1
|
Nayanathara U, Yang F, Zhang C, Wang Y, Rossi Herling B, Smith SA, Beach MA, Johnston APR, Such GK. Unravelling the endosomal escape of pH-responsive nanoparticles using the split luciferase endosomal escape quantification assay. Biomater Sci 2025; 13:1335-1346. [PMID: 39898829 DOI: 10.1039/d4bm01433b] [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: 02/04/2025]
Abstract
Endosomal escape is a major bottleneck for efficient intracellular delivery of therapeutic cargoes, particularly for macromolecular biological cargoes such as peptides, proteins and nucleic acids. pH-responsive polymeric nanoparticles that can respond to changes in the pH of intracellular microenvironments have generated substantial interest in navigating the endosomal barrier. In this study, we applied the highly sensitive split luciferase endosomal escape quantification (SLEEQ) assay to better understand the endosomal escape efficiency of dual component pH-responsive nanoparticles based on poly(2-(diethylamino) ethyl methacrylate) (PDEAEMA) and poly(2-(diisopropylamino) ethyl methacrylate) (PDPAEMA). Previous work investigated the use of a disulfide-linked HiBiT peptide conjugate encapsulated within the nanoparticle core, which upon meeting the LgBiT protein in the cytosol demonstrated luminescence which could be quantified to assess endosomal escape. However, we were interested in understanding whether this assay could be tuned to understand the endosomal escape of both a therapeutic cargo and a larger carrier. To achieve this, we designed two different HiBiT conjugates by applying a carbonylacrylic-functionalized thioether (non-cleavable) linker, which is more stable in endosomes, and a less stable disulfide (cleavable) linker to attach HiBiT to the nanoparticle core. Nanoparticles with disulfide-linked HiBiT demonstrated a higher endosomal escape efficiency of 6-7%, whereas thioether-linked HiBiT demonstrated <3% endosomal escape efficiency with a twofold decrease in cytosolic delivery. This suggests that degradation of the disulfide linker in endosomes leads to cytosolic delivery of a free HiBiT cargo, while thioether-linked HiBiT polymers are larger and thus fewer HiBiT-carrier conjugates can escape the endosomes. Overall, this work demonstrates that the SLEEQ assay can be tuned to understand the cytosolic delivery of different components based on the use of different linker chemistries and thus it is an important tool for designing therapeutic delivery systems in the future.
Collapse
Affiliation(s)
- Umeka Nayanathara
- School of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia.
| | - Fan Yang
- School of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia.
| | - Changhe Zhang
- School of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia.
| | - Yufu Wang
- School of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia.
| | - Bruna Rossi Herling
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia.
| | - Samuel A Smith
- School of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia.
| | - Maximilian A Beach
- School of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia.
| | - Angus P R Johnston
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia.
| | - Georgina K Such
- School of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia.
| |
Collapse
|
2
|
Laurent Q, Bona BL, Asohan J, Rosati M, Faiad S, Bombelli FB, Metrangolo P, Sleiman HF. Self-Assembly and Biological Properties of Highly Fluorinated Oligonucleotide Amphiphiles. Angew Chem Int Ed Engl 2025; 64:e202419996. [PMID: 39636686 PMCID: PMC11811686 DOI: 10.1002/anie.202419996] [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: 10/16/2024] [Revised: 11/24/2024] [Accepted: 12/04/2024] [Indexed: 12/07/2024]
Abstract
Nucleic acids, used as therapeutics to silence disease-related genes, offer significant advantages over small molecule drugs: they provide high specificity, the ability to target "undruggable" molecules, and adaptability to a wide range of disease phenotypes. However, their instability in biological media, as well their rapid clearance from the organism limit their applicability, necessitating the use of nanocarriers to overcome these challenges. Among these strategies, spherical nucleic acids (SNA)-composed of a densely packed corona of oligonucleotides around a nanoparticle-have emerged as a powerful tool, in particular when self-assembled from DNA amphiphiles. This non-covalent strategy however has caveats, especially when it comes to stability in complex biological media, where these SNAs disassemble in contact to serum proteins. Here, we developed highly fluorinated DNA amphiphiles that readily self-assemble into SNAs and have tunable stability profiles in biological media. They are made of branched fluorinated moieties with potentially improved biodegradability as compared to their linear counterparts. Depending on the number of fluorophilic interactions, the self-assembled SNAs can have excellent serum stabilities-up to days-and readily deliver nucleic acid therapeutics for gene silencing applications. These systems show great potential as promising candidates for nucleic acid-based therapies.
Collapse
Affiliation(s)
- Quentin Laurent
- Department of ChemistryMcGill University801 Sherbrooke St. WQC-H3A 0B8MontrealCanada
| | - Beatrice L. Bona
- SupraBioNano LabDepartment of ChemistryMaterialsand Chemical Engineering “Giulio Natta”Politecnico di MilanoVia Luigi Mancinelli 7MI-20131MilanoItaly
| | - Jathavan Asohan
- Department of ChemistryMcGill University801 Sherbrooke St. WQC-H3A 0B8MontrealCanada
| | - Marta Rosati
- SupraBioNano LabDepartment of ChemistryMaterialsand Chemical Engineering “Giulio Natta”Politecnico di MilanoVia Luigi Mancinelli 7MI-20131MilanoItaly
| | - Sinan Faiad
- Department of ChemistryMcGill University801 Sherbrooke St. WQC-H3A 0B8MontrealCanada
| | - Francesca Baldelli Bombelli
- SupraBioNano LabDepartment of ChemistryMaterialsand Chemical Engineering “Giulio Natta”Politecnico di MilanoVia Luigi Mancinelli 7MI-20131MilanoItaly
| | - Pierangelo Metrangolo
- SupraBioNano LabDepartment of ChemistryMaterialsand Chemical Engineering “Giulio Natta”Politecnico di MilanoVia Luigi Mancinelli 7MI-20131MilanoItaly
| | - Hanadi F. Sleiman
- Department of ChemistryMcGill University801 Sherbrooke St. WQC-H3A 0B8MontrealCanada
| |
Collapse
|
3
|
Kaviani S, Bai H, Das T, Asohan J, Elmanzalawy A, Marlyn J, Choueiri LE, Damha MJ, Laurent Q, Sleiman HF. Photochemical Stabilization of Self-Assembled Spherical Nucleic Acids. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2407742. [PMID: 39790078 PMCID: PMC11840461 DOI: 10.1002/smll.202407742] [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: 08/30/2024] [Revised: 11/08/2024] [Indexed: 01/12/2025]
Abstract
Oligonucleotide therapeutics, including antisense oligonucleotides and small interfering RNA, offer promising avenues for modulating the expression of disease-associated proteins. However, challenges such as nuclease degradation, poor cellular uptake, and unspecific targeting hinder their application. To overcome these obstacles, spherical nucleic acids have emerged as versatile tools for nucleic acid delivery in biomedical applications. Our laboratory has introduced sequence-defined DNA amphiphiles which self-assemble in aqueous solutions. Despite their advantages, self-assembled SNAs can be inherently fragile due to their reliance on non-covalent interactions and fall apart in biologically relevant conditions, specifically by interaction with serum proteins. Herein, this challenge is addressed by introducing two methods of covalent crosslinking of SNAs via UV irradiation. Thymine photodimerization or disulfide crosslinking at the micellar interface enhance SNA stability against human serum albumin binding. This enhanced stability, particularly for disulfide crosslinked SNAs, leads to increased cellular uptake. Furthermore, this crosslinking results in sustained activity and accessibility for release of the therapeutic nucleic acid, along with improvement in unaided gene silencing. The findings demonstrate the efficient stabilization of SNAs through UV crosslinking, influencing their cellular uptake, therapeutic release, and ultimately, gene silencing activity. These studies offer promising avenues for further optimization and exploration of pre-clinical, in vivo studies.
Collapse
Affiliation(s)
- Sepideh Kaviani
- Department of ChemistryMcGill University801, Sherbrooke St. WestMontrealQCH3A 0B8Canada
| | - Haochen Bai
- Department of ChemistryMcGill University801, Sherbrooke St. WestMontrealQCH3A 0B8Canada
| | - Trishalina Das
- Department of ChemistryMcGill University801, Sherbrooke St. WestMontrealQCH3A 0B8Canada
| | - Jathavan Asohan
- Department of ChemistryMcGill University801, Sherbrooke St. WestMontrealQCH3A 0B8Canada
| | | | - Julian Marlyn
- Department of ChemistryMcGill University801, Sherbrooke St. WestMontrealQCH3A 0B8Canada
| | - Lea El Choueiri
- Department of ChemistryMcGill University801, Sherbrooke St. WestMontrealQCH3A 0B8Canada
| | - Masad J. Damha
- Department of ChemistryMcGill University801, Sherbrooke St. WestMontrealQCH3A 0B8Canada
| | - Quentin Laurent
- Department of ChemistryMcGill University801, Sherbrooke St. WestMontrealQCH3A 0B8Canada
- University Grenoble Alpes, DCM UMR 5250Grenoble Cedex 938058France
| | - Hanadi F. Sleiman
- Department of ChemistryMcGill University801, Sherbrooke St. WestMontrealQCH3A 0B8Canada
| |
Collapse
|
4
|
Sabet MJ, Hasanzadeh A, Vahabi A, Hosseini ES, Saeedi S, Chegeni BK, Kiani J, Azar BKY, Molabashi ZA, Shamsara M, Hamblin MR, Karimi M, Roustazadeh A. Metal-Coordinated Histidine-Functionalized Redox-Responsive Polyethyleneimine as a Smart Gene Delivery Vector. Mol Biotechnol 2025:10.1007/s12033-024-01360-x. [PMID: 39806116 DOI: 10.1007/s12033-024-01360-x] [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: 03/10/2024] [Accepted: 12/12/2024] [Indexed: 01/16/2025]
Abstract
Despite significant advancements in gene delivery and CRISPR technology, several challenges remain. Chief among these are overcoming serum inhibition and achieving high transfection efficiency with minimal cytotoxicity. To address these issues, there is a need for novel vectors that exhibit lower toxicity, maintain stability in serum-rich environments, and effectively deliver plasmids of various sizes across diverse cell types. In this study, to convert common polyethylenimine (PEI1.8k) into high-performance DNA delivery vectors, an innovative multifunctional vector was constructed based on histidine linked to PEI1.8k by redox-responsive disulfide bonds. Apart from highly efficient transfection of both small and large plasmids into HEK 293T (Human Embryonic Kidney 293T cells) with negligible cytotoxicity, PEI1.8k-S-S-His showed great transfection potential even at low plasmid doses (0.5 µg), as well as at serum concentrations ranging from 5 to 30% into HEK 293T cells, and achieved excellent plasmid transfection into NIH/3T3 (Mouse Embryonic Fibroblast cells), and MCF7 (Human Breast Cancer cells). Additionally, several metals were tested (Co, Cu, Cd, Ni, Zn, and Mn) to promote the plasmid packaging functionality and improve transfection efficiency. We observed that, in comparison to PEI1.8k-S-S-His, the manganese-functionalized nanocarrier (PEI1.8k-S-S-His-Mn) could transfect a large plasmid with equal efficiency (~ 30%) into MSCs (Mesenchymal Stem Cells). Interestingly, PEI1.8k-S-S-His-Mn showed higher transfection efficiency with the small plasmid (~ 90%) and the large one (~ 80%) into HEK 293T cells, even better than its backbone. We propose that the presence of metal-coordinated His ligand, redox-responsive S-S bonds, and the cationic polymer can synergistically provide robust DNA binding, efficient endosomal disruption, tolerance of serum protein adsorption, and low cytotoxicity. These new vectors could be promising for gene delivery and may be therapeutically relevant.
Collapse
Affiliation(s)
- Makkieh Jahanpeimay Sabet
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran, Iran
| | - Akbar Hasanzadeh
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran, Iran
| | - Amirhossein Vahabi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran, Iran
| | - Elaheh Sadat Hosseini
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran, Iran
| | - Sara Saeedi
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Neuroscience Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Beheshteh Khodadadi Chegeni
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran, Iran
| | - Jafar Kiani
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Behjat Kheiri Yeghaneh Azar
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Student Research Committee, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Asghari Molabashi
- Department of Plant Molecular Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Mehdi Shamsara
- Department of Animal Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028, South Africa
| | - Mahdi Karimi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran, Iran.
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Research Center for Science and Technology in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Applied Biotechnology Research Centre, Tehran Medical Science, Islamic Azad University, Tehran, Iran.
| | - Abazar Roustazadeh
- Noncommunicable Disease Research Center, Jahrom University of Medical Sciences, Jahrom, Iran.
- Dapartment of Biochemistry, Jahrom University of Medical Sciences, Jahrom, Iran.
- Department of Advanced Medical Sciences and Technologies, Jahrom University of Medical Sciences, Jahrom, Iran.
| |
Collapse
|
5
|
Yang C, Zhao Y, Jiao Y, Yang L, He B, Dai W, Zhang H, Zhang Q, Wang X. Organelle-Level Trafficking and Metabolism Kinetics for Redox-Responsive Paclitaxel Prodrug Nanoparticles Characterized by Experimental and Modeling Analysis. ACS NANO 2024. [PMID: 39688493 DOI: 10.1021/acsnano.4c09704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Redox-responsive self-assembled prodrug nanoparticles have received extensive attention for their high loading efficiency and environmentally responsive properties. However, the intracellular metabolism and transportation kinetics were poorly understood, which limited the rational design and development of this delivery system. Herein, tetraphenylporphyrin-paclitaxel (TxP) prodrugs with thioether, disulfide, and dicarbon linkers (TsP, TssP, and TccP) were synthesized and self-assembled as nanoparticles. The redox responsiveness was investigated both in the simulated medium and in tumor cells via mass spectrometry. TxP NP and PTX concentrations in 4T1 whole cells, endosomal systems, and cytoplasm over time were quantified by UPLC-MS/MS and modeled using the nonlinear mixed effect (NLME) approach. Cytotoxicity was studied in 4T1 and MCF-7 cell lines, and antitumor efficacy was analyzed in 4T1 tumor-bearing mice. Mass spectrometry identified both oxidative and reductive metabolites in redox simulants for TssP NPs and TsP NPs. In 4T1 cells, only reductive metabolites for TssP NPs were detected, while both oxidative and reductive metabolites for TsP NPs were detected. The developed subcellular pharmacokinetic model suggested that the estimated metabolism rates of TxP NPs in endosomal systems were 10 to 27 times of the rates in cytoplasm, indicating that endosomal systems were the dominant place for intracellular metabolism. These rates were numerically higher for TsP NPs than TssP NPs in endosomal systems (1.7-fold) and the cytoplasm (2.5-fold). The internalization of nanoparticles was identified to be slow (kmax,int, 0.015 h-1) and saturable. The transportation rate constant across the endosomal membranes was fast for PTX (27.1 h-1) and slow for TxP NPs (0.098 h-1). TsP and TssP NPs had comparable in vivo antitumor efficacy, which was higher than that of TccP NPs. This study quantified the organelle-level transportation and metabolism kinetics for three prodrug nanoparticles with redox-responsive or inert linkers using a combined experimental and modeling approach. These findings and the modeling framework might inform future studies for redox-responsive prodrug design and drug delivery systems.
