1
|
Aghajanpour S, Amiriara H, Ebrahimnejad P, Slavcev RA. Advancing ocular gene therapy: a machine learning approach to enhance delivery, uptake and gene expression. Drug Discov Today 2025; 30:104359. [PMID: 40228736 DOI: 10.1016/j.drudis.2025.104359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 03/30/2025] [Accepted: 04/09/2025] [Indexed: 04/16/2025]
Abstract
Ocular gene therapy offers a promising approach for treating various eye diseases, centered on the process of transfection, including delivery, cellular uptake and gene expression. This study addresses anatomical and physiological barriers, such as the eyelids, tear film, conjunctiva, cornea, sclera, choroid and retina, affecting therapeutic success. A three-step machine-learning approach is proposed. The first step predicts gene delivery efficacy by integrating molecular characteristics of the ocular gene therapy product, ocular barrier properties and patient demographics. The second step predicts cellular uptake rates, analyzing product penetration and cellular interactions. The final step forecasts gene expression levels, considering factors like nucleic acid type and endosomal escape. An artificial neural network model is recommended to capture complex, nonlinear relationships, enhancing our understanding of therapeutic and biological interactions.
Collapse
Affiliation(s)
- Sareh Aghajanpour
- Pharmaceutical Sciences Research Center, Institute of Herbal Medicines and Metabolic Disorders, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hamid Amiriara
- Department of Electrical Engineering, Faculty of Engineering and Technology, University of Mazandaran, Mazandaran, Iran
| | - Pedram Ebrahimnejad
- Pharmaceutical Sciences Research Center, Institute of Herbal Medicines and Metabolic Disorders, Mazandaran University of Medical Sciences, Sari, Iran; Centre for Eye and Vision Research, Unit 901-903, Building 17W, Hong Kong Science Park, Pak Shek Kok, Shatin, Hong Kong; School of Pharmacy, University of Waterloo, 10A Victoria St S, Kitchener N2G 1C5, Canada.
| | - Roderick A Slavcev
- Centre for Eye and Vision Research, Unit 901-903, Building 17W, Hong Kong Science Park, Pak Shek Kok, Shatin, Hong Kong; School of Pharmacy, University of Waterloo, 10A Victoria St S, Kitchener N2G 1C5, Canada; Mediphage Bioceuticals, 661 University Avenue, Suite 1300, Toronto, ON M5G 0B7, Canada.
| |
Collapse
|
2
|
George M, Boukherroub R, Sanyal A, Szunerits S. Treatment of lung diseases via nanoparticles and nanorobots: Are these viable alternatives to overcome current treatments? Mater Today Bio 2025; 31:101616. [PMID: 40124344 PMCID: PMC11930446 DOI: 10.1016/j.mtbio.2025.101616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/25/2025] Open
Abstract
Challenges Respiratory diseases remain challenging to treat, with current efforts primarily focused on managing symptoms rather than maintaining overall lung health. Traditional treatment methods, such as oral or parenteral administration of antiviral, antibacterial, and anti-inflammatory drugs, face limitations. These include difficulty in delivering therapeutic agents to pathogens residing deep in the airways and the risk of severe side effects due to high systemic drug concentrations. The growing threat of drug-resistant pathogens further complicates infection management. Advancements The lung's large surface area offers an attractive target for inhalation-based drug delivery. Nanoparticles (NP) enable uniform and sustained drug distribution across the alveolar network, overcoming challenges posed by complex lung anatomy. Recent breakthroughs in nanorobots (NR) have demonstrated precise navigation through biological environments, delivering therapies directly to affected lung areas with enhanced accuracy. Nanotechnology has also shown promise in treating lung cancer, with nanoparticles engineered to overcome biological barriers, improve drug solubility, and enable controlled drug release. Future scope This review explores the progress of NP and NR in addressing challenges in pulmonary drug delivery. These innovations allow targeted delivery of nucleic acids, drugs, or peptides to the pulmonary epithelium with unprecedented accuracy, offering significant potential for improving therapeutic effectiveness in respiratory disorders.
Collapse
Affiliation(s)
- Meekha George
- Laboratory for Life Sciences and Technology (LiST), Faculty of Medicine and Dentistry, Danube Private University (DPU), Viktor-Kaplan-Straße 2, Geb. E, 2700, Wiener Neustadt, Austria
| | - Rabah Boukherroub
- Univ. Lille, CNRS, Univ. Polytechnique, Hauts-de-France, UMR 8520 - IEMN, F-59000, Lille, France
| | - Amitav Sanyal
- Department of Chemistry, Bogazici University, Bebek, 34342, Istanbul, Turkey
| | - Sabine Szunerits
- Laboratory for Life Sciences and Technology (LiST), Faculty of Medicine and Dentistry, Danube Private University (DPU), Viktor-Kaplan-Straße 2, Geb. E, 2700, Wiener Neustadt, Austria
- Univ. Lille, CNRS, Univ. Polytechnique, Hauts-de-France, UMR 8520 - IEMN, F-59000, Lille, France
| |
Collapse
|
3
|
Bayraktutan H, Symonds P, Brentville VA, Moloney C, Galley C, Bennett CL, Mata A, Durrant L, Alexander C, Gurnani P. Sparsely PEGylated poly(beta-amino ester) polyplexes enhance antigen specific T-cell response of a bivalent SARS-CoV-2 DNA vaccine. Biomaterials 2024; 311:122647. [PMID: 38878479 DOI: 10.1016/j.biomaterials.2024.122647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 08/06/2024]
Abstract
DNA technology has emerged as a promising route to accelerated manufacture of sequence agnostic vaccines. For activity, DNA vaccines must be protected and delivered to the correct antigen presenting cells. However, the physicochemical properties of the vector must be carefully tuned to enhance interaction with immune cells and generate sufficient immune response for disease protection. In this study, we have engineered a range of polymer-based nanocarriers based on the poly(beta-amino ester) (PBAE) polycation platform to investigate the role that surface poly(ethylene glycol) (PEG) density has on pDNA encapsulation, formulation properties and gene transfectability both in vitro and in vivo. We achieved this by synthesising a non-PEGylated and PEGylated PBAE and produced formulations containing these PBAEs, and mixed polyplexes to tune surface PEG density. All polymers and co-formulations produced small polyplex nanoparticles with almost complete encapsulation of the cargo in all cases. Despite high gene transfection in HEK293T cells, only the fully PEGylated and mixed formulations displayed significantly higher expression of the reporter gene than the negative control in dendritic cells. Further in vivo studies with a bivalent SARS-CoV-2 pDNA vaccine revealed that only the mixed formulation led to strong antigen specific T-cell responses, however this did not translate into the presence of serum antibodies indicating the need for further studies into improving immunisation with polymer delivery systems.
Collapse
Affiliation(s)
- Hulya Bayraktutan
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK; Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Peter Symonds
- Scancell Ltd, University of Nottingham Biodiscovery Institute, Nottingham, NG7 2RD, UK
| | - Victoria A Brentville
- Scancell Ltd, University of Nottingham Biodiscovery Institute, Nottingham, NG7 2RD, UK
| | - Cara Moloney
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK; Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Charlotte Galley
- Department of Haematology, UCL Cancer Institute, 72 Huntley Street, University College London, London, WC1E 6DD, UK
| | - Clare L Bennett
- Department of Haematology, UCL Cancer Institute, 72 Huntley Street, University College London, London, WC1E 6DD, UK
| | - Alvaro Mata
- Division of Regenerative Medicine and Cellular Therapies, School of Pharmacy, University of Nottingham, NG7 2RD, UK; Department of Chemical and Environmental Engineering, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Lindy Durrant
- Scancell Ltd, University of Nottingham Biodiscovery Institute, Nottingham, NG7 2RD, UK
| | - Cameron Alexander
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK.
| | - Pratik Gurnani
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK.
| |
Collapse
|
4
|
Norimatsu J, Mizuno HL, Watanabe T, Obara T, Nakakido M, Tsumoto K, Cabral H, Kuroda D, Anraku Y. Triphenylphosphonium-modified catiomers enhance in vivo mRNA delivery through stabilized polyion complexation. MATERIALS HORIZONS 2024; 11:4711-4721. [PMID: 38988276 DOI: 10.1039/d4mh00325j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
Nanocarriers based on cationic materials play a central role in the success of mRNA-based therapies. Traditionally, amine-bearing lipids and polymers have been successfully employed for creating mRNA-loaded nanocarriers, though they still present challenges, such as physical and biological instability, limiting both delivery efficiency and therapeutic potential. Non-amine cations could be a promising avenue in addressing these limitations. However, such alternatives remain notably underexplored. Herein, we introduced triphenylphosphonium (TPP) as an alternative cationic moiety for mRNA delivery, leveraging its advantageous properties for nucleic acid complexation. Through the modification of amine-bearing catiomers, we replaced traditional amine-based counterparts with TPP to create innovative polymeric micelles as mRNA nanocarriers. A comprehensive analysis, encompassing physicochemical, thermodynamic, and computational approaches, revealed that the TPP substitution significantly influenced polymer self-assembly, mRNA binding, and the overall stability of mRNA-loaded polymeric micelles. Upon intravenous injection, TPP-bearing micelles demonstrated a remarkable increase in mRNA bioavailability, facilitating efficient protein production in solid tumors. These findings provide a compelling rationale for substituting amines with TPP, emphasizing their potential for advancing mRNA therapeutics.
Collapse
Affiliation(s)
- Jumpei Norimatsu
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| | - Hayato L Mizuno
- Department of Materials Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology, 2-12-1 Ookayama, Meguro-ku, Tokyo 152-8550, Japan.
| | - Takayoshi Watanabe
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| | - Takumi Obara
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| | - Makoto Nakakido
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Kouhei Tsumoto
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
- The Institute of Medical Sciences, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kanagawa 210-0821, Japan
| | - Daisuke Kuroda
- Research Center of Drug and Vaccine Development, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan.
| | - Yasutaka Anraku
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
- Department of Materials Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology, 2-12-1 Ookayama, Meguro-ku, Tokyo 152-8550, Japan.
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kanagawa 210-0821, Japan
| |
Collapse
|
5
|
Qiu B, Manzanares D, Li Y, Wang X, Li Z, Terreau S, He Z, Lyu J, Wang W, Lara-Sáez I. Highly branched poly β-amino ester/CpG-depleted CFTR plasmid nanoparticles for non-viral gene therapy in lung cystic fibrosis disease. Mol Ther Methods Clin Dev 2024; 32:101292. [PMID: 40017666 PMCID: PMC11866167 DOI: 10.1016/j.omtm.2024.101292] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 06/21/2024] [Indexed: 03/01/2025]
Abstract
Lung cystic fibrosis (CF) is a lethal inherited disease caused by mutations in the CF transmembrane conductance regulator (CFTR) gene, leading to a dysfunctional CFTR protein. Gene therapy offers promise for the treatment of lung CF. However, the development and clinical application of CF gene therapy have long been hampered by the absence of safe and highly efficient delivery vectors. In this work, a novel polymer-based gene replacement treatment approach was developed. A series of poly (β-amino esters) (PAEs) with various topological structures and chemical compositions were screened to create non-viral therapeutic systems for CFTR restoration in lung CF disease. A nanoparticle, formed by the selected highly branched PAE (HPAE) with a CpG-depleted CFTR plasmid, demonstrated CFTR gene expression and biocompatibility in lung epithelial cells, outperforming leading commercial gene transfection reagents such as Lipofectamine 3000 and Xfect. The newly developed gene therapy system successfully restored functional CFTR protein production in lung CF epithelial monolayers. This therapeutic approach holds great potential for use as an efficient and safe non-viral treatment for CF patients.
