reference Morrissey DV, Lockridge JA, Shaw L, et al. Potent and persistent in vivo anti-HBV activity of chemically modified siRNAs. Nat Biotechnol 2005 Aug; 23(8): 1002–7PubMedCrossRef Morrissey DV, Lockridge JA, Shaw L, et al. Potent and persistent in vivo anti-HBV activity of chemically modified siRNAs. Nat Biotechnol 2005 Aug; 23(8): 1002–7PubMedCrossRef
79.
go back to reference Judge AD, Robbins M, Tavakoli I, et al. Confirming the RNAi-mediated mechanism of action of siRNA-based cancer therapeutics in mice. J Clin Invest 2009 Mar; 119(3): 661–73PubMedCrossRef Judge AD, Robbins M, Tavakoli I, et al. Confirming the RNAi-mediated mechanism of action of siRNA-based cancer therapeutics in mice. J Clin Invest 2009 Mar; 119(3): 661–73PubMedCrossRef
80.
go back to reference Kinoshita M, Hynynen K. A novel method for the intracellular delivery of siRNA using microbubble-enhanced focused ultrasound. Biochem Biophys Res Commun 2005 Sep 23; 335(2): 393–9PubMedCrossRef Kinoshita M, Hynynen K. A novel method for the intracellular delivery of siRNA using microbubble-enhanced focused ultrasound. Biochem Biophys Res Commun 2005 Sep 23; 335(2): 393–9PubMedCrossRef
81.
go back to reference Kinoshita M, Hynynen K. Key factors that affect sonoporation efficiency in in vitro settings: the importance of standing wave in sonoporation. Biochem Biophys Res Commun 2007 Aug 10; 359(4): 860–5PubMedCrossRef Kinoshita M, Hynynen K. Key factors that affect sonoporation efficiency in in vitro settings: the importance of standing wave in sonoporation. Biochem Biophys Res Commun 2007 Aug 10; 359(4): 860–5PubMedCrossRef
82.
go back to reference Otani K, Yamahara K, Ohnishi S, et al. Nonviral delivery of siRNA into mesenchymal stem cells by a combination of ultrasound and microbubbles. J Control Release 2009 Jan 19; 133(2): 146–53PubMedCrossRef Otani K, Yamahara K, Ohnishi S, et al. Nonviral delivery of siRNA into mesenchymal stem cells by a combination of ultrasound and microbubbles. J Control Release 2009 Jan 19; 133(2): 146–53PubMedCrossRef
83.
go back to reference Negishi Y, Endo Y, Fukuyama T, et al. Delivery of siRNA into the cytoplasm by liposomal bubbles and ultrasound. J Control Release 2008 Dec 8; 132(2): 124–30PubMedCrossRef Negishi Y, Endo Y, Fukuyama T, et al. Delivery of siRNA into the cytoplasm by liposomal bubbles and ultrasound. J Control Release 2008 Dec 8; 132(2): 124–30PubMedCrossRef
84.
go back to reference MacDiarmid JA, Mugridge NB, Weiss JC, et al. Bacterially derived 400 nm particles for encapsulation and cancer cell targeting of chemotherapeutics. Cancer Cell 2007 May; 11(5): 431–45PubMedCrossRef MacDiarmid JA, Mugridge NB, Weiss JC, et al. Bacterially derived 400 nm particles for encapsulation and cancer cell targeting of chemotherapeutics. Cancer Cell 2007 May; 11(5): 431–45PubMedCrossRef
85.
go back to reference MacDiarmid JA, Amaro-Mugridge NB, Madrid-Weiss J, et al. Sequential treatment of drug-resistant tumors with targeted minicells containing siRNA or a cytotoxic drug. Nat Biotechnol 2009 Jul; 27(7): 643–51PubMedCrossRef MacDiarmid JA, Amaro-Mugridge NB, Madrid-Weiss J, et al. Sequential treatment of drug-resistant tumors with targeted minicells containing siRNA or a cytotoxic drug. Nat Biotechnol 2009 Jul; 27(7): 643–51PubMedCrossRef
86.
go back to reference Minakuchi Y, Takeshita F, Kosaka N, et al. Atelocollagen-mediated synthetic small interfering RNA delivery for effective gene silencing in vitro and in vivo. Nucleic Acids Res 2004 Jul 22; 32(13): e109PubMedCrossRef Minakuchi Y, Takeshita F, Kosaka N, et al. Atelocollagen-mediated synthetic small interfering RNA delivery for effective gene silencing in vitro and in vivo. Nucleic Acids Res 2004 Jul 22; 32(13): e109PubMedCrossRef
87.
