301
|
Baigude H, Su J, McCarroll J, Rana TM. In Vivo Delivery of RNAi by Reducible Interfering Nanoparticles (iNOPs). ACS Med Chem Lett 2013; 4:720-723. [PMID: 24319542 PMCID: PMC3850243 DOI: 10.1021/ml4001003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Accepted: 06/18/2013] [Indexed: 11/30/2022] Open
Abstract
![]()
RNA interference (RNAi) has considerable
potential as a therapeutic
strategy, but the development of efficient in vivo RNA delivery methods
remains challenging. To this end, we designed and synthesized chemically
modified interfering nanoparticles (iNOPs) composed of functionalized
poly-l-lysine dendrimers modified with reducible spacers
to facilitate release of small interfering RNAs (siRNAs) in vivo.
We show that the novel siRNA–iNOP complexes mediate efficient
gene-specific RNAi in cultured cells and in mice, where they display
enhanced tissue-targeting capabilities. At a clinically feasible dose
of 1 mg kg–1, apolipoprotein B (apoB) siRNA–iNOP
complexes achieved ∼40–45% reduction of liver apoB mRNA
and plasma apoB protein levels within 48 h of administration to mice,
without apparent toxicity. Collectively, these findings demonstrate
that siRNA delivery by the modified reducible iNOPs can provide a
clinically significant and potentially tissue-specific new approach
for RNAi therapy.
Collapse
Affiliation(s)
- Huricha Baigude
- Program for RNA Biology, Sanford−Burnham Medical Research Institute,
10901 North Torrey Pines Road, La Jolla, California 92037, United
States
- Department of Biochemistry
and
Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
| | - Jie Su
- Department of Biochemistry
and
Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
| | - Joshua McCarroll
- Department of Biochemistry
and
Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
| | - Tariq M. Rana
- Program for RNA Biology, Sanford−Burnham Medical Research Institute,
10901 North Torrey Pines Road, La Jolla, California 92037, United
States
- Department of Biochemistry
and
Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
- Department of Pediatrics and Biomedical
Sciences Graduate Program, University of California, San Diego, California 92093, United States
| |
Collapse
|
302
|
Shu Y, Shu D, Haque F, Guo P. Fabrication of pRNA nanoparticles to deliver therapeutic RNAs and bioactive compounds into tumor cells. Nat Protoc 2013; 8:1635-59. [PMID: 23928498 DOI: 10.1038/nprot.2013.097] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
RNA nanotechnology is a term that refers to the design, fabrication and use of nanoparticles that are mainly composed of RNAs via bottom-up self-assembly. The packaging RNA (pRNA) of the bacteriophage phi29 DNA packaging motor has been developed into a nanodelivery platform. This protocol describes the synthesis, assembly and functionalization of pRNA nanoparticles on the basis of three 'toolkits' derived from pRNA structural features: interlocking loops for hand-in-hand interactions, palindrome sequences for foot-to-foot interactions and an RNA three-way junction for branch extension. siRNAs, ribozymes, aptamers, chemical ligands, fluorophores and other functionalities can also be fused to the pRNA before the assembly of the nanoparticles, so as to ensure the production of homogeneous nanoparticles and the retention of appropriate folding and function of the incorporated modules. The resulting self-assembled multivalent pRNA nanoparticles are thermodynamically and chemically stable, and they remain intact at ultralow concentrations. Gene-silencing effects are progressively enhanced with increasing numbers of siRNAs in each pRNA nanoparticle. Systemic injection of the pRNA nanoparticles into xenograft-bearing mice has revealed strong binding to tumors without accumulation in vital organs or tissues. The pRNA-based nanodelivery scaffold paves a new way for nanotechnological application of pRNA-based nanoparticles for disease detection and treatment. The time required for completing one round of this protocol is 3-4 weeks when including in vitro functional assays, or 2-3 months when including in vivo studies.
Collapse
Affiliation(s)
- Yi Shu
- Nanobiotechnology Center, Markey Cancer Center, Lexington, Kentucky, USA
| | | | | | | |
Collapse
|
303
|
Multiparametric approach for the evaluation of lipid nanoparticles for siRNA delivery. Proc Natl Acad Sci U S A 2013; 110:12881-6. [PMID: 23882076 DOI: 10.1073/pnas.1306529110] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Nanoparticle-mediated siRNA delivery is a complex process that requires transport across numerous extracellular and intracellular barriers. As such, the development of nanoparticles for efficient delivery would benefit from an understanding of how parameters associated with these barriers relate to the physicochemical properties of nanoparticles. Here, we use a multiparametric approach for the evaluation of lipid nanoparticles (LNPs) to identify relationships between structure, biological function, and biological activity. Our results indicate that evaluation of multiple parameters associated with barriers to delivery such as siRNA entrapment, pKa, LNP stability, and cell uptake as a collective may serve as a useful prescreening tool for the advancement of LNPs in vivo. This multiparametric approach complements the use of in vitro efficacy results alone for prescreening and improves in vitro-in vivo translation by minimizing false negatives. For the LNPs used in this work, the evaluation of multiple parameters enabled the identification of LNP pKa as one of the key determinants of LNP function and activity both in vitro and in vivo. It is anticipated that this type of analysis can aid in the identification of meaningful structure-function-activity relationships, improve the in vitro screening process of nanoparticles before in vivo use, and facilitate the future design of potent nanocarriers.
Collapse
|
304
|
Chen J, Zhao M, Feng F, Sizovs A, Wang J. Tunable Thioesters as “Reduction” Responsive Functionality for Traceless Reversible Protein PEGylation. J Am Chem Soc 2013; 135:10938-41. [DOI: 10.1021/ja405261u] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
| | | | | | | | - Jin Wang
- Department of Bioengineering, Rice University, Houston, Texas 77030, United States
| |
Collapse
|
305
|
Hu Y, Haynes MT, Wang Y, Liu F, Huang L. A highly efficient synthetic vector: nonhydrodynamic delivery of DNA to hepatocyte nuclei in vivo. ACS NANO 2013; 7:5376-84. [PMID: 23647441 PMCID: PMC3718078 DOI: 10.1021/nn4012384] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Multifunctional membrane-core nanoparticles, composed of calcium phosphate cores, arginine-rich peptides, cationic and PEGylated lipid membranes, and galactose targeting ligands, have been developed as synthetic vectors for efficient nuclear delivery of plasmid DNA and subsequent gene expression in hepatocytes in vivo. Targeted particles exhibited rapid and extensive hepatic accumulation and were predominantly internalized by hepatocytes, while the inclusion of such peptides in LCP was sufficient to elicit high degrees of nuclear translocation of plasmid DNA. Monocyclic CR8C significantly enhanced in vivo gene expression over 10-fold more than linear CR8C, likely due to a release-favoring mechanism of the DNA/peptide complex. Though 100-fold lower in activity than that achieved via hydrodynamic injection, this formulation presents as a much less invasive alternative. To our knowledge, this is the most effective synthetic vector for liver gene transfer.
Collapse
Affiliation(s)
- Yunxia Hu
- The Center for Nanotechnology in Drug Delivery, Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill NC 27599, USA
- Correspondence: Yunxia Hu,
| | - Matthew T. Haynes
- The Center for Nanotechnology in Drug Delivery, Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill NC 27599, USA
| | - Yuhua Wang
- The Center for Nanotechnology in Drug Delivery, Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill NC 27599, USA
| | - Feng Liu
- The Center for Nanotechnology in Drug Delivery, Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill NC 27599, USA
| | - Leaf Huang
- The Center for Nanotechnology in Drug Delivery, Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill NC 27599, USA
- Correspondence: Leaf Huang,
| |
Collapse
|
306
|
Stegh AH. Toward personalized cancer nanomedicine - past, present, and future. Integr Biol (Camb) 2013; 5:48-65. [PMID: 22858688 DOI: 10.1039/c2ib20104f] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Tumors are composed of highly proliferate, migratory, invasive, and therapy-evading cells. These characteristics are conferred by an enormously complex landscape of genomic, (epi-)genetic, and proteomic aberrations. Recent efforts to comprehensively catalogue these reversible and irreversible modifications have began to identify molecular mechanisms that contribute to cancer pathophysiology, serve as novel therapeutic targets, and may constitute biomarkers for early diagnosis and prediction of therapy responses. With constantly evolving technologies that will ultimately enable a complete survey of cancer genomes, the challenges for discovery cancer science and drug development are daunting. Bioinformatic and functional studies must differentiate cancer-driving and -contributing mutations from mere bystanders or 'noise', and have to delineate their molecular mechanisms of action as a function of collaborating oncogenic and tumor suppressive signatures. In addition, the translation of these genomic discoveries into meaningful clinical endpoints requires the development of co-extinction strategies to therapeutically target multiple cancer genes, to robustly deliver therapeutics to tumor sites, and to enable widespread dissemination of therapies within tumor tissue. In this perspective, I will describe the most current paradigms to study and validate cancer gene function. I will highlight advances in the area of nanotechnology, in particular, the development of RNA interference (RNAi)-based platforms to more effectively deliver therapeutic agents to tumor sites, and to modulate critical cancer genes that are difficult to target using conventional small-molecule- or antibody-based approaches. I will conclude with an outlook on the deluge of challenges that genomic and bioengineering sciences must overcome to make the long-awaited era of personalized nano-medicine a clinical reality for cancer patients.
