1
|
Wu L, Sun W, Huang L, Sun L, Dou J, Lu G. Calcium Imaging in Vivo: How to Correctly Select and Apply Fiber Optic Photometric Indicators. Organogenesis 2025; 21:2489667. [PMID: 40186873 PMCID: PMC11980459 DOI: 10.1080/15476278.2025.2489667] [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: 12/02/2024] [Revised: 03/11/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025] Open
Abstract
Fiber-photometric is a novel optogenetic method for recording neural activity in vivo, which allows the use of calcium indicators to observe and study the relationship between neural activity and behavior in free-ranging animals. Calcium indicators also convert changes in calcium concentration in cells or tissues into recordable fluorescent signals, which can then be observed using the system of fiber-photometric. To date, there is a paucity of relevant literature on the proper selection and application of fiber-photometric indicators. Therefore, this paper will detail how to correctly select and apply fiber-photometer indicators in four sections: the basic principle of optical fiber photometry, the selection of calcium fluorescent probes and viral vector systems, and the measurement of specific expression of fluorescent proteins in specific tissues. Therefore, the correct use of suitable fiber optic recording indicators will greatly assist researchers in exploring the link between neuronal activity and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Lanxia Wu
- School of Psychology, Shandong Second Medical University, Weifang, Shandong, China
| | - Wenxuan Sun
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
| | - Linjie Huang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
| | - Lin Sun
- School of Psychology, Shandong Second Medical University, Weifang, Shandong, China
| | - Jinhua Dou
- Mental Health Education Center, Shandong Second Medical University, Weifang, Shandong, China
| | - Guohua Lu
- School of Psychology, Shandong Second Medical University, Weifang, Shandong, China
| |
Collapse
|
2
|
Le TP, Le TT, Jin S, Shin SA, Lyu AR, Park YH, Choi JS, Huh KM. Injectable polyplex-loaded glycol chitosan thermogel for efficient and safe inner ear gene delivery. J Control Release 2025; 380:1095-1108. [PMID: 39986472 DOI: 10.1016/j.jconrel.2025.02.060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 02/16/2025] [Accepted: 02/19/2025] [Indexed: 02/24/2025]
Abstract
Gene therapy has emerged as a promising approach for treating inner ear disorders, such as hearing loss and balance impairments, which require precise and efficient delivery mechanisms. This study introduces a dual-component delivery system combining RH-PAMAM G2 dendrimer-based polyplexes with hexanoyl glycol chitosan (HGC) thermogel to enhance gene delivery to the inner ear. The RH-PAMAM G2 dendrimers, modified with histidine and arginine, demonstrated high DNA binding affinity, low cytotoxicity, and effective cellular uptake, facilitating stable plasmid DNA (pDNA) transfection in HEI-OC1 auditory cells. Encapsulating these polyplexes within the HGC thermogel, an injectable and thermosensitive hydrogel, resulted in a supportive matrix that protects against premature clearance and provides sustained gene release upon intratympanic administration. In vivo studies in a mouse model confirmed substantial gene expression in the cochlear tissues, with widespread distribution in regions including the spiral ganglion and organ of Corti, compared to polyplexes alone. The HGC thermogel also exhibited favorable biocompatibility, with no observed inflammation or adverse effects in the middle ear tissues. This novel polyplex-HGC thermogel system demonstrates potential as a safe, efficient injectable gene delivery platform, offering a significant advance in gene therapy for inner ear disorders.
Collapse
Affiliation(s)
- Thi Phuc Le
- Department of Polymer Science and Engineering, Chungnam National University, Daejeon 34134, South Korea
| | - Thi Thuy Le
- Department of Biochemistry, Chungnam National University, Daejeon 34134, South Korea; Department of Biochemistry and Molecular Biology, Faculty of Biology, VNU University of Science, Vietnam National University, 334 Nguyen Trai, Thanh Xuan, Hanoi, Viet Nam
| | - Sheng Jin
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, South Korea
| | - Sun-Ae Shin
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, South Korea; Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Chungnam National University and Chungnam National University Hospital, Daejeon 35015, South Korea; Brain Research Institute, College of Medicine, Chungnam National University, Daejeon 35015, South Korea
| | - Ah-Ra Lyu
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, South Korea; Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Chungnam National University and Chungnam National University Hospital, Daejeon 35015, South Korea; Brain Research Institute, College of Medicine, Chungnam National University, Daejeon 35015, South Korea
| | - Yong-Ho Park
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, South Korea; Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Chungnam National University and Chungnam National University Hospital, Daejeon 35015, South Korea; Brain Research Institute, College of Medicine, Chungnam National University, Daejeon 35015, South Korea.
| | - Joon Sig Choi
- Department of Biochemistry, Chungnam National University, Daejeon 34134, South Korea.
| | - Kang Moo Huh
- Department of Polymer Science and Engineering, Chungnam National University, Daejeon 34134, South Korea; Department of Materials Science and Engineering, Chungnam National University, Daejeon 34134, South Korea.
| |
Collapse
|
3
|
Krabbes M, Kampik V, Krieghoff J, Haas V, Heizmann M, Morawietz M, Kalwa H, Kaysser L, Schoeder CT, Mäder K, Schmelzer CEH, Schulz-Siegmund M, Wölk C. Development and functionality analysis of lipoplex-loaded polysaccharide-based surface coatings for local nucleic acid delivery. Int J Pharm 2025; 673:125330. [PMID: 39956410 DOI: 10.1016/j.ijpharm.2025.125330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/27/2025] [Accepted: 02/06/2025] [Indexed: 02/18/2025]
Abstract
Although therapeutic nucleic acids reached the clinical application for a decade, the success of these new drugs is dependent on their delivery strategies, which are still a challenge. In particular, local delivery of nucleic acids is a promising approach to develop therapies with a spatially controlled site of action. However, compared to techniques for systemic administration, local nucleic acid delivery systems are still rarely described. In this study, we present a promising approach to fill this gap by the design of surface coatings based on polysaccharides for local delivery of nucleic acids. An automatized Layer-by-Layer deposition approach was applied using hyaluronic acid and chitosan to form polyelectrolyte multilayer systems, into which lipid nanoparticles, more specific lipoplexes, were embedded as nucleic acid carriers. Different manufacturing parameters, in particular the number of deposited polyelectrolyte layers and the preparation buffer, were varied. The multilayer film characteristics were investigated systematically regarding their physical properties, with a focus on thickness and topology as well as lipoplex deposition, to identify a system with efficient transfection properties. The multilayer systems prepared in acetate buffer were characterized by a good lipoplex embedding with a more uniform distribution and lower tendency for formation of large lipoplex aggregates in the polyelectrolyte film. Additionally, we were able to demonstrate the functionality of the developed system for nucleic acid delivery. The nucleic acids were successfully transferred into cells in a contact-triggered manner. Furthermore, we could demonstrate the enzymatic degradation-based release of nucleic acid cargo from the delivery system caused by hyaluronidase, followed by successful in vitro transfection.
Collapse
Affiliation(s)
- Maria Krabbes
- Institute of Pharmacy, Faculty of Medicine, Leipzig University, Leipzig, Saxony, Germany
| | - Vincent Kampik
- Institute of Pharmacy, Faculty of Medicine, Leipzig University, Leipzig, Saxony, Germany
| | - Jan Krieghoff
- Institute of Pharmacy, Faculty of Medicine, Leipzig University, Leipzig, Saxony, Germany
| | - Vivian Haas
- Institute for Drug Discovery, Faculty of Medicine, Leipzig University, Leipzig, Saxony, Germany
| | - Mathilde Heizmann
- Institute for Drug Discovery, Faculty of Medicine, Leipzig University, Leipzig, Saxony, Germany
| | - Maria Morawietz
- Fraunhofer Institute for Microstructure of Materials and Systems IMWS, Halle (Saale), Saxony-Anhalt, Germany
| | - Hermann Kalwa
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Faculty of Medicine, Leipzig University, Leipzig, Saxony, Germany
| | - Leonard Kaysser
- Institute for Drug Discovery, Faculty of Medicine, Leipzig University, Leipzig, Saxony, Germany
| | - Clara T Schoeder
- Institute for Drug Discovery, Faculty of Medicine, Leipzig University, Leipzig, Saxony, Germany
| | - Karsten Mäder
- Institute of Pharmacy, Faculty of Science I, MLU Halle-Wittenberg, Halle (Saale), Saxony-Anhalt, Germany
| | - Christian E H Schmelzer
- Fraunhofer Institute for Microstructure of Materials and Systems IMWS, Halle (Saale), Saxony-Anhalt, Germany
| | | | - Christian Wölk
- Institute of Pharmacy, Faculty of Medicine, Leipzig University, Leipzig, Saxony, Germany.
| |
Collapse
|
4
|
Razzaq S, Fatima I, Moafian Z, Rahdar A, Fathi-Karkan S, Kharaba Z, Shirzad M, Khan A, Pandey S. Nanomedicine innovations in colon and rectal cancer: advances in targeted drug and gene delivery systems. Med Oncol 2025; 42:113. [PMID: 40097759 DOI: 10.1007/s12032-025-02670-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Accepted: 03/04/2025] [Indexed: 03/19/2025]
Abstract
Nanotechnology has revolutionized cancer diagnostics and therapy, offering unprecedented possibilities to overcome the constraints of conventional treatments. This study provides a detailed overview of the current progress and difficulties in the creation of nanostructured materials, with a specific emphasis on their use in drug and gene delivery systems. The study examines tactics that attempt to improve the effectiveness and safety of chemotherapeutic drugs such as doxorubicin (Dox) by focusing on the potential of antibody-drug conjugates and functionalized nanoparticles. Moreover, it clarifies the challenges encountered in administering nanoparticles orally for gastrointestinal treatments, emphasizing the crucial physicochemical properties that affect their behavior in the gastrointestinal system. This study highlights the transformational potential of nanostructured materials in precision oncology by examining advanced breakthroughs such cell membrane-camouflaged nanoparticles and inorganic nanoparticles designed for gastrointestinal disorders. The text investigates the processes involved in the absorption of nanoparticles and their destruction in lysosomes, revealing the many methods in which enterocytes take up these particles. This study strongly supports the use of advanced nanoparticle-based methods to reduce the harmful effects on the whole body and improve the effectiveness of therapy, based on a thorough examination of current experiments on animals and humans. The main objective of this paper is to provide a fundamental comprehension that will stimulate more investigation and practical use in the field of cancer nanomedicine, advancing its boundaries.
Collapse
Affiliation(s)
- Sobia Razzaq
- School of Pharmacy, University of Management and Technology, Lahore, Punjab, Pakistan
| | - Iqra Fatima
- Department of Pharmacy, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Zeinab Moafian
- Department of Chemistry and Biochemistry, University of Delaware, Newark, USA
| | - Abbas Rahdar
- Department of Physics, Faculty of Sciences, University of Zabol, Zabol, 538-98615, Iran.
| | - Sonia Fathi-Karkan
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, 94531-55166, Iran.
- Department of Advanced Sciences and Technologies in Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, 9414974877, Iran.
| | - Zelal Kharaba
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, UAE
| | - Maryam Shirzad
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ahmad Khan
- Department of Pharmacy, Quaid-i-Azam University, Islamabad, 45320, Pakistan.
| | - Sadanand Pandey
- School of Bioengineering and Food Technology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, 173229, Himachal Pradesh, India.
- Department of Chemistry, College of Natural Science, Yeungnam University, 280 Daehak-Ro, Gyeongsan, Gyeongbuk, 38541, Republic of Korea.
| |
Collapse
|
5
|
Gonzalez JC, Park KW, Evans DB, Sharma R, Sahaym O, Gopalakrishnan S, dar AI, Valdez TA, Sharma A. Nano Approaches to Nucleic Acid Delivery: Barriers, Solutions, and Current Landscape. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2025; 17:e70010. [PMID: 40223402 PMCID: PMC11994986 DOI: 10.1002/wnan.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 03/07/2025] [Accepted: 03/27/2025] [Indexed: 04/15/2025]
Abstract
Nucleic acid (NA) therapy holds tremendous potential for treating a wide range of genetic diseases by the delivery of therapeutic genes into target cells. However, significant challenges exist in safely and effectively delivering these genes to their intended locations. Viral vectors, though efficient, pose risks such as immunogenicity and mutagenesis. This has resulted in growing interest in non-viral, nanoparticle-based NA delivery systems. This review article describes various physiological barriers to NA delivery and explores nanoparticle-based NA delivery systems, including bioengineered nanoparticles, peptides, lipid nanoparticles, and polymeric nanoparticles, highlighting their unique features to overcome in vivo barriers for NA delivery. While these nanoparticle-based NA delivery systems offer a promising alternative to viral vectors, challenges related to cytotoxicity, reproducible synthesis, and cost need to be addressed. The current clinical landscape of NA delivery is also discussed, emphasizing the need for safer, scalable, and cost-effective solutions. Nanoparticles represent a promising future in NA therapy, with the possibility of developing clinically relevant, non-toxic, stable, and non-immunogenic delivery vehicles, paving the way for broader therapeutic applications and improved clinical outcomes.
