1
|
Martinez Junior AM, Ruiz TFR, Vilamaior PSL, Tiera VADO, Taboga SR, Tiera MJ. Topical delivery of siRNA to psoriatic skin model using high molecular weight chitosan derivatives: In vitro and in vivo studies. Drug Deliv Transl Res 2025:10.1007/s13346-025-01800-4. [PMID: 39907973 DOI: 10.1007/s13346-025-01800-4] [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] [Accepted: 01/17/2025] [Indexed: 02/06/2025]
Abstract
Psoriasis is a chronic inflammatory skin disease that, like other immune-mediated conditions, may benefit from small interfering RNA (siRNA)-based therapies, which are emerging as a promising alternative by addressing several limitations of current treatments. In this study, topical formulations of chitosan-based vectors were developed to deliver siRNA targeting tumor necrosis factor alpha (TNFα) to inflamed skin. Grafting diisopropylethylamine (DIPEA) and polyethylene glycol (PEG) onto the chitosan backbone enhanced siRNA delivery efficiency under physiological conditions, forming robust polymeric vectors with high structural and colloidal stability. These vectors provided siRNA protection against RNAse degradation and oxidative damage. Additionally, the chitosan derivatives displayed lysozyme-mediated biodegradability comparable to native chitosan, while PEG was released in response to reductive environments, supporting controlled vector disassembly. The PEGylated DIPEA-chitosan/siRNA polyplexes demonstrated positive zeta potentials (up to + 11 mV), particle sizes of 100-200 nm, and very low cytotoxicity in keratinocyte and fibroblast cell lines. In vitro, the polyplexes achieved TNFα knockdown levels (65%) in RAW macrophages, comparable to those obtained with Lipofectamine™. Topical formulations showed enhanced interaction of vectors with skin models (Strat-M® and porcine ear skin) compared to naked siRNA. Furthermore, in vivo studies indicated that hair follicles were a key route for polyplexes to penetrate deeper skin layers. A rodent model of psoriasis induced by imiquimod was treated topically with these vectors, resulting in approximately a 50% reduction in TNFα levels at inflammation sites, decreased immune cell infiltration, and preservation of epidermal structure. These findings collectively underscore the potential of DIPEA-chitosan-based vectors for topical siRNA-based therapies.
Collapse
Affiliation(s)
- André Miguel Martinez Junior
- Department of Chemistry and Environmental Sciences, Institute of Biosciences, Humanities and Exact Sciences (IBILCE), São Paulo State University (UNESP), R. Cristóvão Colombo 2265, 15054-000, São José do Rio Preto, São Paulo, Brazil
| | - Thalles Fernando Rocha Ruiz
- Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences (IBILCE), São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| | - Patrícia Simone Leite Vilamaior
- Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences (IBILCE), São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| | - Vera Aparecida de Oliveira Tiera
- Department of Chemistry and Environmental Sciences, Institute of Biosciences, Humanities and Exact Sciences (IBILCE), São Paulo State University (UNESP), R. Cristóvão Colombo 2265, 15054-000, São José do Rio Preto, São Paulo, Brazil
| | - Sebastião Roberto Taboga
- Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences (IBILCE), São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| | - Marcio José Tiera
- Department of Chemistry and Environmental Sciences, Institute of Biosciences, Humanities and Exact Sciences (IBILCE), São Paulo State University (UNESP), R. Cristóvão Colombo 2265, 15054-000, São José do Rio Preto, São Paulo, Brazil.
| |
Collapse
|
2
|
Yu Y, Tao Y, Ma J, Li J, Song Z. Targeting the tumor microenvironment with mesenchymal stem cells based delivery approach for efficient delivery of anticancer agents: An updated review. Biochem Pharmacol 2025; 232:116725. [PMID: 39746456 DOI: 10.1016/j.bcp.2024.116725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/14/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025]
Abstract
Drug delivery to cancer cells continues to present a major therapeutic challenge. Mesenchymal stem cells (MSCs) possess an intrinsic ability to migrate specifically to tumor tissues, making them promising candidates for targeted drug delivery. Evidence from preclinical studies indicates that MSCs loaded with therapeutic anti-cancer agents exhibit considerable anti-tumor activity. Moreover, several clinical trials are currently evaluating their effectiveness in cancer patients. The integration of MSCs with synthetic nanoparticles (NPs) enhances their therapeutic potential, particularly through the use of cell membrane-coated NPs, which represent a significant advancement in the field. This review systematically investigates the tumor microenvironment, the sources of MSCs, the tumor homing mechanisms, and the methods of loading and releasing anticancer drugs from MSCs. Furthermore, cutting-edge strategies to improve the efficacy of MSCs based drug delivery systems (DDS) including the innovative use of MSC membrane coated nanoparticles have been discussed. The study concludes with an overview of the therapeutic use of MSCs as drug carriers, including a detailed analysis of the mechanisms by which MSCs deliver therapeutics to cancer cells, enabling targeted drug delivery. It aims to elucidate the current state of this approach, identify key areas for development, and outline potential future directions for advancing MSCs based cancer therapies.
Collapse
Affiliation(s)
- Yang Yu
- Department of Emergency and Critical Care, the Second Hospital of Jilin University, Changchun 130000, China
| | - Ying Tao
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun 130000, China
| | - Jingru Ma
- Department of Clinical Laboratory, the Second Hospital of Jilin University, Changchun 130000, China
| | - Jian Li
- Department of Emergency and Critical Care, the Second Hospital of Jilin University, Changchun 130000, China
| | - Zhidu Song
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun 130000, China.
| |
Collapse
|
3
|
Kheiriabad S, Jafari A, Namvar Aghdash S, Ezzati Nazhad Dolatabadi J, Andishmand H, Jafari SM. Applications of Advanced Nanomaterials in Biomedicine, Pharmaceuticals, Agriculture, and Food Industry. BIONANOSCIENCE 2024; 14:4298-4321. [DOI: 10.1007/s12668-024-01506-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/15/2024] [Indexed: 01/06/2025]
|
4
|
Liu J, Li Y, Zhou K, Zhang S, Wang Y, Wang X, Lan X, Chen Q, Zhao Y. In Situ Polymerization for Manufacture of Multifunctional Delivery Systems for Transcellular Delivery of Nucleic Acids. Bioconjug Chem 2024; 35:1417-1428. [PMID: 39225485 DOI: 10.1021/acs.bioconjchem.4c00319] [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: 09/04/2024]
Abstract
Electrostatic self-assembly between negatively charged nucleic acids and cationic materials is the basis for the formulation of the delivery systems. Nevertheless, structural disintegration occurs because their colloidal stabilities are frequently insufficient in a hostile biological environment. To overcome the sequential biological barriers encountered during transcellular gene delivery, we attempted to use in situ polymerization onto plasmid DNA (pDNA) with a variety of functional monomers, including N-(3-aminopropyl)methacrylate, (aminopropyl)methacrylamide hydrochloride, 1-vinylimidazole, and 2-methacryloyloxyethylphosphorylcholine and N,N'-bis(acryloyl) cystamine. The covalently linked monomers could polymerize into a network structure on top of pDNA, providing excellent structural stability. Additionally, the significant proton buffering capacity of 1-vinylimidazole is expected to aid in the release of pDNA payloads from acidic and digestive endolysosomes. In addition, the redox-mediated cleavage of the disulfide bond in N,N'-bis(acryloyl)cystamine allows for the selective cleavage of the covalently linked network in the cytosolic microenvironment. This is due to the high intracellular level of glutathione, which promotes the liberation of pDNA payloads in the cell interiors. The proposed polymerization strategies resulted in well-defined nanoscale pDNA delivery systems. Excellent colloidal stabilities were observed, even when incubated in the presence of high concentrations of heparin (10 mg/mL). In contrast, the release of pDNA was confirmed upon incubation in the presence of glutathione, mimicking the intracellular microenvironment. Cell transfection experiments verified their efficient cellular uptake and gene expression activities in the hard-transfected MCF-7 cells. Hence, the polymerization strategy used in the fabrication of covalently linked nonviral gene delivery systems shows promise in creating high-performance gene delivery systems with diverse functions. This could open new avenues in cellular microenvironment engineering.
Collapse
Affiliation(s)
- Jun Liu
- School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
- Jiaxing Qingzhun Pharmaceutical Technology Co., Ltd, Western Kechuang Bay Valley, Tongxiang Town, Jiaxing, Zhejiang 314500, China
| | - Yanhua Li
- International Medical Department, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian 116027, China
| | - Kehui Zhou
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang 310022, China
- Department of Thyroid Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China
| | - Shijia Zhang
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang 310022, China
- Department of Thyroid Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China
| | - Yue Wang
- Department of Gastric Surgery, Cancer Hospital of Dalian University of Technology, No. 44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning 110042, China
- Department of Gastric Surgery, Cancer Hospital of China Medical University, No. 44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning 110042, China
- Provincial Key Laboratory of Interdisciplinary Medical Engineering for Gastrointestinal Carcinoma, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning 110042, China
| | - Xiumei Wang
- School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Xiabin Lan
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang 310022, China
- Department of Thyroid Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China
| | - Qixian Chen
- Jiaxing Qingzhun Pharmaceutical Technology Co., Ltd, Western Kechuang Bay Valley, Tongxiang Town, Jiaxing, Zhejiang 314500, China
- Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, Zhejiang 314100, China
| | - Yan Zhao
- Department of Gastric Surgery, Cancer Hospital of Dalian University of Technology, No. 44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning 110042, China
- Department of Gastric Surgery, Cancer Hospital of China Medical University, No. 44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning 110042, China
- Provincial Key Laboratory of Interdisciplinary Medical Engineering for Gastrointestinal Carcinoma, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning 110042, China
| |
Collapse
|
5
|
Gong Y, Hu X, Chen M, Wang J. Recent progress of iron-based nanomaterials in gene delivery and tumor gene therapy. J Nanobiotechnology 2024; 22:309. [PMID: 38825720 PMCID: PMC11145874 DOI: 10.1186/s12951-024-02550-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 05/14/2024] [Indexed: 06/04/2024] Open
Abstract
Gene therapy aims to modify or manipulate gene expression and change the biological characteristics of living cells to achieve the purpose of treating diseases. The safe, efficient, and stable expression of exogenous genes in cells is crucial for the success of gene therapy, which is closely related to the vectors used in gene therapy. Currently, gene therapy vectors are mainly divided into two categories: viral vectors and non-viral vectors. Viral vectors are widely used due to the advantages of persistent and stable expression, high transfection efficiency, but they also have certain issues such as infectivity, high immunological rejection, randomness of insertion mutation, carcinogenicity, and limited vector capacity. Non-viral vectors have the advantages of non-infectivity, controllable chemical structure, and unlimited vector capacity, but the transfection efficiency is low. With the rapid development of nanotechnology, the unique physicochemical properties of nanomaterials have attracted increasing attention in the field of drug and gene delivery. Among many nanomaterials, iron-based nanomaterials have attracted much attention due to their superior physicochemical properties, such as Fenton reaction, magnetic resonance imaging, magnetothermal therapy, photothermal therapy, gene delivery, magnetically-assisted drug delivery, cell and tissue targeting, and so on. In this paper, the research progress of iron-based nanomaterials in gene delivery and tumor gene therapy is reviewed, and the future application direction of iron-based nanomaterials is further prospected.
Collapse
Affiliation(s)
- Ya Gong
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, Chongqing, 400037, China
| | - Xiaoyan Hu
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100864, China
| | - Ming Chen
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University, Chongqing, 400038, China.
| | - Jun Wang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
6
|
Wu L, Yuan R, Wen T, Qin Y, Wang Y, Luo X, Liu JW. Recent advances in functional nucleic acid decorated nanomaterials for cancer imaging and therapy. Biomed Pharmacother 2024; 174:116546. [PMID: 38603885 DOI: 10.1016/j.biopha.2024.116546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/19/2024] [Accepted: 04/04/2024] [Indexed: 04/13/2024] Open
Abstract
Nanomaterials possess unusual physicochemical properties including unique optical, magnetic, electronic properties, and large surface-to-volume ratio. However, nanomaterials face some challenges when they were applied in the field of biomedicine. For example, some nanomaterials suffer from the limitations such as poor selectivity and biocompatibility, low stability, and solubility. To address the above-mentioned obstacles, functional nucleic acid has been widely served as a powerful and versatile ligand for modifying nanomaterials because of their unique characteristics, such as ease of modification, excellent biocompatibility, high stability, predictable intermolecular interaction and recognition ability. The functionally integrating functional nucleic acid with nanomaterials has produced various kinds of nanocomposites and recent advances in applications of functional nucleic acid decorated nanomaterials for cancer imaging and therapy were summarized in this review. Further, we offer an insight into the future challenges and perspectives of functional nucleic acid decorated nanomaterials.