Collapse
Affiliation(s)
- Alexander H Stegh
- Ken and Ruth Davee Department of Neurology, The Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
307
|
Wong SC, Klein JJ, Hamilton HL, Chu Q, Frey CL, Trubetskoy VS, Hegge J, Wakefield D, Rozema DB, Lewis DL. Co-injection of a targeted, reversibly masked endosomolytic polymer dramatically improves the efficacy of cholesterol-conjugated small interfering RNAs in vivo. Nucleic Acid Ther 2013. [PMID: 23181701 DOI: 10.1089/nat.2012.0389] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Effective in vivo delivery of small interfering (siRNA) has been a major obstacle in the development of RNA interference therapeutics. One of the first attempts to overcome this obstacle utilized intravenous injection of cholesterol-conjugated siRNA (chol-siRNA). Although studies in mice revealed target gene knockdown in the liver, delivery was relatively inefficient, requiring 3 daily injections of 50 mg/kg of chol-siRNA to obtain measurable reduction in gene expression. Here we present a new delivery approach that increases the efficacy of the chol-siRNA over 500-fold and allows over 90% reduction in target gene expression in mice and, for the first time, high levels of gene knockdown in non-human primates. This improved efficacy is achieved by the co-injection of a hepatocyte-targeted and reversibly masked endosomolytic polymer. We show that knockdown is absolutely dependent on the presence of hepatocyte-targeting ligand on the polymer, the cognate hepatocyte receptor, and the cholesterol moiety of the siRNA. Importantly, we provide evidence that this increase in efficacy is not dependent on interactions between the chol-siRNA with the polymer prior to injection or in the bloodstream. The simplicity of the formulation and efficacy of this mode of siRNA delivery should prove beneficial in the use of siRNA as a therapeutic.
Collapse
Affiliation(s)
- So C Wong
- Arrowhead Madison Inc., Arrowhead Research Corporation, Madison, Wisconsin, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
308
|
Shi B, Abrams M. Technologies for investigating the physiological barriers to efficient lipid nanoparticle-siRNA delivery. J Histochem Cytochem 2013; 61:407-20. [PMID: 23504369 PMCID: PMC3715328 DOI: 10.1369/0022155413484152] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 02/20/2013] [Indexed: 11/22/2022] Open
Abstract
Small interfering RNA (siRNA) therapeutics have advanced from bench to clinical trials in recent years, along with new tools developed to enable detection of siRNA delivered at the organ, cell, and subcellular levels. Preclinical models of siRNA delivery have benefitted from methodologies such as stem-loop quantitative polymerase chain reaction, histological in situ immunofluorescent staining, endosomal escape assay, and RNA-induced silencing complex loading assay. These technologies have accelerated the detection and optimization of siRNA platforms to overcome the challenges associated with delivering therapeutic oligonucleotides to the cytosol of specific target cells. This review focuses on the methodologies and their application in the biodistribution of siRNA delivered by lipid nanoparticles.
Collapse
Affiliation(s)
- Bin Shi
- Department of RNA Therapeutics, Merck Research Laboratories, Merck & Co., Inc., West Point, Pennsylvania, USA.
| | | |
Collapse
|
309
|
Abstract
Though the pharmaceutical industry's infatuation with the therapeutic potential of RNA interference (RNAi) technology has finally come down from its initial lofty levels,[1] hope is by no means lost for the once-burgeoning enterprise, as recent clinical trials are beginning to show efficacy in areas ranging from amyloidosis to hypercholesterolemia to muscular dystrophy. With such resurgence comes a more informed perspective on the needs of such therapeutics: a renewed focus on true RNA drug development, and a desire for enhanced site-specific delivery.[2] In this review, we will discuss the latter with regard to hepatic targeting by synthetic vectors, covering the implications of organ and cellular physiology on conjugate structure, particle morphology, and active targeting. In presenting efficacy in a variety of disease models, we emphasize as well the extraordinary degree to which synthetic formulation improves upon and coordinates efforts with oligonucleotide development. Such advances in the understanding of and the technology behind RNAi have the potential to finally stabilize the long-term prospects RNA therapeutic development.
Collapse
|
310
|
Takemoto H, Miyata K, Hattori S, Ishii T, Suma T, Uchida S, Nishiyama N, Kataoka K. Acidic pH-Responsive siRNA Conjugate for Reversible Carrier Stability and Accelerated Endosomal Escape with Reduced IFNα-Associated Immune Response. Angew Chem Int Ed Engl 2013; 52:6218-21. [DOI: 10.1002/anie.201300178] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 02/25/2013] [Indexed: 01/30/2023]
|
311
|
Takemoto H, Miyata K, Hattori S, Ishii T, Suma T, Uchida S, Nishiyama N, Kataoka K. Acidic pH-Responsive siRNA Conjugate for Reversible Carrier Stability and Accelerated Endosomal Escape with Reduced IFNα-Associated Immune Response. Angew Chem Int Ed Engl 2013. [DOI: 10.1002/ange.201300178] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
312
|
Alexander C, Fernandez Trillo F. Bioresponsive Polyplexes and Micelleplexes. SMART MATERIALS FOR DRUG DELIVERY 2013. [DOI: 10.1039/9781849736800-00256] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The delivery of nucleic acids (NAs) is hindered by several factors, such as the size of the biomolecule (micron size for plasmid DNA), the presence of different biological barriers or the degradation of NAs. Most of these limitations are avoided by complexation with polycationic species, which collapse NAs into nanometer-sized polyplexes that can be efficiently internalized into the target cells. Because there are subtle changes in physiological conditions, such as the drop in pH at the endosome, or the increase in temperature in tumor tissue, stimuli responsive synthetic polymers are ideal candidates for the synthesis of efficient gene delivery vehicles. In this chapter, representative examples of “smart” polypexes that exploit these changes in physiological environment for the delivery of NAs are described, and the transfection efficiency of pH-, redox-, temperature- and light-responsive polyplexes is analyzed.
Collapse
|
313
|
Uchino K, Ochiya T, Takeshita F. RNAi therapeutics and applications of microRNAs in cancer treatment. Jpn J Clin Oncol 2013; 43:596-607. [PMID: 23592885 DOI: 10.1093/jjco/hyt052] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
RNA interference-based therapies are proving to be powerful tools for combating various diseases, including cancer. Scientists are researching the development of safe and efficient systems for the delivery of small RNA molecules, which are extremely fragile in serum, to target organs and cells in the human body. A dozen pre-clinical and clinical trials have been under way over the past few years involving biodegradable nanoparticles, lipids, chemical modification and conjugation. On the other hand, microRNAs, which control the balance of cellular biological processes, have been studied as attractive therapeutic targets in cancer treatment. In this review, we provide an overview of RNA interference-based therapeutics in clinical trials and discuss the latest technology for the systemic delivery of nucleic acid drugs. Furthermore, we focus on dysregulated microRNAs in human cancer, which have progressed in pre-clinical trials as therapeutic targets, and describe a wide range of strategies to control the expression levels of endogenous microRNAs. Further development of RNA interference technologies and progression of clinical trials will contribute to the achievement of practical applications of nucleic acid drugs.
Collapse
Affiliation(s)
- Keita Uchino
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, 1-1, Tsukiji 5-chome, Chuo-ku, Tokyo 104-0045, Japan
| | | | | |
Collapse
|
314
|
Gooding M, Browne LP, Quinteiro FM, Selwood DL. siRNA delivery: from lipids to cell-penetrating peptides and their mimics. Chem Biol Drug Des 2013; 80:787-809. [PMID: 22974319 DOI: 10.1111/cbdd.12052] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
To deliver siRNA for therapeutic use, several hurdles must be addressed. Metabolic degradation must be blocked, and the RNAi cellular machinery is located in the cytoplasm, while double-stranded siRNA is large, highly charged and impermeable to cell membranes. To date, the solutions to the delivery issues have mostly involved different forms of lipid particle encapsulation. Cell-penetrating peptides and their mimics or analogues offer a different approach and this is an emerging field with the first in vivo examples now reported. Recent reports point to lipid receptors being involved in the cellular uptake of both types of transporter. This review examines the delivery of siRNA with a focus on cell-penetrating peptides and their small molecule and oligomeric mimics. The current status of siRNA delivery methods in clinical trials is examined. It now seems that the goal of delivering siRNA therapeutically is achievable but will they form part of a sustainable healthcare portfolio for the future.
