1
|
Moazzam M, Zhang M, Hussain A, Yu X, Huang J, Huang Y. The landscape of nanoparticle-based siRNA delivery and therapeutic development. Mol Ther 2024; 32:284-312. [PMID: 38204162 PMCID: PMC10861989 DOI: 10.1016/j.ymthe.2024.01.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 10/01/2023] [Accepted: 01/05/2024] [Indexed: 01/12/2024] Open
Abstract
Five small interfering RNA (siRNA)-based therapeutics have been approved by the Food and Drug Administration (FDA), namely patisiran, givosiran, lumasiran, inclisiran, and vutrisiran. Besides, siRNA delivery to the target site without toxicity is a big challenge for researchers, and naked-siRNA delivery possesses several challenges, including membrane impermeability, enzymatic degradation, mononuclear phagocyte system (MPS) entrapment, fast renal excretion, endosomal escape, and off-target effects. The siRNA therapeutics can silence any disease-specific gene, but their intracellular and extracellular barriers limit their clinical applications. For this purpose, several modifications have been employed to siRNA for better transfection efficiency. Still, there is a quest for better delivery systems for siRNA delivery to the target site. In recent years, nanoparticles have shown promising results in siRNA delivery with minimum toxicity and off-target effects. Patisiran is a lipid nanoparticle (LNP)-based siRNA formulation for treating hereditary transthyretin-mediated amyloidosis that ultimately warrants the use of nanoparticles from different classes, especially lipid-based nanoparticles. These nanoparticles may belong to different categories, including lipid-based, polymer-based, and inorganic nanoparticles. This review briefly discusses the lipid, polymer, and inorganic nanoparticles and their sub-types for siRNA delivery. Finally, several clinical trials related to siRNA therapeutics are addressed, followed by the future prospects and conclusions.
Collapse
Affiliation(s)
- Muhammad Moazzam
- Faculty of Engineering and Science, University of Greenwich, Medway Campus, Chatham Maritime, Kent ME4 4TB, UK
| | - Mengjie Zhang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Abid Hussain
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Xiaotong Yu
- Department of Immunology, School of Basic Medical Sciences, Key Laboratory of Medical Immunology of Ministry of Health, Peking University, Beijing 100191, China.
| | - Jia Huang
- Department of Hepatobiliary Surgery, China-Japan Friendship Hospital, Beijing 100029, China.
| | - Yuanyu Huang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing 100081, China; Rigerna Therapeutics Co. Ltd., Suzhou 215127, China.
| |
Collapse
|
2
|
Vetter VC, Wagner E. Targeting nucleic acid-based therapeutics to tumors: Challenges and strategies for polyplexes. J Control Release 2022; 346:110-135. [PMID: 35436520 DOI: 10.1016/j.jconrel.2022.04.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/07/2022] [Accepted: 04/10/2022] [Indexed: 12/18/2022]
Abstract
The current medical reality of cancer gene therapy is reflected by more than ten approved products on the global market, including oncolytic and other viral vectors and CAR T-cells as ex vivo gene-modified cell therapeutics. The development of synthetic antitumoral nucleic acid therapeutics has been proceeding at a lower but steady pace, fueled by a plethora of alternative nucleic acid platforms (from various antisense oligonucleotides, siRNA, microRNA, lncRNA, sgRNA, to larger mRNA and DNA) and several classes of physical and chemical delivery technologies. This review summarizes the challenges and strategies for tumor-targeted nucleic acid delivery. Focusing primarily on polyplexes (polycation complexes) as nanocarriers, delivery options across multiple barriers into tumor cells are illustrated.
Collapse
Affiliation(s)
- Victoria C Vetter
- Pharmaceutical Biotechnology, Center for System-based Drug Research, Ludwig-Maximilians-Universität, Munich 81377, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Center for System-based Drug Research, Ludwig-Maximilians-Universität, Munich 81377, Germany; Center for NanoScience (CeNS), Ludwig-Maximilians-Universität, Munich 81377, Germany.
| |
Collapse
|
3
|
Benli-Hoppe T, Göl Öztürk Ş, Öztürk Ö, Berger S, Wagner E, Yazdi M. Transferrin Receptor Targeted Polyplexes Completely Comprised of Sequence-Defined Components. Macromol Rapid Commun 2021; 43:e2100602. [PMID: 34713524 DOI: 10.1002/marc.202100602] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 10/20/2021] [Indexed: 11/08/2022]
Abstract
Human transferrin protein (Tf) modified polyplexes have already displayed encouraging potential for receptor-mediated nucleic acid delivery into tumors. The use of a blood-derived targeting protein and polydisperse macromolecular cationic subunits however presents a practical challenge for pharmaceutical grade production. Here, Tf receptor (TfR) targeted small interfering RNA (siRNA) polyplexes are designed that are completely composed of synthetic, monodisperse, and sequence-defined subunits generated by solid-phase supported synthesis. An optimized cationizable lipo-oligoaminoamide (lipo-OAA) is used for siRNA core polyplex formation, and a retro-enantio peptide (reTfR) attached via a monodisperse polyethylene glycol (PEG) spacer via click chemistry is applied for targeting. Improved gene silencing is demonstrated in TfR-expressing KB and DU145 cells. Analogous plasmid DNA (pDNA) polyplexes are successfully used for receptor-mediated gene delivery in TfR-rich K562 cells and Neuro2a cells. Six lipo-OAAs differing in their lipidic domain and redox-sensitive attachment of lipid residues are tested in order to evaluate the impact of core polyplex stability on receptor-dependent gene transfer.
Collapse
Affiliation(s)
- Teoman Benli-Hoppe
- Pharmaceutical Biotechnology, Center for Drug Research, and Center for Nanoscience, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, Munich, 81377, Germany
| | - Şurhan Göl Öztürk
- Pharmaceutical Biotechnology, Center for Drug Research, and Center for Nanoscience, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, Munich, 81377, Germany
| | - Özgür Öztürk
- Pharmaceutical Biotechnology, Center for Drug Research, and Center for Nanoscience, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, Munich, 81377, Germany
| | - Simone Berger
- Pharmaceutical Biotechnology, Center for Drug Research, and Center for Nanoscience, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, Munich, 81377, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Center for Drug Research, and Center for Nanoscience, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, Munich, 81377, Germany
| | - Mina Yazdi
- Pharmaceutical Biotechnology, Center for Drug Research, and Center for Nanoscience, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, Munich, 81377, Germany
| |
Collapse
|
4
|
Xia X, Pollock N, Zhou J, Rossi J. Tissue-Specific Delivery of Oligonucleotides. Methods Mol Biol 2020; 2036:17-50. [PMID: 31410789 DOI: 10.1007/978-1-4939-9670-4_2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
From the initial discovery of short-interfering RNA (siRNA) and antisense oligonucleotides for specific gene knockdown at the posttranscriptional level to the current CRISPR-Cas9 system offering gene editing at the genomic level, oligonucleotides, in addition to their biological functions in storing and conveying genetic information, provide the most prominent solutions to targeted gene therapies. Nonetheless, looking into the future of curing cancer and acute diseases, researchers are only cautiously optimistic as the cellular delivery of these polyanionic biomacromolecules is still the biggest hurdle for their therapeutic realization. To overcome the delivery obstacle, oligonucleotides have been encapsulated within or conjugated with delivery vehicles for enhanced membrane penetration, improved payload, and tissue-specific delivery. Such delivery systems include but not limited to virus-based vehicles, gold-nanoparticle vehicles, formulated liposomes, and synthetic polymers. In this chapter, delivery challenges imposed by biological barriers are briefly discussed; followed by recent advances in tissue-specific oligonucleotide delivery utilizing both viral and nonviral delivery vectors, discussing their advantages, and how judicious design and formulation could improve and expand their potential as delivery vehicles.
