1
|
López Espinar A, Mulder LM, Elkhashab M, Khan Z, Czarnocki-Cieciura M, Aburto MR, Vucen S, Kowalski PS. Tailoring Alkyl Side Chains of Ionizable Amino-Polyesters for Enhanced In Vivo mRNA Delivery. ACS APPLIED BIO MATERIALS 2025; 8:3958-3971. [PMID: 40293247 DOI: 10.1021/acsabm.5c00116] [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] [Indexed: 04/30/2025]
Abstract
Lipid nanoparticles (LNPs) containing ionizable lipids are the most clinically advanced platform for mRNA delivery, but their application beyond the liver remains challenging. Polymer-lipid hybrid nanoparticles offer a promising alternative, combining the synthetic versatility and unique properties of polymers with the biocompatibility of lipid excipients. While the significance of alkyl tail design is well-recognized for ionizable lipids, the impact of the polymer side chain composition on interactions with lipid excipients, mRNA delivery efficacy, and tissue specificity remains poorly understood. Here, we focus on a class of ionizable amino-polyesters (APEs) that exhibit features desired for potential clinical applications, including narrow molecular weight distribution and a good safety profile, and investigate the effect of polymer side chain composition on the formulation of APE lipid nanoparticles (APE-LNPs) for mRNA delivery. A library of 36 APEs was synthesized via ring-opening polymerization of chemically diverse tertiary amino-alcohols and lactone monomers with distinct alkyl side chain compositions, including variations in length and unsaturation. We show that optimal alkyl side chain length is critical for the assembly of stable mRNA nanoparticles and efficient mRNA delivery both in vitro and in vivo. Top-performing APE-LNPs display superior delivery efficacy in vitro and in extrahepatic tissues compared to benchmark LNPs, including DLin-MC3-DMA ionizable lipid. The polymer chain composition affects the tissue selectivity of APE-LNPs, with shorter side chains (4-5 carbons) effectively targeting the spleen and lungs, while longer chains (7-9 carbons) show enhanced liver delivery. We also explored the relevance of lipid excipients in APE-LNPs, demonstrating the essential role of unsaturated phospholipids in enhancing cellular uptake and mRNA delivery, and the limited relevance of cholesterol. These findings provide valuable insights into the design of polymers for use in the LNP context, which could aid the development of polymeric alternatives to ionizable lipids and expand the utility of mRNA LNP technology to nonliver tissues.
Collapse
Affiliation(s)
| | - Lianne M Mulder
- School of Pharmacy, University College Cork, Cork T12 K8AF, Ireland
| | | | - Zahra Khan
- School of Pharmacy, University College Cork, Cork T12 K8AF, Ireland
| | - Mariusz Czarnocki-Cieciura
- Laboratory of Protein Structure, International Institute of Molecular and Cell Biology, Warsaw 02-109, Poland
| | - Maria R Aburto
- APC Microbiome Ireland, University College Cork, Cork T12 K8AF, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork T12 K8AF, Ireland
| | - Sonja Vucen
- SSPC, Research Ireland Centre for Pharmaceuticals, School of Pharmacy, University College Cork, Cork T12 K8AF, Ireland
| | - Piotr S Kowalski
- School of Pharmacy, University College Cork, Cork T12 K8AF, Ireland
- APC Microbiome Ireland, University College Cork, Cork T12 K8AF, Ireland
| |
Collapse
|
2
|
Honrath S, Burger M, Leroux JC. Hurdles to healing: Overcoming cellular barriers for viral and nonviral gene therapy. Int J Pharm 2025; 674:125470. [PMID: 40112901 DOI: 10.1016/j.ijpharm.2025.125470] [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: 01/20/2025] [Revised: 02/26/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
Gene delivery offers great potential for treating various diseases, yet its success requires overcoming several biological barriers. These hurdles span from extracellular degradation, reaching the target cells, and inefficient cellular uptake to endosomal entrapment, cytoplasmic transport, nuclear entry, and transcription limitations. Viruses and non-viral vectors deal with these barriers via different mechanisms. Viral vectors, such as adenoviruses, adeno-associated viruses, and lentiviruses use natural mechanisms to efficiently deliver genetic material but face limitations including immunogenicity, cargo capacity, and production complexity. Nonviral vectors, including lipid nanoparticles, polymers, and protein-based systems, offer scalable and safer alternatives but often fall short in overcoming intracellular barriers and achieving high transfection efficiencies. Recent advancements in vector engineering have partially overcome several of these challenges. Ionizable lipids improve endosomal escape while minimizing toxicity. Biodegradable polymers balance efficacy with safety, and engineered protein systems, inspired by viral or bacterial entry mechanisms, integrate multifunctionality for enhanced delivery. Despite these advances, challenges, particularly in achieving robust in vivo translatability, scalability, and reduced immunogenicity, remain. This review synthesizes current knowledge of cellular barriers and the approaches to overcome them, providing a roadmap for designing more efficient gene delivery systems. By addressing these barriers, the field can advance toward safer, and more effective therapies.
Collapse
Affiliation(s)
- Steffen Honrath
- ETH Zurich, Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, Vladimir-Prelog-Weg 3, 8093 Zurich, Switzerland
| | - Michael Burger
- ETH Zurich, Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, Vladimir-Prelog-Weg 3, 8093 Zurich, Switzerland.
| | - Jean-Christophe Leroux
- ETH Zurich, Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, Vladimir-Prelog-Weg 3, 8093 Zurich, Switzerland.
| |
Collapse
|
3
|
Liu M, Wang Y, Zhang Y, Hu D, Tang L, Zhou B, Yang L. Landscape of small nucleic acid therapeutics: moving from the bench to the clinic as next-generation medicines. Signal Transduct Target Ther 2025; 10:73. [PMID: 40059188 PMCID: PMC11891339 DOI: 10.1038/s41392-024-02112-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/23/2024] [Accepted: 12/13/2024] [Indexed: 03/17/2025] Open
Abstract
The ability of small nucleic acids to modulate gene expression via a range of processes has been widely explored. Compared with conventional treatments, small nucleic acid therapeutics have the potential to achieve long-lasting or even curative effects via gene editing. As a result of recent technological advances, efficient small nucleic acid delivery for therapeutic and biomedical applications has been achieved, accelerating their clinical translation. Here, we review the increasing number of small nucleic acid therapeutic classes and the most common chemical modifications and delivery platforms. We also discuss the key advances in the design, development and therapeutic application of each delivery platform. Furthermore, this review presents comprehensive profiles of currently approved small nucleic acid drugs, including 11 antisense oligonucleotides (ASOs), 2 aptamers and 6 siRNA drugs, summarizing their modifications, disease-specific mechanisms of action and delivery strategies. Other candidates whose clinical trial status has been recorded and updated are also discussed. We also consider strategic issues such as important safety considerations, novel vectors and hurdles for translating academic breakthroughs to the clinic. Small nucleic acid therapeutics have produced favorable results in clinical trials and have the potential to address previously "undruggable" targets, suggesting that they could be useful for guiding the development of additional clinical candidates.
Collapse
Affiliation(s)
- Mohan Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yusi Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yibing Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Die Hu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lin Tang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Bailing Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Li Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
4
|
Zhang Y, Huang Q, Lei F, Qian W, Zhang C, Wang Q, Liu C, Ji H, Wang F. Exploring New Bioorthogonal Catalysts: Scaffold Diversity in Catalysis for Chemical Biology. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2404431. [PMID: 39921286 PMCID: PMC11884534 DOI: 10.1002/advs.202404431] [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: 04/25/2024] [Revised: 01/11/2025] [Indexed: 02/10/2025]
Abstract
Bioorthogonal catalysis has revolutionized the field of chemical biology by enabling selective and controlled chemical transformations within living systems. Research has converged on the development of innovative catalyst scaffolds, seeking to broaden the scope of bioorthogonal reactions, boost their efficiency, and surpass the limitations of conventional catalysts. This review provides a comprehensive overview of the latest advancements in bioorthogonal catalyst research based on different scaffold materials. Through an in-depth analysis of fabrication strategies and applications of bioorthogonal catalysts, this review discusses the design principles, mechanisms of action, and applications of these novel catalysts in chemical biology. Current challenges and future directions in exploring the scaffold diversity are also highlighted. The integration of diverse catalyst scaffolds offers exciting prospects for precise manipulation of biomolecules and the development of innovative therapeutic strategies in chemical biology. In addition, the review fills in the gaps in previous reviews, such as in fully summarizing the presented scaffold materials applied in bioorthogonal catalysts, emphasizing the potential impact on advancing bioorthogonal chemistry, and offering prospects for future development in this field.