Collapse
Affiliation(s)
- Bei Qiu
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Dublin D04V1W8, Ireland
| | - Darío Manzanares
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Dublin D04V1W8, Ireland
| | - Yinghao Li
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Dublin D04V1W8, Ireland
| | - Xianqing Wang
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Dublin D04V1W8, Ireland
| | - Zishan Li
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Dublin D04V1W8, Ireland
| | - Sébastien Terreau
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Dublin D04V1W8, Ireland
| | - Zhonglei He
- Institute of Precision Medicine (AUST-IPM), Anhui University of Science and Technology, Huainan 232001, China
- School of Public Health, Anhui University of Science and Technology, Huainan 232001, China
| | - Jing Lyu
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Dublin D04V1W8, Ireland
| | - Wenxin Wang
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Dublin D04V1W8, Ireland
- Institute of Precision Medicine (AUST-IPM), Anhui University of Science and Technology, Huainan 232001, China
- School of Public Health, Anhui University of Science and Technology, Huainan 232001, China
| | - Irene Lara-Sáez
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Dublin D04V1W8, Ireland
| |
Collapse
|
6
|
Turuvekere Vittala Murthy N, Vlasova K, Renner J, Jozic A, Sahay G. A new era of targeting cystic fibrosis with non-viral delivery of genomic medicines. Adv Drug Deliv Rev 2024; 209:115305. [PMID: 38626860 DOI: 10.1016/j.addr.2024.115305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 03/27/2024] [Accepted: 04/09/2024] [Indexed: 04/21/2024]
Abstract
Cystic fibrosis (CF) is a complex genetic respiratory disorder that necessitates innovative gene delivery strategies to address the mutations in the gene. This review delves into the promises and challenges of non-viral gene delivery for CF therapy and explores strategies to overcome these hurdles. Several emerging technologies and nucleic acid cargos for CF gene therapy are discussed. Novel formulation approaches including lipid and polymeric nanoparticles promise enhanced delivery through the CF mucus barrier, augmenting the potential of non-viral strategies. Additionally, safety considerations and regulatory perspectives play a crucial role in navigating the path toward clinical translation of gene therapy.
Collapse
Affiliation(s)
| | - Kseniia Vlasova
- Department of Pharmaceutical Sciences, College of Pharmacy at Oregon State University, Corvallis, OR 97331, USA
| | - Jonas Renner
- Department of Pharmaceutical Sciences, College of Pharmacy at Oregon State University, Corvallis, OR 97331, USA
| | - Antony Jozic
- Department of Pharmaceutical Sciences, College of Pharmacy at Oregon State University, Corvallis, OR 97331, USA
| | - Gaurav Sahay
- Department of Pharmaceutical Sciences, College of Pharmacy at Oregon State University, Corvallis, OR 97331, USA; Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR 97201, USA; Department of Biomedical Engineering, Robertson Life Sciences Building, Oregon Health & Science University, Portland, OR 97201, USA.
| |
Collapse
|
7
|
Feng X, Shi Y, Zhang Y, Lei F, Ren R, Tang X. Opportunities and Challenges for Inhalable Nanomedicine Formulations in Respiratory Diseases: A Review. Int J Nanomedicine 2024; 19:1509-1538. [PMID: 38384321 PMCID: PMC10880554 DOI: 10.2147/ijn.s446919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 01/24/2024] [Indexed: 02/23/2024] Open
Abstract
Lungs experience frequent interactions with the external environment and have an abundant supply of blood; therefore, they are susceptible to invasion by pathogenic microorganisms and tumor cells. However, the limited pharmacokinetics of conventional drugs in the lungs poses a clinical challenge. The emergence of different nano-formulations has been facilitated by advancements in nanotechnology. Inhaled nanomedicines exhibit better targeting and prolonged therapeutic effects. Although nano-formulations have great potential, they still present several unknown risks. Herein, we review the (1) physiological anatomy of the lungs and their biological barriers, (2) pharmacokinetics and toxicology of nanomaterial formulations in the lungs; (3) current nanomaterials that can be applied to the respiratory system and related design strategies, and (4) current applications of inhaled nanomaterials in treating respiratory disorders, vaccine design, and imaging detection based on the characteristics of different nanomaterials. Finally, (5) we analyze and summarize the challenges and prospects of nanomaterials for respiratory disease applications. We believe that nanomaterials, particularly inhaled nano-formulations, have excellent prospects for application in respiratory diseases. However, we emphasize that the simultaneous toxic side effects of biological nanomaterials must be considered during the application of these emerging medicines. This study aims to offer comprehensive guidelines and valuable insights for conducting research on nanomaterials in the domain of the respiratory system.
Collapse
Affiliation(s)
- Xujun Feng
- Department of Respiratory and Critical Care Medicine, Sleep Medicine Center, Mental Health Center, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Yuan Shi
- Department of Respiratory and Critical Care Medicine, Sleep Medicine Center, Mental Health Center, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Ye Zhang
- Department of Respiratory and Critical Care Medicine, Sleep Medicine Center, Mental Health Center, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Fei Lei
- Department of Respiratory and Critical Care Medicine, Sleep Medicine Center, Mental Health Center, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Rong Ren
- Department of Respiratory and Critical Care Medicine, Sleep Medicine Center, Mental Health Center, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Xiangdong Tang
- Department of Respiratory and Critical Care Medicine, Sleep Medicine Center, Mental Health Center, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| |
Collapse
|
8
|
Sp S, Mitra RN, Zheng M, Chrispell JD, Wang K, Kwon YS, Weiss ER, Han Z. Gene augmentation for autosomal dominant retinitis pigmentosa using rhodopsin genomic loci nanoparticles in the P23H +/- knock-in murine model. Gene Ther 2023; 30:628-640. [PMID: 36935427 DOI: 10.1038/s41434-023-00394-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 02/13/2023] [Accepted: 02/28/2023] [Indexed: 03/21/2023]
Abstract
Gene therapy for autosomal dominant retinitis pigmentosa (adRP) is challenged by the dominant inheritance of the mutant genes, which would seemingly require a combination of mutant suppression and wild-type replacement of the appropriate gene. We explore the possibility that delivery of a nanoparticle (NP)-mediated full-length mouse genomic rhodopsin (gRho) or human genomic rhodopsin (gRHO) locus can overcome the dominant negative effects of the mutant rhodopsin in the clinically relevant P23H+/--knock-in heterozygous mouse model. Our results demonstrate that mice in both gRho and gRHO NP-treated groups exhibit significant structural and functional recovery of the rod photoreceptors, which lasted for 3 months post-injection, indicating a promising reduction in photoreceptor degeneration. We performed miRNA transcriptome analysis using next generation sequencing and detected differentially expressed miRNAs as a first step towards identifying miRNAs that could potentially be used as rhodopsin gene expression enhancers or suppressors for sustained photoreceptor rescue. Our results indicate that delivering an intact genomic locus as a transgene has a greater chance of success compared to the use of the cDNA for treatment of this model of adRP, emphasizing the importance of gene augmentation using a gDNA that includes regulatory elements.
Collapse
Affiliation(s)
- Simna Sp
- Department of Ophthalmology, the University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Rajendra N Mitra
- Department of Ophthalmology, the University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Min Zheng
- Department of Ophthalmology, the University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Jared D Chrispell
- Department of Cell Biology and Physiology, the University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Kai Wang
- Department of Ophthalmology, the University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Yong-Su Kwon
- Department of Ophthalmology, the University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Ellen R Weiss
- Department of Cell Biology and Physiology, the University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Zongchao Han
- Department of Ophthalmology, the University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Carolina Institute for NanoMedicine, the University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Division of Pharmacoengineering & Molecular Pharmaceutics, Eshelman School of Pharmacy, the University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
9
|
Kwak G, Lee D, Suk JS. Advanced approaches to overcome biological barriers in respiratory and systemic routes of administration for enhanced nucleic acid delivery to the lung. Expert Opin Drug Deliv 2023; 20:1531-1552. [PMID: 37946533 PMCID: PMC10872418 DOI: 10.1080/17425247.2023.2282535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 11/08/2023] [Indexed: 11/12/2023]
Abstract
INTRODUCTION Numerous delivery strategies, primarily novel nucleic acid delivery carriers, have been developed and explored to enable therapeutically relevant lung gene therapy. However, its clinical translation is yet to be achieved despite over 30 years of efforts, which is attributed to the inability to overcome a series of biological barriers that hamper efficient nucleic acid transfer to target cells in the lung. AREAS COVERED This review is initiated with the fundamentals of nucleic acid therapy and a brief overview of previous and ongoing efforts on clinical translation of lung gene therapy. We then walk through the nature of biological barriers encountered by nucleic acid carriers administered via respiratory and/or systemic routes. Finally, we introduce advanced strategies developed to overcome those barriers to achieve therapeutically relevant nucleic acid delivery efficiency in the lung. EXPERT OPINION We are now stepping close to the clinical translation of lung gene therapy, thanks to the discovery of novel delivery strategies that overcome biological barriers via comprehensive preclinical studies. However, preclinical findings should be cautiously interpreted and validated to ultimately realize meaningful therapeutic outcomes with newly developed delivery strategies in humans. In particular, individual strategies should be selected, tailored, and implemented in a manner directly relevant to specific therapeutic applications and goals.
Collapse
Affiliation(s)
- Gijung Kwak
- Department of Neurosurgery and Medicine Institute for Neuroscience Discovery (UM-MIND), University of Maryland School of Medicine, Baltimore, MD, USA
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daiheon Lee
- Department of Neurosurgery and Medicine Institute for Neuroscience Discovery (UM-MIND), University of Maryland School of Medicine, Baltimore, MD, USA
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jung Soo Suk
- Department of Neurosurgery and Medicine Institute for Neuroscience Discovery (UM-MIND), University of Maryland School of Medicine, Baltimore, MD, USA
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
10
|
Uskoković V. Lessons from the history of inorganic nanoparticles for inhalable diagnostics and therapeutics. Adv Colloid Interface Sci 2023; 315:102903. [PMID: 37084546 DOI: 10.1016/j.cis.2023.102903] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 04/04/2023] [Accepted: 04/10/2023] [Indexed: 04/23/2023]
Abstract
The respiratory tract is one of the most accessible ones to exogenous nanoparticles, yet drug delivery by their means to it is made extraordinarily challenging because of the plexus of aerodynamic, hemodynamic and biomolecular factors at cellular and extracellular levels that synergistically define the safety and efficacy of this process. Here, the use of inorganic nanoparticles (INPs) for inhalable diagnostics and therapies of the lung is viewed through the prism of the history of studies on the interaction of INPs with the lower respiratory tract. The most conceptually and methodologically innovative and illuminative studies are referred to in the chronological order, as they were reported in the literature, and the trends in the progress of understanding this interaction of immense therapeutic and toxicological significance are being deduced from it. The most outstanding actual trends delineated include the diminishment of toxicity via surface functionalization, cell targeting, tagging and tracking via controlled binding and uptake, hybrid INP treatments, magnetic guidance, combined drug and gene delivery, use as adjuvants in inhalable vaccines, and other. Many of the understudied research directions, which have been accomplished by the nanostructured organic polymers in the pulmonary niche, are discussed. The progress in the use of INPs as inhalable diagnostics or therapeutics has been hampered by their well-recognized inflammatory potential and toxicity in the respiratory tract. However, the annual numbers of methodologically innovative studies have been on the rise throughout the past two decades, suggesting that this is a prolific direction of research, its comparatively poor commercial takings notwithstanding. Still, the lack of consensus on the effects of many INP compositions at low but therapeutically effective doses, the plethora of contradictory reports on ostensibly identical chemical compositions and NP properties, and the many cases of antagonism in combinatorial NP treatments imply that the rational design of inhalable medical devices based on INPs must rely on qualitative principles for the most part and embrace a partially stochastic approach as well. At the same time, the fact that the most studied INPs for pulmonary applications have been those with some of the thickest records of pulmonary toxicity, e.g., carbon, silver, gold, silica and iron oxide, is a silent call for the expansion of the search for new inorganic compositions for use in inhalable therapies to new territories.