Collapse
Affiliation(s)
- Xin Xia
- Department of Molecular and Cellular Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Nicolette Pollock
- Department of Molecular and Cellular Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Jiehua Zhou
- Department of Molecular and Cellular Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - John Rossi
- Department of Molecular and Cellular Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA.
| |
Collapse
|
5
|
Kopeina GS, Prokhorova EA, Lavrik IN, Zhivotovsky B. Alterations in the nucleocytoplasmic transport in apoptosis: Caspases lead the way. Cell Prolif 2018; 51:e12467. [PMID: 29947118 DOI: 10.1111/cpr.12467] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 04/19/2018] [Indexed: 12/13/2022] Open
Abstract
Apoptosis is a mode of regulated cell death that is indispensable for the morphogenesis, development and homeostasis of multicellular organisms. Caspases are cysteine-dependent aspartate-specific proteases, which function as initiators and executors of apoptosis. Caspases are cytosolic proteins that can cleave substrates located in different intracellular compartments during apoptosis. Many years ago, the involvement of caspases in the regulation of nuclear changes, a hallmark of apoptosis, was documented. Accumulated data suggest that apoptosis-associated alterations in nucleocytoplasmic transport are also linked to caspase activity. Here, we aim to discuss the current state of knowledge regarding this process. Particular attention will be focused on caspase nuclear entry and their functions in the demolition of the nucleus upon apoptotic stimuli.
Collapse
Affiliation(s)
- Gelina S Kopeina
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | | | - Inna N Lavrik
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia.,Translational Inflammation Research, Medical Faculty, Otto von Guericke University, Magdeburg, Germany
| | - Boris Zhivotovsky
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia.,Division of Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
6
|
Alamoudi K, Martins P, Croissant JG, Patil S, Omar H, Khashab NM. Thermoresponsive pegylated bubble liposome nanovectors for efficient siRNA delivery via endosomal escape. Nanomedicine (Lond) 2017; 12:1421-1433. [PMID: 28524721 DOI: 10.2217/nnm-2017-0021] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
AIM Improving the delivery of siRNA into cancer cells via bubble liposomes. Designing a thermoresponsive pegylated liposome through the introduction of ammonium bicarbonate salt into liposomes so as to control their endosomal escape for gene therapy. METHODS A sub-200 nm nanovector was fully characterized and examined for cellular uptake, cytotoxicity, endosomal escape and gene silencing. RESULTS The siRNA-liposomes were internalized into cancer cells within 5 min and then released siRNAs in the cytosol prior to lysosomal degradation upon external temperature elevation. This was confirmed by confocal bioimaging and gene silencing reaching up to 90% and further demonstrated by the protein inhibition of both target genes. CONCLUSION The thermoresponsiveness of ammonium bicarbonate containing liposomes enabled the rapid endosomal escape of the particles and resulted in an efficient gene silencing.
Collapse
Affiliation(s)
- Kholod Alamoudi
- Smart Hybrid Materials Laboratory, Advanced Membranes & Porous Materials Center, King Abdullah University of Science & Technology, Thuwal, Saudi Arabia
| | - Patricia Martins
- Smart Hybrid Materials Laboratory, Advanced Membranes & Porous Materials Center, King Abdullah University of Science & Technology, Thuwal, Saudi Arabia
| | - Jonas G Croissant
- Smart Hybrid Materials Laboratory, Advanced Membranes & Porous Materials Center, King Abdullah University of Science & Technology, Thuwal, Saudi Arabia
| | - Sachin Patil
- Smart Hybrid Materials Laboratory, Advanced Membranes & Porous Materials Center, King Abdullah University of Science & Technology, Thuwal, Saudi Arabia
| | - Haneen Omar
- Smart Hybrid Materials Laboratory, Advanced Membranes & Porous Materials Center, King Abdullah University of Science & Technology, Thuwal, Saudi Arabia
| | - Niveen M Khashab
- Smart Hybrid Materials Laboratory, Advanced Membranes & Porous Materials Center, King Abdullah University of Science & Technology, Thuwal, Saudi Arabia
| |
Collapse
|
7
|
Chen J, Guan X, Hu Y, Tian H, Chen X. Peptide-Based and Polypeptide-Based Gene Delivery Systems. Top Curr Chem (Cham) 2017; 375:32. [DOI: 10.1007/s41061-017-0115-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 01/28/2017] [Indexed: 12/15/2022]
|
8
|
Zhang W, Müller K, Kessel E, Reinhard S, He D, Klein PM, Höhn M, Rödl W, Kempter S, Wagner E. Targeted siRNA Delivery Using a Lipo-Oligoaminoamide Nanocore with an Influenza Peptide and Transferrin Shell. Adv Healthc Mater 2016; 5:1493-504. [PMID: 27109317 DOI: 10.1002/adhm.201600057] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 02/23/2016] [Indexed: 01/06/2023]
Abstract
Developing RNA-interference-based therapeutic approaches with efficient and targeted cytosolic delivery of small interfering RNA (siRNA) is remaining a critical challenge since two decades. Herein, a multifunctional transferrin receptor (TfR)-targeted siRNA delivery system (Tf&INF7) is designed based on siRNA complexes formed with the cationic lipo-oligoamino amide 454, sequentially surface-modified with polyethylene glycol-linked transferrin (Tf) for receptor targeting and the endosomolytic peptide INF7 for efficient cytosolic release of the siRNA. Effective Tf&INF7 polyplex internalization and target gene silencing are demonstrated for the TfR overexpressing tumor cell lines (K562, D145, and N2a). Treatment with antitumoral EG5 siRNA results in a block of tumor cell growth and triggered apoptosis. Tf-modified polyplexes are far more effective than the corresponding albumin- (Alb) or nonmodified 454 polyplexes. Competition experiments with excess of Tf demonstrate TfR target specificity. As alternative to the ligand Tf, an anti-murine TfR antibody is incorporated into the polyplexes for specific targeting and gene silencing in the murine N2a cell line. In vivo distribution studies not only demonstrate an enhanced tumor residence of siRNA in N2a tumor-bearing mice with the Tf&INF7 as compared to the 454 polyplex group but also a reduced siRNA nanoparticle stability limiting the in vivo performance.
Collapse
Affiliation(s)
- Wei Zhang
- Pharmaceutical Biotechnology Department of Pharmacy Ludwig‐Maximilians‐Universität München (LMU) Butenandtstrasse 5‐13 D‐81377 Munich Germany
| | - Katharina Müller
- Pharmaceutical Biotechnology Department of Pharmacy Ludwig‐Maximilians‐Universität München (LMU) Butenandtstrasse 5‐13 D‐81377 Munich Germany
| | - Eva Kessel
- Pharmaceutical Biotechnology Department of Pharmacy Ludwig‐Maximilians‐Universität München (LMU) Butenandtstrasse 5‐13 D‐81377 Munich Germany
- Nanosystems Initiative Munich Schellingstrasse 4 D‐80799 Munich Germany
| | - Sören Reinhard
- Pharmaceutical Biotechnology Department of Pharmacy Ludwig‐Maximilians‐Universität München (LMU) Butenandtstrasse 5‐13 D‐81377 Munich Germany
| | - Dongsheng He
- Pharmaceutical Biotechnology Department of Pharmacy Ludwig‐Maximilians‐Universität München (LMU) Butenandtstrasse 5‐13 D‐81377 Munich Germany
- Nanosystems Initiative Munich Schellingstrasse 4 D‐80799 Munich Germany
| | - Philipp M. Klein
- Pharmaceutical Biotechnology Department of Pharmacy Ludwig‐Maximilians‐Universität München (LMU) Butenandtstrasse 5‐13 D‐81377 Munich Germany
| | - Miriam Höhn
- Pharmaceutical Biotechnology Department of Pharmacy Ludwig‐Maximilians‐Universität München (LMU) Butenandtstrasse 5‐13 D‐81377 Munich Germany
| | - Wolfgang Rödl
- Pharmaceutical Biotechnology Department of Pharmacy Ludwig‐Maximilians‐Universität München (LMU) Butenandtstrasse 5‐13 D‐81377 Munich Germany
| | - Susanne Kempter
- Department of Physics Ludwig‐Maximilians‐Universität München Geschwister‐Scholl‐Platz 1 80539 Munich Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology Department of Pharmacy Ludwig‐Maximilians‐Universität München (LMU) Butenandtstrasse 5‐13 D‐81377 Munich Germany
- Nanosystems Initiative Munich Schellingstrasse 4 D‐80799 Munich Germany
| |
Collapse
|
9
|
Kumar MD, Dravid A, Kumar A, Sen D. Gene therapy as a potential tool for treating neuroblastoma-a focused review. Cancer Gene Ther 2016; 23:115-24. [PMID: 27080224 DOI: 10.1038/cgt.2016.16] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 03/14/2016] [Accepted: 03/15/2016] [Indexed: 12/12/2022]
Abstract
Neuroblastoma, a solid tumor caused by rapid division of undifferentiated neuroblasts, is the most common childhood malignancy affecting children aged <5 years. Several approaches and strategies developed and tested to cure neuroblastoma have met with limited success due to different reasons. Many oncogenes are deregulated during the onset and development of neuroblastoma and thus offer an opportunity to circumvent this disease if the expression of these genes is restored to normalcy. Gene therapy is a powerful tool with the potential to inhibit the deleterious effects of oncogenes by inserting corrected/normal genes into the genome. Both viral and non-viral vector-based gene therapies have been developed and adopted to deliver the target genes into neuroblastoma cells. These attempts have given hope to bringing in a new regime of treatment against neuroblastoma. A few gene-therapy-based treatment strategies have been tested in limited clinical trials yielding some positive results. This mini review is an attempt to provide an overview of the available options of gene therapy to treat neuroblastoma.