Collapse
Affiliation(s)
- Canyu Yang
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China
| | - Yao Zhao
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yuanyuan Jiao
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pharmacy, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Long Yang
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Bing He
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Wenbing Dai
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Hua Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Qiang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xueqing Wang
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
6
|
Zhang Z, Lu Y, Liu W, Huang Y. Nanomaterial-assisted delivery of CpG oligodeoxynucleotides for boosting cancer immunotherapy. J Control Release 2024; 376:184-199. [PMID: 39368710 DOI: 10.1016/j.jconrel.2024.09.044] [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: 05/09/2024] [Revised: 08/03/2024] [Accepted: 09/26/2024] [Indexed: 10/07/2024]
Abstract
Cancer immunotherapy aims to improve immunity to not only eliminate the primary tumor but also inhibit metastasis and recurrence. It is considered an extremely promising therapeutic approach that breaks free from the traditional paradigm of oncological treatment. As the medical community learns more about the immune system's mechanisms that "turn off the brake" and "step on the throttle", there is increasingly successful research on immunomodulators. However, there are still more restrictions than countermeasures with immunotherapy related to immunomodulators, such as low responsiveness and immune-related adverse events that cause multiple adverse reactions. Therefore, medical experts and materials scientists attempted to the efficacy of immunomodulatory treatments through various methods, especially nanomaterial-assisted strategies. CpG oligodeoxynucleotides (CpG) not only act as an adjuvant to promote immune responses, but also induce autophagy. In this review, the enhancement of immunotherapy using nanomaterial-based CpG formulations is systematically elaborated, with a focus on the delivery, protection, synergistic promotion of CpG efficacy by nanomaterials, and selection of the timing of treatment. In addition, we also discuss and prospect the existing problems and future directions of research on nanomaterials in auxiliary CpG therapy.
Collapse
Affiliation(s)
- Zhiyu Zhang
- Department of Pharmacology, Beijing Chest Hospital, Capital Medical University/Beijing Key Laboratory of Drug Resistance Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Yu Lu
- Department of Pharmacology, Beijing Chest Hospital, Capital Medical University/Beijing Key Laboratory of Drug Resistance Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China.
| | - Wenjing Liu
- Department of Pharmacology, Beijing Chest Hospital, Capital Medical University/Beijing Key Laboratory of Drug Resistance Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China.
| | - Yuanyu Huang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
7
|
Zeng H, Zhou S, Zhang X, Liang Q, Yan M, Xu Y, Guo Y, Hu X, Jiang L, Kong B. Super-assembled periodic mesoporous organosilica membranes with hierarchical channels for efficient glutathione sensing. Analyst 2024; 149:3522-3529. [PMID: 38787653 DOI: 10.1039/d4an00559g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Abstract
Bioinspired nanochannel-based sensors have elicited significant interest because of their excellent sensing performance, and robust mechanical and tunable chemical properties. However, the existing designs face limitations due to material constraints, which hamper broader application possibilities. Herein, a heteromembrane system composed of a periodic mesoporous organosilica (PMO) layer with three-dimensional (3D) network nanochannels is constructed for glutathione (GSH) detection. The unique hierarchical pore architecture provides a large surface area, abundant reaction sites and plentiful interconnected pathways for rapid ionic transport, contributing to efficient and sensitive detection. Moreover, the thioether groups in nanochannels can be selectively cleaved by GSH to generate hydrophilic thiol groups. Benefiting from the increased hydrophilic surface, the proposed sensor achieves efficient GSH detection with a detection limit of 1.2 μM by monitoring the transmembrane ionic current and shows good recovery ranges in fetal bovine serum sample detection. This work paves an avenue for designing and fabricating nanofluidic sensing systems for practical and biosensing applications.
Collapse
Affiliation(s)
- Hui Zeng
- Department of Chemistry, Shanghai Key Lab of Molecular Catalysis and Innovative Materials and Collaborative Innovation Center of Chemistry for Energy Materials, Fudan University, Shanghai 200438, P. R. China.
| | - Shan Zhou
- Department of Chemistry, Shanghai Key Lab of Molecular Catalysis and Innovative Materials and Collaborative Innovation Center of Chemistry for Energy Materials, Fudan University, Shanghai 200438, P. R. China.
| | - Xin Zhang
- Department of Chemistry, Shanghai Key Lab of Molecular Catalysis and Innovative Materials and Collaborative Innovation Center of Chemistry for Energy Materials, Fudan University, Shanghai 200438, P. R. China.
| | - Qirui Liang
- Department of Chemistry, Shanghai Key Lab of Molecular Catalysis and Innovative Materials and Collaborative Innovation Center of Chemistry for Energy Materials, Fudan University, Shanghai 200438, P. R. China.
| | - Miao Yan
- Department of Chemistry, Shanghai Key Lab of Molecular Catalysis and Innovative Materials and Collaborative Innovation Center of Chemistry for Energy Materials, Fudan University, Shanghai 200438, P. R. China.
| | - Yeqing Xu
- Department of Chemistry, Shanghai Key Lab of Molecular Catalysis and Innovative Materials and Collaborative Innovation Center of Chemistry for Energy Materials, Fudan University, Shanghai 200438, P. R. China.
| | - Yaxin Guo
- Department of Chemistry, Shanghai Key Lab of Molecular Catalysis and Innovative Materials and Collaborative Innovation Center of Chemistry for Energy Materials, Fudan University, Shanghai 200438, P. R. China.
| | - Xiaomeng Hu
- Department of Chemistry, Shanghai Key Lab of Molecular Catalysis and Innovative Materials and Collaborative Innovation Center of Chemistry for Energy Materials, Fudan University, Shanghai 200438, P. R. China.
| | - Lei Jiang
- CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Science, Beijing 100190, P. R. China
| | - Biao Kong
- Department of Chemistry, Shanghai Key Lab of Molecular Catalysis and Innovative Materials and Collaborative Innovation Center of Chemistry for Energy Materials, Fudan University, Shanghai 200438, P. R. China.
- Yiwu Research Institute of Fudan University, Yiwu, Zhejiang 322000, P. R. China
- Shandong Research Institute, Fudan University, Jinan, Shandong 250103, P. R. China
| |
Collapse
|
8
|
Ooi YJ, Huang C, Lau K, Chew SY, Park JG, Chan-Park MB. Nontoxic, Biodegradable Hyperbranched Poly(β-amino ester)s for Efficient siRNA Delivery and Gene Silencing. ACS APPLIED MATERIALS & INTERFACES 2024; 16:14093-14112. [PMID: 38449351 DOI: 10.1021/acsami.3c10620] [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: 03/08/2024]
Abstract
RNA interference (RNAi)-mediated gene silencing is a promising therapeutic approach to treat various diseases, but safe and efficient delivery remains a major challenge to its clinical application. Non-viral gene vectors, such as poly(β-amino esters) (pBAEs), have emerged as a potential candidate due to their biodegradability, low toxicity profile, ease of synthesis, and high gene transfection efficiency for both DNA and siRNA delivery. However, achieving significant gene silencing using pBAEs often requires a large amount of polymer carrier (with polymer/siRNA weight ratio >100) or high siRNA dose (>100 nM), which might potentially exacerbate toxicity concerns during delivery. To overcome these barriers, we designed and optimized a series of hyperbranched pBAEs capable of efficiently condensing siRNA and achieving excellent silencing efficiency at a lower polymer/siRNA weight ratio (w/w) and siRNA dose. Through modulation of monomer combinations and branching density, we identified the top-performing hyperbranched pBAEs, named as h(A2B3)-1, which possess good siRNA condensation ability, low cytotoxicity, and high cellular uptake efficiency. Compared with Lipofectamine 2000, h(A2B3)-1 achieved lower cytotoxicity and higher siRNA silencing efficiency in HeLa cells at a polymer/siRNA weight ratio of 30 and 30 nM siRNA dose. Notably, h(A2B3)-1 enhanced the gene uptake in primary neural cells and effectively silenced the target gene in hard-to-transfect primary cortical neurons and oligodendrocyte progenitor cells, with gene knockdown efficiencies of 34.8 and 53.4% respectively. By incorporating a bioreducible disulfide compartment into the polymer backbone, the cytocompatibility of the h(A2B3)-1 was greatly enhanced while maintaining their good transfection efficiency. Together, the low cytotoxicity and high siRNA transfection efficiency of hyperbranched h(A2B3)-1 in this study demonstrated their great potential as a non-viral gene vector for efficient siRNA delivery and RNAi-mediated gene silencing. This provides valuable insight into the future development of safe and efficient non-viral siRNA delivery systems as well as their translation into clinical applications.
Collapse
Affiliation(s)
- Ying Jie Ooi
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637459, Singapore
| | - Chongquan Huang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637459, Singapore
- Neuroscience@NTU, Interdisciplinary Graduate Programme, Nanyang Technological University, Singapore 637459, Singapore
| | - Kieran Lau
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637459, Singapore
| | - Sing Yian Chew
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637459, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921, Singapore
- School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Jong Gu Park
- Welgene Inc, 693, Namcheon-ro, Namcheon-myeon, Gyeongsan-si, Gyeongsangbuk-do 38695, Republic of Korea
| | - Mary B Chan-Park
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637459, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921, Singapore
| |
Collapse
|
9
|
Liu W, Cao J, Ma C, Zhou H, Guo T. Zinc(II)-Quinoline module and ROS cleavable crosslinker functionalized self-fluorescent siRNA vectors for selective gene silencing of cancer cells. J Control Release 2024; 366:366-374. [PMID: 38184231 DOI: 10.1016/j.jconrel.2023.12.055] [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: 12/04/2023] [Revised: 12/27/2023] [Accepted: 12/31/2023] [Indexed: 01/08/2024]
Abstract
A highly efficient siRNA vector (Zn-PQD) capable of selectively silencing genes in cancer cells was obtained by using ROS-cleavable DED to crosslink low molecular weight (LMW) polyethylene imine (PEI) modified by self-fluorescent metal coordinatied multifunctional module Zn-QS. Under the combined action of DED cross-linking and Zn-QS modification, Zn-PQD performs well in the siRNA delivery process in cancer cells, including siRNA condensation, cell uptake, endosome escape, and siRNA release. Zn-PQD exhibited higher transfection efficiency than commercial PEI25k and Lipo2k in multiple cancer cell lines including HepG2, HeLa, 4 T1, H520 and PANC-1, as well as cancer treatment-related stem cell rADSC. Ultimately, Zn-PQD can achieve extremely high and selective gene silencing effects in cancer cells (with a gene silencing rate of 98.3% in HepG2). This work is expected to provide an efficient and safe siRNA carrier for the future tumor siRNA therapy and its study of fluorescence mediated mechanism.
Collapse
Affiliation(s)
- Weijie Liu
- Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry and Frontier Science Center for the Creation of New Organic Substances, Nankai University, Tianjin 300071, China
| | - Junpeng Cao
- Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry and Frontier Science Center for the Creation of New Organic Substances, Nankai University, Tianjin 300071, China
| | - Chunchao Ma
- Department of Neurology, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin 300192, China.
| | - Hao Zhou
- Department of Biochemistry and Molecular Biology, College of Life Science, Nankai University, Tianjin 300071, China
| | - Tianying Guo
- Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry and Frontier Science Center for the Creation of New Organic Substances, Nankai University, Tianjin 300071, China.
| |
Collapse
|
10
|
Đorđević S, Medel M, Hillaert J, Masiá E, Conejos-Sánchez I, Vicent MJ. Critical Design Strategies Supporting Optimized Drug Release from Polymer-Drug Conjugates. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2303157. [PMID: 37752780 DOI: 10.1002/smll.202303157] [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: 04/14/2023] [Revised: 08/19/2023] [Indexed: 09/28/2023]
Abstract
The importance of an adequate linking moiety design that allows controlled drug(s) release at the desired site of action is extensively studied for polymer-drug conjugates (PDCs). Redox-responsive self-immolative linkers bearing disulfide moieties (SS-SIL) represent a powerful strategy for intracellular drug delivery; however, the influence of drug structural features and linker-associated spacers on release kinetics remains relatively unexplored. The influence of drug/spacer chemical structure and the chemical group available for conjugation on drug release and the biological effect of resultant PDCs is evaluated. A "design of experiments" tool is implemented to develop a liquid chromatography-mass spectrometry method to perform the comprehensive characterization required for this systematic study. The obtained fit-for-purpose analytical protocol enables the quantification of low drug concentrations in drug release studies and the elucidation of metabolite presence. and provides the first data that clarifies how drug structural features influence the drug release from SS-SIL and demonstrates the non-universal nature of the SS-SIL. The importance of rigorous linker characterization in understanding structure-function correlations between linkers, drug chemical functionalities, and in vitro release kinetics from a rationally-designed polymer-drug nanoconjugate, a critical strategic crafting methodology that should remain under consideration when using a reductive environment as an endogenous drug release trigger.