Collapse
Affiliation(s)
- Yan Zhang
- Institute of Special Environmental MedicineNantong UniversityNantong226019China
| | - Qizhen Huang
- School of Public HealthNantong UniversityNantong226019China
| | - Fang Lei
- School of Public HealthNantong UniversityNantong226019China
| | - Wanlong Qian
- Institute of Special Environmental MedicineNantong UniversityNantong226019China
| | - Chengfeng Zhang
- Institute of Special Environmental MedicineNantong UniversityNantong226019China
| | - Qi Wang
- School of Public HealthNantong UniversityNantong226019China
| | - Chaoqun Liu
- School of PharmacyHenan UniversityKaifeng475004China
| | - Haiwei Ji
- School of Public HealthNantong UniversityNantong226019China
| | - Faming Wang
- School of Public HealthNantong UniversityNantong226019China
| |
Collapse
|
5
|
Jia W, Wu Y, Xie Y, Yu M, Chen Y. Advanced Polymeric Nanoparticles for Cancer Immunotherapy: Materials Engineering, Immunotherapeutic Mechanism and Clinical Translation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2413603. [PMID: 39797474 DOI: 10.1002/adma.202413603] [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: 09/10/2024] [Revised: 12/13/2024] [Indexed: 01/13/2025]
Abstract
Cancer immunotherapy, which leverages immune system components to treat malignancies, has emerged as a cornerstone of contemporary therapeutic strategies. Yet, critical concerns about the efficacy and safety of cancer immunotherapies remain formidable. Nanotechnology, especially polymeric nanoparticles (PNPs), offers unparalleled flexibility in manipulation-from the chemical composition and physical properties to the precision control of nanoassemblies. PNPs provide an optimal platform to amplify the potency and minimize systematic toxicity in a broad spectrum of immunotherapeutic modalities. In this comprehensive review, the basics of polymer chemistry, and state-of-the-art designs of PNPs from a physicochemical standpoint for cancer immunotherapy, encompassing therapeutic cancer vaccines, in situ vaccination, adoptive T-cell therapies, tumor-infiltrating immune cell-targeted therapies, therapeutic antibodies, and cytokine therapies are delineated. Each immunotherapy necessitates distinctively tailored design strategies in polymeric nanoplatforms. The extensive applications of PNPs, and investigation of their mechanisms of action for enhanced efficacy are particularly focused on. The safety profiles of PNPs and clinical research progress are discussed. Additionally, forthcoming developments and emergent trends of polymeric nano-immunotherapeutics poised to transform cancer treatment paradigms into clinics are explored.
Collapse
Affiliation(s)
- Wencong Jia
- School of Medicine, Shanghai University, Shanghai China, 200444, China
| | - Ye Wu
- School of Medicine, Shanghai University, Shanghai China, 200444, China
| | - Yujie Xie
- School of Medicine, Shanghai University, Shanghai China, 200444, China
| | - Meihua Yu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, China
- Shanghai Institute of Materdicine, Shanghai, 200051, China
| |
Collapse
|
6
|
Hu Y, Luo Z, Bao Y. Trends in Photopolymerization 3D Printing for Advanced Drug Delivery Applications. Biomacromolecules 2025; 26:85-117. [PMID: 39625843 PMCID: PMC11733939 DOI: 10.1021/acs.biomac.4c01004] [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: 07/19/2024] [Revised: 11/21/2024] [Accepted: 11/21/2024] [Indexed: 01/14/2025]
Abstract
Since its invention in the 1980s, photopolymerization-based 3D printing has attracted significant attention for its capability to fabricate complex microstructures with high precision, by leveraging light patterning to initiate polymerization and cross-linking in liquid resin materials. Such precision makes it particularly suitable for biomedical applications, in particular, advanced and customized drug delivery systems. This review summarizes the latest advancements in photopolymerization 3D printing technology and the development of biocompatible and/or biodegradable materials that have been used or shown potential in the field of drug delivery. The drug loading methods and release characteristics of the 3D printing drug delivery systems are summarized. Importantly, recent trends in the drug delivery applications based on photopolymerization 3D printing, including oral formulations, microneedles, implantable devices, microrobots and recently emerging systems, are analyzed. In the end, the challenges and opportunities in photopolymerization 3D printing for customized drug delivery are discussed.
Collapse
Affiliation(s)
- Yu Hu
- Department
of Biomedical Engineering, Southern University
of Science and Technology, Shenzhen 518055, Guangdong, P.R. China
| | - Zhi Luo
- Department
of Biomedical Engineering, Southern University
of Science and Technology, Shenzhen 518055, Guangdong, P.R. China
| | - Yinyin Bao
- Department
of Chemistry and Applied Biosciences, ETH
Zurich, Vladimir-Prelog-Weg 1, 8093 Zurich, Switzerland
- Department
of Chemistry, Faculty of Science, University
of Helsinki, 00014 Helsinki, Finland
| |
Collapse
|
7
|
Wang H, Cheng Y. Polymers for mRNA Delivery. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2025; 17:e70002. [PMID: 39763235 DOI: 10.1002/wnan.70002] [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: 05/30/2024] [Revised: 10/22/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025]
Abstract
mRNA delivery has emerged as a transformative approach in biotechnology and medicine, offering a versatile platform for the development of novel therapeutics. Unlike traditional small molecule drugs or protein-based biologics, mRNA therapeutics have the unique ability to direct cells to generate therapeutic proteins, allowing for precise modulation of biological processes. The delivery of mRNA into target cells is a critical step in realizing the therapeutic potential of this technology. In this review, our focus is on the latest advancements in designing functional polymers to achieve efficient mRNA delivery. Biodegradable polymers and low molecular weight polymers in addressing the balance in mRNA binding and release are summarized. Benefiting from the excellent performance of lipid nanoparticles in mRNA delivery, polymer/lipid hybrid nanostructures are also included. Finally, the challenges and future prospects in the development of polymer-based mRNA delivery systems are discussed.
Collapse
Affiliation(s)
- Hui Wang
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou, China
| | - Yiyun Cheng
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| |
Collapse
|
8
|
Rocher EE, Luly KM, Tzeng SY, Sunshine JC, Green JJ. Efficient Polymeric Nanoparticle Gene Delivery Enabled Via Tri- and Tetrafunctional Branching. Biomacromolecules 2024; 25:7260-7273. [PMID: 39466232 DOI: 10.1021/acs.biomac.4c00954] [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: 10/29/2024]
Abstract
Poly(β-amino ester) (PBAE) nanoparticles (NPs) show great promise for nonviral gene delivery. Recent studies suggest branched PBAEs (BPBAEs) offer advantages over linear counterparts, but the effect of polymer structure has not been well investigated across many chemical constituents. Here, a library of BPBAEs was synthesized with tri- and tetrafunctional branching. These polymers self-assemble with DNA to form highly cationic, monodisperse NPs with notably small size (∼50 nm). Optimal transfection occurred with polymer structures that featured moderate PBAE branching, enabling complete DNA encapsulation, rapid NP uptake, and robust expression at low DNA doses and polymer amounts. Optimized NPs enabled efficient DNA delivery to diverse cell types in vitro while maintaining high cellular viability, demonstrating significant improvements over a well-performing linear PBAE counterpart. BPBAEs also facilitated efficient mRNA and siRNA delivery, highlighting the versatility of these structures and demonstrating the broad utility of BPBAE NPs as vectors for nucleic acid delivery.
