Targeted delivery of RNAi therapeutics for cancer therapy.

Since the discovery of RNA interference (RNAi) [1], siRNA) has become the focus of attention for pharmaceutical development. siRNA is more potent than antisense oligo­ nucleotides owing to the endogenous mecha­ nism of action catalyzed by the RNA­induced silencing complex [2]. Many oncogenic targets involved in survival, specifically antiapoptosis, angiogenesis and drug resistance, have been tested for siRNA­mediated cancer therapy. However, siRNA can only become an anti cancer agent when it is specifically and effectively deliv­ ered to the target cells in vivo [3]. An ideal siRNA delivery vehicle for cancer therapy must be able to evade the reticuloendothelial system to be effectively taken up by the tumor cells and to escape from the endosome after endocytosis. The existence of a leaky vasculature in most of the solid tumors supports the enhanced per­ meability and retention effect for nanoparticle (NP)­mediated delivery of anticancer agents, for example the successful Doxil formulation of doxorubicin. A well­designed NP of less than 200 nm in diameter is suitable for siRNA delivery to the tumor. NPs composed of differ­ ent materials have been developed for siRNA delivery. Two major classes of biomaterials have been employed for siRNA delivery. They will be discussed separately in this article.

[1]  Vladimir P Torchilin,et al.  Effective stabilization and delivery of siRNA: reversible siRNA-phospholipid conjugate in nanosized mixed polymeric micelles. , 2010, Bioconjugate chemistry.

[2]  K. G. Rajeev,et al.  Targeted Delivery of RNAi Therapeutics With Endogenous and Exogenous Ligand-Based Mechanisms. , 2010, Molecular therapy : the journal of the American Society of Gene Therapy.

[3]  Jeong Won Lee,et al.  Targeted Gene Silencing Using RGD-Labeled Chitosan Nanoparticles , 2010, Clinical Cancer Research.

[4]  Yu-cheng Tseng,et al.  Biodegradable calcium phosphate nanoparticle with lipid coating for systemic siRNA delivery. , 2010, Journal of controlled release : official journal of the Controlled Release Society.

[5]  Mark E. Davis,et al.  Evidence of RNAi in humans from systemically administered siRNA via targeted nanoparticles , 2010, Nature.

[6]  K. G. Rajeev,et al.  Rational design of cationic lipids for siRNA delivery , 2010, Nature Biotechnology.

[7]  Kathryn A. Whitehead,et al.  Lipid-like materials for low-dose, in vivo gene silencing , 2010, Proceedings of the National Academy of Sciences.

[8]  Leaf Huang,et al.  Nanoparticles evading the reticuloendothelial system: role of the supported bilayer. , 2009, Biochimica et biophysica acta.

[9]  Michael S. Goldberg,et al.  Development of lipidoid-siRNA formulations for systemic delivery to the liver. , 2009, Molecular therapy : the journal of the American Society of Gene Therapy.

[10]  A. Judge,et al.  Confirming the RNAi-mediated mechanism of action of siRNA-based cancer therapeutics in mice. , 2009, The Journal of clinical investigation.

[11]  Daniel G. Anderson,et al.  Knocking down barriers: advances in siRNA delivery , 2009, Nature Reviews Drug Discovery.

[12]  Leaf Huang,et al.  An efficient and low immunostimulatory nanoparticle formulation for systemic siRNA delivery to the tumor. , 2008, Journal of controlled release : official journal of the Controlled Release Society.

[13]  Leaf Huang,et al.  Reactive oxygen species play a central role in the activity of cationic liposome based cancer vaccine. , 2008, Journal of controlled release : official journal of the Controlled Release Society.

[14]  Leaf Huang,et al.  Efficient oncogene silencing and metastasis inhibition via systemic delivery of siRNA. , 2008, Molecular therapy : the journal of the American Society of Gene Therapy.

[15]  Robert Langer,et al.  A combinatorial library of lipid-like materials for delivery of RNAi therapeutics , 2008, Nature Biotechnology.

[16]  Darren H. Wakefield,et al.  Dynamic PolyConjugates for targeted in vivo delivery of siRNA to hepatocytes , 2007, Proceedings of the National Academy of Sciences.

[17]  Soo Hyun Lee,et al.  PEG conjugated VEGF siRNA for anti-angiogenic gene therapy. , 2006, Journal of controlled release : official journal of the Controlled Release Society.

[18]  Anastasia Khvorova,et al.  Off-target effects by siRNA can induce toxic phenotype. , 2006, RNA.

[19]  L. Lim,et al.  Position-specific chemical modification of siRNAs reduces "off-target" transcript silencing. , 2006, RNA.

[20]  Matthias John,et al.  RNAi-mediated gene silencing in non-human primates , 2006, Nature.

[21]  Mark E. Davis,et al.  Sequence-specific knockdown of EWS-FLI1 by targeted, nonviral delivery of small interfering RNA inhibits tumor growth in a murine model of metastatic Ewing's sarcoma. , 2005, Cancer research.

[22]  T. Ochiya,et al.  Efficient delivery of small interfering RNA to bone-metastatic tumors by using atelocollagen in vivo. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[23]  Anil K Sood,et al.  Therapeutic EphA2 gene targeting in vivo using neutral liposomal small interfering RNA delivery. , 2005, Cancer research.

[24]  Judy Lieberman,et al.  Antibody mediated in vivo delivery of small interfering RNAs via cell-surface receptors , 2005, Nature Biotechnology.

[25]  L. Jeffs,et al.  A Scalable, Extrusion-Free Method for Efficient Liposomal Encapsulation of Plasmid DNA , 2005, Pharmaceutical Research.

[26]  J. M. Harris,et al.  Effect of pegylation on pharmaceuticals , 2003, Nature Reviews Drug Discovery.

[27]  P. Cullis,et al.  On the mechanism whereby cationic lipids promote intracellular delivery of polynucleic acids , 2001, Gene Therapy.

[28]  P. Sharp,et al.  RNAi Double-Stranded RNA Directs the ATP-Dependent Cleavage of mRNA at 21 to 23 Nucleotide Intervals , 2000, Cell.

[29]  Sanchita Bhattacharya,et al.  Characterization of cationic lipid-protamine–DNA (LPD) complexes for intravenous gene delivery , 1998, Gene Therapy.

[30]  A. Fire,et al.  Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans , 1998, Nature.

[31]  L. Huang,et al.  In vivo gene transfer via intravenous administration of cationic lipid–protamine–DNA (LPD) complexes , 1997, Gene Therapy.

[32]  F. Szoka,et al.  Mechanism of DNA release from cationic liposome/DNA complexes used in cell transfection. , 1996, Biochemistry.

[33]  Yun Chen,et al.  Tumor-targeted delivery of siRNA by self-assembled nanoparticles. , 2008, Molecular therapy : the journal of the American Society of Gene Therapy.