Collapse
Affiliation(s)
- Snežana Đorđević
- Polymer Therapeutics Laboratory, Príncipe Felipe Research Center (CIPF) and CIBERONC, Eduardo Primo Yúfera 3, Valencia, 46012, Spain
| | - María Medel
- Polymer Therapeutics Laboratory, Príncipe Felipe Research Center (CIPF) and CIBERONC, Eduardo Primo Yúfera 3, Valencia, 46012, Spain
| | - Justine Hillaert
- Polymer Therapeutics Laboratory, Príncipe Felipe Research Center (CIPF) and CIBERONC, Eduardo Primo Yúfera 3, Valencia, 46012, Spain
| | - Esther Masiá
- Polymer Therapeutics Laboratory, Príncipe Felipe Research Center (CIPF) and CIBERONC, Eduardo Primo Yúfera 3, Valencia, 46012, Spain
- Screening Platform, Príncipe Felipe Research Center (CIPF), Eduardo Primo Yúfera 3, Valencia, 46012, Spain
| | - Inmaculada Conejos-Sánchez
- Polymer Therapeutics Laboratory, Príncipe Felipe Research Center (CIPF) and CIBERONC, Eduardo Primo Yúfera 3, Valencia, 46012, Spain
| | - María J Vicent
- Polymer Therapeutics Laboratory, Príncipe Felipe Research Center (CIPF) and CIBERONC, Eduardo Primo Yúfera 3, Valencia, 46012, Spain
- Screening Platform, Príncipe Felipe Research Center (CIPF), Eduardo Primo Yúfera 3, Valencia, 46012, Spain
| |
Collapse
|
11
|
Aono R, Nomura K, Yuba E, Harada A. Reversible Stabilization of Nanofiber-Polyplexes through Introducing Cross-Linkages. J Funct Biomater 2023; 15:14. [PMID: 38248681 PMCID: PMC10817492 DOI: 10.3390/jfb15010014] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 12/27/2023] [Accepted: 12/28/2023] [Indexed: 01/23/2024] Open
Abstract
Non-viral gene delivery systems are typically designed vector systems with contradictory properties, namely sufficient stability before cellular uptake and instability to ensure the release of nucleic acid cargoes in the transcription process after being taken up into cells. We reported previously that poly-(L-lysine) terminally bearing a multi-arm PEG (maPEG-PLL) formed nanofiber-polyplexes that suppressed excessive DNA condensation via steric repulsion among maPEGs and exhibited effective transcriptional capability in PCR amplification experiments and a cell-free gene expression system. In this study, the reversible stabilization of a nanofiber-polyplex without impairing the effective transcriptional capability was investigated by introducing cross-links between the PLL side chains within the polyplex using a cross-linking reagent with disulfide (SS) bonds that can be disrupted under reducing conditions. In the presence of dextran sulfate and/or dithiothreitol, the stability of the polyplex and the reactivity of the pDNA were evaluated using agarose gel electrophoresis and real-time PCR. We succeeded in reversibly stabilizing nanofiber-polyplexes using dithiobis (succinimidyl propionate) (DSP) as the cross-linking reagent. The effect of the reversible stabilization was confirmed in experiments using cultured cells, and the DSP-crosslinked polyplexes exhibited gene expression superior to that of polyethyleneimine polyplexes, which are typical polyplexes.
Collapse
Affiliation(s)
- Ryuta Aono
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Sakai 599-8531, Osaka, Japan
| | - Kenta Nomura
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Sakai 599-8531, Osaka, Japan
| | - Eiji Yuba
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Sakai 599-8531, Osaka, Japan
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Metropolitan University, 1-1 Gakuen-cho, Sakai 599-8531, Osaka, Japan
| | - Atsushi Harada
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Sakai 599-8531, Osaka, Japan
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Metropolitan University, 1-1 Gakuen-cho, Sakai 599-8531, Osaka, Japan
| |
Collapse
|
12
|
Mao L, Ma P, Luo X, Cheng H, Wang Z, Ye E, Loh XJ, Wu YL, Li Z. Stimuli-Responsive Polymeric Nanovaccines Toward Next-Generation Immunotherapy. ACS NANO 2023; 17:9826-9849. [PMID: 37207347 DOI: 10.1021/acsnano.3c02273] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
The development of nanovaccines that employ polymeric delivery carriers has garnered substantial interest in therapeutic treatment of cancer and a variety of infectious diseases due to their superior biocompatibility, lower toxicity and reduced immunogenicity. Particularly, stimuli-responsive polymeric nanocarriers show great promise for delivering antigens and adjuvants to targeted immune cells, preventing antigen degradation and clearance, and increasing the uptake of specific antigen-presenting cells, thereby sustaining adaptive immune responses and improving immunotherapy for certain diseases. In this review, the most recent advances in the utilization of stimulus-responsive polymer-based nanovaccines for immunotherapeutic applications are presented. These sophisticated polymeric nanovaccines with diverse functions, aimed at therapeutic administration for disease prevention and immunotherapy, are further classified into several active domains, including pH, temperature, redox, light and ultrasound-sensitive intelligent nanodelivery systems. Finally, the potential strategies for the future design of multifunctional next-generation polymeric nanovaccines by integrating materials science with biological interface are proposed.
Collapse
Affiliation(s)
- Liuzhou Mao
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Panqin Ma
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Xi Luo
- BE/Phase I Clinical Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361000, China
| | - Hongwei Cheng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Zhanxiang Wang
- BE/Phase I Clinical Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361000, China
| | - Enyi Ye
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2), Agency for Science, Technology and Research (A*STAR), 1 Pesek Road, Jurong Island, Singapore 627833, Republic of Singapore
| | - Xian Jun Loh
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2), Agency for Science, Technology and Research (A*STAR), 1 Pesek Road, Jurong Island, Singapore 627833, Republic of Singapore
| | - Yun-Long Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Zibiao Li
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2), Agency for Science, Technology and Research (A*STAR), 1 Pesek Road, Jurong Island, Singapore 627833, Republic of Singapore
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Republic of Singapore
| |
Collapse
|
13
|
Zyryanov GV, Kopchuk DS, Kovalev IS, Santra S, Majee A, Ranu BC. Pillararenes as Promising Carriers for Drug Delivery. Int J Mol Sci 2023; 24:ijms24065167. [PMID: 36982244 PMCID: PMC10049520 DOI: 10.3390/ijms24065167] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 02/19/2023] [Accepted: 02/21/2023] [Indexed: 03/30/2023] Open
Abstract
Since their discovery in 2008 by N. Ogoshi and co-authors, pillararenes (PAs) have become popular hosts for molecular recognition and supramolecular chemistry, as well as other practical applications. The most useful property of these fascinating macrocycles is their ability to accommodate reversibly guest molecules of various kinds, including drugs or drug-like molecules, in their highly ordered rigid cavity. The last two features of pillararenes are widely used in various pillararene-based molecular devices and machines, stimuli-responsive supramolecular/host-guest systems, porous/nonporous materials, organic-inorganic hybrid systems, catalysis, and, finally, drug delivery systems. In this review, the most representative and important results on using pillararenes for drug delivery systems for the last decade are presented.
Collapse
Affiliation(s)
- Grigory V Zyryanov
- Chemical Engineering Institute, Ural Federal University, 19 Mira Street, 620002 Yekaterinburg, Russia
- I. Ya. Postovskiy Institute of Organic Synthesis, Ural Division of the Russian Academy of Sciences, 22 S. Kovalevskoy Street, 620219 Yekaterinburg, Russia
| | - Dmitry S Kopchuk
- Chemical Engineering Institute, Ural Federal University, 19 Mira Street, 620002 Yekaterinburg, Russia
- I. Ya. Postovskiy Institute of Organic Synthesis, Ural Division of the Russian Academy of Sciences, 22 S. Kovalevskoy Street, 620219 Yekaterinburg, Russia
| | - Igor S Kovalev
- Chemical Engineering Institute, Ural Federal University, 19 Mira Street, 620002 Yekaterinburg, Russia
- I. Ya. Postovskiy Institute of Organic Synthesis, Ural Division of the Russian Academy of Sciences, 22 S. Kovalevskoy Street, 620219 Yekaterinburg, Russia
| | - Sougata Santra
- Chemical Engineering Institute, Ural Federal University, 19 Mira Street, 620002 Yekaterinburg, Russia
| | - Adinath Majee
- Department of Chemistry, Visva-Bharati University, Santiniketan 731235, India
| | - Brindaban C Ranu
- Chemical Engineering Institute, Ural Federal University, 19 Mira Street, 620002 Yekaterinburg, Russia
- School of Chemical Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| |
Collapse
|
14
|
Khakpour E, Salehi S, Naghib SM, Ghorbanzadeh S, Zhang W. Graphene-based nanomaterials for stimuli-sensitive controlled delivery of therapeutic molecules. Front Bioeng Biotechnol 2023; 11:1129768. [PMID: 36845181 PMCID: PMC9947473 DOI: 10.3389/fbioe.2023.1129768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 01/19/2023] [Indexed: 02/11/2023] Open
Abstract
Stimuli-responsive drug delivery has attracted tremendous attention in the past decades. It provides a spatial- and temporal-controlled release in response to different triggers, thus enabling highly efficient drug delivery and minimizing drug side effects. Graphene-based nanomaterials have been broadly explored, and they show great potential in smart drug delivery due to their stimuli-responsive behavior and high loading capacity for an extended range of drug molecules. These characteristics are a result of high surface area, mechanical stability and chemical stability, and excellent optical, electrical, and thermal properties. Their great and infinite functionalization potential also allows them to be integrated into several types of polymers, macromolecules, or other nanoparticles, leading to the fabrication of novel nanocarriers with enhanced biocompatibility and trigger-sensitive properties. Thus, numerous studies have been dedicated to graphene modification and functionalization. In the current review, we introduce graphene derivatives and different graphene-based nanomaterials utilized in drug delivery and discuss the most important advances in their functionalization and modification. Also, their potential and progress in an intelligent drug release in response to different types of stimuli either endogenous (pH, redox conditions, and reactive oxygen species (ROS)) or exogenous (temperature, near-infrared (NIR) radiation, and electric field) will be debated.
Collapse
Affiliation(s)
- Elnaz Khakpour
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology and Biomaterials and Tissue Engineering Department, Breast Cancer Research Center, Motamed Cancer Institute, IUST, ACECR, Tehran, Iran
| | - Saba Salehi
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology and Biomaterials and Tissue Engineering Department, Breast Cancer Research Center, Motamed Cancer Institute, IUST, ACECR, Tehran, Iran
| | - Seyed Morteza Naghib
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology and Biomaterials and Tissue Engineering Department, Breast Cancer Research Center, Motamed Cancer Institute, IUST, ACECR, Tehran, Iran,*Correspondence: Seyed Morteza Naghib, ; Wei Zhang,
| | - Sadegh Ghorbanzadeh
- State Key Laboratory of Structure Analysis for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian, China
| | - Wei Zhang
- State Key Laboratory of Structure Analysis for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian, China,*Correspondence: Seyed Morteza Naghib, ; Wei Zhang,
| |
Collapse
|
15
|
Li J, Zhu F, Lou K, Tian H, Luo Q, Dang Y, Liu X, Wang P, Wu L. Tumor microenvironment enhanced NIR II fluorescence imaging for tumor precise surgery navigation via tetrasulfide mesoporous silica-coated Nd-based rare-earth nanocrystals. Mater Today Bio 2022; 16:100397. [PMID: 36081578 PMCID: PMC9445393 DOI: 10.1016/j.mtbio.2022.100397] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/08/2022] [Accepted: 08/08/2022] [Indexed: 11/28/2022] Open
Abstract
In vivo fluorescent imaging by using the new contrast agents emitted at short-wavelength infrared region (NIR II, 1000-1700 nm) presents an unprecedent advantages in imaging sensitivity and spatial resolution over traditional near-infrared (NIR) light. Recently, Nd-based rare-earth nanocrystals have attracted considerable attention due to the high quantum yield (∼40%) of their emission at NIR II. However, undesirable capture by reticuloendothelial system to bring strong background signal is unsatisfying for tumor discrimination. Here, GSH-sensitive tetrasulfide bond incorporated mesoporous silica shell has entrusted onto Nd-based down-conversion nanocrystals (DCNPs) surface to totally quench the fluorescence of DCNPs. After RGD conjugation on the silica surface, the NIR II contrast agents could actively target to liver tumors. Then tetrasulfide bonds can be broken during the silica framework decomposing in cytoplasm under high GSH concentration to result in NIR II fluorescence explosive recover. Benefiting from this specific response under tumor microenvironment, the NIR II signal in other organs was markedly reduced, while the signal-to-background ratio is prominently enhanced in tumors. Then, solid liver tumors were successfully resected under the guidance of our GSH responsive NIR II fluorescent imaging with no recurrence after 20-day of surgery. Meanwhile, by combining with the ignorable side effects, the Nd-based nanoprobes vastly improved the imaging resolution of tumor margin, opening a paradigm of NIR II fluorescent imaging-guided surgery.