Collapse
Affiliation(s)
- M D Kumar
- School of Biosciences and Technology, Vellore Institute of Technology University, Vellore, Tamil Nadu, India
| | - A Dravid
- School of Biosciences and Technology, Vellore Institute of Technology University, Vellore, Tamil Nadu, India
| | - A Kumar
- School of Biosciences and Technology, Vellore Institute of Technology University, Vellore, Tamil Nadu, India
| | - D Sen
- School of Biosciences and Technology, Vellore Institute of Technology University, Vellore, Tamil Nadu, India.,Cellular and Molecular Therapeutics Laboratory, Centre for Biomaterials, Cellular and Molecular Theranostics, Vellore Institute of Technology University, Vellore, Tamil Nadu, India
| |
Collapse
|
10
|
Frère A, Kawalec M, Tempelaar S, Peixoto P, Hendrick E, Peulen O, Evrard B, Dubois P, Mespouille L, Mottet D, Piel G. Impact of the Structure of Biocompatible Aliphatic Polycarbonates on siRNA Transfection Ability. Biomacromolecules 2015; 16:769-79. [DOI: 10.1021/bm501676p] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
| | - Michal Kawalec
- Laboratory
of Polymeric and Composite Materials, Center of Innovation and Research
in Materials and Polymers (CIRMAP), Research Institute for Health
Sciences and Technology, University of Mons, Place du Parc 20 - 7000, Mons, Belgium
| | - Sarah Tempelaar
- Laboratory
of Polymeric and Composite Materials, Center of Innovation and Research
in Materials and Polymers (CIRMAP), Research Institute for Health
Sciences and Technology, University of Mons, Place du Parc 20 - 7000, Mons, Belgium
| | | | | | | | | | - Philippe Dubois
- Laboratory
of Polymeric and Composite Materials, Center of Innovation and Research
in Materials and Polymers (CIRMAP), Research Institute for Health
Sciences and Technology, University of Mons, Place du Parc 20 - 7000, Mons, Belgium
| | - Laetitia Mespouille
- Laboratory
of Polymeric and Composite Materials, Center of Innovation and Research
in Materials and Polymers (CIRMAP), Research Institute for Health
Sciences and Technology, University of Mons, Place du Parc 20 - 7000, Mons, Belgium
| | | | | |
Collapse
|
11
|
Jia H, Aw W, Saito K, Hanate M, Hasebe Y, Kato H. Eggshell membrane ameliorates hepatic fibrogenesis in human C3A cells and rats through changes in PPARγ-Endothelin 1 signaling. Sci Rep 2014; 4:7473. [PMID: 25503635 PMCID: PMC5378949 DOI: 10.1038/srep07473] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 11/20/2014] [Indexed: 12/22/2022] Open
Abstract
Our previous nutrigenomic findings indicate that eggshell membrane (ESM) may prevent liver fibrosis. Here we investigated the effects and mechanisms underlying ESM intervention against liver injury by using DNA microarray analysis and comparative proteomics. In vitro hydrolyzed ESM attenuated the TGFβ1-induced procollagen production of human hepatocyte C3A cells and inhibited the expression of Endothelin 1 (EDN1) and its two receptors, and extracellular matrix components. In vivo male Wistar rats were allocated into a normal control group, a CCl4 group (hypodermic injection of 50% CCl4 2×/wk) and an ESM group (20 g ESM/kg diet with CCl4 injection) for 7 wks. Dietary ESM ameliorated the elevated activity of ALT/AST, oxidative stress and collagen accumulation in liver, accompanied by the down-regulated expression of Edn1 signaling and notable profibrogenic genes and growth factors as well as peroxisome proliferator-activated receptor gamma (PPARγ). Concomitantly, the decreased expressions of Galectin-1 and Desmin protein in the ESM group indicated the deactivation of hepatic stellate cells (HSCs). Through a multifaceted integrated omics approach, we have demonstrated that ESM can exert an antifibrotic effect by suppressing oxidative stress and promoting collagen degradation by inhibiting HSCs' transformation, potentially via a novel modulation of the PPARγ-Endothelin 1 interaction signaling pathway.
Collapse
Affiliation(s)
- Huijuan Jia
- Corporate Sponsored Research Program "Food for Life, " Organization for Interdisciplinary Research Projects, The University of Tokyo, Tokyo, Japan
| | - Wanping Aw
- Institute of Advanced Biosciences, Keio University, Yamagata, Japan
| | - Kenji Saito
- Corporate Sponsored Research Program "Food for Life, " Organization for Interdisciplinary Research Projects, The University of Tokyo, Tokyo, Japan
| | - Manaka Hanate
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | | | - Hisanori Kato
- 1] Corporate Sponsored Research Program "Food for Life, " Organization for Interdisciplinary Research Projects, The University of Tokyo, Tokyo, Japan [2] Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
12
|
Abstract
RNA interference (RNAi) therapeutics appear to offer substantial opportunities for future therapy. However, post-administration RNAi effectors are typically unable to reach disease target cells in vivo without the assistance of a delivery system or vector. The main focus of this review is on lipid-based nanoparticle (LNP) delivery systems in current research and development that have at least been shown to act as effective delivery systems for functional delivery of RNAi effectors to disease target cells in vivo. The potential utility of these LNP delivery systems is growing rapidly, and LNPs are emerging as the preferred synthetic delivery systems in preclinical studies and current nonviral RNAi effector clinical trials. Moreover, studies on LNP-mediated delivery in vivo are leading to the emergence of useful biophysical parameters and physical organic chemistry rules that provide a framework for understanding in vivo delivery behaviors and outcomes. These same parameters and rules should also suggest ways and means to develop next generations of LNPs with genuine utility and long-term clinical viability.
