1
|
Lin J, Ma Z, Zuo W, Zhu M. Triple-function porphyrin in glycopolymeric photosensitizers: from photoATRP to targeted PDT. Chem Sci 2024; 15:20388-20396. [PMID: 39583554 PMCID: PMC11579898 DOI: 10.1039/d4sc06466f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 11/09/2024] [Indexed: 11/26/2024] Open
Abstract
Porphyrin derivatives serve as photocatalysts in reversible-deactivation radical polymerization and as photosensitizers in photodynamic therapy (PDT). Herein, a triple function porphyrin, ZnTPPC6Br, was synthesized as a photocatalyst and initiator for photoATRP. Oxygen-tolerant photoATRP produced fructose-based star-shaped glycopolymers as targeted photosensitizers for PDT. ZnTPPC6Br/CuII/PMDETA could synthesize polymer photosensitizers with predictable M n and low Đ. Mechanistic studies unveiled the transition of ZnTPPC6Br from a singlet excited state (1PC*) to a triplet excited state (3PC*), enabling the activator CuI/L generation and initiating photoATRP. The excess ligands facilitate return of the active species to the ground state, while the presence of DMSO assists in oxygen depletion. Three fructose-based monomers with different polymerizable groups (acrylated, methacrylated, and p-vinylbenzoated) were employed to scale up polymerization, yielding glycopolymeric photosensitizers post-deprotection. In vitro cellular studies showed enhanced PDT efficacy of glycopolymeric photosensitizers against MCF-7 cells, attributed to specific GLUT5 binding for targeted endocytosis, highlighting their potential for precise cancer treatment compared to L929 cells. The multifunctional capabilities of ZnTPPC6Br are anticipated to serve as a strategic avenue for the advancement of polymer photosensitizers with potential PDT applications.
Collapse
Affiliation(s)
- Jiahui Lin
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University 2999 North Renmin Road Shanghai 201620 China
| | - Zhiyuan Ma
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University 2999 North Renmin Road Shanghai 201620 China
| | - Weiwei Zuo
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University 2999 North Renmin Road Shanghai 201620 China
| | - Meifang Zhu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University 2999 North Renmin Road Shanghai 201620 China
| |
Collapse
|
2
|
Liu L, Qi G, Wang M, He J, Zheng Y, Guan J, Lv P, Zeng D. Construction of intelligent response gene vector based on MOF/Fe 3O 4/AuNRs for tumor-targeted gene delivery. Int J Biol Macromol 2024; 277:134313. [PMID: 39098672 DOI: 10.1016/j.ijbiomac.2024.134313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/28/2024] [Accepted: 07/28/2024] [Indexed: 08/06/2024]
Abstract
Metal-organic frameworks (MOFs) have the potential to efficiently carry cargo due to their excellent porosity and high surface area. Nevertheless, conventional MOFs and their derivatives exhibit low efficiency in transporting nucleic acids and other small molecules, as well as having poor colloidal stability. In this study, a ZIF-90 loaded with iron oxide nanoparticles and Au nanorods was prepared, and then surface-functionalized with polyethyleneimine (PEI) to create a multifunctional nanocomposite (AFZP25k) with pH, photothermal, and magnetic responsiveness. AFZP25k can condense plasmid DNA to form AFZP25k/DNA complexes, with a maximum binding efficiency of 92.85 %. DNA release assay showed significant light and pH responsiveness, with over 80 % cumulative release after 6 h of incubation. When an external magnetic field is applied, the cellular uptake efficiency in HeLa cells reached 81.51 %, with low cytotoxicity and specific distribution. In vitro transfection experiments demonstrated a gene transfection efficiency of 44.77 % in HeLa cells. Following near-infrared irradiation, the uptake efficiency and transfection efficiency of AFZP25k in HeLa cells increased by 21.3 % and 13.59 % respectively. The findings indicate the potential of AFZP25k as an efficient and targeted gene delivery vector in cancer gene therapy.
Collapse
Affiliation(s)
- Liang Liu
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China.
| | - Guowei Qi
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Mingjie Wang
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Jiayu He
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Yuqiu Zheng
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Jintao Guan
- School of Chemical and Environmental Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Peiwen Lv
- School of Chemical and Environmental Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Dong Zeng
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China.
| |
Collapse
|
3
|
Kachanov A, Kostyusheva A, Brezgin S, Karandashov I, Ponomareva N, Tikhonov A, Lukashev A, Pokrovsky V, Zamyatnin AA, Parodi A, Chulanov V, Kostyushev D. The menace of severe adverse events and deaths associated with viral gene therapy and its potential solution. Med Res Rev 2024; 44:2112-2193. [PMID: 38549260 DOI: 10.1002/med.22036] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 03/05/2024] [Accepted: 03/07/2024] [Indexed: 08/09/2024]
Abstract
Over the past decade, in vivo gene replacement therapy has significantly advanced, resulting in market approval of numerous therapeutics predominantly relying on adeno-associated viral vectors (AAV). While viral vectors have undeniably addressed several critical healthcare challenges, their clinical application has unveiled a range of limitations and safety concerns. This review highlights the emerging challenges in the field of gene therapy. At first, we discuss both the role of biological barriers in viral gene therapy with a focus on AAVs, and review current landscape of in vivo human gene therapy. We delineate advantages and disadvantages of AAVs as gene delivery vehicles, mostly from the safety perspective (hepatotoxicity, cardiotoxicity, neurotoxicity, inflammatory responses etc.), and outline the mechanisms of adverse events in response to AAV. Contribution of every aspect of AAV vectors (genomic structure, capsid proteins) and host responses to injected AAV is considered and substantiated by basic, translational and clinical studies. The updated evaluation of recent AAV clinical trials and current medical experience clearly shows the risks of AAVs that sometimes overshadow the hopes for curing a hereditary disease. At last, a set of established and new molecular and nanotechnology tools and approaches are provided as potential solutions for mitigating or eliminating side effects. The increasing number of severe adverse reactions and, sadly deaths, demands decisive actions to resolve the issue of immune responses and extremely high doses of viral vectors used for gene therapy. In response to these challenges, various strategies are under development, including approaches aimed at augmenting characteristics of viral vectors and others focused on creating secure and efficacious non-viral vectors. This comprehensive review offers an overarching perspective on the present state of gene therapy utilizing both viral and non-viral vectors.
Collapse
Affiliation(s)
- Artyom Kachanov
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
| | - Anastasiya Kostyusheva
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
| | - Sergey Brezgin
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
- Division of Biotechnology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia
| | - Ivan Karandashov
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
| | - Natalia Ponomareva
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
- Division of Biotechnology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia
| | - Andrey Tikhonov
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
| | - Alexander Lukashev
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
| | - Vadim Pokrovsky
- Laboratory of Biochemical Fundamentals of Pharmacology and Cancer Models, Blokhin Cancer Research Center, Moscow, Russia
- Department of Biochemistry, People's Friendship University, Russia (RUDN University), Moscow, Russia
| | - Andrey A Zamyatnin
- Division of Biotechnology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
- Belozersky Research, Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Alessandro Parodi
- Division of Biotechnology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia
| | - Vladimir Chulanov
- Division of Biotechnology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia
- Faculty of Infectious Diseases, Sechenov University, Moscow, Russia
| | - Dmitry Kostyushev
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
- Division of Biotechnology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
4
|
Chen J, Zhang H, Zhao T, Yu Y, Song J, Zhao Y, Alshawwa H, Zou X, Zhang Z. Oxygen Self-Supplied Nanoplatform for Enhanced Photodynamic Therapy against Enterococcus Faecalis within Root Canals. Adv Healthc Mater 2024; 13:e2302926. [PMID: 38273674 DOI: 10.1002/adhm.202302926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 01/10/2024] [Indexed: 01/27/2024]
Abstract
The successful treatment of persistent and recurrent endodontic infections hinges upon the eradication of residual microorganisms within the root canal system, which urgently needs novel drugs to deliver potent yet gentle antimicrobial effects. Antibacterial photodynamic therapy (aPDT) is a promising tool for root canal infection management. Nevertheless, the hypoxic microenvironment within the root canal system significantly limits the efficacy of this treatment. Herein, a nanohybrid drug, Ce6/CaO2/ZIF-8@polyethylenimine (PEI), is developed using a bottom-up strategy to self-supply oxygen for enhanced aPDT. PEI provides a positively charged surface, which enables precise targeting of bacteria. CaO2 reacts with H2O to generate O2, which alleviates the hypoxia in the root canal and serves as a substrate for Ce6 under 660 nm laser irradiation, leading to the successful eradication of planktonic Enterococcus faecalis (E. faecalis) and biofilm in vitro and, moreover, the effective elimination of mature E. faecalis biofilm in situ within the root canal system. This smart design offers a viable alternative for mitigating hypoxia within the root canal system to overcome the restricted efficacy of photosensitizers, providing an exciting prospect for the clinical management of persistent endodontic infection.