Collapse
Affiliation(s)
- Matt Gooding
- The Wolfson Institute for Biomedical Research, UCL, Gower Street, London WC1E 6BT, UK
| | | | | | | |
Collapse
|
315
|
Zhou J, Ke F, Xia Y, Sun J, Xu N, Li ZC, Liang D. Complexation of DNA with poly-(L-lysine) and its copolymers in dimethylformamide. POLYMER 2013. [DOI: 10.1016/j.polymer.2013.03.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
316
|
Parmar RG, Busuek M, Walsh ES, Leander KR, Howell BJ, Sepp-Lorenzino L, Kemp E, Crocker LS, Leone A, Kochansky CJ, Carr BA, Garbaccio RM, Colletti SL, Wang W. Endosomolytic Bioreducible Poly(amido amine disulfide) Polymer Conjugates for the in Vivo Systemic Delivery of siRNA Therapeutics. Bioconjug Chem 2013; 24:640-7. [PMID: 23496378 DOI: 10.1021/bc300600a] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Rubina Giare Parmar
- Department of Medicinal Chemistry, ‡Department of RNA Biology, §Department of Pharmaceutical Sciences, and ∥Department of Pharmacokinetic Pharmacodynamic Drug Metabolism, Merck & Co. Inc, West Point, Pennsylvania, United States
| | - Marina Busuek
- Department of Medicinal Chemistry, ‡Department of RNA Biology, §Department of Pharmaceutical Sciences, and ∥Department of Pharmacokinetic Pharmacodynamic Drug Metabolism, Merck & Co. Inc, West Point, Pennsylvania, United States
| | - Eileen S. Walsh
- Department of Medicinal Chemistry, ‡Department of RNA Biology, §Department of Pharmaceutical Sciences, and ∥Department of Pharmacokinetic Pharmacodynamic Drug Metabolism, Merck & Co. Inc, West Point, Pennsylvania, United States
| | - Karen R. Leander
- Department of Medicinal Chemistry, ‡Department of RNA Biology, §Department of Pharmaceutical Sciences, and ∥Department of Pharmacokinetic Pharmacodynamic Drug Metabolism, Merck & Co. Inc, West Point, Pennsylvania, United States
| | - Bonnie J. Howell
- Department of Medicinal Chemistry, ‡Department of RNA Biology, §Department of Pharmaceutical Sciences, and ∥Department of Pharmacokinetic Pharmacodynamic Drug Metabolism, Merck & Co. Inc, West Point, Pennsylvania, United States
| | - Laura Sepp-Lorenzino
- Department of Medicinal Chemistry, ‡Department of RNA Biology, §Department of Pharmaceutical Sciences, and ∥Department of Pharmacokinetic Pharmacodynamic Drug Metabolism, Merck & Co. Inc, West Point, Pennsylvania, United States
| | - Eric Kemp
- Department of Medicinal Chemistry, ‡Department of RNA Biology, §Department of Pharmaceutical Sciences, and ∥Department of Pharmacokinetic Pharmacodynamic Drug Metabolism, Merck & Co. Inc, West Point, Pennsylvania, United States
| | - Louis S. Crocker
- Department of Medicinal Chemistry, ‡Department of RNA Biology, §Department of Pharmaceutical Sciences, and ∥Department of Pharmacokinetic Pharmacodynamic Drug Metabolism, Merck & Co. Inc, West Point, Pennsylvania, United States
| | - Anthony Leone
- Department of Medicinal Chemistry, ‡Department of RNA Biology, §Department of Pharmaceutical Sciences, and ∥Department of Pharmacokinetic Pharmacodynamic Drug Metabolism, Merck & Co. Inc, West Point, Pennsylvania, United States
| | - Christopher J. Kochansky
- Department of Medicinal Chemistry, ‡Department of RNA Biology, §Department of Pharmaceutical Sciences, and ∥Department of Pharmacokinetic Pharmacodynamic Drug Metabolism, Merck & Co. Inc, West Point, Pennsylvania, United States
| | - Brian A. Carr
- Department of Medicinal Chemistry, ‡Department of RNA Biology, §Department of Pharmaceutical Sciences, and ∥Department of Pharmacokinetic Pharmacodynamic Drug Metabolism, Merck & Co. Inc, West Point, Pennsylvania, United States
| | - Robert M. Garbaccio
- Department of Medicinal Chemistry, ‡Department of RNA Biology, §Department of Pharmaceutical Sciences, and ∥Department of Pharmacokinetic Pharmacodynamic Drug Metabolism, Merck & Co. Inc, West Point, Pennsylvania, United States
| | - Steven L. Colletti
- Department of Medicinal Chemistry, ‡Department of RNA Biology, §Department of Pharmaceutical Sciences, and ∥Department of Pharmacokinetic Pharmacodynamic Drug Metabolism, Merck & Co. Inc, West Point, Pennsylvania, United States
| | - Weimin Wang
- Department of Medicinal Chemistry, ‡Department of RNA Biology, §Department of Pharmaceutical Sciences, and ∥Department of Pharmacokinetic Pharmacodynamic Drug Metabolism, Merck & Co. Inc, West Point, Pennsylvania, United States
| |
Collapse
|
317
|
Abstract
RNA interference (RNAi) drugs have significant therapeutic potential, but delivery systems with appropriate efficacy and toxicity profiles are still needed. Here, we describe a neutral, ampholytic polymeric delivery system based on conjugatable diblock polymer micelles. The diblock copolymer contains a hydrophilic poly[N-(2-hydroxypropyl)methacrylamide-co-N-(2-(pyridin-2-yldisulfanyl)ethyl)methacrylamide) (poly[HPMA-co-PDSMA]) segment to promote aqueous stability and facilitate thiol-disulfide exchange reactions and a second ampholytic block composed of propylacrylic acid (PAA), dimethylaminoethyl methacrylate (DMAEMA), and butyl methacrylate (BMA). The poly[(HPMA-co-PDSMA)-b-(PAA-co-DMAEMA-co-BMA)] was synthesized using reversible addition-fragmentation chain transfer (RAFT) polymerization with an overall molecular weight of 22 000 g/mol and a PDI of 1.88. Dynamic light scattering and fluorescence measurements indicated that the diblock copolymers self-assemble under aqueous conditions to form polymeric micelles with a hydrodynamic radius and critical micelle concentration of 25 nm and 25 μg/mL, respectively. Red blood cell hemolysis experiments show that the neutral hydrophilic micelles have potent membrane destabilizing activity at endosomal pH values. Thiolated siRNA targeting glyceraldehyde 3-phosphate dehydrogenase (GAPDH) was directly conjugated to the polymeric micelles via thiol exchange reactions with the pyridal disulfide groups present in the micelle corona. Maximum silencing activity in HeLa cells was observed at a 1:10 molar ratio of siRNA to polymer following a 48 h incubation period. Under these conditions 90% mRNA knockdown and 65% protein knockdown at 48 h was achieved with negligible toxicity. In contrast the polymeric micelles lacking a pH-responsive endosomalytic segment demonstrated negligible mRNA and protein knockdown under these conditions. The potent mRNA knockdown and excellent biocompatibility of the neutral siRNA conjugates demonstrate the potential utility of this carrier design for delivering therapeutic siRNA drugs.
Collapse
Affiliation(s)
- Brittany B. Lundy
- Department of Bioengineering, University of Washington, Seattle, WA 98195
| | - Anthony Convertine
- Department of Bioengineering, University of Washington, Seattle, WA 98195
| | - Martina Miteva
- Department of Bioengineering, University of Washington, Seattle, WA 98195
| | - Patrick S. Stayton
- Department of Bioengineering, University of Washington, Seattle, WA 98195
| |
Collapse
|
318
|
Wooddell CI, Rozema DB, Hossbach M, John M, Hamilton HL, Chu Q, Hegge JO, Klein JJ, Wakefield DH, Oropeza CE, Deckert J, Roehl I, Jahn-Hofmann K, Hadwiger P, Vornlocher HP, McLachlan A, Lewis DL. Hepatocyte-targeted RNAi therapeutics for the treatment of chronic hepatitis B virus infection. Mol Ther 2013; 21:973-85. [PMID: 23439496 PMCID: PMC3666629 DOI: 10.1038/mt.2013.31] [Citation(s) in RCA: 239] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
RNA interference (RNAi)-based therapeutics have the potential to treat chronic hepatitis B virus (HBV) infection in a fundamentally different manner than current therapies. Using RNAi, it is possible to knock down expression of viral RNAs including the pregenomic RNA from which the replicative intermediates are derived, thus reducing viral load, and the viral proteins that result in disease and impact the immune system's ability to eliminate the virus. We previously described the use of polymer-based Dynamic PolyConjugate (DPC) for the targeted delivery of siRNAs to hepatocytes. Here, we first show in proof-of-concept studies that simple coinjection of a hepatocyte-targeted, N-acetylgalactosamine-conjugated melittin-like peptide (NAG-MLP) with a liver-tropic cholesterol-conjugated siRNA (chol-siRNA) targeting coagulation factor VII (F7) results in efficient F7 knockdown in mice and nonhuman primates without changes in clinical chemistry or induction of cytokines. Using transient and transgenic mouse models of HBV infection, we show that a single coinjection of NAG-MLP with potent chol-siRNAs targeting conserved HBV sequences resulted in multilog repression of viral RNA, proteins, and viral DNA with long duration of effect. These results suggest that coinjection of NAG-MLP and chol-siHBVs holds great promise as a new therapeutic for patients chronically infected with HBV.
