1
|
Cullis PR, Felgner PL. The 60-year evolution of lipid nanoparticles for nucleic acid delivery. Nat Rev Drug Discov 2024; 23:709-722. [PMID: 38965378 DOI: 10.1038/s41573-024-00977-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2024] [Indexed: 07/06/2024]
Abstract
Delivery of genetic information to the interior of target cells in vivo has been a major challenge facing gene therapies. This barrier is now being overcome, owing in part to dramatic advances made by lipid-based systems that have led to lipid nanoparticles (LNPs) that enable delivery of nucleic acid-based vaccines and therapeutics. Examples include the clinically approved COVID-19 LNP mRNA vaccines and Onpattro (patisiran), an LNP small interfering RNA therapeutic to treat transthyretin-induced amyloidosis (hATTR). In addition, a host of promising LNP-enabled vaccines and gene therapies are in clinical development. Here, we trace this success to two streams of research conducted over the past 60 years: the discovery of the transfection properties of lipoplexes composed of positively charged cationic lipids complexed with nucleic acid cargos and the development of lipid nanoparticles using ionizable cationic lipids. The fundamental insights gained from these two streams of research offer potential delivery solutions for most forms of gene therapies.
Collapse
Affiliation(s)
- P R Cullis
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada.
| | - P L Felgner
- Department of Physiology & Biophysics, University of California, Irvine, CA, USA.
| |
Collapse
|
2
|
Chu R, Wang Y, Kong J, Pan T, Yang Y, He J. Lipid nanoparticles as the drug carrier for targeted therapy of hepatic disorders. J Mater Chem B 2024; 12:4759-4784. [PMID: 38682294 DOI: 10.1039/d3tb02766j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
The liver, a complex and vital organ in the human body, is susceptible to various diseases, including metabolic disorders, acute hepatitis, cirrhosis, and hepatocellular carcinoma. In recent decades, these diseases have significantly contributed to global morbidity and mortality. Currently, liver transplantation remains the most effective treatment for hepatic disorders. Nucleic acid therapeutics offer a selective approach to disease treatment through diverse mechanisms, enabling the regulation of relevant genes and providing a novel therapeutic avenue for hepatic disorders. It is expected that nucleic acid drugs will emerge as the third generation of pharmaceuticals, succeeding small molecule drugs and antibody drugs. Lipid nanoparticles (LNPs) represent a crucial technology in the field of drug delivery and constitute a significant advancement in gene therapies. Nucleic acids encapsulated in LNPs are shielded from the degradation of enzymes and effectively delivered to cells, where they are released and regulate specific genes. This paper provides a comprehensive review of the structure, composition, and applications of LNPs in the treatment of hepatic disorders and offers insights into prospects and challenges in the future development of LNPs.
Collapse
Affiliation(s)
- Runxuan Chu
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, P. R. China.
| | - Yi Wang
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tung, Hong Kong SAR, P. R. China.
| | - Jianglong Kong
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tung, Hong Kong SAR, P. R. China.
| | - Ting Pan
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, P. R. China.
- Department of Pharmaceutics School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Yani Yang
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, P. R. China.
| | - Jun He
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, P. R. China.
| |
Collapse
|
3
|
Zhong Q, Zhang H. Preparation of Small Unilamellar Vesicle Liposomes Using Detergent Dialysis Method. Methods Mol Biol 2023; 2622:49-56. [PMID: 36781748 DOI: 10.1007/978-1-0716-2954-3_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
Small unilamellar liposomes are commonly used as model biomembranes and carriers for drug and gene delivery. Although methods which employ mechanical forces or organic solvents can be used to reduce the liposome size and the number of lipid bilayers, they are not suitable options when the purpose is to incorporate biologically active proteins into lipid bilayers or to encapsulate nucleic acid into liposomes. Detergent dialysis is a simple and inexpensive procedure to produce homogeneous small unilamellar vesicles (SUVs). Lipids are solubilized by detergent at a concentration much higher than its critical micellar concentration (CMC) in an aqueous dispersion medium. Free monomer detergent molecules in the dispersion medium are removed during dialysis, while the supramolecular detergent-lipid mixed micelles (MMs) are kept inside the dialysis tubing, leading to dissociation of detergent molecules from MMs. SUVs are formed after the micelle to vesicle transition (MVT).
Collapse
Affiliation(s)
- Qingyue Zhong
- Department of Pharmaceutical Sciences, School of Pharmacy-Boston, MCPHS University, Boston, MA, USA
| | - Hongwei Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy-Boston, MCPHS University, Boston, MA, USA
| |
Collapse
|
4
|
Sun D, Lu ZR. Structure and Function of Cationic and Ionizable Lipids for Nucleic Acid Delivery. Pharm Res 2023; 40:27-46. [PMID: 36600047 PMCID: PMC9812548 DOI: 10.1007/s11095-022-03460-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 12/08/2022] [Indexed: 01/05/2023]
Abstract
Hereditary genetic diseases, cancer, and infectious diseases are affecting global health and become major health issues, but the treatment development remains challenging. Gene therapies using DNA plasmid, RNAi, miRNA, mRNA, and gene editing hold great promise. Lipid nanoparticle (LNP) delivery technology has been a revolutionary development, which has been granted for clinical applications, including mRNA vaccines against SARS-CoV-2 infections. Due to the success of LNP systems, understanding the structure, formulation, and function relationship of the lipid components in LNP systems is crucial for design more effective LNP. Here, we highlight the key considerations for developing an LNP system. The evolution of structure and function of lipids as well as their LNP formulation from the early-stage simple formulations to multi-components LNP and multifunctional ionizable lipids have been discussed. The flexibility and platform nature of LNP enable efficient intracellular delivery of a variety of therapeutic nucleic acids and provide many novel treatment options for the diseases that are previously untreatable.
Collapse
Affiliation(s)
- Da Sun
- Department of Biomedical Engineering, Case Western Reserve University, Wickenden 427, Mail Stop 7207, 10900 Euclid Avenue, Cleveland, OH, 44106, USA
| | - Zheng-Rong Lu
- Department of Biomedical Engineering, Case Western Reserve University, Wickenden 427, Mail Stop 7207, 10900 Euclid Avenue, Cleveland, OH, 44106, USA.
| |
Collapse
|
5
|
Han S, Chen X, Huang L. The tumor therapeutic potential of long non-coding RNA delivery and targeting. Acta Pharm Sin B 2022; 13:1371-1382. [PMID: 37139413 PMCID: PMC10149988 DOI: 10.1016/j.apsb.2022.12.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/25/2022] [Accepted: 11/11/2022] [Indexed: 12/15/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) is a type of RNA over 200 nt long without any protein coding ability, which has been investigated relating to crucial biological function in cells. There are many key lncRNAs in tumor/normal cells that serve as a biological marker or a new target for tumor treatment. However, compared to some small non-coding RNA, lncRNA-based drugs are limited in clinical application. Different from other non-coding RNA, like microRNAs, most lncRNAs have a high molecular weight and conserved secondary structure, making the delivery of lncRNAs more complex than the small non-coding RNAs. Considering that lncRNAs constitute the most abundant part of the mammalian genome, it is critical to further explore lncRNA delivery and the subsequent functional studies for potential clinical application. In this review, we will discuss the function and mechanism of lncRNAs in diseases, especially cancer, and different approaches for lncRNA transfection using multiple biomaterials.
Collapse
|
6
|
Hald Albertsen C, Kulkarni JA, Witzigmann D, Lind M, Petersson K, Simonsen JB. The role of lipid components in lipid nanoparticles for vaccines and gene therapy. Adv Drug Deliv Rev 2022; 188:114416. [PMID: 35787388 PMCID: PMC9250827 DOI: 10.1016/j.addr.2022.114416] [Citation(s) in RCA: 288] [Impact Index Per Article: 144.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 06/03/2022] [Accepted: 06/28/2022] [Indexed: 12/21/2022]
Abstract
Lipid nanoparticles (LNPs) play an important role in mRNA vaccines against COVID-19. In addition, many preclinical and clinical studies, including the siRNA-LNP product, Onpattro®, highlight that LNPs unlock the potential of nucleic acid-based therapies and vaccines. To understand what is key to the success of LNPs, we need to understand the role of the building blocks that constitute them. In this Review, we discuss what each lipid component adds to the LNP delivery platform in terms of size, structure, stability, apparent pKa, nucleic acid encapsulation efficiency, cellular uptake, and endosomal escape. To explore this, we present findings from the liposome field as well as from landmark and recent articles in the LNP literature. We also discuss challenges and strategies related to in vitro/in vivo studies of LNPs based on fluorescence readouts, immunogenicity/reactogenicity, and LNP delivery beyond the liver. How these fundamental challenges are pursued, including what lipid components are added and combined, will likely determine the scope of LNP-based gene therapies and vaccines for treating various diseases.