Collapse
Affiliation(s)
- Jiawen Chen
- Department of Endodontics, Jilin Provincial Key Laboratory of Oral Biomedical Engineering, School and Hospital of Stomatology, Jilin University, Changchun, 130021, P. R. China
| | - Hong Zhang
- Department of Endodontics, Jilin Provincial Key Laboratory of Oral Biomedical Engineering, School and Hospital of Stomatology, Jilin University, Changchun, 130021, P. R. China
| | - Tiancong Zhao
- College of Chemistry and Materials, Department of Chemistry and Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, State Key Laboratory of Molecular Engineering of Polymers, Collaborative Innovation Center of Chemistry for Energy Materials (2011-iChEM), Fudan University, Shanghai, 200433, P. R. China
| | - Yiyan Yu
- Department of Endodontics, Jilin Provincial Key Laboratory of Oral Biomedical Engineering, School and Hospital of Stomatology, Jilin University, Changchun, 130021, P. R. China
| | - Jiazhuo Song
- Department of Endodontics, Jilin Provincial Key Laboratory of Oral Biomedical Engineering, School and Hospital of Stomatology, Jilin University, Changchun, 130021, P. R. China
| | - Yuanhang Zhao
- Department of Endodontics, Jilin Provincial Key Laboratory of Oral Biomedical Engineering, School and Hospital of Stomatology, Jilin University, Changchun, 130021, P. R. China
| | - Hamed Alshawwa
- Department of Endodontics, Jilin Provincial Key Laboratory of Oral Biomedical Engineering, School and Hospital of Stomatology, Jilin University, Changchun, 130021, P. R. China
| | - Xinying Zou
- Department of Endodontics, Jilin Provincial Key Laboratory of Oral Biomedical Engineering, School and Hospital of Stomatology, Jilin University, Changchun, 130021, P. R. China
| | - Zhimin Zhang
- Department of Endodontics, Jilin Provincial Key Laboratory of Oral Biomedical Engineering, School and Hospital of Stomatology, Jilin University, Changchun, 130021, P. R. China
| |
Collapse
|
5
|
Yue NN, Xu HM, Xu J, Zhu MZ, Zhang Y, Tian CM, Nie YQ, Yao J, Liang YJ, Li DF, Wang LS. Therapeutic potential of gene therapy for gastrointestinal diseases: Advancements and future perspectives. Mol Ther Oncolytics 2023; 30:193-215. [PMID: 37663132 PMCID: PMC10471515 DOI: 10.1016/j.omto.2023.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023] Open
Abstract
Advancements in understanding the pathogenesis mechanisms underlying gastrointestinal diseases, encompassing inflammatory bowel disease, gastrointestinal cancer, and gastroesophageal reflux disease, have led to the identification of numerous novel therapeutic targets. These discoveries have opened up exciting possibilities for developing gene therapy strategies to treat gastrointestinal diseases. These strategies include gene replacement, gene enhancement, gene overexpression, gene function blocking, and transgenic somatic cell transplantation. In this review, we introduce the important gene therapy targets and targeted delivery systems within the field of gastroenterology. Furthermore, we provide a comprehensive overview of recent progress in gene therapy related to gastrointestinal disorders and shed light on the application of innovative gene-editing technologies in treating these conditions. These developments are fueling a revolution in the management of gastrointestinal diseases. Ultimately, we discuss the current challenges (particularly regarding safety, oral efficacy, and cost) and explore potential future directions for implementing gene therapy in the clinical settings for gastrointestinal diseases.
Collapse
Affiliation(s)
- Ning-ning Yue
- Department of Gastroenterology, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University), Shenzhen 518000, China
| | - Hao-ming Xu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou 510000, China
| | - Jing Xu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou 510000, China
| | - Min-zheng Zhu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510000, China
| | - Yuan Zhang
- Department of Medical Administration, Huizhou Institute of Occupational Diseases Control and Prevention, Huizhou, Guangdong 516000, China
| | - Cheng-Mei Tian
- Department of Emergency, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University, the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518000, China
| | - Yu-qiang Nie
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou 510000, China
| | - Jun Yao
- Department of Gastroenterology, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University, the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518000, China
| | - Yu-jie Liang
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, Shenzhen 518000, China
| | - De-feng Li
- Department of Gastroenterology, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University, the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518000, China
| | - Li-sheng Wang
- Department of Gastroenterology, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University, the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518000, China
| |
Collapse
|
6
|
Liu S, Wu J, Feng Y, Guo X, Li T, Meng M, Chen J, Chen D, Tian H. CD47KO/CRT dual-bioengineered cell membrane-coated nanovaccine combined with anti-PD-L1 antibody for boosting tumor immunotherapy. Bioact Mater 2023; 22:211-224. [PMID: 36246666 PMCID: PMC9535270 DOI: 10.1016/j.bioactmat.2022.09.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 12/02/2022] Open
Abstract
Tumor vaccines trigger tumor-specific immune responses to prevent or treat tumors by activating the hosts' immune systems, and therefore, these vaccines have potential clinical applications. However, the low immunogenicity of the tumor antigen itself and the low efficiency of the vaccine delivery system hinder the efficacy of tumor vaccines that cannot produce high-efficiency and long-lasting antitumor immune effects. Here, we constructed a nanovaccine by integrating CD47KO/CRT dual-bioengineered B16F10 cancer cell membranes and the unmethylated cytosine-phosphate-guanine (CpG) adjuvant. Hyperbranched PEI25k was used to load unmethylated cytosine-phosphate-guanine (CpG) through electrostatic adsorption to prepare PEI25k/CpG nanoparticles (PEI25k/CpG-NPs). CD47KO/CRT dual-bioengineered cells were obtained by CRISPR-Cas9 gene editing technology, followed by the cell surface translocation of calreticulin (CRT) to induce immunogenic cell death (ICD) in vitro. Finally, the extracted cell membranes were coextruded with PEI25k/CpG-NPs to construct the CD47KO/CRT dual-bioengineered cancer cell membrane-coated nanoparticles (DBE@CCNPs). DBE@CCNPs could promote endocytosis of antigens and adjuvants in murine bone marrow derived dendritic cells (BMDCs) and induce their maturation and antigen cross-presentation. To avoid immune checkpoint molecule-induced T cell dysfunction, the immune checkpoint inhibitor, the anti-PD-L1 antibody, was introduced to boost tumor immunotherapy through a combination with the DBE@CCNPs nanovaccine. This combination therapy strategy can significantly alleviate tumor growth and may open up a potential strategy for clinical tumor immunotherapy.