Collapse
Affiliation(s)
- Christine I Wooddell
- Arrowhead Research Corporation, Arrowhead Madison, Madison, Wisconsin 53711, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
319
|
Goldberg MS. siRNA delivery for the treatment of ovarian cancer. Methods 2013; 63:95-100. [PMID: 23403216 DOI: 10.1016/j.ymeth.2013.01.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 01/23/2013] [Accepted: 01/24/2013] [Indexed: 12/21/2022] Open
Abstract
Short interfering RNAs (siRNAs) mediate the catalytic sequence-specific cleavage of target messenger RNA (mRNA) molecules, resulting in the silencing of gene products in an efficient and precise manner. One apparent application of this technology is the knockdown of genes responsible for cancer progression, including pro-proliferative oncogenes, inhibitors of apoptosis, and mediators of angiogenesis. Delivery of siRNAs into particular cells has remained the principal obstacle to the realization of the potential of RNA interference (RNAi) in the clinic. Several groups have worked to develop carriers that facilitate siRNA delivery into ovarian cancer cells in mouse models of ovarian cancer. The results have been promising, often leading to significant survival extension. Such benefit is critical for a disease that is characterized by very poor outcomes and demands novel treatment options. This review describes advancements in siRNA delivery for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Michael S Goldberg
- Dana-Farber Cancer Institute, 450 Brookline Ave., Boston, MA 02215, United States.
| |
Collapse
|
320
|
Khormaee S, Choi Y, Shen MJ, Xu B, Wu H, Griffiths GL, Chen R, Slater NKH, Park JK. Endosomolytic anionic polymer for the cytoplasmic delivery of siRNAs in localized in vivo applications. ADVANCED FUNCTIONAL MATERIALS 2013; 23:10.1002/adfm.201201945. [PMID: 24273480 PMCID: PMC3834980 DOI: 10.1002/adfm.201201945] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
The use of small interfering RNAs (siRNAs) to down-regulate the expression of disease-associated proteins carries significant promise for the treatment of a variety of clinical disorders. One of the main barriers to the widespread clinical use of siRNAs, however, is their entrapment and degradation within the endolysosomal pathway of target cells. Here we report the trafficking and function of PP75, a non-toxic, biodegradable, lipid membrane disruptive anionic polymer composed of phenylalanine derivatized poly(L-lysine iso-phthalamide). PP75 is readily endocytosed by cells, safely permeabilizes endolysosomes in a pH dependent manner and facilitates the transfer of co-endocytosed materials directly into the cytoplasm. The covalent attachment of siRNAs to PP75 using disulfide linkages generates conjugates that effectively traffic siRNAs to the cytoplasm of target cells both in vitro and in vivo. In a subcutaneous malignant glioma tumor model, a locally delivered PP75-stathmin siRNA conjugate decreases stathmin expression in tumor cells and, in combination with the nitrosourea chemotherapy carmustine, is highly effective at inhibiting tumor growth. PP75 may be clinically useful for the local delivery of siRNAs, in particular for the treatment of solid tumors.
Collapse
Affiliation(s)
- Sariah Khormaee
- Surgical and Molecular Neuro-oncology Unit, NINDS, NIH, Bethesda, MD, 20892, USA
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB2 3RA, UK
| | - Yong Choi
- Surgical and Molecular Neuro-oncology Unit, NINDS, NIH, Bethesda, MD, 20892, USA
| | - Michael J. Shen
- Surgical and Molecular Neuro-oncology Unit, NINDS, NIH, Bethesda, MD, 20892, USA
| | - Biying Xu
- Imaging Probe Development Center, NHLBI, NIH, Rockville, MD, 20850, USA
| | - Haitao Wu
- Imaging Probe Development Center, NHLBI, NIH, Rockville, MD, 20850, USA
| | - Gary L. Griffiths
- Imaging Probe Development Center, NHLBI, NIH, Rockville, MD, 20850, USA
| | - Rongjun Chen
- Centre for Molecular Nanoscience, School of Chemistry, University of Leeds, Leeds LS2 9JT, UK
| | - Nigel K. H. Slater
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB2 3RA, UK
| | - John K. Park
- Surgical and Molecular Neuro-oncology Unit, NINDS, NIH, Bethesda, MD, 20892, USA
| |
Collapse
|
321
|
Tabernero J, Shapiro GI, LoRusso PM, Cervantes A, Schwartz GK, Weiss GJ, Paz-Ares L, Cho DC, Infante JR, Alsina M, Gounder MM, Falzone R, Harrop J, White ACS, Toudjarska I, Bumcrot D, Meyers RE, Hinkle G, Svrzikapa N, Hutabarat RM, Clausen VA, Cehelsky J, Nochur SV, Gamba-Vitalo C, Vaishnaw AK, Sah DWY, Gollob JA, Burris HA. First-in-humans trial of an RNA interference therapeutic targeting VEGF and KSP in cancer patients with liver involvement. Cancer Discov 2013; 3:406-17. [PMID: 23358650 DOI: 10.1158/2159-8290.cd-12-0429] [Citation(s) in RCA: 503] [Impact Index Per Article: 41.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
UNLABELLED RNA interference (RNAi) is a potent and specific mechanism for regulating gene expression. Harnessing RNAi to silence genes involved in disease holds promise for the development of a new class of therapeutics. Delivery is key to realizing the potential of RNAi, and lipid nanoparticles (LNP) have proved effective in delivery of siRNAs to the liver and to tumors in animals. To examine the activity and safety of LNP-formulated siRNAs in humans, we initiated a trial of ALN-VSP, an LNP formulation of siRNAs targeting VEGF and kinesin spindle protein (KSP), in patients with cancer. Here, we show detection of drug in tumor biopsies, siRNA-mediated mRNA cleavage in the liver, pharmacodynamics suggestive of target downregulation, and antitumor activity, including complete regression of liver metastases in endometrial cancer. In addition, we show that biweekly intravenous administration of ALN-VSP was safe and well tolerated. These data provide proof-of-concept for RNAi therapeutics in humans and form the basis for further development in cancer. SIGNIFICANCE The fi ndings in this report show safety, pharmacokinetics, RNAi mechanism of action, and clinical activity with a novel fi rst-in-class LNP-formulated RNAi therapeutic in patients with cancer. The ability to harness RNAi to facilitate specifi c multitargeting, as well as increase the number of druggable targets, has important implications for future drug development in oncology.
Collapse
Affiliation(s)
- Josep Tabernero
- Medical Oncology Department, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
322
|
Protein-resistant, reductively dissociable polyplexes for in vivo systemic delivery and tumor-targeting of siRNA. Biomaterials 2013; 34:2370-9. [PMID: 23294546 DOI: 10.1016/j.biomaterials.2012.12.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 12/09/2012] [Indexed: 01/19/2023]
Abstract
Small interfering RNA (siRNA) has been considered as a very attractive therapeutic alternative to chemical drugs; however, the chemical and biological instability and poor delivery efficiency of siRNA limit its success in clinical applications. Here we report a protein-resistant, reductively dissociable siRNA delivery system based on self-assembled polyelectrolyte complexes of dextran-siRNA conjugates linked by disulfide bonds. The prepared polyplexes exhibit excellent dispersion stability in the presence of serum because of the anti-fouling property of dextran exposed onto the complex surface. The enzymatic degradation of siRNA is also effectively suppressed within the complex. Folates are introduced as an active tumor-targeting moiety via the conjugation of folates to the hydroxyl groups of dextran. An in vivo investigation with a xenograft tumor mouse model shows that the folate-decorated dextran-siRNA conjugates are very efficiently targeted to cancer cells and induce sequence-specific gene silencing.