Collapse
Affiliation(s)
- Joan Castaneda Gonzalez
- Department of ChemistryCollege of Arts and Sciences, Washington State UniversityPullmanWashingtonUSA
| | - Ki Wan Park
- Department of Otolaryngology−Head & Neck Surgery DivisionsStanford University School of MedicineStanfordCaliforniaUSA
| | - Dallin Brian Evans
- Department of ChemistryCollege of Arts and Sciences, Washington State UniversityPullmanWashingtonUSA
| | - Rishi Sharma
- Department of ChemistryCollege of Arts and Sciences, Washington State UniversityPullmanWashingtonUSA
| | - Om Sahaym
- Department of ChemistryCollege of Arts and Sciences, Washington State UniversityPullmanWashingtonUSA
| | - Shamila Gopalakrishnan
- Department of ChemistryCollege of Arts and Sciences, Washington State UniversityPullmanWashingtonUSA
| | - Aqib Iqbal dar
- Department of ChemistryCollege of Arts and Sciences, Washington State UniversityPullmanWashingtonUSA
| | - Tulio A. Valdez
- Department of Otolaryngology−Head & Neck Surgery DivisionsStanford University School of MedicineStanfordCaliforniaUSA
| | - Anjali Sharma
- Department of ChemistryCollege of Arts and Sciences, Washington State UniversityPullmanWashingtonUSA
| |
Collapse
|
6
|
Yao Z, Liu T, Wang J, Fu Y, Zhao J, Wang X, Li Y, Yang X, He Z. Targeted delivery systems of siRNA based on ionizable lipid nanoparticles and cationic polymer vectors. Biotechnol Adv 2025; 81:108546. [PMID: 40015385 DOI: 10.1016/j.biotechadv.2025.108546] [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: 09/23/2024] [Revised: 02/04/2025] [Accepted: 02/23/2025] [Indexed: 03/01/2025]
Abstract
As an emerging therapeutic tool, small interfering RNA (siRNA) had the capability to down-regulate nearly all human mRNAs via sequence-specific gene silencing. Numerous studies have demonstrated the substantial potential of siRNA in the treatment of broad classes of diseases. With the discovery and development of various delivery systems and chemical modifications, six siRNA-based drugs have been approved by 2024. The utilization of siRNA-based therapeutics has significantly propelled efforts to combat a wide array of previously incurable diseases and advanced at a rapid pace, particularly with the help of potent targeted delivery systems. Despite encountering several extracellular and intracellular challenges, the efficiency of siRNA delivery has been gradually enhanced. Currently, targeted strategies aimed at improving potency and reducing toxicity played a crucial role in the druggability of siRNA. This review focused on recent advancements on ionizable lipid nanoparticles (LNPs) and cationic polymer (CP) vectors applied for targeted siRNA delivery. Based on various types of targeted modifications, we primarily described delivery systems modified with receptor ligands, peptides, antibodies, aptamers and amino acids. Finally, we discussed the challenges and opportunities associated with siRNA delivery systems based on ionizable LNPs and CPs vectors.
Collapse
Affiliation(s)
- Ziying Yao
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Taiqing Liu
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jingwen Wang
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yunhai Fu
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jinhua Zhao
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaoyu Wang
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yinqi Li
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaodong Yang
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhiyao He
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
7
|
Nerella SG, Eldridge MAG, Innis RB, Pike VW. PET Reporter Probes for Brain Imaging of Transduced Gene and Cell Expression: Status and Challenges. J Med Chem 2025; 68:2198-2218. [PMID: 39879224 DOI: 10.1021/acs.jmedchem.4c02326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Gene therapy and cell transduction are gaining interest as therapeutic strategies for neurological and psychiatric disorders. Positron emission tomography (PET) has been established as a uniquely powerful modality for brain molecular imaging in vivo. The utility of PET depends on the development and application of suitably specific radiotracers and/or reporter probes. PET probes are potentially useful to confirm the success of gene therapy or cell transduction without the need for brain biopsy or necroscopy. Probes are needed to target proteins expressed by specific exogenous transgenes or cells and could play a crucial role in elucidating neurobiological mechanisms and in longitudinal tracking of expression for therapeutic applications. This perspective article describes the current status and ongoing challenges for the design and development of PET reporter probes for verifying the expression of reporter genes and cells in the brain. Radiochemical aspects, applications, and translational challenges for diagnostic and therapeutic interventions are highlighted.
Collapse
Affiliation(s)
- Sridhar Goud Nerella
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, 20892 United States
| | - Mark A G Eldridge
- Laboratory of Neuropsychology, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, 20892 United States
- Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, NE1 7RU, U.K
| | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, 20892 United States
| | - Victor W Pike
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, 20892 United States
| |
Collapse
|
8
|
Hosseini-Kharat M, Bremmell KE, Grubor-Bauk B, Prestidge CA. Enhancing non-viral DNA delivery systems: Recent advances in improving efficiency and target specificity. J Control Release 2025; 378:170-194. [PMID: 39647508 DOI: 10.1016/j.jconrel.2024.12.002] [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: 06/07/2024] [Revised: 11/23/2024] [Accepted: 12/02/2024] [Indexed: 12/10/2024]
Abstract
DNA-based therapies are often limited by challenges such as stability, long-term integration, low transfection efficiency, and insufficient targeted DNA delivery. This review focuses on recent progress in the design of non-viral delivery systems for enhancing targeted DNA delivery and modulation of therapeutic efficiency. Cellular uptake and intracellular trafficking mechanisms play a crucial role in optimizing gene delivery efficiency. There are two main strategies employed to improve the efficiency of gene delivery vectors: (i) explore different administration routes (e.g., mucosal, intravenous, intramuscular, subcutaneous, intradermal, intratumoural, and intraocular) that best facilitates optimal uptake into the targeted cells and organs and (ii) modify the delivery vectors with cell-specific ligands (e.g., natural ligands, antibodies, peptides, carbohydrates, or aptamers) that enable targeted uptake to specific cells with higher specificity and improved biodistribution. We describe how recent progress in employing these DNA delivery strategies is advancing the field and increasing the clinical translation and ultimate clinical application of DNA therapies.
Collapse
Affiliation(s)
- Mahboubeh Hosseini-Kharat
- Clinical and Health Sciences, Centre for Pharmaceutical Innovation, University of South Australia, Adelaide, SA 5000, Australia
| | - Kristen E Bremmell
- Clinical and Health Sciences, Centre for Pharmaceutical Innovation, University of South Australia, Adelaide, SA 5000, Australia
| | - Branka Grubor-Bauk
- Viral Immunology Group, Adelaide Medical School, University of Adelaide and Basil Hetzel Institute for Translational Health Research, Adelaide, SA, Australia
| | - Clive A Prestidge
- Clinical and Health Sciences, Centre for Pharmaceutical Innovation, University of South Australia, Adelaide, SA 5000, Australia.
| |
Collapse
|
9
|
Zwi-Dantsis L, Mohamed S, Massaro G, Moeendarbary E. Adeno-Associated Virus Vectors: Principles, Practices, and Prospects in Gene Therapy. Viruses 2025; 17:239. [PMID: 40006994 PMCID: PMC11861813 DOI: 10.3390/v17020239] [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: 12/22/2024] [Revised: 01/27/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Gene therapy offers promising potential as an efficacious and long-lasting therapeutic option for genetic conditions, by correcting defective mutations using engineered vectors to deliver genetic material to host cells. Among these vectors, adeno-associated viruses (AAVs) stand out for their efficiency, versatility, and safety, making them one of the leading platforms in gene therapy. The enormous potential of AAVs has been demonstrated through their use in over 225 clinical trials and the FDA's approval of six AAV-based gene therapy products, positioning these vectors at the forefront of the field. This review highlights the evolution and current applications of AAVs in gene therapy, focusing on their clinical successes, ongoing developments, and the manufacturing processes required for the rapid commercial growth anticipated in the AAV therapy market. It also discusses the broader implications of these advancements for future therapeutic strategies targeting more complex and multi-systemic conditions and biological processes such as aging. Finally, we explore some of the major challenges currently confronting the field.
Collapse
Affiliation(s)
- Limor Zwi-Dantsis
- Department of Mechanical Engineering, Roberts Building, University College London, London WC1E 6BT, UK
| | - Saira Mohamed
- Department of Mechanical Engineering, Roberts Building, University College London, London WC1E 6BT, UK
| | - Giulia Massaro
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK
| | - Emad Moeendarbary
- Department of Mechanical Engineering, Roberts Building, University College London, London WC1E 6BT, UK
| |
Collapse
|
10
|
Nunziata G, Nava M, Lacroce E, Pizzetti F, Rossi F. Thermo-Responsive Polymer-Based Nanoparticles: From Chemical Design to Advanced Applications. Macromol Rapid Commun 2025:e2401127. [PMID: 39895239 DOI: 10.1002/marc.202401127] [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: 12/20/2024] [Revised: 01/20/2025] [Indexed: 02/04/2025]
Abstract
Thermo-responsive polymers have emerged as a cutting-edge tool in nanomedicine, paving the way for innovative approaches to targeted drug delivery and advanced therapeutic strategies. These "smart" polymers respond to temperature changes, enabling controlled drug release in pathological environments characterized by high temperatures. By exploiting their unique phase transition, occurring at the lower or upper critical solution temperatures (LCST and UCST), these systems ensure localized therapeutic action, minimizing collateral damage to healthy tissues. The integration of these polymers into nanoparticles with hydrophilic shells and hydrophobic cores enhances their stability and biocompatibility. Furthermore, advanced polymer engineering allows precise modulation of LCST and UCST through adjustments in composition and hydrophilic-lipophilic balance, optimizing their responsiveness for specific applications. In addition to drug delivery, thermo-responsive nanoparticles are gaining attention in several fields such as gene therapy and imaging. Therefore, this review explores the chemical and structural diversity of thermo-responsive nanoparticles, emphasizing their ability to encapsulate and release drugs effectively. Second, this review highlights the potential of thermo-responsive nanoparticles to redefine treatment paradigms, providing a comprehensive understanding of their mechanisms, applications, and future perspectives in biomedical research.
Collapse
Affiliation(s)
- Giuseppe Nunziata
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, via Mancinelli 7, Milano, 20131, Italy
| | - Marco Nava
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, via Mancinelli 7, Milano, 20131, Italy
| | - Elisa Lacroce
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, via Mancinelli 7, Milano, 20131, Italy
| | - Fabio Pizzetti
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, via Mancinelli 7, Milano, 20131, Italy
| | - Filippo Rossi
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, via Mancinelli 7, Milano, 20131, Italy
| |
Collapse
|
11
|
Zhang J, Yang X, Chang Z, Zhu W, Ma Y, He H. Polymeric nanocarriers for therapeutic gene delivery. Asian J Pharm Sci 2025; 20:101015. [PMID: 39931356 PMCID: PMC11808530 DOI: 10.1016/j.ajps.2025.101015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 11/12/2024] [Accepted: 11/19/2024] [Indexed: 02/13/2025] Open
Abstract
The recent commercialization of gene products has sparked significant interest in gene therapy, necessitating efficient and precise gene delivery via various vectors. Currently, viral vectors and lipid-based nanocarriers are the predominant choices and have been extensively investigated and reviewed. Beyond these vectors, polymeric nanocarriers also hold the promise in therapeutic gene delivery owing to their versatile functionalities, such as improving the stability, cellar uptake and endosomal escape of nucleic acid drugs, along with precise delivery to targeted tissues. This review presents a brief overview of the status quo of the emerging polymeric nanocarriers for therapeutic gene delivery, focusing on key cationic polymers, nanocarrier types, and preparation methods. It also highlights targeted diseases, strategies to improve delivery efficiency, and potential future directions in this research area. The review is hoped to inspire the development, optimization, and clinical translation of highly efficient polymeric nanocarriers for therapeutic gene delivery.
Collapse
Affiliation(s)
- Jiayuan Zhang
- Key Laboratory of Smart Drug Delivery of Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
- Key Laboratory for Tibet Plateau Phytochemistry of Qinghai Province, School of Pharmacy, Qinghai Minzu University, Xining 810007, China
| | - Xinyu Yang
- Key Laboratory of Smart Drug Delivery of Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Zhichao Chang
- Key Laboratory of Smart Drug Delivery of Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Wenwei Zhu
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yuhua Ma
- Key Laboratory for Tibet Plateau Phytochemistry of Qinghai Province, School of Pharmacy, Qinghai Minzu University, Xining 810007, China
| | - Haisheng He
- Key Laboratory of Smart Drug Delivery of Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| |
Collapse
|
12
|
Jonjaroen V, Jitrakorn S, Charoonnart P, Kaewsaengon P, Thinkohkaew K, Payongsri P, Surarit R, Saksmerprome V, Niamsiri N. Optimizing chitosan nanoparticles for oral delivery of double-stranded RNA in treating white spot disease in shrimp: Key insights and practical implications. Int J Biol Macromol 2025; 290:138970. [PMID: 39706429 DOI: 10.1016/j.ijbiomac.2024.138970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/11/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Delivering double-stranded RNA (dsRNA) in shrimp is challenging due to the lack of an effective carrier system. This study optimized chitosan nanoparticles (CNs) from two sources-α-chitosan from shrimp and β-chitosan from squid-to encapsulate antiviral dsRNA for oral administration via shrimp feed. Using response surface methodology (RSM), formulations were refined for encapsulation efficiency, particle size, polydispersity index, and zeta potential. Both types of CNs demonstrated high encapsulation efficiency (>95 %), small sizes (<300 nm), and stable zeta potential (>20 mV). Shrimp-derived CNs provided superior RNase protection and controlled release, while squid-derived CNs showed a burst release. Incorporated into feed, both types of CNs remained stable for over a month. Shrimp-derived CNs offered greater dsRNA protection (>70 %) and improved efficacy against white spot syndrome virus (WSSV), significantly reducing mortality. These results position shrimp-derived CNs as promising dsRNA carriers for combating WSSV in shrimp aquaculture.
Collapse
Affiliation(s)
- Veasarach Jonjaroen
- Department of Biotechnology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand.
| | - Sarocha Jitrakorn
- Center of Excellence for Shrimp Molecular Biology and Biotechnology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; National Center of Genetic Engineering and Biotechnology, (BIOTEC), Thailand Science Park, Pathum Thani 12120, Thailand.
| | - Patai Charoonnart
- Center of Excellence for Shrimp Molecular Biology and Biotechnology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; National Center of Genetic Engineering and Biotechnology, (BIOTEC), Thailand Science Park, Pathum Thani 12120, Thailand.
| | - Parichart Kaewsaengon
- Center of Excellence for Shrimp Molecular Biology and Biotechnology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; National Center of Genetic Engineering and Biotechnology, (BIOTEC), Thailand Science Park, Pathum Thani 12120, Thailand.
| | - Korlid Thinkohkaew
- Department of Material Science, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand.
| | - Panwajee Payongsri
- Department of Biotechnology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand.
| | - Rudee Surarit
- Department of Oral Biology, Faculty of Dentistry, Mahidol University, Bangkok 10400, Thailand; Faculty of Dentistry, Siam University, Bangkok 10160, Thailand.
| | - Vanvimon Saksmerprome
- Center of Excellence for Shrimp Molecular Biology and Biotechnology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; National Center of Genetic Engineering and Biotechnology, (BIOTEC), Thailand Science Park, Pathum Thani 12120, Thailand.
| | - Nuttawee Niamsiri
- Department of Biotechnology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand.
| |
Collapse
|
13
|
Palmer CR, Pastora LE, Kimmel BR, Pagendarm HM, Kwiatkowski AJ, Stone PT, Arora K, Francini N, Fedorova O, Pyle AM, Wilson JT. Covalent Polymer-RNA Conjugates for Potent Activation of the RIG-I Pathway. Adv Healthc Mater 2025; 14:e2303815. [PMID: 38648653 PMCID: PMC11493851 DOI: 10.1002/adhm.202303815] [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: 11/02/2023] [Revised: 04/13/2024] [Indexed: 04/25/2024]
Abstract
RNA ligands of retinoic acid-inducible gene I (RIG-I) are a promising class of oligonucleotide therapeutics with broad potential as antiviral agents, vaccine adjuvants, and cancer immunotherapies. However, their translation has been limited by major drug delivery barriers, including poor cellular uptake, nuclease degradation, and an inability to access the cytosol where RIG-I is localized. Here this challenge is addressed by engineering nanoparticles that harness covalent conjugation of 5'-triphospate RNA (3pRNA) to endosome-destabilizing polymers. Compared to 3pRNA loaded into analogous nanoparticles via electrostatic interactions, it is found that covalent conjugation of 3pRNA improves loading efficiency, enhances immunostimulatory activity, protects against nuclease degradation, and improves serum stability. Additionally, it is found that 3pRNA could be conjugated via either a disulfide or thioether linkage, but that the latter is only permissible if conjugated distal to the 5'-triphosphate group. Finally, administration of 3pRNA-polymer conjugates to mice significantly increases type-I interferon levels relative to analogous carriers that use electrostatic 3pRNA loading. Collectively, these studies have yielded a next-generation polymeric carrier for in vivo delivery of 3pRNA, while also elucidating new chemical design principles for covalent conjugation of 3pRNA with potential to inform the further development of therapeutics and delivery technologies for pharmacological activation of RIG-I.