Collapse
Affiliation(s)
- Andrew D Miller
- Institute of Pharmaceutical Science, King's College London, Franklin-Wilkins Building, Waterloo Campus, 150 Stamford Street, London SE1 9NH , UK and GlobalAcorn Limited , London , UK
| |
Collapse
|
13
|
Hsc70 contributes to cancer cell survival by preventing Rab1A degradation under stress conditions. PLoS One 2014; 9:e96785. [PMID: 24801886 PMCID: PMC4011886 DOI: 10.1371/journal.pone.0096785] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 04/11/2014] [Indexed: 12/20/2022] Open
Abstract
Heat shock cognate protein 70 (Hsc70) acts as a molecular chaperone for the maintenance of intracellular proteins, which allows cancer cells to survive under proteotoxic stress. We attempted to use Hsc70 to identify key molecules in cancer cell survival. Here, we performed mass-spectrometry-based proteomics analysis utilizing affinity purification with anti-Hsc70 antibodies; as a result, 83 differentially expressed proteins were identified under stress conditions. This result implies that there was a change in the proteins with which Hsc70 interacted in response to stress. Among the proteins identified under both serum-depleted and 5-fluorouracil-treated conditions, Rab1A was identified as an essential molecule for cancer cell survival. Hsc70 interacted with Rab1A in a chaperone-dependent manner. In addition, Hsc70 knockdown decreased the level of Rab1A and increased the level of its ubiquitination under stress conditions, suggesting that Hsc70 prevented the degradation of Rab1A denatured by stress exposure. We also found that Rab1A knockdown induced cell death by inhibition of autophagosome formation. Rab1A may therefore contribute to overcoming proteotoxic insults, which allows cancer cells to survive under stress conditions. Analysis of Hsc70 interactors provided insight into changes of intracellular status. We expect further study of the Hsc70 interactome to provide a more comprehensive understanding of cancer cell physiology.
Collapse
|
14
|
Abstract
Small non-coding RNA (ncRNA) therapeutics make use of small ncRNA effectors for desired therapeutic purposes that are essentially short (10–20 kD) RNA segments. These small ncRNA effectors are potentially tremendously powerful therapeutic agents, but are typically unable to reach disease target cells in vivo without the assistance of a delivery system or vector. The main focus of this review is the use of lipid-based nanoparticles (LNPs) for the functional delivery of small ncRNA effectors in vivo. LNPs appear to be amongst the most effective delivery systems currently available for this purpose. Moreover, studies on LNP-mediated delivery in vivo are leading to the emergence of useful biophysical parameters and physical organic chemistry rules that provide a framework for understanding LNP-mediated in vivo delivery behaviors and outcomes. These same parameters and rules should also suggest ways and means to develop next generations of LNPs with genuine utility and long-term clinical viability.
Collapse
|
15
|
Abstract
For the last five decades cationic polymers have been used for nucleic acids transfection. Our understanding of polymer-nucleic acid interactions and their rational use in delivery has continuously increased. The great improvements in macromolecular chemistry and the recognition of distinct biological extra- and intracellular delivery hurdles triggered several breakthrough developments, including the discovery of natural and synthetic polycations for compaction of nucleic acids into stable nanoparticles termed polyplexes; the incorporation of targeting ligands and surface-shielding of polyplexes to enable receptor-mediated gene delivery into defined target tissues; and strongly improved intracellular transfer efficacy by better endosomal escape of vesicle-trapped polyplexes into the cytosol. These experiences triggered the development of second-generation polymers with more dynamic properties, such as endosomal pH-responsive release mechanisms, or biodegradable units for improved biocompatibility and intracellular release of the nucleic acid pay load. Despite a better biological understanding, significant challenges such as efficient nuclear delivery and persistence of gene expression persist. The therapeutic perspectives widened from pDNA-based gene therapy to application of novel therapeutic nucleic acids including mRNA, siRNA, and microRNA. The finding that different therapeutic pay loads require different tailor-made carriers complicates preclinical developments. Convincing evidence of medical efficacy still remains to be demonstrated. Bioinspired multifunctional polyplexes resembling "synthetic viruses" appear as attractive opportunity, but provide additional challenges: how to identify optimum combinations of functional delivery units, and how to prepare such polyplexes reproducibly in precise form? Design of sequence-defined polymers, screening of combinatorial polymer and polyplex libraries are tools for further chemical evolution of polyplexes.
Collapse
Affiliation(s)
- Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-University Munich, and Nanosystems Initiative Munich (NIM), Munich, Germany
| |
Collapse
|
16
|
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
|
17
|
Resnier P, Montier T, Mathieu V, Benoit JP, Passirani C. A review of the current status of siRNA nanomedicines in the treatment of cancer. Biomaterials 2013; 34:6429-43. [PMID: 23727262 DOI: 10.1016/j.biomaterials.2013.04.060] [Citation(s) in RCA: 162] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 04/27/2013] [Indexed: 12/11/2022]
Abstract
RNA interference currently offers new opportunities for gene therapy by the specific extinction of targeted gene(s) in cancer diseases. However, the main challenge for nucleic acid delivery still remains its efficacy through intravenous administration. Over the last decade, many delivery systems have been developed and optimized to encapsulate siRNA and to specifically promote their delivery into tumor cells and improve their pharmacokinetics for anti-cancer purposes. This review aims to sum up the potential targets in numerous pathways and the properties of recently optimized siRNA synthetic nanomedicines with their preclinical applications and efficacy. Future perspectives in cancer treatment are discussed including promising concomitant treatment with chemotherapies or other siRNA. The outcomes in human clinical trials are also presented.
Collapse
|
18
|
Edinger D, Kläger R, Troiber C, Dohmen C, Wagner E. Gene silencing and antitumoral effects of Eg5 or Ran siRNA oligoaminoamide polyplexes. Drug Deliv Transl Res 2013; 4:84-95. [DOI: 10.1007/s13346-013-0146-y] [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]
|
19
|
|
20
|
Järver P, Coursindel T, Andaloussi SEL, Godfrey C, Wood MJA, Gait MJ. Peptide-mediated Cell and In Vivo Delivery of Antisense Oligonucleotides and siRNA. MOLECULAR THERAPY. NUCLEIC ACIDS 2012; 1:e27. [PMID: 23344079 PMCID: PMC3390225 DOI: 10.1038/mtna.2012.18] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Revised: 04/26/2012] [Accepted: 04/26/2012] [Indexed: 12/22/2022]
Affiliation(s)
- Peter Järver
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | | | - Samir EL Andaloussi
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
- Department of Laboratory Medicine, Karolinska Institute, Hudidnge, Sweden
| | - Caroline Godfrey
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Matthew JA Wood
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Michael J Gait
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| |
Collapse
|
21
|
Cationic albumin nanoparticles for enhanced drug delivery to treat breast cancer: preparation and in vitro assessment. JOURNAL OF DRUG DELIVERY 2011; 2012:686108. [PMID: 22187654 PMCID: PMC3236506 DOI: 10.1155/2012/686108] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Accepted: 09/10/2011] [Indexed: 11/20/2022]
Abstract
Most anticancer drugs are greatly limited by the serious side effects that they cause. Doxorubicin (DOX) is an antineoplastic agent, commonly used against breast cancer. However, it may lead to irreversible cardiotoxicity, which could even result in congestive heart failure. In order to avoid these harmful side effects to the patients and to improve the therapeutic efficacy of doxorubicin, we developed DOX-loaded polyethylenimine- (PEI-) enhanced human serum albumin (HSA) nanoparticles. The formed nanoparticles were ~137 nm in size with a surface zeta potential of ~+15 mV, prepared using 20 μg of PEI added per mg of HSA. Cytotoxicity was not observed with empty PEI-enhanced HSA nanoparticles, formed with low-molecular weight (25 kDa) PEI, indicating biocompatibility and safety of the nanoparticle formulation. Under optimized transfection conditions, approximately 80% of cells were transfected with HSA nanoparticles containing tetramethylrhodamine-conjugated bovine serum albumin. Conclusively, PEI-enhanced HSA nanoparticles show potential for developing into an effective carrier for anticancer drugs.