Collapse
Affiliation(s)
- Shengyang Liu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, PR China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China
| | - Jiayan Wu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China
| | - Yuanji Feng
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China
| | - Xiaoya Guo
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China
- College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, PR China
| | - Tong Li
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China
- College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, PR China
| | - Meng Meng
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China
- College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, PR China
| | - Jie Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China
| | - Daquan Chen
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, PR China
| | - Huayu Tian
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China
- College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, PR China
| |
Collapse
|
7
|
Shtykalova S, Deviatkin D, Freund S, Egorova A, Kiselev A. Non-Viral Carriers for Nucleic Acids Delivery: Fundamentals and Current Applications. Life (Basel) 2023; 13:903. [PMID: 37109432 PMCID: PMC10142071 DOI: 10.3390/life13040903] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/21/2023] [Accepted: 03/24/2023] [Indexed: 03/31/2023] Open
Abstract
Over the past decades, non-viral DNA and RNA delivery systems have been intensively studied as an alternative to viral vectors. Despite the most significant advantage over viruses, such as the lack of immunogenicity and cytotoxicity, the widespread use of non-viral carriers in clinical practice is still limited due to the insufficient efficacy associated with the difficulties of overcoming extracellular and intracellular barriers. Overcoming barriers by non-viral carriers is facilitated by their chemical structure, surface charge, as well as developed modifications. Currently, there are many different forms of non-viral carriers for various applications. This review aimed to summarize recent developments based on the essential requirements for non-viral carriers for gene therapy.
Collapse
Affiliation(s)
- Sofia Shtykalova
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint-Petersburg, Russia
- Faculty of Biology, Saint-Petersburg State University, Universitetskaya Embankment 7-9, 199034 Saint-Petersburg, Russia
| | - Dmitriy Deviatkin
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint-Petersburg, Russia
- Faculty of Biology, Saint-Petersburg State University, Universitetskaya Embankment 7-9, 199034 Saint-Petersburg, Russia
| | - Svetlana Freund
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint-Petersburg, Russia
- Faculty of Biology, Saint-Petersburg State University, Universitetskaya Embankment 7-9, 199034 Saint-Petersburg, Russia
| | - Anna Egorova
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint-Petersburg, Russia
| | - Anton Kiselev
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint-Petersburg, Russia
| |
Collapse
|
8
|
Santo D, Mendonça PV, Serra AC, Coelho JFJ, Faneca H. Targeted downregulation of MYC mediated by a highly efficient lactobionic acid-based glycoplex to enhance chemosensitivity in human hepatocellular carcinoma cells. Int J Pharm 2023; 637:122865. [PMID: 36940837 DOI: 10.1016/j.ijpharm.2023.122865] [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: 10/15/2022] [Revised: 01/31/2023] [Accepted: 03/14/2023] [Indexed: 03/23/2023]
Abstract
The chemosensitization of tumor cells by gene therapy represents a promising strategy for hepatocellular carcinoma (HCC) treatment. In this regard, HCC-specific and highly efficient gene delivery nanocarriers are urgently needed. For this purpose, novel lactobionic acid-based gene delivery nanosystems were developed to downregulate c-MYC expression and sensitize tumor cells to low concentration of sorafenib (SF). A library of tailor-made cationic glycopolymers, based on poly(2-aminoethyl methacrylate hydrochloride) (PAMA) and poly(2-lactobionamidoethyl methacrylate) (PLAMA) were synthesized by a straightforward activators regenerated by electron transfer atom transfer radical polymerization. The nanocarriers prepared with PAMA114-co-PLAMA20 glycopolymer were the most efficient for gene delivery. These glycoplexes specifically bound to the asialoglycoprotein receptor and were internalized through the clathrin-coated pit endocytic pathway. c-MYC expression was significantly downregulated by MYC short-hairpin RNA (MYC shRNA), resulting in efficient inhibition of tumor cells proliferation and a high levels apoptosis in 2D and 3D HCC-tumor models. Moreover, c-MYC silencing increased the sensitivity of HCC cells to SF (IC50 for MYC shRNA+ SF 1.9 μM compared to 6.9 μM for control shRNA + SF). Overall, the data obtained demonstrated the great potential of PAMA114-co-PLAMA20/MYC shRNA nanosystems combined with low doses of SF for the treatment of HCC.
Collapse
Affiliation(s)
- Daniela Santo
- University of Coimbra, Center for Neuroscience and Cell Biology, Coimbra, Portugal; University of Coimbra, Institute for Interdisciplinary Research, Coimbra, Portugal
| | - Patrícia V Mendonça
- University of Coimbra, Centre for Mechanical Engineering, Materials and Processes, Department of Chemical Engineering, Coimbra, Portugal
| | - Arménio C Serra
- University of Coimbra, Centre for Mechanical Engineering, Materials and Processes, Department of Chemical Engineering, Coimbra, Portugal
| | - Jorge F J Coelho
- University of Coimbra, Centre for Mechanical Engineering, Materials and Processes, Department of Chemical Engineering, Coimbra, Portugal; IPN, Instituto Pedro Nunes, Associação para a Inovação e Desenvolvimento em Ciência e Tecnologia, Rua Pedro Nunes, 3030-199 Coimbra, Portugal
| | - Henrique Faneca
- University of Coimbra, Center for Neuroscience and Cell Biology, Coimbra, Portugal; University of Coimbra, Institute for Interdisciplinary Research, Coimbra, Portugal.
| |
Collapse
|
9
|
Santo D, Cordeiro RA, Mendonça P, Serra A, Coelho JFJ, Faneca H. Glycopolymers Mediate Suicide Gene Therapy in ASGPR-Expressing Hepatocellular Carcinoma Cells in Tandem with Docetaxel. Biomacromolecules 2023; 24:1274-1286. [PMID: 36780314 PMCID: PMC10015461 DOI: 10.1021/acs.biomac.2c01329] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
Cationic glycopolymers stand out as gene delivery nanosystems due to their inherent biocompatibility and high binding affinity to the asialoglycoprotein receptor (ASGPR), a target receptor overexpressed in hepatocellular carcinoma (HCC) cells. However, their synthesis procedure remains laborious and complex, with problems of solubilization and the need for protection/deprotection steps. Here, a mini-library of well-defined poly(2-aminoethyl methacrylate hydrochloride-co-poly(2-lactobionamidoethyl methacrylate) (PAMA-co-PLAMA) glycopolymers was synthesized by activators regenerated by electron transfer (ARGET) ATRP to develop an efficient gene delivery nanosystem. The glycoplexes generated had suitable physicochemical properties and showed high ASGPR specificity and high transfection efficiency. Moreover, the HSV-TK/GCV suicide gene therapy strategy, mediated by PAMA144-co-PLAMA19-based nanocarriers, resulted in high antitumor activity in 2D and 3D culture models of HCC, which was significantly enhanced by the combination with small amounts of docetaxel. Overall, our results demonstrated the potential of primary-amine polymethacrylate-containing-glycopolymers as HCC-targeted suicide gene delivery nanosystems and highlight the importance of combined strategies for HCC treatment.