Collapse
Affiliation(s)
- Liu Wu
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Ruitao Yuan
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Tong Wen
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Yingfeng Qin
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Yumin Wang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China.
| | - Xiaoling Luo
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China.
| | - Jin-Wen Liu
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China.
| |
Collapse
|
7
|
Chaix A, Cueto‐Diaz E, Dominguez‐Gil S, Spiteri C, Lichon L, Maynadier M, Dumail X, Aggad D, Delalande A, Bessière A, Pichon C, Chiappini C, Sailor MJ, Bettache N, Gary‐Bobo M, Durand J, Nguyen C, Cunin F. Two-Photon Light Trigger siRNA Transfection of Cancer Cells Using Non-Toxic Porous Silicon Nanoparticles. Adv Healthc Mater 2023; 12:e2301052. [PMID: 37499629 PMCID: PMC11468491 DOI: 10.1002/adhm.202301052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 07/23/2023] [Indexed: 07/29/2023]
Abstract
The concept of using two-photon excitation in the NIR for the spatiotemporal control of biological processes holds great promise. However, its use for the delivery of nucleic acids has been very scarcely described and the reported procedures are not optimal as they often involve potentially toxic materials and irradiation conditions. This work prepares a simple system made of biocompatible porous silicon nanoparticles (pSiNP) for the safe siRNA photocontrolled delivery and gene silencing in cells upon two-photon excitation. PSiNP are linked to an azobenzene moiety, which possesses a lysine group (pSiNP@ICPES-azo@Lys) to efficiently complex siRNA. Non-linear excitation of the two-photon absorber system (pSiNP) followed by intermolecular energy transfer (FRET) to trans azobenzene moiety, result in the photoisomerization of the azobenzene from trans to cis and in the destabilization of the azobenzene-siRNA complex, thus inducing the delivery of the cargo siRNA to the cytoplasm of cells. Efficient silencing in MCF-7 expressing stable firefly luciferase with siRNAluc against luciferase is observed. Furthermore, siRNA against inhibitory apoptotic protein (IAP) leads to over 70% of MCF-7 cancer cell death. The developed technique using two-photon light allows a unique high spatiotemporally controlled and safe siRNA delivery in cells in few seconds of irradiation.
Collapse
Affiliation(s)
- Arnaud Chaix
- ICGMCNRSENSCMUniversity of MontpellierMontpellier34293France
| | | | | | - Chantelle Spiteri
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 9RTUK
- London Centre for NanotechnologyKing's College LondonLondonWC2R 2LSUK
| | - Laure Lichon
- IBMMUniv. MontpellierCNRSENSCMMontpellier34093France
| | - Marie Maynadier
- NanoMedSyn Avenue Charles FlahaultMontpellier Cedex 0534093France
| | - Xavier Dumail
- ICGMCNRSENSCMUniversity of MontpellierMontpellier34293France
| | - Dina Aggad
- IBMMUniv. MontpellierCNRSENSCMMontpellier34093France
| | - Anthony Delalande
- Centre de Biophysique MoléculaireCNRS UPR4301Orléans cedex 02F‐45071France
- Inserm UMS 55ART ARNm and University of OrléansOrléansF‐45100France
- Institut Universitaire de France1 rue DescartesParisF‐75035France
| | | | - Chantal Pichon
- Centre de Biophysique MoléculaireCNRS UPR4301Orléans cedex 02F‐45071France
- Inserm UMS 55ART ARNm and University of OrléansOrléansF‐45100France
- Institut Universitaire de France1 rue DescartesParisF‐75035France
| | - Ciro Chiappini
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 9RTUK
- London Centre for NanotechnologyKing's College LondonLondonWC2R 2LSUK
| | - Michael J. Sailor
- University of CaliforniaSan DiegoDepartment of Chemistry and Biochemistry9500 Gilman Drive, m/c 0358La JollaCA92093USA
| | | | | | | | | | | |
Collapse
|
8
|
Kumar Shukla M, Parihar A, Karthikeyan C, Kumar D, Khan R. Multifunctional GQDs for receptor targeting, drug delivery, and bioimaging in pancreatic cancer. NANOSCALE 2023; 15:14698-14716. [PMID: 37655476 DOI: 10.1039/d3nr03161f] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
Pancreatic cancer is a devastating disease with a low survival rate and limited treatment options. Graphene quantum dots (GQDs) have recently become popular as a promising platform for cancer diagnosis and treatment due to their exceptional physicochemical properties, such as biocompatibility, stability, and fluorescence. This review discusses the potential of multifunctional GQDs as a platform for receptor targeting, drug delivery, and bioimaging in pancreatic cancer. The current studies emphasized the ability of GQDs to selectively target pancreatic cancer cells by overexpressing binding receptors on the cell surface. Additionally, this review discussed the uses of GQDs as drug delivery vehicles for the controlled and targeted release of therapeutics for pancreatic cancer cells. Finally, the potential of GQDs as imaging agents for pancreatic cancer detection and monitoring has been discussed. Overall, multifunctional GQDs showed great promise as a versatile platform for the diagnosis and treatment of pancreatic cancer. Further investigation of multifunctional GQDs in terms of their potential and optimization in the context of pancreatic cancer therapy is needed.
Collapse
Affiliation(s)
- Monu Kumar Shukla
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan 173229, India
| | - Arpana Parihar
- Industrial Waste Utilization, Nano and Biomaterials, CSIR-Advanced Materials and Processes Research Institute (AMPRI), Hoshangabad Road, Bhopal 462026, Madhya Pradesh, India.
| | | | - Deepak Kumar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan 173229, India
| | - Raju Khan
- Industrial Waste Utilization, Nano and Biomaterials, CSIR-Advanced Materials and Processes Research Institute (AMPRI), Hoshangabad Road, Bhopal 462026, Madhya Pradesh, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
9
|
Cheng Q, Kang Y, Yao B, Dong J, Zhu Y, He Y, Ji X. Genetically Engineered-Cell-Membrane Nanovesicles for Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302131. [PMID: 37409429 PMCID: PMC10502869 DOI: 10.1002/advs.202302131] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/13/2023] [Indexed: 07/07/2023]
Abstract
The advent of immunotherapy has marked a new era in cancer treatment, offering significant clinical benefits. Cell membrane as drug delivery materials has played a crucial role in enhancing cancer therapy because of their inherent biocompatibility and negligible immunogenicity. Different cell membranes are prepared into cell membrane nanovesicles (CMNs), but CMNs have limitations such as inefficient targeting ability, low efficacy, and unpredictable side effects. Genetic engineering has deepened the critical role of CMNs in cancer immunotherapy, enabling genetically engineered-CMN (GCMN)-based therapeutics. To date, CMNs that are surface modified by various functional proteins have been developed through genetic engineering. Herein, a brief overview of surface engineering strategies for CMNs and the features of various membrane sources is discussed, followed by a description of GCMN preparation methods. The application of GCMNs in cancer immunotherapy directed at different immune targets is addressed as are the challenges and prospects of GCMNs in clinical translation.
Collapse
Affiliation(s)
| | - Yong Kang
- Academy of Medical Engineering and Translational MedicineMedical CollegeTianjin UniversityTianjin300072China
| | - Bin Yao
- Academy of Medical Engineering and Translational MedicineMedical CollegeTianjin UniversityTianjin300072China
| | - Jinrui Dong
- Academy of Medical Engineering and Translational MedicineMedical CollegeTianjin UniversityTianjin300072China
| | - Yalan Zhu
- Jinhua Municipal Central HospitalJinhua321000China
| | - Yiling He
- Jinhua Municipal Central HospitalJinhua321000China
| | - Xiaoyuan Ji
- Academy of Medical Engineering and Translational MedicineMedical CollegeTianjin UniversityTianjin300072China
- Medical CollegeLinyi UniversityLinyi276000China
| |
Collapse
|
10
|
Annu, Rehman S, Nabi B, Sartaj A, Md S, Sahoo PK, Baboota S, Ali J. Nanoparticle Mediated Gene Therapy: A Trailblazer Armament to Fight CNS Disorders. Curr Med Chem 2023; 30:304-315. [PMID: 34986767 DOI: 10.2174/0929867329666220105122318] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 10/10/2021] [Accepted: 10/28/2021] [Indexed: 11/22/2022]
Abstract
Central nervous system (CNS) disorders account for boundless socioeconomic burdens with devastating effects among the population, especially the elderly. The major symptoms of these disorders are neurodegeneration, neuroinflammation, and cognitive dysfunction caused by inherited genetic mutations or by genetic and epigenetic changes due to injury, environmental factors, and disease-related events. Currently available clinical treatments for CNS diseases, i.e., Alzheimer's disease, Parkinson's disease, stroke, and brain tumor, have significant side effects and are largely unable to halt the clinical progression. So gene therapy displays a new paradigm in the treatment of these disorders with some modalities, varying from the suppression of endogenous genes to the expression of exogenous genes. Both viral and non-viral vectors are commonly used for gene therapy. Viral vectors are quite effective but associated with severe side effects, like immunogenicity and carcinogenicity, and poor target cell specificity. Thus, non-viral vectors, mainly nanotherapeutics like nanoparticles (NPs), turn out to be a realistic approach in gene therapy, achieving higher efficacy. NPs demonstrate a new avenue in pharmacotherapy for the delivery of drugs or genes to their selective cells or tissue, thus providing concentrated and constant drug delivery to targeted tissues, minimizing systemic toxicity and side effects. The current review will emphasize the role of NPs in mediating gene therapy for CNS disorders treatment. Moreover, the challenges and perspectives of NPs in gene therapy will be summarized.
Collapse
Affiliation(s)
- Annu
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-110062, India
| | - Saleha Rehman
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-110062, India
| | - Bushra Nabi
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-110062, India
| | - Ali Sartaj
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-110062, India
| | - Shadab Md
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - P K Sahoo
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Sciences and Research, New Delhi-110017, India
| | - Sanjula Baboota
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-110062, India
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-110062, India
| |
Collapse
|
11
|
Shahcheraghi SH, Ayatollahi J, Lotfi M, Aljabali AAA, Al-Zoubi MS, Panda PK, Mishra V, Satija S, Charbe NB, Serrano-Aroca Á, Bahar B, Takayama K, Goyal R, Bhatia A, Almutary AG, Alnuqaydan AM, Mishra Y, Negi P, Courtney A, McCarron PA, Bakshi HA, Tambuwala MM. Gene Therapy for Neuropsychiatric Disorders: Potential Targets and Tools. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2023; 22:51-65. [PMID: 35249508 DOI: 10.2174/1871527321666220304153719] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/16/2022] [Accepted: 01/16/2022] [Indexed: 01/01/2023]
Abstract
Neuropsychiatric disorders that affect the central nervous system cause considerable pressures on the health care system and have a substantial economic burden on modern societies. The present treatments based on available drugs are mostly ineffective and often costly. The molecular process of neuropsychiatric disorders is closely connected to modifying the genetic structures inherited or caused by damage, toxic chemicals, and some current diseases. Gene therapy is presently an experimental concept for neurological disorders. Clinical applications endeavor to alleviate the symptoms, reduce disease progression, and repair defective genes. Implementing gene therapy in inherited and acquired neurological illnesses entails the integration of several scientific disciplines, including virology, neurology, neurosurgery, molecular genetics, and immunology. Genetic manipulation has the power to minimize or cure illness by inducing genetic alterations at endogenous loci. Gene therapy that involves treating the disease by deleting, silencing, or editing defective genes and delivering genetic material to produce therapeutic molecules has excellent potential as a novel approach for treating neuropsychiatric disorders. With the recent advances in gene selection and vector design quality in targeted treatments, gene therapy could be an effective approach. This review article will investigate and report the newest and the most critical molecules and factors in neuropsychiatric disorder gene therapy. Different genome editing techniques available will be evaluated, and the review will highlight preclinical research of genome editing for neuropsychiatric disorders while also evaluating current limitations and potential strategies to overcome genome editing advancements.