Collapse
Affiliation(s)
- Camilla Hald Albertsen
- Explorative Formulation & Technologies, CMC Design and Development, LEO Pharma A/S, Industriparken 55, 2750 Ballerup, Denmark
| | - Jayesh A Kulkarni
- NanoVation Therapeutics Inc., 2405 Wesbrook Mall, 4th Floor, Vancouver BC V6T 1Z3, Canada
| | - Dominik Witzigmann
- NanoVation Therapeutics Inc., 2405 Wesbrook Mall, 4th Floor, Vancouver BC V6T 1Z3, Canada
| | - Marianne Lind
- Explorative Formulation & Technologies, CMC Design and Development, LEO Pharma A/S, Industriparken 55, 2750 Ballerup, Denmark
| | - Karsten Petersson
- Explorative Formulation & Technologies, CMC Design and Development, LEO Pharma A/S, Industriparken 55, 2750 Ballerup, Denmark
| | - Jens B Simonsen
- Explorative Formulation & Technologies, CMC Design and Development, LEO Pharma A/S, Industriparken 55, 2750 Ballerup, Denmark.
| |
Collapse
|
7
|
Taina-González L, de la Fuente M. The Potential of Nanomedicine to Unlock the Limitless Applications of mRNA. Pharmaceutics 2022; 14:460. [PMID: 35214191 PMCID: PMC8879057 DOI: 10.3390/pharmaceutics14020460] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/10/2022] [Accepted: 02/14/2022] [Indexed: 01/27/2023] Open
Abstract
The year 2020 was a turning point in the way society perceives science. Messenger RNA (mRNA) technology finally showed and shared its potential, starting a new era in medicine. However, there is no doubt that commercialization of these vaccines would not have been possible without nanotechnology, which has finally answered the long-term question of how to deliver mRNA in vivo. The aim of this review is to showcase the importance of this scientific milestone for the development of additional mRNA therapeutics. Firstly, we provide a full description of the marketed vaccine formulations and disclose LNPs' pharmaceutical properties, including composition, structure, and manufacturing considerations Additionally, we review different types of lipid-based delivery technologies currently in preclinical and clinical development, namely lipoplexes and cationic nanoemulsions. Finally, we highlight the most promising clinical applications of mRNA in different fields such as vaccinology, immuno-oncology, gene therapy for rare genetic diseases and gene editing using CRISPR Cas9.
Collapse
Affiliation(s)
- Laura Taina-González
- Nano-Oncology and Translational Therapeutics Group, Health Research Institute of Santiago de Compostela (IDIS), SERGAS, 15706 Santiago de Compostela, Spain;
- Universidad de Santiago de Compostela (USC), 15782 Santiago de Compostela, Spain
| | - María de la Fuente
- Nano-Oncology and Translational Therapeutics Group, Health Research Institute of Santiago de Compostela (IDIS), SERGAS, 15706 Santiago de Compostela, Spain;
- Cancer Network Research (CIBERONC), 28029 Madrid, Spain
- DIVERSA Technologies, 15782 Santiago de Compostela, Spain
| |
Collapse
|
8
|
Coelho F, Salonen LM, Silva BFB. Hemiacetal-linked pH-sensitive PEG-lipids for non-viral gene delivery. NEW J CHEM 2022. [DOI: 10.1039/d2nj02217f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Cationic lipid–DNA complexes containing a novel hemiacetal PEG-lipid for endosomal escape were characterized in terms of pH-response, stability, and biological activity.
Collapse
Affiliation(s)
- Filipe Coelho
- International Iberian Nanotechnology Laboratory (INL), Av. Mestre José Veiga, 4715-330 Braga, Portugal
| | - Laura M. Salonen
- International Iberian Nanotechnology Laboratory (INL), Av. Mestre José Veiga, 4715-330 Braga, Portugal
| | - Bruno F. B. Silva
- International Iberian Nanotechnology Laboratory (INL), Av. Mestre José Veiga, 4715-330 Braga, Portugal
| |
Collapse
|
9
|
Chan C, Du S, Dong Y, Cheng X. Computational and Experimental Approaches to Investigate Lipid Nanoparticles as Drug and Gene Delivery Systems. Curr Top Med Chem 2021; 21:92-114. [PMID: 33243123 PMCID: PMC8191596 DOI: 10.2174/1568026620666201126162945] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 10/16/2020] [Accepted: 10/22/2020] [Indexed: 02/06/2023]
Abstract
Lipid nanoparticles (LNPs) have been widely applied in drug and gene delivery. More than twenty years ago, DoxilTM was the first LNPs-based drug approved by the US Food and Drug Administration (FDA). Since then, with decades of research and development, more and more LNP-based therapeutics have been used to treat diverse diseases, which often offer the benefits of reduced toxicity and/or enhanced efficacy compared to the active ingredients alone. Here, we provide a review of recent advances in the development of efficient and robust LNPs for drug/gene delivery. We emphasize the importance of rationally combining experimental and computational approaches, especially those providing multiscale structural and functional information of LNPs, to the design of novel and powerful LNP-based delivery systems.
Collapse
Affiliation(s)
- Chun Chan
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Shi Du
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Yizhou Dong
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
- Department of Biomedical Engineering; The Center for Clinical and Translational Science; The Comprehensive Cancer Center; Dorothy M. Davis Heart & Lung Research Institute; Department of Radiation Oncology, The Ohio State University, Columbus, OH 43210, USA
| | - Xiaolin Cheng
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
- Biophysics Graduate Program, Translational Data Analytics Institute, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
10
|
Tanaka H, Sakurai Y, Anindita J, Akita H. Development of lipid-like materials for RNA delivery based on intracellular environment-responsive membrane destabilization and spontaneous collapse. Adv Drug Deliv Rev 2020; 154-155:210-226. [PMID: 32650040 DOI: 10.1016/j.addr.2020.07.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/26/2020] [Accepted: 07/02/2020] [Indexed: 01/01/2023]
Abstract
Messenger RNA and small interfering RNA are attractive modalities for curing diseases by complementation or knock-down of proteins. For success of these RNAs, a drug delivery system (DDS) is required to control a pharmacokinetics, to enhance cellular uptake, to overcome biological membranes, and to release the cargo into the cytoplasm. Based on past research, developing nanoparticles that are neutrally charged have been the mainstream of their development. Also, the materials are further mounted with pH- and/or reducing environment-responsive units. In this review, we summarize progress made in the molecular design of these materials. We also focus on the importance of the hydrophobic scaffold for tissue/cell targeting, intracellular trafficking, and immune responses. As a practical example, the design concept of the SS-cleavable and pH-activated lipid-like material (ssPalm) and subsequent molecular modification tailored to the RNA-based medical application is discussed.
Collapse
|
11
|
Witzigmann D, Kulkarni JA, Leung J, Chen S, Cullis PR, van der Meel R. Lipid nanoparticle technology for therapeutic gene regulation in the liver. Adv Drug Deliv Rev 2020; 159:344-363. [PMID: 32622021 PMCID: PMC7329694 DOI: 10.1016/j.addr.2020.06.026] [Citation(s) in RCA: 203] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 06/12/2020] [Accepted: 06/25/2020] [Indexed: 02/08/2023]
Abstract
Hereditary genetic disorders, cancer, and infectious diseases of the liver affect millions of people around the globe and are a major public health burden. Most contemporary treatments offer limited relief as they generally aim to alleviate disease symptoms. Targeting the root cause of diseases originating in the liver by regulating malfunctioning genes with nucleic acid-based drugs holds great promise as a therapeutic approach. However, employing nucleic acid therapeutics in vivo is challenging due to their unfavorable characteristics. Lipid nanoparticle (LNP) delivery technology is a revolutionary development that has enabled clinical translation of gene therapies. LNPs can deliver siRNA, mRNA, DNA, or gene-editing complexes, providing opportunities to treat hepatic diseases by silencing pathogenic genes, expressing therapeutic proteins, or correcting genetic defects. Here we discuss the state-of-the-art LNP technology for hepatic gene therapy including formulation design parameters, production methods, preclinical development and clinical translation.
Collapse
Affiliation(s)
- Dominik Witzigmann
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada; NanoMedicines Innovation Network (NMIN), University of British Columbia, Vancouver, BC, Canada
| | - Jayesh A Kulkarni
- NanoMedicines Innovation Network (NMIN), University of British Columbia, Vancouver, BC, Canada; Centre for Molecular Medicine and Therapeutics, Department of Medical Genetics, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada; Evonik Canada, Vancouver, BC, Canada
| | - Jerry Leung
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Sam Chen
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada; Integrated Nanotherapeutics, Vancouver, BC, Canada
| | - Pieter R Cullis
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada; NanoMedicines Innovation Network (NMIN), University of British Columbia, Vancouver, BC, Canada.
| | - Roy van der Meel
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| |
Collapse
|
12
|
Samaridou E, Heyes J, Lutwyche P. Lipid nanoparticles for nucleic acid delivery: Current perspectives. Adv Drug Deliv Rev 2020; 154-155:37-63. [PMID: 32526452 DOI: 10.1016/j.addr.2020.06.002] [Citation(s) in RCA: 293] [Impact Index Per Article: 73.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/21/2020] [Accepted: 06/03/2020] [Indexed: 12/11/2022]
Abstract
Nucleic Acid (NA) based therapeutics are poised to disrupt modern medicine and augment traditional pharmaceutics in a meaningful way. However, a key challenge to advancing NA therapies into the clinical setting and on to the market is the safe and effective delivery to the target tissue and cell. Lipid Nanoparticles (LNP) have been extensively investigated and are currently the most advanced vector for the delivery of NA drugs, as evidenced by the approval of Onpattro for treatment of Amyloidosis in the US and EU in 2018. This article provides a comprehensive review of the state-of-the-art for LNP technology. We discuss key advances in the design and development of LNP, leading to a broad range of therapeutic applications. Finally, the current status of this technology in clinical trials and its future prospects are discussed.
