1
|
Chi WY, Hu Y, Huang HC, Kuo HH, Lin SH, Kuo CTJ, Tao J, Fan D, Huang YM, Wu AA, Hung CF, Wu TC. Molecular targets and strategies in the development of nucleic acid cancer vaccines: from shared to personalized antigens. J Biomed Sci 2024; 31:94. [PMID: 39379923 PMCID: PMC11463125 DOI: 10.1186/s12929-024-01082-x] [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: 07/19/2024] [Accepted: 09/01/2024] [Indexed: 10/10/2024] Open
Abstract
Recent breakthroughs in cancer immunotherapies have emphasized the importance of harnessing the immune system for treating cancer. Vaccines, which have traditionally been used to promote protective immunity against pathogens, are now being explored as a method to target cancer neoantigens. Over the past few years, extensive preclinical research and more than a hundred clinical trials have been dedicated to investigating various approaches to neoantigen discovery and vaccine formulations, encouraging development of personalized medicine. Nucleic acids (DNA and mRNA) have become particularly promising platform for the development of these cancer immunotherapies. This shift towards nucleic acid-based personalized vaccines has been facilitated by advancements in molecular techniques for identifying neoantigens, antigen prediction methodologies, and the development of new vaccine platforms. Generating these personalized vaccines involves a comprehensive pipeline that includes sequencing of patient tumor samples, data analysis for antigen prediction, and tailored vaccine manufacturing. In this review, we will discuss the various shared and personalized antigens used for cancer vaccine development and introduce strategies for identifying neoantigens through the characterization of gene mutation, transcription, translation and post translational modifications associated with oncogenesis. In addition, we will focus on the most up-to-date nucleic acid vaccine platforms, discuss the limitations of cancer vaccines as well as provide potential solutions, and raise key clinical and technical considerations in vaccine development.
Collapse
Affiliation(s)
- Wei-Yu Chi
- Physiology, Biophysics and Systems Biology Graduate Program, Weill Cornell Medicine, New York, NY, USA
| | - Yingying Hu
- Tri-Institutional PhD Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hsin-Che Huang
- Tri-Institutional PhD Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hui-Hsuan Kuo
- Pharmacology PhD Program, Weill Cornell Medicine, New York, NY, USA
| | - Shu-Hong Lin
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas Graduate School of Biomedical Sciences at Houston and MD Anderson Cancer Center, Houston, TX, USA
| | - Chun-Tien Jimmy Kuo
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Julia Tao
- Department of Pathology, Johns Hopkins School of Medicine, 1550 Orleans St, CRB II Room 309, Baltimore, MD, 21287, USA
| | - Darrell Fan
- Department of Pathology, Johns Hopkins School of Medicine, 1550 Orleans St, CRB II Room 309, Baltimore, MD, 21287, USA
| | - Yi-Min Huang
- Department of Pathology, Johns Hopkins School of Medicine, 1550 Orleans St, CRB II Room 309, Baltimore, MD, 21287, USA
| | - Annie A Wu
- Department of Pathology, Johns Hopkins School of Medicine, 1550 Orleans St, CRB II Room 309, Baltimore, MD, 21287, USA
| | - Chien-Fu Hung
- Department of Pathology, Johns Hopkins School of Medicine, 1550 Orleans St, CRB II Room 309, Baltimore, MD, 21287, USA
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Obstetrics and Gynecology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - T-C Wu
- Department of Pathology, Johns Hopkins School of Medicine, 1550 Orleans St, CRB II Room 309, Baltimore, MD, 21287, USA.
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Department of Obstetrics and Gynecology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
2
|
Mapfumo P, Reichel LS, Leer K, Egger J, Dzierza A, Peneva K, Fischer D, Traeger A. Harnessing Guanidinium and Imidazole Functional Groups: A Dual-Charged Polymer Strategy for Enhanced Gene Delivery. ACS Macro Lett 2024; 13:1000-1007. [PMID: 39052525 PMCID: PMC11340021 DOI: 10.1021/acsmacrolett.4c00321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/30/2024] [Accepted: 07/10/2024] [Indexed: 07/27/2024]
Abstract
Histidine and arginine are two amino acids that exhibit beneficial properties for gene delivery. In particular, the imidazole group of histidine facilitates endosomal release, while the guanidinium group of arginine promotes cellular entry. Consequently, a dual-charged copolymer library based on these amino acids was synthesized via reversible addition-fragmentation chain transfer (RAFT) polymerization. The content of the N-acryloyl-l-histidine (His) monomer was systematically increased, while maintaining consistent levels of methyl N-acryloyl-l-argininate hydrochloride (ArgOMe) or N-(4-guanidinobutyl)acrylamide hydrochloride (GBAm). The resulting polymers formed stable, nanosized polyplexes when complexed with nucleic acids. Remarkably, candidates with increased His content exhibited reduced cytotoxicity profiles and enhanced transfection efficiency, particularly retaining this performance level at lower pDNA concentrations. Furthermore, endosomal release studies revealed that increased His content improved endosomal release, while ArgOMe improved cellular entry. These findings underscore the potential of customized dual-charged copolymers and the synergistic effects of His and ArgOMe/GBAm in enhancing gene delivery.
Collapse
Affiliation(s)
- Prosper
P. Mapfumo
- Institute
of Organic Chemistry and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany
| | - Liên S. Reichel
- Institute
of Organic Chemistry and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany
| | - Katharina Leer
- Institute
of Organic Chemistry and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany
| | - Jan Egger
- Division
of Pharmaceutical Technology and Biopharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Cauerstr. 4, 91058 Erlangen, Germany
| | - Andreas Dzierza
- Division
of Pharmaceutical Technology and Biopharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Cauerstr. 4, 91058 Erlangen, Germany
| | - Kalina Peneva
- Institute
of Organic Chemistry and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany
- Jena
Center for Soft Matter (JCSM), Friedrich
Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
- Center
for Energy and Environmental Chemistry Jena (CEEC), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Dagmar Fischer
- Jena
Center for Soft Matter (JCSM), Friedrich
Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
- Division
of Pharmaceutical Technology and Biopharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Cauerstr. 4, 91058 Erlangen, Germany
- FAU NeW -
Research Center New Bioactive Compounds, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Anja Traeger
- Institute
of Organic Chemistry and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany
- Jena
Center for Soft Matter (JCSM), Friedrich
Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| |
Collapse
|
3
|
Wang C, Yuan F. A comprehensive comparison of DNA and RNA vaccines. Adv Drug Deliv Rev 2024; 210:115340. [PMID: 38810703 PMCID: PMC11181159 DOI: 10.1016/j.addr.2024.115340] [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: 03/28/2024] [Revised: 05/06/2024] [Accepted: 05/18/2024] [Indexed: 05/31/2024]
Abstract
Nucleic acid technology has revolutionized vaccine development, enabling rapid design and production of RNA and DNA vaccines for prevention and treatment of diseases. The successful deployment of mRNA and plasmid DNA vaccines against COVID-19 has further validated the technology. At present, mRNA platform is prevailing due to its higher efficacy, while DNA platform is undergoing rapid evolution because it possesses unique advantages that can potentially overcome the problems associated with the mRNA platform. To help understand the recent performances of the two vaccine platforms and recognize their clinical potentials in the future, this review compares the advantages and drawbacks of mRNA and DNA vaccines that are currently known in the literature, in terms of development timeline, financial cost, ease of distribution, efficacy, safety, and regulatory approval of products. Additionally, the review discusses the ongoing clinical trials, strategies for improvement, and alternative designs of RNA and DNA platforms for vaccination.
Collapse
Affiliation(s)
- Chunxi Wang
- Department of Biomedical Engineering, Duke University, Durham, NC 27705, United States
| | - Fan Yuan
- Department of Biomedical Engineering, Duke University, Durham, NC 27705, United States.
| |
Collapse
|
4
|
Zhang Y, Zhang M, Song H, Dai Q, Liu C. Tumor Microenvironment-Responsive Polymer-Based RNA Delivery Systems for Cancer Treatment. SMALL METHODS 2024:e2400278. [PMID: 38803312 DOI: 10.1002/smtd.202400278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 03/30/2024] [Indexed: 05/29/2024]
Abstract
Ribonucleic acid (RNA) therapeutics offer a broad prospect in cancer treatment. However, their successful application requires overcoming various physiological barriers to effectively deliver RNAs to the target sites. Currently, a number of RNA delivery systems based on polymeric nanoparticles are developed to overcome these barriers in RNA delivery. This work provides an overview of the existing RNA therapeutics for cancer gene therapy, and particularly summarizes those that are entering the clinical phase. This work then discusses the core features and latest research developments of tumor microenvironment-responsive polymer-based RNA delivery carriers which are designed based on the pathological characteristics of the tumor microenvironment. Finally, this work also proposes opportunities for the transformation of RNA therapies into cancer immunotherapy methods in clinical applications.
Collapse
Affiliation(s)
- Yahan Zhang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Ming Zhang
- Department of Pathology, Peking University International Hospital, Beijing, 102206, China
| | - Haiqin Song
- Department of General Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200025, China
| | - Qiong Dai
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Chaoyong Liu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
5
|
Genedy HH, Humbert P, Laoulaou B, Le Moal B, Fusellier M, Passirani C, Le Visage C, Guicheux J, Lepeltier É, Clouet J. MicroRNA-targeting nanomedicines for the treatment of intervertebral disc degeneration. Adv Drug Deliv Rev 2024; 207:115214. [PMID: 38395361 DOI: 10.1016/j.addr.2024.115214] [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: 12/13/2023] [Revised: 02/09/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024]
Abstract
Low back pain stands as a pervasive global health concern, afflicting almost 80% of adults at some point in their lives with nearly 40% attributable to intervertebral disc degeneration (IVDD). As only symptomatic relief can be offered to patients there is a dire need for innovative treatments.Given the accumulating evidence that multiple microRNAs (miRs) are dysregulated during IVDD, they could have a huge potential against this debilitating condition. The way miRs can profoundly modulate signaling pathways and influence several cellular processes at once is particularly exciting to tackle this multifaceted disorder. However, miR delivery encounters extracellular and intracellular biological barriers. A promising technology to address this challenge is the vectorization of miRs within nanoparticles, providing both protection and enhancing their uptake within the scarce target cells of the degenerated IVD. This comprehensive review presents the diverse spectrum of miRs' connection with IVDD and demonstrates their therapeutic potential when vectorized in nanomedicines.
Collapse
Affiliation(s)
- Hussein H Genedy
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, Nantes, France; Univ Angers, INSERM, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| | - Paul Humbert
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, Nantes, France
| | - Bilel Laoulaou
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, Nantes, France; Univ Angers, INSERM, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| | - Brian Le Moal
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, Nantes, France; Univ Angers, INSERM, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| | - Marion Fusellier
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, Nantes, France; Department of Diagnostic Imaging, CRIP, ONIRIS, College of Veterinary Medicine, Food Science and Engineering, Nantes F-44307, France
| | | | - Catherine Le Visage
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, Nantes, France
| | - Jérôme Guicheux
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, Nantes, France
| | - Élise Lepeltier
- Univ Angers, INSERM, CNRS, MINT, SFR ICAT, F-49000 Angers, France; Institut Universitaire de France (IUF), France.
| | - Johann Clouet
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, Nantes, France
| |
Collapse
|
6
|
Tang F, Ding A, Xu Y, Ye Y, Li L, Xie R, Huang W. Gene and Photothermal Combination Therapy: Principle, Materials, and Amplified Anticancer Intervention. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307078. [PMID: 37775950 DOI: 10.1002/smll.202307078] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/19/2023] [Indexed: 10/01/2023]
Abstract
Gene therapy (GT) and photothermal therapy (PTT) have emerged as promising alternatives to chemotherapy and radiotherapy for cancer treatment, offering noninvasiveness and reduced side effects. However, their efficacy as standalone treatments is limited. GT exhibits slow response rates, while PTT is confined to local tumor ablation. The convergence of GT and PTT, known as GT-PTT, facilitated by photothermal gene nanocarriers, has attracted considerable attention across various disciplines. In this integrated approach, GT reciprocates PTT by sensitizing cellular response to heat, while PTT benefits GT by improving gene translocation, unpacking, and expression. Consequently, this integration presents a unique opportunity for cancer therapy with rapid response and improved effectiveness. Extensive efforts over the past few years have been dedicated to the development of GT-PTT, resulting in notable achievements and rapid progress from the laboratory to potential clinical applications. This comprehensive review outlines recent advances in GT-PTT, including synergistic mechanisms, material systems, imaging-guided therapy, and anticancer applications. It also explores the challenges and future prospects in this nascent field. By presenting innovative ideas and insights into the implementation of GT-PTT for enhanced cancer therapy, this review aims to inspire further progress in this promising area of research.