Collapse
Affiliation(s)
- Christian R. Palmer
- Department of Chemical and Biomolecular EngineeringVanderbilt UniversityNashvilleTN37235USA
| | - Lucinda E. Pastora
- Department of Chemical and Biomolecular EngineeringVanderbilt UniversityNashvilleTN37235USA
| | - Blaise R. Kimmel
- Department of Chemical and Biomolecular EngineeringVanderbilt UniversityNashvilleTN37235USA
| | - Hayden M. Pagendarm
- Department of Biomedical EngineeringVanderbilt UniversityNashvilleTN37235USA
| | | | - Payton T. Stone
- Department of Chemical and Biomolecular EngineeringVanderbilt UniversityNashvilleTN37235USA
| | - Karan Arora
- Department of Chemical and Biomolecular EngineeringVanderbilt UniversityNashvilleTN37235USA
| | - Nora Francini
- Department of Biomedical EngineeringVanderbilt UniversityNashvilleTN37235USA
| | - Olga Fedorova
- Department of MolecularCellular and Developmental BiologyYale UniversityNew HavenCT06511USA
- Howard Hughes Medical InstituteChevy ChaseMD20815USA
| | - Anna M. Pyle
- Department of MolecularCellular and Developmental BiologyYale UniversityNew HavenCT06511USA
- Howard Hughes Medical InstituteChevy ChaseMD20815USA
- Department of ChemistryYale UniversityNew HavenCT06511USA
| | - John T. Wilson
- Department of Chemical and Biomolecular EngineeringVanderbilt UniversityNashvilleTN37235USA
- Department of Biomedical EngineeringVanderbilt UniversityNashvilleTN37235USA
- Department of PathologyMicrobiologyand ImmunologyVanderbilt University Medical CenterNashvilleTN37232USA
- Vanderbilt‐Ingram Cancer CenterVanderbilt University Medical CenterNashvilleTN37232USA
| |
Collapse
|
14
|
Psarrou M, Vamvakaki M, Karatasos K, Rissanou AN. Interfacial interactions between DNA and polysaccharide-coated magnetic nanoparticles: Insight from simulations and experiments. Colloids Surf B Biointerfaces 2025; 246:114386. [PMID: 39603198 DOI: 10.1016/j.colsurfb.2024.114386] [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: 09/17/2024] [Revised: 11/05/2024] [Accepted: 11/16/2024] [Indexed: 11/29/2024]
Abstract
In this work we examine the structural and energetic stability and the interactions between dextran-coated magnetic nanoparticles (MNPs) and a DNA oligonucleotide at ionic strength conditions that are relevant to physiological gene delivery processes. All-atom Molecular Dynamics simulations provided information at the atomic-level regarding the mechanisms responsible for the physical adsorption of Dextran on the magnetic surface and the conditions under which a successful DNA-Dextran complexation can be accomplished. Coulombic interactions were found to play the main role for the formation of the Dextran interfacial layer onto the magnetic surface while hydrogen bonding between the Dextran molecules enhanced the structural integrity of this layer. The Dextran-DNA complexation was also driven by electrostatic interactions between the two moieties. An increase of the salt concentration was found to promote DNA complexation with the DX-coated magnetic nanoparticles, through the modification of the Coulombic interactions between the DX and DNA chains, which worked synergistically with the increase in hydrogen bonding between the two macromolecules. Comparison of the behavior of the coated with the uncoated magnetic nanoparticles, highlighted the significant role of the DX interfacial layer on the DNA association to the magnetic surface. Relevant experimental results provided complementary information for the coated nanoparticle/DNA interactions at different (larger) length scales. A good qualitative agreement was found between the simulation and experimental findings. This study demonstrates that tailoring the nanoparticle coating and ionic strength can optimize the delivery of DNA by fine-tuning the favorable interfacial forces and thus the DNA/MNP binding stability.
Collapse
Affiliation(s)
- Maria Psarrou
- Department of Materials Science and Technology, University of Crete, Heraklion, Crete 700 13, Greece; Institute of Electronic Structure and Laser, FORTH, Heraklion, Crete 700 13, Greece
| | - Maria Vamvakaki
- Department of Materials Science and Technology, University of Crete, Heraklion, Crete 700 13, Greece; Institute of Electronic Structure and Laser, FORTH, Heraklion, Crete 700 13, Greece
| | - Kostas Karatasos
- Department of Chemical Engineering, University of Thessaloniki, P.O. BOX 420, Thessaloniki 54124, Greece
| | - Anastassia N Rissanou
- Theoretical & Physical Chemistry Institute, National Hellenic Research Foundation, 48 Vassileos Constantinou Avenue, Athens 11635, Greece.
| |
Collapse
|
15
|
Jing X, Mackay JP, Passioura T. Macrocyclic peptides as a new class of targeted protein degraders. RSC Chem Biol 2025:d4cb00199k. [PMID: 39822773 PMCID: PMC11733494 DOI: 10.1039/d4cb00199k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 01/01/2025] [Indexed: 01/19/2025] Open
Abstract
Targeted protein degraders, in the form of proteolysis targeting chimaeras (PROTACs) and molecular glues, leverage the ubiquitin-proteasome system to catalytically degrade specific target proteins of interest. Because such molecules can be extremely potent, they have attracted considerable attention as a therapeutic modality in recent years. However, while targeted degraders have great potential, they are likely to face many of the same challenges as more traditional small molecules when it comes to their development as therapeutics. In particular, existing targeted degrader design is largely only applicable to the same set of protein targets as traditional small molecules (i.e., ∼15% of the human proteome). Here, we consider the potential of macrocyclic peptides to overcome this limitation. Such molecules possess several features that make them well-suited for the role, including the ability to induce the formation of ternary protein complexes that can involve relatively flat surfaces and their structural commonality with E3 ligase-recruiting peptide degrons. For these reasons, macrocyclic peptides provide the opportunity both to broaden the number of targets accessible to degrader activity and to broaden the number of E3 ligases that can be harnessed to mediate that activity.
Collapse
Affiliation(s)
- Xuefei Jing
- School of Life and Environmental Sciences, The University of Sydney Sydney NSW 2006 Australia
| | - Joel P Mackay
- School of Life and Environmental Sciences, The University of Sydney Sydney NSW 2006 Australia
| | - Toby Passioura
- School of Chemistry, The University of Sydney Sydney NSW 2006 Australia
- Insamo South Pty Ltd Chippendale NSW 2008 Australia
| |
Collapse
|
16
|
Müllerová M, Tarach P, Strašák T, Cuřínová P, Petrickovic R, Závodná T, Topinka J, Janaszewska A, Klajnert-Maculewicz B, Červenková Št’astná L. Comparative Study of Functionalized Carbosilane Dendrimers for siRNA Delivery: Synthesis, Cytotoxicity, and Biophysical Properties. ACS OMEGA 2025; 10:1047-1060. [PMID: 39829590 PMCID: PMC11740622 DOI: 10.1021/acsomega.4c08314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 01/06/2025]
Abstract
Efficient and safe carriers of genetic material are crucial for advancing gene therapy. Three new series of cationic dendritic nanocarriers based on a carbosilane scaffold, differentiated by peripheral modifications: saccharide (CS-glyco), amine (CS-N), and phosphonium dendrimers (CS-P) were designed for binding, protecting, and releasing polyanionic compounds like therapeutic siRNA. Besides introducing synthetic methodology, this study brings a unique direct interstructural comparison of 16 dendritic nanovector's characteristics, addressing a gap in typical research that focuses on uniform structural types. The study evaluates the dendrimer's in vitro cytotoxicity, biophysical properties, and complexation capabilities in comparison with widely used PAMAM dendrimers. CS-glyco and PAMAMs were significantly less toxic to MCF-7 and THP-1 cell lines than were CS-N and CS-P, despite having the same peripheral charge density. Notably, CS-glyco maintained biocompatibility comparable to analogous neutral CS glycodendrimers, underscoring the exceptional capability of sugar coating to reduce toxicity. Dendriplexes formed from these nanocarriers protected siRNA from RNase degradation and facilitated its release in the presence of heparin, highlighting its potential in gene delivery applications. The study provides a background for future in-depth investigations into the introduced dendritic nanocarriers, which show significant potential for advancing drug delivery.
Collapse
Affiliation(s)
- Monika Müllerová
- Institute
of Chemical Process Fundamentals Czech Academy of Sciences, Rozvojová 135, Prague 165 02, Czech Republic
- Institute
of Experimental Medicine, Czech Academy
of Sciences, Vídeňská
1083, Prague 142 00, Czech Republic
| | - Piotr Tarach
- Faculty of
Biology and Environmental Protection, Department of General Biophysics, University of Lodz, Pomorska 141/143, Lodz 90-236, Poland
| | - Tomáš Strašák
- Institute
of Chemical Process Fundamentals Czech Academy of Sciences, Rozvojová 135, Prague 165 02, Czech Republic
| | - Petra Cuřínová
- Institute
of Chemical Process Fundamentals Czech Academy of Sciences, Rozvojová 135, Prague 165 02, Czech Republic
- Department
of Organic Chemistry, University of Chemistry
and Technology Prague, Technická 5, Prague 6 166 28, Czech Republic
| | - Roman Petrickovic
- Institute
of Chemical Process Fundamentals Czech Academy of Sciences, Rozvojová 135, Prague 165 02, Czech Republic
| | - Táňa Závodná
- Institute
of Experimental Medicine, Czech Academy
of Sciences, Vídeňská
1083, Prague 142 00, Czech Republic
| | - Jan Topinka
- Institute
of Experimental Medicine, Czech Academy
of Sciences, Vídeňská
1083, Prague 142 00, Czech Republic
| | - Anna Janaszewska
- Faculty of
Biology and Environmental Protection, Department of General Biophysics, University of Lodz, Pomorska 141/143, Lodz 90-236, Poland
| | - Barbara Klajnert-Maculewicz
- Faculty of
Biology and Environmental Protection, Department of General Biophysics, University of Lodz, Pomorska 141/143, Lodz 90-236, Poland
| | - Lucie Červenková Št’astná
- Institute
of Chemical Process Fundamentals Czech Academy of Sciences, Rozvojová 135, Prague 165 02, Czech Republic
| |
Collapse
|
17
|
Zheng D, Chen T, Yang K, Yin G, Chen Y, Gui J, Xu C, Lv S. Microfluidic Synthesis of miR-200c-3p Lipid Nanoparticles: Targeting ZEB2 to Alleviate Chondrocyte Damage in Osteoarthritis. Int J Nanomedicine 2025; 20:505-521. [PMID: 39830158 PMCID: PMC11742371 DOI: 10.2147/ijn.s491711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 12/21/2024] [Indexed: 01/22/2025] Open
Abstract
Introduction Osteoarthritis (OA) is a degenerative joint disease characterized by articular cartilage degeneration. Chondrocyte inflammation, apoptosis, and extracellular matrix degradation accelerated OA progression. MicroRNA (miRNA) has the potential to be a therapeutic method for osteoarthritis. However, it is difficult to penetrate the cell to exercise its biological function, and its extracellular effect is unclear. Methods lipo-AgPEI-miR-200c-3p was created by combining miR-200c-3p with silver nitrate polyvinylimine nanoparticles on a microfluidic device. The drug release curve, stability, temperature sensitivity, cytotoxicity, and the impact of lipo-AgPEI-miR-200c-3p on the expression of proteins linked to matrix disintegration, apoptosis, and inflammatory factors were all detected. Results Results showed that the particle size of Lipo-AgPEI-miR-200c-3p was about 130 nm, the Zeta potential was lowered to 1.08±0.12 mV. Lipo-AgPEI-miR-200c-3p could increase cell viability, prevent cell apoptosis, and decrease the expression levels of TNF-α, IL-6, IL-1β, and MCP-1 in ADTC5 cells following LPS stimulation. MMP3, MMP13, and ADAMTS-4 expression was downregulated whereas collagen II expression was upregulated. The ZEB2 expression was greatly elevated following LPS stimulation and dramatically decreased following transfection of miR-200c-3p. Collagen II expression rose following transfection of si-ZEB2, whereas the expression levels of inflammatory factors, apoptosis-related proteins, MMP3, MMP13, and ADAMTS-4 decreased. The dual luciferase experiment demonstrated that ZEB2 was the target gene of miR-200c-3p. Conclusion The synergistic effect of AgPEI and miR-200c-3p can inhibit the inflammatory response, apoptosis, and matrix degradation of chondrocytes. Lipo-AgPEI-miR-200c-3p can also improve transfection efficiency and obtain good physicochemical properties of drugs. miR-200c-3p may be crucial in the development of OA and can influence the target gene ZEB2, control the inflammatory response, apoptosis, and chondrocyte matrix breakdown.