Collapse
Affiliation(s)
- Jiaqi Li
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China
- Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, 350002, PR China
| | - Fukai Zhu
- Collaborative Innovation Center of Mushroom Health Industry, Minnan Normal University, Zhangzhou, Fujian, 363000, PR China
| | - Kangliang Lou
- Cancer Center & Department of Breast and Thyroid Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361100, Fujian, China
| | - Haina Tian
- Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, 350002, PR China
- Department of Biomaterials, College of Materials, Research Center of Biomedical Engineering of Xiamen & Key Laboratory of Biomedical Engineering of Fujian Province, Xiamen University, Xiamen, 361005, PR China
| | - Qiang Luo
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China
- Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, 350002, PR China
| | - Yongying Dang
- Cancer Center & Department of Breast and Thyroid Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361100, Fujian, China
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China
- Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, 350002, PR China
| | - Peiyuan Wang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China
- Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, 350002, PR China
| | - Liming Wu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang Provincial Key Laboratory of Pancreatic Disease, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| |
Collapse
|
16
|
Amarsy I, Papot S, Gasser G. Stimuli‐Responsive Metal Complexes for Biomedical Applications. Angew Chem Int Ed Engl 2022; 61:e202205900. [DOI: 10.1002/anie.202205900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Indexed: 11/09/2022]
Affiliation(s)
- Ivanna Amarsy
- Chimie ParisTech PSL University, CNRS Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology 75005 Paris France
| | - Sébastien Papot
- Institut de Chimie des Milieux et des Matériaux de Poitiers (IC2MP) Université de Poitiers, CNRS Equipe Labellisée Ligue Contre le Cancer 4 rue Michel Brunet, TSA 51106 86073 Poitiers France
| | - Gilles Gasser
- Chimie ParisTech PSL University, CNRS Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology 75005 Paris France
| |
Collapse
|
17
|
Chen F, Liu Q, Xiong Y, Xu L. Nucleic acid strategies for infectious disease treatments: The nanoparticle-based oral delivery route. Front Pharmacol 2022; 13:984981. [PMID: 36105233 PMCID: PMC9465296 DOI: 10.3389/fphar.2022.984981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Therapies based on orally administrated nucleic acids have significant potential for the treatment of infectious diseases, including chronic inflammatory diseases such as inflammatory bowel disease (IBD)-associated with the gastrointestinal (GI) tract, and infectious and acute contagious diseases like coronavirus disease 2019 (COVID-19). This is because nucleic acids could precisely regulate susceptibility genes in regulating the pro- and anti-inflammatory cytokines expression related to the infections. Unfortunately, gene delivery remains a major hurdle due to multiple intracellular and extracellular barriers. This review thoroughly discusses the challenges of nanoparticle-based nucleic acid gene deliveries and strategies for overcoming delivery barriers to the inflammatory sites. Oral nucleic acid delivery case studies were also present as vital examples of applications in infectious diseases such as IBD and COVID-19.
Collapse
Affiliation(s)
- Fengqian Chen
- Translational Research Program, Department of Anesthesiology and Center for Shock Trauma Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Qi Liu
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Yang Xiong
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Xu
- Department of Anorectal Surgery, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- *Correspondence: Li Xu,
| |
Collapse
|
18
|
Teng XQ, Qu J, Li GH, Zhuang HH, Qu Q. Small Interfering RNA for Gliomas Treatment: Overcoming Hurdles in Delivery. Front Cell Dev Biol 2022; 10:824299. [PMID: 35874843 PMCID: PMC9304887 DOI: 10.3389/fcell.2022.824299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Gliomas are central nervous system tumors originating from glial cells, whose incidence and mortality rise in coming years. The current treatment of gliomas is surgery combined with chemotherapy or radiotherapy. However, developing therapeutic resistance is one of the significant challenges. Recent research suggested that small interfering RNA (siRNA) has excellent potential as a therapeutic to silence genes that are significantly involved in the manipulation of gliomas’ malignant phenotypes, including proliferation, invasion, metastasis, therapy resistance, and immune escape. However, it is challenging to deliver the naked siRNA to the action site in the cells of target tissues. Therefore, it is urgent to develop delivery strategies to transport siRNA to achieve the optimal silencing effect of the target gene. However, there is no systematic discussion about siRNAs’ clinical potential and delivery strategies in gliomas. This review mainly discusses siRNAs’ delivery strategies, especially nanotechnology-based delivery systems, as a potential glioma therapy. Moreover, we envisage the future orientation and challenges in translating these findings into clinical applications.
Collapse
Affiliation(s)
- Xin-Qi Teng
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Pharmacy, The Second Xiangya Hospital, Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Jian Qu
- Department of Pharmacy, The Second Xiangya Hospital, Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Guo-Hua Li
- Department of Pharmacy, The Second Xiangya Hospital, Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Hai-Hui Zhuang
- Department of Pharmacy, The Second Xiangya Hospital, Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Qiang Qu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Qiang Qu,
| |
Collapse
|
19
|
Kozlovskaya V, Dolmat M, Kharlampieva E. Two-Dimensional and Three-Dimensional Ultrathin Multilayer Hydrogels through Layer-by-Layer Assembly. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:7867-7888. [PMID: 35686955 DOI: 10.1021/acs.langmuir.2c00630] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Stimuli-responsive multilayer hydrogels have opened new opportunities to design hierarchically organized networks with properties controlled at the nanoscale. These multilayer materials integrate structural, morphological, and compositional versatility provided by alternating layer-by-layer polymer deposition with the capability for dramatic and reversible changes in volumes upon environmental triggers, a characteristic of chemically cross-linked responsive networks. Despite their intriguing potential, there has been limited knowledge about the structure-property relationships of multilayer hydrogels, partly because of the challenges in regulating network structural organization and the limited set of the instrumental pool to resolve structure and properties at nanometer spatial resolution. This Feature Article highlights our recent studies on advancing assembly technologies, fundamentals, and applications of multilayer hydrogels. The fundamental relationships among synthetic strategies, chemical compositions, and hydrogel architectures are discussed, and their impacts on stimuli-induced volume changes, morphology, and mechanical responses are presented. We present an overview of our studies on thin multilayer hydrogel coatings, focusing on controlling and quantifying the degree of layer intermixing, which are crucial issues in the design of hydrogels with predictable properties. We also uncover the behavior of stratified "multicompartment" hydrogels in response to changes in pH and temperature. We summarize the mechanical responses of free-standing multilayer hydrogels, including planar thin coatings and films with closed geometries such as hollow microcapsules and nonhollow hydrogel microparticles with spherical and nonspherical shapes. Finally, we will showcase potential applications of pH- and temperature-sensitive multilayer hydrogels in sensing and drug delivery. The knowledge about multilayer hydrogels can advance the rational design of polymer networks with predictable and well-tunable properties, contributing to modern polymer science and broadening hydrogel applications.
Collapse
|
20
|
Amarsy I, Papot S, Gasser G. Stimuli‐Responsive Metal Complexes for Biomedical Applications. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202205900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Ivanna Amarsy
- Chimie ParisTech - PSL: Ecole nationale superieure de chimie de Paris PSL University FRANCE
| | - Sébastien Papot
- Université de Poitiers: Universite de Poitiers Institut de Chimie des Milieux et des Matériaux de Poitiers (IC2MP) FRANCE
| | - Gilles Gasser
- Universite PSL Chimie ParisTech 11, rue Pierre et Marie Curie 75005 Paris FRANCE
| |
Collapse
|
21
|
Burger M, Kaelin S, Leroux J. The TFAMoplex-Conversion of the Mitochondrial Transcription Factor A into a DNA Transfection Agent. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104987. [PMID: 35038234 PMCID: PMC8922101 DOI: 10.1002/advs.202104987] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Indexed: 06/14/2023]
Abstract
Non-viral gene delivery agents, such as cationic lipids, polymers, and peptides, mainly rely on charge-based and hydrophobic interactions for the condensation of DNA molecules into nanoparticles. The human protein mitochondrial transcription factor A (TFAM), on the other hand, has evolved to form nanoparticles with DNA through highly specific protein-protein and protein-DNA interactions. Here, the properties of TFAM are repurposed to create a DNA transfection agent by means of protein engineering. TFAM is covalently fused to Listeria monocytogenes phospholipase C (PLC), an enzyme that lyses lipid membranes under acidic conditions, to enable endosomal escape and human vaccinia-related kinase 1 (VRK1), which is intended to protect the DNA from cytoplasmic defense mechanisms. The TFAM/DNA complexes (TFAMoplexes) are stabilized by cysteine point mutations introduced rationally in the TFAM homodimerization site, resulting in particles, which show maximal activity when formed in 80% serum and transfect HeLa cells in vitro after 30 min of incubation under challenging cell culture conditions. The herein developed TFAM-based DNA scaffolds combine interesting characteristics in an easy-to-use system and can be readily expanded with further protein factors. This makes the TFAMoplex a promising tool in protein-based gene delivery.
Collapse
Affiliation(s)
- Michael Burger
- Swiss Federal Institute of Technology Zurich (ETHZ)Department of Chemistry and Applied BiosciencesInstitute of Pharmaceutical SciencesVladimir‐Prelog‐Weg 3Zurich8093Switzerland
| | - Seraina Kaelin
- Swiss Federal Institute of Technology Zurich (ETHZ)Department of Chemistry and Applied BiosciencesInstitute of Pharmaceutical SciencesVladimir‐Prelog‐Weg 3Zurich8093Switzerland
| | - Jean‐Christophe Leroux
- Swiss Federal Institute of Technology Zurich (ETHZ)Department of Chemistry and Applied BiosciencesInstitute of Pharmaceutical SciencesVladimir‐Prelog‐Weg 3Zurich8093Switzerland
| |
Collapse
|
22
|
Xiang Q, Wu Z, Tian EK, Nong S, Liao W, Zheng W. Gold Nanoparticle Drug Delivery System: Principle and Application. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.2934] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In recent years, gold nanoparticles (GNPs) have gradually become a major choice of drug delivery cargoes due to unique properties. Compared to traditional bulk solid gold, GNPs have basic physical and chemical advantages, such as a larger surface area-to-volume ratio and easier surface
modification. Furthermore, these have excellent biocompatibility, can induce the directional adsorption and enrichment of biological macromolecules, help retain biological macromolecule activity, and cause low harm to the human body. All these make GNPs good drug delivery cargoes. The present
study introduces the properties of GNPs, including factors that affect the properties and synthesis. Then, focus was given on the application in drug delivery, not only on the molecular mechanism, but also on the clinical application. Furthermore, the properties and applications of peptide
GNPs were also introduced. Finally, the challenges and prospects of GNPs for drug delivery were summarized.
Collapse
Affiliation(s)
- Qianrong Xiang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Sichuan University, West China School of Stomatology, Chengdu 610064, China
| | - Zhuoxuan Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Sichuan University, West China School of Stomatology, Chengdu 610064, China
| | - Er-Kang Tian
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Sichuan University, West China School of Stomatology, Chengdu 610064, China
| | - Shiqi Nong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Sichuan University, West China School of Stomatology, Chengdu 610064, China
| | - Wen Liao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Sichuan University, West China School of Stomatology, Chengdu 610064, China
| | - Wenyue Zheng
- Departments of Obstetrics & Gynecology and Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
23
|
Kanwal S, Naveed M, Arshad A, Arshad A, Firdous F, Faisal A, Yameen B. Reduction-Sensitive Dextran-Paclitaxel Polymer-Drug Conjugate: Synthesis, Self-Assembly into Nanoparticles, and In Vitro Anticancer Efficacy. Bioconjug Chem 2021; 32:2516-2529. [PMID: 34762796 DOI: 10.1021/acs.bioconjchem.1c00492] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Delivery systems that can encapsulate a precise amount of drug and offer a spatiotemporally controlled drug release are being actively sought for safe yet effective cancer therapy. Compared to polymer nanoparticle (NP)-based delivery systems that rely on physical drug encapsulation, NPs derived from stimuli-sensitive covalent polymer-drug conjugates (PDCs) have emerged as promising alternatives offering precise control over drug dosage and spatiotemporal drug release. Herein, we report a reduction-sensitive PDC "Dex-SS-PTXL" synthesized by conjugating dextran and paclitaxel (PTXL) through a disulfide bond-bearing linker. The synthesized Dex-SS-PTXL PDC with a precise degree of substitution in terms of the percentage of repeat units of dextran covalently conjugated to PTXL (27 ± 0.6%) and the amount of drug carried by the PDC (39 ± 1.4 wt %) was found to self-assemble into spherical NPs with an average size of 110 ± 34 nm and a ζ-potential of -14.09 ± 8 mV. The reduction-sensitive Dex-SS-PTXL NPs were found to release PTXL exclusively in response to the reducing agent concentration reflective of the intracellular reducing environment of the tumor cells. Challenging BT-549 and MCF-7 cells with Dex-SS-PTXL NPs revealed significant cytotoxicity, while the IC50 values and the mode of action (mitotic arrest) of Dex-SS-PTXL NPs were found to be comparable to those of free PTXL, highlighting the active nature of the intracellularly released drug. The developed PDC with its unique ability to self-assemble into NPs and stimuli-responsive drug release can enhance the success of the NP-based drug delivery systems during clinical translation.
Collapse
Affiliation(s)
- Sidra Kanwal
- Department of Chemistry and Chemical Engineering, Syed Babar Ali School of Science and Engineering (SBASSE), Lahore University of Management Sciences (LUMS), Lahore 54792, Pakistan
| | - Muhammad Naveed
- Cancer Therapeutics Laboratory, Department of Biology, Syed Babar Ali School of Science and Engineering (SBASSE), Lahore University of Management Sciences (LUMS), Lahore 54792, Pakistan
| | - Ali Arshad
- Department of Chemistry and Chemical Engineering, Syed Babar Ali School of Science and Engineering (SBASSE), Lahore University of Management Sciences (LUMS), Lahore 54792, Pakistan
| | - Azka Arshad
- Department of Chemistry and Chemical Engineering, Syed Babar Ali School of Science and Engineering (SBASSE), Lahore University of Management Sciences (LUMS), Lahore 54792, Pakistan
| | - Farhat Firdous
- Department of Chemistry and Chemical Engineering, Syed Babar Ali School of Science and Engineering (SBASSE), Lahore University of Management Sciences (LUMS), Lahore 54792, Pakistan
| | - Amir Faisal
- Cancer Therapeutics Laboratory, Department of Biology, Syed Babar Ali School of Science and Engineering (SBASSE), Lahore University of Management Sciences (LUMS), Lahore 54792, Pakistan
| | - Basit Yameen
- Department of Chemistry and Chemical Engineering, Syed Babar Ali School of Science and Engineering (SBASSE), Lahore University of Management Sciences (LUMS), Lahore 54792, Pakistan
| |
Collapse
|
24
|
Khan F, Atif M, Haseen M, Kamal S, Khan MS, Shahid S, Nami SAA. Synthesis, classification and properties of hydrogels: their applications in drug delivery and agriculture. J Mater Chem B 2021; 10:170-203. [PMID: 34889937 DOI: 10.1039/d1tb01345a] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Absorbent polymers or hydrogel polymer materials have an enhanced water retention capacity and are widely used in agriculture and medicine. The controlled release of bioactive molecules (especially drug proteins) by hydrogels and the encapsulation of living cells are some of the active areas of drug discovery research. Hydrogel-based delivery systems may result in a therapeutically advantageous outcome for drug delivery. They can provide various sequential therapeutic agents including macromolecular drugs, small molecule drugs, and cells to control the release of molecules. Due to their controllable degradability, ability to protect unstable drugs from degradation and flexible physical properties, hydrogels can be used as a platform in which various chemical and physical interactions with encapsulated drugs for controlled release in the system can be studied. Practically, hydrogels that possess biodegradable properties have aroused greater interest in drug delivery systems. The original three-dimensional structure gets broken down into non-toxic substances, thus confirming the excellent biocompatibility of the gel. Chemical crosslinking is a resource-rich method for forming hydrogels with excellent mechanical strength. But in some cases the crosslinker used in the synthesis of the hydrogels may cause some toxicity. However, the physically cross-linked hydrogel preparative method is an alternative solution to overcome the toxicity of cross-linkers. Hydrogels that are responsive to stimuli formed from various natural and synthetic polymers can show significant changes in their properties under external stimuli such as temperature, pH, light, ion changes, and redox potential. Stimulus-responsive hydrogels have a wider range of applications in biomedicine including drug delivery, gene delivery and tissue regeneration. Stimulus-responsive hydrogels loaded with multiple drugs show controlled and sustained drug release and can act as drug carriers. By integrating stimulus-responsive hydrogels, such as those with improved thermal responsiveness, pH responsiveness and dual responsiveness, into textile materials, advanced functions can be imparted to the textile materials, thereby improving the moisture and water retention performance, environmental responsiveness, aesthetic appeal, display and comfort of textiles. This review explores the stimuli-responsive hydrogels in drug delivery systems and examines super adsorbent hydrogels and their application in the field of agriculture.