Collapse
Affiliation(s)
- Fang Tang
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen, 361005, China
- Future Display Institute in Xiamen, Xiamen, 361005, China
| | - Aixiang Ding
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen, 361005, China
| | - Yao Xu
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen, 361005, China
| | - Yingsong Ye
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen, 361005, China
| | - Lin Li
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen, 361005, China
- Future Display Institute in Xiamen, Xiamen, 361005, China
- Frontiers Science Center for Flexible Electronics (IFE), Northwestern Polytechnical University, Xi'an, 710072, China
| | - Rongjun Xie
- Fujian Key Laboratory of Materials Genome, College of Materials, Xiamen University, Xiamen, 361005, China
| | - Wei Huang
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen, 361005, China
- Future Display Institute in Xiamen, Xiamen, 361005, China
- Frontiers Science Center for Flexible Electronics (IFE), Northwestern Polytechnical University, Xi'an, 710072, China
| |
Collapse
|
7
|
Xiu K, Zhang J, Xu J, Chen YE, Ma PX. Recent progress in polymeric gene vectors: Delivery mechanisms, molecular designs, and applications. BIOPHYSICS REVIEWS 2023; 4:011313. [PMID: 37008888 PMCID: PMC10062053 DOI: 10.1063/5.0123664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 03/07/2023] [Indexed: 03/30/2023]
Abstract
Gene therapy and gene delivery have drawn extensive attention in recent years especially when the COVID-19 mRNA vaccines were developed to prevent severe symptoms caused by the corona virus. Delivering genes, such as DNA and RNA into cells, is the crucial step for successful gene therapy and remains a bottleneck. To address this issue, vehicles (vectors) that can load and deliver genes into cells are developed, including viral and non-viral vectors. Although viral gene vectors have considerable transfection efficiency and lipid-based gene vectors become popular since the application of COVID-19 vaccines, their potential issues including immunologic and biological safety concerns limited their applications. Alternatively, polymeric gene vectors are safer, cheaper, and more versatile compared to viral and lipid-based vectors. In recent years, various polymeric gene vectors with well-designed molecules were developed, achieving either high transfection efficiency or showing advantages in certain applications. In this review, we summarize the recent progress in polymeric gene vectors including the transfection mechanisms, molecular designs, and biomedical applications. Commercially available polymeric gene vectors/reagents are also introduced. Researchers in this field have never stopped seeking safe and efficient polymeric gene vectors via rational molecular designs and biomedical evaluations. The achievements in recent years have significantly accelerated the progress of polymeric gene vectors toward clinical applications.
Collapse
Affiliation(s)
- Kemao Xiu
- Department of Biologic and Materials Sciences and Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | | | | | - Peter X. Ma
- Author to whom correspondence should be addressed:. Tel.: (734) 764-2209
| |
Collapse
|
8
|
Delivery of CRISPR/Cas9 Plasmid DNA by Hyperbranched Polymeric Nanoparticles Enables Efficient Gene Editing. Cells 2022; 12:cells12010156. [PMID: 36611948 PMCID: PMC9818138 DOI: 10.3390/cells12010156] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/22/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023] Open
Abstract
Gene editing nucleases such as CRISPR/Cas9 have enabled efficient and precise gene editing in vitro and hold promise of eventually achieving in vivo gene editing based therapy. However, a major challenge for their use is the lack of a safe and effective virus-free system to deliver gene editing nuclease elements. Polymers are a promising class of delivery vehicle due to their higher safety compared to currently used viral vectors, but polymers suffer from lower transfection efficiency. Polymeric vectors have been used for small nucleotide delivery but have yet to be used successfully with plasmid DNA (pDNA), which is often several hundred times larger than small nucleotides, presenting an engineering challenge. To address this, we extended our previously reported hyperbranched polymer (HP) delivery system for pDNA delivery by synthesizing several variants of HPs: HP-800, HP-1.8K, HP-10K, HP-25K. We demonstrate that all HPs have low toxicity in various cultured cells, with HP-25K being the most efficient at packaging and delivering pDNA. Importantly, HP-25K mediated delivery of CRISPR/Cas9 pDNA resulted in higher gene-editing rates than all other HPs and Lipofectamine at several clinically significant loci in different cell types. Consistently, HP-25K also led to more robust base editing when delivering the CRISPR base editor "BE4-max" pDNA to cells compared with Lipofectamine. The present work demonstrates that HP nanoparticles represent a promising class of vehicle for the non-viral delivery of pDNA towards the clinical application of gene-editing therapy.
Collapse
|
9
|
Vlach M, Coppens-Exandier H, Jamin A, Berchel M, Scaviner J, Chesné C, Montier T, Jaffrès PA, Corlu A, Loyer P. Liposome-Mediated Gene Transfer in Differentiated HepaRG™ Cells: Expression of Liver Specific Functions and Application to the Cytochrome P450 2D6 Expression. Cells 2022; 11:cells11233904. [PMID: 36497165 PMCID: PMC9737581 DOI: 10.3390/cells11233904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 11/19/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
The goal of this study was to establish a procedure for gene delivery mediated by cationic liposomes in quiescent differentiated HepaRG™ human hepatoma cells. We first identified several cationic lipids promoting efficient gene transfer with low toxicity in actively dividing HepG2, HuH7, BC2 and progenitor HepaRG™ human hepatoma cells. The lipophosphoramidate Syn1-based nanovector, which allowed the highest transfection efficiencies of progenitor HepaRG™ cells, was next used to transfect differentiated HepaRG™ cells. Lipofection of these cells using Syn1-based liposome was poorly efficient most likely because the differentiated HepaRG™ cells are highly quiescent. Thus, we engineered the differentiated HepaRG™ Mitogenic medium supplement (ADD1001) that triggered robust proliferation of differentiated cells. Importantly, we characterized the phenotypical changes occurring during proliferation of differentiated HepaRG™ cells and demonstrated that mitogenic stimulation induced a partial and transient decrease in the expression levels of some liver specific functions followed by a fast recovery of the full differentiation status upon removal of the mitogens. Taking advantage of the proliferation of HepaRG™ cells, we defined lipofection conditions using Syn1-based liposomes allowing transient expression of the cytochrome P450 2D6, a phase I enzyme poorly expressed in HepaRG cells, which opens new means for drug metabolism studies in HepaRG™ cells.
Collapse
Affiliation(s)
- Manuel Vlach
- Institut NUMECAN (Nutrition Metabolisms and Cancer), F-35000 Rennes, France
- Institut AGRO Rennes-Angers, F-35042 Rennes, France
| | - Hugo Coppens-Exandier
- Institut NUMECAN (Nutrition Metabolisms and Cancer), F-35000 Rennes, France
- Biopredic International, F-35760 Saint Grégoire, France
| | - Agnès Jamin
- Biopredic International, F-35760 Saint Grégoire, France
| | - Mathieu Berchel
- Univ. Brest, CNRS, CEMCA, UMR 6521, F-29238 Brest, France
- Plateforme BiogenOuest SynNanoVect, F-44035 Nantes, France
| | - Julien Scaviner
- Institut NUMECAN (Nutrition Metabolisms and Cancer), F-35000 Rennes, France
- Biopredic International, F-35760 Saint Grégoire, France
| | | | - Tristan Montier
- Plateforme BiogenOuest SynNanoVect, F-44035 Nantes, France
- Univ. Brest, INSERM, EFS, UMR 1078, GGB-GTCA, F-29200 Brest, France
| | - Paul-Alain Jaffrès
- Univ. Brest, CNRS, CEMCA, UMR 6521, F-29238 Brest, France
- Plateforme BiogenOuest SynNanoVect, F-44035 Nantes, France
| | - Anne Corlu
- Institut NUMECAN (Nutrition Metabolisms and Cancer), F-35000 Rennes, France
- Correspondence: (A.C.); (P.L.); Tel.: +33-(02)-23233873 (P.L.)
| | - Pascal Loyer
- Institut NUMECAN (Nutrition Metabolisms and Cancer), F-35000 Rennes, France
- Plateforme BiogenOuest SynNanoVect, F-44035 Nantes, France
- Correspondence: (A.C.); (P.L.); Tel.: +33-(02)-23233873 (P.L.)
| |
Collapse
|
10
|
Padmakumar S, D'Souza A, Parayath NN, Bleier BS, Amiji MM. Nucleic acid therapies for CNS diseases: Pathophysiology, targets, barriers, and delivery strategies. J Control Release 2022; 352:121-145. [PMID: 36252748 DOI: 10.1016/j.jconrel.2022.10.018] [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: 06/23/2022] [Revised: 09/10/2022] [Accepted: 10/10/2022] [Indexed: 11/08/2022]
Abstract
Nucleic acid therapeutics have emerged as one of the very advanced and efficacious treatment approaches for debilitating health conditions, including those diseases affecting the central nervous system (CNS). Precise targeting with an optimal control over gene regulation confers long-lasting benefits through the administration of nucleic acid payloads via viral, non-viral, and engineered vectors. The current review majorly focuses on the development and clinical translational potential of non-viral vectors for treating CNS diseases with a focus on their specific design and targeting approaches. These carriers must be able to surmount the various intracellular and extracellular barriers, to ensure successful neuronal transfection and ultimately attain higher therapeutic efficacies. Additionally, the specific challenges associated with CNS administration also include the presence of blood-brain barrier (BBB), the complex pathophysiological and biochemical changes associated with different disease conditions and the existence of non-dividing cells. The advantages offered by lipid-based or polymeric systems, engineered proteins, particle-based systems coupled with various approaches of neuronal targeting have been discussed in the context of a variety of CNS diseases. The possibilities of rapid yet highly efficient gene modifications rendered by the breakthrough methodologies for gene editing and gene manipulation have also opened vast avenues of research in neuroscience and CNS disease therapy. The current review also underscores the extensive scientific efforts to optimize specialized, efficacious yet non-invasive and safer administration approaches to overcome the therapeutic delivery challenges specifically posed by the CNS transport barriers and the overall obstacles to clinical translation.