Collapse
Affiliation(s)
- Vuk Uskoković
- Advanced Materials and Nanobiotechnology Laboratory, TardigradeNano LLC, 7 Park Vista, Irvine, CA 92604, USA; Department of Mechanical Engineering, San Diego State University, 5500 Campanile Dr., San Diego, CA 92182, USA.
| |
Collapse
|
11
|
Kole E, Jadhav K, Sirsath N, Dudhe P, Verma RK, Chatterjee A, Naik J. Nanotherapeutics for pulmonary drug delivery: An emerging approach to overcome respiratory diseases. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
|
12
|
Lee D, Kwak G, Johnson TV, Suk JS. Formulation and Evaluation of Polymer-Based Nanoparticles for Intravitreal Gene-Delivery Applications. Curr Protoc 2022; 2:e607. [PMID: 36469609 PMCID: PMC9731353 DOI: 10.1002/cpz1.607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The advent of the first-ever retinal gene therapy product, involving subretinal administration of a virus-based gene delivery platform, has garnered hope that this state-of-the-art therapeutic modality may benefit a broad spectrum of patients with diverse retinal disorders. On the other hand, clinical studies have revealed limitations of the applied delivery strategy that may restrict its universal use. To this end, intravitreal administration of synthetic gene-delivery platforms, such as polymer-based nanoparticles (PNPs), has emerged as an attractive alternative to the current mainstay. To achieve success, however, it is imperative that synthetic platforms overcome key biological barriers in human eyes encountered following intravitreal administration, including the vitreous gel and inner limiting membrane (ILM). Here, we introduce a series of experiments, from the fabrication of PNPs to a comprehensive evaluation in relevant experimental models, to determine whether PNPs overcome these barriers and efficiently deliver therapeutic gene payloads to retinal cells. We conclude the article by discussing a few important considerations for successful implementation of the strategy. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Preparation and characterization of PNPs Basic Protocol 2: Evaluation of in vitro transfection efficacy Basic Protocol 3: Evaluation of PNP diffusion in vitreous gel Basic Protocol 4: Ex vivo assessment of PNP penetration within vitreoretinal explant culture Basic Protocol 5: Assessment of in vivo transgene expression mediated by intravitreally administered PNPs.
Collapse
Affiliation(s)
- Daiheon Lee
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- These authors contributed equally to this work
| | - Gijung Kwak
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- These authors contributed equally to this work
| | - Thomas V. Johnson
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jung Soo Suk
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
13
|
Maurya S, Srivastava R, Arfin S, Hawthorne S, Jha NK, Agrawal K, Raj S, Rathi B, Kumar A, Raj R, Agrawal S, Paiva-Santos AC, Malik AA, Dua K, Rana R, Ojha S, Jha SK, Sharma A, Kumar D, El-Zahaby SA, Nagar A. Exploring state-of-the-art advances in targeted nanomedicines for managing acute and chronic inflammatory lung diseases. Nanomedicine (Lond) 2022; 17:2245-2264. [PMID: 36975758 DOI: 10.2217/nnm-2021-0437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/30/2023] [Indexed: 03/29/2023] Open
Abstract
Diagnosis and treatment of lung diseases pose serious challenges. Currently, diagnostic as well as therapeutic methods show poor efficacy toward drug-resistant bacterial infections, while chemotherapy causes toxicity and nonspecific delivery of drugs. Advanced treatment methods that cure lung-related diseases, by enabling drug bioavailability via nasal passages during mucosal formation, which interferes with drug penetration to targeted sites, are in demand. Nanotechnology confers several advantages. Currently, different nanoparticles, or their combinations, are being used to enhance targeted drug delivery. Nanomedicine, a combination of nanoparticles and therapeutic agents, that delivers drugs to targeted sites increases the bioavailability of drugs at these sites. Thus, nanotechnology is superior to conventional chemotherapeutic strategies. Here, the authors review the latest advancements in nanomedicine-based drug-delivery methods for managing acute and chronic inflammatory lung diseases.
Collapse
Affiliation(s)
- Sujata Maurya
- School of Health Sciences & Technology, UPES University, Dehradun, Uttarakhand, 248007, India
| | - Rashi Srivastava
- Chemical & Biochemical Engineering, Indian Institute of Technology, Patna, 801106, India
| | - Saniya Arfin
- School of Health Sciences & Technology, UPES University, Dehradun, Uttarakhand, 248007, India
| | - Susan Hawthorne
- SAAD Building, School of Pharmacy & Pharmaceutical Sciences, Ulster University, Coleraine, BT52 1SA, UK
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Greater Noida, Uttar Pradesh, 201310, India
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Kirti Agrawal
- School of Health Sciences & Technology, UPES University, Dehradun, Uttarakhand, 248007, India
| | - Sibi Raj
- School of Health Sciences & Technology, UPES University, Dehradun, Uttarakhand, 248007, India
| | - Brijesh Rathi
- Department of Chemistry, Hansraj College, Delhi University, New Delhi, 110007, Delhi, India
| | - Arun Kumar
- Mahavir Cancer Institute & Research Centre Patna, Bihar, 800002, India
| | - Riya Raj
- Department of Biochemistry, Bangalore University, Bangalore, 560056, Karnataka, India
| | - Sharad Agrawal
- Department of Life Sciences, School of Basic Science & Research, Sharda University, Greater Noida, Uttar Pradesh, 201310, India
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal
| | - Asrar Ahmad Malik
- Department of Life Sciences, School of Basic Science & Research, Sharda University, Greater Noida, Uttar Pradesh, 201310, India
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW, 2007, Australia
| | - Rakesh Rana
- MSD, HILLEMAN LABS, Analytical Division, New Delhi, 110062, India
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Greater Noida, Uttar Pradesh, 201310, India
- Department of Biotechnology Engineering & Food Technology, Chandigarh University, Mohali, 140413, India
- Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun 248007, India
| | - Ankur Sharma
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Cathedral Street, Glasgow, G10RE, Scotland, UK
| | - Dhruv Kumar
- School of Health Sciences & Technology, UPES University, Dehradun, Uttarakhand, 248007, India
| | - Sally A El-Zahaby
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt
| | - Amka Nagar
- Department of Life Sciences, School of Basic Science & Research, Sharda University, Greater Noida, Uttar Pradesh, 201310, India
| |
Collapse
|
14
|
Prasher P, Sharma M, Singh SK, Gulati M, Jha NK, Gupta PK, Gupta G, Chellappan DK, Zacconi F, de Jesus Andreoli Pinto T, Chan Y, Liu G, Paudel K, Hansbro PM, George Oliver BG, Dua K. Targeting mucus barrier in respiratory diseases by chemically modified advanced delivery systems. Chem Biol Interact 2022; 365:110048. [PMID: 35932910 DOI: 10.1016/j.cbi.2022.110048] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 06/30/2022] [Accepted: 07/13/2022] [Indexed: 11/26/2022]
Abstract
Mucus gel constitutes of heavily cross-linked mucin fibers forming a viscoelastic, dense porous network that coats all the exposed epithelia not covered with the skin. The layer provides protection to the underlying gastrointestinal, respiratory, and female reproductive tracts, in addition to the organs such as the surface of eye by trapping the pathogens, irritants, environmental fine particles, and potentially hazardous foreign matter. However, this property of mucus gel poses a substantial challenge for realizing the localized and sustained drug delivery across the mucosal surfaces. The mucus permeating particles that spare the protective properties of mucus gel improve the therapeutic potency of the drugs aimed at the management of diseases, including sexually transmitted infections, lung cancer, irritable bowel disease, degenerative eye diseases and infections, and cystic fibrosis. As such, the mucoadhesive materials conjugated with drug molecules display a prolonged retention time in the mucosal gel that imparts a sustained release of the deliberated drug molecules across the mucosa. The contemporarily developed mucus penetrating materials for drug delivery applications comprise of a finer size, appreciable hydrophilicity, and a neutral surface to escape the entrapment within the cross-inked mucus fibers. Pertaining to the mucus secretion as a first line of defence in respiratory tract in response to the invading physical, chemical, and biological pathogens, the development of mucus penetrating materials hold promise as a stalwart approach for revolutionizing the respiratory drug delivery paradigm. The present review provides an epigrammatic collation of the mucus penetrating/mucoadhesive materials for achieving a controlled/sustained release of the cargo pharmaceutics and drug molecules across the respiratory mucus barrier.
Collapse
Affiliation(s)
- Parteek Prasher
- Department of Chemistry, University of Petroleum & Energy Studies, Dehradun, 248007, India
| | - Mousmee Sharma
- Department of Chemistry, Uttaranchal University, Dehradun, 248007, India
| | - Sachin Kumar Singh
- School of Pharmacy and Pharmaceutical Science, Lovely Professional University, India
| | - Monica Gulati
- School of Pharmacy and Pharmaceutical Science, Lovely Professional University, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology (SET), Sharda University, Greater Noida, 201310, UP, India
| | - Piyush Kumar Gupta
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, 201310, Uttar Pradesh, India
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jaipur, Rajasthan, India
| | - Dinesh Kumar Chellappan
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur, 57000, Malaysia
| | - Flavia Zacconi
- Departamento de Quimica Organica, Facultad de Quimica y de Farmacia, Pontificia Universidad Catolica de Chile, Av. Vicuna Mackenna 4860, Macul, Santiago, 7820436, Chile; Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, 7820436, Chile
| | - Terezinha de Jesus Andreoli Pinto
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of São Paulo, Professor Lineu Prestes Street, São Paulo, 05508-000, Brazil
| | - Yinghan Chan
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur, 57000, Malaysia
| | - Gang Liu
- Centre for Inflammation, Centenary Institute, Sydney, NSW 2050, Australia; School of Life Sciences, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Keshav Paudel
- Centre for Inflammation, Centenary Institute, Sydney, NSW 2050, Australia; School of Life Sciences, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute, Sydney, NSW 2050, Australia; School of Life Sciences, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Brian Gregory George Oliver
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW, 2007, Australia; Woolcock Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia.
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW, 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia.
| |
Collapse
|
15
|
Yamasaki Y, Kumekawa D, Yamauchi S, Omuro H. Re-examination of Peptide-Sequence-Dependent Gene Expression of Cysteine-Installed Pegylated Oligolysine/DNA Complexes. ACS OMEGA 2022; 7:15478-15487. [PMID: 35571853 PMCID: PMC9096824 DOI: 10.1021/acsomega.2c00122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 04/08/2022] [Indexed: 06/15/2023]
Abstract
We previously synthesized cysteine-installed C-terminally PEGylated oligolysines with 20 amino acid residues to form cross-linked polymeric micelles (PMs) with luciferase-coding plasmid DNA as a candidate for artificial gene vectors. Luciferase gene expression in HeLa cells mediated by PEG-CK18C, PEG-CK9CK9, and PEG-K9CK9C was reported to be 35-, 5.4-, and 1.3-fold higher than that mediated by cysteine-uninstalled PEGylated oligolysine PEG-K20, respectively. However, after the publication, the survival rate of HeLa cells used in the previous study was found to be lower than usual when subcutaneously implanted into mice to create a xenograft model. In this study, to re-examine the peptide sequence-dependent gene expression, gene expression efficacy mediated by PEG-peptide PMs was compared with the PM cellular uptake results using newly obtained HeLa cell lines and the additional cell lines Huh-7, PANC-1, and BxPC3. As a result, PEG-K9CK9C PMs mediated the maximum gene expression in all cell lines, and the corresponding cellular uptake was also obtained. Therefore, we concluded that our previous results were erroneously obtained due to normality-depleted HeLa cells. A comparison of physicochemical characterizations, gene expression efficacy, and cellular uptake of PEG-peptide PMs is discussed in detail.
Collapse
|
16
|
Application of bioengineered elastin-like polypeptide-based system for targeted gene delivery in tumor cells. BIOMATERIALS AND BIOSYSTEMS 2022; 6:100050. [PMID: 36824163 PMCID: PMC9934475 DOI: 10.1016/j.bbiosy.2022.100050] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 03/28/2022] [Accepted: 04/15/2022] [Indexed: 11/20/2022] Open
Abstract
Successful gene delivery depends on the entry of negatively charged DNAs and oligonucleotides across the various barriers of the tumor cells and localization into the nucleus for its transcription and protein translation. Here, we have reported a thermal responsive self-assemble and highly biocompatible, targeted ELP-based gene delivery system. These systems consist of cell-penetrating peptides, Tat and single or multiple repeats of IL-4 receptor targeting peptide AP-1 along the backbone of ELP. Cell-penetrating peptides were introduced for nuclear localization of genes of interest, AP-1 for targeting IL-4R highly expressed tumor cells and ELP for stable condensation favoring protection of nucleic acids. The designed multidomain fusion ELPs referred to as Tat-ELP, Tat-A1E28 and Tat-A4V48 were employed to generate formulation with pEGFP-N1. Profound formulation of stable complexes occurred at different molar ratios owing to electrostatic interactions of positively charged amino acids in polymers with negatively charged nucleic acids. Among the complexes, Tat-A4V48 containing four copies of AP-1 showed maximum complexation with pEGFP-N1 in lower molar ratio. The polymer-pEGFP complexes were further analyzed for its transfection efficiency in different cancer cell lines. Both the targeted polymers, Tat-A4V48 and Tat-A1E28 upon transfection displayed significant EGFP-expression with low toxicity in different cancer cells. Therefore, both Tat-A4V48 and Tat-A1E28 can be considered as novel transfection system for successful gene delivery with therapeutic applications.