Collapse
|
9
|
Zhang S, Fang H, Tian H. Recent Advances in Degradable Biomedical Polymers for Prevention, Diagnosis and Treatment of Diseases. Biomacromolecules 2024; 25:7015-7057. [PMID: 39420482 DOI: 10.1021/acs.biomac.4c01193] [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: 10/19/2024]
Abstract
Biomedical polymers play a key role in preventing, diagnosing, and treating diseases, showcasing a wide range of applications. Their unique advantages, such as rich source, good biocompatibility, and excellent modifiability, make them ideal biomaterials for drug delivery, biomedical imaging, and tissue engineering. However, conventional biomedical polymers suffer from poor degradation in vivo, increasing the risks of bioaccumulation and potential toxicity. To address these issues, degradable biomedical polymers can serve as an alternative strategy in biomedicine. Degradable biomedical polymers can efficiently relieve bioaccumulation in vivo and effectively reduce patient burden in disease management. This review comprehensively introduces the classification and properties of biomedical polymers and the recent research progress of degradable biomedical polymers in various diseases. Through an in-depth analysis of their classification, properties, and applications, we aim to provide strong guidance for promoting basic research and clinical translation of degradable biomedical polymers.
Collapse
Affiliation(s)
- Siting Zhang
- State Key Laboratory of Physical Chemistry of Solid Surfaces, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Huapan Fang
- State Key Laboratory of Physical Chemistry of Solid Surfaces, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
- Shenzhen Research Institute of Xiamen University, Shenzhen 518000, China
| | - Huayu Tian
- State Key Laboratory of Physical Chemistry of Solid Surfaces, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| |
Collapse
|
10
|
Liu X, Zhou C, Xie Q, Xia L, Liu L, Bao W, Lin H, Xiong X, Zhang H, Zheng Z, Zhao J, Liang W. Recent advances in layer-by-layer assembly scaffolds for co-delivery of bioactive molecules for bone regeneration: an updated review. J Transl Med 2024; 22:1001. [PMID: 39501263 PMCID: PMC11539823 DOI: 10.1186/s12967-024-05809-0] [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: 06/07/2024] [Accepted: 10/25/2024] [Indexed: 11/08/2024] Open
Abstract
Orthopedic implants have faced challenges in treating bone defects due to various factors, including inadequate osseointegration, oxidative stress, bacterial infection, immunological rejection, and poor individualized treatment. These challenges profoundly affect both the results of treatment and patients' daily lives. There is great promise for the layer-by-layer (LbL) assembly method in tissue engineering. The method primarily relies on electrostatic attraction and entails the consecutive deposition of electrolyte complexes with opposite charges onto a substrate, leading to the formation of homogeneous single layers that can be quickly deposited to produce nanolayer films. LbL has attracted considerable interest as a coating technology because of its ease of production, cost-effectiveness, and capability to apply diverse biomaterial coatings without compromising the primary bio-functional properties of the substrate materials. This review will look into the fundamentals and evolution of LbL in orthopedics, provide an analysis of the chemical strategy used to prepare bone implants with LbL and introduce the application of LbL bone implants in orthopedics over recent years. Among the many potential uses of LbL, such as the implementation of sustained-release and programmed drug delivery, which in turn promotes the osseointegration and the development of new blood vessels, as well as antibacterial, antioxidant, and other similar applications. In addition, we offer a thorough examination of cell behavior and biomaterial interaction to facilitate the advancement of next-generation LbL films for tissue engineering.
Collapse
Affiliation(s)
- Xiankun Liu
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, 355 Xinqiao Road, Dinghai District, Zhoushan, 316000, Zhejiang, People's Republic of China
| | - Chao Zhou
- Department of Orthopedics, Zhoushan Guanghua Hospital, Zhoushan, 316000, Zhejiang, China
| | - Qiong Xie
- Department of Orthopedics, Zhoushan Guanghua Hospital, Zhoushan, 316000, Zhejiang, China
| | - Linying Xia
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Lu Liu
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Wenwen Bao
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Hongming Lin
- Department of Orthopedics, Zhoushan Guanghua Hospital, Zhoushan, 316000, Zhejiang, China
| | - Xiaochun Xiong
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, 355 Xinqiao Road, Dinghai District, Zhoushan, 316000, Zhejiang, People's Republic of China
| | - Hao Zhang
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, 355 Xinqiao Road, Dinghai District, Zhoushan, 316000, Zhejiang, People's Republic of China
| | - Zeping Zheng
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, 355 Xinqiao Road, Dinghai District, Zhoushan, 316000, Zhejiang, People's Republic of China
| | - Jiayi Zhao
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, 355 Xinqiao Road, Dinghai District, Zhoushan, 316000, Zhejiang, People's Republic of China.
| | - Wenqing Liang
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, 355 Xinqiao Road, Dinghai District, Zhoushan, 316000, Zhejiang, People's Republic of China.
| |
Collapse
|
11
|
Chen B, Ren Q, Jiang P, Wu Q, Shuai Q, Yan Y. Combinatorial Synthesis of Alkyl Chain-Capped Poly(β-Amino Ester)s for Effective siRNA Delivery. Macromol Biosci 2024; 24:e2400168. [PMID: 39052313 DOI: 10.1002/mabi.202400168] [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: 04/08/2024] [Revised: 06/18/2024] [Indexed: 07/27/2024]
Abstract
Poly (β-amino ester) (PBAE) is a class of biodegradable polymers containing ester bonds in their main chain, extensively investigated as cationic polymer carriers for siRNA. Most current PBAE carriers rely on termination with hydrophilic or charged amines. In this study, a polymer platform consisting of 168 PBAE polymers with hydrophobic alkyl chain terminals is constructed through sequential aza-Michael addition. A large number of effective carriers are identified through in vitro screening of the PBAE platform for siLuc delivery to HeLa-Luc cells. Specifically, PA8-C6 and PA8-C8 achieve remarkable gene knockdown efficacies of up to 80% with low cytotoxicity. Certain materials from the PA2 and PA5 series demonstrate potent siRNA delivery capabilities associated with elevated cytotoxicity. The pKa value of PBAE is predominantly determined by the hydrophilic amine side chains rather than the end-capping groups. A pKa range of ≈6.2-6.5 may contribute to the excellent delivery capability for PA8 series carriers. The co-formulation of PBAE carriers with helper lipids leads to the reduced size and surface charges of the polyplex NPs with siRNA, consequently decreasing the cytotoxicity and enhancing siRNA delivery efficacy. These findings hold significant implications for the development of novel degradable polymer carriers for siRNA delivery.
Collapse
Affiliation(s)
- Baiqiu Chen
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Qidi Ren
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Pingge Jiang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Qiong Wu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Qi Shuai
- College of Pharmaceutical Sciences and Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Yunfeng Yan
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| |
Collapse
|
12
|
Zhang Y, Lei F, Qian W, Zhang C, Wang Q, Liu C, Ji H, Liu Z, Wang F. Designing intelligent bioorthogonal nanozymes: Recent advances of stimuli-responsive catalytic systems for biomedical applications. J Control Release 2024; 373:929-951. [PMID: 39097195 DOI: 10.1016/j.jconrel.2024.07.073] [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: 04/29/2024] [Revised: 07/28/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024]
Abstract
Bioorthogonal nanozymes have emerged as a potent tool in biomedicine due to their unique ability to perform enzymatic reactions that do not interfere with native biochemical processes. The integration of stimuli-responsive mechanisms into these nanozymes has further expanded their potential, allowing for controlled activation and targeted delivery. As such, intelligent bioorthogonal nanozymes have received more and more attention in developing therapeutic approaches. This review provides a comprehensive overview of the recent advances in the development and application of stimuli-responsive bioorthogonal nanozymes. By summarizing the design outlines for anchoring bioorthogonal nanozymes with stimuli-responsive capability, this review seeks to offer valuable insights and guidance for the rational design of these remarkable materials. This review highlights the significant progress made in this exciting field with different types of stimuli and the various applications. Additionally, it also examines the current challenges and limitations in the design, synthesis, and application of these systems, and proposes potential solutions and research directions. This review aims to stimulate further research toward the development of more efficient and versatile stimuli-responsive bioorthogonal nanozymes for biomedical applications.