Collapse
Affiliation(s)
- Smrithi Padmakumar
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA 02115, USA
| | - Anisha D'Souza
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA 02115, USA; Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 20115, USA
| | - Neha N Parayath
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA 02115, USA
| | - Benjamin S Bleier
- Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 20115, USA
| | - Mansoor M Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA 02115, USA; Department of Chemical Engineering, College of Engineering, Northeastern University, Boston, MA 02115, USA.
| |
Collapse
|
11
|
Sachdev S, Potočnik T, Rems L, Miklavčič D. Revisiting the role of pulsed electric fields in overcoming the barriers to in vivo gene electrotransfer. Bioelectrochemistry 2022; 144:107994. [PMID: 34930678 DOI: 10.1016/j.bioelechem.2021.107994] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 10/15/2021] [Accepted: 11/02/2021] [Indexed: 12/21/2022]
Abstract
Gene therapies are revolutionizing medicine by providing a way to cure hitherto incurable diseases. The scientific and technological advances have enabled the first gene therapies to become clinically approved. In addition, with the ongoing COVID-19 pandemic, we are witnessing record speeds in the development and distribution of gene-based vaccines. For gene therapy to take effect, the therapeutic nucleic acids (RNA or DNA) need to overcome several barriers before they can execute their function of producing a protein or silencing a defective or overexpressing gene. This includes the barriers of the interstitium, the cell membrane, the cytoplasmic barriers and (in case of DNA) the nuclear envelope. Gene electrotransfer (GET), i.e., transfection by means of pulsed electric fields, is a non-viral technique that can overcome these barriers in a safe and effective manner. GET has reached the clinical stage of investigations where it is currently being evaluated for its therapeutic benefits across a wide variety of indications. In this review, we formalize our current understanding of GET from a biophysical perspective and critically discuss the mechanisms by which electric field can aid in overcoming the barriers. We also identify the gaps in knowledge that are hindering optimization of GET in vivo.
Collapse
Affiliation(s)
- Shaurya Sachdev
- University of Ljubljana, Faculty of Electrical Engineering, Tržaška cesta 25, 1000 Ljubljana, Slovenia
| | - Tjaša Potočnik
- University of Ljubljana, Faculty of Electrical Engineering, Tržaška cesta 25, 1000 Ljubljana, Slovenia
| | - Lea Rems
- University of Ljubljana, Faculty of Electrical Engineering, Tržaška cesta 25, 1000 Ljubljana, Slovenia
| | - Damijan Miklavčič
- University of Ljubljana, Faculty of Electrical Engineering, Tržaška cesta 25, 1000 Ljubljana, Slovenia.
| |
Collapse
|
12
|
Hooshmand SE, Sabet MJ, Hasanzadeh A, Mousavi SMK, Moghadam NH, Hooshmand SA, Rabiee N, Liu Y, Hamblin MR, Karimi M. Histidine‐enhanced gene delivery systems: The state of the art. J Gene Med 2022; 24:e3415. [DOI: 10.1002/jgm.3415] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/26/2022] [Accepted: 01/29/2022] [Indexed: 11/08/2022] Open
Affiliation(s)
- Seyyed Emad Hooshmand
- 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
| | - Makkieh Jahanpeimay Sabet
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine Iran University of Medical Sciences Tehran Iran
| | - Akbar Hasanzadeh
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine Iran University of Medical Sciences Tehran Iran
| | - Seyede Mahtab Kamrani Mousavi
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine Iran University of Medical Sciences Tehran Iran
| | - Niloofar Haeri Moghadam
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine Iran University of Medical Sciences Tehran Iran
| | - Seyed Aghil Hooshmand
- Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics University of Tehran Tehran Iran
| | - Navid Rabiee
- Department of Physics Sharif University of Technology Tehran Iran
- School of Engineering Macquarie University Sydney New South Wales Australia
| | - Yong Liu
- Institute of Functional Nano & Soft Materials (FUNSOM) Soochow University Suzhou Jiangsu China
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science University of Johannesburg South Africa
| | - Mahdi Karimi
- 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
- Oncopathology Research Center Iran University of Medical Sciences Tehran Iran
- Research Center for Science and Technology in Medicine Tehran University of Medical Sciences Tehran Iran
| |
Collapse
|
13
|
Qin J, Xue L, Gong N, Zhang H, Shepherd SJ, Haley RM, Swingle KL, Mitchell MJ. RGD peptide-based lipids for targeted mRNA delivery and gene editing applications. RSC Adv 2022; 12:25397-25404. [PMID: 36199352 PMCID: PMC9450108 DOI: 10.1039/d2ra02771b] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 08/08/2022] [Indexed: 11/21/2022] Open
Abstract
mRNA therapeutics are promising platforms for protein replacement therapies and gene editing technologies. When delivered via non-viral gene delivery systems, such as lipid nanoparticles (LNPs), mRNA therapeutics are easy to produce and show low toxicity and immunogenicity. However, LNPs show limited delivery efficiency and tissue specificity in certain applications. To overcome this, we designed RGD peptide (Arg-Gly-Asp) based ionizable lipids, which can be formulated into LNPs for integrin binding on cells and targeted mRNA delivery. RGD-LNPs were formulated using microfluidic devices and screened in vitro for size, mRNA encapsulation efficiency, transfection efficiency, and cell viability. A lead candidate, 1A RGD-based hybrid LNP, showed effective mRNA encapsulation and transfection, and was selected for further testing, including the co-delivery of Cas9 mRNA and sgRNA for gene editing applications. In vitro, 1A RGD-based hybrid LNP outperformed a non-targeted control LNP and showed GFP knockout efficiencies up to 90%. Further, the improved cellular uptake was reversed in the presence of soluble RGD, supporting the hypothesis that this improved uptake is RGD-dependent. In vivo, 1A RGD-based hybrid LNPs showed comparable mRNA delivery to the liver and spleen, when compared to a non-targeted control, and had increased expression in the whole body. Overall, this RGD-based hybrid LNP system is a promising platform for targeted mRNA delivery, which may allow for mRNA-based protein replacement and gene editing in a more efficient and specific manner with reduced off-target effects. We developed RGD peptide based ionizable lipids, which can be formulated into LNPs for integrin-dependent targeted mRNA delivery and gene editing applications.![]()
Collapse
Affiliation(s)
- Jingya Qin
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lulu Xue
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ningqiang Gong
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hanwen Zhang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sarah J. Shepherd
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rebecca M. Haley
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kelsey L. Swingle
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael J. Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19014, USA
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
| |
Collapse
|
14
|
Betker JL, Anchordoquy TJ. The Effect of Repeat Administration of Lipoplexes on Gene Delivery, Biodistribution, and Cytokine Response in Immunocompetent Tumor-bearing Mice. J Pharm Sci 2021; 111:1926-1936. [PMID: 34929156 DOI: 10.1016/j.xphs.2021.12.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 12/14/2021] [Accepted: 12/14/2021] [Indexed: 12/19/2022]
Abstract
It is becoming increasingly clear that the intravenous administration of nanoparticles elicits an immune response that compromises delivery efficiency and can be life threatening. This study investigated both the systemic and tissue-level cytokine response to repeat administration of lipoplexes coated with either lactose or PEG. We report that blood cytokine levels differ significantly from that observed in individual tissues. While we consistently observed a reduced cytokine response to lactosylated particles, this did not result in enhanced delivery or expression as compared to PEGylated formulations. We also document that repeat injection did not increase plasmid levels in the liver, lung, or spleen, but delivery to the tumor was enhanced under these conditions. In addition, we show that changes in neither blood nor tissue cytokines correlated strongly with reporter gene expression, and we observed relatively constant expression efficiencies (RLU/ng plasmid) across all tissues despite a considerably reduced cytokine response in the tumor. Together, these results indicate that both biodistribution and cytokine responses are dramatically altered by a repeat intravenous injection of lipoplexes, and that the mechanisms regulating reporter gene expression are not straightforward.
Collapse
Affiliation(s)
- Jamie L Betker
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| | - Thomas J Anchordoquy
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States.
| |
Collapse
|
15
|
Kanazawa T, Hoashi Y, Ibaraki H, Takashima Y, Okada H. Electroporation-Based ex Vivo Gene Delivery into Dendritic Cells by Anionic Polymer-Coated Versatile Nuclear Localization Signal/pDNA Complex. Biol Pharm Bull 2021; 44:1866-1871. [PMID: 34853269 DOI: 10.1248/bpb.b21-00559] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In this study, we focused on a nuclear localization signal (NLS)-based versatile peptide vector, designed by us, combined with electroporation (EP) to establish an efficient gene delivery system to non-dividing or slow growing dendritic cells. We determined the intranuclear transport, gene expression, and cell viability in JAWS II mouse dendritic cells transfected with the green fluorescent protein (GFP) expression plasmid DNA alone (naked pEGFP); positive charged complex of NLS derivative STR-CH2SV40H2C, and pEGFP (binary complex); or negative charged complex of the binary complex with a biocompatible polyanion, γ-polyglutamic acid (ternary complex) combined with or without EP application. Although the binary complex showed higher nuclear transport and GFP expression in the absence of EP than those for naked pEGFP, the combination of EP significantly decreased the cell viability and did not improve the efficiency of compared gene expression. However, in the ternary complex, the intranuclear transport and GFP expression efficiency were significantly higher than those of naked pEGFP and the binary complex when combined with EP, and there was no decrease in cell viability. The results suggest that polyanion-coated ternary complex with EP is useful for non-viral gene delivery system into non-dividing cells for ex vivo gene/cell therapy.
Collapse
Affiliation(s)
- Takanori Kanazawa
- School of Pharmacy, Tokyo University of Pharmacy and Life Sciences.,School of Pharmaceutical Sciences, University of Shizuoka
| | - Yuki Hoashi
- School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| | - Hisako Ibaraki
- School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| | - Yuuki Takashima
- School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| | - Hiroaki Okada
- School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| |
Collapse
|
16
|
Amani A, Dustparast M, Noruzpour M, Zakaria RA, Ebrahimi HA. Design and Invitro Characterization of Green Synthesized Magnetic Nanoparticles Conjugated with Multitargeted Poly Lactic Acid Copolymers for Co-delivery of siRNA and Paclitaxel. Eur J Pharm Sci 2021; 167:106007. [PMID: 34520835 DOI: 10.1016/j.ejps.2021.106007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 08/21/2021] [Accepted: 09/10/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND The self-assembling of various amphipathic copolymers is a simple method that allows the preparation of complex nanoparticles with several useful properties. In the present study, the polylactic acid-polyethylene glycol-folate (PLA-PEG-FA) (PPF), PLA-PEG-T7 peptide (PPT) and PLA-Chitosan-Spermine (PCS) copolymers were synthesized separately. METHODS These copolymers combined with Fe3O4 magnetic core and loaded with paclitaxel (PTX)/siRNA-FAM to form magnetic PCS/PPF/PPT/PTX/siRNA micelles (MPCSFT/PTX/siRNA) and were characterized using physicochemical and biological analysis. RESULTS The results revealed that the MPCSPFT/PTX/siRNA had spherical morphology with particle size and zeta potential about 197 nm and -7.8 mV, respectively. Release assay was determined under neutral (pH=7.4) and acidic pH (pH=6) conditions to simulate PTX and siRNA release profile from MPCSPFT/PTX/siRNA micelles in normal and cancerous tissues. The ability of MPCSPFT for co-delivery of PTX and siRNA into MCF-7 cells was determined by MTT and flow cytometry tests, respectively. The results revealed that the release rate of siRNA and PTX from MPCSPFT/PTX/siRNA nanoparticles under an acidic environment (pH=6) was significantly higher than that of their release rate in a neutral medium (pH=7.4). CONCLUSION Conjugation of both folic acid and T7-peptide on the surface of micelles compared to separate conjugation of one of these ligands, increased the efficiency of drug and siRNA delivery to breast cancer cells.