Collapse
|
22
|
Verissimo CS, Molenaar JJ, Fitzsimons CP, Vreugdenhil E. Neuroblastoma therapy: what is in the pipeline? Endocr Relat Cancer 2011; 18:R213-31. [PMID: 21971288 DOI: 10.1530/erc-11-0251] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Despite the expansion of knowledge about neuroblastoma (NB) in recent years, the therapeutic outcome for children with a high-risk NB has not significantly improved. Therefore, more effective therapies are needed. This might be achieved by aiming future efforts at recently proposed but not yet developed targets for NB therapy. In this review, we discuss the recently proposed molecular targets that are in clinical trials and, in particular, those that are not yet explored in the clinic. We focus on the selection of these molecular targets for which promising in vitro and in vivo results have been obtained by silencing/inhibiting them. In addition, these selected targets are involved at least in one of the NB tumorigenic processes: proliferation, anti-apoptosis, angiogenesis and/or metastasis. In particular, we will review a recently proposed target, the microtubule-associated proteins (MAPs) encoded by doublecortin-like kinase gene (DCLK1). DCLK1-derived MAPs are crucial for proliferation and survival of neuroblasts and are highly expressed not only in NB but also in other tumours such as gliomas. Additionally, we will discuss neuropeptide Y, its Y2 receptor and cathepsin L as examples of targets to decrease angiogenesis and metastasis of NB. Furthermore, we will review the micro-RNAs that have been proposed as therapeutic targets for NB. Detailed investigation of these not yet developed targets as well as exploration of multi-target approaches might be the key to a more effective NB therapy, i.e. increasing specificity, reducing toxicity and avoiding long-term side effects.
Collapse
Affiliation(s)
- Carla S Verissimo
- Division of Medical Pharmacology, Leiden/Amsterdam Center for Drug Research, Leiden University Medical Center, Gorlaeus Laboratories, The Netherlands
| | | | | | | |
Collapse
|
23
|
Steele TWJ, Zhao X, Tarcha P, Kissel T. Factors influencing polycation/siRNA colloidal stability toward aerosol lung delivery. Eur J Pharm Biopharm 2011; 80:14-24. [PMID: 21924355 DOI: 10.1016/j.ejpb.2011.08.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2011] [Revised: 08/24/2011] [Accepted: 08/31/2011] [Indexed: 01/25/2023]
Abstract
Hexanediol diacrylate cross-linked oligoethylenimine (OEI-HD) is a non-viral polymeric vector designed to deliver siRNA. To achieve safe and effective in vivo siRNA delivery using this vector, the polyplex must have sufficient colloidal stability if administered intravenously or nebulized for delivery by the pulmonary route. In this study, polyplexes from OEI-HD and siRNA were formulated for aerosol-based lung delivery, regarding their colloidal stability, optimal particle size, and in vitro biological activity. Herein, we describe how these properties are dependent upon the polymer-to siRNA weight ratios, buffer composition they were complexed in, PEG-grafting, and the addition of commercial lung surfactants and/or non-ionic surfactants to the formulation. Lastly, the effects of nebulization of the formulation into aerosol droplets, on the polyplex particle size and transfection efficiency, were evaluated. Polyplex size was monitored for up to 2 h after polyplex formation to determine the extent of aggregation and final particle sizes when stability was achieved. Our results suggest that PEG-grafting and polyethylenimine-PEG mixing were effective in achieving colloidal stability in isotonic saline buffers. In addition, colloidal stability was achieved in isotonic glucose buffers using commercially available non-ionic surfactant Pluronic™ P68 or the lung-derived surfactant Alveofact™. The smallest particle size, 140 nm, was obtained with Pluronic™ F68. For transfection efficiency, both Alveofact™ and Pluronic™ F68 achieved equal or better transfection when added to the OEI-HD/siRNA polyplexes. For long term storage of OEI-HD/siRNA formulations, we propose a lyophilization method that created in situ polyplexes upon addition of water. Preparation of OEI-HD/siRNA polyplexes by this method allowed dry storage at room temperature for up to the 3 months. In conclusion, we have identified approaches to achieve formulation and colloidal stability of OEI-HD/siRNA complexes, a step toward successful application of polyplexes for in vivo siRNA delivery.
Collapse
Affiliation(s)
- Terry W J Steele
- School of Material Science & Engineering, Nanyang Technological University, Singapore, Singapore.
| | | | | | | |
Collapse
|
24
|
Guo J, Bourre L, Soden DM, O'Sullivan GC, O'Driscoll C. Can non-viral technologies knockdown the barriers to siRNA delivery and achieve the next generation of cancer therapeutics? Biotechnol Adv 2011; 29:402-17. [DOI: 10.1016/j.biotechadv.2011.03.003] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Revised: 03/08/2011] [Accepted: 03/13/2011] [Indexed: 12/22/2022]
|
25
|
Ming X. Cellular delivery of siRNA and antisense oligonucleotides via receptor-mediated endocytosis. Expert Opin Drug Deliv 2011; 8:435-49. [PMID: 21381985 DOI: 10.1517/17425247.2011.561313] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION There is great potential for antisense and siRNA oligonucleotides to become mainstream therapeutic entities thanks to their high specificity and wide therapeutic target space compared with small molecules. Despite this potential, the pharmacological targets within the cells are less accessible to oligonucleotides that are hydrophilic and often charged. Oligonucleotides access their intracellular targets mainly by means of endocytosis, but only a fraction of them reach their targets, as delivery requires functional synergy of cellular uptake and intracellular trafficking. AREAS COVERED This review provides an update on the progress of receptor-targeted delivery of oligonucleotides over the last 15 years and summarizes various targeting moieties for oligonucleotide delivery and coupling strategies. To inspire new strategies that can lead to oligonucleotides in the clinic, this review highlights how oligonucleotides successfully reach their intracellular targets by means of receptor-mediated endocytosis. EXPERT OPINION Understanding the mechanisms of oligonucleotide internalization has led to greater cellular uptake and superior endosomal release through the rational design of receptor-targeted delivery systems. Further improvements will again depend on a better understanding of the intracellular trafficking of oligonucleotides.
Collapse
Affiliation(s)
- Xin Ming
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
26
|
Philipp A, Meyer M, Zintchenko A, Wagner E. Functional modification of amide-crosslinked oligoethylenimine for improved siRNA delivery. REACT FUNCT POLYM 2011. [DOI: 10.1016/j.reactfunctpolym.2010.10.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
27
|
Tamura A, Nagasaki Y. Smart siRNA delivery systems based on polymeric nanoassemblies and nanoparticles. Nanomedicine (Lond) 2011; 5:1089-102. [PMID: 20874023 DOI: 10.2217/nnm.10.76] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
RNA interference is a post-transcriptional gene-silencing pathway induced by double-stranded small interfering RNA (siRNA). The potential use of siRNA as a therapeutic agent has attracted great attention as a novel approach to the treatment of several intractable diseases. Despite the rapid progress in the therapeutic use of siRNA, systemic application is still controversial due to the limitations of siRNA, such as low enzymatic tolerability, cellular internalization and body distribution after systemic administration. This review describes the recent progress and strategies of siRNA delivery systems based on polyion complexes. Numerous siRNA-containing polyion complex systems bound together through electrostatic interactions between the negatively charged siRNA and positively charged components, including synthetic polymers, biopolymers and nanoparticles, have been developed for the therapeutic application of siRNA. Additionally, stimulus-sensitive smart siRNA carrier systems, including bioreducible polycations and hydrophilic polymer-siRNA conjugates, have been developed to enhance the gene-silencing efficacy of siRNAs.