Collapse
Affiliation(s)
- Yan Zhang
- Institute of Special Environmental Medicine, Nantong University, Nantong 226019, China
| | - Fang Lei
- School of Public Health, Nantong University, Nantong 226019, China
| | - Wanlong Qian
- Institute of Special Environmental Medicine, Nantong University, Nantong 226019, China
| | - Chengfeng Zhang
- Institute of Special Environmental Medicine, Nantong University, Nantong 226019, China
| | - Qi Wang
- School of Public Health, Nantong University, Nantong 226019, China
| | - Chaoqun Liu
- School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Haiwei Ji
- School of Public Health, Nantong University, Nantong 226019, China
| | - Zhengwei Liu
- Precision Immunology Institute, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York 10029, USA.
| | - Faming Wang
- School of Public Health, Nantong University, Nantong 226019, China.
| |
Collapse
|
13
|
Wang C, Zhang Y, Kong W, Rong X, Zhong Z, Jiang L, Chen S, Li C, Zhang F, Jiang J. Delivery of miRNAs Using Nanoparticles for the Treatment of Osteosarcoma. Int J Nanomedicine 2024; 19:8641-8660. [PMID: 39188861 PMCID: PMC11346496 DOI: 10.2147/ijn.s471900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 07/31/2024] [Indexed: 08/28/2024] Open
Abstract
Osteosarcoma is the predominant primary malignant bone tumor that poses a significant global health challenge. MicroRNAs (miRNAs) that regulate gene expression are associated with osteosarcoma pathogenesis. Thus, miRNAs are potential therapeutic targets for osteosarcoma. Nanoparticles, widely used for targeted drug delivery, facilitate miRNA-based osteosarcoma treatment. Numerous studies have focused on miRNA delivery using nanoparticles to inhibit the progress of osteosarcoma. Polymer-based, lipid-based, inorganic-based nanoparticles and extracellular vesicles were used to deliver miRNAs for the treatment of osteosarcoma. They can be modified to enhance drug loading and delivery capabilities. Also, miRNA delivery was combined with traditional therapies, for example chemotherapy, to treat osteosarcoma. Consequently, miRNA delivery offers promising therapeutic avenues for osteosarcoma, providing renewed hope for patients. This review emphasizes the studies utilizing nanoparticles for miRNA delivery in osteosarcoma treatment, then introduced and summarized the nanoparticles in detail. And it also discusses the prospects for clinical applications.
Collapse
Affiliation(s)
- Chengran Wang
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Yihong Zhang
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Weihui Kong
- Department of Stomatology, the First Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Xin’ao Rong
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Ziming Zhong
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Lei Jiang
- Department of Geriatric Medicine, Changchun Central Hospital, Changchun, Jilin Province, People’s Republic of China
| | - Shuhan Chen
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Chuang Li
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Fuqiang Zhang
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Jinlan Jiang
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| |
Collapse
|
14
|
Wang L, Wang D, Lei W, Sun T, Gu B, Dong H, Taniguchi Y, Liu Y, Ling Y. Trigonometric Bundling Disulfide Unit Starship Synergizes More Effectively to Promote Cellular Uptake. Int J Mol Sci 2024; 25:7518. [PMID: 39062760 PMCID: PMC11277142 DOI: 10.3390/ijms25147518] [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: 05/20/2024] [Revised: 07/06/2024] [Accepted: 07/07/2024] [Indexed: 07/28/2024] Open
Abstract
A small molecule disulfide unit technology platform based on dynamic thiol exchange chemistry at the cell membrane has the potential for drug delivery. However, the alteration of the CSSC dihedral angle of the disulfide unit caused by diverse substituents directly affects the effectiveness of this technology platform as well as its own chemical stability. The highly stable open-loop relaxed type disulfide unit plays a limited role in drug delivery due to its low dihedral angle. Here, we have built a novel disulfide unit starship based on the 3,4,5-trihydroxyphenyl skeleton through trigonometric bundling. The intracellular delivery results showed that the trigonometric bundling of the disulfide unit starship effectively promoted cellular uptake without any toxicity, which is far more than 100 times more active than that of equipment with a single disulfide unit in particular. Then, the significant reduction in cell uptake capacity (73-93%) using thiol erasers proves that the trigonometric bundling of the disulfide starship is an endocytosis-independent internalization mechanism via a dynamic covalent disulfide exchange mediated by thiols on the cell surface. Furthermore, analysis of the molecular dynamics simulations demonstrated that trigonometric bundling of the disulfide starship can significantly change the membrane curvature while pushing lipid molecules in multiple directions, resulting in a significant distortion in the membrane structure and excellent membrane permeation performance. In conclusion, the starship system we built fully compensates for the inefficiency deficiencies induced by poor dihedral angles.
Collapse
Affiliation(s)
- Lei Wang
- School of Pharmacy, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, China; (D.W.); (W.L.); (T.S.); (B.G.); (H.D.); (Y.L.)
| | - Dezhi Wang
- School of Pharmacy, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, China; (D.W.); (W.L.); (T.S.); (B.G.); (H.D.); (Y.L.)
| | - Wenzhuo Lei
- School of Pharmacy, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, China; (D.W.); (W.L.); (T.S.); (B.G.); (H.D.); (Y.L.)
| | - Tiantian Sun
- School of Pharmacy, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, China; (D.W.); (W.L.); (T.S.); (B.G.); (H.D.); (Y.L.)
| | - Bei Gu
- School of Pharmacy, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, China; (D.W.); (W.L.); (T.S.); (B.G.); (H.D.); (Y.L.)
| | - Han Dong
- School of Pharmacy, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, China; (D.W.); (W.L.); (T.S.); (B.G.); (H.D.); (Y.L.)
| | - Yosuke Taniguchi
- School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan;
| | - Yichang Liu
- School of Pharmacy, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, China; (D.W.); (W.L.); (T.S.); (B.G.); (H.D.); (Y.L.)
| | - Yong Ling
- School of Pharmacy, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, China; (D.W.); (W.L.); (T.S.); (B.G.); (H.D.); (Y.L.)
| |
Collapse
|
15
|
Yang R, Yao T, Xu J, Liu X, Yang Y, Ding J, Cao C, Su R, Li S, Lu W, Gao X. Peptide-TLR7/8a-Coordinated DNA Vaccines Elicit Enhanced Immune Responses against Infectious Diseases. ACS Biomater Sci Eng 2024; 10:4374-4387. [PMID: 38869358 DOI: 10.1021/acsbiomaterials.4c00749] [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: 06/14/2024]
Abstract
DNA vaccines represent an innovative approach for the immunization of diverse diseases. However, their clinical trial outcomes are constrained by suboptimal transfection efficiency and immunogenicity. In this work, we present a universal methodology involving the codelivery of Toll-like receptor 7/8 agonists (TLR7/8a) and antigen gene using TLR7/8a-conjugated peptide-coated poly(β-amino ester) (PBAE) nanoparticles (NPs) to augment delivery efficiency and immune response. Peptide-TLR7/8a-coated PBAE NPs exhibit advantageous biophysical attributes, encompassing diminutive particle dimensions, nearly neutral ζ potential, and stability in the physiological environment. This synergistic approach not only ameliorates the stability of plasmid DNA (pDNA) and gene delivery efficacy but also facilitates subsequent antigen production. Furthermore, under optimal formulation conditions, the TLR7/8a-conjugated peptide coated PBAE NPs exhibit a potent capacity to induce robust immune responses. Collectively, this nanoparticulate gene delivery system demonstrates heightened transfection efficacy, stability, biodegradability, immunostimulatory effect, and low toxicity, making it a promising platform for the clinical advancement of DNA vaccines.