Collapse
Affiliation(s)
- Daniela Santo
- Center
for Neuroscience and Cell Biology, University
of Coimbra, Coimbra 3004-504, Portugal
- Institute
for Interdisciplinary Research, University
of Coimbra, Coimbra 3030-789, Portugal
| | - Rosemeyre A. Cordeiro
- Center
for Neuroscience and Cell Biology, University
of Coimbra, Coimbra 3004-504, Portugal
- Institute
for Interdisciplinary Research, University
of Coimbra, Coimbra 3030-789, Portugal
| | - Patrícia
V. Mendonça
- Centre
for Mechanical Engineering, Materials and Processes, Department of
Chemical Engineering, University of Coimbra, Coimbra 3030-790, Portugal
| | - Arménio
C. Serra
- Centre
for Mechanical Engineering, Materials and Processes, Department of
Chemical Engineering, University of Coimbra, Coimbra 3030-790, Portugal
| | - Jorge F. J. Coelho
- Centre
for Mechanical Engineering, Materials and Processes, Department of
Chemical Engineering, University of Coimbra, Coimbra 3030-790, Portugal
- Associação
para a Inovação e Desenvolvimento Em Ciência
e Tecnologia, IPN—Instituto Pedro
Nunes, Rua Pedro Nunes, 3030-199 Coimbra, Portugal
| | - Henrique Faneca
- Center
for Neuroscience and Cell Biology, University
of Coimbra, Coimbra 3004-504, Portugal
- Institute
for Interdisciplinary Research, University
of Coimbra, Coimbra 3030-789, Portugal
- . Phone: +351-239-820-190. Fax: +351- 239-853-607
| |
Collapse
|
10
|
Mishra AK, Pandey M, Dewangan HK, Sl N, Sahoo PK. A Comprehensive Review on Liver Targeting: Emphasis on Nanotechnology- based Molecular Targets and Receptors Mediated Approaches. Curr Drug Targets 2022; 23:1381-1405. [PMID: 36065923 DOI: 10.2174/1389450123666220906091432] [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: 11/10/2021] [Revised: 02/10/2022] [Accepted: 02/25/2022] [Indexed: 01/25/2023]
Abstract
BACKGROUND The pathogenesis of hepatic diseases involves several cells, which complicates the delivery of pharmaceutical agents. Many severe liver diseases affecting the worldwide population cannot be effectively treated. Major hindrances or challenges are natural physiological barriers and non-specific targeting of drugs administered, leading to inefficient treatment. Hence, there is an earnest need to look for novel therapeutic strategies to overcome these hindrances. A kind of literature has reported that drug safety and efficacy are incredibly raised when a drug is incorporated inside or attached to a polymeric material of either hydrophilic or lipophilic nature. This has driven the dynamic investigation for developing novel biodegradable materials, drug delivery carriers, target-specific drug delivery systems, and many other novel approaches. OBJECTIVE Present review is devoted to summarizing receptor-based liver cell targeting using different modified novel synthetic drug delivery carriers. It also highlights recent progress in drug targeting to diseased liver mediated by various receptors, including asialoglycoprotein, mannose and galactose receptor, Fc receptor, low-density lipoprotein, glycyrrhetinic, and bile acid receptor. The essential consideration is given to treating liver cancer targeting using nanoparticulate systems, proteins, viral and non-viral vectors, homing peptides and gene delivery. CONCLUSION Receptors based targeting approach is one such approach that was explored by researchers to develop novel formulations which can ensure site-specific drug delivery. Several receptors are on the surfaces of liver cells, which are highly overexpressed in various disease conditions. They all are helpful for the treatment of liver cancer.
Collapse
Affiliation(s)
- Ashwini Kumar Mishra
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Sciences and Research, Sector 3, MB Road Pushp Vihar, Delhi 110017, India
| | - Mukesh Pandey
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Sciences and Research, Sector 3, MB Road Pushp Vihar, Delhi 110017, India
| | - Hitesh Kumar Dewangan
- University Institute of Pharma Sciences (UIPS), Chandigarh University NH-05, Chandigarh Ludhiana Highway, Mohali Punjab, Pin: 160101, India
| | - Neha Sl
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Sciences and Research, Sector 3, MB Road Pushp Vihar, Delhi 110017, India
| | - Pravat Kumar Sahoo
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Sciences and Research, Sector 3, MB Road Pushp Vihar, Delhi 110017, India
| |
Collapse
|
11
|
Abstract
INTRODUCTION Gene delivery vectors are a crucial determinant for gene therapeutic efficacy. Usually, it is necessary to use an excess of cationic vectors to achieve better transfection efficiency. However, it will cause severe cytotoxicity. In addition, cationic vectors are not resistant to serum, suffering from reduced transfection efficiency by forming large aggregates. Therefore, there is an urgent need to develop optimized gene delivery vectors. Recently, fluorination of vectors has been extensively applied to increase the gene delivery performance because of the unique properties of both hydrophobicity and lipophobicity, and chemical and biological inertness. AREAS COVERED This review will discuss the fluorophilic effects that impact gene delivery efficiency, and chemical modification approaches for fluorination. Next, recent advances and applications of fluorinated polymeric and lipidic vectors in gene therapy and gene editing are summarized. EXPERT OPINION Fluorinated vectors are a promising candidate for gene delivery. However, it still needs further studies to obtain pure and well-defined fluorinated polymers, guarantee the biosafety, and clarify the detailed mechanism. Apart from the improvements in gene delivery, exploiting other versatility of fluorinated vectors, such as oxygen-carrying ability, high affinity with fluorine-containing drugs, and imaging property upon introducing 19F, will further facilitate their applications in gene therapy.
Collapse
Affiliation(s)
- Yu Wan
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Yuhan Yang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Mingyu Wu
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Shun Feng
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| |
Collapse
|
12
|
Thalji MR, Ibrahim AA, Chong KF, Soldatov AV, Ali GAM. Glycopolymer-Based Materials: Synthesis, Properties, and Biosensing Applications. Top Curr Chem (Cham) 2022; 380:45. [PMID: 35951265 PMCID: PMC9366760 DOI: 10.1007/s41061-022-00395-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 06/02/2022] [Indexed: 11/30/2022]
Abstract
Glycopolymer materials have emerged as a significant biopolymer class that has piqued the scientific community's attention due to their potential applications. Recently, they have been found to be a unique synthetic biomaterial; glycopolymer materials have also been used for various applications, including direct therapeutic methods, medical adhesives, drug/gene delivery systems, and biosensor applications. Therefore, for the next stage of biomaterial research, it is essential to understand current breakthroughs in glycopolymer-based materials research. This review discusses the most widely utilized synthetic methodologies for glycopolymer-based materials, their properties based on structure-function interactions, and the significance of these materials in biosensing applications, among other topics. When creating glycopolymer materials, contemporary polymerization methods allow precise control over molecular weight, molecular weight distribution, chemical activity, and polymer architecture. This review concludes with a discussion of the challenges and complexities of glycopolymer-based biosensors, in addition to their potential applications in the future.
Collapse
Affiliation(s)
- Mohammad R. Thalji
- School of Chemical Engineering, Yeungnam University, Gyeongsan, 38541 Gyeongbuk South Korea
| | - Amal Amin Ibrahim
- Polymers and pigments department, Chemical industries research institute, National Research Centre, El-Bohouth St, Dokki, Cairo, 12622 Egypt
| | - Kwok Feng Chong
- Faculty of Industrial Sciences and Technology, Universiti Malaysia Pahang, Gambang, 26300 Kuantan, Malaysia
| | - Alexander V. Soldatov
- The Smart Materials Research Institute, Southern Federal University, Sladkova Str. 178/24, Rostov-on-Don, Russian Federation
| | - Gomaa A. M. Ali
- Chemistry Department, Faculty of Science, Al-Azhar University, Assiut, 71524 Egypt
| |
Collapse
|
13
|
Yan Y, Zhang G, Wu C, Ren Q, Liu X, Huang F, Cao Y, Ye W. Structural Exploration of Polycationic Nanoparticles for siRNA Delivery. ACS Biomater Sci Eng 2022; 8:1964-1974. [PMID: 35380797 DOI: 10.1021/acsbiomaterials.2c00196] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
RNA interference (RNAi) is a promising approach to the treatment of genetic diseases by the specific knockdown of target genes. Functional polymers are potential vehicles for the effective delivery of vulnerable small interfering RNA (siRNA), which is required for the broad application of RNAi-based therapeutics. The development of methods for the facile modulation of chemical structures of polymeric carriers and an elucidation of detailed delivery mechanisms remain important areas of research. In this paper, we synthesized a series of methacrylate-based polymers with controllable structures and narrow distributions by atom transfer radical polymerization using various combinations of cationic monomers (2-dimethylaminoethyl methacrylate, 2-diethylaminoethyl methacrylate, and 2-dibutylaminoethyl methacrylate) and hydrophobic monomers (2-butyl methacrylate (BMA), cyclohexyl methacrylate, and 2-ethylhexyl methacrylate). These polymers exhibited varying hydrophobicities, charge densities, and pKa values, enabling the discovery of effective carriers for siRNA by in vitro delivery assays. For the polymers with BMA segments, 50% of cationic segments were beneficial to the formation of siRNA nanoparticles (NPs) and the in vitro delivery of siRNA. The optimal ratio varied for different combinations of cationic and hydrophobic segments. In particular, 20k PMB 0.5, PME 0.5, and PEB 1.0 showed >75% luciferase knockdown. Efficacious delivery was dependent on high siRNA binding, the small size of NPs, and balanced hydrophobicity and charge density. Cellular uptake and endosomal escape experiments indicated that carboxybetaine modification of 20k PMB 0.5 did not remarkably affect the internalization of corresponding NPs after incubation for 6 h but significantly reduced the endosomal escape of NPs, which leads to the notable decrease in delivery efficacy of polymers. These results provide insights into the mechanism of polymer-based siRNA delivery and may inspire the development of novel polymeric carriers.