Collapse
Affiliation(s)
- Eleni Samaridou
- Genevant Sciences Corp., 155 - 887 Great Northern Way, Vancouver, British Columbia V5T 4T5, Canada
| | - James Heyes
- Genevant Sciences Corp., 155 - 887 Great Northern Way, Vancouver, British Columbia V5T 4T5, Canada
| | - Peter Lutwyche
- Genevant Sciences Corp., 155 - 887 Great Northern Way, Vancouver, British Columbia V5T 4T5, Canada.
| |
Collapse
|
13
|
Zhang M, Lemay SG. Interaction of Anionic Bulk Nanobubbles with Cationic Liposomes: Evidence for Reentrant Condensation. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:4146-4151. [PMID: 30811209 PMCID: PMC6427481 DOI: 10.1021/acs.langmuir.8b03927] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 02/25/2019] [Indexed: 06/09/2023]
Abstract
We investigated the interaction of bulk nanobubbles with cationic liposomes composed of 1,2-dioleoyl- sn-glycero-3-ethylphosphocholine and anionic liposomes assembled from 1-palmitoyl-2-oleoyl- sn-glycero-3-phospho-(1'- rac-glycerol). We employed dynamic light scattering and fluorescence microscopy to investigate both the hydrodynamic and electrophoretic properties of the nanobubble/liposome complexes. These optical techniques permit direct visualization of structural changes as a function of the bubble/liposome ratio. We observed reentrant condensation with cationic liposomes and gas nucleation with anionic liposomes. This is the first report of charge inversion and reentrant condensation of cationic liposomes induced by bulk nanobubbles.
Collapse
Affiliation(s)
- Minmin Zhang
- MESA+ Institute for Nanotechnology
& Faculty of Science and Technology, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - Serge G. Lemay
- MESA+ Institute for Nanotechnology
& Faculty of Science and Technology, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| |
Collapse
|
14
|
Kulkarni JA, Cullis PR, van der Meel R. Lipid Nanoparticles Enabling Gene Therapies: From Concepts to Clinical Utility. Nucleic Acid Ther 2018; 28:146-157. [DOI: 10.1089/nat.2018.0721] [Citation(s) in RCA: 230] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Jayesh A. Kulkarni
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Pieter R. Cullis
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Roy van der Meel
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
15
|
Nishimura T, Yamada A, Umezaki K, Sawada SI, Mukai SA, Sasaki Y, Akiyoshi K. Self-Assembled Polypeptide Nanogels with Enzymatically Transformable Surface as a Small Interfering RNA Delivery Platform. Biomacromolecules 2017; 18:3913-3923. [DOI: 10.1021/acs.biomac.7b00937] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Tomoki Nishimura
- Department
of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
- ERATO
Bio-nanotransporter Project, Japan Science and Technology Agency (JST), Kyoto University, Katsura,
Nishikyo-ku, Kyoto 615-8530, Japan
| | - Akina Yamada
- Department
of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Kaori Umezaki
- ERATO
Bio-nanotransporter Project, Japan Science and Technology Agency (JST), Kyoto University, Katsura,
Nishikyo-ku, Kyoto 615-8530, Japan
| | - Shin-ichi Sawada
- Department
of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
- ERATO
Bio-nanotransporter Project, Japan Science and Technology Agency (JST), Kyoto University, Katsura,
Nishikyo-ku, Kyoto 615-8530, Japan
| | - Sada-atsu Mukai
- Department
of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
- ERATO
Bio-nanotransporter Project, Japan Science and Technology Agency (JST), Kyoto University, Katsura,
Nishikyo-ku, Kyoto 615-8530, Japan
| | - Yoshihiro Sasaki
- Department
of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Kazunari Akiyoshi
- Department
of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
- ERATO
Bio-nanotransporter Project, Japan Science and Technology Agency (JST), Kyoto University, Katsura,
Nishikyo-ku, Kyoto 615-8530, Japan
| |
Collapse
|
16
|
Fan M, Zeng Y, Ruan H, Zhang Z, Gong T, Sun X. Ternary Nanoparticles with a Sheddable Shell Efficiently Deliver MicroRNA-34a against CD44-Positive Melanoma. Mol Pharm 2017; 14:3152-3163. [PMID: 28759238 DOI: 10.1021/acs.molpharmaceut.7b00377] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Minmin Fan
- Key Laboratory of
Drug Targeting and Drug Delivery Systems, Ministry of Education, West
China School of Pharmacy, Sichuan University, Chengdu 610041, China
- State Key Laboratory of Polymer Materials
Engineering, Polymer Research Institute of Sichuan University, Chengdu 610065, China
| | - Ye Zeng
- Key Laboratory of
Drug Targeting and Drug Delivery Systems, Ministry of Education, West
China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Huitong Ruan
- Key Laboratory of
Drug Targeting and Drug Delivery Systems, Ministry of Education, West
China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zhirong Zhang
- Key Laboratory of
Drug Targeting and Drug Delivery Systems, Ministry of Education, West
China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Tao Gong
- Key Laboratory of
Drug Targeting and Drug Delivery Systems, Ministry of Education, West
China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xun Sun
- Key Laboratory of
Drug Targeting and Drug Delivery Systems, Ministry of Education, West
China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
17
|
Angelov B, Garamus VM, Drechsler M, Angelova A. Structural analysis of nanoparticulate carriers for encapsulation of macromolecular drugs. J Mol Liq 2017. [DOI: 10.1016/j.molliq.2016.11.064] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
18
|
Zhou Z, Liu X, Zhu D, Wang Y, Zhang Z, Zhou X, Qiu N, Chen X, Shen Y. Nonviral cancer gene therapy: Delivery cascade and vector nanoproperty integration. Adv Drug Deliv Rev 2017; 115:115-154. [PMID: 28778715 DOI: 10.1016/j.addr.2017.07.021] [Citation(s) in RCA: 281] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 07/25/2017] [Accepted: 07/27/2017] [Indexed: 02/07/2023]
Abstract
Gene therapy represents a promising cancer treatment featuring high efficacy and limited side effects, but it is stymied by a lack of safe and efficient gene-delivery vectors. Cationic polymers and lipid-based nonviral gene vectors have many advantages and have been extensively explored for cancer gene delivery, but their low gene-expression efficiencies relative to viral vectors limit their clinical translations. Great efforts have thus been devoted to developing new carrier materials and fabricating functional vectors aimed at improving gene expression, but the overall efficiencies are still more or less at the same level. This review analyzes the cancer gene-delivery cascade and the barriers, the needed nanoproperties and the current strategies for overcoming these barriers, and outlines PEGylation, surface-charge, size, and stability dilemmas in vector nanoproperties to efficiently accomplish the cancer gene-delivery cascade. Stability, surface, and size transitions (3S Transitions) are proposed to resolve those dilemmas and strategies to realize these transitions are comprehensively summarized. The review concludes with a discussion of the future research directions to design high-performance nonviral gene vectors.
Collapse
Affiliation(s)
- Zhuxian Zhou
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, China
| | - Xiangrui Liu
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, China
| | - Dingcheng Zhu
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, China
| | - Yue Wang
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, China
| | - Zhen Zhang
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, China
| | - Xuefei Zhou
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, China
| | - Nasha Qiu
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, China
| | - Xuesi Chen
- Changchun Institute of Applied Chemistry, Key Lab of Polymer Ecomaterials, Changchun, China
| | - Youqing Shen
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, China.
| |
Collapse
|
19
|
Ansari AS, Santerre PJ, Uludağ H. Biomaterials for polynucleotide delivery to anchorage-independent cells. J Mater Chem B 2017; 5:7238-7261. [DOI: 10.1039/c7tb01833a] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Comparison of various chemical vectors used for polynucleotide delivery to mammalian anchorage-independent cells.