Collapse
Affiliation(s)
- Rong Yang
- Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity Fudan University, Shanghai 200032, China
| | - Tingting Yao
- Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity Fudan University, Shanghai 200032, China
| | - Jinliang Xu
- Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity Fudan University, Shanghai 200032, China
| | - Xiaoxiao Liu
- Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity Fudan University, Shanghai 200032, China
| | - Yaqi Yang
- Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity Fudan University, Shanghai 200032, China
| | - Junqiang Ding
- School of Pharmacy, Shanghai Pudong Hospital & Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Cong Cao
- Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity Fudan University, Shanghai 200032, China
| | - Runping Su
- Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity Fudan University, Shanghai 200032, China
| | - Sha Li
- Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity Fudan University, Shanghai 200032, China
| | - Wuyuan Lu
- Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity Fudan University, Shanghai 200032, China
| | - Xihui Gao
- Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity Fudan University, Shanghai 200032, China
| |
Collapse
|
16
|
Vishwakarma M, Agrawal P, Soni S, Tomar S, Haider T, Kashaw SK, Soni V. Cationic nanocarriers: A potential approach for targeting negatively charged cancer cell. Adv Colloid Interface Sci 2024; 327:103160. [PMID: 38663154 DOI: 10.1016/j.cis.2024.103160] [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: 12/31/2023] [Revised: 04/04/2024] [Accepted: 04/16/2024] [Indexed: 05/07/2024]
Abstract
Cancer, a widespread and lethal disease, necessitates precise therapeutic interventions to mitigate its devastating impact. While conventional chemotherapy remains a cornerstone of cancer treatment, its lack of specificity towards cancer cells results in collateral damage to healthy tissues, leading to adverse effects. Thus, the quest for targeted strategies has emerged as a critical focus in cancer research. This review explores the development of innovative targeting methods utilizing novel drug delivery systems tailored to recognize and effectively engage cancer cells. Cancer cells exhibit morphological and metabolic traits, including irregular morphology, unchecked proliferation, metabolic shifts, genetic instability, and a higher negative charge, which serve as effective targeting cues. Central to these strategies is the exploitation of the unique negative charge characteristic of cancer cells, attributed to alterations in phospholipid composition and the Warburg effect. Leveraging this distinct feature, researchers have devised cationic carrier systems capable of enhancing the specificity of therapeutic agents towards cancer cells. The review delineates the underlying causes of the negative charge in cancer cells and elucidates various targeting approaches employing cationic compounds for drug delivery systems. Furthermore, it delves into the methods employed for the preparation of these systems. Beyond cancer treatment, the review also underscores the multifaceted applications of cationic carrier systems, encompassing protein and peptide delivery, imaging, photodynamic therapy, gene delivery, and antimicrobial applications. This comprehensive exploration underscores the potential of cationic carrier systems as versatile tools in the fight against cancer and beyond.
Collapse
Affiliation(s)
- Monika Vishwakarma
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, MP, India
| | - Poornima Agrawal
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, MP, India
| | - Sakshi Soni
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, MP, India
| | - Surbhi Tomar
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, MP, India
| | - Tanweer Haider
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, MP, India; Amity Institute of Pharmacy, Amity University Madhya Pradesh, Gwalior 474005, MP, India
| | - Sushil K Kashaw
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, MP, India
| | - Vandana Soni
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, MP, India.
| |
Collapse
|
17
|
Jia B, Zhang B, Li J, Qin J, Huang Y, Huang M, Ming Y, Jiang J, Chen R, Xiao Y, Du J. Emerging polymeric materials for treatment of oral diseases: design strategy towards a unique oral environment. Chem Soc Rev 2024; 53:3273-3301. [PMID: 38507263 DOI: 10.1039/d3cs01039b] [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: 03/22/2024]
Abstract
Oral diseases are prevalent but challenging diseases owing to the highly movable and wet, microbial and inflammatory environment. Polymeric materials are regarded as one of the most promising biomaterials due to their good compatibility, facile preparation, and flexible design to obtain multifunctionality. Therefore, a variety of strategies have been employed to develop materials with improved therapeutic efficacy by overcoming physicobiological barriers in oral diseases. In this review, we summarize the design strategies of polymeric biomaterials for the treatment of oral diseases. First, we present the unique oral environment including highly movable and wet, microbial and inflammatory environment, which hinders the effective treatment of oral diseases. Second, a series of strategies for designing polymeric materials towards such a unique oral environment are highlighted. For example, multifunctional polymeric materials are armed with wet-adhesive, antimicrobial, and anti-inflammatory functions through advanced chemistry and nanotechnology to effectively treat oral diseases. These are achieved by designing wet-adhesive polymers modified with hydroxy, amine, quinone, and aldehyde groups to provide strong wet-adhesion through hydrogen and covalent bonding, and electrostatic and hydrophobic interactions, by developing antimicrobial polymers including cationic polymers, antimicrobial peptides, and antibiotic-conjugated polymers, and by synthesizing anti-inflammatory polymers with phenolic hydroxy and cysteine groups that function as immunomodulators and electron donors to reactive oxygen species to reduce inflammation. Third, various delivery systems with strong wet-adhesion and enhanced mucosa and biofilm penetration capabilities, such as nanoparticles, hydrogels, patches, and microneedles, are constructed for delivery of antibiotics, immunomodulators, and antioxidants to achieve therapeutic efficacy. Finally, we provide insights into challenges and future development of polymeric materials for oral diseases with promise for clinical translation.
Collapse
Affiliation(s)
- Bo Jia
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangdong, China
| | - Beibei Zhang
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.
- Department of Polymeric Materials, School of Materials Science and Engineering, Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Jianhua Li
- Department of Polymeric Materials, School of Materials Science and Engineering, Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Jinlong Qin
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.
- Department of Polymeric Materials, School of Materials Science and Engineering, Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Yisheng Huang
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangdong, China
| | - Mingshu Huang
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangdong, China
| | - Yue Ming
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangdong, China
| | - Jingjing Jiang
- Department of Polymeric Materials, School of Materials Science and Engineering, Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Ran Chen
- Department of Polymeric Materials, School of Materials Science and Engineering, Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Yufen Xiao
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.
- Department of Polymeric Materials, School of Materials Science and Engineering, Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Jianzhong Du
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.
- Department of Polymeric Materials, School of Materials Science and Engineering, Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| |
Collapse
|
18
|
Pegoraro C, Domingo-Ortí I, Conejos-Sánchez I, Vicent MJ. Unlocking the Mitochondria for Nanomedicine-based Treatments: Overcoming Biological Barriers, Improving Designs, and Selecting Verification Techniques. Adv Drug Deliv Rev 2024; 207:115195. [PMID: 38325562 DOI: 10.1016/j.addr.2024.115195] [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: 10/24/2023] [Revised: 01/13/2024] [Accepted: 02/02/2024] [Indexed: 02/09/2024]
Abstract
Enhanced targeting approaches will support the treatment of diseases associated with dysfunctional mitochondria, which play critical roles in energy generation and cell survival. Obstacles to mitochondria-specific targeting include the presence of distinct biological barriers and the need to pass through (or avoid) various cell internalization mechanisms. A range of studies have reported the design of mitochondrially-targeted nanomedicines that navigate the complex routes required to influence mitochondrial function; nonetheless, a significant journey lies ahead before mitochondrially-targeted nanomedicines become suitable for clinical use. Moving swiftly forward will require safety studies, in vivo assays confirming effectiveness, and methodologies to validate mitochondria-targeted nanomedicines' subcellular location/activity. From a nanomedicine standpoint, we describe the biological routes involved (from administration to arrival within the mitochondria), the features influencing rational design, and the techniques used to identify/validate successful targeting. Overall, rationally-designed mitochondria-targeted-based nanomedicines hold great promise for precise subcellular therapeutic delivery.