Collapse
Affiliation(s)
- Faisal Khan
- Department of Chemistry, Aligarh Muslim University, Aligarh 202002, India.
| | - Mohd Atif
- Department of Chemistry, Aligarh Muslim University, Aligarh 202002, India.
| | - Mohd Haseen
- Department of Chemistry, Aligarh Muslim University, Aligarh 202002, India.
| | - Shahid Kamal
- Department of Chemistry, Aligarh Muslim University, Aligarh 202002, India.
| | - Mohd Shoeb Khan
- Interdisciplinary Nanotechnology Centre, Aligarh Muslim University, Aligarh 202002, India
| | - Shumaila Shahid
- Division of Plant Pathology, ICAR-Indian Agricultural Research Institute, New Delhi 110012, India
| | - Shahab A A Nami
- Department of Chemistry, Aligarh Muslim University, Aligarh 202002, India.
| |
Collapse
|
25
|
Zhang R, Nie T, Fang Y, Huang H, Wu J. Poly(disulfide)s: From Synthesis to Drug Delivery. Biomacromolecules 2021; 23:1-19. [PMID: 34874705 DOI: 10.1021/acs.biomac.1c01210] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bioresponsive polymers have been widely used in drug delivery because of their degradability. For example, poly(disulfide)s with repeating disulfide bonds in the main chain have attracted considerable research attention. The characteristics of the disulfide bonds, including their dynamic and reversible properties and their responsiveness to stimuli such as reductants, light, heat, and mechanical force, make them ideal platforms for on-demand drug delivery. This review introduces the synthesis methods and applications of poly(disulfide)s. Furthermore, the synthesis methods of poly(disulfide)s are classified on the basis of the monomers used: oxidative step-growth polymerization with dithiols, ring-opening polymerization with cyclic disulfides, and polymerization with linear disulfides. In addition, recent advances in poly(disulfide)s for the delivery of small-molecule or biomacromolecular drugs are discussed. Quantum-dot-loaded poly(disulfide) delivery systems for imaging are also included. This review provides an overview of the various design strategies employed in the construction of poly(disulfide) platforms to inspire new applications in the field of drug delivery.
Collapse
Affiliation(s)
- Ruhe Zhang
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province, School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| | - Tianqi Nie
- The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Yifen Fang
- Department of Cardiology, The Affiliated TCM Hospital of Guangzhou Medical University, Guangzhou 510180, China
| | - Hai Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jun Wu
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province, School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| |
Collapse
|
26
|
García-Garrido E, Cordani M, Somoza Á. Modified Gold Nanoparticles to Overcome the Chemoresistance to Gemcitabine in Mutant p53 Cancer Cells. Pharmaceutics 2021; 13:2067. [PMID: 34959348 PMCID: PMC8703659 DOI: 10.3390/pharmaceutics13122067] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/27/2021] [Accepted: 11/28/2021] [Indexed: 12/29/2022] Open
Abstract
Mutant p53 proteins result from missense mutations in the TP53 gene, the most mutated in human cancer, and have been described to contribute to cancer initiation and progression. Therapeutic strategies for targeting mutant p53 proteins in cancer cells are limited and have proved unsuitable for clinical application due to problems related to drug delivery and toxicity to healthy tissues. Therefore, the discovery of efficient and safe therapeutic strategies that specifically target mutant p53 remains challenging. In this study, we generated gold nanoparticles (AuNPs) chemically modified with low molecular branched polyethylenimine (bPEI) for the efficient delivery of gapmers targeting p53 mutant protein. The AuNPs formulation consists of a combination of polymeric mixed layer of polyethylene glycol (PEG) and PEI, and layer-by-layer assembly of bPEI through a sensitive linker. These nanoparticles can bind oligonucleotides through electrostatic interactions and release them in the presence of a reducing agent as glutathione. The nanostructures generated here provide a non-toxic and powerful system for the delivery of gapmers in cancer cells, which significantly downregulated mutant p53 proteins and altered molecular markers related to cell growth and apoptosis, thus overcoming chemoresistance to gemcitabine.
Collapse
Affiliation(s)
- Eduardo García-Garrido
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Faraday 9, 28049 Madrid, Spain
| | - Marco Cordani
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Faraday 9, 28049 Madrid, Spain
| | - Álvaro Somoza
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Faraday 9, 28049 Madrid, Spain
- Unidad Asociada al Centro Nacional de Biotecnología (CSIC), Darwin 3, 28049 Madrid, Spain
| |
Collapse
|
27
|
Moradi P, Hasanzadeh A, Radmanesh F, Rajai Daryasarei S, Hosseini ES, Kiani J, Shahbazi A, Nourizadeh H, Eslami M, Dorgalaleh A, Sahlolbei M, Hamblin MR, Karimi M. Smart arginine-equipped polycationic nanoparticles for p/CRISPR delivery into cells. NANOTECHNOLOGY 2021; 33:075104. [PMID: 34727527 DOI: 10.1088/1361-6528/ac357a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 11/02/2021] [Indexed: 06/13/2023]
Abstract
An efficient and safe delivery system for the transfection of CRISPR plasmid (p/CRISPR) into target cells can open new avenues for the treatment of various diseases. Herein, we design a novel nonvehicle by integrating an arginine-disulfide linker with low-molecular-weight PEI (PEI1.8k) for the delivery of p/CRISPR. These PEI1.8k-Arg nanoparticles facilitate the plasmid release and improve both membrane permeability and nuclear localization, thereby exhibiting higher transfection efficiency compared to native PEI1.8kin the delivery of nanocomplexes composed of PEI1.8k-Arg and p/CRISPR into conventional cells (HEK 293T). This nanovehicle is also able to transfect p/CRISPR in a wide variety of cells, including hard-to-transfect primary cells (HUVECs), cancer cells (HeLa), and neuronal cells (PC-12) with nearly 5-10 times higher efficiency compared to the polymeric gold standard transfection agent. Furthermore, the PEI1.8k-Arg nanoparticles can edit the GFP gene in the HEK 293T-GFP reporter cell line by delivering all possible forms of CRISPR/Cas9 system (e.g. plasmid encoding Cas9 and sgRNA targeting GFP, and Cas9/sgRNA ribonucleoproteins (RNPs) as well as Cas9 expression plasmid andin vitro-prepared sgRNA) into HEK 293T-GFP cells. The successful delivery of p/CRISPR into local brain tissue is also another remarkable capability of these nanoparticles. In view of all the exceptional benefits of this safe nanocarrier, it is expected to break new ground in the field of gene editing, particularly for therapeutic purposes.
Collapse
Affiliation(s)
- Pardis Moradi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran, Iran
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Akbar Hasanzadeh
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Radmanesh
- Uro-Oncology Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Saideh Rajai Daryasarei
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran, Iran
| | - Elaheh Sadat Hosseini
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran, Iran
| | - Jafar Kiani
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Shahbazi
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Helena Nourizadeh
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Eslami
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Applied Biotechnology Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Akbar Dorgalaleh
- Department of Hematology and Blood Transfusion, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Sahlolbei
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Mahdi Karimi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran, Iran
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Research Center for Science and Technology in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Applied Biotechnology Research Centre, Tehran Medical Science, Islamic Azad University, Tehran, Iran
| |
Collapse
|
28
|
Yao H, Jiang D, Dong G, Sun J, Sun S, Li L, Zheng F, Xiong W. Near infrared imaging of intracellular GSH by AuNCs@MnO 2 core-shell nanoparticles based on the absorption competition mechanism. Analyst 2021; 146:5115-5123. [PMID: 34269357 DOI: 10.1039/d1an00839k] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Dynamically monitoring intracellular glutathione (GSH), a crucial biomarker of oxidative stress, is of significance for the diagnosis and treatment of certain diseases. Although manganese dioxide (MnO2) based GSH fluorescent sensors have exhibited high sensitivity and good selectivity owing to the specific reactivity between GSH and MnO2, near-infrared (NIR) MnO2 based nanoprobes for GSH detection are scarce. Herein, we have developed a NIR activatable fluorescence nanoprobe for the imaging and determination of intracellular GSH based on a core-shell nanoparticle, consisting of NIR emitted gold nanocluster doped silica as the fluorescent core and manganese dioxide as the GSH-responsive shell (named AuNCs@MnO2). Due to the absorption competition mechanism, the outer MnO2 shell rather than the inner AuNCs core preferentially absorbed the excitation light, thus leading to fluorescence quenching of the inner AuNCs core. Upon addition of GSH, the fluorescence of the nanoprobe restored along with the reduction of MnO2 to Mn2+ because of the absorption competition disappearance-induced emission. The activatable fluorescence linearly increased upon changing the GSH concentration in the range of 2 to 5000 μM with a detection limit of 0.67 μM. The cytotoxicity test shows that the AuNCs@MnO2 nanoprobes have a good biocompatibility. After entering the cancer cells, the intracellular GSH degraded the outermost MnO2 shell and initiated the NIR fluorescence restoration of AuNCs, which can be used to monitor the dynamic change of intracellular GSH. This strategy provides an NIR-activatable way to detect GSH levels in living cells and offers a promising platform for the diagnosis and treatment of GSH-related diseases.
Collapse
Affiliation(s)
- Haiyang Yao
- School of Environmental & Chemical Engineering, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu 212003, China.
| | - Difei Jiang
- School of Environmental & Chemical Engineering, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu 212003, China.
| | - Gaoqiu Dong
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Jiamin Sun
- School of Environmental & Chemical Engineering, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu 212003, China.
| | - Shasha Sun
- School of Environmental & Chemical Engineering, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu 212003, China.
| | - Lingling Li
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Fenfen Zheng
- School of Environmental & Chemical Engineering, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu 212003, China.
| | - Weiwei Xiong
- School of Environmental & Chemical Engineering, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu 212003, China. and Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| |
Collapse
|
29
|
Kumar R, Santa Chalarca CF, Bockman MR, Bruggen CV, Grimme CJ, Dalal RJ, Hanson MG, Hexum JK, Reineke TM. Polymeric Delivery of Therapeutic Nucleic Acids. Chem Rev 2021; 121:11527-11652. [PMID: 33939409 DOI: 10.1021/acs.chemrev.0c00997] [Citation(s) in RCA: 196] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The advent of genome editing has transformed the therapeutic landscape for several debilitating diseases, and the clinical outlook for gene therapeutics has never been more promising. The therapeutic potential of nucleic acids has been limited by a reliance on engineered viral vectors for delivery. Chemically defined polymers can remediate technological, regulatory, and clinical challenges associated with viral modes of gene delivery. Because of their scalability, versatility, and exquisite tunability, polymers are ideal biomaterial platforms for delivering nucleic acid payloads efficiently while minimizing immune response and cellular toxicity. While polymeric gene delivery has progressed significantly in the past four decades, clinical translation of polymeric vehicles faces several formidable challenges. The aim of our Account is to illustrate diverse concepts in designing polymeric vectors towards meeting therapeutic goals of in vivo and ex vivo gene therapy. Here, we highlight several classes of polymers employed in gene delivery and summarize the recent work on understanding the contributions of chemical and architectural design parameters. We touch upon characterization methods used to visualize and understand events transpiring at the interfaces between polymer, nucleic acids, and the physiological environment. We conclude that interdisciplinary approaches and methodologies motivated by fundamental questions are key to designing high-performing polymeric vehicles for gene therapy.
Collapse
Affiliation(s)
- Ramya Kumar
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | | | - Matthew R Bockman
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Craig Van Bruggen
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Christian J Grimme
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Rishad J Dalal
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Mckenna G Hanson
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Joseph K Hexum
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
30
|
Jia H, Wang X, Chen Z, Liu S, Zhou T, Zhou H, Guo T. Zinc(II)-Dipicolylamine Analogs Mediated PEI1.8k/pDNA Vector: Effect of Ligand Structure on the Gene Transport Process. Macromol Biosci 2021; 21:e2100048. [PMID: 33861507 DOI: 10.1002/mabi.202100048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 03/08/2021] [Indexed: 11/06/2022]
Abstract
Zinc ion complexes of dipicolylamine analogs, due to the strong synergistic effect between the Zn2+ complex of containing polypyridine derivatives and polycations in each key step of pDNA transport, have been used as the third component to mediate polyethyleneimine with molecular weight 1.8 kDa (PEI1.8k)/DNA gene delivery system. And the effects of different structural characteristics, such as the number of pyridinamine ligands, the hydrophilic-hydrophobicity of the adjacent groups, on the in vitro transfection performance of the ternary complex are systematically investigated. This ternary hybrid system provides an effective strategy to improve the gene delivery of cationic polymers.