Collapse
Affiliation(s)
- Seyed H Shahcheraghi
- Department of Medical Genetics, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Infectious Diseases Research Center, Shahid Sadoughi Hospital, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Jamshid Ayatollahi
- Infectious Diseases Research Center, Shahid Sadoughi Hospital, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Marzieh Lotfi
- Abortion Research Center, Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Alaa A A Aljabali
- Department of Pharmaceutics & Pharmaceutical Technology, Yarmouk University, Irbid, Jordan
| | - Mazhar S Al-Zoubi
- Yarmouk University, Faculty of Medicine, Department of Basic Medical Sciences, Irbid, Jordan
| | - Pritam K Panda
- Condensed Matter Theory Group, Materials Theory Division, Department of Physics and Astronomy, Uppsala University, Box 516, 75120 Uppsala, Sweden
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Saurabh Satija
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Nitin B Charbe
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M Health Science Center, Kingsville, TX 78363, USA
| | - Ángel Serrano-Aroca
- Biomaterials and Bioengineering Lab, Translational Research Centre San Alberto Magno, Catholic University of Valencia San Vicente Mártir, C/Guillem de Castro 94, 46001 Valencia, Spain
| | - Bojlul Bahar
- Nutrition Sciences and Applied Food Safety Studies, Research Centre for Global Development, School of Sport & Health Sciences, University of Central Lancashire, Preston, PR1 2HE, UK
| | - Kazuo Takayama
- Center for IPS Cell Research and Application, Kyoto University, Kyoto, 606-8397, Japan
| | - Rohit Goyal
- Neuropharmacology Laboratory, School of Pharmaceutical Sciences, Shoolini University, Post Box No. 9, Solan, Himachal Pradesh 173212, India
| | - Amit Bhatia
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Punjab 151001, India
| | - Abdulmajeed G Almutary
- Department of Medical Biotechnology, College of Applied Medical Sciences, Qassim University, Saudi Arabia
| | - Abdullah M Alnuqaydan
- Department of Medical Biotechnology, College of Applied Medical Sciences, Qassim University, Saudi Arabia
| | - Yachana Mishra
- Shri Shakti Degree College, Sankhahari, Ghatampur 209206, India
| | - Poonam Negi
- Shoolini University of Biotechnology and Management Sciences, Solan 173 212, India
| | - Aaron Courtney
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine, County Londonderry, BT52 1SA, Northern Ireland, United Kingdom
| | - Paul A McCarron
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine, County Londonderry, BT52 1SA, Northern Ireland, United Kingdom
| | - Hamid A Bakshi
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine, County Londonderry, BT52 1SA, Northern Ireland, United Kingdom
| | - Murtaza M Tambuwala
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine, County Londonderry, BT52 1SA, Northern Ireland, United Kingdom
| |
Collapse
|
12
|
Chrysostomou V, Foryś A, Trzebicka B, Demetzos C, Pispas S. Amphiphilic Copolymer-Lipid Chimeric Nanosystems as DNA Vectors. Polymers (Basel) 2022; 14:polym14224901. [PMID: 36433029 PMCID: PMC9699196 DOI: 10.3390/polym14224901] [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: 10/14/2022] [Revised: 11/08/2022] [Accepted: 11/11/2022] [Indexed: 11/15/2022] Open
Abstract
Lipid-polymer chimeric (hybrid) nanosystems are promising platforms for the design of effective gene delivery vectors. In this regard, we developed DNA nanocarriers comprised of a novel poly[(stearyl methacrylate-co-oligo(ethylene glycol) methyl ether methacrylate] [P(SMA-co-OEGMA)] amphiphilic random copolymer, the cationic 1,2-dioleoyl-3-(trimethylammonium) propane (DOTAP), and the zwitterionic L-α-phosphatidylcholine, hydrogenated soybean (soy) (HSPC) lipids. Chimeric HSPC:DOTAP:P[(SMA-co-OEGMA)] nanosystems, and pure lipid nanosystems as reference, were prepared in several molar ratios of the components. The colloidal dispersions obtained presented well-defined physicochemical characteristics and were further utilized for the formation of lipoplexes with a model DNA of linear topology containing 113 base pairs. Nanosized complexes were formed through the electrostatic interaction of the cationic lipid and phosphate groups of DNA, as observed by dynamic, static, and electrophoretic light scattering techniques. Ultraviolet-visible (UV-Vis) and fluorescence spectroscopy disclosed the strong binding affinity of the chimeric and also the pure lipid nanosystems to DNA. Colloidally stable chimeric/lipid complexes were formed, whose physicochemical characteristics depend on the N/P ratio and on the molar ratio of the building components. Cryogenic transmission electron microscopy (Cryo-TEM) revealed the formation of nanosystems with vesicular morphology. The results suggest the successful fabrication of these novel chimeric nanosystems with well-defined physicochemical characteristics, which can form stable lipoplexes.
Collapse
Affiliation(s)
- Varvara Chrysostomou
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, 15771 Athens, Greece
- Theoretical and Physical Chemistry Institute, National Hellenic Research Foundation, 48 Vassileos Constantinou Avenue, 11635 Athens, Greece
| | - Aleksander Foryś
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, 34 ul. M. Curie-Skłodowskiej, 41-819 Zabrze, Poland
| | - Barbara Trzebicka
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, 34 ul. M. Curie-Skłodowskiej, 41-819 Zabrze, Poland
| | - Costas Demetzos
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, 15771 Athens, Greece
| | - Stergios Pispas
- Theoretical and Physical Chemistry Institute, National Hellenic Research Foundation, 48 Vassileos Constantinou Avenue, 11635 Athens, Greece
- Correspondence: ; Tel.: +30-2107273824
| |
Collapse
|
13
|
Yang JB, Wu CY, Liu XY, Yu XM, Guo XR, Zhang YJ, Liu R, Lu ZL, Huang HW. Red fluorescent AIEgens based multifunctional nonviral gene vectors for the efficient combination of gene therapy and photodynamic therapy in anti-cancer. Colloids Surf B Biointerfaces 2022; 218:112765. [PMID: 35981470 DOI: 10.1016/j.colsurfb.2022.112765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/17/2022] [Accepted: 08/07/2022] [Indexed: 10/15/2022]
Abstract
Precise molecular engineering of AIEgens-based cationic delivery systems for high transfection efficiency (TE) and effective photodynamic therapy (PDT) holds a huge potential for cancer treatment. Herein, three amphiphiles (DT-C6/8/12-M) consisting of di(triazole-[12]aneN3) (M) and 1,1-dicyano-2-phenyl-2-(4-diphenylamino)phenyl-ethylene (DT) units have been developed to achieve luminescent tracking, efficient TE, and effective PDT in vitro and in vivo. These compounds exhibited strong aggregated induced emission (AIE) at 630 nm and mega Stokes shifts of up to 160 nm. They were able to bind DNA into nanoparticles with suitable sizes, positive surface potential, and good biocompatibility in the presence of DOPE. Among them, vector DT-C12-M/DOPE with n-dodecyl linker achieved a transfection efficiency as high as 42.3 folds that of Lipo2000 in PC-3 cell lines. DT-C12-M/DOPE exhibited the capability of successful endo/lysosomal escape and rapid nuclear delivery of pDNA, and the gene delivery process was clearly monitored via confocal laser scanning microscopy. Moreover, efficient reactive oxygen species (ROS) generation by DT-C12-M upon light irradiation led to effective PDT in vitro . We further show that combination of p53 gene therapy and PDT dramatically enhanced cancer therapeutic outcome in vivo. This "three birds, one stone" strategy offers a novel and promising approach for real-time tracking of gene delivery and better cancer treatment.
Collapse
Affiliation(s)
- Jing-Bo Yang
- College of Chemistry, Beijing Normal University, Key Laboratory of Radiopharmaceuticals, Ministry of Education, Beijing 100875, PR China
| | - Cheng-Yan Wu
- College of Chemistry, Beijing Normal University, Key Laboratory of Radiopharmaceuticals, Ministry of Education, Beijing 100875, PR China
| | - Xu-Ying Liu
- College of Chemistry, Beijing Normal University, Key Laboratory of Radiopharmaceuticals, Ministry of Education, Beijing 100875, PR China
| | - Xiao-Man Yu
- College of Chemistry, Beijing Normal University, Key Laboratory of Radiopharmaceuticals, Ministry of Education, Beijing 100875, PR China
| | - Xiao-Ru Guo
- College of Chemistry, Beijing Normal University, Key Laboratory of Radiopharmaceuticals, Ministry of Education, Beijing 100875, PR China
| | - Yi-Jing Zhang
- College of Chemistry, Beijing Normal University, Key Laboratory of Radiopharmaceuticals, Ministry of Education, Beijing 100875, PR China
| | - Rui Liu
- College of Chemistry, Beijing Normal University, Key Laboratory of Radiopharmaceuticals, Ministry of Education, Beijing 100875, PR China
| | - Zhong-Lin Lu
- College of Chemistry, Beijing Normal University, Key Laboratory of Radiopharmaceuticals, Ministry of Education, Beijing 100875, PR China.
| | - Hai-Wei Huang
- China National Institute for Food and Drug Control, Institute of Chemical Drug Control, HuaTuo Road 29, Beijing 102629, PR China.
| |
Collapse
|
14
|
Luo M, Lee LKC, Peng B, Choi CHJ, Tong WY, Voelcker NH. Delivering the Promise of Gene Therapy with Nanomedicines in Treating Central Nervous System Diseases. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2201740. [PMID: 35851766 PMCID: PMC9475540 DOI: 10.1002/advs.202201740] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/19/2022] [Indexed: 06/01/2023]
Abstract
Central Nervous System (CNS) diseases, such as Alzheimer's diseases (AD), Parkinson's Diseases (PD), brain tumors, Huntington's disease (HD), and stroke, still remain difficult to treat by the conventional molecular drugs. In recent years, various gene therapies have come into the spotlight as versatile therapeutics providing the potential to prevent and treat these diseases. Despite the significant progress that has undoubtedly been achieved in terms of the design and modification of genetic modulators with desired potency and minimized unwanted immune responses, the efficient and safe in vivo delivery of gene therapies still poses major translational challenges. Various non-viral nanomedicines have been recently explored to circumvent this limitation. In this review, an overview of gene therapies for CNS diseases is provided and describes recent advances in the development of nanomedicines, including their unique characteristics, chemical modifications, bioconjugations, and the specific applications that those nanomedicines are harnessed to deliver gene therapies.
Collapse
Affiliation(s)
- Meihua Luo
- Monash Institute of Pharmaceutics ScienceMonash UniversityParkville Campus, 381 Royal ParadeParkvilleVIC3052Australia
- Australian Institute for Bioengineering and Nanotechnologythe University of QueenslandSt LuciaQLD4072Australia
| | - Leo Kit Cheung Lee
- Department of Biomedical EngineeringThe Chinese University of Hong KongShatinNew TerritoriesHong Kong
| | - Bo Peng
- Monash Institute of Pharmaceutics ScienceMonash UniversityParkville Campus, 381 Royal ParadeParkvilleVIC3052Australia
- Frontiers Science Center for Flexible ElectronicsXi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical materials & EngineeringNorthwestern Polytechnical UniversityXi'an710072China
| | - Chung Hang Jonathan Choi
- Department of Biomedical EngineeringThe Chinese University of Hong KongShatinNew TerritoriesHong Kong
| | - Wing Yin Tong
- Monash Institute of Pharmaceutics ScienceMonash UniversityParkville Campus, 381 Royal ParadeParkvilleVIC3052Australia
| | - Nicolas H. Voelcker
- Monash Institute of Pharmaceutics ScienceMonash UniversityParkville Campus, 381 Royal ParadeParkvilleVIC3052Australia
- Commonwealth Scientific and Industrial Research Organization (CSIRO)ClaytonVIC3168Australia
- Melbourne Centre for NanofabricationVictorian Node of the Australian National Fabrication Facility151 Wellington RoadClaytonVIC3168Australia
- Materials Science and EngineeringMonash University14 Alliance LaneClaytonVIC3800Australia
| |
Collapse
|
15
|
Cheng D, Theivendran S, Tang J, Cai L, Zhang J, Song H, Yu C. Surface chemistry of spiky silica nanoparticles tailors polyethyleneimine binding and intracellular DNA delivery. J Colloid Interface Sci 2022; 628:297-305. [PMID: 35998455 DOI: 10.1016/j.jcis.2022.08.038] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/27/2022] [Accepted: 08/06/2022] [Indexed: 11/17/2022]
Abstract
Cellular delivery of DNA using silica nanoparticles has attracted great attention. Typically, polyethyleneimine (PEI) is used to form a silica/PEI composite vector. Understanding the interactions at the silica and PEI interface is important for successful DNA delivery and transfection, especially for silica with different surface functionality. Herein, we report that a higher content of hydrogen boning formed between PEI molecules and phosphonate modified silica nanoparticles could slow down the PEI dissolution from the freeze-dried solid composites into aqueous solution than the bare silica counterpart. The pronounced PEI retention ability through phosphonation of silica nanoparticles effectively improves the transfection efficiency due to the high DNA binding affinity extracellularly, effective lysosome escape and high nuclear entry of both PEI and DNA intracellularly. Our study provides a fundamental understanding on designing effective silica-PEI-based nano-vectors for DNA delivery applications.