Collapse
Affiliation(s)
- Camilla Pegoraro
- Polymer Therapeutics Laboratory and CIBERONC, Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - Inés Domingo-Ortí
- Polymer Therapeutics Laboratory and CIBERONC, Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - Inmaculada Conejos-Sánchez
- Polymer Therapeutics Laboratory and CIBERONC, Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - María J Vicent
- Polymer Therapeutics Laboratory and CIBERONC, Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| |
Collapse
|
19
|
Wang X, Li Y, A S, Lyu J, Wang X, He Z, Lara-Sáez I, Li M, Wang W. Cyclization-enhanced poly(β-amino ester)s vectors for efficient CRISPR gene editing therapy. J Control Release 2024; 368:444-452. [PMID: 38401849 DOI: 10.1016/j.jconrel.2024.02.032] [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: 11/28/2023] [Revised: 01/29/2024] [Accepted: 02/21/2024] [Indexed: 02/26/2024]
Abstract
Among non-viral gene delivery vectors, poly(β-amino ester)s (PAEs) are one of the most versatile candidates because of their wide monomer availability, high polymer flexibility, and superior gene transfection performance both in vitro and in vivo. Over two decades, PAEs have evolved from linear to highly branched structures, significantly enhancing gene delivery efficacy. Building on the proven efficient sets of monomers in highly branched PAEs (HPAEs), this work introduced a new class of cyclic PAEs (CPAEs) constructed via an A2 + B4 + C2 cyclization synthesis strategy and identified their markedly improved gene transfection capabilities in gene delivery applications. Two sets of cyclic PAEs (CPAEs) with rings of different sizes and topologies were obtained. Their chemical structures were confirmed via two-dimensional nuclear magnetic resonance and the photoluminescence phenomena, and their DNA delivery behaviours were investigated and compared with the HPAE counterparts. In vitro assessments demonstrated that the CPAEs with a macrocyclic architecture (MCPAEs), significantly enhanced DNA intracellular uptake and facilitated efficient gene expression while maintaining perfect biocompatibility. The top-performance MCPAEs have been further employed to deliver a plasmid coding dual single guide RNA-guided CRISPR-Cas9 machinery to delete COL7A1 exon 80 containing the c.6527dupC mutation. In recessive dystrophic epidermolysis bullosa (RDEB) patient-derived epidermal keratinocytes, MCPAEs facilitated the CRISPR plasmid delivery and achieved efficient targeted gene editing in multiple colonies.
Collapse
Affiliation(s)
- Xianqing Wang
- Charles Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Yinghao Li
- Charles Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Sigen A
- School of Medicine, Anhui University of Science and Technology, Huainan 232001, China
| | - Jing Lyu
- Charles Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland.
| | - Xi Wang
- Institute of Precision Medicine (AUST-IPM), Anhui University of Science and Technology, Huainan 232001, China
| | - Zhonglei He
- Institute of Precision Medicine (AUST-IPM), Anhui University of Science and Technology, Huainan 232001, China
| | - Irene Lara-Sáez
- Charles Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Ming Li
- Department of Dermatology, Children's Hospital of Fudan University, National Children's Medical Center, 399 Wanyuan Road, Shanghai, China
| | - Wenxin Wang
- Charles Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland.
| |
Collapse
|
20
|
Kasza K, Richards B, Jones S, Romero M, Robertson SN, Hardie KR, Gurnani P, Cámara M, Alexander C. Ciprofloxacin Poly(β-amino ester) Conjugates Enhance Antibiofilm Activity and Slow the Development of Resistance. ACS APPLIED MATERIALS & INTERFACES 2024; 16:5412-5425. [PMID: 38289032 PMCID: PMC10859900 DOI: 10.1021/acsami.3c14357] [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: 09/25/2023] [Revised: 12/17/2023] [Accepted: 01/05/2024] [Indexed: 02/09/2024]
Abstract
To tackle the emerging antibiotic resistance crisis, novel antimicrobial approaches are urgently needed. Bacterial biofilms are a particular concern in this context as they are responsible for over 80% of bacterial infections and are inherently more recalcitrant toward antimicrobial treatments. The high tolerance of biofilms to conventional antibiotics has been attributed to several factors, including reduced drug diffusion through the dense exopolymeric matrix and the upregulation of antimicrobial resistance machinery with successful biofilm eradication requiring prolonged high doses of multidrug treatments. A promising approach to tackle bacterial infections involves the use of polymer drug conjugates, shown to improve upon free drug toxicity and bioavailability, enhance drug penetration through the thick biofilm matrix, and evade common resistance mechanisms. In the following study, we conjugated the antibiotic ciprofloxacin (CIP) to a small library of biodegradable and biocompatible poly(β-amino ester) (PBAE) polymers with varying central amine functionality. The suitability of the polymers as antibiotic conjugates was then verified in a series of assays including testing of efficacy and resistance response in planktonic Gram-positive and Gram-negative bacteria and the reduction of viability in mono- and multispecies biofilm models. The most active polymer within the prepared PBAE-CIP library was shown to achieve an over 2-fold increase in the reduction of biofilm viability in a Pseudomonas aeruginosa monospecies biofilm and superior elimination of all the species present within the multispecies biofilm model. Hence, we demonstrate that CIP conjugation to PBAEs can be employed to achieve improved antibiotic efficacy against clinically relevant biofilm models.
Collapse
Affiliation(s)
- Karolina Kasza
- Division
of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, U.K.
- National
Biofilms Innovation Centre, School of Life Sciences, Biodiscovery
Institute, University Park, University of
Nottingham, Nottingham NG7 2RD, U.K.
| | - Brogan Richards
- National
Biofilms Innovation Centre, School of Life Sciences, Biodiscovery
Institute, University Park, University of
Nottingham, Nottingham NG7 2RD, U.K.
| | - Sal Jones
- Division
of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, U.K.
| | - Manuel Romero
- National
Biofilms Innovation Centre, School of Life Sciences, Biodiscovery
Institute, University Park, University of
Nottingham, Nottingham NG7 2RD, U.K.
- Department
of Microbiology and Parasitology, Faculty of Biology-CIBUS, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Shaun N. Robertson
- National
Biofilms Innovation Centre, School of Life Sciences, Biodiscovery
Institute, University Park, University of
Nottingham, Nottingham NG7 2RD, U.K.
| | - Kim R. Hardie
- National
Biofilms Innovation Centre, School of Life Sciences, Biodiscovery
Institute, University Park, University of
Nottingham, Nottingham NG7 2RD, U.K.
| | - Pratik Gurnani
- UCL
School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, U.K.
| | - Miguel Cámara
- National
Biofilms Innovation Centre, School of Life Sciences, Biodiscovery
Institute, University Park, University of
Nottingham, Nottingham NG7 2RD, U.K.
| | - Cameron Alexander
- Division
of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, U.K.
| |
Collapse
|
21
|
Friesen JJ, Blakney AK. Trends in the synthetic polymer delivery of RNA. J Gene Med 2024; 26:e3672. [PMID: 38380796 DOI: 10.1002/jgm.3672] [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: 06/01/2023] [Revised: 11/27/2023] [Accepted: 01/27/2024] [Indexed: 02/22/2024] Open
Abstract
Ribonucleic acid (RNA) has emerged as one of the most promising therapeutic payloads in the field of gene therapy. There are many unique types of RNA that allow for a range of applications including vaccination, protein replacement therapy, autoimmune disease treatment, gene knockdown and gene editing. However, RNA triggers the host immune system, is vulnerable to degradation and has a low proclivity to enter cells spontaneously. Therefore, a delivery vehicle is required to facilitate the protection and uptake of RNA therapeutics into the desired host cells. Lipid nanoparticles have emerged as one of the only clinically approved vehicles for genetic payloads, including in the COVID-19 messenger RNA vaccines. While lipid nanoparticles have distinct advantages, they also have drawbacks, including strong immune stimulation, complex manufacturing and formulation heterogeneity. In contrast, synthetic polymers are a widely studied group of gene delivery vehicles and boast distinct advantages, including biocompatibility, tunability, inexpensiveness, simple formulation and ease of modification. Some classes of polymers enhance efficient transfection efficiency, and lead to lower stimulation of the host immune system, making them more viable candidates for non-vaccine-related applications of RNA medicines. This review aims to identify the most promising classes of synthetic polymers, summarize recent research aimed at moving them into the clinic and postulate the future steps required for unlocking their full potential.