Collapse
Affiliation(s)
- Huiting Jia
- Key Laboratory, of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Xindong Wang
- Key Laboratory, of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Zhaoming Chen
- Key Laboratory, of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Shuai Liu
- Key Laboratory, of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Tongtong Zhou
- Key Laboratory, of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Hao Zhou
- Department of Biochemistry and Molecular Biology, College of Life Science, Nankai University, Tianjin, 300071, China
| | - Tianying Guo
- Key Laboratory, of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| |
Collapse
|
31
|
Poudel K, Banstola A, Gautam M, Soe ZC, Pham LM, Jeong JH, Choi HG, Ku SK, Yong CS, Tran TH, Kim JO. Redox/photo dual-responsive, self-targeted, and photosensitizer-laden bismuth sulfide nanourchins for combination therapy in cancer. NANOSCALE 2021; 13:1231-1247. [PMID: 33406178 DOI: 10.1039/d0nr07736d] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Targeted and stimuli-sensitive nanobombs for the release of therapeutic agents after laser irradiation of the tumor site are gaining widespread attention as personalized anticancer regimens. In this study, redox and photo dual-responsive, folate receptor-targeted nanourchin carriers for chemo-, photodynamic, and photothermal therapy were constructed by the amalgamation of an outer layer of polyethylene glycol (PEG)-S-S-methotrexate (MTX) and an inner core of indocyanine green (ICG)-loaded bismuth sulfide (Bi2S3) nanoparticles for cancer treatment. MTX introduces the carrier to folate receptors resulting in the internalization of nanoparticles into cancer cells, specifically and increasingly. In the reducing environment inside cancer cells, MTX was cleaved, resulting in a burst release that effectively inhibited tumor growth. Simultaneously, the fusion of Bi2S3 and ICG in the inner core absorbed energy from a near-infrared radiation (NIR) laser to generate heat and reactive oxygen species, which further ablated the tumors and synergistically enhanced the anticancer activity of MTX. These results indicate the successful preparation of combined nanourchins (NUs) showing GSH-induced and laser-responsive release of MTX and ICG, accompanied by hyperthermia via Bi2S3 and ICG. Effective in vitro cellular internalization, cellular cytotoxicity, and pro-apoptotic behavior of the nanosystem were achieved through a targeting, redox, and NIR-responsive combination strategy. In vivo biodistribution and photothermal imaging also revealed tumor-selective and -retentive, as well as thermally responsive attributes. Ultimately, this in vivo antitumor study shows an effective tumor ablation by these nanourchins without affecting the vital organs. Our findings indicate that using these targeted redox- and laser-responsive combination therapeutic carriers can be a promising strategy in folate receptor-expressing tumors.
Collapse
Affiliation(s)
- Kishwor Poudel
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea.
| | - Asmita Banstola
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| | - Milan Gautam
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea.
| | - Zar Chi Soe
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea.
| | - Le Minh Pham
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea.
| | - Jee-Heon Jeong
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea.
| | - Han-Gon Choi
- College of Pharmacy, Hanyang University, 55, Hanyangdaehak-ro, Sangnok-gu, Ansan 426-791, Republic of Korea
| | - Sae Kwang Ku
- Department of Anatomy and Histology, College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Republic of Korea.
| | - Chul Soon Yong
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea.
| | - Tuan Hiep Tran
- Faculty of Pharmacy, PHENIKAA University, Yen Nghia, Ha Dong, Hanoi 12116, Vietnam. and PHENIKAA Research and Technology Institute (PRATI), A&A Green Phoenix Group JSC, No. 167 Hoang Ngan, Trung Hoa, Cau Giay, Hanoi 11313, Vietnam
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea.
| |
Collapse
|
32
|
Xu C, Yu B, Qi Y, Zhao N, Xu F. Versatile Types of Cyclodextrin-Based Nucleic Acid Delivery Systems. Adv Healthc Mater 2021; 10:e2001183. [PMID: 32935932 DOI: 10.1002/adhm.202001183] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/23/2020] [Indexed: 12/16/2022]
Abstract
Nowadays, nucleic acid therapy has become a promising way for the treatment of various malignant diseases. Cyclodextrin (CD)-based nucleic acid delivery systems have attracted widespread attention due to the favorable chemical structures and excellent biological properties of CD. Recently, a variety of CD-based nucleic acid delivery systems has been designed according to the different functions of CD for flexible gene therapies. In this review, the construction strategies and biomedical applications of CD-based nucleic acid delivery systems are mainly focused on. The review begins with an introduction to the synthesis and properties of simple CD-grafted polycations. Thereafter, CD-related supramolecular assemblies based on different guest components are discussed in detail. Finally, different CD-based organic/inorganic nanohybrids and their relevant functions are demonstrated. It is hoped that this brief review will motivate the delicate design of CD-based nucleic acid delivery systems for potential clinical applications.
Collapse
Affiliation(s)
- Chen Xu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology Ministry of Education) Beijing Laboratory of Biomedical Materials Beijing University of Chemical Technology Beijing 100029 China
| | - Bingran Yu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology Ministry of Education) Beijing Laboratory of Biomedical Materials Beijing University of Chemical Technology Beijing 100029 China
| | - Yu Qi
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology Ministry of Education) Beijing Laboratory of Biomedical Materials Beijing University of Chemical Technology Beijing 100029 China
| | - Nana Zhao
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology Ministry of Education) Beijing Laboratory of Biomedical Materials Beijing University of Chemical Technology Beijing 100029 China
| | - Fu‐Jian Xu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology Ministry of Education) Beijing Laboratory of Biomedical Materials Beijing University of Chemical Technology Beijing 100029 China
| |
Collapse
|
33
|
Jeong HY, Kim H, Lee M, Hong J, Lee JH, Kim J, Choi MJ, Park YS, Kim SC. Development of HER2-Specific Aptamer-Drug Conjugate for Breast Cancer Therapy. Int J Mol Sci 2020; 21:ijms21249764. [PMID: 33371333 PMCID: PMC7767363 DOI: 10.3390/ijms21249764] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/18/2020] [Accepted: 12/18/2020] [Indexed: 02/07/2023] Open
Abstract
In this study, HER2 RNA aptamers were conjugated to mertansine (DM1) and the anti-cancer effectiveness of the conjugate was evaluated in HER2-overexpressing breast cancer models. The conjugate of HER2 aptamer and anticancer drug DM1 (aptamer-drug conjugate, ApDC) was prepared and analyzed using HPLC and mass spectrometry. The cell-binding affinity and cytotoxicity of the conjugate were determined using confocal microscopy and WST-1 assay. The in vivo anti-tumoral efficacy of ApDC was also evaluated in mice carrying BT-474 breast tumors overexpressing HER2. The synthesized HER2-specific RNA aptamers were able to specifically and efficiently bind to HER-positive BT-474 breast cancer cells, but not to HER2-negative MDA-MB-231 breast cancer cells. Also, the HER2-specific ApDC showed strong toxicity to the target cells, BT-474, but not to MDA-MB-231 cells. According to the in vivo analyses drawn from the mouse xenografts of BT-747 tumor, the ApDC was able to more effectively inhibit the tumor growth. Compared to the control group, the mice treated with the ApDC showed a significant reduction of tumor growth. Besides, any significant body weight losses or hepatic toxicities were monitored in the ApDC-treated mice. This research suggests the HER2 aptamer-DM1 conjugate as a target-specific anti-cancer modality and provides experimental evidence supporting its enhanced effectiveness for HER2-overexpressing target tumors. This type of aptamer-conjugated anticancer drug would be utilized as a platform structure for the development of versatile targeted high-performance anticancer drugs by adopting the easy deformability and high affinity of aptamers.
Collapse
Affiliation(s)
- Hwa Yeon Jeong
- Biois Co., Ltd., Seoul 08390, Korea; (H.Y.J.); (H.K.); (M.L.); (J.H.); (J.H.L.); (J.K.)
| | - Hyeri Kim
- Biois Co., Ltd., Seoul 08390, Korea; (H.Y.J.); (H.K.); (M.L.); (J.H.); (J.H.L.); (J.K.)
| | - Myunghwa Lee
- Biois Co., Ltd., Seoul 08390, Korea; (H.Y.J.); (H.K.); (M.L.); (J.H.); (J.H.L.); (J.K.)
| | - Jinju Hong
- Biois Co., Ltd., Seoul 08390, Korea; (H.Y.J.); (H.K.); (M.L.); (J.H.); (J.H.L.); (J.K.)
| | - Joo Han Lee
- Biois Co., Ltd., Seoul 08390, Korea; (H.Y.J.); (H.K.); (M.L.); (J.H.); (J.H.L.); (J.K.)
| | - Jeonghyeon Kim
- Biois Co., Ltd., Seoul 08390, Korea; (H.Y.J.); (H.K.); (M.L.); (J.H.); (J.H.L.); (J.K.)
| | - Moon Jung Choi
- Department of Biomedical Laboratory Science, Yonsei University, Wonju 26493, Korea;
| | - Yong Serk Park
- Department of Biomedical Laboratory Science, Yonsei University, Wonju 26493, Korea;
- Correspondence: (Y.S.P.); (S.-C.K.); Tel.: +82-33-760-2448 (Y.S.P.); +82-2-6959-0363 (S.-C.K.)
| | - Sung-Chun Kim
- Biois Co., Ltd., Seoul 08390, Korea; (H.Y.J.); (H.K.); (M.L.); (J.H.); (J.H.L.); (J.K.)
- Correspondence: (Y.S.P.); (S.-C.K.); Tel.: +82-33-760-2448 (Y.S.P.); +82-2-6959-0363 (S.-C.K.)
| |
Collapse
|
34
|
Thomas RG, Surendran SP, Jeong YY. Tumor Microenvironment-Stimuli Responsive Nanoparticles for Anticancer Therapy. Front Mol Biosci 2020; 7:610533. [PMID: 33392264 PMCID: PMC7775573 DOI: 10.3389/fmolb.2020.610533] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 11/23/2020] [Indexed: 12/14/2022] Open
Abstract
Cancer is a disease that affects a large number of people all over the world. For treating cancer, nano-drug delivery system has been introduced recently with objective of increasing therapeutic efficiency of chemotherapeutic drug. The main characteristics of this system are the encapsulation of the insoluble chemotherapeutic cargo, increasing the period of circulation in the body, as well as the delivery of the drug at that specific site. Currently, the nano-drug delivery system based on the stimuli response is becoming more popular because of the extra features for controlling the drug release based on the internal atmosphere of cancer. This review provides a summary of different types of internal (pH, redox, enzyme, ROS, hypoxia) stimuli-responsive nanoparticle drug delivery systems as well as perspective for upcoming times.
Collapse
Affiliation(s)
- Reju George Thomas
- Department of Radiology, Chonnam National University Hwasun Hospital, Hwasun, South Korea
- BioMolecular Theranostics (BiT) Laboratory, Department of Biomedical Sciences, Chonnam National University Medical School, Chonnam National University Hwasun Hospital, Gwangju, South Korea
| | - Suchithra Poilil Surendran
- Department of Radiology, Chonnam National University Hwasun Hospital, Hwasun, South Korea
- BioMolecular Theranostics (BiT) Laboratory, Department of Biomedical Sciences, Chonnam National University Medical School, Chonnam National University Hwasun Hospital, Gwangju, South Korea
| | - Yong Yeon Jeong
- Department of Radiology, Chonnam National University Hwasun Hospital, Hwasun, South Korea
- BioMolecular Theranostics (BiT) Laboratory, Department of Biomedical Sciences, Chonnam National University Medical School, Chonnam National University Hwasun Hospital, Gwangju, South Korea
| |
Collapse
|
35
|
McErlean EM, Ziminska M, McCrudden CM, McBride JW, Loughran SP, Cole G, Mulholland EJ, Kett V, Buckley NE, Robson T, Dunne NJ, McCarthy HO. Rational design and characterisation of a linear cell penetrating peptide for non-viral gene delivery. J Control Release 2020; 330:1288-1299. [PMID: 33227336 DOI: 10.1016/j.jconrel.2020.11.037] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 11/16/2020] [Accepted: 11/19/2020] [Indexed: 01/19/2023]
Abstract
The design of a non-viral gene delivery system that can release a functional nucleic acid at the intracellular destination site is an exciting but also challenging proposition. The ideal gene delivery vector must be non-toxic, non-immunogenic, overcome extra- and intra-cellular barriers, protect the nucleic acid cargo from degradation with stability over a range of temperatures. A new 15 amino acid linear peptide termed CHAT was designed in this study with the goal of delivering DNA with high efficiency into cells in vitro and tissues in vivo. Rational design involved incorporation of key amino acids including arginine for nucleic acid complexation and cellular uptake, tryptophan to enhance hydrophobic interaction with cell membranes, histidine to facilitate endosomal escape and cysteine for stability and controlled cargo release. Six linear peptides were synthesised with strategic sequences and amino acid substitutions. Data demonstrated that all six peptides complexed pDNA to produce cationic nanoparticles less than 200 nm in diameter, but not all peptides resulted in successful transfection; indicating the influence of peptide design for endosomal escape. Peptide 4, now termed CHAT, was non-cytotoxic, traversed the plasma membrane of breast and prostate cancer cell lines, and elicited reporter-gene expression following intra-tumoural and intravenous delivery in vivo. CHAT presents an exciting new peptide for the delivery of nucleic acid therapeutics.