Collapse
|
17
|
Bisserier M, Sun XQ, Fazal S, Turnbull IC, Bonnet S, Hadri L. Novel Insights into the Therapeutic Potential of Lung-Targeted Gene Transfer in the Most Common Respiratory Diseases. Cells 2022; 11:984. [PMID: 35326434 PMCID: PMC8947048 DOI: 10.3390/cells11060984] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/08/2022] [Accepted: 03/10/2022] [Indexed: 12/10/2022] Open
Abstract
Over the past decades, a better understanding of the genetic and molecular alterations underlying several respiratory diseases has encouraged the development of new therapeutic strategies. Gene therapy offers new therapeutic alternatives for inherited and acquired diseases by delivering exogenous genetic materials into cells or tissues to restore physiological protein expression and/or activity. In this review, we review (1) different types of viral and non-viral vectors as well as gene-editing techniques; and (2) the application of gene therapy for the treatment of respiratory diseases and disorders, including pulmonary arterial hypertension, idiopathic pulmonary fibrosis, cystic fibrosis, asthma, alpha-1 antitrypsin deficiency, chronic obstructive pulmonary disease, non-small-cell lung cancer, and COVID-19. Further, we also provide specific examples of lung-targeted therapies and discuss the major limitations of gene therapy.
Collapse
Affiliation(s)
- Malik Bisserier
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY 10029, USA; (M.B.); (S.F.); (I.C.T.)
| | - Xiao-Qing Sun
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands;
| | - Shahood Fazal
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY 10029, USA; (M.B.); (S.F.); (I.C.T.)
| | - Irene C. Turnbull
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY 10029, USA; (M.B.); (S.F.); (I.C.T.)
| | - Sébastien Bonnet
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec, QC G1V4G5, Canada;
- Department of Medicine, Laval University, Québec, QC G1V4G5, Canada
| | - Lahouaria Hadri
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY 10029, USA; (M.B.); (S.F.); (I.C.T.)
| |
Collapse
|
18
|
Saw PE, Cui GH, Xu X. Nanoparticles-mediated CRISPR/Cas gene editing delivery system. ChemMedChem 2022; 17:e202100777. [PMID: 35261159 DOI: 10.1002/cmdc.202100777] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 03/06/2022] [Indexed: 11/09/2022]
Abstract
The CRISPR/Cas gene editing system utilizes CRISPR RNA to guide the endonuclease in specifically breaking target gene, and then repairs genomic DNA by the means of homology directed repair (HDR) and non-homologous end joining (NHEJ). The gene editing system can only play its role in gene editing when it enters the nucleus. This crucial step in the process of gene editing is the major hurdle to gene therapy as it is still a huge challenge to efficiently deliver the CRISPR/Cas system to target tissues and cells. The low delivery efficiency hinders the clinical transformation of this technology. At present, delivery systems mainly include physical methods, viral vectors, and non-viral vectors. Due to the advantages of nanomaterial, it is currently being used rapidly in developing non-viral delivery systems. This review focuses on the mechanism of CRISPR/Cas and the delivery of gene editing system, following the research progress of nanoparticle-mediated gene editing.
Collapse
Affiliation(s)
- Phei Er Saw
- Sun Yat-Sen Memorial Hospital, 107 West Yanjiang Road, 510000, Guangzhou, CHINA
| | - Guo-Hui Cui
- Sun Yat-sen University Zhongshan School of Medicine, Bio-safety Laboratory, CHINA
| | - Xiaoding Xu
- Sun Yat-Sen Memorial Hospital, Medical Research Center, CHINA
| |
Collapse
|
19
|
Aerosol-Mediated Non-Viral Lung Gene Therapy: The Potential of Aminoglycoside-Based Cationic Liposomes. Pharmaceutics 2021; 14:pharmaceutics14010025. [PMID: 35056921 PMCID: PMC8778791 DOI: 10.3390/pharmaceutics14010025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/29/2021] [Accepted: 12/14/2021] [Indexed: 11/17/2022] Open
Abstract
Aerosol lung gene therapy using non-viral delivery systems represents a credible therapeutic strategy for chronic respiratory diseases, such as cystic fibrosis (CF). Progress in CF clinical setting using the lipidic formulation GL67A has demonstrated the relevance of such a strategy while emphasizing the need for more potent gene transfer agents. In recent years, many novel non-viral gene delivery vehicles were proposed as potential alternatives to GL67 cationic lipid. However, they were usually evaluated using procedures difficult or even impossible to implement in clinical practice. In this study, a clinically-relevant administration protocol via aerosol in murine lungs was used to conduct a comparative study with GL67A. Diverse lipidic compounds were used to prepare a series of formulations inspired by the composition of GL67A. While some of these formulations were ineffective at transfecting murine lungs, others demonstrated modest-to-very-efficient activities and a series of structure-activity relationships were unveiled. Lipidic aminoglycoside derivative-based formulations were found to be at least as efficient as GL67A following aerosol delivery of a luciferase-encoding plasmid DNA. A single aerosol treatment with one such formulation was found to mediate long-term lung transgene expression, exceeding half the animal's lifetime. This study clearly supports the potential of aminoglycoside-based cationic lipids as potent GL67-alternative scaffolds for further enhanced aerosol non-viral lung gene therapy for diseases such as CF.
Collapse
|
20
|
Liu Y, Wu W, Wang Y, Han S, Yuan Y, Huang J, Shuai X, Peng Z. Recent development of gene therapy for pancreatic cancer using non-viral nanovectors. Biomater Sci 2021; 9:6673-6690. [PMID: 34378568 DOI: 10.1039/d1bm00748c] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Pancreatic cancer (PC), characterized by its dense desmoplastic stroma and hypovascularity, is one of the most lethal cancers with a poor prognosis in the world. Traditional treatments such as chemotherapy, radiotherapy, and targeted therapy show little benefit in the survival rate in patients with advanced PC due to the poor penetration and resistance of drugs, low radiosensitivity, or severe side effects. Gene therapy can modify the morbific and drug-resistant genes as well as insert the tumor-suppressing genes, which has been shown to have great potential in PC treatment. The development of safe non-viral vectors for the highly efficient delivery of nucleic acids is essential for effective gene therapy, and has been attracting much attention. In this review, we first summarized the PC-promoting genes and gene therapies using plasmid DNA, mRNA, miRNA/siRNA-based RNA interference technology, and genome editing technology. Second, the commonly used non-viral nanovector and theranostic gene delivery nanosystem, especially the tumor microenvironment-sensitive delivery nanosystem and the cell/tumor-penetrating delivery nanosystem, were introduced. Third, a combination of non-viral nanovector-based gene therapy and other therapies, such as immunotherapy, chemotherapy, photothermal therapy (PTT), and photodynamic therapy (PDT), for PDAC treatment was discussed. Finally, a number of clinical trials have demonstrated the proof-of-principle that gene therapy or the combination of gene therapy and chemotherapy using non-viral vectors can inhibit the progression of PC. Although most of the non-viral vector-based gene therapies and their combination therapy are still under preclinical research, the development of genetics, molecular biology, and novel vectors would promote the clinical transformation of gene therapy.
Collapse
Affiliation(s)
- Yu Liu
- Department of Medical Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| | - Wei Wu
- Department of Medical Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| | - Yiyao Wang
- Department of Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China.
| | - Shisong Han
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, China.
| | - Yuanyuan Yuan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jinsheng Huang
- Department of Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China.
| | - Xintao Shuai
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, China.
| | - Zhao Peng
- Department of Medical Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| |
Collapse
|
21
|
Sanchez-Martos M, Martinez-Navarrete G, Bernabeu-Zornoza A, Humphreys L, Fernandez E. Evaluation and Optimization of Poly-d-Lysine as a Non-Natural Cationic Polypeptide for Gene Transfer in Neuroblastoma Cells. NANOMATERIALS 2021; 11:nano11071756. [PMID: 34361142 PMCID: PMC8308159 DOI: 10.3390/nano11071756] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 06/26/2021] [Accepted: 06/29/2021] [Indexed: 11/17/2022]
Abstract
Cationic polypeptides and cationic polymers have cell-penetrating capacities and have been used in gene transfer studies. In this study, we investigate the capability of a polymer of d-lysine (PDL), a chiral form of α–Poly-lysine, as a possible nonviral vector for releasing genetic materials to neuroblastoma cells and evaluate its stability against proteases. We tested and compared its transfection effectiveness in vitro as a vehicle for the EGFP plasmid DNA (pDNA) reporter in the SH-SY5Y human neuroblastoma, HeLa, and 3T3 cell lines. Using fluorescent microscopy and flow cytometry, we demonstrated high transfection efficiencies based on EGFP fluorescence in SH-SY5Y cells, compared with HeLa and 3T3. Our results reveal PDL as an efficient vector for gene delivery specifically in the SH-SY5Y cell line and suggest that PDL can be used as a synthetic cell-penetrating polypeptide for gene therapy in neuroblastoma cells.
Collapse
Affiliation(s)
- Miguel Sanchez-Martos
- Neuroprothesis and Neuroengineering Research Group, Miguel Hernández University, 03201 Elche, Spain; (M.S.-M.); (G.M.-N.); (A.B.-Z.); (L.H.)
| | - Gema Martinez-Navarrete
- Neuroprothesis and Neuroengineering Research Group, Miguel Hernández University, 03201 Elche, Spain; (M.S.-M.); (G.M.-N.); (A.B.-Z.); (L.H.)
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Adela Bernabeu-Zornoza
- Neuroprothesis and Neuroengineering Research Group, Miguel Hernández University, 03201 Elche, Spain; (M.S.-M.); (G.M.-N.); (A.B.-Z.); (L.H.)
| | - Lawrence Humphreys
- Neuroprothesis and Neuroengineering Research Group, Miguel Hernández University, 03201 Elche, Spain; (M.S.-M.); (G.M.-N.); (A.B.-Z.); (L.H.)
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Eduardo Fernandez
- Neuroprothesis and Neuroengineering Research Group, Miguel Hernández University, 03201 Elche, Spain; (M.S.-M.); (G.M.-N.); (A.B.-Z.); (L.H.)
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-965222001
| |
Collapse
|
22
|
Non-adhesive and highly stable biodegradable nanoparticles that provide widespread and safe transgene expression in orthotopic brain tumors. Drug Deliv Transl Res 2021; 10:572-581. [PMID: 32323162 DOI: 10.1007/s13346-020-00759-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Several generations of poly(β-amino ester) (PBAE) polymers have been developed for efficient cellular transfection. However, PBAE-based gene vectors, similar to other cationic materials, cannot readily provide widespread gene transfer in the brain due to adhesive interactions with the extracellular matrix (ECM). We thus engineered eight vector candidates using previously identified lead PBAE polymer variants but endowed them with non-adhesive surface coatings to facilitate their spread through brain ECM. Specifically, we screened for the ability to provide widespread gene transfer in tumor spheroids and healthy mouse brains. We then confirmed that a lead formulation provided widespread transgene expression in orthotopically established brain tumor models with an excellent in vivo safety profile. Lastly, we developed a method to store it long-term while fully retaining its brain-penetrating property. This new platform provides a broad utility in evaluating novel genetic targets for gene therapy of brain tumors and neurological disorders in preclinical and clinical settings. Graphical abstract We engineered biodegradable DNA-loaded brain-penetrating nanoparticles (DNA-BPN) possessing small particle diameters (< 70 nm) and non-adhesive surface coatings to facilitate their spread through brain tumor extracellular matrix (ECM). These DNA-BPN provide widespread gene transfer in models recapitulating the ECM barrier, including three-dimensional multicellular tumor spheroids and mice with orthotopically established brain tumor.