Collapse
|
323
|
Gallas A, Alexander C, Davies MC, Puri S, Allen S. Chemistry and formulations for siRNA therapeutics. Chem Soc Rev 2013; 42:7983-97. [DOI: 10.1039/c3cs35520a] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
324
|
|
325
|
|
326
|
Schlenk F, Grund S, Fischer D. Recent developments and perspectives on gene therapy using synthetic vectors. Ther Deliv 2013; 4:95-113. [PMID: 23323783 DOI: 10.4155/tde.12.128] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025] Open
Abstract
Nonviral vector technology is attracting increasing importance in the biomedical community owing to unique advantages and prospects for the treatment of severe diseases by gene therapy. In this review, synthetic vectors that allow the controlled design of efficient and biocompatible carriers are highlighted. The current benefits, potentials, problems and unmet needs of synthetic gene delivery systems, as well as the strategies to overcome the obstacles are also discussed. Common design principles and structure-activity trends have been established that are important for stable and targeted transport to regions of interest in the body, efficient uptake into cells as well as controlled release of drugs inside the cells, for example, in specialized compartments. The status quo of the use of these systems in preclinical and clinical trials is also considered.
Collapse
Affiliation(s)
- Florian Schlenk
- Department of Pharmaceutical Technology, Friedrich-Schiller-University Jena, Jena, Germany
| | | | | |
Collapse
|
327
|
Marra E, Palombo F, Ciliberto G, Aurisicchio L. Kinesin spindle protein SiRNA slows tumor progression. J Cell Physiol 2012; 228:58-64. [PMID: 22552964 DOI: 10.1002/jcp.24103] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The kinesin spindle protein (KSP), a member of the kinesin superfamily of microtubule-based motors, plays a critical role in mitosis as it mediates centrosome separation and bipolar spindle assembly and maintenance. Inhibition of KSP function leads to cell cycle arrest at mitosis with the formation of monoastral microtubule arrays, and ultimately, to cell death. Several KSP inhibitors are currently being studied in clinical trials and provide new opportunities for the development of novel anticancer therapeutics. RNA interference (RNAi) may represent a powerful strategy to interfere with key molecular pathways involved in cancer. In this study, we have established an efficient method for intratumoral delivery of siRNA. We evaluated short interfering RNA (siRNA) duplexes targeting luciferase as surrogate marker or KSP sequence. To examine the potential feasibility of RNAi therapy, the siRNA was transfected into pre-established lesions by means of intratumor electro-transfer of RNA therapeutics (IERT). This technology allowed cell permeation of the nucleic acids and to efficiently knock down gene expression, albeit transiently. The KSP-specific siRNA drastically reduced outgrowth of subcutaneous melanoma and ovarian cancer lesions. Our results show that intratumoral electro-transfer of siRNA is feasible and KSP-specific siRNA may provide a novel strategy for therapeutic intervention.
Collapse
|
328
|
Guo P, Haque F, Hallahan B, Reif R, Li H. Uniqueness, advantages, challenges, solutions, and perspectives in therapeutics applying RNA nanotechnology. Nucleic Acid Ther 2012; 22:226-45. [PMID: 22913595 DOI: 10.1089/nat.2012.0350] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The field of RNA nanotechnology is rapidly emerging. RNA can be manipulated with the simplicity characteristic of DNA to produce nanoparticles with a diversity of quaternary structures by self-assembly. Additionally RNA is tremendously versatile in its function and some RNA molecules display catalytic activities much like proteins. Thus, RNA has the advantage of both worlds. However, the instability of RNA has made many scientists flinch away from RNA nanotechnology. Other concerns that have deterred the progress of RNA therapeutics include the induction of interferons, stimulation of cytokines, and activation of other immune systems, as well as short pharmacokinetic profiles in vivo. This review will provide some solutions and perspectives on the chemical and thermodynamic stability, in vivo half-life and biodistribution, yield and production cost, in vivo toxicity and side effect, specific delivery and targeting, as well as endosomal trapping and escape.
Collapse
Affiliation(s)
- Peixuan Guo
- Nanobiotechnology Center, Markey Cancer Center and Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky 40536, USA.
| | | | | | | | | |
Collapse
|
329
|
Wang HX, Xiong MH, Wang YC, Zhu J, Wang J. N-acetylgalactosamine functionalized mixed micellar nanoparticles for targeted delivery of siRNA to liver. J Control Release 2012; 166:106-14. [PMID: 23266452 DOI: 10.1016/j.jconrel.2012.12.017] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Revised: 11/23/2012] [Accepted: 12/10/2012] [Indexed: 01/12/2023]
Abstract
Due to its efficient and specific gene silencing ability, RNA interference has shown great potential in the treatment of liver diseases. However, achieving in vivo delivery of siRNA to critical liver cells remains the biggest obstacle for this technique to be a real clinic therapeutic modality. Here, we describe a promising liver targeting siRNA delivery system based on N-acetylgalactosamine functionalized mixed micellar nanoparticles (Gal-MNP), which can efficiently deliver siRNA to hepatocytes and silence the target gene expression after systemic administration. The Gal-MNP were assembled in aqueous solution from mixed N-acetylgalactosamine functionalized poly(ethylene glycol)-b-poly(ε-caprolactone) and cationic poly(ε-caprolactone)-b-poly(2-aminoethyl ethylene phosphate) (PCL-b-PPEEA); the properties of nanoparticles, including particle size, zeta potential and the density of poly(ethylene glycol) could be easily regulated. The hepatocyte-targeting effect of Gal-MNP was demonstrated by significant enriching of fluorescent siRNA in primary hepatocytes in vitro and in vivo. Successful down-regulation of liver-specific apolipoprotein B (apoB) expression was achieved in mouse liver, at both the transcriptional and protein level, following intravenous injection of Gal-MNP/siapoB to BALB/c mice. Systemic delivery of Gal-MNP/siRNA did not induce the innate immune response or positive hepatotoxicity. The results of this study suggested therapeutic potential for the Gal-MNP/siRNA system in liver disease.
Collapse
Affiliation(s)
- Hong-Xia Wang
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, PR China
| | | | | | | | | |
Collapse
|
330
|
Schellinger JG, Pahang JA, Johnson RN, Chu DSH, Sellers DL, Maris DO, Convertine AJ, Stayton PS, Horner PJ, Pun SH. Melittin-grafted HPMA-oligolysine based copolymers for gene delivery. Biomaterials 2012; 34:2318-26. [PMID: 23261217 DOI: 10.1016/j.biomaterials.2012.09.072] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Accepted: 09/28/2012] [Indexed: 10/27/2022]
Abstract
Non-viral gene delivery systems capable of transfecting cells in the brain are critical in realizing the potential impact of nucleic acid therapeutics for diseases of the central nervous system. In this study, the membrane-lytic peptide melittin was incorporated into block copolymers synthesized by reversible addition-fragmentation chain transfer (RAFT) polymerization. The first block, designed for melittin conjugation, was composed of N-(2-hydroxypropyl)methacrylamide (HPMA) and pyridyl disulfide methacrylamide (PDSMA) and the second block, designed for DNA binding, was composed of oligo-l-lysine (K10) and HPMA. Melittin modified with cysteine at the C-terminus was conjugated to the polymers through the pyridyl disulfide pendent groups via disulfide exchange. The resulting pHgMelbHK10 copolymers are more membrane-lytic than melittin-free control polymers, and efficiently condensed plasmid DNA into salt-stable particles (~100-200 nm). The melittin-modified polymers transfected both HeLa and neuron-like PC-12 cells more efficiently than melittin-free polymers although toxicity associated with the melittin peptide was observed. Optimized formulations containing the luciferase reporter gene were delivered to mouse brain by intraventricular brain injections. Melittin-containing polyplexes produced about 35-fold higher luciferase activity in the brain compared to polyplexes without melittin. Thus, the melittin-containing block copolymers described in this work are promising materials for gene delivery to the brain.
Collapse
Affiliation(s)
- Joan G Schellinger
- Department of Bioengineering, Univeristy of Washington, Seattle, WA 98195, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
331
|
Juliano RL, Carver K, Cao C, Ming X. Receptors, endocytosis, and trafficking: the biological basis of targeted delivery of antisense and siRNA oligonucleotides. J Drug Target 2012; 21:27-43. [PMID: 23163768 DOI: 10.3109/1061186x.2012.740674] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The problem of targeted delivery of antisense and siRNA oligonucleotides can be resolved into two distinct aspects. The first concerns devising ligand-oligonucleotide or ligand-carrier moieties that bind with high selectivity to receptors on the cell type of interest and that are efficiently internalized by endocytosis. The second concerns releasing oligonucleotides from pharmacologically inert endomembrane compartments so that they can access RNA in the cytosol or nucleus. In this review, we will address both of these aspects. Thus, we present information on three important receptor families, the integrins, the receptor tyrosine kinases, and the G protein-coupled receptors in terms of their suitability for targeted delivery of oligonucleotides. This includes discussion of receptor abundance, internalization and trafficking pathways, and the availability of suitable high affinity ligands. We also consider the process of oligonucleotide uptake and intracellular trafficking and discuss approaches to modulating these processes in a pharmacologically productive manner. Hopefully, the basic information presented in this review will be of value to investigators involved in designing delivery approaches for oligonucleotides.