Collapse
Affiliation(s)
- Josh J Friesen
- Michael Smith Laboratories, University of British Columbia, Vancouver, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
| | - Anna K Blakney
- Michael Smith Laboratories, University of British Columbia, Vancouver, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
| |
Collapse
|
22
|
Brito J, Moon J, Hlushko R, Aliakseyeu A, Andrianov AK, Sukhishvili SA. Engineering Degradation Rate of Polyphosphazene-Based Layer-by-Layer Polymer Coatings. J Funct Biomater 2024; 15:26. [PMID: 38391879 PMCID: PMC10889497 DOI: 10.3390/jfb15020026] [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: 12/10/2023] [Revised: 01/13/2024] [Accepted: 01/21/2024] [Indexed: 02/24/2024] Open
Abstract
Degradable layer-by-layer (LbL) polymeric coatings have distinct advantages over traditional biomedical coatings due to their precision of assembly, versatile inclusion of bioactive molecules, and conformality to the complex architectures of implantable devices. However, controlling the degradation rate while achieving biocompatibility has remained a challenge. This work employs polyphosphazenes as promising candidates for film assembly due to their inherent biocompatibility, tunability of chemical composition, and the buffering capability of degradation products. The degradation of pyrrolidone-functionalized polyphosphazenes was monitored in solution, complexes and LbL coatings (with tannic acid), providing the first to our knowledge comparison of solution-state degradation to solid-state LbL degradation. In all cases, the rate of degradation accelerated in acidic conditions. Importantly, the tunability of the degradation rate of polyphosphazene-based LbL films was achieved by varying film assembly conditions. Specifically, by slightly increasing the ionization of tannic acid (near neutral pH), we introduce electrostatic "defects" to the hydrogen-bonded pairs that accelerate film degradation. Finally, we show that replacing the pyrrolidone side group with a carboxylic acid moiety greatly reduces the degradation rate of the LbL coatings. In practical applications, these coatings have the versatility to serve as biocompatible platforms for various biomedical applications and controlled release systems.
Collapse
Affiliation(s)
- Jordan Brito
- Department of Materials Science & Engineering, Texas A&M University, College Station, TX 77840, USA
| | - Junho Moon
- Department of Materials Science & Engineering, Texas A&M University, College Station, TX 77840, USA
| | - Raman Hlushko
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850, USA
| | - Aliaksei Aliakseyeu
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Alexander K Andrianov
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850, USA
| | - Svetlana A Sukhishvili
- Department of Materials Science & Engineering, Texas A&M University, College Station, TX 77840, USA
| |
Collapse
|
23
|
Wang X, Wang X, Li Y, A S, Qiu B, Bushmalyova A, He Z, Wang W, Lara-Sáez I. CRISPR-Cas9-based non-viral gene editing therapy for topical treatment of recessive dystrophic epidermolysis bullosa. Mol Ther Methods Clin Dev 2023; 31:101134. [PMID: 38027067 PMCID: PMC10630779 DOI: 10.1016/j.omtm.2023.101134] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023]
Abstract
Recessive dystrophic epidermolysis bullosa (RDEB) is an autosomal monogenic skin disease caused by mutations in COL7A1 gene and lack of functional type VII collagen (C7). Currently, there is no cure for RDEB, and most of the gene therapies under development have been designed as ex vivo strategies because of the shortage of efficient and safe carriers for gene delivery. Herein, we designed, synthesized, and screened a new group of highly branched poly(β amino ester)s (HPAEs) as non-viral carriers for the delivery of plasmids encoding dual single-guide RNA (sgRNA)-guided CRISPR-Cas9 machinery to delete COL7A1 exon 80 containing the c.6527dupC mutation. The selected HPAEs (named PTTA-DATOD) showed robust transfection efficiency, comparable with or surpassing that of leading commercial gene transfection reagents such as Lipofectamine 3000, Xfect, and jetPEI, while maintaining negligible cytotoxicity. Furthermore, CRISPR-Cas9 plasmids delivered by PTTA-DATOD achieved efficient targeted deletion and restored bulk C7 production in RDEB patient keratinocyte polyclones. The non-viral CRISPR-Cas9-based COL7A1 exon deletion approach developed here has great potential to be used as a topical treatment for RDEB patients with mutations in COL7A1 exon 80. Besides, this therapeutic strategy can easily be adapted for mutations in other COL7A1 exons, other epidermolysis bullosa subtypes, and other genetic diseases.
Collapse
Affiliation(s)
- Xianqing Wang
- Charles Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Xi Wang
- Research and Clinical Translation Center of Gene Medicine and Tissue Engineering, School of Public Health, Anhui University of Science and Technology, Huainan 232001, China
| | - Yinghao Li
- Charles Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Sigen A
- Research and Clinical Translation Center of Gene Medicine and Tissue Engineering, School of Public Health, Anhui University of Science and Technology, Huainan 232001, China
| | - Bei Qiu
- Charles Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Albina Bushmalyova
- Charles Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Zhonglei He
- Research and Clinical Translation Center of Gene Medicine and Tissue Engineering, School of Public Health, Anhui University of Science and Technology, Huainan 232001, China
| | - Wenxin Wang
- Charles Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
- Research and Clinical Translation Center of Gene Medicine and Tissue Engineering, School of Public Health, Anhui University of Science and Technology, Huainan 232001, China
| | - Irene Lara-Sáez
- Charles Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| |
Collapse
|
24
|
Kasza K, Elsherbeny A, Moloney C, Hardie KR, Cámara M, Alexander C, Gurnani P. Hybrid Poly( β-amino ester) Triblock Copolymers Utilizing a RAFT Polymerization Grafting-From Methodology. MACROMOL CHEM PHYS 2023; 224:2300262. [PMID: 38495072 PMCID: PMC10941699 DOI: 10.1002/macp.202300262] [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: 07/21/2023] [Revised: 08/25/2023] [Indexed: 03/19/2024]
Abstract
The biocompatibility, biodegradability, and responsiveness of poly(β-amino esters) (PBAEs) has led to their widespread use as biomaterials for drug and gene delivery. Nonetheless, the step-growth polymerization mechanism that yields PBAEs limits the scope for their structural optimization toward specific applications because of limited monomer choice and end-group modifications. Moreover, to date the post-synthetic functionalization of PBAEs has relied on grafting-to approaches, challenged by the need for efficient polymer-polymer coupling and potentially difficult post-conjugation purification. Here a novel grafting-from approach to grow reversible addition-fragmentation chain transfer (RAFT) polymers from a PBAE scaffold is described. This is achieved through PBAE conversion into a macromolecular chain transfer agent through a multistep capping procedure, followed by RAFT polymerization with a range of monomers to produce PBAE-RAFT hybrid triblock copolymers. Following successful synthesis, the potential biological applications of these ABA triblock copolymers are illustrated through assembly into polymeric micelles and encapsulation of a model hydrophobic drug, followed by successful nanoparticle (NP) uptake in breast cancer cells. The findings demonstrate this novel synthetic methodology can expand the scope of PBAEs as biomaterials.