Collapse
|
23
|
Delivery of Genetic Information: Viral Vector and Nonviral Vector Gene Therapies. Int Ophthalmol Clin 2021; 61:35-57. [PMID: 34196317 DOI: 10.1097/iio.0000000000000360] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
24
|
Ciulla T, Pennesi ME, Kiss S, Cunningham ET. DNA- and RNA-based Gene Therapies in Ophthalmology. Int Ophthalmol Clin 2021; 61:3-16. [PMID: 34196315 DOI: 10.1097/iio.0000000000000359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
25
|
Mercier J, Ruffin M, Corvol H, Guillot L. Gene Therapy: A Possible Alternative to CFTR Modulators? Front Pharmacol 2021; 12:648203. [PMID: 33967785 PMCID: PMC8097140 DOI: 10.3389/fphar.2021.648203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 02/24/2021] [Indexed: 12/12/2022] Open
Abstract
Cystic fibrosis (CF) is a rare genetic disease that affects several organs, but lung disease is the major cause of morbidity and mortality. The gene responsible for CF, the CFTR (Cystic Fibrosis Transmembrane Conductance Regulator) gene, has been discovered in 1989. Since then, gene therapy i.e., defective gene replacement by a functional one, remained the ultimate goal but unfortunately, it has not yet been achieved. However, patients care and symptomatic treatments considerably increased CF patients’ life expectancy ranging from 5 years old in the 1960s to 40 today. In the last decade, research works on CFTR protein structure and activity led to the development of new drugs which, by readdressing CFTR to the plasma membrane (correctors) or by enhancing its transport activity (potentiators), allow, alone or in combination, an improvement of CF patients’ lung function and quality of life. While expected, it is not yet known whether taking these drugs from an early age and for years will improve the quality of life of CF patients in the long term and further increase their life expectancy. Besides, these molecules are not available (specific variants of CFTR) or accessible (national health policies) for all patients and there is still no curative treatment. Another alternative that could benefit from new technologies, such as gene therapy, is therefore still attractive, although it is not yet offered to patients. Faced with the development of new CFTR correctors and potentiators, the question arises as to whether there is still a place for gene therapy and this is discussed in this perspective.
Collapse
Affiliation(s)
- J Mercier
- Sorbonne Université, Inserm, Centre de Recherche, Saint Antoine, F-75012, Paris, France
| | - M Ruffin
- Sorbonne Université, Inserm, Centre de Recherche, Saint Antoine, F-75012, Paris, France
| | - H Corvol
- Sorbonne Université, Inserm, Centre de Recherche, Saint Antoine, F-75012, Paris, France.,Pneumologie Pédiatrique, APHP, Hôpital Trousseau, Paris, France
| | - L Guillot
- Sorbonne Université, Inserm, Centre de Recherche, Saint Antoine, F-75012, Paris, France
| |
Collapse
|
26
|
Abstract
Delivery of genetic material to tissues in vivo is an important technique used in research settings and is the foundation upon which clinical gene therapy is built. The lung is a prime target for gene delivery due to a host of genetic, acquired, and infectious diseases that manifest themselves there, resulting in many pathologies. However, the in vivo delivery of genetic material to the lung remains a practical problem clinically and is considered the major obstacle needed to be overcome for gene therapy. Currently there are four main strategies for in vivo gene delivery to the lung: viral vectors, liposomes, nanoparticles, and electroporation. Viral delivery uses several different genetically modified viruses that enter the cell and express desired genes that have been inserted to the viral genome. Liposomes use combinations of charged and neutral lipids that can encapsulate genetic cargo and enter cells through endogenous mechanisms, thereby delivering their cargoes. Nanoparticles are defined by their size (typically less than 100 nm) and are made up of many different classes of building blocks, including biological and synthetic polymers, cell penetrant and other peptides, and dendrimers, that also enter cells through endogenous mechanisms. Electroporation uses mild to moderate electrical pulses to create pores in the cell membrane through which delivered genetic material can enter a cell. An emerging fifth category, exosomes and extracellular vesicles, may have advantages of both viral and non-viral approaches. These extracellular vesicles bud from cellular membranes containing receptors and ligands that may aid cell targeting and which can be loaded with genetic material for efficient transfer. Each of these vectors can be used for different gene delivery applications based on mechanisms of action, side-effects, and other factors, and their use in the lung and possible clinical considerations is the primary focus of this review.
Collapse
Affiliation(s)
- Uday K Baliga
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642, USA
- Department of Pathology, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642, USA
| | - David A Dean
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642, USA
| |
Collapse
|
27
|
Majumder J, Minko T. Targeted Nanotherapeutics for Respiratory Diseases: Cancer, Fibrosis, and Coronavirus. ADVANCED THERAPEUTICS 2021; 4:2000203. [PMID: 33173809 PMCID: PMC7646027 DOI: 10.1002/adtp.202000203] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 09/27/2020] [Indexed: 12/13/2022]
Abstract
Systemic delivery of therapeutics for treatment of lung diseases has several limitations including poor organ distribution of delivered payload with relatively low accumulation of active substances in the lungs and severe adverse side effects. In contrast, nanocarrier based therapeutics provide a broad range of opportunities due to their ability to encapsulate substances with different aqueous solubility, transport distinct types of cargo, target therapeutics specifically to the deceased organ, cell, or cellular organelle limiting adverse side effects and increasing the efficacy of therapy. Moreover, many nanotherapeutics can be delivered by inhalation locally to the lungs avoiding systemic circulation. In addition, nanoscale based delivery systems can be multifunctional, simultaneously carrying out several tasks including diagnostics, treatment and suppression of cellular resistance to the treatment. Nanoscale delivery systems improve the clinical efficacy of conventional therapeutics allowing new approaches for the treatment of respiratory diseases which are difficult to treat or possess intrinsic or acquired resistance to treatment. The present review summarizes recent advances in the development of nanocarrier based therapeutics for local and targeted delivery of drugs, nucleic acids and imaging agents for diagnostics and treatment of various diseases such as cancer, cystic fibrosis, and coronavirus.
Collapse
Affiliation(s)
- Joydeb Majumder
- Department of PharmaceuticsErnest Mario School of Pharmacy, RutgersThe State University of New JerseyPiscatawayNJ08854USA
| | - Tamara Minko
- Department of PharmaceuticsErnest Mario School of Pharmacy, RutgersThe State University of New JerseyPiscatawayNJ08854USA
| |
Collapse
|
28
|
Freitag F, Wagner E. Optimizing synthetic nucleic acid and protein nanocarriers: The chemical evolution approach. Adv Drug Deliv Rev 2021; 168:30-54. [PMID: 32246984 DOI: 10.1016/j.addr.2020.03.005] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/10/2020] [Accepted: 03/30/2020] [Indexed: 12/20/2022]
Abstract
Optimizing synthetic nanocarriers is like searching for a needle in a haystack. How to find the most suitable carrier for intracellular delivery of a specified macromolecular nanoagent for a given disease target location? Here, we review different synthetic 'chemical evolution' strategies that have been pursued. Libraries of nanocarriers have been generated either by unbiased combinatorial chemistry or by variation and novel combination of known functional delivery elements. As in natural evolution, definition of nanocarriers as sequences, as barcode or design principle, may fuel chemical evolution. Screening in appropriate test system may not only provide delivery candidates, but also a refined understanding of cellular delivery including novel, unpredictable mechanisms. Combined with rational design and computational algorithms, candidates can be further optimized in subsequent evolution cycles into nanocarriers with improved safety and efficacy. Optimization of nanocarriers differs for various cargos, as illustrated for plasmid DNA, siRNA, mRNA, proteins, or genome-editing nucleases.
Collapse
|
29
|
Zhong W, Zhang X, Zeng Y, Lin D, Wu J. Recent applications and strategies in nanotechnology for lung diseases. NANO RESEARCH 2021; 14:2067-2089. [PMID: 33456721 PMCID: PMC7796694 DOI: 10.1007/s12274-020-3180-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/11/2020] [Accepted: 10/11/2020] [Indexed: 05/14/2023]
Abstract
Lung diseases, including COVID-19 and lung cancers, is a huge threat to human health. However, for the treatment and diagnosis of various lung diseases, such as pneumonia, asthma, cancer, and pulmonary tuberculosis, are becoming increasingly challenging. Currently, several types of treatments and/or diagnostic methods are used to treat lung diseases; however, the occurrence of adverse reactions to chemotherapy, drug-resistant bacteria, side effects that can be significantly toxic, and poor drug delivery necessitates the development of more promising treatments. Nanotechnology, as an emerging technology, has been extensively studied in medicine. Several studies have shown that nano-delivery systems can significantly enhance the targeting of drug delivery. When compared to traditional delivery methods, several nanoparticle delivery strategies are used to improve the detection methods and drug treatment efficacy. Transporting nanoparticles to the lungs, loading appropriate therapeutic drugs, and the incorporation of intelligent functions to overcome various lung barriers have broad prospects as they can aid in locating target tissues and can enhance the therapeutic effect while minimizing systemic side effects. In addition, as a new and highly contagious respiratory infection disease, COVID-19 is spreading worldwide. However, there is no specific drug for COVID-19. Clinical trials are being conducted in several countries to develop antiviral drugs or vaccines. In recent years, nanotechnology has provided a feasible platform for improving the diagnosis and treatment of diseases, nanotechnology-based strategies may have broad prospects in the diagnosis and treatment of COVID-19. This article reviews the latest developments in nanotechnology drug delivery strategies in the lungs in recent years and studies the clinical application value of nanomedicine in the drug delivery strategy pertaining to the lung.
Collapse
Affiliation(s)
- Wenhao Zhong
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107 China
| | - Xinyu Zhang
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107 China
| | - Yunxin Zeng
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107 China
| | - Dongjun Lin
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107 China
| | - Jun Wu
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107 China
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province, School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, 510006 China
| |
Collapse
|
30
|
Kansara VS, Cooper M, Sesenoglu-Laird O, Muya L, Moen R, Ciulla TA. Suprachoroidally Delivered DNA Nanoparticles Transfect Retina and Retinal Pigment Epithelium/Choroid in Rabbits. Transl Vis Sci Technol 2020; 9:21. [PMID: 33364076 PMCID: PMC7745627 DOI: 10.1167/tvst.9.13.21] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 10/16/2020] [Indexed: 01/04/2023] Open
Abstract
Purpose This study evaluated ocular tolerability and transfectability of nonviral DNA nanoparticles (DNPs) after microneedle-based suprachoroidal (SC) administration, in comparison to subretinal (SR) administration. Methods The DNPs consisted of a single copy of plasmid DNA with a polyubiquitin C/luciferase transcriptional cassette compacted with 10 kDa PEG-substituted lysine 30-mer peptides (CK30PEG10k). New Zealand White rabbits (n = 4 per group) received a unilateral SC injection (0.1 mL via a microneedle technique) of ellipsoid-shaped DNPs, rod-shaped DNPs, or saline (negative control). A cohort of rabbits (n = 4) also received a single unilateral SR injection (0.05 mL via a transvitreal approach) of rod-shaped DNPs. At day 7, luciferase activity was measured in the retina and retinal pigment epithelium (RPE)–choroid via bioluminescence assay. A cohort of rabbits received a SC injection of analogous DNPs to assess spread of DNP injectate in the suprachoroidal space (SCS) via optical coherent tomography and histology. Results Suprachoroidal injection of DNPs resulted in reversible opening of the SCS circumferentially and posteriorly and was generally well tolerated, with no significant ocular examination score changes, intraocular pressure abnormalities, or changes in electroretinography amplitudes on day 7 compared to the baseline. High luciferase activity was observed in the retina and RPE-choroid of eyes that received SC DNPs (rod and ellipsoid shape) and SR DNPs (rod shape) compared to controls. The mean luciferase activity in RPE-choroid and retina was comparable between SC and SR administrations. Transfection in the RPE-choroid was approximately 10-fold higher than in the retina after either SC or SR administration of DNPs. Conclusions Suprachoroidal and SR administration of DNPs resulted in comparable transfection of retina and RPE-choroid. Translational Relevance Suprachoroidal delivery of DNPs offers the potential to precisely target chorioretinal tissues while avoiding surgical risks associated with SR injection, and it may offer an office-based nonsurgical gene therapy option for the treatment of retinal diseases.