Collapse
Affiliation(s)
- Dong Zheng
- Department of Orthopedics, The Affiliated Changzhou No.2 People’s Hospital with Nanjing Medical University, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, People’s Republic of China
| | - Tong Chen
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Kaiyuan Yang
- Department of Orthopedics, The Affiliated Changzhou No.2 People’s Hospital with Nanjing Medical University, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, People’s Republic of China
| | - Guangrong Yin
- Department of Orthopedics, The Affiliated Changzhou No.2 People’s Hospital with Nanjing Medical University, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, People’s Republic of China
| | - Yang Chen
- Changzhou Productivity Development Center, Changzhou, People’s Republic of China
| | - Jianchao Gui
- Department of Sports Medicine and Joint Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Chao Xu
- Department of Orthopedics, The Affiliated Changzhou No.2 People’s Hospital with Nanjing Medical University, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, People’s Republic of China
| | - Songwei Lv
- School of Pharmacy, Changzhou University, Changzhou, People’s Republic of China
| |
Collapse
|
18
|
Tsutsui M, Tsunekawa Y, Wada M, Arima A, Onodera A, Nishina M, Nagoya M, Baba Y, Kawai T, Okada T. Enhanced Discriminability of Viral Vectors in Viscous Nanopores. SMALL METHODS 2025:e2401321. [PMID: 39743980 DOI: 10.1002/smtd.202401321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 12/08/2024] [Indexed: 01/04/2025]
Abstract
Achieving safe and efficient gene therapy hinges upon the inspection of genomes enclosed within individual nano-carriers to mitigate potential health risks associated with empty or fragment-filled vectors. Here solid-state nanopore sensing is reported for identifications of intermediate adeno-associated virus (AAV) vectors in liquid. The method exploits the phenomenon of translocation slowdown induced by the viscosity of salt water-organic mixtures. This enables real-time ionic current measurements allowing precise tracking of the electroosmotic flow-driven motions of recombinant AAV vectors in a nanopore. The resulting ionic signals facilitate discrimination between replicative intermediates carrying ssDNA fragments and its full vector counterparts based on genome length-derived subtle nanometer differences in the viral diameters. This rapid and non-destructive means of genome analysis within virus capsids provides a promising avenue toward a robust methodology for ensuring the integrity of AAV vectors before administration.
Collapse
Affiliation(s)
- Makusu Tsutsui
- The Institute of Scientific and Industrial Research, Osaka University, Mihogaoka 8-1, Ibaraki, Osaka, 567-0047, Japan
| | - Yuji Tsunekawa
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Mikako Wada
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Akihide Arima
- Research Institute for Quantum and Chemical Innovation, Institutes of Innovation for Future Society, Nagoya University, Nagoya, 464-8603, Japan
| | - Azusa Onodera
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Masumi Nishina
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Miho Nagoya
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Yoshinobu Baba
- Research Institute for Quantum and Chemical Innovation, Institutes of Innovation for Future Society, Nagoya University, Nagoya, 464-8603, Japan
- Institute of Quantum Life Science, National Institutes for Quantum and Radiological Science and Technology, Anagawa 4-9-1, Inage-ku, Chiba, 263-8555, Japan
| | - Tomoji Kawai
- The Institute of Scientific and Industrial Research, Osaka University, Mihogaoka 8-1, Ibaraki, Osaka, 567-0047, Japan
| | - Takashi Okada
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| |
Collapse
|
19
|
Upadhyaya A, Dasgupta S, Kumar S, Maiti PK. Stability and conformation of DNA-hairpin in cylindrical confinement. Biophys Chem 2025; 316:107331. [PMID: 39427369 DOI: 10.1016/j.bpc.2024.107331] [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: 07/03/2024] [Revised: 09/21/2024] [Accepted: 09/29/2024] [Indexed: 10/22/2024]
Abstract
We conducted atomistic Molecular Dynamics (MD) simulations of DNA-Hairpin molecules encapsulated within Single-Walled Carbon Nanotubes (SWCNTs) at a temperature of 300 K. Our investigation revealed that the structural integrity of the DNA-Hairpin can be maintained within SWCNTs, provided that the diameter of the SWCNT exceeds a critical threshold value. Conversely, when the SWCNT diameter falls below this critical threshold, the DNA-Hairpin undergoes denaturation, even at a temperature of 300 K. The DNA-Hairpin model we employed consisted of a 12-base pair stem and a 3-base loop, and we studied various SWCNTs with different diameters. Our analyses identified a critical SWCNT diameter of 3.39 nm at 300 K. Examination of key structural features, such as hydrogen bonds (H-bonds), van der Waals (vdW) interactions, and other inter-base interactions, demonstrated a significant reduction in the number of H-bonds, vdW energy, and electrostatic energies among the DNA hairpin's constituent bases when confined within narrower SWCNTs (with diameters of 2.84 nm and 3.25 nm). However, it was observed that the increased interaction energy between the DNA-Hairpin and the inner surface of narrower SWCNTs promoted the denaturation of the DNA-Hairpin. In-depth analysis of electrostatic mapping and hydration status further revealed that the DNA-Hairpin experienced inadequate hydration and non-uniform distribution of counter ions within SWCNTs having diameters below the critical value of 3.39 nm. Our inference is that the inappropriate hydration of counter ions, along with their non-uniform spatial distribution around the DNA hairpin, contributes to the denaturation of the molecule within SWCNTs of smaller diameters. For DNA-Hairpin molecules that remained undenatured within SWCNTs, we investigated their mechanical properties, particularly the elastic properties. Our findings demonstrated an increase in the persistence length of the DNA-Hairpin with increasing SWCNT diameter. Additionally, the stretch modulus and torsional stiffness of the DNA-Hairpin were observed to increase as a function of SWCNT diameter, indicating that confinement within SWCNTs enhances the mechanical flexibility of the DNA-Hairpin.
Collapse
Affiliation(s)
- Anurag Upadhyaya
- Centre for Condensed Matter Theory, Department of Physics, Indian Institute of Science, Bangalore 560012, India.
| | - Subhadeep Dasgupta
- Centre for Condensed Matter Theory, Department of Physics, Indian Institute of Science, Bangalore 560012, India
| | - Sanjay Kumar
- Department of Physics, Banaras Hindu University, Varanasi, India
| | - Prabal K Maiti
- Centre for Condensed Matter Theory, Department of Physics, Indian Institute of Science, Bangalore 560012, India.
| |
Collapse
|
20
|
Rivero-Barbarroja G, López-Fernández J, Juárez-Gonzálvez I, Fernández-Clavero C, Di Giorgio C, Vélaz I, Garrido MJ, Benito JM, Ortiz Mellet C, Mendicuti F, Tros de Ilarduya C, García Fernández JM. β-Cyclodextrin-based geometrically frustrated amphiphiles as one-component, cell-specific and organ-specific nucleic acid delivery systems. Carbohydr Polym 2025; 347:122776. [PMID: 39487000 DOI: 10.1016/j.carbpol.2024.122776] [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: 07/03/2024] [Revised: 09/12/2024] [Accepted: 09/18/2024] [Indexed: 11/04/2024]
Abstract
We introduce an innovative β-cyclodextrin (βCD)-prototype for delivering nucleic acids: "geometrically frustrated amphiphiles (GFAs)." GFAs are designed with cationic centers evenly distributed across the primary O6 and secondary O2 positions of the βCD scaffold, while hydrophobic tails are anchored at the seven O3 positions. Such distribution of functional elements differs from Janus-type architectures and enlarges the capacity for accessing strictly monodisperse variants. Changes at the molecular level can then be correlated with preferred self-assembly and plasmid DNA (pDNA) co-assembly behaviors. Specifically, GFAs undergo pH-dependent transition between bilayered to monolayered vesicles or individual molecules. GFA-pDNA nanocomplexes exhibit topological and internal order characteristics that are also a function of the GFA molecular architecture. Notably, adjusting the pKa of the cationic heads and the hydrophilic-hydrophobic balance, pupa-like arrangements implying axial alignments of GFA units flanked by quasi-parallel pDNA segments are preferred. In vitro cell transfection studies revealed remarkable differences in relative performances, which corresponded to distinct organ targeting outcomes in vivo. This allowed for preferential delivery to the liver and lung, kidney or spleen. The results collectively highlight cyclodextrin-based GFAs as a promising class of molecular vectors capable of finely tuning cell and organ transfection selectivity.
Collapse
Affiliation(s)
| | - José López-Fernández
- Instituto de Investigaciones Químicas (IIQ), CSIC - Universidad de Sevilla, 41092 Sevilla, Spain
| | - Inmaculada Juárez-Gonzálvez
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, 31080 Pamplona, Spain
| | - Carlos Fernández-Clavero
- Departamento de Química Analítica, Química Física e Ingeniería Química and Instituto de Investigación Química "Andrés del Rio", Universidad de Alcalá, Spain
| | | | - Itziar Vélaz
- Department of Chemistry, School of Sciences, University of Navarra, 31080 Pamplona, Spain
| | - María J Garrido
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, 31080 Pamplona, Spain
| | - Juan M Benito
- Instituto de Investigaciones Químicas (IIQ), CSIC - Universidad de Sevilla, 41092 Sevilla, Spain
| | - Carmen Ortiz Mellet
- Department of Organic Chemistry, Faculty of Chemistry, University of Seville, 41012 Sevilla, Spain.
| | - Francisco Mendicuti
- Departamento de Química Analítica, Química Física e Ingeniería Química and Instituto de Investigación Química "Andrés del Rio", Universidad de Alcalá, Spain.
| | - Conchita Tros de Ilarduya
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, 31080 Pamplona, Spain.
| | - José M García Fernández
- Instituto de Investigaciones Químicas (IIQ), CSIC - Universidad de Sevilla, 41092 Sevilla, Spain.
| |
Collapse
|
21
|
Chawathe A, Ahire V, Luthra K, Patil B, Garkhal K, Sharma N. Analytical and drug delivery strategies for short peptides: From manufacturing to market. Anal Biochem 2025; 696:115699. [PMID: 39461693 DOI: 10.1016/j.ab.2024.115699] [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: 08/16/2024] [Revised: 10/17/2024] [Accepted: 10/23/2024] [Indexed: 10/29/2024]
Abstract
In recent times, biopharmaceuticals have gained attention because of their tremendous potential to benefit millions of patients globally by treating widespread diseases such as cancer, diabetes and many rare diseases. Short peptides (SP), also termed as oligopeptides, are one such class of biopharmaceuticals, that are majorly involved in efficient functioning of biological systems. Peptide chains that are 2-20 amino acids long are considered as oligopeptides by researchers and are some of the functionally vital compounds with widespread applications including self-assembly material for drug delivery, targeting ligands for precise/specific targeting and other biological uses. Using functionalised biomacromolecules such as short chained peptides, helps in improving pharmacokinetic properties and biodistribution profile of the drug. Apart from this, functionalised SP are being employed as cell penetrating peptides and prodrug to specifically and selectively target tumor sites. In order to minimize any unwanted interaction and adverse effects, the stability and safety of SP should be ensured throughout its development from manufacturing to market. Formulation development and characterization strategies of these potential molecules are described in the following review along with various applications and details of marketed formulations.
Collapse
Affiliation(s)
- Ashwini Chawathe
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research-Ahmedabad, Opposite Air Force Station, Palaj, Gandhinagar, Gujarat, 382355, India
| | - Vishal Ahire
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research-Ahmedabad, Opposite Air Force Station, Palaj, Gandhinagar, Gujarat, 382355, India
| | - Kshitiz Luthra
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Ahmedabad, Opposite Air Force Station, Palaj, Gandhinagar, Gujarat, 382355, India
| | - Bhumika Patil
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Ahmedabad, Opposite Air Force Station, Palaj, Gandhinagar, Gujarat, 382355, India
| | - Kalpna Garkhal
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Ahmedabad, Opposite Air Force Station, Palaj, Gandhinagar, Gujarat, 382355, India.
| | - Nitish Sharma
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research-Ahmedabad, Opposite Air Force Station, Palaj, Gandhinagar, Gujarat, 382355, India.
| |
Collapse
|
22
|
Taeb S, Rostamzadeh D, Amini SM, Rahmati M, Golshekan M, Abedinzade M, Ahmadi E, Neha S, Najafi M. Revolutionizing Cancer Treatment: Harnessing the Power of Mesenchymal Stem Cells for Precise Targeted Therapy in the Tumor Microenvironment. Curr Top Med Chem 2025; 25:243-262. [PMID: 38797895 DOI: 10.2174/0115680266299112240514103048] [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: 12/17/2023] [Revised: 04/03/2024] [Accepted: 04/16/2024] [Indexed: 05/29/2024]
Abstract
In recent years, mesenchymal stem cells (MSCs) have emerged as promising anti-- cancer mediators with the potential to treat several cancers. MSCs have been modified to produce anti-proliferative, pro-apoptotic, and anti-angiogenic molecules that could be effective against a variety of malignancies. Additionally, customizing MSCs with cytokines that stimulate pro-tumorigenic immunity or using them as vehicles for traditional chemical molecules with anti-cancer characteristics. Even though the specific function of MSCs in tumors is still challenged, promising outcomes from preclinical investigations of MSC-based gene therapy for a variety of cancers inspire the beginning of clinical trials. In addition, the tumor microenvironment (TME) could have a substantial influence on normal tissue stem cells, which can affect the treatment outcomes. To overcome the complications of TME in cancer development, MSCs could provide some signs of hope for converting TME into unequivocal therapeutic tools. Hence, this review focuses on engineered MSCs (En-MSCs) as a promising approach to overcoming the complications of TME.