Collapse
Affiliation(s)
- Yunfeng Yan
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Guangliang Zhang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Chengfan Wu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Qidi Ren
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Xiaomin Liu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Fangqian Huang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Yi Cao
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Wenbo Ye
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| |
Collapse
|
14
|
Lin M, Lin A, Huang S, Liu T, Ke F, Qiu D, Lin X, Luo D. Development of a novel vector for
siRNA
Delivery based on Arginine Modified Polyvinylamine. POLYM INT 2022. [DOI: 10.1002/pi.6386] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Mei Lin
- Department of Medicinal Chemistry, School of Pharmacy Fujian Medical University Fuzhou China
| | - Aizhu Lin
- Key Laboratory of Technical Evaluation of Fertility Regulation of Non Human Primate National Health Commission Fuzhou 350013 Fujian China
- Fujian Obstetrics and Gynecology Hospital Fuzhou 350012 Fujian China
| | - Sheng Huang
- Department of Medicinal Chemistry, School of Pharmacy Fujian Medical University Fuzhou China
| | - Tianhui Liu
- Department of Medicinal Chemistry, School of Pharmacy Fujian Medical University Fuzhou China
| | - Fang Ke
- Department of Medicinal Chemistry, School of Pharmacy Fujian Medical University Fuzhou China
| | - Deshun Qiu
- Department of Medicinal Chemistry, School of Pharmacy Fujian Medical University Fuzhou China
| | - Xuefeng Lin
- Department of Medicinal Chemistry, School of Pharmacy Fujian Medical University Fuzhou China
| | - Daoshu Luo
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences Fujian Medical University Fuzhou China
| |
Collapse
|
15
|
Wang Y, Xu X, Chen X, Li J. Multifunctional Biomedical Materials Derived from Biological Membranes. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 34:e2107406. [PMID: 34739155 DOI: 10.1002/adma.202107406] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/24/2021] [Indexed: 02/06/2023]
Abstract
The delicate structure and fantastic functions of biological membranes are the successful evolutionary results of a long-term natural selection process. Their excellent biocompatibility and biofunctionality are widely utilized to construct multifunctional biomedical materials mainly by directly camouflaging materials with single or mixed biological membranes, decorating or incorporating materials with membrane-derived vesicles (e.g., exosomes), and designing multifunctional materials with the structure/functions of biological membranes. Here, the structure-function relationship of some important biological membranes and biomimetic membranes are discussed, such as various cell membranes, extracellular vesicles, and membranes from bacteria and organelles. Selected literature examples of multifunctional biomaterials derived from biological membranes for biomedical applications, such as drug- and gene-delivery systems, tissue-repair scaffolds, bioimaging, biosensors, and biological detection, are also highlighted. These designed materials show excellent properties, such as long circulation time, disease-targeted therapy, excellent biocompatibility, and selective recognition. Finally, perspectives and challenges associated with the clinical applications of biological-membrane-derived materials are discussed.
Collapse
Affiliation(s)
- Yuemin Wang
- College of Polymer Science and Engineering State Key Laboratory of Polymer Materials Engineering Sichuan University Chengdu 610065 P. R. China
| | - Xinyuan Xu
- College of Polymer Science and Engineering State Key Laboratory of Polymer Materials Engineering Sichuan University Chengdu 610065 P. R. China
| | - Xingyu Chen
- College of Polymer Science and Engineering State Key Laboratory of Polymer Materials Engineering Sichuan University Chengdu 610065 P. R. China
- College of Medicine Southwest Jiaotong University Chengdu 610003 China
| | - Jianshu Li
- College of Polymer Science and Engineering State Key Laboratory of Polymer Materials Engineering Sichuan University Chengdu 610065 P. R. China
- State Key Laboratory of Oral Diseases West China Hospital of Stomatology Med‐X Center for Materials Sichuan University Chengdu 610041 China
| |
Collapse
|
16
|
Liu L, Chen Y, Liu C, Yan Y, Yang Z, Chen X, Liu G. Effect of Extracellular Matrix Coating on Cancer Cell Membrane-Encapsulated Polyethyleneimine/DNA Complexes for Efficient and Targeted DNA Delivery In Vitro. Mol Pharm 2021; 18:2803-2822. [PMID: 34086466 DOI: 10.1021/acs.molpharmaceut.1c00359] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Polyethyleneimine (PEI) has a good spongy proton effect and is an excellent nonviral gene vector, but its high charge density leads to the instability and toxicity of PEI/DNA complexes. Cell membrane (CM) capsules provide a universal and natural solution for this problem. Here, CM-coated PEI/DNA capsules (CPDcs) were prepared through extrusion, and the extracellular matrix was coated on CPDcs (ECM-CPDcs) for improved targeting. The results showed that compared with PEI/DNA complexes, CPDcs had core-shell structures (PEI/DNA complexes were coated by a 6-10 nm layer), lower cytotoxicity, and obvious homologous targeting. The internalization and transfection efficiency of 293T-CM-coated PEI70k/DNA capsules (293T-CP70Dcs) were 91.8 and 74.5%, respectively, which were higher than those of PEI70k/DNA complexes. Then, the internalization and transfection efficiency of 293T-CP70Dcs were further improved by ECM coating, which were 94.7 and 78.9%, respectively. Then, the internalization and transfection efficiency of 293T-CP70Dcs were further improved by ECM coating, which were 94.7 and 78.9%, respectively. Moreover, the homologous targeting of various CPDcs was improved by ECM coating, and other CPDcs also showed similar effects as 293T-CP70Dcs after ECM coating. These findings suggest that tumor-targeted CPDcs may have considerable advantages in gene delivery.
Collapse
Affiliation(s)
- Liang Liu
- College of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Yiran Chen
- College of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Chaobing Liu
- College of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Yujian Yan
- College of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Zhaojun Yang
- College of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Xin Chen
- College of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Gang Liu
- School of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| |
Collapse
|
17
|
Luo MX, Hua S, Shang QY. Application of nanotechnology in drug delivery systems for respiratory diseases (Review). Mol Med Rep 2021; 23:325. [PMID: 33760125 PMCID: PMC7974419 DOI: 10.3892/mmr.2021.11964] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 01/28/2021] [Indexed: 12/17/2022] Open
Abstract
Respiratory disease is a common disease with a high incidence worldwide, which is a serious threat to human health, and is considered a societal and economic burden. The application of nanotechnology in drug delivery systems has created new treatments for respiratory diseases. Within this context, the present review systematically introduced the physicochemical properties of nanoparticles (NPs); reviewed the current research status of different nanocarriers in the treatment of respiratory diseases, including liposomes, solid lipid nanocarriers, polymeric nanocarriers, dendrimers, inorganic nanocarriers and protein nanocarriers; and discussed the main advantages and limitations of therapeutic nanomedicine in this field. The application of nanotechnology overcomes drug inherent deficiencies to a certain extent, and provides unlimited potential for the development of drugs to treat respiratory diseases. However, most of the related research work is in the preclinical experimental stage and safety assessment is still a challenging task. Future studies are needed to focus on the performance modification, molecular mechanism and potential toxicity of therapeutic nanomedicine.