Collapse
Affiliation(s)
- Atsushi Tamura
- Graduate School of Pure & Applied Sciences, University of Tsukuba. 1-1-1 Ten-noudai, Tsukuba, Ibaraki, Japan
| | | |
Collapse
|
28
|
Functional Polymer Conjugates for Medicinal Nucleic Acid Delivery. POLYMERS IN NANOMEDICINE 2011. [DOI: 10.1007/12_2011_148] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
29
|
Sato Y, Hatakeyama H, Harashima H. Ornithine and tryptophan analogs as efficient polycations for short interference RNA delivery to tumor cells. Biol Pharm Bull 2010; 33:1246-9. [PMID: 20606322 DOI: 10.1248/bpb.33.1246] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The delivery of nucleic acids to cancer cells represents a potentially useful strategy. Previously, we developed a multifunctional envelope-type nano device (MEND) for the efficient delivery of plasmid DNA. In addition, we successfully delivered short interference RNA (siRNA) into cytoplasm using a MEND which contains siRNA particles that were produced using stearyl octaarginine (STR-R8). In the present study, to achieve further gene silencing activity compared with STR-R8, various additional polycations were screened. We used protamine and 10 different polypeptides containing random sequence of basic amino acids. The ability of these polycations to form nano particles with siRNA were evaluated by measuring the size and zeta-potential of produced nano particles, and as a consequence, 6 of the polycations were selected for further evaluation. We then prepared MENDs containing the particles. The lipid composition of the MEND consisted of dioleoylphosphatidyl ethanolamine (DOPE)/phosphatidic acid (PA) (7/2). For cellular uptake and endosomal escape, the MEND was modified with PPD (polyethylene glycol (PEG)-peptide-DOPE), STR-R8 and GALA, pH-sensitive fusogenic peptide. The resulting MEND had a diameter of 120-170 nm and a zeta-potential of 15-25 mV. The MEND was transfected into HeLa cells stably expressing luciferase and the silencing activity of the polycations was compared. Most of the polycations failed to knockdown luciferase activity. However, the polypeptide containing ornithine and tryptophan (Orn/Trp) induced a higher knockdown than STR-R8. In addition, Orn/Trp induced a silencing effect at lower doses than STR-R8, as evidenced by dose-response data. In conclusion, the findings suggest that Orn/Trp is a superior polycation to STR-R8 for siRNA delivery.
Collapse
Affiliation(s)
- Yusuke Sato
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | | | | |
Collapse
|
30
|
Gao W, Chan JM, Farokhzad OC. pH-Responsive nanoparticles for drug delivery. Mol Pharm 2010; 7:1913-20. [PMID: 20836539 DOI: 10.1021/mp100253e] [Citation(s) in RCA: 696] [Impact Index Per Article: 46.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
First-generation nanoparticles (NPs) have been clinically translated as pharmaceutical drug delivery carriers for their ability to improve on drug tolerability, circulation half-life, and efficacy. Toward the development of the next-generation NPs, researchers have designed novel multifunctional platforms for sustained release, molecular targeting, and environmental responsiveness. This review focuses on environmentally responsive mechanisms used in NP designs, and highlights the use of pH-responsive NPs in drug delivery. Different organs, tissues, and subcellular compartments, as well as their pathophysiological states, can be characterized by their pH levels and gradients. When exposed to these pH stimuli, pH-responsive NPs respond with physicochemical changes to their material structure and surface characteristics. These include swelling, dissociating or surface charge switching, in a manner that favors drug release at the target site over surrounding tissues. The novel developments described here may revise the classical outlook that NPs are passive delivery vehicles, in favor of responsive, sensing vehicles that use environmental cues to achieve maximal drug potency.
Collapse
Affiliation(s)
- Weiwei Gao
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women's Hospital, Boston, Massachusetts 02115, United States
| | | | | |
Collapse
|
31
|
Barrès V, Ouellet V, Lafontaine J, Tonin PN, Provencher DM, Mes-Masson AM. An essential role for Ran GTPase in epithelial ovarian cancer cell survival. Mol Cancer 2010; 9:272. [PMID: 20942967 PMCID: PMC2964620 DOI: 10.1186/1476-4598-9-272] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2010] [Accepted: 10/13/2010] [Indexed: 12/03/2022] Open
Abstract
Background We previously identified that Ran protein, a member of the Ras GTPase family, is highly expressed in high grade and high stage serous epithelial ovarian cancers, and that its overexpression is associated with a poor prognosis. Ran is known to contribute to both nucleocytoplasmic transport and cell cycle progression, but its role in ovarian cancer is not well defined. Results Using a lentivirus-based tetracycline-inducible shRNA approach, we show that downregulation of Ran expression in aggressive ovarian cancer cell lines affects cellular proliferation by inducing a caspase-3 associated apoptosis. Using a xenograft tumor assay, we demonstrate that depletion of Ran results in decreased tumorigenesis, and eventual tumor formation is associated with tumor cells that express Ran protein. Conclusion Our results suggest a role for Ran in ovarian cancer cell survival and tumorigenicity and suggest that this critical GTPase may be suitable as a therapeutic target.
Collapse
Affiliation(s)
- Véronique Barrès
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal/Institut du cancer de Montréal,1560 Sherbrooke Est, Montreal H2L 4M1, Canada
| | | | | | | | | | | |
Collapse
|
32
|
Edinger D, Wagner E. Bioresponsive polymers for the delivery of therapeutic nucleic acids. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2010; 3:33-46. [DOI: 10.1002/wnan.97] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Daniel Edinger
- Pharmaceutical Biotechnology, LMU University, Butenandtstrasse 5‐13, D‐81377 Munich, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, LMU University, Butenandtstrasse 5‐13, D‐81377 Munich, Germany
| |
Collapse
|
33
|
Abstract
Plasmid DNA and siRNA have a large potential for use as therapeutic nucleic acids in medicine. The way to the target cell and its proper compartment is full of obstacles. Polymeric carriers help to overcome the encountered barriers. Cationic polymers can interact with the nucleic acid in a nondamaging way but still require optimization with regard to transfer efficiency and biocompatibility. Aiming at virus-like features, as viruses are the most efficient natural gene carriers, the design of bioresponsive polymers shows promising results regarding DNA and siRNA delivery. By specific chemical modifications dynamic structures are created, programmed to respond towards changing demands on the delivery pathway by cleavage of labile bonds or conformational changes, thus enhancing biocompatible gene delivery.
Collapse
|
34
|
Gao W, Xiao Z, Radovic-Moreno A, Shi J, Langer R, Farokhzad OC. Progress in siRNA delivery using multifunctional nanoparticles. Methods Mol Biol 2010; 629:53-67. [PMID: 20387142 DOI: 10.1007/978-1-60761-657-3_4] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Nanoparticles made from synthetic polymers have been developed to deliver small interfering RNA (siRNA). For successful siRNA delivery, these nanoparticles need to efficiently encapsulate siRNA, actively target sites of interest, and release siRNA intracellularly. This chapter reviews recent progress using a multifunctional approach to design and engineer polymeric nanoparticles for siRNA delivery.
Collapse
Affiliation(s)
- Weiwei Gao
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesia, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | |
Collapse
|
35
|
Abstract
Small interfering RNA (siRNA) technology holds great promise as a therapeutic intervention for targeted gene silencing in cancer and other diseases. However, in vivo systemic delivery of siRNA-based therapeutics to tumour tissues/cells remains a challenge. The major limitations against the use of siRNA as a therapeutic tool are its degradation by serum nucleases, poor cellular uptake and rapid renal clearance following systemic administration. Several siRNA-based loco-regional therapeutics are already in clinical trials. Further development of siRNAs for anti-cancer therapy depends on the development of safe and effective nanocarriers for systemic administration. To overcome these hurdles, nuclease-resistant chemically modified siRNAs and variety of synthetic and natural biodegradable lipids and polymers have been developed to systemically deliver siRNA with different efficacy and safety profiles. Cationic liposomes have emerged as one of the most attractive carriers because of their ability to form complexes with negatively charged siRNA and high in vitro transfection efficiency. However, their effectiveness as potential therapeutic carriers is limited by potential for pulmonary toxicity. Recently, our laboratories described the use of neutral 1,2-dioleoyl-sn-glycero-3-phosphatidylcholine based nanoliposomes in murine tumour models. We found this approach to be safe and 10- and 30-fold more effective than cationic liposomes and naked siRNA, respectively, for systemic delivery of siRNA into tumour tissues. Here, we review potential approaches for systemic delivery of siRNA for cancer therapy.