Collapse
Affiliation(s)
- Shahram Taeb
- Department of Radiology, School of Paramedical Sciences, Guilan University of Medical Sciences, Rasht, Iran
| | - Davoud Rostamzadeh
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030, Connecticut, USA
| | - Seyed Mohammad Amini
- Radiation Biology Research center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Rahmati
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Mostafa Golshekan
- Guilan Road Trauma Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Mahmoud Abedinzade
- Department of Medical Physiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Elham Ahmadi
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030, Connecticut, USA
| | - Singh Neha
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030, Connecticut, USA
| | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
23
|
Zanganeh S, Zahedi AM, Sattarzadeh Bardsiri M, Bazi A, Bastanifard M, Shool S, Kouhbananinejad SM, Farsinejad A, Afgar A, Shahabi A, Mirzaei-Parsa MJ. Recent advances and applications of the CRISPR-Cas system in the gene therapy of blood disorders. Gene 2024; 931:148865. [PMID: 39168259 DOI: 10.1016/j.gene.2024.148865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 08/11/2024] [Accepted: 08/14/2024] [Indexed: 08/23/2024]
Affiliation(s)
- Saeed Zanganeh
- Stem Cells and Regenerative Medicine Innovation Center, Kerman University of Medical Sciences, Kerman, Iran; Research Center for Hydatid Disease in Iran, Kerman University of Medical Sciences, Kerman, Iran.
| | - Amir Mohammad Zahedi
- Stem Cells and Regenerative Medicine Innovation Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Mahla Sattarzadeh Bardsiri
- Stem Cells and Regenerative Medicine Innovation Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Ali Bazi
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mahdieh Bastanifard
- Stem Cells and Regenerative Medicine Innovation Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Sanaz Shool
- Stem Cells and Regenerative Medicine Innovation Center, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Alireza Farsinejad
- Stem Cells and Regenerative Medicine Innovation Center, Kerman University of Medical Sciences, Kerman, Iran; Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Ali Afgar
- Research Center for Hydatid Disease in Iran, Kerman University of Medical Sciences, Kerman, Iran
| | - Arman Shahabi
- Stem Cells and Regenerative Medicine Innovation Center, Kerman University of Medical Sciences, Kerman, Iran
| | | |
Collapse
|
24
|
Wang J, Chen Z, Zhao P, Wang Y, Chen J, Lin Q. PDGFR-α shRNA-polyplex for uveal melanoma treatment via EMT mediated vasculogenic mimicry interfering. J Nanobiotechnology 2024; 22:797. [PMID: 39726008 DOI: 10.1186/s12951-024-03077-0] [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: 09/05/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024] Open
Abstract
Up to 50% of individuals with uveal melanoma (UM), a frequent cancer of the eye, pass away from metastases. One of the major challenges in treating UM is the role of receptor tyrosine kinases (RTKs), which mediate the epithelial-mesenchymal transition (EMT) of tumors. RTKs are involved in binding multiple growth factors, leading to angiogenesis and vasculogenic mimicry (VM) phenomena. Currently, most anti-angiogenic drugs have shown a tendency to increase the VM of tumors in clinical trials, resulting in limited efficacy. The existing gap in UM treatment lies in the lack of effective strategies to target RTK-mediated EMT and VM. While some approaches have been attempted, there is still a need for novel therapeutic interventions that can specifically interfere with these processes. This research employed the gene vector PEI-g-PEG to interfere with the platelet derived growth factor-alpha receptor (PDGFR-α)-mediated EMT process, thereby retarding the growth of UM. The cell experiments demonstrated that the gene polyplex exhibited favorable cell uptake and lysosome escape properties, effectively suppressing the expression of PDGFR-α protein and EMT marker proteins and the occurrence of VM phenomenon. In vivo animal studies also inhibited the growth of UM, and PAS assays showed that the treatment reduced the generation of VM in tumor tissue. This study broadens the application of PEI-g-PEG while interfering with the RTK-mediated tumor EMT process with the help of RNAi technology, providing a new idea for tumor reduction research.
Collapse
Affiliation(s)
- Jiahao Wang
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Zhirong Chen
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Peiyi Zhao
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Yajia Wang
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Jiang Chen
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Quankui Lin
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
| |
Collapse
|
25
|
Yang X, Xiao J, Staveness D, Zang X. Efficient mRNA Delivery In Vitro and In Vivo Using a Polycharged Biodegradable Nanomaterial. Int J Mol Sci 2024; 25:13620. [PMID: 39769382 PMCID: PMC11728123 DOI: 10.3390/ijms252413620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/14/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025] Open
Abstract
As RNA rises as one of the most significant modalities for clinical applications and life science research, efficient tools for delivering and integrating RNA molecules into biological systems become essential. Herein, we report a formulation using a polycharged biodegradable nano-carrier, N1-501, which demonstrates superior efficiency and versatility in mRNA encapsulation and delivery in both cell and animal models. N1-501 is a polymeric material designed to function through a facile one-step formulation process suitable for various research settings. Its capability for mRNA transfection is investigated across a wide range of mRNA doses and in different biological models, including 18 tested cell lines and mouse models. This study also comprehensively analyzes N1-501's application for mRNA transfection by examining factors such as buffer composition and pH, incubation condition, and media type. Additionally, N1-501's superior in vivo mRNA transfection capability ensures its potential as an efficient and consistent tool for advancing mRNA-based therapies and genetic research.
Collapse
Affiliation(s)
| | | | | | - Xiaoyu Zang
- N1 Life, Inc., 446 S Hillview Dr, Milpitas, CA 95035, USA
| |
Collapse
|
26
|
Tzimou K, Catalán-Tatjer D, Nielsen LK, Lavado-García J. Unlocking DOE potential by selecting the most appropriate design for rAAV optimization. Mol Ther Methods Clin Dev 2024; 32:101329. [PMID: 39296857 PMCID: PMC11406035 DOI: 10.1016/j.omtm.2024.101329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 08/23/2024] [Indexed: 09/21/2024]
Abstract
Producing recombinant adeno-associated virus (rAAV) for gene therapy via triple transfection is an intricate process involving many cellular interactions. Each of the different elements encoded in the three required plasmids-pHelper, pRepCap, and pGOI-plays a distinct role, affecting different cellular pathways when producing rAAVs. The required expression balance emphasizes the critical need to fine-tune the concentration of all these different elements. The use of design of experiments (DOE) to find optimal ratios is a powerful method to streamline the process. However, the choice of the DOE method and design construction is crucial to avoid misleading results. In this work, we examined and compared four distinct DOE approaches: rotatable central composite design (RCCD), Box-Behnken design (BBD), face-centered central composite design (FCCD), and mixture design (MD). We compared the abilities of the different models to predict optimal ratios and interactions among the plasmids and the transfection reagent. Our findings revealed that blocking is essential to reduce the variability caused by uncontrolled random effects and that MD coupled with FCCD outperformed all other approaches, improving volumetric productivity 109-fold. These outcomes underscore the importance of selecting a model that can effectively account for the biological context, ultimately yielding superior results in optimizing rAAV production.
Collapse
Affiliation(s)
- Konstantina Tzimou
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - David Catalán-Tatjer
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Lars K Nielsen
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jesús Lavado-García
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| |
Collapse
|
27
|
Zhang X, Zhang Y, Rong X, Tang C, Liu H, Yue L, Su R, Wang Y, Qi W. Alkylated RALA-Derived Peptides for Efficient Gene Delivery. Biomacromolecules 2024; 25:8046-8057. [PMID: 39535929 DOI: 10.1021/acs.biomac.4c01355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
RALA is an amphipathic cationic peptide demonstrated to be a low-toxicity and high-efficiency delivery platform for the systemic delivery of nucleic acid therapeutics. This work reports three RALA-derived peptides modified with N-terminal palmitic acid, engineered through amino acid substitutions and truncated sequences. All three peptides have good nucleic acid encapsulation, release and uptake, biocompatibility, and endolysosome escape. The siRNA transfection efficiency is about 90%, and the silencing rate of GA (C16-GLFWHHHARLARALARHLARALRA) exceeds that of lipofectamine 2000 (siRNA concentration = 50 nM). Truncating the peptide chain while retaining a certain amount of arginine ensures an effective particle size. Replacing glutamic acid with three histidines ensures an effective zeta potential and accelerates the endosome escape process through the proton sponge phenomenon. Introducing phenylalanine enhances the carrier-cell interaction. We believe that they are powerful carriers of siRNA therapy and may have good application prospects in treating various diseases.
Collapse
Affiliation(s)
- Xuelin Zhang
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Yexi Zhang
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Xi Rong
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Chuanmei Tang
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Huiye Liu
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Lei Yue
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Rongxin Su
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin 300072, P. R. China
- Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin 300072, P. R. China
| | - Yuefei Wang
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China
- Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin 300072, P. R. China
- Beyonpep Biotechnology Limited, Tianjin 300110, P. R. China
| | - Wei Qi
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin 300072, P. R. China
- Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin 300072, P. R. China
| |
Collapse
|
28
|
Haque MA, Shrestha A, Mikelis CM, Mattheolabakis G. Comprehensive analysis of lipid nanoparticle formulation and preparation for RNA delivery. Int J Pharm X 2024; 8:100283. [PMID: 39309631 PMCID: PMC11415597 DOI: 10.1016/j.ijpx.2024.100283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 08/21/2024] [Accepted: 09/07/2024] [Indexed: 09/25/2024] Open
Abstract
Nucleic acid-based therapeutics are a common approach that is increasingly popular for a wide spectrum of diseases. Lipid nanoparticles (LNPs) are promising delivery carriers that provide RNA stability, with strong transfection efficiency, favorable and tailorable pharmacokinetics, limited toxicity, and established translatability. In this review article, we describe the lipid-based delivery systems, focusing on lipid nanoparticles, the need of their use, provide a comprehensive analysis of each component, and highlight the advantages and disadvantages of the existing manufacturing processes. We further summarize the ongoing and completed clinical trials utilizing LNPs, indicating important aspects/questions worth of investigation, and analyze the future perspectives of this significant and promising therapeutic approach.
Collapse
Affiliation(s)
- Md. Anamul Haque
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Archana Shrestha
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Constantinos M. Mikelis
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras 26504, Greece
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - George Mattheolabakis
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| |
Collapse
|
29
|
Polash SA, Poddar A, Ahmady F, Kannourakis G, Jayachandran A, Shukla R. Impact of Ligand Concentration on the Properties of Nucleic-Acid-Encapsulated MOFs and Inflammation Modulation in Prostate Cancer Cells. ACS APPLIED BIO MATERIALS 2024; 7:7635-7645. [PMID: 39497260 DOI: 10.1021/acsabm.4c01185] [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] [Indexed: 11/19/2024]
Abstract
The zeolitic imidazolate framework (ZIF) is one of the most explored metal-organic-framework-based systems for nucleic acid delivery to cancer cells. Current nucleic acid delivery tools exhibit several drawbacks, such as high manufacturing costs, endosomal entrapment, toxicity, and immunogenicity. However, the biomimetic mineralization of Zn-based ZIFs offers a low-cost and facile encapsulation of nucleic acids at room temperature in aqueous conditions. The efficiency of nucleic acid delivery and its subsequent impact on inflammation in cells are influenced by the physicochemical properties of the material. The imidazole content determines the formation and crystallinity of ZIF, and an optimal ratio ensures the formation of well-defined and highly crystalline structures. In this study, a series of siRNA-encapsulated ZIFs (siRNA@ZIF) were systematically prepared by varying ligand-to-metal (L/M) molar ratios. Our study demonstrates that variations in ligand concentrations influence the crystalline structures, particle size, and shape of siRNA@ZIF particles. At low L/M, two-dimensional siRNA@ZIF particles form with a size of 1 μm. As the L/M ratio increases gradually, the particle size decreases, resulting in three-dimensional particles ∼200 nm in size. We also observed better stability of siRNA@ZIF in water prepared using high L/M values and time-dependent cellular uptake by the cells. Additionally, no significant impact of the biocomposites on inflammation was found, indicating the lack of an unwanted immune response and nonimmunotoxic nature over longer periods (96 h). These findings highlight the necessity of fine-tuning ligand concentrations and synthesis chemistry in designing efficient and optimal ZIF-based systems as versatile delivery platforms for nucleic acids.
Collapse
Affiliation(s)
- Shakil Ahmed Polash
- Ian Potter NanoBiosensing Facility, NanoBiotechnology Research Laboratory, School of Science, RMIT University, Melbourne, VIC 3001, Australia
- Centre for Advanced Materials and Industrial Chemistry, RMIT University, Melbourne, VIC 3001, Australia
| | - Arpita Poddar
- Ian Potter NanoBiosensing Facility, NanoBiotechnology Research Laboratory, School of Science, RMIT University, Melbourne, VIC 3001, Australia
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3350, Australia
| | - Farah Ahmady
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3350, Australia
| | | | | | - Ravi Shukla
- Ian Potter NanoBiosensing Facility, NanoBiotechnology Research Laboratory, School of Science, RMIT University, Melbourne, VIC 3001, Australia
- Centre for Advanced Materials and Industrial Chemistry, RMIT University, Melbourne, VIC 3001, Australia
| |
Collapse
|
30
|
Wang D, Tang L, Chen M, Gong Z, Fan C, Qu H, Liu Y, Shi L, Mo Y, Wang Y, Yan Q, Chen P, Xiang B, Liao Q, Zeng Z, Li G, Jiang W, Wu SX, Xiong W. Nanocarriers Targeting Circular RNA ADARB1 Boost Radiosensitivity of Nasopharyngeal Carcinoma through Synergically Promoting Ferroptosis. ACS NANO 2024; 18:31055-31075. [PMID: 39467079 DOI: 10.1021/acsnano.4c07676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
Nasopharyngeal carcinoma (NPC) is a common malignant tumor of the head and neck, prevalent in regions such as Southern China and Southeast Asia. Radiotherapy serves as the primary clinical treatment for this carcinoma. However, resistance to radiotherapy is a fundamental cause of treatment failure and patient mortality, with the underlying mechanisms yet to be fully elucidated. We identified a recently characterized circular RNA, circADARB1, which is markedly upregulated in NPC tissues and closely associated with poor prognosis and radiotherapy resistance. Both in vitro and in vivo experiments demonstrated that circADARB1 inhibited ferroptosis, thereby inducing radiotherapy resistance in NPC cells. Building on these findings, we synthesized a biomimetic nanomaterial consisting of semiconducting polymer nanoparticles wrapped in cell membranes, designed to deliver both siRNA targeting circADARB1 and iron ions. The application of this nanomaterial not only efficiently suppressed the expression of circADARB1 and boosted intracellular iron concentrations, but also enhanced ferroptosis induced by radiotherapy, improving the radiosensitivity of NPC cells. Furthermore, our study revealed that circADARB1 upregulated the expression of heat shock protein HSP90B1, which repaired misfolded SLC7A11 and GPX4 proteins triggered by radiotherapy, thereby preserving their stability and biological functions. Mechanistically, SLC7A11 facilitated cysteine transportation into cells and glutathione synthesis, while GPX4 employed glutathione to mitigate intracellular lipid peroxidation induced by radiotherapy, shielding cells from oxidative damage and inhibiting ferroptosis, and ultimately leading to radiotherapy resistance in NPC cells. Our investigation elucidates molecular mechanisms with substantial clinical relevance, highlights the promising application prospects of nanotechnology in precision cancer therapy.