Collapse
Affiliation(s)
- Ming-Xin Luo
- Department of Respiratory Medicine, Anhui Provincial Children's Hospital, Hefei, Anhui 230000, P.R. China
| | - Shan Hua
- Department of Respiratory Medicine, Anhui Provincial Children's Hospital, Hefei, Anhui 230000, P.R. China
| | - Qi-Yun Shang
- Department of Respiratory Medicine, Anhui Provincial Children's Hospital, Hefei, Anhui 230000, P.R. China
| |
Collapse
|
18
|
Singh R, Kumar P. Disaccharide-polyethylenimine organic nanoparticles as non-toxic in vitro gene transporters and their anticancer potential. Bioorg Chem 2021; 112:104918. [PMID: 33932768 DOI: 10.1016/j.bioorg.2021.104918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 04/01/2021] [Accepted: 04/13/2021] [Indexed: 12/26/2022]
Abstract
Polyethylenimines (PEIs) have been shown as efficient gene delivery vectors due to their unique properties, however, toxicity as well as non-specific interactions with the tissues/cells because of high charge density have hampered their use in clinical applications. To counter these concerns, here, we have prepared disachharide-PEI organic nanoparticles by mixing PEIs with non-reducing disaccharides, i.e. trehalose (TPONs) and sucrose (SPONs), under mild conditions. The fabricated nanoparticles were complexed with pDNA and size of these complexes was found in the range of ~130-162 nm with zeta potential ~ +8-25 mV. Further evaluation of these nanoparticles revealed that substitution of disaccharides on PEIs successfully augmented cell viability. Transfection efficiency exhibited by these complexes was significantly higher than the unmodified polymer and the standard, Lipofectamine, complexes. Fabrication of organic nanoparticles did not alter the buffering capacity considerably which was found to be instrumental during endosomal escape of the complexes. Among both the series of nanoparticles, trehalose-PEI organic nanoparticles (TPONs) exhibited greater pDNA transportation potential than sucrose-PEI organic nanoparticles (SPONs) which was also established by flow cytometric data, wherein percent cells expressing GFP was higher in case of TP/pDNA complexes as compared to SP/pDNA complexes. Interestingly, TPONs also showed promising anticancer activity on cancer cell lines i.e. Mg63, MCF-7 and HepG2. Overall, the results advocate promising potential of disaccharide-PEI organic nanoparticles as efficient gene delivery agents which can be used effectively in future gene therapy applications along with anti-cancer competence of TPONs.
Collapse
Affiliation(s)
- Reena Singh
- Nucleic Acids Research Laboratory, CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Pradeep Kumar
- Nucleic Acids Research Laboratory, CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
19
|
Liu R, Zuo R, Hudalla GA. Harnessing molecular recognition for localized drug delivery. Adv Drug Deliv Rev 2021; 170:238-260. [PMID: 33484737 PMCID: PMC8274479 DOI: 10.1016/j.addr.2021.01.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/06/2021] [Accepted: 01/08/2021] [Indexed: 12/18/2022]
Abstract
A grand challenge in drug delivery is providing the right dose, at the right anatomic location, for the right duration of time to maximize therapeutic efficacy while minimizing off-target toxicity and other deleterious side-effects. Two general modalities are receiving broad attention for localized drug delivery. In the first, referred to as "targeted accumulation", drugs or drug carriers are engineered to have targeting moieties that promote their accumulation at a specific tissue site from circulation. In the second, referred to as "local anchoring", drugs or drug carriers are inserted directly into the tissue site of interest where they persist for a specified duration of time. This review surveys recent advances in harnessing molecular recognition between proteins, peptides, nucleic acids, lipids, and carbohydrates to mediate targeted accumulation and local anchoring of drugs and drug carriers.
Collapse
Affiliation(s)
- Renjie Liu
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Ran Zuo
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Gregory A Hudalla
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
20
|
Wang J, Wang D, Zhang Y, Dong J. Synthesis and Biopharmaceutical Applications of Sugar-Based Polymers: New Advances and Future Prospects. ACS Biomater Sci Eng 2021; 7:963-982. [PMID: 33523642 DOI: 10.1021/acsbiomaterials.0c01710] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The rapid rise in research interest in carbohydrate-based polymers is undoubtedly due to the nontoxic nature of such materials in an in vivo environment and the versatile roles that the polymers can play in cellular functions. Such polymers have served as therapeutic tools for drug delivery, including antigens, proteins, and genes, as well as diagnostic devices. Our focus in the first half of this Review is on synthetic methods based on ring-opening polymerization and enzyme-catalyzed polymerization, along with controlled radical polymerization. In the second half of this Review, sugar-based polymers are discussed on the basis of their remarkable success in competitive receptor binding, as multifunctional nanocarriers of targeting inhibitors for cancer treatment, in genome-editing delivery, in immunotherapy based on endogenous antibody recruitment, and in treatment of respiratory diseases, including influenza A. Particular emphasis is put on the synthesis and biopharmaceutical applications of sugar-based polymers published in the most recent 5 years. A noticeable attribute of carbohydrate-based polymers is that the sugar-receptor interactions can be facilitated by the cooperative effect of multiple sugar units. Their diversified topology and structures will drive the development of new synthetic strategies and bring about important applications, including coronavirus-related drug therapy.
Collapse
Affiliation(s)
- Jie Wang
- College of Chemistry and Chemical Engineering, Shaoxing University, 508 Huancheng West Road, Shaoxing, Zhejiang Province 312000, China
| | - Dong Wang
- College of Chemistry and Chemical Engineering, Shaoxing University, 508 Huancheng West Road, Shaoxing, Zhejiang Province 312000, China
| | - Yixian Zhang
- College of Chemistry and Chemical Engineering, Shaoxing University, 508 Huancheng West Road, Shaoxing, Zhejiang Province 312000, China
| | - Jian Dong
- College of Chemistry and Chemical Engineering, Shaoxing University, 508 Huancheng West Road, Shaoxing, Zhejiang Province 312000, China
| |
Collapse
|
21
|
Peng H, Ji W, Zhao R, Lu Z, Yang J, Li Y, Zhang X. pH-sensitive zwitterionic polycarboxybetaine as a potential non-viral vector for small interfering RNA delivery. RSC Adv 2020; 10:45059-45066. [PMID: 35516239 PMCID: PMC9058814 DOI: 10.1039/d0ra09359a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 12/01/2020] [Indexed: 11/21/2022] Open
Abstract
Small interfering RNA (siRNA) has great potential for the treatment of various diseases. However, its intrinsic deficiencies seriously limit its application. Herein, pH-sensitive zwitterionic polymer polycarboxybetaine (PCB) was developed as a non-viral vector for siRNA. The PCB could be protonated in an acidic environment and become positively charged from a cancer site. After protonation, PCB could complex siRNA via electrostatic interaction, and its loading ability was enhanced with a decrease of pH value. Compared with the PEI 10k, PCB50 with a similar molecular weight had comparable siRNA loading ability and lower cytotoxicity. Besides, siRNA loaded by PCB50 could escape from endosomes and reduce the loss of drugs, and based on the excellent uptake and obvious apoptotic effect on HeLa cells, the pH-sensitive PCB with low cytotoxicity could be used as a non-viral vector for safe siRNA delivery for cancer treatment. pH-sensitive zwitterionic polycarboxybetaine could complex siRNA in an acidic environment and could be used as a non-viral vector for safe siRNA delivery.![]()