Collapse
Affiliation(s)
- Dan Cheng
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Shevanuja Theivendran
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jie Tang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Larry Cai
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jun Zhang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Hao Song
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Chengzhong Yu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
16
|
Wang F, Liu LS, Li P, Lau CH, Leung HM, Chin YR, Tin C, Lo PK. Cellular uptake, tissue penetration, biodistribution, and biosafety of threose nucleic acids: Assessing in vitro and in vivo delivery. Mater Today Bio 2022; 15:100299. [PMID: 35637854 PMCID: PMC9142632 DOI: 10.1016/j.mtbio.2022.100299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/03/2022] [Accepted: 05/16/2022] [Indexed: 11/28/2022]
|
17
|
Lin G, Huang J, Zhang M, Chen S, Zhang M. Chitosan-Crosslinked Low Molecular Weight PEI-Conjugated Iron Oxide Nanoparticle for Safe and Effective DNA Delivery to Breast Cancer Cells. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:584. [PMID: 35214917 PMCID: PMC8876741 DOI: 10.3390/nano12040584] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/30/2022] [Accepted: 02/04/2022] [Indexed: 02/06/2023]
Abstract
Breast cancer has attracted tremendous research interest in treatment development as one of the major threats to public health. The use of non-viral carriers for therapeutic DNA delivery has shown promise in treating various cancer types, including breast cancer, due to their high DNA loading capacity, high cell transfection efficiency, and design versatility. However, cytotoxicity and large sizes of non-viral DNA carriers often raise safety concerns and hinder their applications in the clinic. Here we report the development of a novel nanoparticle formulation (termed NP-Chi-xPEI) that can safely and effectively deliver DNA into breast cancer cells for successful transfection. The nanoparticle is composed of an iron oxide core coated with low molecular weight (800 Da) polyethyleneimine crosslinked with chitosan via biodegradable disulfide bonds. The NP-Chi-xPEI can condense DNA into a small nanoparticle with the overall size of less than 100 nm and offer full DNA protection. Its biodegradable coating of small-molecular weight xPEI and mildly positive surface charge confer extra biocompatibility. NP-Chi-xPEI-mediated DNA delivery was shown to achieve high transfection efficiency across multiple breast cancer cell lines with significantly lower cytotoxicity as compared to the commercial transfection agent Lipofectamine 3000. With demonstrated favorable physicochemical properties and functionality, NP-Chi-xPEI may serve as a reliable vehicle to deliver DNA to breast cancer cells.
Collapse
Affiliation(s)
| | | | | | | | - Miqin Zhang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA; (G.L.); (J.H.); (M.Z.); (S.C.)
| |
Collapse
|
18
|
Hooshmand SE, Sabet MJ, Hasanzadeh A, Mousavi SMK, Moghadam NH, Hooshmand SA, Rabiee N, Liu Y, Hamblin MR, Karimi M. Histidine‐enhanced gene delivery systems: The state of the art. J Gene Med 2022; 24:e3415. [DOI: 10.1002/jgm.3415] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/26/2022] [Accepted: 01/29/2022] [Indexed: 11/08/2022] Open
Affiliation(s)
- Seyyed Emad Hooshmand
- Cellular and Molecular Research Center Iran University of Medical Sciences Tehran Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine Iran University of Medical Sciences Tehran Iran
| | - Makkieh Jahanpeimay Sabet
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine Iran University of Medical Sciences Tehran Iran
| | - Akbar Hasanzadeh
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine Iran University of Medical Sciences Tehran Iran
| | - Seyede Mahtab Kamrani Mousavi
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine Iran University of Medical Sciences Tehran Iran
| | - Niloofar Haeri Moghadam
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine Iran University of Medical Sciences Tehran Iran
| | - Seyed Aghil Hooshmand
- Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics University of Tehran Tehran Iran
| | - Navid Rabiee
- Department of Physics Sharif University of Technology Tehran Iran
- School of Engineering Macquarie University Sydney New South Wales Australia
| | - Yong Liu
- Institute of Functional Nano & Soft Materials (FUNSOM) Soochow University Suzhou Jiangsu China
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science University of Johannesburg South Africa
| | - Mahdi Karimi
- Cellular and Molecular Research Center Iran University of Medical Sciences Tehran Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine Iran University of Medical Sciences Tehran Iran
- Oncopathology Research Center Iran University of Medical Sciences Tehran Iran
- Research Center for Science and Technology in Medicine Tehran University of Medical Sciences Tehran Iran
| |
Collapse
|
19
|
Jiang Q, Guan S, Zhang Y, Sun Y, Jiang X. Targeted and fluorescence traceable multifunctional host-guest supramolecular gene delivery platform based on poly(cyclodextrin) and rhodamine conjugated disulfide-containing azobenzene-terminated branched polymer. INT J POLYM MATER PO 2022. [DOI: 10.1080/00914037.2022.2029438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Qimin Jiang
- Jiangsu Key Laboratory of Environmentally Friendly Polymeric Materials, School of Materials Science and Engineering, Jiangsu Collaborative Innovation Center of Photovolatic Science and Engineering, Changzhou University, Changzhou, P. R. China
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, P. R. China
| | - Shuyi Guan
- Jiangsu Key Laboratory of Environmentally Friendly Polymeric Materials, School of Materials Science and Engineering, Jiangsu Collaborative Innovation Center of Photovolatic Science and Engineering, Changzhou University, Changzhou, P. R. China
| | - Yunti Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, P. R. China
| | - Yuhua Sun
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R. China
| | - Xulin Jiang
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, P. R. China
| |
Collapse
|
20
|
Karimi Jabali M, Allafchian AR, Jalali SAH, Shakeripour H, Mohammadinezhad R, Rahmani F. Design of a pDNA nanocarrier with ascorbic acid modified chitosan coated on superparamagnetic iron oxide nanoparticles for gene delivery. Colloids Surf A Physicochem Eng Asp 2022. [DOI: 10.1016/j.colsurfa.2021.127743] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
21
|
Amani A, Dustparast M, Noruzpour M, Zakaria RA, Ebrahimi HA. Design and Invitro Characterization of Green Synthesized Magnetic Nanoparticles Conjugated with Multitargeted Poly Lactic Acid Copolymers for Co-delivery of siRNA and Paclitaxel. Eur J Pharm Sci 2021; 167:106007. [PMID: 34520835 DOI: 10.1016/j.ejps.2021.106007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 08/21/2021] [Accepted: 09/10/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND The self-assembling of various amphipathic copolymers is a simple method that allows the preparation of complex nanoparticles with several useful properties. In the present study, the polylactic acid-polyethylene glycol-folate (PLA-PEG-FA) (PPF), PLA-PEG-T7 peptide (PPT) and PLA-Chitosan-Spermine (PCS) copolymers were synthesized separately. METHODS These copolymers combined with Fe3O4 magnetic core and loaded with paclitaxel (PTX)/siRNA-FAM to form magnetic PCS/PPF/PPT/PTX/siRNA micelles (MPCSFT/PTX/siRNA) and were characterized using physicochemical and biological analysis. RESULTS The results revealed that the MPCSPFT/PTX/siRNA had spherical morphology with particle size and zeta potential about 197 nm and -7.8 mV, respectively. Release assay was determined under neutral (pH=7.4) and acidic pH (pH=6) conditions to simulate PTX and siRNA release profile from MPCSPFT/PTX/siRNA micelles in normal and cancerous tissues. The ability of MPCSPFT for co-delivery of PTX and siRNA into MCF-7 cells was determined by MTT and flow cytometry tests, respectively. The results revealed that the release rate of siRNA and PTX from MPCSPFT/PTX/siRNA nanoparticles under an acidic environment (pH=6) was significantly higher than that of their release rate in a neutral medium (pH=7.4). CONCLUSION Conjugation of both folic acid and T7-peptide on the surface of micelles compared to separate conjugation of one of these ligands, increased the efficiency of drug and siRNA delivery to breast cancer cells.
Collapse
Affiliation(s)
- Amin Amani
- Department of Pharmaceutics, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran; Department of Agronomy and Plant Breeding, Faculty of Agriculture and Natural Resources, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Mohammad Dustparast
- Department of Pharmaceutics, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mehran Noruzpour
- Department of Agronomy and Plant Breeding, Faculty of Agriculture and Natural Resources, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Rasool Asghari Zakaria
- Department of Agronomy and Plant Breeding, Faculty of Agriculture and Natural Resources, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Hossein Ali Ebrahimi
- Department of Pharmaceutics, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
22
|
Krishnan S, Thirunavukarasu A, Jha NK, Gahtori R, Roy AS, Dholpuria S, Kesari KK, Singh SK, Dua K, Gupta PK. Nanotechnology-based therapeutic formulations in the battle against animal coronaviruses: an update. JOURNAL OF NANOPARTICLE RESEARCH : AN INTERDISCIPLINARY FORUM FOR NANOSCALE SCIENCE AND TECHNOLOGY 2021; 23:229. [PMID: 34690535 PMCID: PMC8520458 DOI: 10.1007/s11051-021-05341-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 10/03/2021] [Indexed: 06/13/2023]
Abstract
Outbreak of infectious diseases imposes a serious threat to human population and also causes a catastrophic impact on global economy. Animal coronaviruses remain as one of the intriguing problems, known to cause deadly viral diseases on economically important animal population, and also these infections may spread to other animals and humans. Through isolation of the infected animals from others and providing appropriate treatment using antiviral drugs, it is possible to prevent the virus transmission from animals to other species. In recent times, antiviral drug-resistant strains are being emerged as a deadly virus which are known to cause pandemic. To overcome this, nanoparticles-based formulations are developed as antiviral agent which attacks the animal coronaviruses at multiple sites in the virus replication cycle. Nanovaccines are also being formulated to protect the animals from coronaviruses. Nanoformulations contain particles of one or more dimensions in nano-scale (few nanometers to 1000 nm), which could be inorganic or organic in nature. This review presents the comprehensive outline of the nanotechnology-based therapeutics formulated against animal coronaviruses, which includes the nanoparticles-based antiviral formulations and nanoparticles-based adjuvant vaccines. The mechanism of action of these nanoparticles-based antivirals against animal coronavirus is also discussed using relevant examples. In addition, the scope of repurposing the existing nano-enabled antivirals and vaccines to combat the coronavirus infections in animals is elaborated.