Collapse
Affiliation(s)
- Aysha S. Ansari
- Department of Chemical & Materials Engineering
- Faculty of Engineering
- University of Alberta
- Edmonton
- Canada
| | - Paul J. Santerre
- Institute of Biomaterials & Biomedical Engineering
- University of Toronto
- Toronto
- Canada
| | - Hasan Uludağ
- Department of Chemical & Materials Engineering
- Faculty of Engineering
- University of Alberta
- Edmonton
- Canada
| |
Collapse
|
20
|
DeRosa F, Guild B, Karve S, Smith L, Love K, Dorkin JR, Kauffman KJ, Zhang J, Yahalom B, Anderson DG, Heartlein MW. Therapeutic efficacy in a hemophilia B model using a biosynthetic mRNA liver depot system. Gene Ther 2016; 23:699-707. [PMID: 27356951 PMCID: PMC5059749 DOI: 10.1038/gt.2016.46] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 04/06/2016] [Accepted: 05/20/2016] [Indexed: 12/11/2022]
Abstract
DNA-based gene therapy has considerable therapeutic potential, but the challenges associated with delivery continue to limit progress. Messenger RNA (mRNA) has the potential to provide for transient production of therapeutic proteins, without the need for nuclear delivery and without the risk of insertional mutagenesis. Here we describe the sustained delivery of therapeutic proteins in vivo in both rodents and non-human primates via nanoparticle-formulated mRNA. Nanoparticles formulated with lipids and lipid-like materials were developed for delivery of two separate mRNA transcripts encoding either human erythropoietin (hEPO) or factor IX (hFIX) protein. Dose-dependent protein production was observed for each mRNA construct. Upon delivery of hEPO mRNA in mice, serum EPO protein levels reached several orders of magnitude (>125 000-fold) over normal physiological values. Further, an increase in hematocrit (Hct) was established, demonstrating that the exogenous mRNA-derived protein maintained normal activity. The capacity of producing EPO in non-human primates via delivery of formulated mRNA was also demonstrated as elevated EPO protein levels were observed over a 72-h time course. Exemplifying the possible broad utility of mRNA drugs, therapeutically relevant amounts of human FIX (hFIX) protein were achieved upon a single intravenous dose of hFIX mRNA-loaded lipid nanoparticles in mice. In addition, therapeutic value was established within a hemophilia B (FIX knockout (KO)) mouse model by demonstrating a marked reduction in Hct loss following injury (incision) to FIX KO mice.
Collapse
Affiliation(s)
- F DeRosa
- Shire Pharmaceuticals, Lexington, MA, USA
| | - B Guild
- Shire Pharmaceuticals, Lexington, MA, USA
| | - S Karve
- Shire Pharmaceuticals, Lexington, MA, USA
| | - L Smith
- Shire Pharmaceuticals, Lexington, MA, USA
| | - K Love
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - J R Dorkin
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - K J Kauffman
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - J Zhang
- Shire Pharmaceuticals, Lexington, MA, USA
| | - B Yahalom
- Biomedical Research Models, Inc., Worcester, MA, USA
| | - D G Anderson
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard-MIT Division of Health Sciences & Technology, Cambridge, MA, USA
- Institute of Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | |
Collapse
|
21
|
Tiffany M, Szoka FC. Co-localization of fluorescent labeled lipid nanoparticles with specifically tagged subcellular compartments by single particle tracking at low nanoparticle to cell ratios. J Drug Target 2016; 24:857-864. [DOI: 10.1080/1061186x.2016.1233976] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Matthew Tiffany
- Department of Bioengineering, Therapeutic Sciences and Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
- Department of Pediatrics and Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Francis C. Szoka
- Department of Bioengineering, Therapeutic Sciences and Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| |
Collapse
|
22
|
Chertok B, Langer R, Anderson DG. Spatial Control of Gene Expression by Nanocarriers Using Heparin Masking and Ultrasound-Targeted Microbubble Destruction. ACS NANO 2016; 10:7267-7278. [PMID: 27472268 PMCID: PMC5240524 DOI: 10.1021/acsnano.6b01199] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
We developed a method to spatially control gene expression following nonviral delivery of DNA. This method includes surface-modifying DNA nanocarriers with heparin to inhibit passive gene transfer in both the target and the off-target tissues and using ultrasound-targeted microbubble destruction (UTMD) to selectively activate heparin-inhibited gene transfer at the target site. We observed that the engraftment of heparin onto the surface of cationic liposomes reduced off-target gene expression in the liver, a major site of nanoplex accumulation, by more than 700-fold compared to the nonheparinized PEGylated liposomes. We further observed that tumor-directed UTMD increased gene transfer with heparin-modified nanoplexes by more than 10-fold. This method augmented tumor-to-liver selectivity of gene expression by 4000-fold compared to controls. We conclude that heparinization of DNA nanocarriers in conjunction with localized activation of gene transfer by UTMD may enable greater spatial control over genetic therapy.
Collapse
Affiliation(s)
- Beata Chertok
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan , Ann Arbor, Michigan 48109, United States
- Department of Biomedical Engineering, College of Engineering, University of Michigan , Ann Arbor, Michigan 48109, United States
| | - Robert Langer
- Department of Chemical Engineering, MIT , Cambridge, Massachusetts 02139, United States
- David H. Koch Institute for Integrative Cancer Research, MIT , Cambridge, Massachusetts 02139, United States
- Institute for Medical Engineering & Science, MIT , Cambridge, Massachusetts 02139, United States
- Harvard-MIT Division of Health Sciences & Technology, Cambridge, Massachusetts 02139, United States
| | - Daniel G Anderson
- Department of Chemical Engineering, MIT , Cambridge, Massachusetts 02139, United States
- David H. Koch Institute for Integrative Cancer Research, MIT , Cambridge, Massachusetts 02139, United States
- Institute for Medical Engineering & Science, MIT , Cambridge, Massachusetts 02139, United States
- Harvard-MIT Division of Health Sciences & Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
23
|
Tanaka H, Sato Y, Harashima H, Akita H. Cellular environment-responsive nanomaterials for use in gene and siRNA delivery: molecular design for biomembrane destabilization and intracellular collapse. Expert Opin Drug Deliv 2016; 13:1015-27. [DOI: 10.1517/17425247.2016.1154531] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Hiroki Tanaka
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Yusuke Sato
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | | | - Hidetaka Akita
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| |
Collapse
|
24
|
Abstract
The delivery of genetic materials into cells to elicit cellular response has been extensively studied by biomaterials scientists globally.
Collapse
Affiliation(s)
- Xian Jun Loh
- Institute of Materials Research and Engineering
- A*STAR
- (Agency for Science
- Technology and Research)
- Singapore 117602
| | - Tung-Chun Lee
- UCL Institute for Materials Discovery and Department of Chemistry
- University College London
- London WC1H 0AJ
- UK
| | - Qingqing Dou
- Institute of Materials Research and Engineering
- A*STAR
- (Agency for Science
- Technology and Research)
- Singapore 117602
| | - G. Roshan Deen
- Soft Materials Laboratory
- Natural Sciences and Science Education
- National Institute of Education
- Nanyang Technological University
- 637616 Singapore
| |
Collapse
|
25
|
Chan CL, Ewert KK, Majzoub RN, Hwu YK, Liang KS, Leal C, Safinya CR. Optimizing cationic and neutral lipids for efficient gene delivery at high serum content. J Gene Med 2015; 16:84-96. [PMID: 24753287 DOI: 10.1002/jgm.2762] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 04/14/2014] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Cationic liposome (CL)-DNA complexes are promising gene delivery vectors with potential application in gene therapy. A key challenge in creating CL-DNA complexes for application is that their transfection efficiency (TE) is adversely affected by serum. In particular, little is known about the effects of a high serum content on TE, even though this may provide design guidelines for application in vivo. METHODS We prepared CL-DNA complexes in which we varied the neutral lipid [1,2-dioleoyl-sn-glycerophosphatidylcholine, glycerol-monooleate (GMO), cholesterol], the headgroup charge and chemical structure of the cationic lipid, and the ratio of neutral to cationic lipid; we then measured the TE of these complexes as a function of serum content and assessed their cytotoxicity. We tested selected formulations in two human cancer cell lines (M21/melanoma and PC-3/prostate cancer). RESULTS In the absence of serum, all CL-DNA complexes of custom-synthesized multivalent lipids show high TE. Certain combinations of multivalent lipids and neutral lipids, such as MVL5(5+)/GMO-DNA complexes or complexes based on the dendritic-headgroup lipid TMVLG3(8+) exhibited high TE both in the absence and presence of serum. Although their TE still dropped to a small extent in the presence of serum, it reached or surpassed that of benchmark commercial transfection reagents, particularly at a high serum content. CONCLUSIONS Two-component vectors (one multivalent cationic lipid and one neutral lipid) can rival or surpass benchmark reagents at low and high serum contents (up to 50%, v/v). We propose guidelines for optimizing the serum resistance of CL-DNA complexes based on a given cationic lipid.