Collapse
Affiliation(s)
- Huan Peng
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences Beijing 100190 China .,University of Chinese Academy of Sciences Beijing 100049 China
| | - Weihong Ji
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences Beijing 100190 China .,University of Chinese Academy of Sciences Beijing 100049 China
| | - Ruichen Zhao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences Beijing 100190 China .,University of Chinese Academy of Sciences Beijing 100049 China
| | - Zhiguo Lu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences Beijing 100190 China .,University of Chinese Academy of Sciences Beijing 100049 China
| | - Jun Yang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences Beijing 100190 China
| | - Yan Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences Beijing 100190 China
| | - Xin Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences Beijing 100190 China
| |
Collapse
|
22
|
Wang T, Meng Z, Kang Z, Ding G, Zhao B, Han Z, Zheng Z, Wang C, Meng Q. Peptide Gene Delivery Vectors for Specific Transfection of Glioma Cells. ACS Biomater Sci Eng 2020; 6:6778-6789. [DOI: 10.1021/acsbiomaterials.0c01336] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Taoran Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P. R. China
| | - Zhao Meng
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P. R. China
| | - Ziyao Kang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P. R. China
| | - Guihua Ding
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P. R. China
| | - Baoquan Zhao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P. R. China
| | - Zhenbin Han
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P. R. China
| | - Zhibing Zheng
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P. R. China
| | - Chenhong Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P. R. China
| | - Qingbin Meng
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P. R. China
- Key Laboratory of Natural Resources and Functional Molecules of the Changbai Mountain, Affiliated Ministry of Education, College of Pharmacy, Yanbian University, Yanji, Jilin 133002, China
| |
Collapse
|
23
|
Preparation and performance of a poly(ethyleneimine) embedded N-acetyl-L-phenylalanine mixed-mode stationary phase for HPLC. Microchem J 2020. [DOI: 10.1016/j.microc.2020.105021] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
24
|
Guo X, Fang Z, Zhang M, Yang D, Wang S, Liu K. A Co-Delivery System of Curcumin and p53 for Enhancing the Sensitivity of Drug-Resistant Ovarian Cancer Cells to Cisplatin. Molecules 2020; 25:molecules25112621. [PMID: 32512936 PMCID: PMC7321199 DOI: 10.3390/molecules25112621] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/25/2020] [Accepted: 06/01/2020] [Indexed: 11/16/2022] Open
Abstract
In order to enhance the sensitivity of drug-resistant ovarian cancer cells to cisplatin (DDP), a co-delivery system was designed for simultaneous delivery of curcumin (CUR) and p53 DNA. Firstly, the bifunctional peptide K14 composed of tumor targeting peptide (tLyP-1) and nuclear localization signal (NLS) was synthesized. A nonviral carrier (PEI-K14) was synthesized by cross-linking low molecular weight polyethyleneimine (PEI) with K14. Then, CUR was coupled to PEI-K14 by matrix metalloproteinase 9 (MMP9)-cleavable peptide to prepare CUR-PEI-K14. A co-delivery system, named CUR-PEI-K14/p53, was obtained by CUR-PEI-K14 and p53 self-assembly. Furthermore, the physicochemical properties and gene transfection efficiency were evaluated. Finally, ovarian cancer cisplatin-resistant (SKOV3-DDP) cells were selected to evaluate the effect of CUR-PEI-K14/p53 on enhancing the sensitivity of drug-resistant cells to DDP. The CUR-PEI-K14/DNA complexes appeared uniformly dispersed and spherical. The particle size was around 20-150 nm and the zeta potential was around 18-37 mV. It had good stability, high transfection efficiency, and low cytotoxicity. CUR-PEI-K14/p53 could significantly increase the sensitivity of SKOV3-DDP cells to DDP, and this effect was better as combined with DDP. The sensitizing effect might be related to the upregulation of p53 messenger RNA (mRNA), the downregulation of P-glycoprotein (P-gp) mRNA, and the upregulation of BCL2-Associated X (bax) mRNA. CUR-PEI-K14/p53 can be used as an effective strategy to enhance the sensitivity of drug-resistant ovarian cancer cells to DDP.
Collapse
Affiliation(s)
| | | | | | | | | | - Kehai Liu
- Correspondence: ; Tel.: +86-216-190-0388
| |
Collapse
|
25
|
Ribeiro JPM, Mendonça PV, Coelho JFJ, Matyjaszewski K, Serra AC. Glycopolymer Brushes by Reversible Deactivation Radical Polymerization: Preparation, Applications, and Future Challenges. Polymers (Basel) 2020; 12:E1268. [PMID: 32492977 PMCID: PMC7362234 DOI: 10.3390/polym12061268] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/26/2020] [Accepted: 05/29/2020] [Indexed: 12/27/2022] Open
Abstract
The cellular surface contains specific proteins, also known as lectins, that are carbohydrates receptors involved in different biological events, such as cell-cell adhesion, cell recognition and cell differentiation. The synthesis of well-defined polymers containing carbohydrate units, known as glycopolymers, by reversible deactivation radical polymerization (RDRP) methods allows the development of tailor-made materials with high affinity for lectins because of their multivalent interaction. These polymers are promising candidates for the biomedical field, namely as novel diagnostic disease markers, biosensors, or carriers for tumor-targeted therapy. Although linear glycopolymers are extensively studied for lectin recognition, branched glycopolymeric structures, such as polymer brushes can establish stronger interactions with lectins. This specific glycopolymer topology can be synthesized in a bottlebrush form or grafted to/from surfaces by using RDRP methods, allowing a precise control over molecular weight, grafting density, and brush thickness. Here, the preparation and application of glycopolymer brushes is critically discussed and future research directions on this topic are suggested.
Collapse
Affiliation(s)
- Jessica P. M. Ribeiro
- Department of Chemical Engineering, Centre for Mechanical Engineering, Materials and Processes, University of Coimbra, Rua Sílvio Lima-Polo II, 3030-790 Coimbra, Portugal; (J.P.M.R.); (J.F.J.C.)
| | - Patrícia V. Mendonça
- Department of Chemical Engineering, Centre for Mechanical Engineering, Materials and Processes, University of Coimbra, Rua Sílvio Lima-Polo II, 3030-790 Coimbra, Portugal; (J.P.M.R.); (J.F.J.C.)
| | - Jorge F. J. Coelho
- Department of Chemical Engineering, Centre for Mechanical Engineering, Materials and Processes, University of Coimbra, Rua Sílvio Lima-Polo II, 3030-790 Coimbra, Portugal; (J.P.M.R.); (J.F.J.C.)
| | - Krzysztof Matyjaszewski
- Department of Materials Science & Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, PA 15213, USA;
| | - Arménio C. Serra
- Department of Chemical Engineering, Centre for Mechanical Engineering, Materials and Processes, University of Coimbra, Rua Sílvio Lima-Polo II, 3030-790 Coimbra, Portugal; (J.P.M.R.); (J.F.J.C.)