Collapse
Affiliation(s)
- Amin Amani
- Department of Pharmaceutics, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran; Department of Agronomy and Plant Breeding, Faculty of Agriculture and Natural Resources, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Mohammad Dustparast
- Department of Pharmaceutics, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mehran Noruzpour
- Department of Agronomy and Plant Breeding, Faculty of Agriculture and Natural Resources, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Rasool Asghari Zakaria
- Department of Agronomy and Plant Breeding, Faculty of Agriculture and Natural Resources, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Hossein Ali Ebrahimi
- Department of Pharmaceutics, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
17
|
Tian X, Qiu N, Shen Y, Zhang S, Dang C, Hu G, Wang Z, Chen J, Ding S. Preparation of poly(DMA-co-DBA) and its application in gene delivery. INT J POLYM MATER PO 2021. [DOI: 10.1080/00914037.2021.1999952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Xiaoxiao Tian
- College of Chemical Engineering, Huaiyin Institute of Technology, Huaian, P. R. China
| | - Nasha Qiu
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, P. R. China
| | - Youqing Shen
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, P. R. China
| | - Shizhong Zhang
- College of Chemical Engineering, Huaiyin Institute of Technology, Huaian, P. R. China
| | - Chun Dang
- College of Chemical Engineering, Huaiyin Institute of Technology, Huaian, P. R. China
| | - Guang Hu
- College of Chemical Engineering, Huaiyin Institute of Technology, Huaian, P. R. China
| | - Zhihui Wang
- College of Chemical Engineering, Huaiyin Institute of Technology, Huaian, P. R. China
| | - Jing Chen
- College of Chemical Engineering, Huaiyin Institute of Technology, Huaian, P. R. China
| | - Shijie Ding
- College of Chemical Engineering, Huaiyin Institute of Technology, Huaian, P. R. China
| |
Collapse
|
18
|
Arora S, Sharma D, Layek B, Singh J. A Review of Brain-Targeted Nonviral Gene-Based Therapies for the Treatment of Alzheimer's Disease. Mol Pharm 2021; 18:4237-4255. [PMID: 34705472 DOI: 10.1021/acs.molpharmaceut.1c00611] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Diseases of the central nervous system (CNS) are difficult to treat owing to the complexity of the brain and the presence of a natural blood-brain-barrier (BBB). Alzheimer's disease (AD) is one of the major progressive and currently incurable neurodegenerative disorders of the CNS, which accounts for 60-80% of cases of dementia. The pathophysiology of AD involves the accumulation of amyloid beta (Aβ) plaques and neurofibrillary tangles (NFTs) in the brain. Additionally, synaptic loss and imbalance of neuronal signaling molecules are characterized as important markers of AD. Existing treatments of AD help in the management of its symptoms and aim toward the maintenance of cognitive functions, behavior, and attenuation of gradual memory loss. Over the past decade, nonviral gene therapy has attracted increasing interest due to its various advantages over its viral counterparts. Moreover, advancements in nonviral gene technology have led to their increasing contributions in clinical trials. However, brain-targeted nonviral gene delivery vectors come across various extracellular and intracellular barriers, limiting their ability to transfer the therapeutic gene into the target cells. Chief barriers to nonviral gene therapy have been discussed briefly in this review. We have also highlighted the rapid advancement of several nonviral gene therapies for AD, which are broadly categorized into physical and chemical methods. These methods aim to modulate Aβ, beta-site amyloid precursor protein (APP) cleaving enzyme 1 (BACE1), apolipoprotein E, or neurotrophic factors' expression in the CNS. Overall, this review discusses challenges and recent advancements of nonviral gene therapy for AD.
Collapse
Affiliation(s)
- Sanjay Arora
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Divya Sharma
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Buddhadev Layek
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Jagdish Singh
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, North Dakota 58105, United States
| |
Collapse
|
19
|
Sharma D, Arora S, Singh J, Layek B. A review of the tortuous path of nonviral gene delivery and recent progress. Int J Biol Macromol 2021; 183:2055-2073. [PMID: 34087309 PMCID: PMC8266766 DOI: 10.1016/j.ijbiomac.2021.05.192] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/24/2021] [Accepted: 05/28/2021] [Indexed: 02/06/2023]
Abstract
Gene therapy encompasses the transfer of exogenous genetic materials into the patient's target cells to treat or prevent diseases. Nevertheless, the transfer of genetic material into desired cells is challenging and often requires specialized tools or delivery systems. For the past 40 years, scientists are mainly pursuing various viruses as gene delivery vectors, and the overall progress has been slow and far from the expectation. As an alternative, nonviral vectors have gained substantial attention due to their several advantages, including superior safety profile, enhanced payload capacity, and stealth abilities. Since nonviral vectors encounter multiple extra- and intra-cellular barriers limiting the transfer of genetic payload into the target cell nucleus, we have discussed these barriers in detail for this review. A direct approach, utilizing physical methods like electroporation, sonoporation, gene gun, eliminate the requirement for a specific carrier for gene delivery. In contrast, chemical methods of gene transfer exploit natural or synthetic compounds as carriers to increase cellular targeting and gene therapy effectiveness. We have also emphasized the recent advancements aimed at enhancing the current nonviral approaches. Therefore, in this review, we have focused on discussing the current evolving state of nonviral gene delivery systems and their future perspectives.
Collapse
Affiliation(s)
- Divya Sharma
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo 58105, ND, USA
| | - Sanjay Arora
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo 58105, ND, USA
| | - Jagdish Singh
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo 58105, ND, USA
| | - Buddhadev Layek
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo 58105, ND, USA.
| |
Collapse
|
20
|
Kim J, Vaughan HJ, Zamboni CG, Sunshine JC, Green JJ. High-throughput evaluation of polymeric nanoparticles for tissue-targeted gene expression using barcoded plasmid DNA. J Control Release 2021; 337:105-116. [PMID: 34097924 DOI: 10.1016/j.jconrel.2021.05.047] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 05/03/2021] [Accepted: 05/31/2021] [Indexed: 12/17/2022]
Abstract
Successful systemic gene delivery requires specific tissue targeting as well as efficient intracellular transfection. Increasingly, research laboratories are fabricating libraries of novel nanoparticles, engineering both new biomaterial structures and composition ratios of multicomponent systems. Yet, methods for screening gene delivery vehicles directly in vivo are often low-throughout, limiting the number of candidate nanoparticles that can be investigated. Here, we report a comprehensive, high-throughput method to evaluate a library of polymeric nanoparticles in vivo for tissue-specific gene delivery. The method involves pairing each nanoparticle formulation with a plasmid DNA (pDNA) that harbors a unique nucleotide sequence serving as the identifying "barcode". Using real time quantitative PCR (qPCR) for detection of the barcoded pDNA and quantitative reverse transcription PCR (RT-qPCR) for transcribed barcoded mRNA, we can quantify accumulation and transfection in tissues of interest. The barcode pDNA and primers were designed with sufficient sensitivity and specificity to evaluate multiple nanoparticle formulations per mouse, improving screening efficiency. Using this platform, we evaluated the biodistribution and transfection of 8 intravenously administered poly(beta-amino ester; PBAE) nanoparticle formulations, each with a PBAE polymer of differential structure. Significant levels of nanoparticle accumulation and gene transfection were observed mainly in organs involved in clearance, including spleen, liver, and kidneys. Interestingly, higher levels of transfection of select organs did not necessarily correlate with higher levels of tissue accumulation, highlighting the importance of directly measuring in vivo transfection efficiency as the key barcoded parameter in gene delivery vector optimization. To validate this method, nanoparticle formulations were used individually for luciferase pDNA delivery in vivo. The distribution of luciferase expression in tissues matched the transfection analysis by the barcode qPCR method, confirming that this platform can be used to accurately evaluate systemic gene delivery.
Collapse
Affiliation(s)
- Jayoung Kim
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Hannah J Vaughan
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Camila G Zamboni
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Joel C Sunshine
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Jordan J Green
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA; Departments of Neurosurgery, Oncology, and Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
21
|
Rezaei A, Hashemi E. A pseudohomogeneous nanocarrier based on carbon quantum dots decorated with arginine as an efficient gene delivery vehicle. Sci Rep 2021; 11:13790. [PMID: 34215792 PMCID: PMC8253742 DOI: 10.1038/s41598-021-93153-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 06/22/2021] [Indexed: 02/06/2023] Open
Abstract
A pseudohomogeneous carrier as an emerging term refers to subnanometric carbon-based vehicle with a high ability to interact with genetic materials to form stable carboplex and successfully transfer them into the cell which will result in inhibiting or expressing of therapeutic genes. Chitosan is a non-toxic polyaminosaccharide used as a precursor in the presence of citric acid to produce carbon quantum dots (CQDs), which decorated with arginine as a surface passivation agent with high amine density in hydrothermal methodology. The Arginine-CQDs are comprehensively characterized by Fourier-transform infrared spectroscopy (FT-IR), Ultraviolet-visible spectroscopy (UV-vis), Atomic force microscopy (AFM), field emission scanning electron microscope (FE-SEM), Energy-dispersive X-ray (EDX) mapping, fluorescence, High-resolution transmission electron microscopy (HR-TEM), zeta potential and X-ray powder diffraction (XRD). In this regard, for the first time, carboplex are formed by electrostatic conjugating of Arginine-CQDs with DNA to protect it from enzymatic degradation. Moreover, the carboplex, like the chitosan precursor, has not shown toxicity against AGS cell line. Interestingly, the Arginine-CQDs have exhibited an excellent ability to overcome cell barriers to deliver into cells compared to chitosan at the same weight ratio. The Arginine-CQDs/pEGFP (W/W) nanocomplex, not only lead to transfection with a relatively higher efficiency than PEI polymer, which is the "golden standard", but carboplex also demonstrates no significant toxicity. Indeed, the EGFP expression level has reached to 2.4 ± 0.2 via Arginine-CQDs carboplex at W/W 50 weight ratio. To the best of our knowledge, this is the first report includes chitosan-based CQDs functionalized by arginine which is applied to serve as a pseudohomogeneous vehicle for gene transfection.
Collapse
Affiliation(s)
- Aram Rezaei
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Ehsan Hashemi
- Department of Animal Biotechnology, National Institute of Genetic Engineering and Biotechnology, P.O. Box: 14965-16, Tehran, Iran
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
22
|
Puchkov PA, Maslov MA. Lipophilic Polyamines as Promising Components of Liposomal Gene Delivery Systems. Pharmaceutics 2021; 13:920. [PMID: 34205825 PMCID: PMC8234823 DOI: 10.3390/pharmaceutics13060920] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/13/2021] [Accepted: 06/17/2021] [Indexed: 12/28/2022] Open
Abstract
Gene therapy requires an effective and safe delivery vehicle for nucleic acids. In the case of non-viral vehicles, including cationic liposomes, the structure of compounds composing them determines the efficiency a lot. Currently, cationic amphiphiles are the most frequently used compounds in liposomal formulations. In their structure, which is a combination of hydrophobic and cationic domains and includes spacer groups, each component contributes to the resulting delivery efficiency. This review focuses on polycationic and disulfide amphiphiles as prospective cationic amphiphiles for gene therapy and includes a discussion of the mutual influence of structural components.
Collapse
Affiliation(s)
| | - Michael A. Maslov
- Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, Vernadsky Ave. 86, 119571 Moscow, Russia;
| |
Collapse
|
23
|
Pressnall MM, Huang A, Groer CE, Huayamares SG, Laird Forrest M, Berkland CJ. Glatiramer acetate enhances tumor retention and innate activation of immunostimulants. Int J Pharm 2021; 605:120812. [PMID: 34144136 DOI: 10.1016/j.ijpharm.2021.120812] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 06/11/2021] [Accepted: 06/13/2021] [Indexed: 10/21/2022]
Abstract
Cancer immunotherapy aims to stimulate immune cells to recognize and attack tumor tissue. The immunostimulatory polyanions polyI:C and CpG induce potent pro-inflammatory immune responses as TLR3 and TLR9 agonists, respectively. Clinical trials of TLR agonists, however, have been fraught with immune-related adverse events, even when injecting intratumorally in an effort to minimize systemic exposure. We identified Glatiramer Acetate (GA), a positively-charged polypeptide approved for multiple sclerosis, as a delivery agent capable of complexing with polyI:C or CpG and reducing the mobility of these actives. Small nanoparticles termed polyplexes form when mixing positively-charged GA and negatively-charged immunostimulant (polyI:C or CpG). The ratio of GA to immunostimulant directly affected the potency of TLR activation and the mobility of these actives in simulated tumor tissue. Polyplexes of GA and CpG were injected intratumorally in a tumor model of head and neck cancer (HNC) and significantly mitigated tumor growth as compared to the vehicle controls. Intratumoral injections of CpG showed the slowest tumor growth but exhibited dramatically higher systemic proinflammatory cytokine levels compared to polyplexes of GA with CpG. Sequencing of RNA from resected tumors revealed a similar pattern of upregulated proinflammatory cytokines for CpG and polyplexes, a finding supported by histological tumor staining showing similar infiltration of immune cells induced by these treatments. Intratumoral administration of polyplexes of GA with immunostimulant represents a translational approach to enhance local immune responses while mitigating systemic immune-related adverse events.