Collapse
Affiliation(s)
- Dan Wang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410078, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha 410078, Hunan, China
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha 410078, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha 410078, Hunan, China
| | - Le Tang
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha 410078, Hunan, China
| | - Mingjian Chen
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha 410078, Hunan, China
| | - Zhaojian Gong
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital of Central South University, Changsha 410078, Hunan, China
| | - Chunmei Fan
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha 410078, Hunan, China
| | - Hongke Qu
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha 410078, Hunan, China
| | - Yixuan Liu
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha 410078, Hunan, China
| | - Lei Shi
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital of Central South University, Changsha 410078, Hunan, China
| | - Yongzhen Mo
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha 410078, Hunan, China
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha 410078, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha 410078, Hunan, China
| | - Yumin Wang
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha 410078, Hunan, China
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha 410078, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha 410078, Hunan, China
| | - Qijia Yan
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha 410078, Hunan, China
| | - Pan Chen
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410078, Hunan, China
| | - Bo Xiang
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha 410078, Hunan, China
- FuRong Laboratory, Changsha 410078, Hunan, China
| | - Qianjin Liao
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410078, Hunan, China
| | - Zhaoyang Zeng
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha 410078, Hunan, China
| | - Guiyuan Li
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha 410078, Hunan, China
| | - Weihong Jiang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha 410078, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha 410078, Hunan, China
- FuRong Laboratory, Changsha 410078, Hunan, China
| | - Steven X Wu
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410078, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha 410078, Hunan, China
- Department of Chemistry and Center for Fluorinated Functional Materials, University of South Dakota, Vermillion, South Dakota 57069, United States
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410078, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha 410078, Hunan, China
- FuRong Laboratory, Changsha 410078, Hunan, China
| |
Collapse
|
31
|
Cheng L, Zhu Y, Ma J, Aggarwal A, Toh WH, Shin C, Sangpachatanaruk W, Weng G, Kumar R, Mao HQ. Machine Learning Elucidates Design Features of Plasmid Deoxyribonucleic Acid Lipid Nanoparticles for Cell Type-Preferential Transfection. ACS NANO 2024; 18:28735-28747. [PMID: 39375194 PMCID: PMC11512640 DOI: 10.1021/acsnano.4c07615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
To broaden the accessibility of cell and gene therapies, it is essential to develop and optimize nonviral, cell type-preferential gene carriers such as lipid nanoparticles (LNPs). While high-throughput screening (HTS) approaches have proven effective in accelerating LNP discovery, they are often costly, labor-intensive, and do not consistently yield actionable design rules that direct screening efforts toward the most relevant chemical and formulation parameters. In this study, we employed a machine learning (ML) workflow, utilizing well-curated plasmid DNA LNP transfection data sets across six cell types, to extract compositional and chemical insights from HTS studies. Our approach achieved prediction errors averaging between 5 and 10%, depending on the cell type. By applying SHapley Additive exPlanations to our ML models, we uncovered key composition-function relationships that govern cell type-preferential LNP transfection efficiency. Notably, we identified consistent LNP composition parameters that enhance in vitro transfection efficiency across diverse cell types, including a helper lipid molar percentage of charged lipids between 9 and 50% and the inclusion of cationic/zwitterionic helper lipids. Additionally, several parameters were found to modulate cell type-preferentiality, such as the total molar percentage of ionizable and helper lipids, N/P ratio, PEGylated lipid molar percentage of uncharged lipids, and hydrophobicity of the helper lipid. This study leverages HTS of compositionally diverse LNP libraries combined with ML analysis to elucidate the interactions between lipid components in LNP formulations, providing insights that contribute to the design of LNP compositions tailored for cell type-preferential transfection.
Collapse
Affiliation(s)
- Leonardo Cheng
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21218, United States
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - Yining Zhu
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21218, United States
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - Jingyao Ma
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Department of Materials Science and Engineering, Whiting School of Engineering. Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Ataes Aggarwal
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21218, United States
- Department of Computer Science, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Wu Han Toh
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Department of Computer Science, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Department of Biology, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Charles Shin
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21218, United States
| | - Will Sangpachatanaruk
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Department of Computer Science, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Gene Weng
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Department of Biophysics, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Ramya Kumar
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, Colorado 80401, United States
| | - Hai-Quan Mao
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21218, United States
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Department of Materials Science and Engineering, Whiting School of Engineering. Johns Hopkins University, Baltimore, Maryland 21218, United States
| |
Collapse
|
32
|
Hoch CC, Hachani K, Han Y, Schmidl B, Wirth M, Multhoff G, Bashiri Dezfouli A, Wollenberg B. The future of interleukin gene therapy in head and neck cancers. Expert Opin Biol Ther 2024; 24:1057-1073. [PMID: 39291462 DOI: 10.1080/14712598.2024.2405568] [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: 06/26/2024] [Accepted: 09/13/2024] [Indexed: 09/19/2024]
Abstract
INTRODUCTION Head and neck cancer (HNC), primarily head and neck squamous cell carcinomas, originates from the squamous epithelium in areas like the oral cavity, lip, larynx, and oropharynx. With high morbidity impacting critical functions, combined treatments like surgery, radiation, and chemotherapy often fall short in advanced stages, highlighting the need for innovative therapies. AREAS COVERED This review critically evaluates interleukin (IL) gene therapy for treating HNC. The discussion extends to key ILs in HNC, various gene therapy techniques and delivery methods. We particularly focus on the application of IL-2, IL-12, and IL-24 gene therapies, examining their mechanisms and outcomes in preclinical studies and clinical trials. The final sections address IL gene therapy challenges in HNC, exploring solutions and critically assessing future therapeutic directions. EXPERT OPINION Despite advancements in genomic and immunotherapy, significant challenges in HNC treatment persist, primarily due to the immunosuppressive nature of the tumor microenvironment and the adverse effects of current therapies. The therapeutic efficacy of IL gene therapy hinges on overcoming these hurdles through refined delivery methods that ensure targeted, tumor-specific gene expression. Future strategies should focus on refining gene delivery methods and combining IL gene therapy with other treatments to optimize efficacy and minimize toxicity.
Collapse
Affiliation(s)
- Cosima C Hoch
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Khouloud Hachani
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Yu Han
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Benedikt Schmidl
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Markus Wirth
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Gabriele Multhoff
- Central Institute for Translational Cancer Research, Technical University of Munich (TranslaTUM), Munich, Germany
- Department of Radiation Oncology, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Ali Bashiri Dezfouli
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
- Central Institute for Translational Cancer Research, Technical University of Munich (TranslaTUM), Munich, Germany
- Department of Radiation Oncology, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Barbara Wollenberg
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| |
Collapse
|
33
|
Narita M, Kohata A, Kageyama T, Watanabe H, Aikawa K, Kawaguchi D, Morihiro K, Okamoto A, Okazoe T. Fluorocarbon-DNA Conjugates for Enhanced Cellular Delivery: Formation of a Densely Packed DNA Nano-Assembly. Chembiochem 2024; 25:e202400436. [PMID: 38858172 DOI: 10.1002/cbic.202400436] [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: 05/31/2024] [Accepted: 06/10/2024] [Indexed: 06/12/2024]
Abstract
Forming nano-assemblies is essential for delivering DNA conjugates into cells, with the DNA density in the nano-assembly playing an important role in determining the uptake efficiency. In this study, we developed a strategy for the facile synthesis of DNA strands bearing perfluoroalkyl (RF) groups (RF-DNA conjugates) and investigated how they affect cellular uptake. An RF-DNA conjugate bearing a long RF group at the DNA terminus forms a nano-assembly with a high DNA density, which results in greatly enhanced cellular uptake. The uptake mechanism is mediated by clathrin-dependent endocytosis. The use of RF groups to densely assemble negatively charged DNA is a useful strategy for designing drug delivery carriers.
Collapse
Grants
- 22UT0019 Ministry of Education, Culture, Sports, Science, and Technology (MEXT)
- 23UT0211 Ministry of Education, Culture, Sports, Science, and Technology (MEXT)
- 23UT1115 Ministry of Education, Culture, Sports, Science, and Technology (MEXT)
- 20K05460 JSPS KAKENHI Grant-in-Aid for Scientific Research
- 23K13852 Grant-in-Aid for Early-Career Scientists
Collapse
Affiliation(s)
- Minako Narita
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 2-11-16 Yayoi, Bunkyo-ku, Tokyo, 113-0032, Japan
| | - Ai Kohata
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 2-11-16 Yayoi, Bunkyo-ku, Tokyo, 113-0032, Japan
- Current address: School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Yokohama-shi, Kanagawa, 226-8501, Japan
| | - Taiichi Kageyama
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 2-11-16 Yayoi, Bunkyo-ku, Tokyo, 113-0032, Japan
| | - Honoka Watanabe
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 2-11-16 Yayoi, Bunkyo-ku, Tokyo, 113-0032, Japan
| | - Kohsuke Aikawa
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 2-11-16 Yayoi, Bunkyo-ku, Tokyo, 113-0032, Japan
| | - Daisuke Kawaguchi
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 2-11-16 Yayoi, Bunkyo-ku, Tokyo, 113-0032, Japan
| | - Kunihiko Morihiro
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Akimitsu Okamoto
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Takashi Okazoe
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 2-11-16 Yayoi, Bunkyo-ku, Tokyo, 113-0032, Japan
- Yokohama Technical Center, AGC Inc., 1-1 Suehiro-cho, Tsurumi-ku, Yokohama, 230-0045, Japan
| |
Collapse
|
34
|
Korzun T, Moses AS, Diba P, Sattler AL, Olson B, Taratula OR, Pejovic T, Marks DL, Taratula O. Development and Perspectives: Multifunctional Nucleic Acid Nanomedicines for Treatment of Gynecological Cancers. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2301776. [PMID: 37518857 PMCID: PMC10827528 DOI: 10.1002/smll.202301776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 07/11/2023] [Indexed: 08/01/2023]
Abstract
Gynecological malignancies are a significant cause of morbidity and mortality across the globe. Due to delayed presentation, gynecological cancer patients are often referred late in the disease's course, resulting in poor outcomes. A considerable number of patients ultimately succumb to chemotherapy-resistant disease, which reoccurs at advanced stages despite treatment interventions. Although efforts have been devoted to developing therapies that demonstrate reduced resistance to chemotherapy and enhanced toxicity profiles, current clinical outcomes remain unsatisfactory due to treatment resistance and unfavorable off-target effects. Consequently, innovative biological and nanotherapeutic approaches are imperative to strengthen and optimize the therapeutic arsenal for gynecological cancers. Advancements in nanotechnology-based therapies for gynecological malignancies offer significant advantages, including reduced toxicity, expanded drug circulation, and optimized therapeutic dosing, ultimately leading to enhanced treatment effectiveness. Recent advances in nucleic acid therapeutics using microRNA, small interfering RNA, and messenger RNA provide novel approaches for cancer therapeutics. Effective single-agent and combinatorial nucleic acid therapeutics for gynecological malignancies have the potential to transform cancer treatment by giving safer, more tailored approaches than conventional therapies. This review highlights current preclinical studies that effectively exploit these approaches for the treatment of gynecological malignant tumors and malignant ascites.
Collapse
Affiliation(s)
- Tetiana Korzun
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, OR, 97201, USA
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 SW Bond Avenue Portland, Portland, OR, 97239, USA
- Medical Scientist Training Program, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
- Papé Family Pediatric Research Institute, Oregon Health & Science University, SW Sam Jackson Park Rd, Mail Code L481, Portland, OR, 97239, USA
| | - Abraham S Moses
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, OR, 97201, USA
| | - Parham Diba
- Medical Scientist Training Program, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
- Papé Family Pediatric Research Institute, Oregon Health & Science University, SW Sam Jackson Park Rd, Mail Code L481, Portland, OR, 97239, USA
| | - Ariana L Sattler
- Papé Family Pediatric Research Institute, Oregon Health & Science University, SW Sam Jackson Park Rd, Mail Code L481, Portland, OR, 97239, USA
- Knight Cancer Institute, Oregon Health & Science University, 2720 S Moody Avenue, Portland, Oregon, 97201, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, 2730 S Moody Avenue, Portland, OR, 97201, USA
| | - Brennan Olson
- Mayo Clinic Department of Otolaryngology-Head and Neck Surgery, 200 First St. SW, Rochester, MN, 55905, USA
| | - Olena R Taratula
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, OR, 97201, USA
| | - Tanja Pejovic
- Departments of Obstetrics and Gynecology and Pathology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Daniel L Marks
- Papé Family Pediatric Research Institute, Oregon Health & Science University, SW Sam Jackson Park Rd, Mail Code L481, Portland, OR, 97239, USA
- Knight Cancer Institute, Oregon Health & Science University, 2720 S Moody Avenue, Portland, Oregon, 97201, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, 2730 S Moody Avenue, Portland, OR, 97201, USA
| | - Oleh Taratula
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, OR, 97201, USA
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 SW Bond Avenue Portland, Portland, OR, 97239, USA
| |
Collapse
|
35
|
Khan M. Polymers as Efficient Non-Viral Gene Delivery Vectors: The Role of the Chemical and Physical Architecture of Macromolecules. Polymers (Basel) 2024; 16:2629. [PMID: 39339093 PMCID: PMC11435517 DOI: 10.3390/polym16182629] [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: 08/26/2024] [Revised: 09/13/2024] [Accepted: 09/15/2024] [Indexed: 09/30/2024] Open
Abstract
Gene therapy is the technique of inserting foreign genetic elements into host cells to achieve a therapeutic effect. Although gene therapy was initially formulated as a potential remedy for specific genetic problems, it currently offers solutions for many diseases with varying inheritance patterns and acquired diseases. There are two major groups of vectors for gene therapy: viral vector gene therapy and non-viral vector gene therapy. This review examines the role of a macromolecule's chemical and physical architecture in non-viral gene delivery, including their design and synthesis. Polymers can boost circulation, improve delivery, and control cargo release through various methods. The prominent examples discussed include poly-L-lysine, polyethyleneimine, comb polymers, brush polymers, and star polymers, as well as hydrogels and natural polymers and their modifications. While significant progress has been made, challenges still exist in gene stabilization, targeting specificity, and cellular uptake. Overcoming cytotoxicity, improving delivery efficiency, and utilizing natural polymers and hybrid systems are vital factors for prospects. This comprehensive review provides an illuminating overview of the field, guiding the way toward innovative non-viral-based gene delivery solutions.