Collapse
Affiliation(s)
- R L Juliano
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA.
| | | | | | | |
Collapse
|
332
|
Sato Y, Hatakeyama H, Sakurai Y, Hyodo M, Akita H, Harashima H. 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. [PMID: 23000694 DOI: 10.1016/j.jconrel.2012.09.009] [Citation(s) in RCA: 223] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Revised: 09/07/2012] [Accepted: 09/13/2012] [Indexed: 11/28/2022]
Abstract
Modification of liposomal siRNA carriers with polyethylene glycol, i.e., PEGylation, is a generally accepted strategy for achieving in vivo stability and delivery to tumor tissue. However, PEGylation significantly inhibits both cellular uptake and the endosomal escape process of the carriers. In a previous study, we reported on the development of a multifunctional envelope-type nano device (MEND) for siRNA delivery and peptide-based functional devices for overcoming the limitations and succeeded in the efficient delivery of siRNA to tumors. In this study, we synthesized a pH-sensitive cationic lipid, YSK05, to overcome the limitations. The YSK05-MEND had a higher ability for endosomal escape than other MENDs containing conventional cationic lipids. The PEGylated YSK05-MEND induced efficient gene silencing and overcame the limitations followed by optimization of the lipid composition. Furthermore, the intratumoral administration of the YSK05-MEND resulted in a more efficient gene silencing compared with MENDs containing conventional cationic lipids. Collectively, these data confirm that YSK05 facilitates the endosomal escape of the MEND and thereby enhances the efficacy of siRNA delivery into cytosol and gene silencing.
Collapse
Affiliation(s)
- Yusuke Sato
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, Japan
| | | | | | | | | | | |
Collapse
|
333
|
Braconi C, Patel T. Non-coding RNAs as therapeutic targets in hepatocellular cancer. Curr Cancer Drug Targets 2012; 12:1073-1080. [PMID: 22873215 PMCID: PMC3916140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Revised: 05/14/2011] [Accepted: 03/11/2012] [Indexed: 06/01/2023]
Abstract
Hepatocellular carcinoma (HCC) is a common malignancy that affects a large number of patients worldwide, with an increasing incidence in the United States and Europe. The therapies that are currently available for patients with inoperable HCC have limited benefits. Although molecular targeted therapies against selected cell signaling pathways have shown some promising results, their impact has been minimal. There is a need to identify and explore other targets for the development of novel therapeutics. Several non-protein coding RNAs (ncRNA) have recently been implicated in hepatocarcinogenesis and tumor progression. These ncRNA genes represent promising targets for cancer. However, therapeutic targeting of ncRNA genes has not been employed for HCC. The use of antisense oligonucleotides and viral vector delivery approaches have been shown to be feasible approaches to modulate ncRNA expression. HCC is an optimal cancer to evaluate novel RNA based therapeutic approaches because of the potential of effective delivery and uptake of therapeutic agents to the liver. In this review, we discuss selected ncRNA that could function as potential targets in HCC treatment and outline approaches to target ncRNA expression. Future challenges include the need to achieve site-specific targeting with acceptable safety and efficacy.
Collapse
Affiliation(s)
- Chiara Braconi
- The Ohio State University Medical Center, 460 West 12 Avenue, Columbus, OH 43212, USA
| | - Tushar Patel
- Mayo Clinic, 4500 San Pablo Boulevard, Jacksonville, FL 32224, USA
| |
Collapse
|
334
|
Naito M, Ishii T, Matsumoto A, Miyata K, Miyahara Y, Kataoka K. A phenylboronate-functionalized polyion complex micelle for ATP-triggered release of siRNA. Angew Chem Int Ed Engl 2012; 51:10751-5. [PMID: 22907668 DOI: 10.1002/anie.201203360] [Citation(s) in RCA: 181] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Revised: 06/26/2012] [Indexed: 12/19/2022]
Affiliation(s)
- Mitsuru Naito
- Department of Materials Engineering, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-8656, Japan
| | | | | | | | | | | |
Collapse
|
335
|
Troiber C, Kasper JC, Milani S, Scheible M, Martin I, Schaubhut F, Küchler S, Rädler J, Simmel FC, Friess W, Wagner E. Comparison of four different particle sizing methods for siRNA polyplex characterization. Eur J Pharm Biopharm 2012; 84:255-64. [PMID: 23079135 DOI: 10.1016/j.ejpb.2012.08.014] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Revised: 08/02/2012] [Accepted: 08/22/2012] [Indexed: 10/27/2022]
Abstract
The ability to reliably determine the size of siRNA polyplexes is the key for the rational design of particles and their formulation, as well as, their safe application in vivo. At the moment, no standard technique for size measurements is available. Each method has different underlying principles and hence may give different results. Here, four different analytical methods were evaluated for their suitability to analyze the characteristics of homogeneous and heterogeneous siRNA polyplexes: dynamic light scattering (DLS), atomic force microscopy (AFM), nanoparticle trafficking analysis (NTA), and fluorescence correlation spectroscopy (FCS). Three different siRNA polyplex compositions generated with different, precise, and hydrophobically modified oligoaminoamides were used in this study. All of the evaluated methods were suitable for analysis of medium sized, homogeneous siRNA polyplexes (~120 nm). Small particles (<40 nm) could not be tracked with NTA, but with the other three methods. Heterogeneous polyplexes were generally difficult to analyze. Only by visualization with AFM, the heterogeneity of those polyplexes was observable. FCS was the only method suitable for measuring polyplex stability in 90% fetal bovine serum. Physico-chemical characteristics of polyplexes are important quality criterions for successful in vivo application and future formulation development. Therefore, a comprehensive analysis by more than one method is of particular importance.
Collapse
Affiliation(s)
- Christina Troiber
- Department of Pharmacy, Pharmaceutical Biotechnology, Ludwig-Maximilians-University, Munich, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
336
|
Daka A, Peer D. RNAi-based nanomedicines for targeted personalized therapy. Adv Drug Deliv Rev 2012; 64:1508-21. [PMID: 22975009 DOI: 10.1016/j.addr.2012.08.014] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Revised: 08/07/2012] [Accepted: 08/13/2012] [Indexed: 12/31/2022]
Abstract
RNA interference (RNAi) has just made it through the pipeline to clinical trials. However, in order for RNAi to serve as an ideal personalized therapeutics and be clinically approved-safe, specific, and potent strategies must be devised for efficient delivery of RNAi payloads to specific cell types, which despite the immense potential, remains a challenge. Through evaluating the recent reported studies in this field, we introduce the progress in designing targeted nano-scaled strategies that are anticipated to overcome the delivery drawbacks and along with the exciting "omics" discipline to personalize RNAi-based therapeutics.
Collapse
Affiliation(s)
- Ala Daka
- Laboratory of Nanomedicine, Department of Cell Research and Immunology, George S. Wise Faculty of Life Science, Israel
| | | |
Collapse
|
337
|
The effect of RAFT-derived cationic block copolymer structure on gene silencing efficiency. Biomaterials 2012; 33:7631-42. [DOI: 10.1016/j.biomaterials.2012.06.090] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Accepted: 06/28/2012] [Indexed: 01/03/2023]
|
338
|
Efficient reduction of serum cholesterol by combining a liver-targeted gene delivery system with chemically modified apolipoprotein B siRNA. J Control Release 2012; 163:119-24. [DOI: 10.1016/j.jconrel.2012.08.030] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2012] [Revised: 08/24/2012] [Accepted: 08/29/2012] [Indexed: 12/19/2022]
|
339
|
Targeted siRNA Delivery and mRNA Knockdown Mediated by Bispecific Digoxigenin-binding Antibodies. MOLECULAR THERAPY. NUCLEIC ACIDS 2012; 1:e46. [PMID: 23344238 PMCID: PMC3464882 DOI: 10.1038/mtna.2012.39] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Bispecific antibodies (bsAbs) that bind to cell surface antigens and to digoxigenin (Dig) were used for targeted small interfering RNA (siRNA) delivery. They are derivatives of immunoglobulins G (IgGs) that bind tumor antigens, such as Her2, IGF1-R, CD22, and LeY, with stabilized Dig-binding variable domains fused to the C-terminal ends of the heavy chains. siRNA that was digoxigeninylated at its 3′end was bound in a 2:1 ratio to the bsAbs. These bsAb–siRNA complexes delivered siRNAs specifically to cells that express the corresponding antigen as demonstrated by flow cytometry and confocal microscopy. The complexes internalized into endosomes and Dig-siRNAs separated from bsAbs, but Dig-siRNA was not released into the cytoplasm; bsAb-targeting alone was thus not sufficient for effective mRNA knockdown. This limitation was overcome by formulating the Dig-siRNA into nanoparticles consisting of dynamic polyconjugates (DPCs) or into lipid-based nanoparticles (LNPs). The resulting complexes enabled bsAb-targeted siRNA-specific messenger RNA (mRNA) knockdown with IC50 siRNA values in the low nanomolar range for a variety of bsAbs, siRNAs, and target cells. Furthermore, pilot studies in mice bearing tumor xenografts indicated mRNA knockdown in endothelial cells following systemic co-administration of bsAbs and siRNA formulated in LNPs that were targeted to the tumor vasculature.