Collapse
Affiliation(s)
- Chia-Ling Chan
- Department of Materials, Department of Physics, and Molecular, Cellular & Developmental Biology Department, University of California at Santa Barbara, California 93106, USA.,Institute of Physics, Academia Sinica, Taipei 11529, Taiwan
| | - Kai K Ewert
- Department of Materials, Department of Physics, and Molecular, Cellular & Developmental Biology Department, University of California at Santa Barbara, California 93106, USA
| | - Ramsey N Majzoub
- Department of Materials, Department of Physics, and Molecular, Cellular & Developmental Biology Department, University of California at Santa Barbara, California 93106, USA
| | - Yeu-Kuang Hwu
- Institute of Physics, Academia Sinica, Taipei 11529, Taiwan
| | - Keng S Liang
- National Synchrotron Radiation Research Center, Hsinchu 30076, Taiwan.,Department of Electrophysics, National Chiao-Tung University, Hsinchu 30010, Taiwan
| | - Cecília Leal
- Department of Materials, Department of Physics, and Molecular, Cellular & Developmental Biology Department, University of California at Santa Barbara, California 93106, USA
| | - Cyrus R Safinya
- Department of Materials, Department of Physics, and Molecular, Cellular & Developmental Biology Department, University of California at Santa Barbara, California 93106, USA
| |
Collapse
|
26
|
Akita H, Ishiba R, Togashi R, Tange K, Nakai Y, Hatakeyama H, Harashima H. A neutral lipid envelope-type nanoparticle composed of a pH-activated and vitamin E-scaffold lipid-like material as a platform for a gene carrier targeting renal cell carcinoma. J Control Release 2014; 200:97-105. [PMID: 25543000 DOI: 10.1016/j.jconrel.2014.12.029] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 11/07/2014] [Accepted: 12/23/2014] [Indexed: 12/21/2022]
Abstract
A renal cell carcinoma (RCC) is one of the refractory tumors, since it readily acquires resistance against chemotherapy. Thus, alternative therapeutic approaches such as obstructing the neovasculature are needed. We previously reported on the development of a plasmid DNA (pDNA)-encapsulating liposomal nanoparticle (LNP) as a hepatic gene delivery system that is applicable to systemic administration. The key molecular component is a SS-cleavable and pH-activated lipid-like material (ssPalm) that mounts dual sensing motifs (ternary amines and disulfide bonding) that are responsive to the intracellular environment. The main purpose of the present study was to expand its application to a tumor-targeting gene delivery system in mice bearing tumors established from a RCC (OS-RC-2). When the modification of the surface of the particle is optimized for the polyethyleneglycol (PEG), stability in the blood circulation is improved, and consequently tumor-selective gene expression can be achieved. Furthermore, gene expression in the tumor was increased slightly when the hydrophobic scaffold of the ssPalm was replaced from the conventionally used myristic acid (ssPalmM) to α-tocopherol succinate (ssPalmE). Moreover, tumor growth was significantly suppressed when the completely CpG-free pDNA encoding the solute form of VEGFR (fms-like tyrosine kinase-1: sFlt-1) was used, especially when it was delivered by the LNP formed with ssPalmE (LNP(ssPalmE)). Thus, the PEG-modified LNP(ssPalmE) is a promising gene carrier for the cancer gene therapy of RCC.
Collapse
Affiliation(s)
- Hidetaka Akita
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Science, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo, Japan.
| | - Ryohei Ishiba
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Science, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo, Japan
| | - Ryohei Togashi
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Science, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo, Japan
| | - Kota Tange
- NOF CORPORATION, 3-3 Chidori-cho, Kawasaki-ku, Kawasaki, Kanagawa 210-0865, Japan
| | - Yuta Nakai
- NOF CORPORATION, 3-3 Chidori-cho, Kawasaki-ku, Kawasaki, Kanagawa 210-0865, Japan
| | - Hiroto Hatakeyama
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Science, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo, Japan
| | - Hideyoshi Harashima
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Science, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo, Japan.
| |
Collapse
|
27
|
Ukawa M, Akita H, Hayashi Y, Ishiba R, Tange K, Arai M, Kubo K, Higuchi Y, Shimizu K, Konishi S, Hashida M, Harashima H. Neutralized nanoparticle composed of SS-cleavable and pH-activated lipid-like material as a long-lasting and liver-specific gene delivery system. Adv Healthc Mater 2014; 3:1222-9. [PMID: 24668914 DOI: 10.1002/adhm.201300629] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Revised: 02/11/2014] [Indexed: 12/17/2022]
Abstract
Charge-neutralized lipid envelope-type nanoparticles formed with SS-cleavable and pH-activated lipid-like materials (ssPalm) accumulate rapidly in the liver without forming aggregates in the blood circulation, and result in a liver-specific gene expression for a long duration (>2 weeks) with neither immunological responses nor hepatotoxicity after intraveneous administration, when it carries pDNA free from CpG-motifs.
Collapse
Affiliation(s)
- Masami Ukawa
- Faculty of Pharmaceutical Sciences, Hokkaido University; Kita12 Nishi6, Kita-ku Sapporo City Hokkaido 060-0812 Japan
| | - Hidetaka Akita
- Faculty of Pharmaceutical Sciences, Hokkaido University; Kita12 Nishi6, Kita-ku Sapporo City Hokkaido 060-0812 Japan
| | - Yasuhiro Hayashi
- Faculty of Pharmaceutical Sciences, Hokkaido University; Kita12 Nishi6, Kita-ku Sapporo City Hokkaido 060-0812 Japan
| | - Ryohei Ishiba
- Faculty of Pharmaceutical Sciences, Hokkaido University; Kita12 Nishi6, Kita-ku Sapporo City Hokkaido 060-0812 Japan
| | - Kota Tange
- NOF Corporation; 3-3 Chidori-cho, Kawasaki-ku Kawasaki Kanagawa 210-0865 Japan
| | - Masaya Arai
- NOF Corporation; 3-3 Chidori-cho, Kawasaki-ku Kawasaki Kanagawa 210-0865 Japan
| | - Kazuhiro Kubo
- NOF Corporation; 3-3 Chidori-cho, Kawasaki-ku Kawasaki Kanagawa 210-0865 Japan
| | - Yuriko Higuchi
- Graduate School of Pharmaceutical Sciences, Kyoto University; 46-28 Yoshida-Shimo-Adachi-cho, Sakyo-ku Kyoto 606-8501 Japan
| | - Kazunori Shimizu
- Graduate School of Pharmaceutical Sciences, Kyoto University; 46-28 Yoshida-Shimo-Adachi-cho, Sakyo-ku Kyoto 606-8501 Japan
- Ritsumeikan-Global Innovation Research Organization, Ritsumeikan University; 1-1-1 Nojihigashi Kusatsu Shiga 525-8577 Japan
| | - Satoshi Konishi
- Ritsumeikan-Global Innovation Research Organization, Ritsumeikan University; 1-1-1 Nojihigashi Kusatsu Shiga 525-8577 Japan
- Department of Micro System Technology; Ritsumeikan University; 1-1-1 Nojihigashi Kusatsu Shiga 525-8577 Japan
| | - Mitsuru Hashida
- Graduate School of Pharmaceutical Sciences, Kyoto University; 46-28 Yoshida-Shimo-Adachi-cho, Sakyo-ku Kyoto 606-8501 Japan
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University; Yoshida-Ushinomiya-cho, Sakyo-ku Kyoto 606-8302 Japan
| | - Hideyoshi Harashima
- Faculty of Pharmaceutical Sciences, Hokkaido University; Kita12 Nishi6, Kita-ku Sapporo City Hokkaido 060-0812 Japan
| |
Collapse
|
28
|
Enhancing physical stability of positively charged catanionic vesicles in the presence of calcium chloride via cholesterol-induced fluidic bilayer characteristic. Colloid Polym Sci 2014. [DOI: 10.1007/s00396-014-3285-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
29
|
Dou C, Lay F, Ansari AM, Rees DJ, Ahmed AK, Kovbasnjuk O, Matsangos AE, Du J, Hosseini SM, Steenbergen C, Fox-Talbot K, Tabor AT, Williams JA, Liu L, Marti GP, Harmon JW. Strengthening the skin with topical delivery of keratinocyte growth factor-1 using a novel DNA plasmid. Mol Ther 2014; 22:752-61. [PMID: 24434934 PMCID: PMC3982499 DOI: 10.1038/mt.2014.2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 12/23/2013] [Indexed: 12/25/2022] Open
Abstract
Fragile skin, susceptible to decubitus ulcers and incidental trauma, is a problem particularly for the elderly and for those with spinal cord injury. Here, we present a simple approach to strengthen the skin by the topical delivery of keratinocyte growth factor-1 (KGF-1) DNA. In initial feasibility studies with the novel minimalized, antibiotic-free DNA expression vector, NTC8385-VA1, the reporter genes luciferase and enhanced green fluorescent protein were delivered. Transfection was documented when luciferase expression significantly increased after transfection. Microscopic imaging of enhanced green fluorescent protein-transfected skin showed green fluorescence in hair follicles, hair shafts, and dermal and superficial epithelial cells. With KGF-1 transfection, KGF-1 mRNA level and protein production were documented with quantitative reverse transcriptase-polymerase chain reaction and immunohistochemistry, respectively. Epithelial thickness of the transfected skin in the KGF group was significantly increased compared with the control vector group (26 ± 2 versus 16 ± 4 µm) at 48 hours (P = 0.045). Dermal thickness tended to be increased in the KGF group (255 ± 36 versus 162 ± 16 µm) at 120 hours (P = 0.057). Biomechanical assessment showed that the KGF-1-treated skin was significantly stronger than control vector-transfected skin. These findings indicate that topically delivered KGF-1 DNA plasmid can increase epithelial thickness and strength, demonstrating the potential of this approach to restore compromised skin.