go back to reference Takeshita F, Minakuchi Y, Nagahara S, et al. Efficient delivery of small interfering RNA to bone-metastatic tumors by using atelocollagen in vivo. Proc Natl Acad Sci U S A 2005 Aug 23; 102(34): 12177–82PubMedCrossRef Takeshita F, Minakuchi Y, Nagahara S, et al. Efficient delivery of small interfering RNA to bone-metastatic tumors by using atelocollagen in vivo. Proc Natl Acad Sci U S A 2005 Aug 23; 102(34): 12177–82PubMedCrossRef
88.
go back to reference Kinouchi N, Ohsawa Y, Ishimaru N, et al. Atelocollagen-mediated local and systemic applications of myostatin-targeting siRNA increase skeletal muscle mass. Gene Ther 2008 Aug; 15(15): 1126–30PubMedCrossRef Kinouchi N, Ohsawa Y, Ishimaru N, et al. Atelocollagen-mediated local and systemic applications of myostatin-targeting siRNA increase skeletal muscle mass. Gene Ther 2008 Aug; 15(15): 1126–30PubMedCrossRef
89.
go back to reference Howard KA, Paludan SR, Behlke MA, et al. Chitosan/siRNA nanoparticle-mediated TNF-α knockdown in peritoneal macrophages for anti-inflammatory treatment in amurine arthritis model. Mol Ther 2009 Jan; 17(1): 162–8PubMedCrossRef Howard KA, Paludan SR, Behlke MA, et al. Chitosan/siRNA nanoparticle-mediated TNF-α knockdown in peritoneal macrophages for anti-inflammatory treatment in amurine arthritis model. Mol Ther 2009 Jan; 17(1): 162–8PubMedCrossRef
90.
go back to reference Kawakami S, Hashida M. Targeted delivery systems of small interfering RNA by systemic administration. Drug Metab Pharmacokinet 2007 Jun; 22(3): 142–51PubMedCrossRef Kawakami S, Hashida M. Targeted delivery systems of small interfering RNA by systemic administration. Drug Metab Pharmacokinet 2007 Jun; 22(3): 142–51PubMedCrossRef
91.
go back to reference Akhtar S, Benter I. Toxicogenomics of non-viral drug delivery systems for RNAi: potential impact on siRNA-mediated gene silencing activity and specificity. Adv Drug Deliv Rev 2007 Mar 30; 59(2–3): 164–82PubMedCrossRef Akhtar S, Benter I. Toxicogenomics of non-viral drug delivery systems for RNAi: potential impact on siRNA-mediated gene silencing activity and specificity. Adv Drug Deliv Rev 2007 Mar 30; 59(2–3): 164–82PubMedCrossRef
92.
93.
go back to reference Akinc A, Thomas M, Klibanov AM, et al. Exploring polyethylenimine-mediated DNA transfection and the proton sponge hypothesis. J Gene Med 2005 May; 7(5): 657–63PubMedCrossRef Akinc A, Thomas M, Klibanov AM, et al. Exploring polyethylenimine-mediated DNA transfection and the proton sponge hypothesis. J Gene Med 2005 May; 7(5): 657–63PubMedCrossRef
94.
go back to reference Saito Y, Higuchi Y, Kawakami S, et al. Immunostimulatory characteristics induced by linear polyethyleneimine/plasmid DNA complexes in cultured macrophages. Hum Gene Ther 2009 Feb; 20(2): 137–45PubMedCrossRef Saito Y, Higuchi Y, Kawakami S, et al. Immunostimulatory characteristics induced by linear polyethyleneimine/plasmid DNA complexes in cultured macrophages. Hum Gene Ther 2009 Feb; 20(2): 137–45PubMedCrossRef
95.
go back to reference Kim YH, Park JH, Lee M, et al. Polyethylenimine with acid-labile linkages as a biodegradable gene carrier. J Control Release 2005 Mar 2; 103(1): 209–19PubMedCrossRef Kim YH, Park JH, Lee M, et al. Polyethylenimine with acid-labile linkages as a biodegradable gene carrier. J Control Release 2005 Mar 2; 103(1): 209–19PubMedCrossRef
96.
go back to reference Woodrow KA, Cu Y, Booth CJ, et al. Intravaginal gene silencing using biodegradable polymer nanoparticles densely loaded with small-interfering RNA. Nat Mater 2009 Jun; 8(6): 526–33PubMedCrossRef Woodrow KA, Cu Y, Booth CJ, et al. Intravaginal gene silencing using biodegradable polymer nanoparticles densely loaded with small-interfering RNA. Nat Mater 2009 Jun; 8(6): 526–33PubMedCrossRef
97.