Collapse
Affiliation(s)
| | | | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology (SET), Sharda University, Plot no. 32 – 34, Knowledge Park III, Greater Noida, 201310 Uttar Pradesh India
| | - Rekha Gahtori
- Department of Biotechnology, Sir J. C. Bose Technical Campus, Kumaun University, Nainital, 263136 Uttarakhand India
| | - Ayush Singha Roy
- Department of Biotechnology, Amity School of Biotechnology, Amity University, Mumbai-Pune Expressway, Mumbai, Maharashtra 410206 India
| | - Sunny Dholpuria
- Department of Life Sciences, J.C. Bose University of Science and Technology, YMCA, Faridabad, 121006 Haryana India
| | | | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab India
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007 Australia
| | - Piyush Kumar Gupta
- Department of Life Sciences, School of Basic Sciences and Research (SBSR), Sharda University, Plot no. 32 – 34, Knowledge Park III, Greater Noida, 201310 Uttar Pradesh India
| |
Collapse
|
23
|
Tarvirdipour S, Skowicki M, Schoenenberger CA, Palivan CG. Peptide-Assisted Nucleic Acid Delivery Systems on the Rise. Int J Mol Sci 2021; 22:9092. [PMID: 34445799 PMCID: PMC8396486 DOI: 10.3390/ijms22169092] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/13/2021] [Accepted: 08/19/2021] [Indexed: 12/12/2022] Open
Abstract
Concerns associated with nanocarriers' therapeutic efficacy and side effects have led to the development of strategies to advance them into targeted and responsive delivery systems. Owing to their bioactivity and biocompatibility, peptides play a key role in these strategies and, thus, have been extensively studied in nanomedicine. Peptide-based nanocarriers, in particular, have burgeoned with advances in purely peptidic structures and in combinations of peptides, both native and modified, with polymers, lipids, and inorganic nanoparticles. In this review, we summarize advances on peptides promoting gene delivery systems. The efficacy of nucleic acid therapies largely depends on cell internalization and the delivery to subcellular organelles. Hence, the review focuses on nanocarriers where peptides are pivotal in ferrying nucleic acids to their site of action, with a special emphasis on peptides that assist anionic, water-soluble nucleic acids in crossing the membrane barriers they encounter on their way to efficient function. In a second part, we address how peptides advance nanoassembly delivery tools, such that they navigate delivery barriers and release their nucleic acid cargo at specific sites in a controlled fashion.
Collapse
Affiliation(s)
- Shabnam Tarvirdipour
- Department of Chemistry, University of Basel, Mattenstrasse 24a, 4058 Basel, Switzerland; (S.T.); (M.S.)
- Department of Biosystem Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Michal Skowicki
- Department of Chemistry, University of Basel, Mattenstrasse 24a, 4058 Basel, Switzerland; (S.T.); (M.S.)
- NCCR-Molecular Systems Engineering, BPR1095, Mattenstrasse 24a, 4058 Basel, Switzerland
| | - Cora-Ann Schoenenberger
- Department of Chemistry, University of Basel, Mattenstrasse 24a, 4058 Basel, Switzerland; (S.T.); (M.S.)
- NCCR-Molecular Systems Engineering, BPR1095, Mattenstrasse 24a, 4058 Basel, Switzerland
| | - Cornelia G. Palivan
- Department of Chemistry, University of Basel, Mattenstrasse 24a, 4058 Basel, Switzerland; (S.T.); (M.S.)
- NCCR-Molecular Systems Engineering, BPR1095, Mattenstrasse 24a, 4058 Basel, Switzerland
| |
Collapse
|
24
|
Tang F, Liu JY, Wu CY, Liang YX, Lu ZL, Ding AX, Xu MD. Two-Photon Near-Infrared AIE Luminogens as Multifunctional Gene Carriers for Cancer Theranostics. ACS APPLIED MATERIALS & INTERFACES 2021; 13:23384-23395. [PMID: 33982571 DOI: 10.1021/acsami.1c02600] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Construction of multifunctional nonviral gene vectors to execute defined tasks holds great potential for the precise and effective treatment of gene-associated diseases. Herein, we have developed four large π-conjugation triphenylamine derivatives bearing two polar [12]aneN3 heads and a lipophilic tail for applications in gene delivery, one/two-photon-triggered near-infrared (NIR) fluorescence bioimaging, and combined photodynamic therapy (PDT) and gene therapy of cancer. These compounds possess typical NIR aggregation-induced emission characteristics, mega Stokes shifts, strong two-photon excitation fluorescence, and excellent DNA condensation abilities. Among them, vector 4 with a tail of n-hexadecane realized a transfection efficiency as high as 6.7 times that of the commercial transfection agent Lipofectamine 2000 in HEK293T cell lines. Using vector 4 as an example, transfection process tracking and ex vivo/in vivo tumoral imaging and retention with high resolution, high brightness, deep tissue penetration, and good biosafety were demonstrated. In addition, efficient singlet oxygen (1O2) generation by the DNA complex formed by vector 4 (4/DNA) resulted in effective PDT. Combined with anticancer gene therapy, collaborative cancer treatment with a dramatically enhanced cancer cell-killing effect was achieved. The development of this "three birds, one stone" approach suggests a new and promising strategy for better cancer treatment and real-time tracking of gene delivery.
Collapse
Affiliation(s)
- Fang Tang
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Jin-Yu Liu
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Cheng-Yan Wu
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Ya-Xuan Liang
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Zhong-Lin Lu
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Ai-Xiang Ding
- College of Chemistry and Chemical Engineering, Xinyang Normal University, Xinyang 464000, China
| | - Ming-Di Xu
- China National Institute for Food and Drug Control, Institute of Chemical Drug Control, Tian Tan XiLi 2, Beijing 100050, China
| |
Collapse
|
25
|
Liu SY, Xu Y, Yang H, Liu L, Zhao M, Yin W, Xu YT, Huang Y, Tan C, Dai Z, Zhang H, Zhang JP, Chen XM. Ultrathin 2D Copper(I) 1,2,4-Triazolate Coordination Polymer Nanosheets for Efficient and Selective Gene Silencing and Photodynamic Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2100849. [PMID: 33797149 DOI: 10.1002/adma.202100849] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/22/2021] [Indexed: 06/12/2023]
Abstract
Gene silencing holds promise for cancer therapeutics because of its potential to inhibit genes involved in tumor development. However, gene silencing is still restricted by its limited efficacy and safety. Nanoscale coordination polymers (CPs) emerge as promising nanocarriers for gene delivery, but their responsiveness and potential therapeutic properties have rarely been explored simultaneously. Here, multifunctional ultrathin 2D nanosheets of Cu(I) 1,2,4-triazolate CP with a thickness of 4.5 ± 0.8 nm are synthesized using a bottom-up method. These CP nanosheets can act as both an effective DNAzyme nanocarrier for gene therapy and an intrinsic photosensitizer for hypoxia-tolerant type I photodynamic therapy (PDT), which is ascribed to the Fenton-like reaction. Because of the glutathione (GSH)-responsiveness of the CP nanosheets, DNAzyme-loaded CP nanosheets exhibit excellent cancer-cell-targeting gene silencing of the early growth response factor-1 (EGR-1), with messenger RNA inhibited by 84% in MCF-7 (human breast cancer cells) and only 6% in MCF-10A (normal human mammary epithelial cells). After tail intravenous injection into MCF-7-tumor-bearing mice, the CP nanosheets loaded with chlorin-e6-modified DNAzyme under photoirradiation show a high antitumor efficacy (88.0% tumor regression), demonstrating a promising therapeutic platform with efficient and selective gene silencing and PDT of cancer.
Collapse
Affiliation(s)
- Si-Yang Liu
- School of Biomedical Engineering, Sun Yat-Sen University, Guangzhou, 510006, China
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, China
- Center for Programmable Materials, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Yuzhi Xu
- Scientific Research Center, Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Huihui Yang
- School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Liping Liu
- School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Meiting Zhao
- Center for Programmable Materials, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Wen Yin
- School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Yan-Tong Xu
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Ying Huang
- Center for Programmable Materials, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Chaoliang Tan
- Department of Electrical Engineering, City University of Hong Kong, Kowloon, Hong Kong, China
| | - Zong Dai
- School of Biomedical Engineering, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Hua Zhang
- Department of Chemistry, City University of Hong Kong, Kowloon, Hong Kong, China
- Hong Kong Branch of National Precious Metals Material Engineering Research Center (NPMM), City University of Hong Kong, Hong Kong, China
| | - Jie-Peng Zhang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Xiao-Ming Chen
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, China
| |
Collapse
|
26
|
Torres-Vanegas JD, Cruz JC, Reyes LH. Delivery Systems for Nucleic Acids and Proteins: Barriers, Cell Capture Pathways and Nanocarriers. Pharmaceutics 2021; 13:428. [PMID: 33809969 PMCID: PMC8004853 DOI: 10.3390/pharmaceutics13030428] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/09/2021] [Accepted: 03/12/2021] [Indexed: 12/27/2022] Open
Abstract
Gene therapy has been used as a potential approach to address the diagnosis and treatment of genetic diseases and inherited disorders. In this line, non-viral systems have been exploited as promising alternatives for delivering therapeutic transgenes and proteins. In this review, we explored how biological barriers are effectively overcome by non-viral systems, usually nanoparticles, to reach an efficient delivery of cargoes. Furthermore, this review contributes to the understanding of several mechanisms of cellular internalization taken by nanoparticles. Because a critical factor for nanoparticles to do this relies on the ability to escape endosomes, researchers have dedicated much effort to address this issue using different nanocarriers. Here, we present an overview of the diversity of nanovehicles explored to reach an efficient and effective delivery of both nucleic acids and proteins. Finally, we introduced recent advances in the development of successful strategies to deliver cargoes.
Collapse
Affiliation(s)
- Julian D. Torres-Vanegas
- Grupo de Diseño de Productos y Procesos (GDPP), Department of Chemical and Food Engineering, Universidad de los Andes, Bogotá 111711, Colombia
| | - Juan C. Cruz
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia
| | - Luis H. Reyes
- Grupo de Diseño de Productos y Procesos (GDPP), Department of Chemical and Food Engineering, Universidad de los Andes, Bogotá 111711, Colombia
| |
Collapse
|
27
|
Lin G, Revia RA, Zhang M. Inorganic Nanomaterial-Mediated Gene Therapy in Combination with Other Antitumor Treatment Modalities. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2007096. [PMID: 34366761 PMCID: PMC8336227 DOI: 10.1002/adfm.202007096] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Indexed: 05/05/2023]
Abstract
Cancer is a genetic disease originating from the accumulation of gene mutations in a cellular subpopulation. Although many therapeutic approaches have been developed to treat cancer, recent studies have revealed an irrefutable challenge that tumors evolve defenses against some therapies. Gene therapy may prove to be the ultimate panacea for cancer by correcting the fundamental genetic errors in tumors. The engineering of nanoscale inorganic carriers of cancer therapeutics has shown promising results in the efficacious and safe delivery of nucleic acids to treat oncological diseases in small-animal models. When these nanocarriers are used for co-delivery of gene therapeutics along with auxiliary treatments, the synergistic combination of therapies often leads to an amplified health benefit. In this review, an overview of the inorganic nanomaterials developed for combinatorial therapies of gene and other treatment modalities is presented. First, the main principles of using nucleic acids as therapeutics, inorganic nanocarriers for medical applications and delivery of gene/drug payloads are introduced. Next, the utility of recently developed inorganic nanomaterials in different combinations of gene therapy with each of chemo, immune, hyperthermal, and radio therapy is examined. Finally, current challenges in the clinical translation of inorganic nanomaterial-mediated therapies are presented and outlooks for the field are provided.