Collapse
Affiliation(s)
- Emma M McErlean
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Monika Ziminska
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Cian M McCrudden
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - John W McBride
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Stephen P Loughran
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Grace Cole
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Eoghan J Mulholland
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Vicky Kett
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Niamh E Buckley
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Tracy Robson
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons Ireland, 111 St Stephen's Green, Dublin 2, Ireland
| | - Nicholas J Dunne
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland; Centre for Medical Engineering Research, School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland; Advanced Manufacturing Research Centre (I-Form), School of Mechanical and Manufacturing Engineering, Dublin City University, Glasnevin, Dublin 9, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin 2, Ireland; Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Helen O McCarthy
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; School of Chemical Sciences, Dublin City University, Dublin 9, Ireland.
| |
Collapse
|
36
|
Pham SH, Choi Y, Choi J. Stimuli-Responsive Nanomaterials for Application in Antitumor Therapy and Drug Delivery. Pharmaceutics 2020; 12:E630. [PMID: 32635539 PMCID: PMC7408499 DOI: 10.3390/pharmaceutics12070630] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 07/03/2020] [Accepted: 07/04/2020] [Indexed: 01/14/2023] Open
Abstract
The new era of nanotechnology has produced advanced nanomaterials applicable to various fields of medicine, including diagnostic bio-imaging, chemotherapy, targeted drug delivery, and biosensors. Various materials are formed into nanoparticles, such as gold nanomaterials, carbon quantum dots, and liposomes. The nanomaterials have been functionalized and widely used because they are biocompatible and easy to design and prepare. This review mainly focuses on nanomaterials responsive to the external stimuli used in drug-delivery systems. To overcome the drawbacks of conventional therapeutics to a tumor, the dual- and multi-responsive behaviors of nanoparticles have been harnessed to improve efficiency from a drug delivery point of view. Issues and future research related to these nanomaterial-based stimuli sensitivities and the scope of stimuli-responsive systems for nanomedicine applications are discussed.
Collapse
Affiliation(s)
| | | | - Jonghoon Choi
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Korea; (S.H.P.); (Y.C.)
| |
Collapse
|
37
|
Blakney AK, Zhu Y, McKay PF, Bouton C, Yeow J, Tang J, Hu K, Samnuan K, Grigsby CL, Shattock RJ, Stevens MM. Big Is Beautiful: Enhanced saRNA Delivery and Immunogenicity by a Higher Molecular Weight, Bioreducible, Cationic Polymer. ACS NANO 2020; 14:5711-5727. [PMID: 32267667 PMCID: PMC7304921 DOI: 10.1021/acsnano.0c00326] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 04/08/2020] [Indexed: 05/18/2023]
Abstract
Self-amplifying RNA (saRNA) vaccines are highly advantageous, as they result in enhanced protein expression compared to mRNA (mRNA), thus minimizing the required dose. However, previous delivery strategies were optimized for siRNA or mRNA and do not necessarily deliver saRNA efficiently due to structural differences of these RNAs, thus motivating the development of saRNA delivery platforms. Here, we engineer a bioreducible, linear, cationic polymer called "pABOL" for saRNA delivery and show that increasing its molecular weight enhances delivery both in vitro and in vivo. We demonstrate that pABOL enhances protein expression and cellular uptake via both intramuscular and intradermal injection compared to commercially available polymers in vivo and that intramuscular injection confers complete protection against influenza challenge. Due to the scalability of polymer synthesis and ease of formulation preparation, we anticipate that this polymer is highly clinically translatable as a delivery vehicle for saRNA for both vaccines and therapeutics.
Collapse
Affiliation(s)
- Anna K. Blakney
- Department
of Infectious Diseases, Imperial College
London, Norfolk Place, London, W2 1PG, U.K.
| | - Yunqing Zhu
- Department
of Materials, Department of Bioengineering, Institute of Biomedical
Engineering, Imperial College London, London, SW7 2AZ, U.K.
- School
of Materials Science and Engineering, Tongji
University, Shanghai, 200092, China
| | - Paul F. McKay
- Department
of Infectious Diseases, Imperial College
London, Norfolk Place, London, W2 1PG, U.K.
| | - Clément
R. Bouton
- Department
of Infectious Diseases, Imperial College
London, Norfolk Place, London, W2 1PG, U.K.
| | - Jonathan Yeow
- Department
of Materials, Department of Bioengineering, Institute of Biomedical
Engineering, Imperial College London, London, SW7 2AZ, U.K.
| | - Jiaqing Tang
- Department
of Materials, Department of Bioengineering, Institute of Biomedical
Engineering, Imperial College London, London, SW7 2AZ, U.K.
| | - Kai Hu
- Department
of Infectious Diseases, Imperial College
London, Norfolk Place, London, W2 1PG, U.K.
| | - Karnyart Samnuan
- Department
of Infectious Diseases, Imperial College
London, Norfolk Place, London, W2 1PG, U.K.
| | - Christopher L. Grigsby
- Department
of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 171 65, Sweden
| | - Robin J. Shattock
- Department
of Infectious Diseases, Imperial College
London, Norfolk Place, London, W2 1PG, U.K.
| | - Molly M. Stevens
- Department
of Materials, Department of Bioengineering, Institute of Biomedical
Engineering, Imperial College London, London, SW7 2AZ, U.K.
- Department
of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 171 65, Sweden
| |
Collapse
|
38
|
Zhu H, Liu C, Zhang H, Jia P, Li Z, Zhang X, Yu Y, Sheng W, Zhu B. A Simple Long-wavelength Fluorescent Probe for Simultaneous Discrimination of Cysteine/Homocysteine and Glutathione/Hydrogen Sulfide with Two Separated Fluorescence Emission Channels by Single Wavelength Excitation. ANAL SCI 2020; 36:255-259. [PMID: 31588065 DOI: 10.2116/analsci.19p214] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Small molecular biothiols, such as cysteine (Cys), homocysteine (Hcy), reduced glutathione (GSH), and hydrogen sulfide (H2S), play crucial parts in regulating the redox balance of life activities, regulating normal physiological activities and preventing various diseases. Quantitative analysis of these important small molecular substances is very important for revealing their diverse physiological and pathological effects. Although many fluorescent probes have been reported to detect biothiols in cells, it is still not sufficiently advanced to detect biothiols with separated fluorescence emission peak by same wavelength excitation. In our work, we designed a simple conjugate of Nile red and NBD (7-nitro-1,2,3-benzoxadiazole) as long-wavelength fluorescent probe NR-NBD for the simultaneous discrimination of these biothiols at single wavelength excitation. Probe NR-NBD could efficiently discriminate Cys/Hcy, GSH and H2S by two separated fluorescence emission channels and absorption spectra. Importantly, probe NR-NBD has excellent specificity and sensitivity towards the monitoring of endogenous/exogenous Cys/Hcy and GSH/H2S in living cells and zebrafish.
Collapse
Affiliation(s)
- Hanchuang Zhu
- School of Water Conservancy and Environment, University of Jinan, Shandong Provincial Engineering Technology Research Center for Ecological Carbon Sink and Capture Utilization
| | - Caiyun Liu
- School of Water Conservancy and Environment, University of Jinan, Shandong Provincial Engineering Technology Research Center for Ecological Carbon Sink and Capture Utilization
| | - Hanming Zhang
- School of Water Conservancy and Environment, University of Jinan, Shandong Provincial Engineering Technology Research Center for Ecological Carbon Sink and Capture Utilization
| | - Pan Jia
- School of Water Conservancy and Environment, University of Jinan, Shandong Provincial Engineering Technology Research Center for Ecological Carbon Sink and Capture Utilization
| | - Zilu Li
- School of Water Conservancy and Environment, University of Jinan, Shandong Provincial Engineering Technology Research Center for Ecological Carbon Sink and Capture Utilization
| | - Xue Zhang
- School of Water Conservancy and Environment, University of Jinan, Shandong Provincial Engineering Technology Research Center for Ecological Carbon Sink and Capture Utilization
| | - Yamin Yu
- School of Water Conservancy and Environment, University of Jinan, Shandong Provincial Engineering Technology Research Center for Ecological Carbon Sink and Capture Utilization
| | - Wenlong Sheng
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences)
| | - Baocun Zhu
- School of Water Conservancy and Environment, University of Jinan, Shandong Provincial Engineering Technology Research Center for Ecological Carbon Sink and Capture Utilization
| |
Collapse
|
39
|
Hao B, Li W, Zhang S, Zhu Y, Li Y, Ding A, Huang X. A facile PEG/thiol-functionalized nanographene oxide carrier with an appropriate glutathione-responsive switch. Polym Chem 2020. [DOI: 10.1039/d0py00110d] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A novel nanographene oxide/PEG-based bioreduction-responsive smart drug delivery system with a GSH-responsive disulfide linker as the controlled release switch can selectively release anti-cancer drugs in cancer cells.
Collapse
Affiliation(s)
- Bingjie Hao
- Key Laboratory of Synthetic and Self-Assembly Chemistry for Organic Functional Molecules
- Center for Excellence in Molecular Synthesis
- Shanghai Institute of Organic Chemistry
- University of Chinese Academy of Sciences
- Chinese Academy of Sciences
| | - Wei Li
- Division of Physical Biology and Bioimaging Center
- Shanghai Synchrotron Radiation Facility
- Shanghai Institute of Applied Physics
- Chinese Academy of Sciences
- University of Chinese Academy of Sciences
| | - Sen Zhang
- Key Laboratory of Synthetic and Self-Assembly Chemistry for Organic Functional Molecules
- Center for Excellence in Molecular Synthesis
- Shanghai Institute of Organic Chemistry
- University of Chinese Academy of Sciences
- Chinese Academy of Sciences
| | - Ying Zhu
- Division of Physical Biology and Bioimaging Center
- Shanghai Synchrotron Radiation Facility
- Shanghai Institute of Applied Physics
- Chinese Academy of Sciences
- University of Chinese Academy of Sciences
| | - Yongjun Li
- Key Laboratory of Synthetic and Self-Assembly Chemistry for Organic Functional Molecules
- Center for Excellence in Molecular Synthesis
- Shanghai Institute of Organic Chemistry
- University of Chinese Academy of Sciences
- Chinese Academy of Sciences
| | - Aishun Ding
- Key Laboratory of Synthetic and Self-Assembly Chemistry for Organic Functional Molecules
- Center for Excellence in Molecular Synthesis
- Shanghai Institute of Organic Chemistry
- University of Chinese Academy of Sciences
- Chinese Academy of Sciences
| | - Xiaoyu Huang
- Key Laboratory of Synthetic and Self-Assembly Chemistry for Organic Functional Molecules
- Center for Excellence in Molecular Synthesis
- Shanghai Institute of Organic Chemistry
- University of Chinese Academy of Sciences
- Chinese Academy of Sciences
| |
Collapse
|
40
|
Su D, Coste M, Diaconu A, Barboiu M, Ulrich S. Cationic dynamic covalent polymers for gene transfection. J Mater Chem B 2020; 8:9385-9403. [DOI: 10.1039/d0tb01836h] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Dynamic covalent polymers have revealed strong potential in gene delivery, thanks to their versatile self-assembly, adaptive and responsive behaviors.
Collapse
Affiliation(s)
- Dandan Su
- Institut Européen des Membranes
- Adaptive Supramolecular Nanosystems Group
- University of Montpellier
- ENSCM
- CNRS
| | - Maëva Coste
- Institut des Biomolécules Max Mousseron (IBMM)
- CNRS
- Université of Montpellier
- ENSCM
- Montpellier
| | - Andrei Diaconu
- Petru Poni” Institute of Macromolecular Chemistry of Romanian Academy
- Iasi
- Romania
| | - Mihail Barboiu
- Institut Européen des Membranes
- Adaptive Supramolecular Nanosystems Group
- University of Montpellier
- ENSCM
- CNRS
| | - Sébastien Ulrich
- Institut des Biomolécules Max Mousseron (IBMM)
- CNRS
- Université of Montpellier
- ENSCM
- Montpellier
| |
Collapse
|
41
|
Pretto C, van Hest JCM. Versatile Reversible Cross-Linking Strategy to Stabilize CCMV Virus Like Particles for Efficient siRNA Delivery. Bioconjug Chem 2019; 30:3069-3077. [PMID: 31765129 PMCID: PMC6923791 DOI: 10.1021/acs.bioconjchem.9b00731] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
![]()
Virus like particles obtained from the Cowpea Chlorotic
Mottle
Virus (CCMV) represent an innovative platform for drug delivery applications.
Their unique reversible self-assembly properties as well as their
suitability for both cargo loading and functionalization make them
a versatile scaffold for numerous purposes. One of the main drawbacks
of this platform is however its limited stability at physiological
conditions. Herein, we report the development of a general reversible
cross-linking strategy involving the homobifunctional cross-linker
DTSSP (3,3′-dithiobis (sulfosuccinimidylpropionate)) which
is suitable for particle stabilization. This methodology is adaptable
to different CCMV variants in the presence or absence of a stabilizing
cargo without varying neither particle shape nor size thus extending
the potential use of these protein cages in nanomedical applications.
Cross-linked particles are stable at neutral pH and 37 °C and
they are capable of protecting loaded cargo against enzymatic digestion.
Furthermore, the reversible nature of the cross-linking ensures particle
disassembly when they are taken up by cells. This was demonstrated
via the highly effective delivery of active siRNA into cells.