Collapse
Affiliation(s)
| | - Mark Cooper
- Copernicus Therapeutics, Inc., Cleveland, OH, USA
| | | | - Leroy Muya
- Clearside Biomedical, Inc., Alpharetta, GA, USA
| | - Robert Moen
- Copernicus Therapeutics, Inc., Cleveland, OH, USA
| | | |
Collapse
|
31
|
Chen D, Liu J, Wu J, Suk JS. Enhancing nanoparticle penetration through airway mucus to improve drug delivery efficacy in the lung. Expert Opin Drug Deliv 2020; 18:595-606. [PMID: 33218265 DOI: 10.1080/17425247.2021.1854222] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Introduction: Airway mucus gel layer serves as a key delivery barrier that limits the performance of inhaled drug delivery nanoparticles. Conventional nanoparticles are readily trapped by the airway mucus and rapidly cleared from the lung via mucus clearance mechanisms. These nanoparticles cannot distribute throughout the lung airways, long-reside in the lung and/or reach the airway epithelium. To address this challenge, strategies to enhance particle penetration through the airway mucus have been developed and proof-of-concept has been established using mucus model systems..Areas covered: In this review, we first overview the biochemical and biophysical characteristics that render the airway mucus a challenging delivery barrier. We then introduce strategies to improve particle penetration through the airway mucus. Specifically, we walk through two classes of approaches, including modification of physicochemical properties of nanoparticles and modulation of barrier properties of airway mucus.Expert opinion: State-of-the-art strategies to overcome the airway mucus barrier have been introduced and experimentally validated. However, data should be interpreted in the comprehensive context of therapeutic delivery from the site of administration to the final destination to determine clinically-relevant approaches. Further, safety should be carefully monitored, particularly when it comes to mucus-altering strategies that may perturb physiological functions of airway mucus.
Collapse
Affiliation(s)
- Daiqin Chen
- The Center for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Ophthalmology, Johns Hopkins University, Baltimore, MD, USA
| | - Jinhao Liu
- The Center for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jerry Wu
- The Center for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jung Soo Suk
- The Center for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Ophthalmology, Johns Hopkins University, Baltimore, MD, USA.,Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
32
|
Peptides as a material platform for gene delivery: Emerging concepts and converging technologies. Acta Biomater 2020; 117:40-59. [PMID: 32966922 DOI: 10.1016/j.actbio.2020.09.027] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 08/27/2020] [Accepted: 09/16/2020] [Indexed: 02/06/2023]
Abstract
Successful gene therapies rely on methods that safely introduce DNA into target cells and enable subsequent expression of proteins. To that end, peptides are an attractive materials platform for DNA delivery, facilitating condensation into nanoparticles, delivery into cells, and subcellular release to enable protein expression. Peptides are programmable materials that can be designed to address biocompatibility, stability, and subcellular barriers that limit efficiency of non-viral gene delivery systems. This review focuses on fundamental structure-function relationships regarding peptide design and their impact on nanoparticle physical properties, biologic activity, and biocompatibility. Recent peptide technologies utilize multi-dimensional structures, non-natural chemistries, and combinations of peptides with lipids to achieve desired properties and efficient transfection. Advances in DNA cargo design are also presented to highlight further opportunities for peptide-based gene delivery. Modern DNA designs enable prolonged expression compared to traditional plasmids, providing an additional component that can be synergized with peptide carriers for improved transfection. Peptide transfection systems are poised to become a flexible and efficient platform incorporating new chemistries, functionalities, and improved DNA cargos to usher in a new era of gene therapy.
Collapse
|
33
|
Mohammadinejad R, Dehshahri A, Sagar Madamsetty V, Zahmatkeshan M, Tavakol S, Makvandi P, Khorsandi D, Pardakhty A, Ashrafizadeh M, Ghasemipour Afshar E, Zarrabi A. In vivo gene delivery mediated by non-viral vectors for cancer therapy. J Control Release 2020; 325:249-275. [PMID: 32634464 PMCID: PMC7334939 DOI: 10.1016/j.jconrel.2020.06.038] [Citation(s) in RCA: 154] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/27/2020] [Accepted: 06/29/2020] [Indexed: 12/17/2022]
Abstract
Gene therapy by expression constructs or down-regulation of certain genes has shown great potential for the treatment of various diseases. The wide clinical application of nucleic acid materials dependents on the development of biocompatible gene carriers. There are enormous various compounds widely investigated to be used as non-viral gene carriers including lipids, polymers, carbon materials, and inorganic structures. In this review, we will discuss the recent discoveries on non-viral gene delivery systems. We will also highlight the in vivo gene delivery mediated by non-viral vectors to treat cancer in different tissue and organs including brain, breast, lung, liver, stomach, and prostate. Finally, we will delineate the state-of-the-art and promising perspective of in vivo gene editing using non-viral nano-vectors.
Collapse
Affiliation(s)
- Reza Mohammadinejad
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Ali Dehshahri
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Vijay Sagar Madamsetty
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL 32224, USA
| | - Masoumeh Zahmatkeshan
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Pooyan Makvandi
- Institute for Polymers, Composites and Biomaterials, National Research Council, IPCB-CNR, Naples, Italy; Chemistry Department, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz 6153753843, Iran
| | - Danial Khorsandi
- Department of Medical Nanotechnology, Faculty of Advanced, Technologies in Medicine, Iran University of Medical Sciences, Tehran 14496-14535, Iran; Department of Biotechnology-Biomedicine, University of Barcelona, Barcelona 08028, Spain
| | - Abbas Pardakhty
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Elham Ghasemipour Afshar
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Istanbul 34956, Turkey; Center of Excellence for Functional Surfaces and Interfaces (EFSUN), Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla, Istanbul 34956, Turkey.
| |
Collapse
|
34
|
Koji K, Yoshinaga N, Mochida Y, Hong T, Miyazaki T, Kataoka K, Osada K, Cabral H, Uchida S. Bundling of mRNA strands inside polyion complexes improves mRNA delivery efficiency in vitro and in vivo. Biomaterials 2020; 261:120332. [PMID: 32877764 DOI: 10.1016/j.biomaterials.2020.120332] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 08/14/2020] [Indexed: 01/08/2023]
Abstract
RNA nanotechnology has promise for developing mRNA carriers with enhanced physicochemical and functional properties. However, the potential synergy for mRNA delivery of RNA nanotechnology in cooperation with established carrier systems remains unknown. This study proposes a combinational system of RNA nanotechnology and mRNA polyplexes, by focusing on mRNA steric structure inside the polyplexes. Firstly, several mRNA strands are bundled through hybridization with RNA oligonucleotide crosslinkers to obtain tight mRNA structure, and then the bundled mRNA is mixed with poly(ethylene glycol) (PEG)-polycation block copolymers to prepare PEG-coated polyplex micelles (PMs). mRNA bundling results in highly condensed mRNA packaging inside PM core with dense PEG chains on the surface, thereby, improving PM stability against polyion exchange reaction and ribonuclease (RNase) attack. Importantly, such stabilization effects are attributed to bundled structure of mRNA rather than the increase in total mRNA amount encapsulated in the PMs, as encapsulation of long mRNA strands without bundling fails to improve PM stability. Consequently, PMs loading bundled mRNA exhibit enhanced stability in mouse blood circulation, and induce efficient protein expression in cultured cells and mouse brain.
Collapse
Affiliation(s)
- Kyoko Koji
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Naoto Yoshinaga
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Yuki Mochida
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan
| | - Taehun Hong
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Takuya Miyazaki
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan; Kanagawa Institute of Industrial Science and Technology, 705-1 Shimoimaizumi, Ebina, Kanagawa, 243-0435, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan; Policy Alternatives Research Institute, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Kensuke Osada
- Department of Molecular Imaging and Theranostics, Quantum Medical Science Directorate, National Institutes for Quantum and Radiological Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba, 263-8555, Japan
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan; Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan.
| | - Satoshi Uchida
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan; Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan.
| |
Collapse
|
35
|
Bañuls L, Pellicer D, Castillo S, Navarro-García MM, Magallón M, González C, Dasí F. Gene Therapy in Rare Respiratory Diseases: What Have We Learned So Far? J Clin Med 2020; 9:E2577. [PMID: 32784514 PMCID: PMC7463867 DOI: 10.3390/jcm9082577] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 07/26/2020] [Accepted: 08/05/2020] [Indexed: 02/06/2023] Open
Abstract
Gene therapy is an alternative therapy in many respiratory diseases with genetic origin and currently without curative treatment. After five decades of progress, many different vectors and gene editing tools for genetic engineering are now available. However, we are still a long way from achieving a safe and efficient approach to gene therapy application in clinical practice. Here, we review three of the most common rare respiratory conditions-cystic fibrosis (CF), alpha-1 antitrypsin deficiency (AATD), and primary ciliary dyskinesia (PCD)-alongside attempts to develop genetic treatment for these diseases. Since the 1990s, gene augmentation therapy has been applied in multiple clinical trials targeting CF and AATD, especially using adeno-associated viral vectors, resulting in a good safety profile but with low efficacy in protein expression. Other strategies, such as non-viral vectors and more recently gene editing tools, have also been used to address these diseases in pre-clinical studies. The first gene therapy approach in PCD was in 2009 when a lentiviral transduction was performed to restore gene expression in vitro; since then, transcription activator-like effector nucleases (TALEN) technology has also been applied in primary cell culture. Gene therapy is an encouraging alternative treatment for these respiratory diseases; however, more research is needed to ensure treatment safety and efficacy.
Collapse
Affiliation(s)
- Lucía Bañuls
- Research group on Rare Respiratory Diseases (ERR), Department of Physiology, School of Medicine, University of Valencia, Avda. Blasco Ibáñez, 15, 46010 Valencia, Spain; (L.B.); (D.P.); (M.M.)
- Research group on Rare Respiratory Diseases (ERR), Instituto de Investigación Sanitaria INCLIVA, Fundación Investigación Hospital Clínico Valencia, Avda. Menéndez y Pelayo, 4, 46010 Valencia, Spain; (S.C.); (M.M.N.-G.); (C.G.)
| | - Daniel Pellicer
- Research group on Rare Respiratory Diseases (ERR), Department of Physiology, School of Medicine, University of Valencia, Avda. Blasco Ibáñez, 15, 46010 Valencia, Spain; (L.B.); (D.P.); (M.M.)
- Research group on Rare Respiratory Diseases (ERR), Instituto de Investigación Sanitaria INCLIVA, Fundación Investigación Hospital Clínico Valencia, Avda. Menéndez y Pelayo, 4, 46010 Valencia, Spain; (S.C.); (M.M.N.-G.); (C.G.)
| | - Silvia Castillo
- Research group on Rare Respiratory Diseases (ERR), Instituto de Investigación Sanitaria INCLIVA, Fundación Investigación Hospital Clínico Valencia, Avda. Menéndez y Pelayo, 4, 46010 Valencia, Spain; (S.C.); (M.M.N.-G.); (C.G.)
- Paediatrics Unit, Hospital Clínico Universitario de Valencia, Avda. Blasco Ibáñez, 17, 46010 Valencia, Spain
| | - María Mercedes Navarro-García
- Research group on Rare Respiratory Diseases (ERR), Instituto de Investigación Sanitaria INCLIVA, Fundación Investigación Hospital Clínico Valencia, Avda. Menéndez y Pelayo, 4, 46010 Valencia, Spain; (S.C.); (M.M.N.-G.); (C.G.)
| | - María Magallón
- Research group on Rare Respiratory Diseases (ERR), Department of Physiology, School of Medicine, University of Valencia, Avda. Blasco Ibáñez, 15, 46010 Valencia, Spain; (L.B.); (D.P.); (M.M.)
- Research group on Rare Respiratory Diseases (ERR), Instituto de Investigación Sanitaria INCLIVA, Fundación Investigación Hospital Clínico Valencia, Avda. Menéndez y Pelayo, 4, 46010 Valencia, Spain; (S.C.); (M.M.N.-G.); (C.G.)
| | - Cruz González
- Research group on Rare Respiratory Diseases (ERR), Instituto de Investigación Sanitaria INCLIVA, Fundación Investigación Hospital Clínico Valencia, Avda. Menéndez y Pelayo, 4, 46010 Valencia, Spain; (S.C.); (M.M.N.-G.); (C.G.)
- Pneumology Unit, Hospital Clínico Universitario de Valencia, Avda. Blasco Ibáñez, 17, 46010 Valencia, Spain
| | - Francisco Dasí
- Research group on Rare Respiratory Diseases (ERR), Department of Physiology, School of Medicine, University of Valencia, Avda. Blasco Ibáñez, 15, 46010 Valencia, Spain; (L.B.); (D.P.); (M.M.)