go back to reference Vandenbroucke RE, De Geest BG, Bonné S, et al. Prolonged gene silencing in hepatoma cells and primary hepatocytes after small interfering RNA delivery with biodegradable poly(beta-amino esters). J Gene Med 2008 Jul; 10(7): 783–94PubMedCrossRef Vandenbroucke RE, De Geest BG, Bonné S, et al. Prolonged gene silencing in hepatoma cells and primary hepatocytes after small interfering RNA delivery with biodegradable poly(beta-amino esters). J Gene Med 2008 Jul; 10(7): 783–94PubMedCrossRef
98.
go back to reference Aouadi M, Tesz GJ, Nicoloro SM, et al. Orally delivered siRNA targeting macrophage Map4k4 suppresses systemic inflammation. Nature 2009 Apr 30; 458(7242): 1180–4PubMedCrossRef Aouadi M, Tesz GJ, Nicoloro SM, et al. Orally delivered siRNA targeting macrophage Map4k4 suppresses systemic inflammation. Nature 2009 Apr 30; 458(7242): 1180–4PubMedCrossRef
99.
go back to reference 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 May–Jun; 6(3): 659–68PubMedCrossRef 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 May–Jun; 6(3): 659–68PubMedCrossRef
100.
go back to reference Eguchi A, Meade BR, Chang YC, et al. Efficient siRNA delivery into primary cells by a peptide transduction domain-dsRNA binding domain fusion protein. Nat Biotechnol 2009 Jun; 27(6): 567–71PubMedCrossRef Eguchi A, Meade BR, Chang YC, et al. Efficient siRNA delivery into primary cells by a peptide transduction domain-dsRNA binding domain fusion protein. Nat Biotechnol 2009 Jun; 27(6): 567–71PubMedCrossRef
101.
go back to reference 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 Jan; 17(1): 95–103PubMedCrossRef 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 Jan; 17(1): 95–103PubMedCrossRef
102.
go back to reference Crombez L, Morris MC, Dufort S, et al. Targeting cyclin B1 through peptide-based delivery of siRNA prevents tumour growth. Nucleic Acids Res 2009 Aug; 37(14): 4559–69PubMedCrossRef Crombez L, Morris MC, Dufort S, et al. Targeting cyclin B1 through peptide-based delivery of siRNA prevents tumour growth. Nucleic Acids Res 2009 Aug; 37(14): 4559–69PubMedCrossRef
103.
go back to reference Kumar P, Wu H, McBride JL, et al. Transvascular delivery of small interfering RNA to the central nervous system. Nature 2007 Jul 5; 448(7149): 39–43PubMedCrossRef Kumar P, Wu H, McBride JL, et al. Transvascular delivery of small interfering RNA to the central nervous system. Nature 2007 Jul 5; 448(7149): 39–43PubMedCrossRef
104.
go back to reference Kumar P, Ban HS, Kim SS, et al. T cell-specific siRNA delivery suppresses HIV-1 infection in humanized mice. Cell 2008 Aug 22; 134(4): 577–86PubMedCrossRef Kumar P, Ban HS, Kim SS, et al. T cell-specific siRNA delivery suppresses HIV-1 infection in humanized mice. Cell 2008 Aug 22; 134(4): 577–86PubMedCrossRef
105.
go back to reference Kam NW, Liu Z, Dai H. Functionalization of carbon nanotubes via cleavable disulfide bonds for efficient intracellular delivery of siRNA and potent gene silencing. J Am Chem Soc 2005 Sep 14; 127(36): 12492–3PubMedCrossRef Kam NW, Liu Z, Dai H. Functionalization of carbon nanotubes via cleavable disulfide bonds for efficient intracellular delivery of siRNA and potent gene silencing. J Am Chem Soc 2005 Sep 14; 127(36): 12492–3PubMedCrossRef
106.
go back to reference Liu Z, Winters M, Holodniy M, et al. siRNA delivery into human T cells and primary cells with carbon-nanotube transporters. Angew Chem Int Ed Engl 2007; 46(12): 2023–7PubMedCrossRef Liu Z, Winters M, Holodniy M, et al. siRNA delivery into human T cells and primary cells with carbon-nanotube transporters. Angew Chem Int Ed Engl 2007; 46(12): 2023–7PubMedCrossRef
107.
go back to reference Wang X, Ren J, Qu X. Targeted RNA interference of cyclin A2 mediated by functionalized single-walled carbon nanotubes induces proliferation arrest and apoptosis in chronic myelogenous leukemia K562 cells. Chem Med Chem 2008 Jun; 3(6): 940–5PubMed Wang X, Ren J, Qu X. Targeted RNA interference of cyclin A2 mediated by functionalized single-walled carbon nanotubes induces proliferation arrest and apoptosis in chronic myelogenous leukemia K562 cells. Chem Med Chem 2008 Jun; 3(6): 940–5PubMed
108.