Collapse
Affiliation(s)
- B Ozpolat
- Department of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030-4009, USA
| | | | | |
Collapse
|
36
|
Oskuee RK, Dehshahri A, Shier WT, Ramezani M. Alkylcarboxylate grafting to polyethylenimine: a simple approach to producing a DNA nanocarrier with low toxicity. J Gene Med 2009; 11:921-32. [PMID: 19634133 DOI: 10.1002/jgm.1374] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Various strategies have been examined to improve both transfection efficiency and cytotoxicity of polyethylenimine (PEI), a widely used polycationic nonviral gene vector. In the present study, we sought to improve PEI transfection efficiency by combining the osmotic burst mechanism for lysing endocytotic vesicles with the lipid depletion mechanism, which was accomplished by maintaining buffering capacity at the same time as adding a lipid-absorbing hydrophobic shell. METHODS PEI was altered via the substitution of various percentages of its primary amines with carboxylate-terminated short, moderate and long alkyl chains, by reaction with bromoacetic, 6-bromohexanoic, 10-bromodecanoic and 16-bromohexadecanoic acids. Modified polymers were complexed with plasmid and the particle size and zeta potential of the polyplexes were determined. Ethidium bromide dye exclusion was used to show the DNA-binding ability of the polymers and their transfection activity and cytotoxicity was evaluated in Neuro2A mammalian cells. RESULTS Decreased DNA-binding ability resulted from increases in either the degree of substitution or hydrocarbon chain length. Particle size and zeta potential measurements demonstrated that modified PEI polymers were able to form nanoparticles in the size range 60-195 nm, and surface charge decreased with an increasing degree of substitution. Higher degrees of substitution resulted in decreased cytotoxicity of polymers. Alkylcarboxylate substitution of primary amines on PEI enhanced transfection efficiencies by up to approximately five-fold relative to underivatized PEI, with the greatest increases occurring with 6-bromohexanoic acid derivatives at degrees of substitution below 10%. CONCLUSIONS The results obtained suggest that an appropriate balance between cationic and hydrophobic regions of alkylated PEI yields the optimal nonviral vector with high transfection efficiency and low toxicity.
Collapse
Affiliation(s)
- Reza K Oskuee
- Pharmaceutical and Biotechnology Research Centers, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | | | | |
Collapse
|
37
|
Schwerdt A, Zintchenko A, Concia M, Roesen N, Fisher K, Lindner LH, Issels R, Wagner E, Ogris M. Hyperthermia-induced targeting of thermosensitive gene carriers to tumors. Hum Gene Ther 2009. [PMID: 19866491 DOI: 10.1089/hgt.2008.064] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Locoregional hyperthermia (HT) can be used for site-directed activation of macromolecular drug delivery systems. We have developed a gene delivery system based on thermosensitive block copolymers (TSCs) with a phase transition temperature of 42 degrees C [Zintchenko, A., Ogris, M., and Wagner, E. (2006). Bioconjug. Chem. 17, 766-772], in which the statistical copolymer of vinylpyrrolidinone and N-isopropylacryamide is grafted on polyethylenimine (PEI). Here we applied polyplexes consisting of plasmid DNA and TSCs systemically in A/J mice bearing a syngeneic Neuro2A neuroblastoma tumor subcutaneously in each hind limb. One limb was selectively treated by HT at 42 degrees C, at the same time that polyplexes were injected via the tail vein. Hyperthermia led to increased accumulation of thermosensitive polymer and aggregation of thermosensitive polyplexes in HT-treated tumors, resulting in up to 10-fold increased DNA deposition compared with non-HT-treated tumor. The level of transgene expression induced by TSC polyplexes in HT-treated tumors was significantly higher and selective for tumor tissue. With nonthermosensitive PEI polyplexes HT did not influence transgene deposition or expression in tumor.
Collapse
Affiliation(s)
- Alenka Schwerdt
- Pharmaceutical Biology-Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität, Munich, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Richardt-Pargmann D, Vollmer J. Stimulation of the immune system by therapeutic antisense oligodeoxynucleotides and small interfering RNAs via nucleic acid receptors. Ann N Y Acad Sci 2009; 1175:40-54. [PMID: 19796076 DOI: 10.1111/j.1749-6632.2009.04971.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Most of the therapeutic applications for synthetic oligodeoxynucleotides (ODN) and oligoribonucleotides (ORN) relate to mechanisms of manipulating gene expression based on Watson-Crick base pairing to endogenous nucleic acids. However, in recent years it has become apparent that the immune system has evolved defense mechanisms against infections that are based on the detection of infecting viral and bacterial nucleic acids. In some cases, synthetic ODN and ORN can trigger these defenses and, therefore, can interfere with or distort the mechanism of action of antisense ODN or small interfering RNAs.
Collapse
|
39
|
Philipp A, Zhao X, Tarcha P, Wagner E, Zintchenko A. Hydrophobically Modified Oligoethylenimines as Highly Efficient Transfection Agents for siRNA Delivery. Bioconjug Chem 2009; 20:2055-61. [DOI: 10.1021/bc9001536] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Alexander Philipp
- Center for Drug Research, Department of Pharmacy, Pharmaceutical Biology-Biotechnology, and Center for NanoScience (CeNS), Ludwig-Maximilians-University, Butenandstr. 5-13, D-81377 Munich, Germany, and Department of Advanced Drug Delivery, Abbott Laboratories, D-R43D, AP31-2, 200 Abbott Park Road, Abbott Park, North Chicago, Illinois 60064
| | - Xiaobin Zhao
- Center for Drug Research, Department of Pharmacy, Pharmaceutical Biology-Biotechnology, and Center for NanoScience (CeNS), Ludwig-Maximilians-University, Butenandstr. 5-13, D-81377 Munich, Germany, and Department of Advanced Drug Delivery, Abbott Laboratories, D-R43D, AP31-2, 200 Abbott Park Road, Abbott Park, North Chicago, Illinois 60064
| | - Peter Tarcha
- Center for Drug Research, Department of Pharmacy, Pharmaceutical Biology-Biotechnology, and Center for NanoScience (CeNS), Ludwig-Maximilians-University, Butenandstr. 5-13, D-81377 Munich, Germany, and Department of Advanced Drug Delivery, Abbott Laboratories, D-R43D, AP31-2, 200 Abbott Park Road, Abbott Park, North Chicago, Illinois 60064
| | - Ernst Wagner
- Center for Drug Research, Department of Pharmacy, Pharmaceutical Biology-Biotechnology, and Center for NanoScience (CeNS), Ludwig-Maximilians-University, Butenandstr. 5-13, D-81377 Munich, Germany, and Department of Advanced Drug Delivery, Abbott Laboratories, D-R43D, AP31-2, 200 Abbott Park Road, Abbott Park, North Chicago, Illinois 60064
| | - Arkadi Zintchenko
- Center for Drug Research, Department of Pharmacy, Pharmaceutical Biology-Biotechnology, and Center for NanoScience (CeNS), Ludwig-Maximilians-University, Butenandstr. 5-13, D-81377 Munich, Germany, and Department of Advanced Drug Delivery, Abbott Laboratories, D-R43D, AP31-2, 200 Abbott Park Road, Abbott Park, North Chicago, Illinois 60064
| |
Collapse
|
40
|
Hatakeyama H, Ito E, Akita H, Oishi M, Nagasaki Y, Futaki S, Harashima H. A pH-sensitive fusogenic peptide facilitates endosomal escape and greatly enhances the gene silencing of siRNA-containing nanoparticles in vitro and in vivo. J Control Release 2009; 139:127-32. [DOI: 10.1016/j.jconrel.2009.06.008] [Citation(s) in RCA: 199] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2008] [Revised: 06/08/2009] [Accepted: 06/13/2009] [Indexed: 11/28/2022]
|
41
|
Oh YK, Park TG. siRNA delivery systems for cancer treatment. Adv Drug Deliv Rev 2009; 61:850-62. [PMID: 19422869 DOI: 10.1016/j.addr.2009.04.018] [Citation(s) in RCA: 481] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2008] [Accepted: 04/28/2009] [Indexed: 02/07/2023]
Abstract
With increasing knowledge on the molecular mechanisms of endogenous RNA interference, small interfering RNAs (siRNAs) have been emerging as innovative nucleic acid medicines for treatment of incurable diseases such as cancers. Although several siRNA candidates for the treatment of ocular and respiratory diseases are undergoing clinical trials, there are challenges inherent in the further development of siRNAs for anti-cancer therapeutics, because systemic administration will be required in most cases. In addition to nonspecific off-target and immune stimulation problems, appropriate delivery remains a major hurdle. The technologies developed for delivery of nucleic acid medicines such as plasmid DNA and antisense oligonucleotides have paved the way to rapid progress for in vivo delivery of siRNAs. Here, we review various in vivo delivery strategies including chemical modification, conjugation, lipid-based techniques, polymer-based nanosystems, and physical methods. Moreover, the current progress in siRNA delivery systems for gynecologic, liver, lung, and prostate cancers is discussed.