- Research group on Rare Respiratory Diseases (ERR), Instituto de Investigación Sanitaria INCLIVA, Fundación Investigación Hospital Clínico Valencia, Avda. Menéndez y Pelayo, 4, 46010 Valencia, Spain; (S.C.); (M.M.N.-G.); (C.G.)
| |
Collapse
|
36
|
Osada K. Structural Polymorphism of Single pDNA Condensates Elicited by Cationic Block Polyelectrolytes. Polymers (Basel) 2020; 12:polym12071603. [PMID: 32707655 PMCID: PMC7408586 DOI: 10.3390/polym12071603] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/15/2020] [Accepted: 07/16/2020] [Indexed: 12/17/2022] Open
Abstract
DNA folding is a core phenomenon in genome packaging within a nucleus. Such a phenomenon is induced by polyelectrolyte complexation between anionic DNA and cationic proteins of histones. In this regard, complexes formed between DNA and cationic polyelectrolytes have been investigated as models to gain insight into genome packaging. Upon complexation, DNA undergoes folding to reduce its occupied volume, which often results in multi-complex associated aggregates. However, when cationic copolymers comprising a polycation block and a neutral hydrophilic polymer block are used instead, DNA undergoes folding as a single molecule within a spontaneously formed polyplex micelle (PM), thereby allowing the observation of the higher-order structures that DNA forms. The DNA complex forms polymorphic structures, including globular, rod-shaped, and ring-shaped (toroidal) structures. This review focuses on the polymorphism of DNA, particularly, to elucidate when, how, and why DNA organizes into these structures with cationic copolymers. The interactions between DNA and the copolymers, and the specific nature of DNA in rigidity; i.e., rigid but foldable, play significant roles in the observed polymorphism. Moreover, PMs serve as potential gene vectors for systemic application. The significance of the controlled DNA folding for such an application is addressed briefly in the last part.
Collapse
Affiliation(s)
- Kensuke Osada
- Quantum Medical Science Directorate, National Institutes for Quantum and Radiological Science and Technology (QST), Anagawa, Inage-ku, Chiba-shi, Chiba 263-8555, Japan
| |
Collapse
|
37
|
French LS, Mellough CB, Chen FK, Carvalho LS. A Review of Gene, Drug and Cell-Based Therapies for Usher Syndrome. Front Cell Neurosci 2020; 14:183. [PMID: 32733204 PMCID: PMC7363968 DOI: 10.3389/fncel.2020.00183] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 05/28/2020] [Indexed: 12/12/2022] Open
Abstract
Usher syndrome is a genetic disorder causing neurosensory hearing loss and blindness from retinitis pigmentosa (RP). Adaptive techniques such as braille, digital and optical magnifiers, mobility training, cochlear implants, or other assistive listening devices are indispensable for reducing disability. However, there is currently no treatment to reduce or arrest sensory cell degeneration. There are several classes of treatments for Usher syndrome being investigated. The present article reviews the progress this research has made towards delivering commercial options for patients with Usher syndrome.
Collapse
Affiliation(s)
- Lucy S French
- Centre for Ophthalmology and Visual Sciences (incorporating Lions Eye Institute), The University of Western Australia, Nedlands, WA, Australia
| | - Carla B Mellough
- Centre for Ophthalmology and Visual Sciences (incorporating Lions Eye Institute), The University of Western Australia, Nedlands, WA, Australia
| | - Fred K Chen
- Centre for Ophthalmology and Visual Sciences (incorporating Lions Eye Institute), The University of Western Australia, Nedlands, WA, Australia.,Department of Ophthalmology, Royal Perth Hospital, Perth, WA, Australia.,Department of Ophthalmology, Perth Children's Hospital, Nedlands, WA, Australia
| | - Livia S Carvalho
- Centre for Ophthalmology and Visual Sciences (incorporating Lions Eye Institute), The University of Western Australia, Nedlands, WA, Australia
| |
Collapse
|
38
|
da Silva AL, Silva LA, Cruz FF, Rocco PRM, Morales MM. Application of novel nanotechnologies in asthma. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:159. [PMID: 32309307 PMCID: PMC7154405 DOI: 10.21037/atm.2019.12.40] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Adriana Lopes da Silva
- Laboratory of Pulmonary Investigation, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Laboratory of Cellular and Molecular Biology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Rede NanoSaúde, Fundação Carlos Chagas Filho de Amparo a Pesquisa do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luisa Andrade Silva
- Laboratory of Pulmonary Investigation, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Rede NanoSaúde, Fundação Carlos Chagas Filho de Amparo a Pesquisa do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernanda Ferreira Cruz
- Laboratory of Pulmonary Investigation, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Rede NanoSaúde, Fundação Carlos Chagas Filho de Amparo a Pesquisa do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patricia Rieken Macedo Rocco
- Laboratory of Pulmonary Investigation, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Rede NanoSaúde, Fundação Carlos Chagas Filho de Amparo a Pesquisa do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo Marcos Morales
- Laboratory of Cellular and Molecular Biology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Rede NanoSaúde, Fundação Carlos Chagas Filho de Amparo a Pesquisa do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
39
|
Melnyk T, Đorđević S, Conejos-Sánchez I, Vicent MJ. Therapeutic potential of polypeptide-based conjugates: Rational design and analytical tools that can boost clinical translation. Adv Drug Deliv Rev 2020; 160:136-169. [PMID: 33091502 DOI: 10.1016/j.addr.2020.10.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 10/09/2020] [Accepted: 10/14/2020] [Indexed: 12/14/2022]
Abstract
The clinical success of polypeptides as polymeric drugs, covered by the umbrella term "polymer therapeutics," combined with related scientific and technological breakthroughs, explain their exponential growth in the development of polypeptide-drug conjugates as therapeutic agents. A deeper understanding of the biology at relevant pathological sites and the critical biological barriers faced, combined with advances regarding controlled polymerization techniques, material bioresponsiveness, analytical methods, and scale up-manufacture processes, have fostered the development of these nature-mimicking entities. Now, engineered polypeptides have the potential to combat current challenges in the advanced drug delivery field. In this review, we will discuss examples of polypeptide-drug conjugates as single or combination therapies in both preclinical and clinical studies as therapeutics and molecular imaging tools. Importantly, we will critically discuss relevant examples to highlight those parameters relevant to their rational design, such as linking chemistry, the analytical strategies employed, and their physicochemical and biological characterization, that will foster their rapid clinical translation.
Collapse
Affiliation(s)
- Tetiana Melnyk
- Centro de Investigación Príncipe Felipe, Polymer Therapeutics Lab, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - Snežana Đorđević
- Centro de Investigación Príncipe Felipe, Polymer Therapeutics Lab, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - Inmaculada Conejos-Sánchez
- Centro de Investigación Príncipe Felipe, Polymer Therapeutics Lab, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - María J Vicent
- Centro de Investigación Príncipe Felipe, Polymer Therapeutics Lab, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| |
Collapse
|
40
|
Zheng M, Mitra RN, Weiss ER, Han Z. Rhodopsin Genomic Loci DNA Nanoparticles Improve Expression and Rescue of Retinal Degeneration in a Model for Retinitis Pigmentosa. Mol Ther 2019; 28:523-535. [PMID: 31879189 DOI: 10.1016/j.ymthe.2019.11.031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 11/12/2019] [Accepted: 11/21/2019] [Indexed: 02/07/2023] Open
Abstract
The use of gene therapy may allow replacement of the defective gene. Minigenes, such as cDNAs, are often used. However, these may not express normal physiological genetic profiles due to lack of crucial endogenous regulatory elements. We constructed DNA nanoparticles (NPs) that contain either the mouse or human full-length rhodopsin genomic locus, including endogenous promoters, all introns, and flanking regulatory sequences of the 15-16 kb genomic rhodopsin DNA inserts. We transduced the NPs into primary retinal cell cultures from the rhodopsin knockout (RKO) mouse in vitro and into the RKO mouse in vivo and compared the effects on different functions to plasmid cDNA NP counterparts that were driven by ubiquitous promoters. Our results demonstrate that genomic DNA vectors resulted in long-term high levels of physiological transgene expression over a period of 5 months. In contrast, the cDNA counterparts exhibited low levels of expression with sensitivity to the endoplasmic reticulum (ER) stress mechanism using the same transgene copy number both in vitro and in vivo. This study demonstrates for the first time the transducing of the rhodopsin genomic locus using compacted DNA NPs.
Collapse
Affiliation(s)
- Min Zheng
- Department of Ophthalmology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Rajendra N Mitra
- Department of Ophthalmology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ellen R Weiss
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Zongchao Han
- Department of Ophthalmology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Division of Pharmacoengineering & Molecular Pharmaceutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
41
|
Shah E, Kadam A, Jubin T, Begum R, Upadhyay P, Soni HP. EDTA‐Capped Iron Oxide Core‐Corona System as Vehicle for Gene Delivery to Transform
E.coli
: Mimicking the Lipid Bilayer Environment. ChemistrySelect 2019. [DOI: 10.1002/slct.201900964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Ekta Shah
- Department of ChemistryFaculty of Science, The Maharaja Sayajirao University of Baroda Vadodara- 390 002, Gujarat India
| | - Ashlesha Kadam
- Department of BiochemistryFaculty of ScienceThe Maharaja Sayajirao University of Baroda Vadodara- 390 002, Gujarat India
| | - Tina Jubin
- Department of BiochemistryFaculty of ScienceThe Maharaja Sayajirao University of Baroda Vadodara- 390 002, Gujarat India
| | - Rasheedunnisa Begum
- Department of BiochemistryFaculty of ScienceThe Maharaja Sayajirao University of Baroda Vadodara- 390 002, Gujarat India
| | - Pratik Upadhyay
- Department of Pharmaceutical TechnologyL. J. Institute of Pharmacy, Ahmedabad, Gujarat India
| | - Hemant P. Soni
- Department of ChemistryFaculty of Science, The Maharaja Sayajirao University of Baroda Vadodara- 390 002, Gujarat India
| |
Collapse
|
42
|
Doroudian M, MacLoughlin R, Poynton F, Prina-Mello A, Donnelly SC. Nanotechnology based therapeutics for lung disease. Thorax 2019; 74:965-976. [PMID: 31285360 DOI: 10.1136/thoraxjnl-2019-213037] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 05/06/2019] [Accepted: 05/13/2019] [Indexed: 11/03/2022]
Abstract
Nanomedicine is a multidisciplinary research field with an integration of traditional sciences such as chemistry, physics, biology and materials science. The application of nanomedicine for lung diseases as a relatively new area of interdisciplinary science has grown rapidly over the last 10 years. Promising research outcomes suggest that nanomedicine will revolutionise the practice of medicine, through the development of new approaches in therapeutic agent delivery, vaccine development and nanotechnology-based medical detections. Nano-based approaches in the diagnosis and treatment of lung diseases will, in the not too distant future, change the way we practise medicine. This review will focus on the current trends and developments in the clinical translation of nanomedicine for lung diseases, such as in the areas of lung cancer, cystic fibrosis, asthma, bacterial infections and COPD.