Collapse
Affiliation(s)
- Karolina Kasza
- Division of Molecular Therapeutics and FormulationSchool of PharmacyUniversity of NottinghamNottinghamNG7 2RDUK
- National Biofilms Innovation CentreSchool of Life Sciences, Biodiscovery InstituteUniversity Park, University of NottinghamNottinghamNG7 2RDUK
| | - Amr Elsherbeny
- Division of Molecular Therapeutics and FormulationSchool of PharmacyUniversity of NottinghamNottinghamNG7 2RDUK
- Ex Vivo Cancer Pharmacology Centre of ExcellenceSchool of MedicineUniversity of NottinghamNottinghamNG7 2RDUK
- School of MedicineBiodiscovery InstituteUniversity Park, University of NottinghamNottinghamNG7 2RDUK
| | - Cara Moloney
- School of MedicineBiodiscovery InstituteUniversity Park, University of NottinghamNottinghamNG7 2RDUK
| | - Kim R. Hardie
- National Biofilms Innovation CentreSchool of Life Sciences, Biodiscovery InstituteUniversity Park, University of NottinghamNottinghamNG7 2RDUK
| | - Miguel Cámara
- National Biofilms Innovation CentreSchool of Life Sciences, Biodiscovery InstituteUniversity Park, University of NottinghamNottinghamNG7 2RDUK
| | - Cameron Alexander
- Division of Molecular Therapeutics and FormulationSchool of PharmacyUniversity of NottinghamNottinghamNG7 2RDUK
| | - Pratik Gurnani
- UCL School of PharmacyUniversity College London29–39 Brunswick SquareLondonWC1N 1AXUK
| |
Collapse
|
25
|
Zhang C, Huang T, Liang J, Cao H, Xu Y, Liu W, Dong Z. A COF template-derived mesoporous CeO 2-supported Au nanoparticles catalyst for the oxidative esterification of benzaldehydes and benzyl alcohols. Dalton Trans 2023; 52:15907-15917. [PMID: 37840534 DOI: 10.1039/d3dt02823b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
The direct oxidative esterification of benzaldehydes and benzyl alcohols to high value-added aromatic esters under mild and green reaction conditions is significant in the fine chemical industry. The accurate design of catalysts with high catalytic performance is crucial for this process. Herein, 2,4,6-trimethylpyridine, benzoic anhydride, and terephthalaldehyde were used to prepare a covalent organic framework (COF) material, which was then used as a template to construct a mesoporous CeO2-supported Au nanoparticles catalyst. The obtained Au@CeO2 catalyst was thoroughly characterized, and it possessed a mesoporous structure with a high surface area. Meanwhile, the as-prepared Au@CeO2 exhibited excellent catalytic performance in the oxidative esterification of benzaldehydes and benzyl alcohols with methanol, affording the corresponding aromatic esters under mild and green reaction conditions. Furthermore, the Au@CeO2 catalyst could also be recycled. Therefore, this study provides a green and sustainable pathway for the synthesis of high-value-added esters through a direct oxidative esterification strategy.
Collapse
Affiliation(s)
- Cheng Zhang
- College of Chemistry and Chemical Engineering, Laboratory of Special Function Materials and Structure Design of the Ministry of Education, Lanzhou University, Lanzhou 730000, PR China.
- Baotou Research Institute of Rare Earths, Baotou 014030, PR China.
| | - Tianhang Huang
- College of Chemistry and Chemical Engineering, Laboratory of Special Function Materials and Structure Design of the Ministry of Education, Lanzhou University, Lanzhou 730000, PR China.
| | - Jinhua Liang
- College of Chemistry and Chemical Engineering, Laboratory of Special Function Materials and Structure Design of the Ministry of Education, Lanzhou University, Lanzhou 730000, PR China.
| | - Hongzhang Cao
- Baotou Research Institute of Rare Earths, Baotou 014030, PR China.
| | - Yanhui Xu
- Baotou Research Institute of Rare Earths, Baotou 014030, PR China.
| | - Weisheng Liu
- College of Chemistry and Chemical Engineering, Laboratory of Special Function Materials and Structure Design of the Ministry of Education, Lanzhou University, Lanzhou 730000, PR China.
| | - Zhengping Dong
- College of Chemistry and Chemical Engineering, Laboratory of Special Function Materials and Structure Design of the Ministry of Education, Lanzhou University, Lanzhou 730000, PR China.
- Baotou Research Institute of Rare Earths, Baotou 014030, PR China.
| |
Collapse
|
26
|
Zhu L, Lu Q, Bian T, Yang P, Yang Y, Zhang L. Fabrication and Characterization of π-π Stacking Peptide-Contained Double Network Hydrogels. ACS Biomater Sci Eng 2023; 9:4761-4769. [PMID: 37424070 DOI: 10.1021/acsbiomaterials.3c00579] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Since the physical properties are similar to native extracellular matrices, double network (DN) hydrogels have been studied extensively in the tissue engineering. However, the double chemical crosslinked DN hydrogel is limited by poor fatigue resistance. π-π stacking is a non-covalent bonding interaction, which is essential to maintain and self-assemble the three-dimensional structure of biological proteins and nucleic acids. In this study, a robust polyethylene glycol diacrylate (PEGDA)/FFK hybrid DN hydrogel was prepared by Michael addition and π-π stacking. The hybrid DN hydrogels with π-π stacking interactions have excellent mechanical strength and fatigue resistance. The DN FFK/PEGDA hydrogels reveal great biocompatibility and hemocompatibility. The DN hydrogels containing π-π stacking have the potential to fabricate robust hybrid DN hydrogels in drug release and tissue engineering.
Collapse
Affiliation(s)
- Linglin Zhu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu, PR China
| | - Qiuyun Lu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu, PR China
| | - Taotao Bian
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu, PR China
| | - Panpan Yang
- Medical School of Nantong University, Nantong University, Nantong 226001, Jiangsu, PR China
| | - Yumin Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu, PR China
| | - Luzhong Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu, PR China
| |
Collapse
|
27
|
Pang Y, Guan L, Zhu Y, Niu R, Zhu S, Lin Q. Gallic acid-grafted chitosan antibacterial hydrogel incorporated with polydopamine-modified hydroxyapatite for enhancing bone healing. Front Bioeng Biotechnol 2023; 11:1162202. [PMID: 37334266 PMCID: PMC10273101 DOI: 10.3389/fbioe.2023.1162202] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 05/18/2023] [Indexed: 06/20/2023] Open
Abstract
An open critical-size bone defect is a major medical problem because of the difficulty in self-healing, leading to an increased risk of bacterial infection owing to wound exposure, resulting in treatment failure. Herein, a composite hydrogel was synthesized by chitosan, gallic acid, and hyaluronic acid, termed "CGH." Hydroxyapatite was modified with polydopamine (PDA@HAP) and introduced to CGH to obtain a mussel-inspired mineralized hydrogel (CGH/PDA@HAP). The CGH/PDA@HAP hydrogel exhibited excellent mechanical performances, including self-healing and injectable properties. Owing to its three-dimensional porous structure and polydopamine modifications, the cellular affinity of the hydrogel was enhanced. When adding PDA@HAP into CGH, Ca2+ and PO4 3- could release and then promoted differentiation of BMSCs into osteoblasts. Without any osteogenic agent or stem cells, the area of new bone at the site of defect was enhanced and the newly formed bone had a dense trabecular structure after implanting of the CGH/PDA@HAP hydrogel for 4 and 8 weeks. Moreover, the growth of Staphylococcus aureus and Escherichia coli was effectively inhibited through the grafting of gallic acid onto chitosan. Above, this study provides a reasonable alternative strategy to manage open bone defects.
Collapse
Affiliation(s)
- Yuxuan Pang
- Department of Prosthodontics, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Lin Guan
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, China
| | - Yanlin Zhu
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Ruijuan Niu
- Meilong Community Health Service Center, Shanghai, China
| | - Song Zhu
- Department of Prosthodontics, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Quan Lin
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, China
| |
Collapse
|