Collapse
Affiliation(s)
- Chiara Pretto
- Eindhoven University of Technology , Institute for Complex Molecular Systems , PO Box 513, 5600 MB Eindhoven , The Netherlands
| | - Jan C M van Hest
- Eindhoven University of Technology , Institute for Complex Molecular Systems , PO Box 513, 5600 MB Eindhoven , The Netherlands
| |
Collapse
|
42
|
Xu Z, Qin T, Zhou X, Wang L, Liu B. Fluorescent probes with multiple channels for simultaneous detection of Cys, Hcy, GSH, and H2S. Trends Analyt Chem 2019. [DOI: 10.1016/j.trac.2019.115672] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
43
|
Affiliation(s)
- Yabin Meng
- Department of Biomedical Engineering, School of EngineeringSun Yat‐sen University Guangzhou 510006 P. R. China
| | - Shuyan Han
- Department of Biomedical Engineering, School of EngineeringSun Yat‐sen University Guangzhou 510006 P. R. China
| | - Zhipeng Gu
- College of Polymer Science and EngineeringState Key Laboratory of Polymer Materials EngineeringSichuan University Chengdu 610065 P. R. China
| | - Jun Wu
- Department of Biomedical Engineering, School of EngineeringSun Yat‐sen University Guangzhou 510006 P. R. China
| |
Collapse
|
44
|
|
45
|
Sang MM, Liu FL, Wang Y, Luo RJ, Huan XX, Han LF, Zhang ZT, Feng F, Qu W, Liu W, Zheng F. A novel redox/pH dual-responsive and hyaluronic acid-decorated multifunctional magnetic complex micelle for targeted gambogic acid delivery for the treatment of triple negative breast cancer. Drug Deliv 2019; 25:1846-1857. [PMID: 30334478 PMCID: PMC6225507 DOI: 10.1080/10717544.2018.1486472] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Gambogic acid (GA) is a naturally derived potent anticancer agent with extremely poor biocompatibility. In the present study, a novel of redox/pH dual-responsive multifunctional magnetic complex micelle (sPEG/HA/CSO-SS-Hex/Fe3O4/GA), which consisted of a reducible hexadecanol-modified chitosan oligosaccharide polymer micelle (CSO-SS-Hex) coated with hyaluronic acid (HA) and DCA grafted sheddable PEG-PLL (sPEG) copolymers and loaded with gambogic acid (GA) and Fe3O4 nanoparticles were developed for parenteral delivery for the treatment of triple negative breast cancer (TNBC). The ex vivo study showed that the sPEG shielded cationic HA/CSO-SS-Hex/Fe3O4/GA core at physiological pH but quickly shed off to re-expose the core due to its charge reversible property. The sPEG/HA/CSO-SS-Hex/Fe3O4/GA micelles effectively facilitated tumor-targeted GA delivery by HA, which is a targeting ligand for CD44 receptor of TNBC cells, meanwhile increase GA uptake at the acidic condition but diminished the drug uptake at neutral pH. The in vitro cellular uptake study and in vivo biodistribution and antitumor activity of the formulations were determined, all results showed that the complex micelle enhanced TNBC tumor cellular uptake and fast drug release due to the combined effect of magnet targeting, CD44 receptor-mediated internalization and redox/pH dual-responsive drug release. Hence, tumor-targeted delivery of GA with redox/pH dual-responsive multifunctional magnetic complex micelle sPEG/HA/CSO-SS-Hex/Fe3O4/GA might have potential implications for the chemotherapy of TNBC.
Collapse
Affiliation(s)
- Mang Mang Sang
- a Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education , China Pharmaceutical University , Nanjing , People's Republic of China.,b Department of Pharmaceutical Analysis , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Fu Lei Liu
- c Department of Natural Medicinal Chemistry , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Yang Wang
- a Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education , China Pharmaceutical University , Nanjing , People's Republic of China.,b Department of Pharmaceutical Analysis , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Ren Jie Luo
- a Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education , China Pharmaceutical University , Nanjing , People's Republic of China.,b Department of Pharmaceutical Analysis , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Xiao Xian Huan
- c Department of Natural Medicinal Chemistry , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Ling Fei Han
- a Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education , China Pharmaceutical University , Nanjing , People's Republic of China.,b Department of Pharmaceutical Analysis , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Zhong Tao Zhang
- c Department of Natural Medicinal Chemistry , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Feng Feng
- c Department of Natural Medicinal Chemistry , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Wei Qu
- c Department of Natural Medicinal Chemistry , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Wenyuan Liu
- a Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education , China Pharmaceutical University , Nanjing , People's Republic of China.,b Department of Pharmaceutical Analysis , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Feng Zheng
- a Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education , China Pharmaceutical University , Nanjing , People's Republic of China.,b Department of Pharmaceutical Analysis , China Pharmaceutical University , Nanjing , People's Republic of China
| |
Collapse
|
46
|
Liu S, Gao Y, Zhou D, Zeng M, Alshehri F, Newland B, Lyu J, O'Keeffe-Ahern J, Greiser U, Guo T, Zhang F, Wang W. Highly branched poly(β-amino ester) delivery of minicircle DNA for transfection of neurodegenerative disease related cells. Nat Commun 2019; 10:3307. [PMID: 31341171 PMCID: PMC6656726 DOI: 10.1038/s41467-019-11190-0] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 06/24/2019] [Indexed: 11/08/2022] Open
Abstract
Current therapies for most neurodegenerative disorders are only symptomatic in nature and do not change the course of the disease. Gene therapy plays an important role in disease modifying therapeutic strategies. Herein, we have designed and optimized a series of highly branched poly(β-amino ester)s (HPAEs) containing biodegradable disulfide units in the HPAE backbone (HPAESS) and guanidine moieties (HPAESG) at the extremities. The optimized polymers are used to deliver minicircle DNA to multipotent adipose derived stem cells (ADSCs) and astrocytes, and high transfection efficiency is achieved (77% in human ADSCs and 52% in primary astrocytes) whilst preserving over 90% cell viability. Furthermore, the top-performing candidate mediates high levels of nerve growth factor (NGF) secretion from astrocytes, causing neurite outgrowth from a model neuron cell line. This synergistic gene delivery system provides a viable method for highly efficient non-viral transfection of ADSCs and astrocytes.
Collapse
Affiliation(s)
- Shuai Liu
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Belfield, 4, Dublin, Ireland
- Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 300071, Tianjin, China
| | - Yongsheng Gao
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Belfield, 4, Dublin, Ireland
| | - Dezhong Zhou
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Belfield, 4, Dublin, Ireland.
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, 710049, Xi'an, China.
| | - Ming Zeng
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Belfield, 4, Dublin, Ireland
| | - Fatma Alshehri
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Belfield, 4, Dublin, Ireland
| | - Ben Newland
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, CF103AT, Cardiff, UK
| | - Jing Lyu
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Belfield, 4, Dublin, Ireland
| | - Jonathan O'Keeffe-Ahern
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Belfield, 4, Dublin, Ireland
| | - Udo Greiser
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Belfield, 4, Dublin, Ireland
| | - Tianying Guo
- Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 300071, Tianjin, China.
| | - Fengzhi Zhang
- School of Pharmaceutical Science, Zhejiang University of Technology, 310014, Hangzhou, China
| | - Wenxin Wang
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Belfield, 4, Dublin, Ireland.
- Department of Polymer Science and Engineering, Zhejiang University, 310027, Hangzhou, China.
| |
Collapse
|
47
|
Urbánek T, Jäger E, Jäger A, Hrubý M. Selectively Biodegradable Polyesters: Nature-Inspired Construction Materials for Future Biomedical Applications. Polymers (Basel) 2019; 11:E1061. [PMID: 31248100 PMCID: PMC6630685 DOI: 10.3390/polym11061061] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/28/2019] [Accepted: 06/14/2019] [Indexed: 12/13/2022] Open
Abstract
In the last half-century, the development of biodegradable polyesters for biomedical applications has advanced significantly. Biodegradable polyester materials containing external stimuli-sensitive linkages are favored in the development of therapeutic devices for pharmacological applications such as delivery vehicles for controlled/sustained drug release. These selectively biodegradable polyesters degrade after particular external stimulus (e.g., pH or redox potential change or the presence of certain enzymes). This review outlines the current development of biodegradable synthetic polyesters materials able to undergo hydrolytic or enzymatic degradation for various biomedical applications, including tissue engineering, temporary implants, wound healing and drug delivery.
Collapse
Affiliation(s)
- Tomáš Urbánek
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského náměstí 2, 162 00 Prague 6, Czech Republic.
| | - Eliézer Jäger
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského náměstí 2, 162 00 Prague 6, Czech Republic.
| | - Alessandro Jäger
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského náměstí 2, 162 00 Prague 6, Czech Republic.
| | - Martin Hrubý
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského náměstí 2, 162 00 Prague 6, Czech Republic.
| |
Collapse
|
48
|
Wang Y, Zhao B, Wang L, Bu W, Liu S, Sun B. Nanoparticles based on retinoic acid caped with ferrocenium: a novel synthesized targetable nanoparticle both with anti-cancer effect and drug loading capacity. RSC Adv 2019; 9:16208-16214. [PMID: 35521379 PMCID: PMC9064344 DOI: 10.1039/c9ra02472g] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 04/30/2019] [Indexed: 11/21/2022] Open
Abstract
To date, there is an urgent need for cancer treatment to improve in many ways in order to successfully cure all cancers. Retinoic acid (RA) is a promising anti-cancer drug through influencing cancer stem cells (CSCs). Taxol is a chemotherapy drug for many cancers. To increase the anti-cancer effects of RA and taxol, we created a novel RA nanoparticle, FCRAN, which has the ability of carrying a second anti-cancer drug, taxol, using nanotechnological methods. The results of this study demonstrated that this RA nanoparticle was water-soluble and retained the same effects as RA on cancer cells, such as inhibiting the proliferation of CSCs, inducing the differentiation of CSCs, and enhancing the sensitivity of CSCs to chemotherapeutic drugs. In addition, this RA nanoparticle can be used to carry a second anticancer drug, taxol, to become FCRAN/T and synergistically enhance the anti-cancer effects of both drugs in vivo. Interestingly, the FCRAN/T is a targetable anti-cancer nanoparticle in the presence of higher levels of glutathione (GSH) in cancer cells. Our results demonstrate that our novel synthesized nanoparticles not only retain the RA functions, but can also carry a second anticancer drug to play a synergistic anticancer role with good water solubility, in particular FCRAN/T can target cancer cells. Therefore, our novel synthesized targetable anti-cancer nanoparticles have a better application prospect than that of RA or taxol alone. A retinoic acid nanoparticle with the ability of carrying a second anti-cancer drug, taxol, was developed. The anti-cancer nanoparticles were shown to have a better application prospect than that of RA or taxol alone.![]()
Collapse
Affiliation(s)
- Yibo Wang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Jilin University Changchun 130041 People's Republic of China
| | - Bin Zhao
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Jilin University Changchun 130041 People's Republic of China.,Department of Periodontosis, School and Hospital of Stomatology, Jilin University Changchun 130041 People's Republic of China
| | - Lu Wang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Jilin University Changchun 130041 People's Republic of China
| | - Wenhuan Bu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Jilin University Changchun 130041 People's Republic of China
| | - Shuwei Liu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University Changchun 130012 People's Republic of China
| | - Bin Sun
- Department of Oral and Maxilloficial Surgery, School and Hospital of Stomatology, Jilin University Changchun 130041 People's Republic of China .,Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Jilin University Changchun 130041 People's Republic of China
| |
Collapse
|
49
|
Combining Inulin Multifunctional Polycation and Magnetic Nanoparticles: Redox-Responsive siRNA-Loaded Systems for Magnetofection. Polymers (Basel) 2019; 11:polym11050889. [PMID: 31096623 PMCID: PMC6571810 DOI: 10.3390/polym11050889] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 05/03/2019] [Accepted: 05/09/2019] [Indexed: 11/16/2022] Open
Abstract
Superparamagnetic Iron Oxide Nanoparticles (SPIONs) are recognized as one of the most promising agents for theranostic applications. Among methods designed for siRNA delivery, magnetofection, that is, nucleic acid cell uptake under the influence of a magnetic field acting on magnetic nucleic acid vectors, is emerging as a unique approach to combining advantages such as strong improvement of the kinetics of the delivery process and the possibility of localizing nucleic acid delivery to an area where the magnetic field is applied. This paper reports on the preparation of siRNA loaded magnetoplexes—named ICD@SS@SPIONs/siRNA—by controlled crosslinking, in the presence of SPIONs, of the polycation INU-C-DETA, synthesized starting from the polysaccharide inulin by grafting diethylenetriamine and cystamine molecules. The obtained ICD@SS@SPIONs/siRNA have suitable chemical-physical characteristics to be employed for iv administration and are also able to release siRNA in a redox-triggered manner thanks to intracellular glutathione (GSH) mediated reduction of disulphide bridges formed during the crosslinking process. Moreover, ICD@SS@SPIONs/siRNA are able to produce magnetic targeting in vitro on breast cancer cells, without appreciable cyto- and hemo-toxic effects, in a wide range of concentrations. Finally, protein binding to nanoparticles revealed that obtained systems are potentially longer circulating and applicable as a smart multifunctional agents for cancer therapy.
Collapse
|
50
|
Xing H, Lu M, Yang T, Liu H, Sun Y, Zhao X, Xu H, Yang L, Ding P. Structure-function relationships of nonviral gene vectors: Lessons from antimicrobial polymers. Acta Biomater 2019; 86:15-40. [PMID: 30590184 DOI: 10.1016/j.actbio.2018.12.041] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 11/22/2018] [Accepted: 12/21/2018] [Indexed: 01/13/2023]
Abstract
In recent years, substantial advances have been achieved in the design and synthesis of nonviral gene vectors. However, lack of effective and biocompatible vectors still remains a major challenge that hinders their application in clinical settings. In the past decade, there has been a rapid expansion of cationic antimicrobial polymers, due to their potent, rapid, and broad-spectrum biocidal activity against resistant microbes, and biocompatible features. Given that antimicrobial polymers share common features with nonviral gene vectors in various aspects, such as membrane affinity, functional groups, physicochemical characteristics, and unique macromolecular architectures, these polymers may provide us with inspirations to overcome challenges in the design of novel vectors toward more safe and efficient gene delivery in clinic. Building off these observations, we provide here an overview of the structure-function relationships of polymers for both antimicrobial applications and gene delivery by elaborating some key structural parameters, including functional groups, charge density, hydrophobic/hydrophilic balance, MW, and macromolecular architectures. By borrowing a leaf from antimicrobial agents, great advancement in the development of newer nonviral gene vectors with high transfection efficiency and biocompatibility will be more promising. STATEMENT OF SIGNIFICANCE: The development of gene delivery is still in the preclinical stage for the lack of effective and biocompatible vectors. Given that antimicrobial polymers share common features with gene vectors in various aspects, such as membrane affinity, functional groups, physicochemical characteristics, and unique macromolecular architectures, these polymers may provide us with inspirations to overcome challenges in the design of novel vectors toward more safe and efficient gene delivery in clinic. In this review, we systematically summarized the structure-function relationships of antimicrobial polymers and gene vectors, with which the design of more advanced nonviral gene vectors is anticipated to be further boosted in the future.
Collapse
Affiliation(s)
- Haonan Xing
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Mei Lu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Tianzhi Yang
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, Husson University, Bangor, ME, USA
| | - Hui Liu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Yanping Sun
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiaoyun Zhao
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Hui Xu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Li Yang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China.
| | - Pingtian Ding
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China.
| |
Collapse
|