Collapse
Affiliation(s)
- Guanyou Lin
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | - Richard A Revia
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | - Miqin Zhang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
28
|
Han X, Zhang L, Zhang Q, Sui X, Qian M, Chen Q, Wang J. Construction of a Novel Reactive Oxygen Species-responsive Cationic Copolymer and Its Performance in Gene Delivery. ACTA CHIMICA SINICA 2021. [DOI: 10.6023/a21030090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
29
|
Kheiriabad S, Dolatabadi JEN, Hamblin MR. Dendrimers for gene therapy. DENDRIMER-BASED NANOTHERAPEUTICS 2021:285-309. [DOI: 10.1016/b978-0-12-821250-9.00026-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
30
|
Wang X, Qiu Y, Wang M, Zhang C, Zhang T, Zhou H, Zhao W, Zhao W, Xia G, Shao R. Endocytosis and Organelle Targeting of Nanomedicines in Cancer Therapy. Int J Nanomedicine 2020; 15:9447-9467. [PMID: 33268987 PMCID: PMC7701161 DOI: 10.2147/ijn.s274289] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 09/25/2020] [Indexed: 12/15/2022] Open
Abstract
Nanomedicines (NMs) have played an increasing role in cancer therapy as carriers to efficiently deliver therapeutics into tumor cells. For this application, the uptake of NMs by tumor cells is usually a prerequisite to deliver the cargo to intracellular locations, which mainly relies on endocytosis. NMs can enter cells through a variety of endocytosis pathways. Different endocytosis pathways exhibit different intracellular trafficking routes and diverse subcellular localizations. Therefore, a comprehensive understanding of endocytosis mechanisms is necessary for increasing cellular entry efficiency and to trace the fate of NMs after internalization. This review focuses on endocytosis pathways of NMs in tumor cells, mainly including clathrin- and caveolae-mediated endocytosis pathways, involving effector molecules, expression difference of those molecules between normal and tumor cells, as well as the intracellular trafficking route of corresponding endocytosis vesicles. Then, the latest strategies for NMs to actively employ endocytosis are described, including improving tumor cellular uptake of NMs by receptor-mediated endocytosis, transporter-mediated endocytosis and enabling drug activity by changing intracellular routes. Finally, active targeting strategies towards intracellular organelles are also mentioned. This review will be helpful not only in explicating endocytosis and the trafficking process of NMs and elucidating anti-tumor mechanisms inside the cell but also in rendering new ideas for the design of highly efficacious and cancer-targeted NMs.
Collapse
Affiliation(s)
- Xiaowei Wang
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Yuhan Qiu
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Mengyan Wang
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Conghui Zhang
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Tianshu Zhang
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Huimin Zhou
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Wenxia Zhao
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Wuli Zhao
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Guimin Xia
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Rongguang Shao
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| |
Collapse
|
31
|
Lugin ML, Lee RT, Kwon YJ. Synthetically Engineered Adeno-Associated Virus for Efficient, Safe, and Versatile Gene Therapy Applications. ACS NANO 2020; 14:14262-14283. [PMID: 33073995 DOI: 10.1021/acsnano.0c03850] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Gene therapy directly targets mutations causing disease, allowing for a specific treatment at a molecular level. Adeno-associated virus (AAV) has been of increasing interest as a gene delivery vehicle, as AAV vectors are safe, effective, and capable of eliciting a relatively contained immune response. With the recent FDA approval of two AAV drugs for treating rare genetic diseases, AAV vectors are now on the market and are being further explored for other therapies. While showing promise in immune privileged tissue, the use of AAV for systemic delivery is still limited due to the high prevalence of neutralizing antibodies (nAbs). To avoid nAb-mediated inactivation, engineered AAV vectors with modified protein capsids, materials tethered to the capsid surface, or fully encapsulated in a second, larger carrier have been explored. Many of these engineered AAVs have added benefits, including avoided immune response, overcoming the genome size limit, targeted and stimuli-responsive delivery, and multimodal therapy of two or more therapeutic modalities in one platform. Native and engineered AAV vectors have been tested to treat a broad range of diseases, including spinal muscular atrophy, retinal diseases, cancers, and tissue damage. This review will cover the benefits of AAV as a promising gene vector by itself, the progress and advantages of engineered AAV vectors, particularly synthetically engineered ones, and the current state of their clinical translation in therapy.
Collapse
|
32
|
Garcia BB, Mertins O, Silva ERD, Mathews PD, Han SW. Arginine-modified chitosan complexed with liposome systems for plasmid DNA delivery. Colloids Surf B Biointerfaces 2020; 193:111131. [DOI: 10.1016/j.colsurfb.2020.111131] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/06/2020] [Accepted: 05/11/2020] [Indexed: 12/13/2022]
|
33
|
Li YQ, Sun W, Liu XY, Chen LQ, Huang W, Lu ZL, He L. Synthesis of Glutathione (GSH)-Responsive Amphiphilic Duplexes and their Application in Gene Delivery. Chempluschem 2020; 84:1060-1069. [PMID: 31943961 DOI: 10.1002/cplu.201900295] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 07/13/2019] [Indexed: 12/16/2022]
Abstract
Oligoamide molecular strands with hydrogen-bonding sequences DADDAD and guanidine (O-1) or 1,5,9-triazacyclododecane ([12]aneN3 ; O-2) side chains and oligoamides with hydrogen-bonding sequences ADAADA and octyl moieties (O-3), were synthesized. Two duplexes (D-1 and D-2) were prepared by conjugating the hydrophilic O-1 or O-2 and hydrophobic O-3 through sequence-specific hydrogen-bond association and cross-linked disulfide bonds. Electrophoresis measurements indicated that O-1, O-2, D-1, and D-2 were able to completely retard the DNA mobiliy at concentrations of 30, 30, 10, and 20 μM, respectively. Reversible DNA release in O-1 and O-2 complexes can be achieved in the presence of heparin sodium, whereas the presence of GSH greatly improved DNA release in D-1 and D-2 complexes. The particles formed were in a size range of 50-170 nm with positively charged surfaces. D-1 and D-2 transfected pEGFP-N1 into HeLa cells successfully.
Collapse
Affiliation(s)
- Yong-Qiang Li
- College of Chemistry, Bejjing Normal University, Xinjiekouwai Street 19, Beijing, China.,State Key laboratory of bioactive substance and Function of Natural Medicines Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Xiannongtan Street 1, Beijing, China
| | - Wan Sun
- College of Chemistry, Bejjing Normal University, Xinjiekouwai Street 19, Beijing, China
| | - Xu-Ying Liu
- College of Chemistry, Bejjing Normal University, Xinjiekouwai Street 19, Beijing, China
| | - Li-Qing Chen
- State Key laboratory of bioactive substance and Function of Natural Medicines Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Xiannongtan Street 1, Beijing, China
| | - Wei Huang
- State Key laboratory of bioactive substance and Function of Natural Medicines Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Xiannongtan Street 1, Beijing, China
| | - Zhong-Lin Lu
- College of Chemistry, Bejjing Normal University, Xinjiekouwai Street 19, Beijing, China
| | - Lan He
- National Institute for Food and Drug Control, Institute of Chemical Drug Control, TianTan XiLi 2, Beijing, 100050, China
| |
Collapse
|
34
|
Chen P, Zhang J, He X, Liu YH, Yu XQ. Hydrophobically modified carbon dots as a multifunctional platform for serum-resistant gene delivery and cell imaging. Biomater Sci 2020; 8:3730-3740. [PMID: 32501458 DOI: 10.1039/d0bm00651c] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The development of novel multifunctional gene delivery systems with high efficiency is significant. Herein, due to the unique physical and optical properties of carbon dots (CDs), CDs prepared from polyethyleneimine (PEI) were modified with various hydrophobic chains and different degrees of substitution via an epoxide ring-opening reaction. The modification and substitution degree were confirmed using several analytical methods including 1H NMR spectroscopy, FT-IR spectroscopy, TEM, and XPS. These CDs were utilized as multifunctional, safe and efficient non-viral gene vectors. The results showed that these materials possessed capability for dual-channel imaging, which enabled the intracellular tracking of the delivered DNA. Both the type and substitution degree of the hydrophobic chain have a large influence on their transfection efficiency. Among the prepared CDs, Ole1.5-CD gave the highest transfection efficiency, which was up to 200 times higher than that of PEI 25 kDa in the presence of serum in A549 cells. Meanwhile, these CD materials showed much less cytotoxicity and better serum tolerance than the traditional cationic polymeric gene vector. The cellular uptake assay further confirmed the good serum tolerance and structure-activity relationship of the CD materials. Thus, these CDs with good biocompatibility, self-imaging and high gene transfection efficiency may serve as a promising platform for both gene delivery and bio-imaging.
Collapse
Affiliation(s)
- Ping Chen
- Key Laboratory of Green Chemistry and Technology (Ministry of Education), College of Chemistry, Sichuan University, Chengdu 610064, P. R. China.
| | | | | | | | | |
Collapse
|
35
|
Heitz M, Zamolo S, Javor S, Reymond JL. Fluorescent Peptide Dendrimers for siRNA Transfection: Tracking pH Responsive Aggregation, siRNA Binding, and Cell Penetration. Bioconjug Chem 2020; 31:1671-1684. [PMID: 32421327 DOI: 10.1021/acs.bioconjchem.0c00231] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Transfecting nucleic acids into various cells is a key procedure in biological research also envisioned for therapeutic applications. In our effort to obtain simple reagents that would be readily accessible from commercial building blocks, we recently reported peptide dendrimers as single component siRNA transfection reagents accessible in pure form by solid-phase peptide synthesis. Here, we extend our studies of these dendrimers by identifying analogs bearing a coumarin or BODIPY fluorescent label in their core and displaying comparable siRNA transfection efficiencies, pH dependent aggregation, siRNA binding, and secondary structures. Fluorescence resonance energy transfer (FRET) studies show that the dendrimers are tightly associated with siRNA within the formed nanoparticles at pH 7.4 but are released into solution at pH 5.0 and can participate in endosome escape by destabilizing the membrane at this pH value. Colocalization studies furthermore suggest that peptide dendrimers and siRNA remain tightly associated throughout the transfection process.
Collapse
Affiliation(s)
- Marc Heitz
- Department of Chemistry and Biochemistry, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland
| | - Susanna Zamolo
- Department of Chemistry and Biochemistry, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland
| | - Sacha Javor
- Department of Chemistry and Biochemistry, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland
| | - Jean-Louis Reymond
- Department of Chemistry and Biochemistry, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland
| |
Collapse
|
36
|
Speranskaya ES, Drozd DD, Pidenko PS, Goryacheva IY. Enzyme modulation of quantum dot luminescence: Application in bioanalysis. Trends Analyt Chem 2020. [DOI: 10.1016/j.trac.2020.115897] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
37
|
Li L, Chen Y, Xu G, Liu D, Yang Z, Chen T, Wang X, Jiang W, Xue D, Lin G. In vivo Comparison of the Biodistribution and Toxicity of InP/ZnS Quantum Dots with Different Surface Modifications. Int J Nanomedicine 2020; 15:1951-1965. [PMID: 32256071 PMCID: PMC7093098 DOI: 10.2147/ijn.s241332] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 03/10/2020] [Indexed: 12/18/2022] Open
Abstract
INTRODUCTION Indium phosphide (InP) quantum dots (QDs) have shown a broad application prospect in the fields of biophotonics and nanomedicine. However, the potential toxicity of InP QDs has not been systematically evaluated. In particular, the effects of different surface modifications on the biodistribution and toxicity of InP QDs are still unknown, which hinders their further developments. The present study aims to investigate the biodistribution and in vivo toxicity of InP/ZnS QDs. METHODS Three kinds of InP/ZnS QDs with different surface modifications, hQDs (QDs-OH), aQDs (QDs-NH2), and cQDs (QDs-COOH) were intravenously injected into BALB/c mice at the dosage of 2.5 mg/kg BW or 25 mg/kg BW, respectively. Biodistribution of three QDs was determined through cryosection fluorescence microscopy and ICP-MS analysis. The subsequent effects of InP/ZnS QDs on histopathology, hematology and blood biochemistry were evaluated at 1, 3, 7, 14 and 28 days post-injection. RESULTS These types of InP/ZnS QDs were rapidly distributed in the major organs of mice, mainly in the liver and spleen, and lasted for 28 days. No abnormal behavior, weight change or organ index were observed during the whole observation period, except that 2 mice died on Day 1 after 25 mg/kg BW hQDs treatment. The results of H&E staining showed that no obvious histopathological abnormalities were observed in the main organs (including heart, liver, spleen, lung, kidney, and brain) of all mice injected with different surface-functionalized QDs. Low concentration exposure of three QDs hardly caused obvious toxicity, while high concentration exposure of the three QDs could cause some changes in hematological parameters or biochemical parameters related to liver function or cardiac function. More attention needs to be paid on cQDs as high-dose exposure of cQDs induced death, acute inflammatory reaction and slight changes in liver function in mice. CONCLUSION The surface modification and exposure dose can influence the biological behavior and in vivo toxicity of QDs. The surface chemistry should be fully considered in the design of InP-based QDs for their biomedical applications.