Collapse
Affiliation(s)
- Melissa M Pressnall
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
| | - Aric Huang
- Bioengineering Program, University of Kansas, Lawrence, KS, United States
| | - Chad E Groer
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States; HylaPharm, LLC, Lawrence, KS, United States
| | | | - M Laird Forrest
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States; HylaPharm, LLC, Lawrence, KS, United States
| | - Cory J Berkland
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States; Bioengineering Program, University of Kansas, Lawrence, KS, United States; Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, KS, United States.
| |
Collapse
|
24
|
Batabyal S, Kim S, Wright W, Mohanty S. Laser-assisted targeted gene delivery to degenerated retina improves retinal function. JOURNAL OF BIOPHOTONICS 2021; 14:e202000234. [PMID: 33026157 DOI: 10.1002/jbio.202000234] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 09/27/2020] [Accepted: 09/28/2020] [Indexed: 06/11/2023]
Abstract
Delivery of therapeutic genes into retina is proving to reverse degeneration and restore vision, however, viral vector-based gene delivery is prone to immunorejection, inflammatory/immune-response and nontargeted. Here, we report nonviral gene delivery and expression of opsin encoding genes in mouse retina in-vitro and in-vivo by use of pulsed femtosecond laser microbeam. In-vitro patch-clamp recording of the opsin-sensitized retinal cells and visually evoked in-vivo electrical recording from laser-transfected eye of mouse with degenerated retina showed functional response. The ultrafast laser-based naked gene delivery showed minimal damage and reliable expression of therapeutic opsin in cell membrane of the selected cells and in targeted retinal region. Laser-based "naked DNA gene therapy" in a spatially targeted manner will pave the way for treatment of inherited retinal diseases.
Collapse
|
25
|
Sousa CFV, Fernandez-Megia E, Borges J, Mano JF. Supramolecular dendrimer-containing layer-by-layer nanoassemblies for bioapplications: current status and future prospects. Polym Chem 2021. [DOI: 10.1039/d1py00988e] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
This review provides a comprehensive and critical overview of the supramolecular dendrimer-containing multifunctional layer-by-layer nanoassemblies driven by a multitude of intermolecular interactions for biological and biomedical applications.
Collapse
Affiliation(s)
- Cristiana F. V. Sousa
- CICECO–Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Eduardo Fernandez-Megia
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - João Borges
- CICECO–Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - João F. Mano
- CICECO–Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| |
Collapse
|
26
|
Amina SJ, Guo B. A Review on the Synthesis and Functionalization of Gold Nanoparticles as a Drug Delivery Vehicle. Int J Nanomedicine 2020; 15:9823-9857. [PMID: 33324054 PMCID: PMC7732174 DOI: 10.2147/ijn.s279094] [Citation(s) in RCA: 196] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 10/23/2020] [Indexed: 12/16/2022] Open
Abstract
Metal nanoparticles are being extensively used in biomedical fields due to their small size-to-volume ratio and extensive thermal stability. Gold nanoparticles (AuNPs) are an obvious choice for biomedical applications due to their amenability of synthesis, stabilization, and functionalization, low toxicity, and ease of detection. In the past few decades, various chemical methods have been used for the synthesis of AuNPs, but recently, newer environment friendly green approaches for the synthesis of AuNPs have gained attention. AuNPs can be conjugated with a number of functionalizing moieties including ligands, therapeutic agents, DNA, amino acids, proteins, peptides, and oligonucleotides. Recently, studies have shown that gold nanoparticles not only infiltrate the blood vessels to reach the site of tumor but also enter inside the organelles, suggesting that they can be employed as effective drug carriers. Moreover, after reaching their target site, gold nanoparticles can release their payload upon an external or internal stimulus. This review focuses on recent advances in various methods of synthesis of AuNPs. In addition, strategies of functionalization and mechanisms of application of AuNPs in drug and bio-macromolecule delivery and release of payloads at target site are comprehensively discussed.
Collapse
Affiliation(s)
- Sundus Jabeen Amina
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Bin Guo
- Department of Pharmacological & Pharmaceutical Sciences, University of Houston, Houston, TX77204, USA
| |
Collapse
|
27
|
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: 26] [Impact Index Per Article: 6.5] [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
|
28
|
Li H, Yang Z, Fu L, Yuan Z, Gao C, Sui X, Liu S, Peng J, Dai Y, Guo Q. Advanced Polymer-Based Drug Delivery Strategies for Meniscal Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2020; 27:266-293. [PMID: 32988289 DOI: 10.1089/ten.teb.2020.0156] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The meniscus plays a critical role in maintaining knee joint homeostasis. Injuries to the meniscus, especially considering the limited self-healing capacity of the avascular region, continue to be a challenge and are often treated by (partial) meniscectomy, which has been identified to cause osteoarthritis. Currently, meniscus tissue engineering focuses on providing extracellular matrix (ECM)-mimicking scaffolds to direct the inherent meniscal regeneration process, and it has been found that various stimuli are essential. Numerous bioactive factors present benefits in regulating cell fate, tissue development, and healing, but lack an optimal delivery system. More recently, bioengineers have developed various polymer-based drug delivery systems (PDDSs), which are beneficial in terms of the favorable properties of polymers as well as novel delivery strategies. Engineered PDDSs aim to provide not only an ECM-mimicking microenvironment but also the controlled release of bioactive factors with release profiles tailored according to the biological concerns and properties of the factors. In this review, both different polymers and bioactive factors involved in meniscal regeneration are discussed, as well as potential candidate systems, with examples of recent progress. This article aims to summarize drug delivery strategies in meniscal regeneration, with a focus on novel delivery strategies rather than on specific delivery carriers. The current challenges and future prospects for the structural and functional regeneration of the meniscus are also discussed. Impact statement Meniscal injury remains a clinical Gordian knot owing to the limited healing potential of the region, restricted surgical approaches, and risk of inducing osteoarthritis. Existing tissue engineering scaffolds that provide mechanical support and a favorable microenvironment also lack biological cues. Advanced polymer-based delivery strategies consisting of polymers incorporating bioactive factors have emerged as a promising direction. This article primarily reviews the types and applications of biopolymers and bioactive factors in meniscal regeneration. Importantly, various carrier systems and drug delivery strategies are discussed with the hope of inspiring further advancements in this field.
Collapse
Affiliation(s)
- Hao Li
- School of Medicine, Nankai University, Tianjin, China.,Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Laboratory of Musculoskeletal Trauma & War Injuries PLA; Beijing, China
| | - Zhen Yang
- School of Medicine, Nankai University, Tianjin, China.,Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Laboratory of Musculoskeletal Trauma & War Injuries PLA; Beijing, China
| | - Liwei Fu
- School of Medicine, Nankai University, Tianjin, China.,Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Laboratory of Musculoskeletal Trauma & War Injuries PLA; Beijing, China
| | - Zhiguo Yuan
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Laboratory of Musculoskeletal Trauma & War Injuries PLA; Beijing, China.,Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Cangjian Gao
- School of Medicine, Nankai University, Tianjin, China.,Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Laboratory of Musculoskeletal Trauma & War Injuries PLA; Beijing, China
| | - Xiang Sui
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Laboratory of Musculoskeletal Trauma & War Injuries PLA; Beijing, China
| | - Shuyun Liu
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Laboratory of Musculoskeletal Trauma & War Injuries PLA; Beijing, China
| | - Jiang Peng
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Laboratory of Musculoskeletal Trauma & War Injuries PLA; Beijing, China
| | - Yongjing Dai
- Department of Orthopedic, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Quanyi Guo
- School of Medicine, Nankai University, Tianjin, China.,Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Laboratory of Musculoskeletal Trauma & War Injuries PLA; Beijing, China
| |
Collapse
|
29
|
Tsafa E, Bentayebi K, Topanurak S, Yata T, Przystal J, Fongmoon D, Hajji N, Waramit S, Suwan K, Hajitou A. Doxorubicin Improves Cancer Cell Targeting by Filamentous Phage Gene Delivery Vectors. Int J Mol Sci 2020; 21:E7867. [PMID: 33114050 PMCID: PMC7660303 DOI: 10.3390/ijms21217867] [Citation(s) in RCA: 13] [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: 07/28/2020] [Revised: 10/18/2020] [Accepted: 10/19/2020] [Indexed: 12/14/2022] Open
Abstract
Merging targeted systemic gene delivery and systemic chemotherapy against cancer, chemovirotherapy, has the potential to improve chemotherapy and gene therapy treatments and overcome cancer resistance. We introduced a bacteriophage (phage) vector, named human adeno-associated virus (AAV)/phage or AAVP, for the systemic targeting of therapeutic genes to cancer. The vector was designed as a hybrid between a recombinant adeno-associated virus genome (rAAV) and a filamentous phage capsid. To achieve tumor targeting, we displayed on the phage capsid the double-cyclic CDCRGDCFC (RGD4C) ligand that binds the alpha-V/beta-3 (αvβ3) integrin receptor. Here, we investigated a combination of doxorubicin chemotherapeutic drug and targeted gene delivery by the RGD4C/AAVP vector. Firstly, we showed that doxorubicin boosts transgene expression from the RGD4C/AAVP in two-dimensional (2D) cell cultures and three-dimensional (3D) tumor spheres established from human and murine cancer cells, while preserving selective gene delivery by RGD4C/AAVP. Next, we confirmed that doxorubicin does not increase vector attachment to cancer cells nor vector cell entry. In contrast, doxorubicin may alter the intracellular trafficking of the vector by facilitating nuclear accumulation of the RGD4C/AAVP genome through destabilization of the nuclear membrane. Finally, a combination of doxorubicin and RGD4C/AAVP-targeted suicide gene therapy exerts a synergistic effect to destroy human and murine tumor cells in 2D and 3D tumor sphere settings.