go back to reference Zhang Z, Yang X, Zhang Y, et al. Delivery of telomerase reverse transcriptase small interfering RNA in complex with positively charged single-walled carbon nanotubes suppresses tumor growth. Clin Cancer Res 2006 Aug 15 Zhang Z, Yang X, Zhang Y, et al. Delivery of telomerase reverse transcriptase small interfering RNA in complex with positively charged single-walled carbon nanotubes suppresses tumor growth. Clin Cancer Res 2006 Aug 15
109.
go back to reference Podesta JE, Al-Jamal KT, Herrero MA, et al. Antitumor activity and prolonged survival by carbon-nanotube-mediated therapeutic siRNA silencing in a human lung xenograft model. Small 2009 May; 5(10): 1176–85PubMedCrossRef Podesta JE, Al-Jamal KT, Herrero MA, et al. Antitumor activity and prolonged survival by carbon-nanotube-mediated therapeutic siRNA silencing in a human lung xenograft model. Small 2009 May; 5(10): 1176–85PubMedCrossRef
110.
go back to reference Herrero MA, Toma FM, Al-Jamal KT, et al. Synthesis and characterization of a carbon nanotube-dendron series for efficient siRNA delivery. J Am Chem Soc 2009 Jul 22; 131(28): 9843–8PubMedCrossRef Herrero MA, Toma FM, Al-Jamal KT, et al. Synthesis and characterization of a carbon nanotube-dendron series for efficient siRNA delivery. J Am Chem Soc 2009 Jul 22; 131(28): 9843–8PubMedCrossRef
111.
go back to reference Bonoiu AC, Mahajan SD, Ding H, et al. Nanotechnology approach for drug addiction therapy: gene silencing using delivery of gold nanorod-siRNA nanoplex in dopaminergic neurons. Proc Natl Acad Sci U S A 2009 Apr 7; 106(14): 5546–50PubMedCrossRef Bonoiu AC, Mahajan SD, Ding H, et al. Nanotechnology approach for drug addiction therapy: gene silencing using delivery of gold nanorod-siRNA nanoplex in dopaminergic neurons. Proc Natl Acad Sci U S A 2009 Apr 7; 106(14): 5546–50PubMedCrossRef
112.
go back to reference Elbakry A, Zaky A, Liebl R, et al. Layer-by-layer assembled gold nanoparticles for siRNA delivery. Nano Lett 2009 May; 9(5): 2059–64PubMedCrossRef Elbakry A, Zaky A, Liebl R, et al. Layer-by-layer assembled gold nanoparticles for siRNA delivery. Nano Lett 2009 May; 9(5): 2059–64PubMedCrossRef
113.
go back to reference Giljohann DA, Seferos DS, Prigodich AE, et al. Gene regulation with polyvalent siRNA-nanoparticle conjugates. J Am Chem Soc 2009 Feb 18; 131(6): 2072–3PubMedCrossRef Giljohann DA, Seferos DS, Prigodich AE, et al. Gene regulation with polyvalent siRNA-nanoparticle conjugates. J Am Chem Soc 2009 Feb 18; 131(6): 2072–3PubMedCrossRef
114.
go back to reference Lee JS, Green JJ, Love KT, et al. Gold, poly(beta-amino ester) nanoparticles for small interfering RNA delivery. Nano Lett 2009 Jun; 9(6): 2402–6PubMedCrossRef Lee JS, Green JJ, Love KT, et al. Gold, poly(beta-amino ester) nanoparticles for small interfering RNA delivery. Nano Lett 2009 Jun; 9(6): 2402–6PubMedCrossRef
115.
go back to reference Braun GB, Pallaoro A, Wu G, et al. Laser-activated gene silencing via gold nanoshell-siRNA conjugates. ACS Nano. Epub 2009 Jun 15 Braun GB, Pallaoro A, Wu G, et al. Laser-activated gene silencing via gold nanoshell-siRNA conjugates. ACS Nano. Epub 2009 Jun 15
Metadata
Title
Strategies for In Vivo Delivery of siRNAs
Recent Progress
Authors
Dr Yuriko Higuchi
Shigeru Kawakami
Mitsuru Hashida
Publication date
01-06-2010
Publisher
Springer International Publishing
Published in
BioDrugs / Issue 3/2010
Print ISSN: 1173-8804
Electronic ISSN: 1179-190X
DOI
https://doi.org/10.2165/11534450-000000000-00000

Other articles of this Issue 3/2010

BioDrugs 3/2010 Go to the issue

Review Article

Dasatinib