Collapse
Affiliation(s)
- Li Li
- Base for International Science and Technology Cooperation: Carson Cancer Stem Cell Vaccines R&D Center, Shenzhen Key Laboratory of Synthetic Biology, Department of Physiology, School of Basic Medical Sciences, Shenzhen University, Shenzhen518055, People’s Republic of China
- Key Laboratory of Optoelectronics Devices and Systems of Ministry of Education/Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen518060, People’s Republic of China
| | - Yajing Chen
- Base for International Science and Technology Cooperation: Carson Cancer Stem Cell Vaccines R&D Center, Shenzhen Key Laboratory of Synthetic Biology, Department of Physiology, School of Basic Medical Sciences, Shenzhen University, Shenzhen518055, People’s Republic of China
| | - Gaixia Xu
- Key Laboratory of Optoelectronics Devices and Systems of Ministry of Education/Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen518060, People’s Republic of China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen518055, People’s Republic of China
| | - Dongmeng Liu
- Base for International Science and Technology Cooperation: Carson Cancer Stem Cell Vaccines R&D Center, Shenzhen Key Laboratory of Synthetic Biology, Department of Physiology, School of Basic Medical Sciences, Shenzhen University, Shenzhen518055, People’s Republic of China
| | - Zhiwen Yang
- Base for International Science and Technology Cooperation: Carson Cancer Stem Cell Vaccines R&D Center, Shenzhen Key Laboratory of Synthetic Biology, Department of Physiology, School of Basic Medical Sciences, Shenzhen University, Shenzhen518055, People’s Republic of China
| | - Tingting Chen
- Base for International Science and Technology Cooperation: Carson Cancer Stem Cell Vaccines R&D Center, Shenzhen Key Laboratory of Synthetic Biology, Department of Physiology, School of Basic Medical Sciences, Shenzhen University, Shenzhen518055, People’s Republic of China
| | - Xiaomei Wang
- Base for International Science and Technology Cooperation: Carson Cancer Stem Cell Vaccines R&D Center, Shenzhen Key Laboratory of Synthetic Biology, Department of Physiology, School of Basic Medical Sciences, Shenzhen University, Shenzhen518055, People’s Republic of China
| | - Wenxiao Jiang
- Base for International Science and Technology Cooperation: Carson Cancer Stem Cell Vaccines R&D Center, Shenzhen Key Laboratory of Synthetic Biology, Department of Physiology, School of Basic Medical Sciences, Shenzhen University, Shenzhen518055, People’s Republic of China
| | - Dahui Xue
- Base for International Science and Technology Cooperation: Carson Cancer Stem Cell Vaccines R&D Center, Shenzhen Key Laboratory of Synthetic Biology, Department of Physiology, School of Basic Medical Sciences, Shenzhen University, Shenzhen518055, People’s Republic of China
| | - Guimiao Lin
- Base for International Science and Technology Cooperation: Carson Cancer Stem Cell Vaccines R&D Center, Shenzhen Key Laboratory of Synthetic Biology, Department of Physiology, School of Basic Medical Sciences, Shenzhen University, Shenzhen518055, People’s Republic of China
| |
Collapse
|
38
|
Hashimoto T, Hirata T, Yuba E, Harada A, Kono K. Light-Activatable Transfection System Using Hybrid Vectors Composed of Thermosensitive Dendron Lipids and Gold Nanorods. Pharmaceutics 2020; 12:pharmaceutics12030239. [PMID: 32156051 PMCID: PMC7150951 DOI: 10.3390/pharmaceutics12030239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 02/28/2020] [Accepted: 03/05/2020] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Gene delivery to target cells is crucially important to establish gene therapy and regenerative medicine. Although various virus-based and synthetic molecule-based gene vectors have been developed to date, selective transfection in a site or a cell level is still challenging. For this study, both light-responsive and temperature-responsive synthetic gene vectors were designed for spatiotemporal control of a transfection system. METHODS 11-Mercaptoundecanoic acid-coated gold nanorods were mixed with polyamidoamine dendron-bearing lipids of two types having amino-terminus or ethoxydiethylene glycol-terminus to obtain hybrid vectors. Hybrid vectors were mixed further with pDNA. Then we investigated their physicochemical properties and transfection efficacy with or without near infrared laser irradiation. RESULTS Hybrid vectors formed complexes with pDNA and exhibited enhanced photothermal property under near infrared laser irradiation compared with parent gold nanorods. Transfection efficacy of complexes was promoted considerably by brief laser irradiation soon after complex application to the cells. Analysis of intracellular distribution revealed that laser irradiation promoted the adsorption of complexes to the cells and cytosolic release of pDNA, which is derived from the change in surface hydrophobicity of complexes through dehydration of temperature-responsive groups. CONCLUSIONS Hybrid vector is promising as a light-activatable transfection system.
Collapse
Affiliation(s)
| | | | - Eiji Yuba
- Correspondence: (E.Y.); (A.H.); Tel.: +81-72-254-9330 (E.Y.); Fax: +81-72-254-9330 (E.Y.)
| | - Atsushi Harada
- Correspondence: (E.Y.); (A.H.); Tel.: +81-72-254-9330 (E.Y.); Fax: +81-72-254-9330 (E.Y.)
| | | |
Collapse
|
39
|
Wu XR, Zhang J, Zhang JH, Xiao YP, He X, Liu YH, Yu XQ. Amino Acid-Linked Low Molecular Weight Polyethylenimine for Improved Gene Delivery and Biocompatibility. Molecules 2020; 25:E975. [PMID: 32098282 PMCID: PMC7070781 DOI: 10.3390/molecules25040975] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/07/2020] [Accepted: 02/19/2020] [Indexed: 12/13/2022] Open
Abstract
The construction of efficient and low toxic non-viral gene delivery vectors is of great significance for gene therapy. Herein, two novel polycations were constructed via Michael addition from low molecular weight polyethylenimine (PEI) 600 Da and amino acid-containing linkages. Lysine and histidine were introduced for the purpose of improved DNA binding and pH buffering capacity, respectively. The ester bonds afforded the polymer biodegradability, which was confirmed by the gel permeation chromatography (GPC) measurement. The polymers could well condense DNA into nanoparticles and protect DNA from degradation by nuclease. Compared with PEI 25 kDa, these polymers showed higher transfection efficiency, lower toxicity, and better serum tolerance. Study of this mechanism revealed that the polyplexes enter the cells mainly through caveolae-mediated endocytosis pathway; this, together with their biodegradability, facilitates the internalization of polyplexes and the release of DNA. The results reveal that the amino acid-linked low molecular weight PEI polymers could serve as promising candidates for non-viral gene delivery.
Collapse
Affiliation(s)
| | - Ji Zhang
- Key Laboratory of Green Chemistry and Technology (Ministry of Education), College of Chemistry, Sichuan University, Chengdu 610064, China; (X.-R.W.); (J.-H.Z.); (Y.-P.X.); (X.H.); (Y.-H.L.)
| | | | | | | | | | - Xiao-Qi Yu
- Key Laboratory of Green Chemistry and Technology (Ministry of Education), College of Chemistry, Sichuan University, Chengdu 610064, China; (X.-R.W.); (J.-H.Z.); (Y.-P.X.); (X.H.); (Y.-H.L.)
| |
Collapse
|
40
|
Tarvirdipour S, Schoenenberger CA, Benenson Y, Palivan CG. A self-assembling amphiphilic peptide nanoparticle for the efficient entrapment of DNA cargoes up to 100 nucleotides in length. SOFT MATTER 2020; 16:1678-1691. [PMID: 31967171 DOI: 10.1039/c9sm01990a] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
To overcome the low efficiency and cytotoxicity associated with most non-viral DNA delivery systems we developed a purely peptidic self-assembling system that is able to entrap single- and double-stranded DNA of up to 100 nucleotides in length. (HR)3gT peptide design consists of a hydrophilic domain prone to undergo electrostatic interactions with DNA cargo, and a hydrophobic domain at a ratio that promotes the self-assembly into multi-compartment micellar nanoparticles (MCM-NPs). Self-assembled (HR)3gT MCM-NPs range between 100 to 180 nm which is conducive to a rapid and efficient uptake by cells. (HR)3gT MCM-NPs had no adverse effects on HeLa cell viability. In addition, they exhibit long-term structural stability at 4 °C but at 37 °C, the multi-micellar organization disassembles overtime which demonstrates their thermo-responsiveness. The comparison of (HR)3gT to a shorter, less charged H3gT peptide indicates that the additional arginine residues result in the incorporation of longer DNA segments, an improved DNA entrapment efficiency and an increase cellular uptake. Our unique non-viral system for DNA delivery sets the stage for developing amphiphilic peptide nanoparticles as candidates for future systemic gene delivery.
Collapse
Affiliation(s)
- Shabnam Tarvirdipour
- Department of Chemistry, University of Basel, Mattenstrasse 24a, 4058 Basel, Switzerland.
| | | | | | | |
Collapse
|
41
|
Mitochondria-targeting nanomedicine self-assembled from GSH-responsive paclitaxel-ss-berberine conjugate for synergetic cancer treatment with enhanced cytotoxicity. J Control Release 2020; 318:38-49. [DOI: 10.1016/j.jconrel.2019.12.011] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 12/03/2019] [Accepted: 12/09/2019] [Indexed: 01/08/2023]
|
42
|
|
43
|
Jiang L, Ding Z, Xia S, Liu Y, Lei S, Zhong M, Chen X. Poly lactic-co-glycolic acid scaffold loaded with plasmid DNA encoding fibroblast growth factor-2 promotes periodontal ligament regeneration of replanted teeth. J Periodontal Res 2020; 55:488-495. [PMID: 31960451 DOI: 10.1111/jre.12734] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 12/12/2019] [Accepted: 12/28/2019] [Indexed: 12/21/2022]
Abstract
OBJECTIVE This study investigated the effects of poly lactic-co-glycolic acid (PLGA) loaded with plasmid DNA encoding fibroblast growth factor-2 (pFGF-2) on human periodontal ligament cells (hPDLCs) in vitro and evaluated the ability of the PLGA/pFGF-2 scaffold to promote periodontal ligament (PDL) regeneration in a beagle dog teeth avulsion animal model. BACKGROUND Growth factor and scaffold play important roles in PDL regeneration. PLGA is a kind of biodegradable and biocompatible polymer that can be used as a carrier to deliver growth factors or genes. FGF-2 can induce potent proliferative responses, promote cell migration and regulate the production of extracellular matrix. Therefore, a gene-activated matrix composed of scaffold and genes is supposed to be a superior approach for promoting tissue regeneration. METHODS In this study, PLGA and PLGA/pFGF-2 scaffolds were fabricated using electrospinning. The characterization of scaffolds was shown by scanning electron microscope (SEM) and transmission electron microscope (TEM). dsDNA HS was used to test the plasmid release of PLGA/pFGF-2 scaffold. The viability and proliferation of hPDLCs on the two kinds of scaffolds were evaluated by the CCK-8 assay, and the expression of collagen I and scleraxis were analysed by RT-qPCR. The roots of avulsed teeth were covered by the two types of scaffolds and replanted into the alveolar pockets in beagles. Haematoxylin-eosin and Masson staining were used to evaluate the effects of PLGA/pFGF-2 scaffold on promoting PDL regeneration. RESULTS The smooth and uniform fibres can be observed in both scaffolds, and the plasmids were randomly distributed in the PLGA/pFGF-2 scaffold. dsDNA HS analysis demonstrated that the PLGA/pFGF-2 scaffold released up to 123 ng pFGF-2 over 21 days in a sustained manner without any obvious burst release. The PLGA/pFGF-2 scaffold promoted the proliferation of hPDLCs and increased the expression levels of collagen I and scleraxis compared with PLGA scaffold. Animal experiments showed that more regular PDL-like tissues and less root surface resorption occurred in the PLGA/pFGF-2 scaffold group compared with the PLGA scaffold group. CONCLUSIONS The PLGA/pFGF-2 scaffold promoted hPDLCs proliferation and facilitated periodontal ligament-related differentiation. The PLGA/pFGF-2 scaffold possesses excellent biological characteristics and could be used as a promising biomaterial for improving the treatment prognosis of replanted tooth.