Collapse
|
42
|
Meyer M, Dohmen C, Philipp A, Kiener D, Maiwald G, Scheu C, Ogris M, Wagner E. Synthesis and Biological Evaluation of a Bioresponsive and Endosomolytic siRNA−Polymer Conjugate. Mol Pharm 2009; 6:752-62. [DOI: 10.1021/mp9000124] [Citation(s) in RCA: 151] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Martin Meyer
- Pharmaceutical Biotechnology, Center for Drug Research, Department of Pharmacy, Department of Chemistry and Biochemistry, and Center for Nanoscience, Ludwig-Maximilians-Universität München, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Christian Dohmen
- Pharmaceutical Biotechnology, Center for Drug Research, Department of Pharmacy, Department of Chemistry and Biochemistry, and Center for Nanoscience, Ludwig-Maximilians-Universität München, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Alexander Philipp
- Pharmaceutical Biotechnology, Center for Drug Research, Department of Pharmacy, Department of Chemistry and Biochemistry, and Center for Nanoscience, Ludwig-Maximilians-Universität München, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Daniel Kiener
- Pharmaceutical Biotechnology, Center for Drug Research, Department of Pharmacy, Department of Chemistry and Biochemistry, and Center for Nanoscience, Ludwig-Maximilians-Universität München, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Gelja Maiwald
- Pharmaceutical Biotechnology, Center for Drug Research, Department of Pharmacy, Department of Chemistry and Biochemistry, and Center for Nanoscience, Ludwig-Maximilians-Universität München, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Christina Scheu
- Pharmaceutical Biotechnology, Center for Drug Research, Department of Pharmacy, Department of Chemistry and Biochemistry, and Center for Nanoscience, Ludwig-Maximilians-Universität München, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Manfred Ogris
- Pharmaceutical Biotechnology, Center for Drug Research, Department of Pharmacy, Department of Chemistry and Biochemistry, and Center for Nanoscience, Ludwig-Maximilians-Universität München, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Center for Drug Research, Department of Pharmacy, Department of Chemistry and Biochemistry, and Center for Nanoscience, Ludwig-Maximilians-Universität München, Butenandtstrasse 5-13, 81377 Munich, Germany
| |
Collapse
|
43
|
Yu B, Zhao X, Lee LJ, Lee RJ. Targeted delivery systems for oligonucleotide therapeutics. AAPS JOURNAL 2009; 11:195-203. [PMID: 19296227 DOI: 10.1208/s12248-009-9096-1] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 10/07/2008] [Accepted: 02/20/2009] [Indexed: 12/11/2022]
Abstract
Oligonucleotides including antisense oligonucleotides and siRNA are emerging as promising therapeutic agents against a variety of diseases. Effective delivery of these molecules is critical to their successful clinical application. Targeted systems can greatly improve the efficiency and specificity of oligonucleotides delivery. Meanwhile, an effective delivery system must successfully overcome a multitude of biological barriers to enable the oligonucleotides to reach the site of action and access their biological targets. Several delivery strategies based on different platform technologies and different targeting ligands have been developed to achieve these objectives. This review aims at providing a summary and perspective on recent progress in this very active area of research.
Collapse
Affiliation(s)
- Bo Yu
- Department of Chemical and Biomolecular Engineering, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | | | | | | |
Collapse
|
44
|
Chen Y, Huang L. Tumor-targeted delivery of siRNA by non-viral vector: safe and effective cancer therapy. Expert Opin Drug Deliv 2009; 5:1301-11. [PMID: 19040393 DOI: 10.1517/17425240802568505] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
RNA interference technology has been developed as a potential therapeutic agent for many indications, including cancer. Silencing a specific oncogene in tumor cells brings about cell death both in vitro and in vivo. However, there is a great need for powerful delivery strategies to enhance the therapeutic effect of small interfering RNA (siRNA). This review summarizes different signaling pathways inhibited by siRNA and the advantages of targeted siRNA as a delivery system.
Collapse
Affiliation(s)
- Yunching Chen
- University of North Carolina at Chapel Hill, Eshelman School of Pharmacy, Division of Molecular Pharmaceutics, Campus Box 7360 Kerr Hall, Chapel Hill, NC 27599, USA
| | | |
Collapse
|
45
|
|
46
|
Sakurai Y, Hatakeyama H, Akita H, Oishi M, Nagasaki Y, Futaki S, Harashima H. Efficient Short Interference RNA Delivery to Tumor Cells Using a Combination of Octaarginine, GALA and Tumor-Specific, Cleavable Polyethylene Glycol System. Biol Pharm Bull 2009; 32:928-32. [DOI: 10.1248/bpb.32.928] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yu Sakurai
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University
- CREST, Japan Science and Technology Agency (JST)
| | - Hiroto Hatakeyama
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University
- CREST, Japan Science and Technology Agency (JST)
| | - Hidetaka Akita
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University
- CREST, Japan Science and Technology Agency (JST)
| | - Motoi Oishi
- Tsukuba Research Center for Interdisciplinary Material Science (TIMS), University of Tsukuba
- CREST, Japan Science and Technology Agency (JST)
| | - Yukio Nagasaki
- Tsukuba Research Center for Interdisciplinary Material Science (TIMS), University of Tsukuba
- CREST, Japan Science and Technology Agency (JST)
| | - Shiro Futaki
- Institute for Chemical Research, Kyoto University
| | - Hideyoshi Harashima
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University
- CREST, Japan Science and Technology Agency (JST)
| |
Collapse
|
47
|
Schwerdt A, Zintchenko A, Concia M, Roesen N, Fisher K, Lindner LH, Issels R, Wagner E, Ogris M. Hyperthermia-Induced Targeting of Thermosensitive Gene Carriers to Tumors. Hum Gene Ther 2008; 19:1283-92. [DOI: 10.1089/hum.2008.064] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Alenka Schwerdt
- Pharmaceutical Biology-Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität, D-81377 Munich, Germany
| | - Arkadi Zintchenko
- Pharmaceutical Biology-Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität, D-81377 Munich, Germany
| | - Massimo Concia
- Pharmaceutical Biology-Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität, D-81377 Munich, Germany
| | - Nick Roesen
- Hybrid Systems, Cherwell Innovation Centre, Upper Heyford, OX25 5HD Oxfordshire, United Kingdom
| | - Kerry Fisher
- Hybrid Systems, Cherwell Innovation Centre, Upper Heyford, OX25 5HD Oxfordshire, United Kingdom
| | - Lars H. Lindner
- Department of Internal Medicine III, Klinikum Grosshadern Medical Center (KGMC), Ludwig-Maximilians-Universität, D-81377 Munich, Germany
- CCG Hyperthermia, Helmholtz Zentrum München-National Research Center for Environment and Health, D-81377 Munich, Germany
| | - Rolf Issels
- Department of Internal Medicine III, Klinikum Grosshadern Medical Center (KGMC), Ludwig-Maximilians-Universität, D-81377 Munich, Germany
- CCG Hyperthermia, Helmholtz Zentrum München-National Research Center for Environment and Health, D-81377 Munich, Germany
| | - Ernst Wagner
- Pharmaceutical Biology-Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität, D-81377 Munich, Germany
| | - Manfred Ogris
- Pharmaceutical Biology-Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität, D-81377 Munich, Germany
| |
Collapse
|