Collapse
Affiliation(s)
- Mohammad Doroudian
- Department of Medicine, Tallaght University Hospital, Dublin 24 & Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Ronan MacLoughlin
- Aerogen, IDA Business Park, Galway, Ireland.,School of Pharmacy, Royal College of Surgeons, Dublin, Ireland.,School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin, Ireland
| | - Fergus Poynton
- Department of Medicine, Tallaght University Hospital, Dublin 24 & Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Adriele Prina-Mello
- CRANN Institute and AMBER Centre, University of Dublin Trinity College, Dublin, Ireland.,Department of Medicine, Laboratory for Biological Characterization of Advanced Materials (LBCAM), Trinity College Dublin, Dublin, Ireland.,Nanomedicine Group, Trinity Translational Medicine Institute (TTMI), Trinity College Dublin, Dublin, Ireland
| | - Seamas C Donnelly
- Department of Medicine, Tallaght University Hospital, Dublin 24 & Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
43
|
Guan S, Munder A, Hedtfeld S, Braubach P, Glage S, Zhang L, Lienenklaus S, Schultze A, Hasenpusch G, Garrels W, Stanke F, Miskey C, Johler SM, Kumar Y, Tümmler B, Rudolph C, Ivics Z, Rosenecker J. Self-assembled peptide-poloxamine nanoparticles enable in vitro and in vivo genome restoration for cystic fibrosis. NATURE NANOTECHNOLOGY 2019; 14:287-297. [PMID: 30692673 DOI: 10.1038/s41565-018-0358-x] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 12/18/2018] [Indexed: 06/09/2023]
Abstract
Developing safe and efficient non-viral delivery systems remains a major challenge for in vivo applications of gene therapy, especially in cystic fibrosis. Unlike conventional cationic polymers or lipids, the emerging poloxamine-based copolymers display promising in vivo gene delivery capabilities. However, poloxamines are invalid for in vitro applications and their in vivo transfection efficiency is still low compared with viral vectors. Here, we show that peptides developed by modular design approaches can spontaneously form compact and monodisperse nanoparticles with poloxamines and nucleic acids via self-assembly. Both messenger RNA and plasmid DNA expression mediated by peptide-poloxamine nanoparticles are greatly boosted in vitro and in the lungs of cystic fibrosis mice with negligible toxicity. Peptide-poloxamine nanoparticles containing integrating vectors enable successful in vitro and in vivo long-term restoration of cystic fibrosis transmembrane conductance regulator deficiency with a safe integration profile. Our dataset provides a new framework for designing non-viral gene delivery systems qualified for in vivo genetic modifications.
Collapse
Affiliation(s)
- Shan Guan
- Department of Pediatrics, Ludwig Maximilian University of Munich, Munich, Germany
| | - Antje Munder
- Clinic for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover, German Center for Lung Research, Hannover, Germany
| | - Silke Hedtfeld
- Clinic for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Peter Braubach
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover, German Center for Lung Research, Hannover, Germany
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Silke Glage
- Institute of Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Longgui Zhang
- Department of Pediatrics, Ludwig Maximilian University of Munich, Munich, Germany
| | - Stefan Lienenklaus
- Institute of Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Anja Schultze
- Department of Pediatrics, Ludwig Maximilian University of Munich, Munich, Germany
| | | | - Wiebke Garrels
- Institute of Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Frauke Stanke
- Clinic for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover, German Center for Lung Research, Hannover, Germany
| | - Csaba Miskey
- Division of Medical Biotechnology, Paul Ehrlich Institute, Langen, Germany
| | - Sarah M Johler
- Department of Pediatrics, Ludwig Maximilian University of Munich, Munich, Germany
| | | | - Burkhard Tümmler
- Clinic for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover, German Center for Lung Research, Hannover, Germany
| | - Carsten Rudolph
- Department of Pediatrics, Ludwig Maximilian University of Munich, Munich, Germany
- Ethris, Planegg, Germany
| | - Zoltan Ivics
- Division of Medical Biotechnology, Paul Ehrlich Institute, Langen, Germany
| | - Joseph Rosenecker
- Department of Pediatrics, Ludwig Maximilian University of Munich, Munich, Germany.
| |
Collapse
|
44
|
Abstract
Cystic fibrosis is a genetic disease affecting more than 70,000 people worldwide. Caused by a mutation in the CFTR gene, cystic fibrosis can result in difficulty breathing, widespread bacterial infections, edema, malnutrition, pancreatitis, and death. Current drug-based treatments struggle to reach the site of action due to the thick mucus, and only manage symptoms such as blocked airways, lung infections, and limited ability to digest food. Nanotechnology opens up possibilities for improved treatment strategies by focusing on drug penetration through the mucus lining, eliminating resulting bacterial infections, and targeting the underlying genetic cause of the disease. In this review, we present recent nanoparticle developments for cystic fibrosis, challenges in nanomedicine therapeutics, and future research directions in gene editing and nonviral vectors for gene delivery.
Collapse
Affiliation(s)
- Victor Ong
- 1 Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Vincent Mei
- 1 Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Lin Cao
- 1 Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Kiana Lee
- 1 Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Eun Ji Chung
- 1 Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
- 2 Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, USA
- 3 Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- 4 Division of Vascular Surgery and Endovascular Repair, Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- 5 Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
- 6 Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
45
|
Abstract
Introduction:
Gene therapy has emerged out as a promising therapeutic pave for the treatment
of genetic and acquired diseases. Gene transfection into target cells using naked DNA is a simple
and safe approach which has been further improved by combining vectors or gene carriers. Both viral
and non-viral approaches have achieved a milestone to establish this technique, but non-viral approaches
have attained a significant attention because of their favourable properties like less immunotoxicity
and biosafety, easy to produce with versatile surface modifications, etc. Literature is rich in evidences
which revealed that undoubtedly, non–viral vectors have acquired a unique place in gene therapy
but still there are number of challenges which are to be overcome to increase their effectiveness and
prove them ideal gene vectors.
Conclusion:
To date, tissue specific expression, long lasting gene expression system, enhanced gene
transfection efficiency has been achieved with improvement in delivery methods using non-viral vectors.
This review mainly summarizes the various physical and chemical methods for gene transfer in vitro
and in vivo.
Collapse
Affiliation(s)
- Aparna Bansal
- Department of Chemistry, Hansraj College, University of Delhi, Delhi-110007, India
| | - Himanshu
- Department of Chemistry, Hansraj College, University of Delhi, Delhi-110007, India
| |
Collapse
|
46
|
Englert C, Brendel JC, Majdanski TC, Yildirim T, Schubert S, Gottschaldt M, Windhab N, Schubert US. Pharmapolymers in the 21st century: Synthetic polymers in drug delivery applications. Prog Polym Sci 2018. [DOI: 10.1016/j.progpolymsci.2018.07.005] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
47
|
Cooney AL, McCray PB, Sinn PL. Cystic Fibrosis Gene Therapy: Looking Back, Looking Forward. Genes (Basel) 2018; 9:genes9110538. [PMID: 30405068 PMCID: PMC6266271 DOI: 10.3390/genes9110538] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 10/30/2018] [Accepted: 10/31/2018] [Indexed: 01/02/2023] Open
Abstract
Cystic fibrosis (CF) is an autosomal recessive disease caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene that encodes a cAMP-regulated anion channel. Although CF is a multi-organ system disease, most people with CF die of progressive lung disease that begins early in childhood and is characterized by chronic bacterial infection and inflammation. Nearly 90% of people with CF have at least one copy of the ΔF508 mutation, but there are hundreds of CFTR mutations that result in a range of disease severities. A CFTR gene replacement approach would be efficacious regardless of the disease-causing mutation. After the discovery of the CFTR gene in 1989, the in vitro proof-of-concept for gene therapy for CF was quickly established in 1990. In 1993, the first of many gene therapy clinical trials attempted to rescue the CF defect in airway epithelia. Despite the initial enthusiasm, there is still no FDA-approved gene therapy for CF. Here we discuss the history of CF gene therapy, from the discovery of the CFTR gene to current state-of-the-art gene delivery vector designs. While implementation of CF gene therapy has proven more challenging than initially envisioned; thanks to continued innovation, it may yet become a reality.
Collapse
Affiliation(s)
- Ashley L Cooney
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA.
| | - Paul B McCray
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA.
| | - Patrick L Sinn
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
48
|
Osman G, Rodriguez J, Chan SY, Chisholm J, Duncan G, Kim N, Tatler AL, Shakesheff KM, Hanes J, Suk JS, Dixon JE. PEGylated enhanced cell penetrating peptide nanoparticles for lung gene therapy. J Control Release 2018; 285:35-45. [PMID: 30004000 PMCID: PMC6573017 DOI: 10.1016/j.jconrel.2018.07.001] [Citation(s) in RCA: 149] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 06/27/2018] [Accepted: 07/02/2018] [Indexed: 11/24/2022]
Abstract
The lung remains an attractive target for the gene therapy of monogenetic diseases such as cystic fibrosis (CF). Despite over 27 clinical trials, there are still very few gene therapy vectors that have shown any improvement in lung function; highlighting the need to develop formulations with improved gene transfer potency and the desirable physiochemical characteristics for efficacious therapy. Herein, we introduce a novel cell penetrating peptide (CPP)-based non-viral vector that utilises glycosaminoglycan (GAG)-binding enhanced transduction (GET) for highly efficient gene transfer. GET peptides couple directly with DNA through electrostatic interactions to form nanoparticles (NPs). In order to adapt the GET peptide for efficient in vivo delivery, we engineered PEGylated versions of the peptide and employed a strategy to form DNA NPs with different densities of PEG coatings. We were able to identify candidate formulations (PEGylation rates ≥40%) that shielded the positively charged surface of particles, maintained colloidal stability in bronchoalveolar lavage fluid (BALF) and retained gene transfer activity in human bronchial epithelial cell lines and precision cut lung slices (PCLS) in vitro. Using multiple particle tracking (MPT) technology, we demonstrated that PEG-GET complexes were able to navigate the mucus mesh and diffuse rapidly through patient CF sputum samples ex vivo. When tested in mouse lung models in vivo, PEGylated particles demonstrated superior biodistribution, improved safety profiles and efficient gene transfer of a reporter luciferase plasmid compared to non-PEGylated complexes. Furthermore, gene expression was significantly enhanced in comparison to polyethylenimine (PEI), a non-viral gene carrier that has been widely tested in pre-clinical settings. This work describes an innovative approach that combines novel GET peptides for enhanced transfection with a tuneable PEG coating for efficacious lung gene therapy.
Collapse
Affiliation(s)
- Gizem Osman
- Wolfson Centre for Stem Cells, Tissue Engineering, and Modelling (STEM), Centre of Biomolecular Sciences, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK
| | - Jason Rodriguez
- The Centre for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Sze Yan Chan
- Wolfson Centre for Stem Cells, Tissue Engineering, and Modelling (STEM), Centre of Biomolecular Sciences, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK
| | - Jane Chisholm
- The Centre for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Gregg Duncan
- The Centre for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Namho Kim
- The Centre for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Amanda L Tatler
- Nottingham NIHR Biomedical Research Centre, Division of Respiratory Medicine, University of Nottingham, Nottingham University Hospitals NHS Trust, City Hospital, Nottingham NG5 1PB, UK
| | - Kevin M Shakesheff
- Wolfson Centre for Stem Cells, Tissue Engineering, and Modelling (STEM), Centre of Biomolecular Sciences, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK
| | - Justin Hanes
- The Centre for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Jung Soo Suk
- The Centre for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| | - James E Dixon
- Wolfson Centre for Stem Cells, Tissue Engineering, and Modelling (STEM), Centre of Biomolecular Sciences, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK.
| |
Collapse
|
49
|
Liang X, Liu L, Wei YQ, Gao GP, Wei XW. Clinical Evaluations of Toxicity and Efficacy of Nanoparticle-Mediated Gene Therapy. Hum Gene Ther 2018; 29:1227-1234. [PMID: 29893153 DOI: 10.1089/hum.2018.069] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Considerable efforts have been devoted to develop safe and efficient gene therapies for life-threatening or inherited diseases. The choice of gene delivery vehicle plays key roles in enhancing the therapeutic effect of nucleic acid cargo. To date, gene therapy approaches involving both viral vectors and nonviral vectors have been evaluated in clinical trials. With improvements in material science and nanotechnologies, positively charged nanoparticles have emerged as potential gene delivery vehicles. In this review, we highlight clinical trials that examined cationic nanocarrier-mediated gene therapy as well as discuss both the toxicity and efficacy of nanocarrier-based therapeutics.
Collapse
Affiliation(s)
- Xiao Liang
- 1 Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children, Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Li Liu
- 2 Laboratory of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yu-Quan Wei
- 1 Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children, Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China .,2 Laboratory of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Guang-Ping Gao
- 3 Horae Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Xia-Wei Wei
- 1 Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children, Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China .,2 Laboratory of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
50
|
Drumm M. Giants in Chest Medicine: Professor Pamela B. Davis, MD, PhD. Chest 2018; 154:240-241. [PMID: 30080499 DOI: 10.1016/j.chest.2018.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 03/12/2018] [Indexed: 10/28/2022] Open
Affiliation(s)
- Mitchell Drumm
- Departments of Pediatrics, and Genetics and Genome Sciences, The Research Institute for Children's Health, Connie and Jim Brown Professor in Cystic Fibrosis Research, Case Western Reserve University, Cleveland, Ohio.
| |
Collapse
|