Collapse
|
340
|
Lee SJ, Son S, Yhee JY, Choi K, Kwon IC, Kim SH, Kim K. Structural modification of siRNA for efficient gene silencing. Biotechnol Adv 2012; 31:491-503. [PMID: 22985697 DOI: 10.1016/j.biotechadv.2012.09.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Revised: 09/07/2012] [Accepted: 09/07/2012] [Indexed: 11/16/2022]
Abstract
Small interfering RNA (siRNA) holds a great promise for the future of genomic medicine because of its highly sequence-specific gene silencing and universality in therapeutic target. The medical use of siRNA, however, has been severely hampered by the inherent physico-chemical properties of siRNA itself, such as low charge density, high structural stiffness and rapid enzymatic degradation; therefore, the establishment of efficient and safe siRNA delivery methodology is an essential prerequisite, particularly for systemic administration. For an efficient systemic siRNA delivery, it is a critical issue to obtain small and compact siRNA polyplexes with cationic condensing reagents including cationic polymers, because the size and surface properties of the polyplexes are major determinants for achieving desirable in vivo fate. Unfortunately, synthetic siRNA is not easily condensed with cationic polymers due to its intrinsic rigid structure and low spatial charge density. Accordingly, the loose siRNA polyplexes inevitably expose siRNA to the extracellular environment during systemic circulation, resulting in low therapeutic efficiency and poor biodistribution. In this review, we highlight the innovative approaches to increase the size of siRNA via structural modification of the siRNA itself. The attempts include several methodologies such as hybridization, chemical polymerization, and micro- and nano-structurization of siRNA. Due to its increased charge density and flexibility, the structured siRNA can produce highly condensed and homogenous polyplexes compared to the classical monomeric siRNA. As a result, stable and compact siRNA polyplexes can enhance serum stability and target delivery efficiency in vivo with desirable biodistribution. The review specifically aims to provide the recent progress of structural modification of siRNA. In addition, the article also briefly and concisely explains the improved physico-chemical properties of structured siRNA with respect to stability, condensation ability and gene silencing efficiency.
Collapse
Affiliation(s)
- So Jin Lee
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul 136-791, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
341
|
Hinton TM, Monaghan P, Green D, Kooijmans SA, Shi S, Breheney K, Tizard M, Nicolazzo JA, Zelikin AN, Wark K. Biodistribution of polymer hydrogel capsules for the delivery of therapeutics. Acta Biomater 2012; 8:3251-60. [PMID: 22659177 DOI: 10.1016/j.actbio.2012.05.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Revised: 05/18/2012] [Accepted: 05/23/2012] [Indexed: 10/28/2022]
Abstract
A key phase in the development of intelligently designed nanoparticle delivery vehicles for new therapeutic agents is to gain an understanding of their interaction with tissues and cells. We report a series of in vitro and in vivo experiments aimed at tracking a potential delivery vehicle for therapeutic agents, including vaccine peptides and drugs derived from poly(methacrylic acid) hydrogel capsules in certain organs and cell types. For the in vitro studies, two immortal liver-derived cell lines (Huh7 and Hepa1-6) and primary cultures of mouse hepatocytes were incubated with Alexa 647 labelled fluorescent capsules to track their internalization and intracellular distribution by confocal microscopy. Capsules, 500nm in diameter, were taken up into the cells in a time-dependent manner in all three cell lines. Capsules were observed in plasma membrane-derived vesicles within the cells. After 24h a significant proportion of the capsules was observed in lysosomes. To understand the behaviour of the capsules in vivo, Alexa 488 labelled fluorescent capsules were intravenously injected into Sprague-Dawley rats and after 24h the fate of the capsules in a number of organs was determined by flow cytometry and confocal microscopy. By flow cytometry, the majority of the capsules were detected in the spleen whilst similar numbers were found in the lung and liver. By confocal microscopy, the majority of the capsules were found in the liver and spleen with significantly less capsules in the lung, heart and kidney. Colocalization of capsules with cell-type specific markers indicated that in lung, heart and kidney, the majority of the capsules were located in endothelial cells. In the spleen ~50% of the capsules were found in CD163-positive cells, whereas in the liver, almost all capsules were located in CD163-positive cells, indicating uptake by Kupffer cells. Electron microscopy confirmed the presence of capsules within Kupffer cells.
Collapse
|
342
|
Abstract
The development of nanoscale delivery vehicles for siRNAs is a current topic of considerable importance. However, little is understood about the exact trafficking mechanisms for siRNA-vehicle complexes across the plasma membrane and into the cytoplasm. While some information can be gleaned from studies on delivery of plasmid DNA, the different delivery requirements for these two vehicles makes drawing specific conclusions a challenge. However, using chemical inhibitors of different endocytosis pathways, studies on which endocytotic pathways are advantageous and deleterious for the delivery of nucleic acid drugs are emerging. Using this information as a guide, it is expected that the future development of effective siRNA delivery vehicles and therapeutics will be greatly improved.
Collapse
Affiliation(s)
- Amanda P Malefyt
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, Michigan 48824
| | | | | |
Collapse
|
343
|
Guzman-Villanueva D, El-Sherbiny IM, Herrera-Ruiz D, Vlassov AV, Smyth HDC. Formulation approaches to short interfering RNA and MicroRNA: challenges and implications. J Pharm Sci 2012; 101:4046-66. [PMID: 22927140 DOI: 10.1002/jps.23300] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Revised: 07/10/2012] [Accepted: 08/02/2012] [Indexed: 11/09/2022]
Abstract
RNA interference has emerged as a potentially powerful tool in the treatment of genetic and acquired diseases by delivering short interfering RNA (siRNA) or microRNA (miRNA) to target genes, resulting in their silencing. However, many physicochemical and biological barriers have to be overcome to obtain efficient in vivo delivery of siRNA and miRNA molecules to the organ/tissue of interest, thereby enabling their effective clinical therapy. This review discusses the challenges associated with the use of siRNA and miRNA and describes the nonviral delivery strategies used in overcoming these barriers. More specifically, emphasis has been placed on those technologies that have progressed to clinical trials for both local and systemic siRNA and miRNA delivery.
Collapse
Affiliation(s)
- Diana Guzman-Villanueva
- Division of Pharmaceutics, College of Pharmacy, The University of Texas at Austin, Texas 78712-0120, USA
| | | | | | | | | |
Collapse
|
344
|
Naito M, Ishii T, Matsumoto A, Miyata K, Miyahara Y, Kataoka K. A Phenylboronate-Functionalized Polyion Complex Micelle for ATP-Triggered Release of siRNA. Angew Chem Int Ed Engl 2012. [DOI: 10.1002/ange.201203360] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
345
|
La Fauce K, Owens L. RNA interference with special reference to combating viruses of crustacea. INDIAN JOURNAL OF VIROLOGY : AN OFFICIAL ORGAN OF INDIAN VIROLOGICAL SOCIETY 2012; 23:226-43. [PMID: 23997446 DOI: 10.1007/s13337-012-0084-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Accepted: 06/26/2012] [Indexed: 11/26/2022]
Abstract
RNA interference has evolved from being a nuisance biological phenomenon to a valuable research tool to determine gene function and as a therapeutic agent. Since pioneering observations regarding RNA interference were first reported in the 1990s from the nematode worm, plants and Drosophila, the RNAi phenomenon has since been reported in all eukaryotic organisms investigated from protozoans, plants, arthropods, fish and mammals. The design of RNAi therapeutics has progressed rapidly to designing dsRNA that can specifically and effectively silence disease related genes. Such technology has demonstrated the effective use of short interfering as therapeutics. In the absence of a B cell lineage in arthropods, and hence no long term vaccination strategy being available, the introduction of using RNA interference in crustacea may serve as an effective control and preventative measure for viral diseases for application in aquaculture.