Collapse
Affiliation(s)
- Liming Jiang
- Department of Paediatric Dentistry, School of Stomatology, China Medical University, Shenyang, Liaoning, China
| | - Zhenjiang Ding
- Department of Paediatric Dentistry, School of Stomatology, China Medical University, Shenyang, Liaoning, China
| | - Shang Xia
- Department of Paediatric Dentistry, School of Stomatology, China Medical University, Shenyang, Liaoning, China
| | - Yao Liu
- Department of Paediatric Dentistry, School of Stomatology, China Medical University, Shenyang, Liaoning, China
| | - Shuang Lei
- Department of Paediatric Dentistry, School of Stomatology, China Medical University, Shenyang, Liaoning, China
| | - Ming Zhong
- Department of Oral Histopathology, School of Stomatology, China Medical University, Shenyang, Liaoning, China
| | - Xu Chen
- Department of Paediatric Dentistry, School of Stomatology, China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
44
|
Dibaba ST, Caputo R, Xi W, Zhang JZ, Wei R, Zhang Q, Zhang J, Ren W, Sun L. NIR Light-Degradable Antimony Nanoparticle-Based Drug-Delivery Nanosystem for Synergistic Chemo-Photothermal Therapy in Vitro. ACS APPLIED MATERIALS & INTERFACES 2019; 11:48290-48299. [PMID: 31802657 DOI: 10.1021/acsami.9b20249] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
A novel drug-delivery nanosystem based on near-infrared (NIR) light-degradable antimony nanoparticles (AMNP) have been developed for synergistic chemo-phototherapy in vitro. The monodispersed AMNP were synthesized by using a simple and cost-effective method. Positively charged doxorubicin hydrochloride (DOX) was loaded onto the negatively charged surface of AMNP via electrostatic interaction and finally modified by polyacrylic acid (PAA) to enhance biocompatibility. Under NIR (808 nm) laser irradiation of the AMNP-DOX-PAA nanosystem, not only was high photothermal conversion efficiency of AMNP achieved but also pH-dependent DOX release was enhanced due to laser-induced hyperthermia. As a consequence, almost all of the HeLa cells (around 97%) were killed because of the combined effects of chemotherapy and photothermal therapy. More interestingly, AMNP showed very fast (about 10 min) laser-induced degradation that may help to minimize long-term toxicity after therapy by using same-wavelength NIR laser irradiation (808 nm). Computational total energy calculations and molecular dynamics simulations based on density functional theory (DFT) suggest that the NIR laser irradiation induces a photothermally activated reaction on the surface of AMNP in water, which can lead to surface degradation via the formation of Sb-H bonds first and then Sb-OH bonds upon further increase of temperature. This work demonstrates a simple platform that has potential applications for synergistic and highly effective chemo-photothermal therapy based on photodegradable nanoparticles.
Collapse
Affiliation(s)
| | | | | | - Jin Z Zhang
- Department of Chemistry and Biochemistry , University of California , Santa Cruz , California 95064 , United States
| | | | | | - Jianhua Zhang
- Key Laboratory of Advanced Display and System Applications of Ministry of Education , Shanghai University , Shanghai 200072 , China
| | | | | |
Collapse
|
45
|
Li L, Tian J, Wang X, Xu G, Jiang W, Yang Z, Liu D, Lin G. Cardiotoxicity of Intravenously Administered CdSe/ZnS Quantum Dots in BALB/c Mice. Front Pharmacol 2019; 10:1179. [PMID: 31649542 PMCID: PMC6791919 DOI: 10.3389/fphar.2019.01179] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 09/13/2019] [Indexed: 12/14/2022] Open
Abstract
Since CdSe quantum dots (QDs) are increasingly used in electronics, medical, and pharmaceutical science due to their excellent optical properties, it is necessary to carry out thorough and systematic studies on their biosafety. Numerous studies have reported the toxicity of QDs on liver, kidney, immune system, and reproductive system. However, few studies have been done on the cardiotoxicity of QDs. In this study, we administered carboxylated CdSe/ZnS QDs in BALB/c mice via the tail vein and analyzed the in vivo cardiotoxicity of CdSe/ZnS QDs. The body weight, hematology, serum biochemistry, histology, heart elements concentration, echocardiography, and heart oxidative stress markers were carried out at different time. There were no significant differences in body weight and heart organ index between QDs group and the control group. Hematology results showed the platelet (PLT) counts on Day 1 and Day 42 in both high dose QDs group and low dose QDs group, and the PLT counts on Day1 in the high dose group were significantly higher than that in control group. Serum biochemistry results showed that lactate dehydrogenase (LDH), creatine kinase (CK), and creatine kinase isoenzyme (CK-MB) of mice exposed to CdSe/ZnS QDs were significantly higher than that of the control group on Day 1, and CK-MB levels still remained high on Day 7. A higher concentration of Cd was observed in the heart of CdSe/ZnS QDs exposed mice on Day 42, whereas no Cd was detected in the control group, which suggested that QDs can accumulate in heart. No significant histopathological changes and cardiac function were observed in all mice at different time after treatment. Increased level of glutathione peroxidase (GPx) and malondialdehyde (MDA) was observed in mice administered with high dose QDs on Day 1, and increased level of total antioxidant capacity (T-AOC) and MDA activities was observed on Day 42. These results indicated that CdSe/ZnS QDs could accumulate in heart, cause some biochemical indicators change, induce oxidative damage, and have cardiotoxicity. Our findings might provide valuable information on the biological safety evaluation of the cardiovascular system of QDs.
Collapse
Affiliation(s)
- Li Li
- Department of Physiology, School of Basic Medical Sciences, Health Sciences Center, Shenzhen University, Shenzhen, China.,Key Laboratory of Optoelectronics Devices and Systems of Ministry of Education/Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, China
| | - Jinglin Tian
- Department of Physiology, School of Basic Medical Sciences, Health Sciences Center, Shenzhen University, Shenzhen, China
| | - Xiaomei Wang
- Department of Physiology, School of Basic Medical Sciences, Health Sciences Center, Shenzhen University, Shenzhen, China
| | - Gaixia Xu
- Key Laboratory of Optoelectronics Devices and Systems of Ministry of Education/Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, China.,National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, China
| | - Wenxiao Jiang
- Department of Physiology, School of Basic Medical Sciences, Health Sciences Center, Shenzhen University, Shenzhen, China
| | - Zhiwen Yang
- Department of Physiology, School of Basic Medical Sciences, Health Sciences Center, Shenzhen University, Shenzhen, China
| | - Dongmeng Liu
- Department of Physiology, School of Basic Medical Sciences, Health Sciences Center, Shenzhen University, Shenzhen, China
| | - Guimiao Lin
- Department of Physiology, School of Basic Medical Sciences, Health Sciences Center, Shenzhen University, Shenzhen, China
| |
Collapse
|
46
|
Slabu I, Roeth AA, Engelmann UM, Wiekhorst F, Buhl EM, Neumann UP, Schmitz-Rode T. Modeling of magnetoliposome uptake in human pancreatic tumor cells in vitro. NANOTECHNOLOGY 2019; 30:184004. [PMID: 30699387 DOI: 10.1088/1361-6528/ab033e] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
The internalization kinetics resulting from magnetic nanoparticle interactions with tumor cells play an important role in nanoparticle-based cancer treatment efficiency. Here, the uptake kinetics of magnetoliposomes (ML) into human pancreatic tumor cells (MiaPaCa-2 and BxPC-3) are quantified using magnetic particle spectrometry. A comparison to the uptake kinetics for healthy L929 cells is given. The experimental results are used for the development of an uptake kinetics model describing the three relevant internalization processes: ML adsorption to the cell membrane, endo- and exocytosis. By fitting of experimental data, the rate constant of each internalization process is determined enabling the prediction of internalized ML at any incubation time. After seven hours incubation time, MiaPaCa-2 internalized three times more ML than BxPC-3 and L929 cells even though their ML adsorption rate constants were nearly the same. As the interaction of the ML with the cell membrane is non-specific, the uptake kinetics mirror the individual cell response to ML internalization. With a new mathematical term to cover the exocytosis contribution to the overall internalization process, the extended uptake kinetics model offers new possibilities to analyze the specific internalization mechanism for other nanoparticle and cell types.
Collapse
Affiliation(s)
- Ioana Slabu
- Institute of Applied Medical Engineering, RWTH Aachen University and University Hospital, Aachen, Germany. Physikalisch-Technische Bundesanstalt, Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
47
|
Wang S, Huang R. Non-viral nucleic acid delivery to the central nervous system and brain tumors. J Gene Med 2019; 21:e3091. [PMID: 30980444 DOI: 10.1002/jgm.3091] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/09/2019] [Accepted: 04/09/2019] [Indexed: 12/20/2022] Open
Abstract
Gene therapy is a rapidly emerging remedial route for many serious incurable diseases, such as central nervous system (CNS) diseases. Currently, nucleic acid medicines, including DNAs encoding therapeutic or destructive proteins, small interfering RNAs or microRNAs, have been successfully delivered to the CNS with gene delivery vectors using various routes of administration and have subsequently exhibited remarkable therapeutic efficiency. Among these vectors, non-viral vectors are favorable for delivering genes into the CNS as a result of their many special characteristics, such as low toxicity and pre-existing immunogenicity, high gene loading efficiency and easy surface modification. In this review, we highlight the main types of therapeutic genes that have been applied in the therapy of CNS diseases and then outline non-viral gene delivery vectors.
Collapse
Affiliation(s)
- Shanshan Wang
- Department of Pharmacy, Zhongshan Hospital, and School of Pharmacy, Fudan University, Shanghai, China
| | - Rongqin Huang
- Department of Pharmacy, Zhongshan Hospital, and School of Pharmacy, Fudan University, Shanghai, China
| |
Collapse
|
48
|
Aghamiri S, Jafarpour A, Gomari MM, Ghorbani J, Rajabibazl M, Payandeh Z. siRNA nanotherapeutics: a promising strategy for anti‐HBV therapy. IET Nanobiotechnol 2019; 13:457-463. [PMCID: PMC8676379 DOI: 10.1049/iet-nbt.2018.5286] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 11/18/2018] [Accepted: 01/28/2019] [Indexed: 07/31/2023] Open
Abstract
Chronic hepatitis B (CHB) is the most common cause of hepatocellular carcinoma (HCC) and liver cirrhosis worldwide. In spite of the numerous advances in the treatment of CHB, drugs and vaccines have failed because of many factors like complexity, resistance, toxicity, and heavy cost. New RNA interference (RNAi)‐based technologies have developed innovative strategies to target Achilles' heel of the several hazardous diseases involving cancer, some genetic disease, autoimmune illnesses, and viral disorders particularly hepatitis B virus (HBV) infections. Naked siRNA delivery has serious challenges including failure to cross the cell membrane, susceptibility to the enzymatic digestion, and excretion by renal filtration, which ideally can be addressed by nanoparticle‐mediated delivery systems. cccDNA formation is a significant problem in obtaining HBV infections complete cure because of strength, durability, and lack of proper immune response. Nano‐siRNA drugs have a great potential to address this problem by silencing specific genes which are involved in cccDNA formation. In this article, the authors describe siRNA nanocarrier‐mediated delivery systems as a promising new strategy for HBV infections therapy. Simultaneously, the authors completely represent the clinical trials which use these strategies for treatment of the HBV infections.
Collapse
Affiliation(s)
- Shahin Aghamiri
- Student research committeeDepartment of Medical BiotechnologySchool of Advanced Technologies in MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Ali Jafarpour
- Students' Scientific Research CenterVirology DivisionDepartment of PathobiologySchool of Public HealthTehran University of Medical SciencesTehranIran
| | | | - Jaber Ghorbani
- Department of Medical BiotechnologySchool of Advanced Technologies in MedicineTehran University of Medical SciencesTehranIran
| | - Masoumeh Rajabibazl
- Department of Clinical BiochemistryFaculty of MedicineShahid Beheshti University of Medical SciencesTehranIran
- Department of Tissue Engineering and Applied Cell SciencesSchool of Advanced Technologies in MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Zahra Payandeh
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
| |
Collapse
|
49
|
Biliuta G, Coseri S. Cellulose: A ubiquitous platform for ecofriendly metal nanoparticles preparation. Coord Chem Rev 2019. [DOI: 10.1016/j.ccr.2019.01.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
50
|
Liu M, Liu B, Liu Q, Du K, Wang Z, He N. Nanomaterial-induced ferroptosis for cancer specific therapy. Coord Chem Rev 2019. [DOI: 10.1016/j.ccr.2018.12.015] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|