Collapse
Affiliation(s)
- Chunqing Dou
- 1] Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chinese PLA General Hospital, Beijing, China [2] Department of Surgery and Hendrix Burn/Wound Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Frank Lay
- Department of Surgery and Hendrix Burn/Wound Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Amir Mehdi Ansari
- Department of Surgery and Hendrix Burn/Wound Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Donald J Rees
- Department of Surgery and Hendrix Burn/Wound Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ali Karim Ahmed
- Department of Surgery and Hendrix Burn/Wound Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Olga Kovbasnjuk
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Aerielle E Matsangos
- Department of Surgery and Hendrix Burn/Wound Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Junkai Du
- Department of Surgery and Hendrix Burn/Wound Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sayed Mohammad Hosseini
- Department of Surgery and Hendrix Burn/Wound Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Charles Steenbergen
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Karen Fox-Talbot
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | - Lixin Liu
- Department of Surgery and Hendrix Burn/Wound Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Guy P Marti
- Department of Surgery and Hendrix Burn/Wound Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - John W Harmon
- Department of Surgery and Hendrix Burn/Wound Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
30
|
Christensen CL, Zandi R, Gjetting T, Cramer F, Poulsen HS. Specifically targeted gene therapy for small-cell lung cancer. Expert Rev Anticancer Ther 2014; 9:437-52. [DOI: 10.1586/era.09.10] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
31
|
Tong HP, Wang LF, Guo YL, Li L, Fan XZ, Ding J, Huang HY. Preparation of protamine cationic nanobubbles and experimental study of their physical properties and in vivo contrast enhancement. ULTRASOUND IN MEDICINE & BIOLOGY 2013; 39:2147-2157. [PMID: 23932278 DOI: 10.1016/j.ultrasmedbio.2013.05.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 05/17/2013] [Accepted: 05/24/2013] [Indexed: 06/02/2023]
Abstract
In this study, we aimed to prepare a novel type of microbubble (MB), protamine cationic nanobubble (NB), to provide a new vector for tumor gene therapy. We prepared cationic NBs with protamine and other lipid components using mechanical oscillation. The protamine cationic NBs had a mean diameter of 521.2 ± 37.57 nm, a zeta potential of +18.5 mV, and a gene-carrying capacity of 15.69 μg androgen receptor (AR) siRNA per 10(8) NBs. The cationic NBs exhibited superior contrast enhancement for in vivo imaging compared with SonoVue (Bracco, Geneva, Switzerland), and their physical properties did not change significantly after 1 wk; meanwhile, the transfection efficiency of the cationic NBs in androgen-independent prostate cancer cells mediated by ultrasound irradiation was better than that of liposomes (82.17 ± 7.4% vs. 55.04 ± 5.4%, p < 0.01). Therefore, the protamine cationic NB can be considered for use as a novel type of gene-loading MB for ultrasound imaging and MB-mediated gene therapy of tumors.
Collapse
Affiliation(s)
- Hai-Peng Tong
- Department of Ultrasound, Southwest Hospital, Third Military Medical University, Chongqing, China
| | | | | | | | | | | | | |
Collapse
|
32
|
Tam YYC, Chen S, Cullis PR. Advances in Lipid Nanoparticles for siRNA Delivery. Pharmaceutics 2013; 5:498-507. [PMID: 24300520 PMCID: PMC3836621 DOI: 10.3390/pharmaceutics5030498] [Citation(s) in RCA: 145] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 09/04/2013] [Accepted: 09/12/2013] [Indexed: 01/21/2023] Open
Abstract
Technological advances in both siRNA (small interfering RNA) and whole genome sequencing have demonstrated great potential in translating genetic information into siRNA-based drugs to halt the synthesis of most disease-causing proteins. Despite its powerful promises as a drug, siRNA requires a sophisticated delivery vehicle because of its rapid degradation in the circulation, inefficient accumulation in target tissues and inability to cross cell membranes to access the cytoplasm where it functions. Lipid nanoparticle (LNP) containing ionizable amino lipids is the leading delivery technology for siRNA, with five products in clinical trials and more in the pipeline. Here, we focus on the technological advances behind these potent systems for siRNA-mediated gene silencing.
Collapse
Affiliation(s)
- Yuen Yi C Tam
- Department of Biochemistry and Molecular Biology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, B.C. V6T 1Z3, Canada.
| | | | | |
Collapse
|
33
|
Bishop CJ, Ketola TM, Tzeng SY, Sunshine JC, Urtti A, Lemmetyinen H, Vuorimaa-Laukkanen E, Yliperttula M, Green JJ. The effect and role of carbon atoms in poly(β-amino ester)s for DNA binding and gene delivery. J Am Chem Soc 2013; 135:6951-7. [PMID: 23570657 PMCID: PMC3838887 DOI: 10.1021/ja4002376] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Polymeric vectors for gene delivery are a promising alternative for clinical applications, as they are generally safer than viral counterparts. Our objective was to further our mechanistic understanding of polymer structure-function relationships to allow the rational design of new biomaterials. Utilizing poly(β-amino ester)s (PBAEs), we investigated polymer-DNA binding by systematically varying the polymer molecular weight, adding single carbons to the backbone and side chain of the monomers that constitute the polymers, and varying the type of polymer end group. We then sought to correlate how PBAE binding affects the polyplex diameter and ζ potential, the transfection efficacy, and its associated cytotoxicity in human breast and brain cancer cells in vitro. Among other trends, we observed in both cell lines that the PBAE-DNA binding constant is biphasic with the transfection efficacy and that the optimal values of the binding constant with respect to the transfection efficacy are in the range (1-6) × 10(4) M(-1). A binding constant in this range is necessary but not sufficient for effective transfection.
Collapse
Affiliation(s)
- Corey J. Bishop
- Department of Biomedical Engineering, the Wilmer Eye Institute, the Institute for Nanobiotechnology, and the Translational Tissue Engineering Center. Johns Hopkins University School of Medicine 400 North Broadway, Baltimore, MD 21231, USA
| | - Tiia-Maaria Ketola
- Department of Chemistry and Bioengineering, Tampere University of Technology, P.O. Box 541, FI33101, Finland
| | - Stephany Y. Tzeng
- Department of Biomedical Engineering, the Wilmer Eye Institute, the Institute for Nanobiotechnology, and the Translational Tissue Engineering Center. Johns Hopkins University School of Medicine 400 North Broadway, Baltimore, MD 21231, USA
| | - Joel C. Sunshine
- Department of Biomedical Engineering, the Wilmer Eye Institute, the Institute for Nanobiotechnology, and the Translational Tissue Engineering Center. Johns Hopkins University School of Medicine 400 North Broadway, Baltimore, MD 21231, USA
| | - Arto Urtti
- Divison of Biopharmacy and Pharmacokinetics, University of Helsinki Viikinkaari 5E, 00014 Helsinki, Finland
| | - Helge Lemmetyinen
- Department of Chemistry and Bioengineering, Tampere University of Technology, P.O. Box 541, FI33101, Finland
| | - Elina Vuorimaa-Laukkanen
- Department of Chemistry and Bioengineering, Tampere University of Technology, P.O. Box 541, FI33101, Finland
| | - Marjo Yliperttula
- Centre for Drug Research, Faculty of Pharmacy, University of Helsinki Viikinkaari 5E, 00014 Helsinki, Finland
| | - Jordan J. Green
- Department of Biomedical Engineering, the Wilmer Eye Institute, the Institute for Nanobiotechnology, and the Translational Tissue Engineering Center. Johns Hopkins University School of Medicine 400 North Broadway, Baltimore, MD 21231, USA
| |
Collapse
|
34
|
Induction of apoptosis by chitosan/HPV16 E7 siRNA complexes in cervical cancer cells. Mol Med Rep 2012; 7:998-1002. [DOI: 10.3892/mmr.2012.1246] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2012] [Accepted: 12/06/2012] [Indexed: 11/05/2022] Open
|
35
|
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: 18.6] [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
|
36
|
The effect of cholesterol domains on PEGylated liposomal gene delivery in vitro. Ther Deliv 2012; 2:451-60. [PMID: 22428082 DOI: 10.4155/tde.11.13] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
AIM PEGylated components have been widely used to reduce particle aggregation in serum and extend circulation lifetime for lipid- and polymer-based gene-delivery systems. However, PEGylation is known to interfere with cell interaction and intracellular trafficking, resulting in decreased biological activity. In the present study, the effect of cholesterol domains on PEGylated liposome-mediated gene delivery was evaluated by PEGylating formulations with and without a cholesterol domain, and also by altering the location of PEG on the particle surface (i.e., within or excluded from the domain). MATERIALS AND METHODS Lipoplexes formulated with PEG-cholesterol or PEG-diacyl lipid were used to transfect various cell lines, including human and mouse cancer cells. Cellular uptake of lipoplexes was also quantified and compared with the transfection results. RESULTS Our findings are consistent with previous work demonstrating that PEGylation reduces transfection rates; however, formulations in which PEG was incorporated into the cholesterol domain did not exhibit this detrimental effect. In some cell lines, the incorporation of PEG into the domain actually increased transfection rates, despite no enhancement of cellular uptake. DISCUSSION These results suggest that the adverse alterations in intracellular trafficking that are a consequence of PEGylation may be avoided by utilizing delivery vehicles that allow PEG to partition into a cholesterol domain.