Collapse
Affiliation(s)
- Effrosyni Tsafa
- Phage Therapy Group, Department of Brain Sciences, Imperial College London, London W12 0NN, UK; (E.T.); (K.B.); (T.Y.); (J.P.); (S.W.)
| | - Kaoutar Bentayebi
- Phage Therapy Group, Department of Brain Sciences, Imperial College London, London W12 0NN, UK; (E.T.); (K.B.); (T.Y.); (J.P.); (S.W.)
| | - Supachai Topanurak
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand;
| | - Teerapong Yata
- Phage Therapy Group, Department of Brain Sciences, Imperial College London, London W12 0NN, UK; (E.T.); (K.B.); (T.Y.); (J.P.); (S.W.)
| | - Justyna Przystal
- Phage Therapy Group, Department of Brain Sciences, Imperial College London, London W12 0NN, UK; (E.T.); (K.B.); (T.Y.); (J.P.); (S.W.)
| | - Duriya Fongmoon
- Department of Medical Services, Lampang Cancer Hospital, Ministry of Public Health, Lampang 52000, Thailand;
| | - Nabil Hajji
- John Fulcher Neuro-Oncology Laboratory, Department of Brain Sciences, Imperial College London, London W12 0NN, UK;
| | - Sajee Waramit
- Phage Therapy Group, Department of Brain Sciences, Imperial College London, London W12 0NN, UK; (E.T.); (K.B.); (T.Y.); (J.P.); (S.W.)
| | - Keittisak Suwan
- Phage Therapy Group, Department of Brain Sciences, Imperial College London, London W12 0NN, UK; (E.T.); (K.B.); (T.Y.); (J.P.); (S.W.)
| | - Amin Hajitou
- Phage Therapy Group, Department of Brain Sciences, Imperial College London, London W12 0NN, UK; (E.T.); (K.B.); (T.Y.); (J.P.); (S.W.)
| |
Collapse
|
30
|
Ma LL, Tang Q, Liu MX, Liu XY, Liu JY, Lu ZL, Gao YG, Wang R. [12]aneN 3-Based Gemini-Type Amphiphiles with Two-Photon Absorption Properties for Enhanced Nonviral Gene Delivery and Bioimaging. ACS APPLIED MATERIALS & INTERFACES 2020; 12:40094-40107. [PMID: 32805811 DOI: 10.1021/acsami.0c10718] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Although a plethora of nonviral gene vectors have been developed for potential gene therapy, imageable gemini surfactants with stimuli-responsiveness and high transfection efficiency are still scarce for gene delivery. Herein, three gemini amphiphiles (DEDPP-4/8/12) consisting of an aggregation-induced emission (AIE) central fluorophore: 5,6-diphenylpyrazine-2,3-diester (DEDPP), decorated with triazole-[12]aneN3 as the hydrophilic moiety and alkyl chains of various lengths as the hydrophobic moiety, were designed and synthesized for trackable gene delivery via optical imaging. All three amphiphiles exhibited ultralow critical micelle concentrations (CMCs) (up to 3.40 × 10-6 M), prominent two-photon absorption properties, and solvatochromic fluorescence. Gel electrophoresis assays demonstrated that the migration of plasmid DNA was completely retarded after condensation with these gemini amphiphiles at low concentrations (up to 10 μM). In addition, the ester bond in these amphiphiles may facilitate vector degradation and DNA release, in response to esterase and the acidic environment inside cells. Upon self-assembly with DOPE to form liposomes, DEDPP-8/DOPE achieved the best transfection efficiency in four cell lines, and the transfection efficiency of DEDPP-8/DOPE in HeLa cell lines was 23.5-fold higher than that of Lipo2000, which is unusually high for small organic molecule-based nonviral vectors. Furthermore, excellent transfection efficiency of DEDPP-8/DOPE was obtained in the presence of serum, and the red fluorescence protein (RFP) gene was successfully transfected in zebrafish embryos. Both one- and two-photon fluorescence imaging clearly demonstrated the delivery process of plasmid DNA. This study demonstrated that gemini-type amphiphiles composed of a two-photon fluorophore core conjugated with triazole-[12]aneN3 via an ester bond afforded an unprecedentedly high transfection efficiency with excellent biocompatibility, which may provide new insights for the design and development of multifunctional nonviral gene vectors for imageable gene delivery.
Collapse
Affiliation(s)
- Le-Le Ma
- Key Laboratory of Radiopharmaceutics, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Quan Tang
- Key Laboratory of Radiopharmaceutics, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Ming-Xuan Liu
- Key Laboratory of Radiopharmaceutics, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Xu-Ying Liu
- Key Laboratory of Radiopharmaceutics, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Jin-Yu Liu
- Key Laboratory of Radiopharmaceutics, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Zhong-Lin Lu
- Key Laboratory of Radiopharmaceutics, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Yong-Guang Gao
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Ruibing Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR 9990078, China
| |
Collapse
|
31
|
Immunostimulant Complexed With Polylysine Limits Transport and Maintains Immune Cell Activation. J Pharm Sci 2020; 109:2836-2846. [DOI: 10.1016/j.xphs.2020.06.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/08/2020] [Accepted: 06/08/2020] [Indexed: 12/30/2022]
|
32
|
Est-Witte SE, Farris AL, Tzeng SY, Hutton DL, Gong DH, Calabresi KG, Grayson WL, Green JJ. Non-viral gene delivery of HIF-1α promotes angiogenesis in human adipose-derived stem cells. Acta Biomater 2020; 113:279-288. [PMID: 32623098 PMCID: PMC8035702 DOI: 10.1016/j.actbio.2020.06.042] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 06/05/2020] [Accepted: 06/26/2020] [Indexed: 02/07/2023]
Abstract
Stable and mature vascular formation is a current challenge in engineering functional tissues. Transient, non-viral gene delivery presents a unique platform for delivering genetic information to cells for tissue engineering purposes and to restore blood flow to ischemic tissue. The formation of new blood vessels can be induced by upregulation of hypoxia-inducible factor-1α (HIF-1), among other factors. We hypothesized that biodegradable polymers could be used to efficiently deliver the HIF-1α gene to human adipose-derived stromal/stem cells (hASCs) and that this treatment could recruit an existing endogenous endothelial cell population to induce angiogenesis in a 3D cell construct in vitro. In this study, end-modified poly(β-amino ester) (PBAE) nanocomplexes were first optimized for transfection of hASCs and a new biodegradable polymer with increased hydrophobicity and secondary amine structures, N'-(3-aminopropyl)-N,N-dimethylpropane-1,3-diamine end-modified poly(1,4-butanediol diacrylate-co-4-amino-1-butanol), was found to be most effective. Optimal PBAE nanocomplexes had a hydrodynamic diameter of approximately 140 nm and had a zeta potential of 30 mV. The PBAE polymer self-assembled with HIF-1α plasmid DNA and treatment of hASCs with these nanocomplexes induced 3D vascularization. Cells transfected with this polymer-DNA complex were found to have 106-fold upregulation HIF-1α expression, an approximately 2-fold increase in secreted VEGF, and caused the formation of vessel tubules compared to an untransfected control. These gene therapy biomaterials may be useful for regenerative medicine. STATEMENT OF SIGNIFICANCE: Not only is the formation of stable vasculature a challenge for engineering human tissues in vitro, but it is also of valuable interest to clinical applications such as peripheral artery disease. Previous studies using HIF-1α to induce vascular formation have been limited by the necessity of hypoxic chambers. It would be advantageous to simulate endogenous responses to hypoxia without the need for physical hypoxia. In this study, 3D vascular formation was shown to be inducible through non-viral gene delivery of HIF-1α with new polymeric nanocomplexes. A biodegradable polymer N'-(3-aminopropyl)-N,N-dimethylpropane-1,3-diamine end-modified poly(1,4-butanediol diacrylate-co-4-amino-1-butanol) demonstrates improved transfection of human adipose-derived stem cells. This nanobiotechnology could be a promising strategy for the creation of vasculature for tissue engineering and clinical applications.
Collapse
Affiliation(s)
- Savannah E Est-Witte
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| | - Ashley L Farris
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| | - Stephany Y Tzeng
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| | - Daphne L Hutton
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| | - Dennis H Gong
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| | - Kaitlyn G Calabresi
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| | - Warren L Grayson
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21231, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21231, USA.
| | - Jordan J Green
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21231, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21231, USA; Department of Oncology and Bloomberg~Kimmel Immunotherapy Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Ophthalmology Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| |
Collapse
|
33
|
Zohrabi T, Hosseinkhani S. Ternary Nanocomplexes of Metallic Nanoclusters and Recombinant Peptides for Fluorescence Imaging and Enhanced Gene Delivery. Mol Biotechnol 2020; 62:495-507. [PMID: 32808172 DOI: 10.1007/s12033-020-00260-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2020] [Indexed: 01/03/2023]
Abstract
The efficient carrier design for transferring therapeutic genes into target cells as well as tracking the delivered agents has attracted lots of attention in the field of DNA-based therapeutics. Here, we demonstrate this concept by a fast and facilitated method using BSA gold nanocluster (BSA AuNcs) conjugated with chimeric peptide with ability of DNA binding/packaging, endosome disruption and cell nuclear localization. An extensive characterization of photoluminescence properties, electrophoresis mobility and size distribution of the nanocarrier demonstrating the stable complexes composed of plasmid DNA, chimeric peptide and BSA AuNcs were successfully formed through electrostatic interactions. In the hybrid complexes, chimeric peptide could effectively decrease the cytotoxicity of AuNcs as well as enhance internalization of plasmid harboring firefly luciferase gene into HEK 293 T. The designed nanocarrier could be a promising vector in gene delivery systems for improved theranostics applications.
Collapse
Affiliation(s)
- Tayebeh Zohrabi
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Saman Hosseinkhani
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
34
|
Haladjova E, Smolíček M, Ugrinova I, Momekova D, Shestakova P, Kroneková Z, Kronek J, Rangelov S. DNA delivery systems based on copolymers of poly (2‐methyl‐2‐oxazoline) and polyethyleneimine: Effect of polyoxazoline moieties on the endo‐lysosomal escape. J Appl Polym Sci 2020. [DOI: 10.1002/app.49400] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Emi Haladjova
- Institute of Polymers, Bulgarian Academy of Sciences Sofia Bulgaria
| | - Maroš Smolíček
- Department for Biomaterials ResearchPolymer Institute, Slovak Academy of Sciences Bratislava Slovakia
- Department of Inorganic Chemistry, Faculty of Natural SciencesComenius University Mlynská dolina Bratislava Slovakia
| | - Iva Ugrinova
- Institute of Molecular Biology, Bulgarian Academy of Sciences Sofia Bulgaria
| | | | - Pavletta Shestakova
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences Sofia Bulgaria
| | - Zuzana Kroneková
- Department for Biomaterials ResearchPolymer Institute, Slovak Academy of Sciences Bratislava Slovakia
| | - Juraj Kronek
- Department for Biomaterials ResearchPolymer Institute, Slovak Academy of Sciences Bratislava Slovakia
| | | |
Collapse
|
35
|
Lavanya K, Chandran SV, Balagangadharan K, Selvamurugan N. Temperature- and pH-responsive chitosan-based injectable hydrogels for bone tissue engineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 111:110862. [DOI: 10.1016/j.msec.2020.110862] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 03/08/2020] [Accepted: 03/16/2020] [Indexed: 01/05/2023]
|
36
|
Hydrophilic/hydrophobic modifications of a PnBA-b-PDMAEA copolymer and complexation behaviour with short DNA. Eur Polym J 2020. [DOI: 10.1016/j.eurpolymj.2020.109636] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
37
|
Zhang H, Rombouts K, Raes L, Xiong R, De Smedt SC, Braeckmans K, Remaut K. Fluorescence-Based Quantification of Messenger RNA and Plasmid DNA Decay Kinetics in Extracellular Biological Fluids and Cell Extracts. ACTA ACUST UNITED AC 2020; 4:e2000057. [PMID: 32402121 DOI: 10.1002/adbi.202000057] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 02/26/2020] [Indexed: 12/22/2022]
Abstract
Extracellular and intracellular degradation of nucleic acids remains an issue in non-viral gene therapy. Understanding biodegradation is critical for the rational design of gene therapeutics in order to maintain stability and functionality at the target site. However, there are only limited methods available that allow determining the stability of genetic materials in biological environments. In this context, the decay kinetics of fluorescently labeled plasmid DNA (pDNA) and messenger RNA (mRNA) in undiluted biological samples (i.e., human serum, human ascites, bovine vitreous) and cell extracts is studied using fluorescence correlation spectroscopy (FCS) and single particle tracking (SPT). It is demonstrated that FCS is suitable to follow mRNA degradation, while SPT is better suited to investigate pDNA integrity. The half-life of mRNA and pDNA is ≈1-2 min and 1-4 h in biological samples, respectively. The resistance against biodegradation drastically improves by complexation with lipid-based carriers. Taken together, FCS and SPT are able to quantify the integrity of mRNA and pDNA, respectively, as a function of time, both in the extracellular biological fluids and cell extracts. This in turn allows to focus on the important but less understood issue of nucleic acids degradation in more detail and to rationally optimize gene delivery system as therapeutics.