Collapse
Affiliation(s)
- Kathy La Fauce
- Microbiology and Immunology, School of Veterinary and Biomedical Science, James Cook University, Townsville, QLD 4811 Australia
| | | |
Collapse
|
346
|
ASGR1 and ASGR2, the Genes that Encode the Asialoglycoprotein Receptor (Ashwell Receptor), Are Expressed in Peripheral Blood Monocytes and Show Interindividual Differences in Transcript Profile. Mol Biol Int 2012; 2012:283974. [PMID: 22919488 PMCID: PMC3419429 DOI: 10.1155/2012/283974] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Revised: 04/20/2012] [Accepted: 04/24/2012] [Indexed: 11/20/2022] Open
Abstract
Background. The asialoglycoprotein receptor (ASGPR) is a hepatic receptor that mediates removal of potentially hazardous glycoconjugates from blood in health and disease. The receptor comprises two proteins, asialoglycoprotein receptor 1 and 2 (ASGR1 and ASGR2), encoded by the genes ASGR1 and ASGR2. Design and Methods. Using reverse transcription amplification (RT-PCR), expression of ASGR1 and ASGR2 was investigated in human peripheral blood monocytes. Results. Monocytes were found to express ASGR1 and ASGR2 transcripts. Correctly spliced transcript variants encoding different isoforms of ASGR1 and ASGR2 were present in monocytes. The profile of transcript variants from both ASGR1 and ASGR2 differed among individuals. Transcript expression levels were compared with the hepatocyte cell line HepG2 which produces high levels of ASGPR. Monocyte transcripts were 4 to 6 orders of magnitude less than in HepG2 but nonetheless readily detectable using standard RT-PCR. The monocyte cell line THP1 gave similar results to monocytes harvested from peripheral blood, indicating it may provide a suitable model system for studying ASGPR function in this cell type. Conclusions. Monocytes transcribe and correctly process transcripts encoding the constituent proteins of the ASGPR. Monocytes may therefore represent a mobile pool of the receptor, capable of reaching sites remote from the liver.
Collapse
|
347
|
Guo P, Haque F, Hallahan B, Reif R, Li H. Uniqueness, advantages, challenges, solutions, and perspectives in therapeutics applying RNA nanotechnology. Nucleic Acid Ther 2012. [PMID: 22913595 DOI: 10.1201/b15152-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023] Open
Abstract
The field of RNA nanotechnology is rapidly emerging. RNA can be manipulated with the simplicity characteristic of DNA to produce nanoparticles with a diversity of quaternary structures by self-assembly. Additionally RNA is tremendously versatile in its function and some RNA molecules display catalytic activities much like proteins. Thus, RNA has the advantage of both worlds. However, the instability of RNA has made many scientists flinch away from RNA nanotechnology. Other concerns that have deterred the progress of RNA therapeutics include the induction of interferons, stimulation of cytokines, and activation of other immune systems, as well as short pharmacokinetic profiles in vivo. This review will provide some solutions and perspectives on the chemical and thermodynamic stability, in vivo half-life and biodistribution, yield and production cost, in vivo toxicity and side effect, specific delivery and targeting, as well as endosomal trapping and escape.
Collapse
Affiliation(s)
- Peixuan Guo
- Nanobiotechnology Center, Markey Cancer Center and Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky 40536, USA.
| | | | | | | | | |
Collapse
|
348
|
Miele E, Spinelli GP, Miele E, Di Fabrizio E, Ferretti E, Tomao S, Gulino A. Nanoparticle-based delivery of small interfering RNA: challenges for cancer therapy. Int J Nanomedicine 2012; 7:3637-57. [PMID: 22915840 PMCID: PMC3418108 DOI: 10.2147/ijn.s23696] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Indexed: 12/18/2022] Open
Abstract
During recent decades there have been remarkable advances and profound changes in cancer therapy. Many therapeutic strategies learned at the bench, including monoclonal antibodies and small molecule inhibitors, have been used at the bedside, leading to important successes. One of the most important advances in biology has been the discovery that small interfering RNA (siRNA) is able to regulate the expression of genes, by a phenomenon known as RNA interference (RNAi). RNAi is one of the most rapidly growing fields of research in biology and therapeutics. Much research effort has gone into the application of this new discovery in the treatment of various diseases, including cancer. However, even though these molecules may have potential and strong utility, some limitations make their clinical application difficult, including delivery problems, side effects due to off-target actions, disturbance of physiological functions of the cellular machinery involved in gene silencing, and induction of the innate immune response. Many researchers have attempted to overcome these limitations and to improve the safety of potential RNAi-based therapeutics. Nanoparticles, which are nanostructured entities with tunable size, shape, and surface, as well as biological behavior, provide an ideal opportunity to modify current treatment regimens in a substantial way. These nanoparticles could be designed to surmount one or more of the barriers encountered by siRNA. Nanoparticle drug formulations afford the chance to improve drug bioavailability, exploiting superior tissue permeability, payload protection, and the “stealth” features of these entities. The main aims of this review are: to explain the siRNA mechanism with regard to potential applications in siRNA-based cancer therapy; to discuss the possible usefulness of nanoparticle-based delivery of certain molecules for overcoming present therapeutic limitations; to review the ongoing relevant clinical research with its pitfalls and promises; and to evaluate critically future perspectives and challenges in siRNA-based cancer therapy.
Collapse
Affiliation(s)
- Evelina Miele
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
349
|
Abstract
The discovery of RNA interference has given a new lease on life to both the chemistry of oligonucleotides and chemical approaches for the intracellular delivery of nucleic acids. In particular, delivery of siRNA, whether in vitro for screening and target validation purposes or in humans as a new class of drugs, may revolutionize our approach to therapy. Their impact could equal that of the bioproduction and various uses of monoclonal antibodies today. Unfortunately, global pharmaceutical companies again seem to be waiting to buy the next Genentech or Genzyme of gene silencing rather than investing research and development into this promising area of research. Gene silencing encounters barriers similar to gene addition and hence may benefit from the extra decade of experience brought by gene therapy. "Chemical" transfection of cells in culture has become routine, and this Account discusses some of the reasons this success has not extended to nonviral gene therapy trials, most of which do not progress beyond the phase 2 stage. The author also discusses a (much debated) mechanism of nucleic acid cell entry and subsequent release of the polycationic particles into the cytoplasm. Both topics should be useful to those interested in delivery of siRNA. The move from gene therapy toward siRNA as an oligonucleotide-based therapy strategy provides a much wider range of druggable targets. Even though these molecules are a hundredfold smaller than a gene, they are delivered via similar cellular mechanisms. Their complexes with cationic polymers are less stable than those with a higher number of phosphate groups, which may be compensated by siRNA concatemerization or by chemical conjugation with the cationic carrier. Thus chemistry is again desperately needed.
Collapse
Affiliation(s)
- Jean-Paul Behr
- Chimie Génétique, Université de Strasbourg, route du Rhin, 67401 Illkirch, and Polyplus-transfection SA, Bioparc, 67400 Illkirch, France
| |
Collapse
|
350
|
Wagner E. Polymers for siRNA delivery: inspired by viruses to be targeted, dynamic, and precise. Acc Chem Res 2012; 45:1005-13. [PMID: 22191535 DOI: 10.1021/ar2002232] [Citation(s) in RCA: 251] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Synthetic small interfering RNA (siRNA) presents an exciting novel medical opportunity. Although researchers agree that siRNA could have a great therapeutic impact, the required extracellular and intracellular delivery of these molecules into the disease-associated target cells presents the primary roadblock for the broader translation of these molecules into medicines. Thus, the design of adequate delivery technologies has utmost importance. Viruses are natural masterpieces of nucleic acid delivery and present chemists and drug delivery experts with a template for the design of artificial carriers for synthetic nucleic acids such as siRNA. They have been developed into gene vectors and have provided convincing successes in gene therapy. Optimized by biological evolution, viruses are programmed to be dynamic and bioresponsive as they enter living cells, and they carry out their functions in a precisely defined sequence. However, because they are synthesized within living cells and with naturally available nucleotides and amino acids, the chemistry of viruses is limited. With the use of diverse synthetic molecules and macromolecules, chemists can provide delivery solutions beyond the scope of the natural evolution of viruses. This Account describes the design and synthesis of "synthetic siRNA viruses." These structures contain elements that mimic the delivery functions of viral particles and surface domains that shield against undesired biological interactions and enable specific host cell receptor binding through the presentation of multiple targeting ligands. For example, cationic polymers can reversibly package one or more siRNA molecules into nanoparticle cores to protect them against a degradative bioenvironment. After internalization by receptor-mediated endocytosis into the acidifying endosomes of cells, synthetic siRNA can escape from these vesicles through the activation of membrane-disruption domains as viruses do and reach the cytoplasm, the location of RNA interference. This multistep task presents an attractive challenge for chemists. Similar to the design of prodrugs, the functional domains of these systems have to be activated in a dynamic mode, triggered by conformational changes or bond cleavages in the relevant microenvironment such as the acidic endosome or disulfide-reducing cytoplasm. These chemical analogues of viral domains are often synthetically simpler and more easily accessible molecules than viral proteins. Their precise assembly into multifunctional macromolecular and supramolecular structures is facilitated by improved analytical techniques, precise orthogonal conjugation chemistries, and sequence-defined polymer syntheses. The chemical evolution of microdomains using chemical libraries and macromolecular and supramolecular evolution could provide key strategies for optimizing siRNA carriers to selected medical indications.
Collapse
Affiliation(s)
- Ernst Wagner
- Pharmaceutical Biotechnology, Center for System-Based Drug Research, and Center for Nanoscience (CeNS), Ludwig-Maximilians-Universität München, D-81377 Munich, Germany
| |
Collapse
|