Collapse
|
37
|
Chan CL, Majzoub RN, Shirazi RS, Ewert KK, Chen YJ, Liang KS, Safinya CR. Endosomal escape and transfection efficiency of PEGylated cationic liposome-DNA complexes prepared with an acid-labile PEG-lipid. Biomaterials 2012; 33:4928-35. [PMID: 22469293 DOI: 10.1016/j.biomaterials.2012.03.038] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Accepted: 03/10/2012] [Indexed: 01/27/2023]
Abstract
Cationic liposome-DNA (CL-DNA) complexes are being pursued as nonviral gene delivery systems for use in applications that include clinic trials. However, to compete with viral vectors for systemic delivery in vivo, their efficiencies and pharmacokinetics need to be improved. The addition of poly (ethylene glycol)-lipids (PEGylation) prolongs circulation lifetimes of liposomes, but inhibits cellular uptake and endosomal escape of CL-DNA complexes. We show that this limits their transfection efficiency (TE) in a manner dependent on the amount of PEG-lipid, the lipid/DNA charge ratio, and the lipid membrane charge density. To improve endosomal escape of PEGylated CL-DNA complexes, we prepared an acid-labile PEG-lipid (HPEG2K-lipid, PEG MW 2000) which is designed to lose its PEG chains at the pH of late endosomes. The HPEG2K-lipid and a similar but acid-stable PEG-lipid were used to prepare PEGylated CL-DNA complexes. TLC and dynamic light scattering showed that HPEG2K-CL-DNA complexes are stable at pH 7.4 for more than 24 h, but the PEG chains are cleaved at pH 5 within 1 h, leading to complex aggregation. The acid-labile HPEG2K-CL-DNA complexes showed enhanced TE over complexes stabilized with the acid-stable PEG-lipid. Live-cell imaging showed that both types of complexes were internalized to quantitatively similar particle distributions within the first 2 h of incubation with cells. Thus, we attribute the increased TE of the HPEG2K-CL-DNA complexes to efficient endosomal escape, enabled by the acid-labile HPEG2K-lipid which sheds its PEG chains in the low pH environment of late endosomes, effectively switching on the electrostatic interactions that promote fusion of the membranes of complex and endosome.
Collapse
Affiliation(s)
- Chia-Ling Chan
- Department of Materials, University of California, Santa Barbara, CA 93106, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Angelov B, Angelova A, Filippov S, Karlsson G, Terrill N, Lesieur S, Štěpánek P. SAXS Study of Sterically Stabilized Lipid Nanocarriers Functionalized by DNA. ACTA ACUST UNITED AC 2012. [DOI: 10.1088/1742-6596/351/1/012004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
39
|
Abstract
SiRNA is the trigger of RNA interference, a mechanism discovered in the late 1990s. To release the therapeutic potential of this versatile but large and fragile molecule, excipients are used which either interact by electrostatic interaction, passively encapsulate siRNA or are covalently attached to enable specific and safe delivery of the drug substance. Controlling the delicate balance between protective complexation and release of siRNA at the right point and time is done by understanding excipients–siRNA interactions. These can be lipids, polymers such as PEI, PLGA, Chitosans, Cyclodextrins, as well as aptamers and peptides. This review describes the mechanisms of interaction of the most commonly used siRNA delivery vehicles, and looks at the results of their clinical and preclinical studies.
Collapse
Affiliation(s)
- Katharina Bruno
- Novartis Pharma AG, Technical Research & Development (TRD), Pharmaceutical and Analytical Development (PHAD), CH-4057 Basel, Switzerland.
| |
Collapse
|
40
|
Gjetting T, Andresen TL, Christensen CL, Cramer F, Poulsen TT, Poulsen HS. A simple protocol for preparation of a liposomal vesicle with encapsulated plasmid DNA that mediate high accumulation and reporter gene activity in tumor tissue. RESULTS IN PHARMA SCIENCES 2011; 1:49-56. [PMID: 25755981 DOI: 10.1016/j.rinphs.2011.08.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Revised: 08/11/2011] [Accepted: 08/12/2011] [Indexed: 11/30/2022]
Abstract
The systemic delivery of gene therapeutics by non-viral methods has proven difficult. Transfection systems that are performing well in vitro have been reported to have disadvantageous properties such as rapid clearance and short circulation time often resulting in poor transfection efficiency when applied in vivo. Large unilaminary vesicles (LUV) with encapsulated nucleic acids designated stabilized-plasmid-lipo-particle (SPLP) have showed promising results in terms of systemic stability and accumulation in tumor tissue due to the enhanced permeability and retention effect (EPR). We have developed a simple protocol for the research-scale preparation of SPLPs from commercially available reagents with high amounts of encapsulated plasmid DNA. The SPLPs show properties of promising accumulation in tumor tissue in comparison to other organs when intravenously injected into xenograft tumor-bearing nude mice. Although transcriptionally targeted suicide gene therapy was not achieved, the SPLPs were capable of mediating reporter gene transfection in subcutaneous flank tumors originating from human small cell lung cancer.
Collapse
Affiliation(s)
- Torben Gjetting
- Department of Radiation Biology, section 6321, Finsen Center, Copenhagen University Hospital, Copenhagen, Denmark ; Department of Micro- and Nanotechnology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Thomas Lars Andresen
- Department of Micro- and Nanotechnology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Camilla Laulund Christensen
- Department of Radiation Biology, section 6321, Finsen Center, Copenhagen University Hospital, Copenhagen, Denmark
| | - Frederik Cramer
- Department of Radiation Biology, section 6321, Finsen Center, Copenhagen University Hospital, Copenhagen, Denmark
| | - Thomas Tuxen Poulsen
- Department of Radiation Biology, section 6321, Finsen Center, Copenhagen University Hospital, Copenhagen, Denmark
| | - Hans Skovgaard Poulsen
- Department of Radiation Biology, section 6321, Finsen Center, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
41
|
Kuntsche J, Horst JC, Bunjes H. Cryogenic transmission electron microscopy (cryo-TEM) for studying the morphology of colloidal drug delivery systems. Int J Pharm 2011; 417:120-37. [DOI: 10.1016/j.ijpharm.2011.02.001] [Citation(s) in RCA: 169] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Revised: 01/29/2011] [Accepted: 02/01/2011] [Indexed: 01/25/2023]
|
42
|
Wang T, Upponi JR, Torchilin VP. Design of multifunctional non-viral gene vectors to overcome physiological barriers: dilemmas and strategies. Int J Pharm 2011; 427:3-20. [PMID: 21798324 DOI: 10.1016/j.ijpharm.2011.07.013] [Citation(s) in RCA: 192] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Revised: 07/11/2011] [Accepted: 07/12/2011] [Indexed: 12/31/2022]
Abstract
Gene-based therapeutics hold great promise for medical advancement and have been used to treat various human diseases with mixed success. However, their therapeutic application in vivo is limited due largely to several physiological barriers. The design of non-viral gene vectors with the ability to overcome delivery obstacles is currently under extensive investigation. These efforts have placed an emphasis on the development of multifunctional vectors able to execute multiple tasks to simultaneously overcome both extracellular and intracellular obstacles. However, the assembly of these different functionalities into a single system to create multifunctional gene vectors faces many conflicts that largely limit the safe and efficient application of lipoplexes and polyplexes in a systemic delivery. In the review, we have described the dilemmas inherent in the design of a viable, non-viral gene vector equipped with multiple functionalities. The strategies directed towards individual delivery barriers are first summarized, followed by a focus on the design of so-called smart multifunctional vectors with the capability to overcome the delivery difficulties of gene medicines, including the so-called the "polycation dilemma", the "PEG dilemma" and the "package and release dilemma".