Collapse
Affiliation(s)
- Heyang Zhang
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, 9000, Belgium
| | - Koen Rombouts
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, 9000, Belgium
| | - Laurens Raes
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, 9000, Belgium
| | - Ranhua Xiong
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, 9000, Belgium
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, 9000, Belgium
| | - Kevin Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, 9000, Belgium
| | - Katrien Remaut
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, 9000, Belgium
| |
Collapse
|
38
|
Husteden C, Doberenz F, Goergen N, Pinnapireddy SR, Janich C, Langner A, Syrowatka F, Repanas A, Erdmann F, Jedelská J, Bakowsky U, Groth T, Wölk C. Contact-Triggered Lipofection from Multilayer Films Designed as Surfaces for in Situ Transfection Strategies in Tissue Engineering. ACS APPLIED MATERIALS & INTERFACES 2020; 12:8963-8977. [PMID: 32003972 DOI: 10.1021/acsami.9b18968] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Biomaterials, which release active compounds after implantation, are an essential tool for targeted regenerative medicine. In this study, thin multilayer films loaded with lipid/DNA complexes (lipoplexes) were designed as surface coatings for in situ transfection applicable in tissue engineering and regenerative medicine. The film production and embedding of lipoplexes were based on the layer-by-layer (LbL) deposition technique. Hyaluronic acid (HA) and chitosan (CHI) were used as the polyelectrolyte components. The embedded plasmid DNA was complexed using a new designed cationic lipid formulation, namely, OH4/DOPE 1/1, the advantageous characteristics of which have been proven already. Three different methods were tested regarding its efficiency of lipid and DNA deposition. Therefore, several surface specific analytics were used to characterize the LbL formation, the lipid DNA embedding, and the surface characteristics of the multilayer films, such as fluorescence microscopy, surface plasmon resonance spectroscopy, ellipsometry, zeta potential measurements, atomic force microscopy, and scanning electron microscopy. Interaction studies were conducted for optimized lipoplex-loaded polyelectrolyte multilayers (PEMs) that showed an efficient attachment of C2C12 cells on the surface. Furthermore, no acute toxic effects were found in cell culture studies, demonstrating biocompatibility. Cell culture experiments with C2C12 cells, a cell line which is hard to transfect, demonstrated efficient transfection of the reporter gene encoding for green fluorescent protein. In vivo experiments using the chicken embryo chorion allantois membrane animal replacement model showed efficient gene-transferring rates in living complex tissues, although the DNA-loaded films were stored over 6 days under wet and dried conditions. Based on these findings, it can be concluded that OH4/DOPE 1/1 lipoplex-loaded PEMs composed of HA and CHI can be an efficient tool for in situ transfection in regenerative medicine.
Collapse
Affiliation(s)
- Catharina Husteden
- Institute of Pharmacy, Department of Medicinal Chemistry , Martin Luther University Halle-Wittenberg , Wolfgang-Langenbeck-Str. 4 , 06120 Halle (Saale) , Germany
| | - Falko Doberenz
- Institute of Pharmacy, Department Biomedical Materials , Martin Luther University Halle-Wittenberg , Heinrich-Damerow-Str. 4 , 06120 Halle (Saale) , Germany
| | - Nathalie Goergen
- Department of Pharmaceutics and Biopharmaceutics , University of Marburg , Robert-Koch-Str. 4 , 35037 Marburg , Germany
| | - Shashank Reddy Pinnapireddy
- Department of Pharmaceutics and Biopharmaceutics , University of Marburg , Robert-Koch-Str. 4 , 35037 Marburg , Germany
| | - Christopher Janich
- Institute of Pharmacy, Department of Medicinal Chemistry , Martin Luther University Halle-Wittenberg , Wolfgang-Langenbeck-Str. 4 , 06120 Halle (Saale) , Germany
| | - Andreas Langner
- Institute of Pharmacy, Department of Medicinal Chemistry , Martin Luther University Halle-Wittenberg , Wolfgang-Langenbeck-Str. 4 , 06120 Halle (Saale) , Germany
| | - Frank Syrowatka
- Interdisciplinary Center of Materials Science , Martin-Luther-University Halle-Wittenberg , Heinrich-Damerow-Str. 4 , 06120 Halle (Saale) , Germany
| | - Alexandros Repanas
- Institute of Pharmacy, Department Biomedical Materials , Martin Luther University Halle-Wittenberg , Heinrich-Damerow-Str. 4 , 06120 Halle (Saale) , Germany
| | - Frank Erdmann
- Institute of Pharmacy, Department of Pharmacology , Martin Luther University Halle-Wittenberg , Wolfgang-Langenbeck-Str. 4 , 06120 Halle (Saale) , Germany
| | - Jarmila Jedelská
- Department of Pharmaceutics and Biopharmaceutics , University of Marburg , Robert-Koch-Str. 4 , 35037 Marburg , Germany
| | - Udo Bakowsky
- Department of Pharmaceutics and Biopharmaceutics , University of Marburg , Robert-Koch-Str. 4 , 35037 Marburg , Germany
| | - Thomas Groth
- Institute of Pharmacy, Department Biomedical Materials , Martin Luther University Halle-Wittenberg , Heinrich-Damerow-Str. 4 , 06120 Halle (Saale) , Germany
- Interdisciplinary Center of Materials Science , Martin-Luther-University Halle-Wittenberg , Heinrich-Damerow-Str. 4 , 06120 Halle (Saale) , Germany
- Laboratory of Biomedical Nanotechnologies, Institute of Bionic Technologies and Engineering , I.M. Sechenov First Moscow State University , Trubetskaya Street 8 , 119991 Moscow , Russian Federation
| | - Christian Wölk
- Institute of Pharmacy, Department of Medicinal Chemistry , Martin Luther University Halle-Wittenberg , Wolfgang-Langenbeck-Str. 4 , 06120 Halle (Saale) , Germany
- Institute of Pharmacy, Pharmaceutical Technology, Faculty of Medicine , Leipzig University , 04317 Leipzig , Germany
| |
Collapse
|
39
|
Liu Y, Tronser T, Peravali R, Reischl M, Levkin PA. High‐Throughput Screening of Cell Transfection Enhancers Using Miniaturized Droplet Microarrays. ACTA ACUST UNITED AC 2020; 4:e1900257. [DOI: 10.1002/adbi.201900257] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/04/2019] [Indexed: 11/07/2022]
Affiliation(s)
- Yanxi Liu
- Institute of Toxicology and Genetics (ITG)Karlsruhe Institute of Technology (KIT) Hermann‐von Helmholtz‐Platz 1 Eggenstein‐Leopoldshafen 76344 Germany
| | - Tina Tronser
- Institute of Toxicology and Genetics (ITG)Karlsruhe Institute of Technology (KIT) Hermann‐von Helmholtz‐Platz 1 Eggenstein‐Leopoldshafen 76344 Germany
| | - Ravindra Peravali
- Institute of Toxicology and Genetics (ITG)Karlsruhe Institute of Technology (KIT) Hermann‐von Helmholtz‐Platz 1 Eggenstein‐Leopoldshafen 76344 Germany
| | - Markus Reischl
- Institute for Automation and Applied Informatics (IAI)Karlsruhe Institute of Technology (KIT) Hermann‐von Helmholtz‐Platz 1 Eggenstein‐Leopoldshafen 76344 Germany
| | - Pavel A. Levkin
- Institute of Toxicology and Genetics (ITG)Karlsruhe Institute of Technology (KIT) Hermann‐von Helmholtz‐Platz 1 Eggenstein‐Leopoldshafen 76344 Germany
- Institute of Organic ChemistryKarlsruhe Institute of Technology (KIT) Fritz‐Haber‐Weg 6 Karlsruhe 76131 Germany
| |
Collapse
|
40
|
Betker JL, Anchordoquy TJ. The Use of Lactose as an Alternative Coating for Nanoparticles. J Pharm Sci 2020; 109:1573-1580. [PMID: 32004536 DOI: 10.1016/j.xphs.2020.01.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 12/16/2019] [Accepted: 01/23/2020] [Indexed: 12/22/2022]
Abstract
Nanoparticle-mediated drug delivery has long utilized PEGylation as a mechanism for reducing uptake by the reticuloendothelial system and extending circulation lifetimes. However, studies over the past 2 decades have established that immune responses to PEG can promote clearance on repeat injection and elicit life-threatening anaphylactic reactions in some patients. As a potential alternative to PEGylation, we explored the ability of utilizing lactose, a naturally occurring sugar that is common on the surface of blood cells, as a coating for lipoplexes. Our data indicate that lactose imparts similar effects as PEG in terms of reducing leukocyte uptake, extending circulation half-life, and enhancing delivery to the tumor and other organs. In addition, measurements of blood cytokine levels after repeat injection indicate that reduced levels of inflammatory cytokines (IL-6, IFN-γ, TNFα) are elicited in response to lipoplexes coated with lactose as compared to PEG. These data indicate that a lactose coating on lipoplexes results in slightly improved tumor accumulation as compared to PEGylated formulations while eliciting a reduced innate immune response.
Collapse
Affiliation(s)
- Jamie L Betker
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Thomas J Anchordoquy
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045.
| |
Collapse
|
41
|
Abstract
Efficient intracellular delivery of small-interfering ribonucleic acid (siRNA) to the target organ or tissues in the body is assumed as the main hurdle for a widespread use of siRNAs in the clinics. Solid lipid-based nanoparticles (SLNs) and derivatives can potentially fit this purpose by enabling to overcome the extracellular and intracellular physiological barriers affecting the delivery. For that, rational formulations and rational process designs are needed. This chapter addresses a comprehensive description and critical appraisal of the main production methods of this particular type of lipid nanoparticles and the leading strategies to prompt a targeted delivery of siRNA.
Collapse
Affiliation(s)
- Andreia Jorge
- Department of Chemistry, Coimbra Chemistry Centre, University of Coimbra, Coimbra, Portugal.
| | - Alberto Pais
- Department of Chemistry, Coimbra Chemistry Centre, University of Coimbra, Coimbra, Portugal
| | - Carla Vitorino
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.
- Center for Neurosciences and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.
- Department of Chemistry, Coimbra Chemistry Centre, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
42
|
Ye M, Wang Y, Zhao Y, Xie R, Yodsanit N, Johnston K, Gong S. Double-Network Nanogel as a Nonviral Vector for DNA Delivery. ACS APPLIED MATERIALS & INTERFACES 2019; 11:42865-42872. [PMID: 31696697 DOI: 10.1021/acsami.9b12492] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
A double-network nanogel, composed of a silane-cross-linked polyethylenimine (PEI) network (i.e., PEI-S) and a pH-responsive poly(2-(hexamethyleneimino) ethyl methacrylate) (PC7A) polymer, was developed for efficient DNA transfection. The chemical cross-linking and hydrophobic interactions in the two networks led to improved stability outside the cell and also pH-triggered intracellular release of DNA. The nanogel with an optimal PEI-S and PC7A weight ratio of 1.3:1 exhibited significantly higher transfection efficiency than Lipofectamine 2000 in multiple cell lines. The nanogel also possessed a small size with a hydrodynamic diameter of 55 nm, low cytotoxicity, and superior stability in serum-containing media. Moreover, besides the PEI-based gene delivery system, we have also demonstrated that addition of the PC7A polymer to several types of cationic polymers commonly used for gene delivery also led to significant transfection enhancement of the resulting nanoparticles, suggesting that the PC7A polymer may be a universal additive that can benefit versatile cationic polymer-based gene delivery systems.