Collapse
Affiliation(s)
- Majad Khan
- Department of Chemistry, King Fahd University of Petroleum & Minerals KFUPM, Dahran 31261, Saudi Arabia
- Interdisciplinary Research Center for Hydrogen Technologies and Carbon Management (IRC-HTCM), King Fahd University of Petroleum & Minerals KFUPM, Dahran 31261, Saudi Arabia
- Interdisciplinary Research Center for Refining and Advanced Chemicals (IRC-CRAC), King Fahd University of Petroleum & Minerals (KFUPM), Dhahran 31261, Saudi Arabia
| |
Collapse
|
36
|
Singh D. Beyond the membrane: Exploring non-viral methods for mitochondrial gene delivery. Mitochondrion 2024; 78:101922. [PMID: 38897397 DOI: 10.1016/j.mito.2024.101922] [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: 04/10/2024] [Revised: 05/22/2024] [Accepted: 06/15/2024] [Indexed: 06/21/2024]
Abstract
Mitochondrial disorders, stemming from mutations in mitochondrial DNA (mtDNA), present a significant therapeutic challenge due to their complex pathophysiology and broad spectrum of clinical manifestations. Traditional gene therapy approaches, primarily reliant on viral vectors, face obstacles such as potential immunogenicity, insertional mutagenesis, and the specificity of targeting mtDNA. This review delves into non-viral methods for mitochondrial gene delivery, emerging as a promising alternative to overcome these limitations. Focusing on lipid-based nanoparticles, polymer-based vectors, and mitochondrial-targeted peptides, the mechanisms of action, advantages, and current applications in treating mitochondrial diseases was well elucidated. Non-viral vectors offer several benefits, including reduced immunogenicity, enhanced safety profiles, and the flexibility to carry a wide range of genetic material. We examine case studies where these methods have been applied, highlighting their potential in correcting pathogenic mtDNA mutations and mitigating disease phenotypes. Despite their promise, challenges such as delivery efficiency, specificity, and long-term expression stability persist. The review underscores the need for ongoing research to refine these delivery systems carry a wide range of genetic material. We examine case studies where these methods settings. As we advance our understanding of mitochondrial biology and gene delivery technologies, non-viral methods hold the potential to revolutionize the treatment of mitochondrial disorders, offering hope for therapies that can precisely target and correct the underlying genetic defects.
Collapse
Affiliation(s)
- Dilpreet Singh
- University Institute of Pharma Sciences, Chandigarh University, Gharuan, Mohali 140413, India; University Centre for Research and Development, Chandigarh University, Gharuan, Mohali 140413, India.
| |
Collapse
|
37
|
Albukhaty S, Sulaiman GM, Al-Karagoly H, Mohammed HA, Hassan AS, Alshammari AAA, Ahmad AM, Madhi R, Almalki FA, Khashan KS, Jabir MS, Yusuf M, Al-aqbi ZT, Sasikumar P, Khan RA. Iron oxide nanoparticles: The versatility of the magnetic and functionalized nanomaterials in targeting drugs, and gene deliveries with effectual magnetofection. J Drug Deliv Sci Technol 2024; 99:105838. [DOI: 10.1016/j.jddst.2024.105838] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
38
|
Habibizadeh M, Lotfollahzadeh S, Mahdavi P, Mohammadi S, Tavallaei O. Nanoparticle-mediated gene delivery of TRAIL to resistant cancer cells: A review. Heliyon 2024; 10:e36057. [PMID: 39247341 PMCID: PMC11379606 DOI: 10.1016/j.heliyon.2024.e36057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 09/10/2024] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), also known as APO2L, has emerged as a highly potential anticancer agent because of its capacity to effectively trigger apoptosis in tumor cells by specifically binding to either of its death receptors (DR4 or DR5) while having no adverse effects on normal cells. Nevertheless, its practical use has been hindered by its inefficient pharmacokinetics characteristics, the challenges involved in its administration and delivery to targeted cells, and the resistance exhibited by most cancer cells towards TRAIL. Gene therapy, as a promising approach would be able to potentially circumvent TRAIL-based cancer therapy challenges mainly through localized TRAIL expression and generating a bystander impact. Among different strategies, using nanoparticles in TRAIL gene delivery allows for precise targeting, and overcoming TRAIL resistance by combination therapy. In this review, we go over potential mechanisms by which cancer cells achieve resistance to TRAIL and provide an overview of different carriers for delivering of the TRAIL gene to resistant cancer cells, focusing on different types of nanoparticles utilized in this context. We will also explore the challenges, and investigate future perspectives of this nanomedicine approach for cancer therapy.
Collapse
Affiliation(s)
- Mina Habibizadeh
- Regenerative Medicine Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Shima Lotfollahzadeh
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Parisa Mahdavi
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Soheila Mohammadi
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Omid Tavallaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
39
|
Zhang L, Lou W, Wang J. Advances in nucleic acid therapeutics: structures, delivery systems, and future perspectives in cancer treatment. Clin Exp Med 2024; 24:200. [PMID: 39196428 PMCID: PMC11358240 DOI: 10.1007/s10238-024-01463-4] [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: 06/19/2024] [Accepted: 08/08/2024] [Indexed: 08/29/2024]
Abstract
Cancer has emerged as a significant threat to human health. Nucleic acid therapeutics regulate the gene expression process by introducing exogenous nucleic acid fragments, offering new possibilities for tumor remission and even cure. Their mechanism of action and high specificity demonstrate great potential in cancer treatment. However, nucleic acid drugs face challenges such as low stability and limited ability to cross physiological barriers in vivo. To address these issues, various nucleic acid delivery vectors have been developed to enhance the stability and facilitate precise targeted delivery of nucleic acid drugs within the body. In this review article, we primarily introduce the structures and principles of nucleic acid drugs commonly used in cancer therapy, as well as their cellular uptake and intracellular transportation processes. We focus on the various vectors commonly employed in nucleic acid drug delivery, highlighting their research progress and applications in recent years. Furthermore, we propose potential trends and prospects of nucleic acid drugs and their carriers in the future.
Collapse
Affiliation(s)
- Leqi Zhang
- Department of Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Wenting Lou
- Department of Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Jianwei Wang
- Department of Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China.
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, 2nd Affiliated Hospital, Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, 310009, China.
| |
Collapse
|
40
|
Vijayakumar KK, Manoharan D, Subbarayan R, Shrestha R, Harshavardhan S. Construction of pVAX-1-based linear covalently closed vector with improved transgene expression. Mol Biol Rep 2024; 51:934. [PMID: 39180671 DOI: 10.1007/s11033-024-09856-0] [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: 04/24/2024] [Accepted: 08/09/2024] [Indexed: 08/26/2024]
Abstract
INTRODUCTION This study presents a Mammalian Linear Expression System (MLES), a linear covalently closed (LCC) vector based on pVAX-1. The purpose of this system was to improve gene expression in mammalian cells and to test the efficacy of MLES in transient transfection and transgene expression using in vitro and in vivo models. Additionally, we aimed to evaluate potential inflammatory responses in vivo. MATERIALS AND METHODS MLES was developed by modifying pVAX-1, and the construct was confirmed by gel electrophoresis. Lipofectamine®2000 was used to assess the transfection efficiency and expression of MLES in various cell lines. In vivo studies were conducted in mice injected with MLES/EGFP, and the resulting transfection efficiency, gene expression, and inflammatory responses were analyzed. RESULTS MLES exhibited higher transfection efficiency and expression levels compared to pVAX-1 when tested on HEK-293, CHO-K1, and NIH-3T3 cells. When tested in vivo, MLES/EGFP showed elevated expression in the heart, kidney, liver, and spleen compared with pVAX-1/EGFP. Minimal changes are observed in the lungs. Additionally, MLES induced a reduced inflammatory response in mice compared with pVAX-1/EGFP. CONCLUSIONS MLES offer improved transfection efficiency and reduced inflammation, representing a significant advancement in gene therapy and recombinant protein production. Further research on MLES-mediated gene expression and immune modulation will enhance gene therapy strategies.
Collapse
Affiliation(s)
- Kevin Kumar Vijayakumar
- Department of Molecular Microbiology, School of Biotechnology, Madurai Kamaraj University, Palkalai Nagar, Madurai, Tamil Nadu, 625021, India
| | - Devaprakash Manoharan
- Department of Molecular Microbiology, School of Biotechnology, Madurai Kamaraj University, Palkalai Nagar, Madurai, Tamil Nadu, 625021, India
| | - Rajasekaran Subbarayan
- Centre for Advanced Biotherapeutic and Regenerative Medicine, Faculty of Research, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
- Centre for Herbal Pharmacology and Environmental Sustainability, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| | - Rupendra Shrestha
- Research and Collaboration, Anka Analytica, Melbourne, Australia
- External Consultant, Independent Researcher, Pittsfield, MA, USA
| | - Shakila Harshavardhan
- Department of Molecular Microbiology, School of Biotechnology, Madurai Kamaraj University, Palkalai Nagar, Madurai, Tamil Nadu, 625021, India.
| |
Collapse
|
41
|
Vinent Genestar J, Auvity S, Christiansen N, Ekelund H, Huys L, McNulty HBØ, Pani M, Pires V, Pourroy B, Stoner N. European Association of Hospital Pharmacists (EAHP) guidance on the pharmacy handling of in vivo gene therapy medicinal products. Eur J Hosp Pharm 2024; 31:390-402. [PMID: 38821721 DOI: 10.1136/ejhpharm-2023-004062] [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: 12/22/2023] [Accepted: 05/06/2024] [Indexed: 06/02/2024] Open
Abstract
Gene therapy is becoming increasingly prevalent, with new gene therapy medicinal products (GTMPs) being approved for use every year. Hospital pharmacists are expected to prepare and dispense these products, but there is substantial heterogeneity in the availability of up-to-date, practical guidance at a national level in Europe. Many institutions have no or very limited experience in handling GTMPs. As such, there is a need for updated, practical guidance to aid hospital pharmacy teams in developing institutional standard operating procedures (SOPs) for the safe handling of GTMPs across the entire workflow. Here, we present the European Association of Hospital Pharmacists' updated guidance on the handling of GTMPs, developed by a team of recognised experts from around Europe. Each aspect of the GTMP handling process is addressed, including receipt and storage, dispensing and reconstitution, transportation, administration, waste disposal, decontamination of spills and accidental exposure. A series of figures are provided to aid the development of practical workflows. This guidance document is intended as a framework to help develop institutional SOPs and should always be used in conjunction with local regulations.
Collapse
Affiliation(s)
| | - Sylvain Auvity
- Necker Hospital, Assistance Publique-Hôpitaux de Paris, University of Paris, Paris, France
| | - Nanna Christiansen
- Women's and Children's Services, Guy's and St Thomas' NHS Trust, London, UK
| | - Heidi Ekelund
- ATMP Center, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Liesbeth Huys
- Ghent University Hospital, Gent, Oost-Vlaanderen, Belgium
| | | | - Marcello Pani
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Lazio, Italy
| | - Vera Pires
- Portugese Institute of Oncology, Lisbon, Portugal
| | - Bertrand Pourroy
- Oncopharma Unit-Pharmacy department, La Timone Unversity Teaching Hospital, Marseille, France
| | - Nicola Stoner
- Pharmacy, Oxford University Hospitals NHS Trust, Oxford, Oxfordshire, UK
| |
Collapse
|
42
|
Kumar J, Karim A, Sweety UH, Sarma H, Nurunnabi M, Narayan M. Bioinspired Approaches for Central Nervous System Targeted Gene Delivery. ACS APPLIED BIO MATERIALS 2024; 7:4975-4997. [PMID: 38100377 DOI: 10.1021/acsabm.3c00842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2023]
Abstract
Disorders of the central nervous system (CNS) which include a wide range of neurodegenerative and neurological conditions have become a serious global issue. The presence of CNS barriers poses a significant challenge to the progress of designing effective therapeutic delivery systems, limiting the effectiveness of drugs, genes, and other therapeutic agents. Natural nanocarriers present in biological systems have inspired researchers to design unique delivery systems through biomimicry. As natural resource derived delivery systems are more biocompatible, current research has been focused on the development of delivery systems inspired by bacteria, viruses, fungi, and mammalian cells. Despite their structural potential and extensive physiological function, making them an excellent choice for biomaterial engineering, the delivery of nucleic acids remains challenging due to their instability in biological systems. Similarly, the efficient delivery of genetic material within the tissues of interest remains a hurdle due to a lack of selectivity and targeting ability. Considering that gene therapies are the holy grail for intervention in diseases, including neurodegenerative disorders such as Alzheimer's disease, Parkinson's Disease, and Huntington's disease, this review centers around recent advances in bioinspired approaches to gene delivery for the prevention of CNS disorders.
Collapse
Affiliation(s)
- Jyotish Kumar
- Department of Chemistry and Biochemistry, The University of Texas at El Paso (UTEP), El Paso, Texas 79968, United States
| | - Afroz Karim
- Department of Chemistry and Biochemistry, The University of Texas at El Paso (UTEP), El Paso, Texas 79968, United States
| | - Ummy Habiba Sweety
- Environmental Science and Engineering, The University of Texas at El Paso (UTEP), El Paso, Texas 79968, United States
| | - Hemen Sarma
- Bioremediation Technology Research Group, Department of Botany, Bodoland University, Rangalikhata, Deborgaon, 783370, Kokrajhar (BTR), Assam, India
| | - Md Nurunnabi
- The Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas at El Paso, El Paso, Texas 79968, United States
| | - Mahesh Narayan
- Department of Chemistry and Biochemistry, The University of Texas at El Paso (UTEP), El Paso, Texas 79968, United States
| |
Collapse
|
43
|
Shahalaei M, Azad AK, Sulaiman WMAW, Derakhshani A, Mofakham EB, Mallandrich M, Kumarasamy V, Subramaniyan V. A review of metallic nanoparticles: present issues and prospects focused on the preparation methods, characterization techniques, and their theranostic applications. Front Chem 2024; 12:1398979. [PMID: 39206442 PMCID: PMC11351095 DOI: 10.3389/fchem.2024.1398979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 07/04/2024] [Indexed: 09/04/2024] Open
Abstract
Metallic nanoparticles (MNPs) have garnered significant attention due to their ability to improve the therapeutic index of medications by reducing multidrug resistance and effectively delivering therapeutic agents through active targeting. In addition to drug delivery, MNPs have several medical applications, including in vitro and in vivo diagnostics, and they improve the biocompatibility of materials and nutraceuticals. MNPs have several advantages in drug delivery systems and genetic manipulation, such as improved stability and half-life in circulation, passive or active targeting into the desired target selective tissue, and gene manipulation by delivering genetic materials. The main goal of this review is to provide current information on the present issues and prospects of MNPs in drug and gene delivery systems. The current study focused on MNP preparation methods and their characterization by different techniques, their applications to targeted delivery, non-viral vectors in genetic manipulation, and challenges in clinical trial translation.