Collapse
Affiliation(s)
- Tao Wang
- Center for Pharmaceutical Biotechnology and Nanomedicine, 312 Mugar Life Sciences Building, 360 Huntington Avenue, Northeastern University, Boston, MA 02115, USA
| | | | | |
Collapse
|
43
|
Kim MH, Na HK, Kim YK, Ryoo SR, Cho HS, Lee KE, Jeon H, Ryoo R, Min DH. Facile synthesis of monodispersed mesoporous silica nanoparticles with ultralarge pores and their application in gene delivery. ACS NANO 2011; 5:3568-3576. [PMID: 21452883 DOI: 10.1021/nn103130q] [Citation(s) in RCA: 243] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Among various nanoparticles, the silica nanoparticle (SiNP) is an attractive candidate as a gene delivery carrier due to advantages such as availability in porous forms for encapsulation of drugs and genes, large surface area to load biomacromolecules, biocompatibility, storage stability, and easy preparation in large quantity with low cost. Here, we report on a facile synthesis of monodispersed mesoporous silica nanoparticles (MMSN) possessing very large pores (>15 nm) and application of the nanoparticles to plasmid DNA delivery to human cells. The aminated MMSN with large pores provided a higher loading capacity for plasmids than those with small pores (∼2 nm), and the complex of MMSN with plasmid DNA readily entered into cells without supplementary polymers such as cationic dendrimers. Furthermore, MMSN with large pores could efficiently protect plasmids from nuclease-mediated degradation and showed much higher transfection efficiency of the plasmids encoding luciferase and green fluorescent protein (pLuc, pGFP) compared to MMSN with small pores (∼2 nm).
Collapse
Affiliation(s)
- Mi-Hee Kim
- Department of Chemistry, KAIST, Daejeon 305-701, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Xu L, Anchordoquy T. Drug delivery trends in clinical trials and translational medicine: challenges and opportunities in the delivery of nucleic acid-based therapeutics. J Pharm Sci 2011; 100:38-52. [PMID: 20575003 DOI: 10.1002/jps.22243] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The ability to deliver nucleic acids (e.g., plasmid DNA, antisense oligonucleotides, siRNA) offers the potential to develop potent vaccines and novel therapeutics. However, nucleic acid-based therapeutics are still in their early stages as a new category of biologics. The efficacy of nucleic acids requires that these molecules be delivered to the interior of the target cell, which greatly complicates delivery strategies and compromises efficiency. Due to the safety concerns of viral vectors, synthetic vectors such as liposomes and polymers are preferred for the delivery of nucleic acid-based therapeutics. Yet, delivery efficiencies of synthetic vectors in the clinic are still too low to obtain therapeutic levels of gene expression. In this review, we focus on some key issues in the field of nucleic acid delivery such as PEGylation, encapsulation and targeted delivery and provide some perspectives for consideration in the development of improved synthetic vectors.
Collapse
Affiliation(s)
- Long Xu
- Department of Pharmaceutical Sciences, University of Colorado, 12700 East Nineteenth Avenue, Aurora, Colorado 80045, USA
| | | |
Collapse
|
45
|
Xu L, Anchordoquy TJ. Effect of cholesterol nanodomains on the targeting of lipid-based gene delivery in cultured cells. Mol Pharm 2010; 7:1311-7. [PMID: 20568694 DOI: 10.1021/mp100097b] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Targeted gene delivery offers immense potential for clinical applications. Liposomes decorated with targeting ligands have been extensively used for both in vitro and in vivo gene delivery. Lipoplexes with high cholesterol content that result in cholesterol domain formation within the complexes have been shown to exhibit enhanced transfection in vitro and resistance to serum-induced aggregation. In the present study, folate was employed as a targeting ligand that was conjugated with either cholesterol or a diacyl lipid (DSPE), and these conjugates were incorporated into lipoplexes formulated with DOTAP/cholesterol (wt/wt: 31/69) that are known to possess cholesterol nanodomains. Cellular uptake and transfection of these lipoplexes in the presence of 50% serum were examined when the ligand was located within or excluded from the cholesterol nanodomain. Lipoplexes with folate-cholesterol exhibited a 50-fold increase in transfection compared to those with folate-DSPE, while the cellular uptake level is only 40% of that with folate-DSPE. These results indicate that the presence of the ligand within the cholesterol domain promotes more productive transfection in cultured cells, and intracellular trafficking of the lipoplexes after entry into cells plays a crucial role in gene delivery.
Collapse
Affiliation(s)
- Long Xu
- School of Pharmacy, University of Colorado Denver, 12700 East 19th Avenue, Aurora, Colorado 80045, USA
| | | |
Collapse
|
46
|
Gjetting T, Arildsen NS, Christensen CL, Poulsen TT, Roth JA, Handlos VN, Poulsen HS. In vitro and in vivo effects of polyethylene glycol (PEG)-modified lipid in DOTAP/cholesterol-mediated gene transfection. Int J Nanomedicine 2010; 5:371-83. [PMID: 20957159 PMCID: PMC2950395 DOI: 10.2147/ijn.s10462] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2010] [Indexed: 11/23/2022] Open
Abstract
Background: DOTAP/cholesterol-based lipoplexes are successfully used for delivery of plasmid DNA in vivo especially to the lungs, although low systemic stability and circulation have been reported. To achieve the aim of discovering the best method for systemic delivery of DNA to disseminated tumors we evaluated the potential of formulating DOTAP/cholesterol lipoplexes with a polyethylene glycol (PEG)-modified lipid, giving the benefit of the shielding and stabilizing properties of PEG in the bloodstream. Method: A direct comparison of properties in vitro and in vivo of 4 different DOTAP/cholesterol-based lipoplexes containing 0%, 2%, 4%, and 10% PEG was performed using reporter gene activity and radioactive tracer lipid markers to monitor biodistribution. Results: We found that 10% PEGylation of lipoplexes caused reduced retention in lung and heart tissues of nude mice compared to nonPEGylated lipoplexes, however no significant delivery to xenograft flank tumors was observed. Although PEGylated and nonPEGylated lipoplexes were delivered to cells the ability to mediate successful transfection is hampered upon PEGylation, presumably due to a changed uptake mechanism and intracellular processing. Conclusion: The eminent in vivo transfection potency of DOTAP/cholesterol-based lipoplexes is well established for expression in lung tumors, but it is unsuitable for expression in non first pass organs such as xenograft flank tumors in mice even after addition of a PEG-lipid in the formulation.
Collapse
Affiliation(s)
- Torben Gjetting
- Department of Radiation Biology, Finsen Center, University Hospital, Copenhagen, Denmark
| | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
Cancer remains a leading cause of morbidity and mortality. Despite advances in understanding, detection, and treatment, it accounts for almost one-fourth of all deaths per year in Western countries. Prostate cancer is currently the most commonly diagnosed noncutaneous cancer in men in Europe and the United States, accounting for 15% of all cancers in men. As life expectancy of individuals increases, it is expected that there will also be an increase in the incidence and mortality of prostate cancer. Prostate cancer may be inoperable at initial presentation, unresponsive to chemotherapy and radiotherapy, or recur following appropriate treatment. At the time of presentation, patients may already have metastases in their tissues. Preventing tumor recurrence requires systemic therapy; however, current modalities are limited by toxicity or lack of efficacy. For patients with such metastatic cancers, the development of alternative therapies is essential. Gene therapy is a realistic prospect for the treatment of prostate and other cancers, and involves the delivery of genetic information to the patient to facilitate the production of therapeutic proteins. Therapeutics can act directly (eg, by inducing tumor cells to produce cytotoxic agents) or indirectly by upregulating the immune system to efficiently target tumor cells or by destroying the tumor's vasculature. However, technological difficulties must be addressed before an efficient and safe gene medicine is achieved (primarily by developing a means of delivering genes to the target cells or tissue safely and efficiently). A wealth of research has been carried out over the past 20 years, involving various strategies for the treatment of prostate cancer at preclinical and clinical trial levels. The therapeutic efficacy observed with many of these approaches in patients indicates that these treatment modalities will serve as an important component of urological malignancy treatment in the clinic, either in isolation or in combination with current approaches.
Collapse
Affiliation(s)
- Mark Tangney
- Cork Cancer Research Centre, Mercy University Hospital, Cork, Ireland.
| | | | | | | |
Collapse
|
48
|
Chen X, Zhang Y, Liu C, Zhang Y, Zhou X, Zhou T, Mao Y, Kan B, Wei YQ, Li J. Retinoic acid and dimethyl sulfoxide promote efficient delivery of transgenes to mouse skin by topically transdermal penetration. Drug Deliv 2010; 17:385-90. [DOI: 10.3109/10717541003762862] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
49
|
Wang KW, Zhou LZ, Sun Y, Wu GJ, Gu HC, Duan YR, Chen F, Zhu YJ. Calcium phosphate/PLGA-mPEG hybrid porous nanospheres: A promising vector with ultrahigh gene loading and transfection efficiency. ACTA ACUST UNITED AC 2010. [DOI: 10.1039/b917441a] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
50
|
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: 13.3] [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]
|