| |
Collapse
|
26
|
Dehshahri A, Sadeghpour H, Mohazzabieh E, Saatchi Avval S, Mohammadinejad R. Targeted double domain nanoplex based on galactosylated polyethylenimine enhanced the delivery ofIL‐12 plasmid. Biotechnol Prog 2020; 36:e3002. [DOI: 10.1002/btpr.3002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/08/2020] [Accepted: 04/08/2020] [Indexed: 12/17/2022]
Affiliation(s)
- Ali Dehshahri
- Department of Pharmaceutical Biotechnology, Shiraz School of Pharmacy Shiraz University of Medical Sciences Shiraz Iran
- Pharmaceutical Sciences Research Center, Shiraz School of Pharmacy Shiraz University of Medical Sciences Shiraz Iran
| | - Hossein Sadeghpour
- Pharmaceutical Sciences Research Center, Shiraz School of Pharmacy Shiraz University of Medical Sciences Shiraz Iran
- Department of Medicinal Chemistry, Shiraz School of Pharmacy Shiraz University of Medical Sciences Shiraz Iran
| | - Erfaneh Mohazzabieh
- Department of Pharmaceutical Biotechnology, Shiraz School of Pharmacy Shiraz University of Medical Sciences Shiraz Iran
- Pharmaceutical Sciences Research Center, Shiraz School of Pharmacy Shiraz University of Medical Sciences Shiraz Iran
| | - Sara Saatchi Avval
- Department of Pharmaceutical Biotechnology, Shiraz School of Pharmacy Shiraz University of Medical Sciences Shiraz Iran
- Pharmaceutical Sciences Research Center, Shiraz School of Pharmacy Shiraz University of Medical Sciences Shiraz Iran
| | - Reza Mohammadinejad
- Pharmaceutics Research Center, Institute of Neuropharmacology Kerman University of Medical Sciences Kerman Iran
| |
Collapse
|
27
|
He M, Wang Y, Chen X, Zhao Y, Lou K, Wang Y, Huang L, Hou X, Xu J, Cai X, Cheng Y, Lan M, Yang Y, Gao F. Spatiotemporally controllable diphtheria toxin expression using a light-switchable transgene system combining multifunctional nanoparticle delivery system for targeted melanoma therapy. J Control Release 2019; 319:1-14. [PMID: 31838205 DOI: 10.1016/j.jconrel.2019.12.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/26/2019] [Accepted: 12/10/2019] [Indexed: 12/16/2022]
Abstract
Gene therapy with external gene insertion (e. g. a suicide gene) and expression specifically in mutated tumor cells has shown to be a promising strategy in treatment of tumors. However, current tumor gene therapy often suffered from low efficiency in gene expression and off-target effects which may cause damage to normal tissues. To address these issues, in this study, a light-switchable transgene nanoparticle delivery system loaded with a diphtheria toxin A (DTA) segment encoded gene, a suicide gene for tumor cells, was developed. The nanoparticles contained vitamin E succinate-grafted polyethyleneimine core and arginylglycylaspartic acid (RGD)-modified pegylated hyaluronic acid shell for targeted delivery of the loaded gene to tumor cells via receptor-mediated (CD44 and αvβ3) endocytosis. Notably, the expression of target proteins in tumor cells could be conveniently regulated by adjusting the blue light intensity in the Light-On system. In in-vitro studies in cultured B16-F10 cells, the pG-DTA-loaded nano-micelles showed greatly improved inhibitory rate compared with the pG-DTA group. Moreover, in the tumor-bearing C57BL/6 mice model, the pG-DTA-loaded nanoparticle exhibited greatly improved efficacy and reduced systemic toxicity with significantly increased survival rate after 21 days. Significantly suppressed tumor angiogenesis was also identified in the nanoparticle-treated group likely due to the targeting ability of the RGD-modified nanoparticle. All the above results indicated that the combination of a light-switchable transgene system with a nanoparticle-based targeted delivery system have great potentials in gene therapy of malignant tumors with improved precision and efficacy.
Collapse
Affiliation(s)
- Muye He
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China; Department of Pharmaceutics, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yan Wang
- Department of Pharmaceutics, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Xianjun Chen
- Shanghai Key Laboratory of New Drug Design, East China University of Science and Technology, Shanghai 200237, China; Synthetic Biology and Biotechnology Laboratory, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China; Optogenetics & Molecular Imaging Interdisciplinary Research Center, CAS Center for Excellence in Brain Science, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
| | - Yuzheng Zhao
- Shanghai Key Laboratory of New Drug Design, East China University of Science and Technology, Shanghai 200237, China; Synthetic Biology and Biotechnology Laboratory, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China; Optogenetics & Molecular Imaging Interdisciplinary Research Center, CAS Center for Excellence in Brain Science, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
| | - Kaiyan Lou
- Department of Pharmaceutics, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yujie Wang
- Department of Pharmaceutics, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Lei Huang
- Department of Pharmaceutics, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Xinyu Hou
- Department of Pharmaceutics, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Jiajun Xu
- Department of Pharmaceutics, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Xiaoran Cai
- Department of Pharmaceutics, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yi Cheng
- Department of Pharmaceutics, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Minbo Lan
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China
| | - Yi Yang
- Shanghai Key Laboratory of New Drug Design, East China University of Science and Technology, Shanghai 200237, China; Synthetic Biology and Biotechnology Laboratory, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China; Optogenetics & Molecular Imaging Interdisciplinary Research Center, CAS Center for Excellence in Brain Science, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China.
| | - Feng Gao
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China; Department of Pharmaceutics, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China; Shanghai Key Laboratory of New Drug Design, East China University of Science and Technology, Shanghai 200237, China.
| |
Collapse
|
28
|
The 40 kDa Linear Polyethylenimine Inhibits Porcine Reproductive and Respiratory Syndrome Virus Infection by Blocking Its Attachment to Permissive Cells. Viruses 2019; 11:v11090876. [PMID: 31546799 PMCID: PMC6784015 DOI: 10.3390/v11090876] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 09/09/2019] [Accepted: 09/17/2019] [Indexed: 12/12/2022] Open
Abstract
Porcine reproductive and respiratory syndrome (PRRS) is one of the most economically devastating infectious diseases in pigs worldwide. The causative agent is the PRRS virus (PRRSV). In this study, we explored polyethylenimine (PEI), a cationic polymer with different forms (linear or branched), to inhibit the replication of PRRSV. Our results demonstrate that the linear but not the 40 kDa branched PEI, or the 25 kDa linear PEI, were well tolerated in cultured cells and exhibited a broad-spectrum inhibition of heterogeneous PRRSV-2 isolates in both MARC-145 cells and primary porcine pulmonary alveolar macrophages (PAMs). Further analysis suggests that PEI could prevent the attachment of PRRSV virions to the susceptible cells. Notably, PEI had a minimal effect on PRRSV internalization in MARC-145 cells, whereas PEI promoted the internalization of PRRSV virions in PAMs, which suggests that these two types of cells might have different internalization processes of PRRSV virions. In conclusion, our data demonstrate that PEI could be used as a novel inhibitor against PRRSV.
Collapse
|
29
|
Chen Y, Diaz-Dussan D, Peng YY, Narain R. Hydroxyl-Rich PGMA-Based Cationic Glycopolymers for Intracellular siRNA Delivery: Biocompatibility and Effect of Sugar Decoration Degree. Biomacromolecules 2019; 20:2068-2074. [PMID: 30970212 DOI: 10.1021/acs.biomac.9b00274] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The ErbB family of proteins, structurally related to the epidermal growth factor receptor (EGFR), is found to be overexpressed in many cancers such as gliomas, a lung and cervical carcinomas. Gene therapy allows to modify the expression of genes like ErbB and has been a promising strategy to target oncogenes and tumor suppressor genes. In the current work, novel hydroxyl-rich poly(glycidyl methacrylate) (PGMA)-based cationic glycopolymers were designed for intracellular small interfering RNA (siRNA) delivery to silence the EGFR gene. The cationic polymers with different sugar decoration degrees (0, 9, and 33%) were synthesized by ring-opening reaction of PGMA with ethanolamine and a lactobionic acid-derived aminosaccharide (Lac-NH2). Specific EGFR knockdown of the protein tyrosine kinase ErbB-overexpressing HeLa cells was achieved using these hydroxyl-rich polycation/siRNA complexes. Higher sugar content improved the biocompatibility of the polymers, but it also seems to decrease the EGFR knockdown capability, which should mainly be related to the surface charge of polyplexes. An optimum balance was observed with PGEL-1 (9% sugar content) formulation, achieving ∼52% knockdown efficiency as well as high cell viability. Considering the specific recognition between galactose residues and asialoglycoprotein receptor in hepatocytes, our novel PGMA-based cationic glycopolymers exhibited promising future to serve as a safe and targeting gene delivery vector to hepatoma cell line like HepG2.
Collapse
Affiliation(s)
- Yangjun Chen
- School of Ophthalmology & Optometry, Eye Hospital , Wenzhou Medical University , Wenzhou 325027 , Zhejiang , China.,Department of Chemical and Materials Engineering , University of Alberta , Edmonton T6G 2G6 , Alberta , Canada
| | - Diana Diaz-Dussan
- Department of Chemical and Materials Engineering , University of Alberta , Edmonton T6G 2G6 , Alberta , Canada
| | - Yi-Yang Peng
- Department of Chemical and Materials Engineering , University of Alberta , Edmonton T6G 2G6 , Alberta , Canada
| | - Ravin Narain
- Department of Chemical and Materials Engineering , University of Alberta , Edmonton T6G 2G6 , Alberta , Canada
| |
Collapse
|