Collapse
|
43
|
Zhong HH, Wang HY, Li J, Huang YZ. TRAIL-based gene delivery and therapeutic strategies. Acta Pharmacol Sin 2019; 40:1373-1385. [PMID: 31444476 PMCID: PMC6889127 DOI: 10.1038/s41401-019-0287-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 07/04/2019] [Indexed: 12/11/2022] Open
Abstract
TRAIL (tumor necrosis factor-related apoptosis-inducing ligand), also known as APO2L, belongs to the tumor necrosis factor family. By binding to the death receptor 4 (DR4) or DR5, TRAIL induces apoptosis of tumor cells without causing side toxicity in normal tissues. In recent years TRAIL-based therapy has attracted great attention for its promise of serving as a cancer drug candidate. However, the treatment efficacy of TRAIL protein was under expectation in the clinical trials because of the short half-life and the resistance of cancer cells. TRAIL gene transfection can produce a "bystander effect" of tumor cell killing and provide a potential solution to TRAIL-based cancer therapy. In this review we focus on TRAIL gene therapy and various design strategies of TRAIL DNA delivery including non-viral vectors and cell-based TRAIL therapy. In order to sensitize the tumor cells to TRAIL-induced apoptosis, combination therapy of TRAIL DNA with other drugs by the codelivery methods for yielding a synergistic antitumor efficacy is summarized. The opportunities and challenges of TRAIL-based gene delivery and therapy are discussed.
Collapse
Affiliation(s)
- Hui-Hai Zhong
- Shanghai University College of Sciences, Shanghai, 200444, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Hui-Yuan Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Jian Li
- Shanghai University College of Sciences, Shanghai, 200444, China
| | - Yong-Zhuo Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| |
Collapse
|
44
|
Hong SJ, Ahn MH, Sangshetti J, Arote RB. Sugar alcohol-based polymeric gene carriers: Synthesis, properties and gene therapy applications. Acta Biomater 2019; 97:105-115. [PMID: 31326667 DOI: 10.1016/j.actbio.2019.07.029] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 07/04/2019] [Accepted: 07/16/2019] [Indexed: 02/07/2023]
Abstract
Advances in the field of nanomedicine have led to the development of various gene carriers with desirable cellular responses. However, unfavorable stability and physicochemical properties have hindered their applications in vivo. Therefore, multifunctional, smart nanocarriers with unique properties to overcome such drawbacks are needed. Among them, sugar alcohol-based nanoparticle with abundant surface chemistry, numerous hydroxyl groups, acceptable biocompatibility and biodegradable property are considered as the recent additions to the growing list of non-viral vectors. In this review, we present some of the major advances in our laboratory in developing sugar-based polymers as non-viral gene delivery vectors to treat various diseases. We also discuss some of the open questions in this field. STATEMENT OF SIGNIFICANCE: Recently, the development of sugar alcohol-based polymers conjugated with polyethylenimine (PEI) has attracted tremendous interest as gene delivery vectors. First, the natural backbone of polymers with their numerous hydroxyl groups display a wide range of hyperosmotic properties and can thereby enhance the cellular uptake of genetic materials via receptor-mediated endocytosis. Second, conjugation of a PEI backbone with sugar alcohols via Michael addition contributes to buffering capacity and thereby the proton sponge effect. Last, sugar alcohol based gene delivery systems improves therapeutic efficacy both in vitro and in vivo.
Collapse
|
45
|
Lam P, Steinmetz NF. Delivery of siRNA therapeutics using cowpea chlorotic mottle virus-like particles. Biomater Sci 2019; 7:3138-3142. [PMID: 31257379 PMCID: PMC6705399 DOI: 10.1039/c9bm00785g] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
While highly promising in medicine, gene therapy requires delivery agents to protect and target nucleic acid therapeutics. We developed a plant viral siRNA delivery platform making use of self-assembling cowpea chlorotic mottle virus (CCMV). CCMV was loaded with siRNAs targeting GFP or FOXA1; to further enhance cell uptake and intracellular trafficking, resulting in more efficient gene knockdown, we appended CCMV with a cell penetrating peptide (CPP), specifically M-lycotoxin peptide L17E.
Collapse
Affiliation(s)
- Patricia Lam
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Nicole F Steinmetz
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA and Departments of NanoEngineering, Bioengineering, Radiology, Moores Cancer Center, University of California San Diego, La Jolla 92093, USA.
| |
Collapse
|
46
|
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
|
47
|
Etchenausia L, Villar-Alvarez E, Forcada J, Save M, Taboada P. Evaluation of cationic core-shell thermoresponsive poly(N-vinylcaprolactam)-based microgels as potential drug delivery nanocarriers. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 104:109871. [PMID: 31499979 DOI: 10.1016/j.msec.2019.109871] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 06/05/2019] [Accepted: 06/06/2019] [Indexed: 01/21/2023]
Abstract
The present work investigates the potentiality of poly(N-vinyl caprolactam) (PVCL)-based thermoresponsive microgels decorated with cationic polymer brushes as drug delivery carriers. The effect of physico-chemical features of the colloids on cell viability response have to be carefully investigated to establish the range of suitable hydrodynamic diameters, crosslinking densities, lengths and ratios of the cationic polyelectrolyte shell which allow their efficient and effective use for cargo loading, transport and delivery. The colloidal stability of all cationic thermoresponsive microgels is maintained over several days of incubation at 37 °C in biological mimicking medium (Dulbecco's Modified Eagle's Medium supplemented with fetal bovine serum). The thin cationic polymer shell covalently anchored does not hinder the all range of microgels to be biocompatible while the higher cytotoxicity of the doxorubicin-loaded microgels on HeLa cells proves their anti-tumor activity. The core-shell PVCL drug delivery nanocarriers allow a sustained release of doxorubicin with a slightly higher viability of HeLa cells incubated in the presence of DOXO-loaded microgels compared to the free DOXO. The nature of the endocytosis pathway is investigated through a quantification of the extent of the cellular survival rate in the presence of various cellular uptake inhibitors. A clathrin-dependent internalization was observed.
Collapse
Affiliation(s)
- Laura Etchenausia
- CNRS, University Pau & Pays Adour, E2S UPPA, Institut des Sciences Analytiques et de Physico-Chimie pour l'Environnement et les Matériaux, IPREM, UMR5254, 64000 Pau, France; Bionanoparticles Group, Department of Applied Chemistry, University of the Basque Country UPV/EHU, Donostia-San Sebastián, Spain
| | - Eva Villar-Alvarez
- Condensed Matter Physics Department, Faculty of Physics, 15782 Campus Sur, Universidad de Santiago de Compostela, Santiago de Compostela, Spain
| | - Jacqueline Forcada
- Bionanoparticles Group, Department of Applied Chemistry, University of the Basque Country UPV/EHU, Donostia-San Sebastián, Spain
| | - Maud Save
- CNRS, University Pau & Pays Adour, E2S UPPA, Institut des Sciences Analytiques et de Physico-Chimie pour l'Environnement et les Matériaux, IPREM, UMR5254, 64000 Pau, France.
| | - Pablo Taboada
- Condensed Matter Physics Department, Faculty of Physics, 15782 Campus Sur, Universidad de Santiago de Compostela, Santiago de Compostela, Spain.
| |
Collapse
|
48
|
Gene delivery to the rat retina by non-viral vectors based on chloroquine-containing cationic niosomes. J Control Release 2019; 304:181-190. [PMID: 31071372 DOI: 10.1016/j.jconrel.2019.05.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 02/15/2019] [Accepted: 05/04/2019] [Indexed: 01/14/2023]
Abstract
The incorporation of chloroquine within nano formulations, rather than as a co-treatment of the cells, could open a new avenue for in vivo retinal gene delivery. In this manuscript, we evaluated the incorporation of chloroquine diphosphate into the cationic niosome formulation composed of poloxamer 188, polysorbate 80 non-ionic surfactants, and 2,3-di (tetradecyloxy) propan-1-amine (hydrochloride salt) cationic lipid, to transfect rat retina. Niosome formulations without and with chloroquine diphosphate (DPP80, and DPP80-CQ, respectively) were prepared by the reverse phase evaporation technique and characterized in terms of size, PDI, zeta potential, and morphology. After the incorporation of the pCMS-EGFP plasmid, the resultant nioplexes -at different cationic lipid/DNA mass ratios- were further evaluated to compact, liberate, and secure the DNA against enzymatic digestion. In vitro procedures were achieved in ARPE-19 cells to assess transfection efficacy and intracellular transportation. Both nioplexes formulations transfected efficiently ARPE-19 cells, although the cell viability was clearly better in the case of DPP80-CQ nioplexes. After subretinal and intravitreal injections, DPP80 nioplexes were not able to transfect the rat retina. However, chloroquine containing vector showed protein expression in many retinal cells, depending on the administration route. These data provide new insights for retinal gene delivery based on chloroquine-containing niosome non-viral vectors.
Collapse
|
49
|
Production, surface modification and biomedical applications of nanodiamonds: A sparkling tool for theranostics. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 97:913-931. [DOI: 10.1016/j.msec.2018.12.073] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 12/21/2018] [Accepted: 12/22/2018] [Indexed: 02/07/2023]
|
50
|
Wang Y, Ye M, Xie R, Gong S. Enhancing the In Vitro and In Vivo Stabilities of Polymeric Nucleic Acid Delivery Nanosystems. Bioconjug Chem 2019; 30:325-337. [PMID: 30592619 PMCID: PMC6941189 DOI: 10.1021/acs.bioconjchem.8b00749] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Gene therapy holds great promise for various medical and biomedical applications. Nonviral gene delivery systems formed by cationic polymer and nucleic acids (e.g., polyplexes) have been extensively investigated for targeted gene therapy; however, their in vitro and in vivo stability is affected by both their intrinsic properties such as chemical compositions (e.g., polymer molecular weight and structure, and N/P ratio) and a number of environmental factors (e.g., shear stress during circulation in the bloodstream, interaction with the serum proteins, and physiological ionic strength). In this review, we surveyed the effects of a number of important intrinsic and environmental factors on the stability of polymeric gene delivery systems, and discussed various strategies to enhance the stability of polymeric gene delivery systems, thereby enabling efficient gene delivery into target cells. Future opportunities and challenges of polymeric nucleic acid delivery nanosystems were also briefly discussed.
Collapse
Affiliation(s)
- Yuyuan Wang
- Department of Materials Science and Engineering, University of Wisconsin–Madison, Madison, Wisconsin 53715, United States
- Wisconsin Institute for Discovery, University of Wisconsin–Madison, Madison, Wisconsin 53715, United States
| | - Mingzhou Ye
- Wisconsin Institute for Discovery, University of Wisconsin–Madison, Madison, Wisconsin 53715, United States
- Department of Biomedical Engineering, University of Wisconsin–Madison, Madison, Wisconsin 53715, United States
| | - Ruosen Xie
- Department of Materials Science and Engineering, University of Wisconsin–Madison, Madison, Wisconsin 53715, United States
- Wisconsin Institute for Discovery, University of Wisconsin–Madison, Madison, Wisconsin 53715, United States
| | - Shaoqin Gong
- Department of Materials Science and Engineering, University of Wisconsin–Madison, Madison, Wisconsin 53715, United States
- Wisconsin Institute for Discovery, University of Wisconsin–Madison, Madison, Wisconsin 53715, United States
- Department of Biomedical Engineering, University of Wisconsin–Madison, Madison, Wisconsin 53715, United States
- Department of Chemistry, University of Wisconsin–Madison, Madison, Wisconsin 53715, United States
| |
Collapse
|