Collapse
Affiliation(s)
- Mona Shahalaei
- Biomaterial Group, Nanotechnology and Advanced Materials Department, Materials and Energy Research Center, Karaj, Iran
| | - Abul Kalam Azad
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University College of MAIWP International (UCMI), Kuala Lumpur, Malaysia
| | - Wan Mohd Azizi Wan Sulaiman
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University College of MAIWP International (UCMI), Kuala Lumpur, Malaysia
| | - Atefeh Derakhshani
- Department of Tissue Engineering and Applied Cell Sciences, Faculty of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Elmira Banaee Mofakham
- Biomaterial Group, Nanotechnology and Advanced Materials Department, Materials and Energy Research Center, Karaj, Iran
| | - Mireia Mallandrich
- Department of Pharmacy, Pharmaceutical Technology and Physical-Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
| | - Vinoth Kumarasamy
- Department of Parasitology and Medical Entomology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Vetriselvan Subramaniyan
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Sunway, Malaysia
| |
Collapse
|
44
|
Oliveira IS, Silva SG, Gomes AC, Real Oliveira MECD, Vale MLCD, Marques EF. Cationic Serine-Based Gemini Surfactant:Monoolein Aggregates as Viable and Efficacious Agents for DNA Complexation and Compaction: A Cytotoxicity and Physicochemical Assessment. J Funct Biomater 2024; 15:224. [PMID: 39194661 DOI: 10.3390/jfb15080224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 08/29/2024] Open
Abstract
Cationic gemini surfactants have emerged as potential gene delivery agents as they can co-assemble with DNA due to a strong electrostatic association. Commonly, DNA complexation is enhanced by the inclusion of a helper lipid (HL), which also plays a key role in transfection efficiency. The formation of lipoplexes, used as non-viral vectors for transfection, through electrostatic and hydrophobic interactions is affected by various physicochemical parameters, such as cationic surfactant:HL molar ratio, (+/-) charge ratio, and the morphological structure of the lipoplexes. Herein, we investigated the DNA complexation ability of mixtures of serine-based gemini surfactants, (nSer)2N5, and monoolein (MO) as a helper lipid. The micelle-forming serine surfactants contain long lipophilic chains (12 to 18 C atoms) and a five CH2 spacer, both linked to the nitrogen atoms of the serine residues by amine linkages. The (nSer)2N5:MO aggregates are non-cytotoxic up to 35-90 µM, depending on surfactant and surfactant/MO mixing ratio, and in general, higher MO content and longer surfactant chain length tend to promote higher cell viability. All systems efficaciously complex DNA, but the (18Ser)2N5:MO one clearly stands as the best-performing one. Incorporating MO into the serine surfactant system affects the morphology and size distribution of the formed mixed aggregates. In the low concentration regime, gemini-MO systems aggregate in the form of vesicles, while at high concentrations the formation of a lamellar liquid crystalline phase is observed. This suggests that lipoplexes might share a similar bilayer-based structure.
Collapse
Affiliation(s)
- Isabel S Oliveira
- CIQUP (Centro de Investigação em Química da Universidade do Porto), IMS (Institute of Molecular Sciences), Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Sandra G Silva
- LAQV-REQUIMTE (Laboratório Associado para a Química Verde-Rede Química e Tecnologia), Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Andreia C Gomes
- CBMA (Centro de Biologia Molecular e Ambiental), Departamento de Biologia, Campus de Gualtar, Universidade do Minho, 4710-057 Braga, Portugal
| | - M Elisabete C D Real Oliveira
- CFUM (Center of Physics), Departamento de Física, Universidade do Minho, Campos de Gualtar, 4710-057 Braga, Portugal
| | - M Luísa C do Vale
- LAQV-REQUIMTE (Laboratório Associado para a Química Verde-Rede Química e Tecnologia), Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Eduardo F Marques
- CIQUP (Centro de Investigação em Química da Universidade do Porto), IMS (Institute of Molecular Sciences), Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| |
Collapse
|
45
|
He W, Kirmizialtin S. Mechanism of Cationic Lipid Induced DNA Condensation: Lipid-DNA Coordination and Divalent Cation Charge Fluctuations. Biomacromolecules 2024; 25:4819-4830. [PMID: 39011747 PMCID: PMC11323003 DOI: 10.1021/acs.biomac.4c00192] [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: 02/11/2024] [Revised: 06/30/2024] [Accepted: 07/01/2024] [Indexed: 07/17/2024]
Abstract
The condensation of nucleic acids by lipids is a widespread phenomenon in biology with crucial implications for drug delivery. However, the mechanisms of DNA assembly in lipid bilayers remain insufficiently understood due to challenges in measuring and assessing each component's contribution in the lipid-DNA-cation system. This study uses all-atom molecular dynamics simulations to investigate DNA condensation in cationic lipid bilayers. Our exhaustive exploration of the thermodynamic factors reveals unique roles for phospholipid head groups and cations. We observed that bridging cations between lipid and DNA drastically reduce charges, while mobile magnesium cations "ping-ponging" between double strands create charge fluctuations. While the first factor stabilizes the DNA-lipid complex, the latter creates attractive forces to induce the spontaneous condensation of DNAs. This novel mechanism not only sheds light on the current data regarding cationic lipid-induced DNA condensation but also provides potential design strategies for creating efficient gene delivery vectors for drug delivery.
Collapse
Affiliation(s)
- Weiwei He
- Chemistry
Program, Science Division, New York University
Abu Dhabi, Abu Dhabi, United Arab Emirates
- Department
of Chemistry, New York University, New York, New York 10003, United States
| | - Serdal Kirmizialtin
- Chemistry
Program, Science Division, New York University
Abu Dhabi, Abu Dhabi, United Arab Emirates
- Department
of Chemistry, New York University, New York, New York 10003, United States
| |
Collapse
|
46
|
Piao X, Li D, Liu H, Guo Q, Yu Y. Advances in Gene and Cellular Therapeutic Approaches for Huntington's Disease. Protein Cell 2024:pwae042. [PMID: 39121016 DOI: 10.1093/procel/pwae042] [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: 04/26/2024] [Indexed: 08/11/2024] Open
Abstract
Huntington's disease (HD) is an inherited neurodegenerative disorder caused by the abnormal expansion of CAG trinucleotide repeats in the Huntingtin gene (HTT) located on chromosome 4. It is transmitted in an autosomal dominant manner and is characterized by motor dysfunction, cognitive decline, and emotional disturbances. To date, there are no curative treatments for HD have been developed; current therapeutic approaches focus on symptom relief and comprehensive care through coordinated pharmacological and non-pharmacological methods to manage the diverse phenotypes of the disease. International clinical guidelines for the treatment of HD are continually being revised in an effort to enhance care within a multidisciplinary framework. Additionally, innovative gene and cell therapy strategies are being actively researched and developed to address the complexities of the disorder and improve treatment outcomes. This review endeavours to elucidate the current and emerging gene and cell therapy strategies for HD, offering a detailed insight into the complexities of the disorder and looking forward to future treatment paradigms. Considering the complexity of the underlying mechanisms driving HD, a synergistic treatment strategy that integrates various factors-such as distinct cell types, epigenetic patterns, genetic components, and methods to improve the cerebral microenvironment-may significantly enhance therapeutic outcomes. In the future, we eagerly anticipate ongoing innovations in interdisciplinary research that will bring profound advancements and refinements in the treatment of HD.
Collapse
Affiliation(s)
- Xuejiao Piao
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | - Dan Li
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | - Hui Liu
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | - Qing Guo
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | - Yang Yu
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| |
Collapse
|
47
|
Feng Q, Li Q, Zhou H, Wang Z, Lin C, Jiang Z, Liu T, Wang D. CRISPR technology in human diseases. MedComm (Beijing) 2024; 5:e672. [PMID: 39081515 PMCID: PMC11286548 DOI: 10.1002/mco2.672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 07/01/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Gene editing is a growing gene engineering technique that allows accurate editing of a broad spectrum of gene-regulated diseases to achieve curative treatment and also has the potential to be used as an adjunct to the conventional treatment of diseases. Gene editing technology, mainly based on clustered regularly interspaced palindromic repeats (CRISPR)-CRISPR-associated protein systems, which is capable of generating genetic modifications in somatic cells, provides a promising new strategy for gene therapy for a wide range of human diseases. Currently, gene editing technology shows great application prospects in a variety of human diseases, not only in therapeutic potential but also in the construction of animal models of human diseases. This paper describes the application of gene editing technology in hematological diseases, solid tumors, immune disorders, ophthalmological diseases, and metabolic diseases; focuses on the therapeutic strategies of gene editing technology in sickle cell disease; provides an overview of the role of gene editing technology in the construction of animal models of human diseases; and discusses the limitations of gene editing technology in the treatment of diseases, which is intended to provide an important reference for the applications of gene editing technology in the human disease.
Collapse
Affiliation(s)
- Qiang Feng
- Laboratory Animal CenterCollege of Animal ScienceJilin UniversityChangchunChina
- Research and Development CentreBaicheng Medical CollegeBaichengChina
| | - Qirong Li
- Laboratory Animal CenterCollege of Animal ScienceJilin UniversityChangchunChina
| | - Hengzong Zhou
- Laboratory Animal CenterCollege of Animal ScienceJilin UniversityChangchunChina
| | - Zhan Wang
- Laboratory Animal CenterCollege of Animal ScienceJilin UniversityChangchunChina
| | - Chao Lin
- School of Grain Science and TechnologyJilin Business and Technology CollegeChangchunChina
| | - Ziping Jiang
- Department of Hand and Foot SurgeryThe First Hospital of Jilin UniversityChangchunChina
| | - Tianjia Liu
- Research and Development CentreBaicheng Medical CollegeBaichengChina
| | - Dongxu Wang
- Laboratory Animal CenterCollege of Animal ScienceJilin UniversityChangchunChina
- Department of Hand and Foot SurgeryThe First Hospital of Jilin UniversityChangchunChina
| |
Collapse
|
48
|
Dooley J, Hughes JG, Needham EJ, Palios KA, Liston A. The potential of gene delivery for the treatment of traumatic brain injury. J Neuroinflammation 2024; 21:183. [PMID: 39069631 DOI: 10.1186/s12974-024-03156-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: 04/01/2024] [Accepted: 06/17/2024] [Indexed: 07/30/2024] Open
Abstract
Therapeutics for traumatic brains injuries constitute a global unmet medical need. Despite the advances in neurocritical care, which have dramatically improved the survival rate for the ~ 70 million patients annually, few treatments have been developed to counter the long-term neuroinflammatory processes and accompanying cognitive impairments, frequent among patients. This review looks at gene delivery as a potential therapeutic development avenue for traumatic brain injury. We discuss the capacity of gene delivery to function in traumatic brain injury, by producing beneficial biologics within the brain. Gene delivery modalities, promising vectors and key delivery routes are discussed, along with the pathways that biological cargos could target to improve long-term outcomes for patients. Coupling blood-brain barrier crossing with sustained local production, gene delivery has the potential to convert proteins with useful biological properties, but poor pharmacodynamics, into effective therapeutics. Finally, we review the limitations and health economics of traumatic brain injury, and whether future gene delivery approaches will be viable for patients and health care systems.
Collapse
Affiliation(s)
- James Dooley
- Department of Pathology, University of Cambridge, Cambridge, UK.
| | - Jasmine G Hughes
- Department of Pathology, University of Cambridge, Cambridge, UK
- Department of Clinical Neuroscience, University of Cambridge, Cambridge, UK
| | - Edward J Needham
- Department of Clinical Neuroscience, University of Cambridge, Cambridge, UK
| | | | - Adrian Liston
- Department of Pathology, University of Cambridge, Cambridge, UK
| |
Collapse
|
49
|
Yan X, Chen Q. Polyamidoamine Dendrimers: Brain-Targeted Drug Delivery Systems in Glioma Therapy. Polymers (Basel) 2024; 16:2022. [PMID: 39065339 PMCID: PMC11280609 DOI: 10.3390/polym16142022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/03/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Glioma is the most common primary intracranial tumor, which is formed by the malignant transformation of glial cells in the brain and spinal cord. It has the characteristics of high incidence, high recurrence rate, high mortality and low cure rate. The treatments for glioma include surgical removal, chemotherapy and radiotherapy. Due to the obstruction of the biological barrier of brain tissue, it is difficult to achieve the desired therapeutic effects. To address the limitations imposed by the brain's natural barriers and enhance the treatment efficacy, researchers have effectively used brain-targeted drug delivery systems (DDSs) in glioma therapy. Polyamidoamine (PAMAM) dendrimers, as branched macromolecular architectures, represent promising candidates for studies in glioma therapy. This review focuses on PAMAM-based DDSs in the treatment of glioma, highlighting their physicochemical characteristics, structural properties as well as an overview of the toxicity and safety profiles.
Collapse
Affiliation(s)
- Xinyi Yan
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China;
| | - Qi Chen
- Interdisciplinary Institute for Medical Engineering, Fuzhou University, Fuzhou 350108, China
| |
Collapse
|
50
|
Singh N, Singh A, Dhanka M, Bhatia D. DNA functionalized programmable hybrid biomaterials for targeted multiplexed applications. J Mater Chem B 2024. [PMID: 38973587 DOI: 10.1039/d4tb00287c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/09/2024]
Abstract
With the advent of DNA nanotechnology, DNA-based biomaterials have emerged as a unique class of materials at the center of various biological advances. Owing to DNA's high modification capacity via programmable Watson-Crick base-pairing, DNA structures of desired design with increased complexity have been developed. However, the limited scalability, along with poor mechanical properties, high synthesis costs, and poor stability, reduced the adaptability of DNA-based materials to complex biological applications. DNA-based hybrid biomaterials were designed to overcome these limitations by conjugating DNA with functional materials. Today, DNA-based hybrid materials have attracted significant attention in biological engineering with broad application prospects in biomedicine, clinical diagnosis, and nanodevices. Here, we summarize the recent advances in DNA-based hybrid materials with an in-depth understanding of general molecular design principles, functionalities, and applications. Finally, the challenges and prospects associated with DNA-based hybrid materials are discussed at the end of this review.
Collapse
Affiliation(s)
- Nihal Singh
- Discipline of Bioengineering, Indian Institute of Technology Gandhinagar, Gujarat, India, 382355.
| | - Ankur Singh
- Discipline of Bioengineering, Indian Institute of Technology Gandhinagar, Gujarat, India, 382355.
| | - Mukesh Dhanka
- Discipline of Bioengineering, Indian Institute of Technology Gandhinagar, Gujarat, India, 382355.
| | - Dhiraj Bhatia
- Discipline of Bioengineering, Indian Institute of Technology Gandhinagar, Gujarat, India, 382355.
| |
Collapse
|