1
|
Alvarez AC, Maguire D, Brannigan RP. Synthetic-polymer-assisted antisense oligonucleotide delivery: targeted approaches for precision disease treatment. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2025; 16:435-463. [PMID: 40166479 PMCID: PMC11956074 DOI: 10.3762/bjnano.16.34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 03/12/2025] [Indexed: 04/02/2025]
Abstract
This review explores the recent advancements in polymer-assisted delivery systems for antisense oligonucleotides (ASOs) and their potential in precision disease treatment. Synthetic polymers have shown significant promise in enhancing the delivery, stability, and therapeutic efficacy of ASOs by addressing key challenges such as cellular uptake, endosomal escape, and reducing cytotoxicity. The review highlights key studies from the past decade demonstrating how these polymers improve gene silencing efficiencies, particularly in cancer and neurodegenerative disease models. Despite the progress achieved, barriers such as immunogenicity, delivery limitations, and scalability still need to be overcome for broader clinical application. Emerging strategies, including stimuli-responsive polymers and advanced nanoparticle systems, offer potential solutions to these challenges. The review underscores the transformative potential of polymer-enhanced ASO delivery in personalised medicine, emphasising the importance of continued innovation to optimise ASO-based therapeutics for more precise and effective disease treatments.
Collapse
Affiliation(s)
- Ana Cubillo Alvarez
- School of Chemical Sciences, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Dylan Maguire
- School of Chemical Sciences, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Ruairí P Brannigan
- School of Chemical Sciences, Dublin City University, Glasnevin, Dublin 9, Ireland
| |
Collapse
|
2
|
Świerczyńska M, Kudzin MH, Chruściel JJ. Poly(lactide)-Based Materials Modified with Biomolecules: A Review. MATERIALS (BASEL, SWITZERLAND) 2024; 17:5184. [PMID: 39517460 PMCID: PMC11546716 DOI: 10.3390/ma17215184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Poly(lactic acid) (PLA) is characterized by unique features, e.g., it is environmentally friendly, biocompatible, has good thermomechanical properties, and is readily available and biodegradable. Due to the increasing pollution of the environment, PLA is a promising alternative that can potentially replace petroleum-derived polymers. Different biodegradable polymers have numerous biomedical applications and are used as packaging materials. Because the pure form of PLA is delicate, brittle, and is characterized by a slow degradation rate and a low thermal resistance and crystallization rate, these disadvantages limit the range of applications of this polymer. However, the properties of PLA can be improved by chemical or physical modification, e.g., with biomolecules. The subject of this review is the modification of PLA properties with three classes of biomolecules: polysaccharides, proteins, and nucleic acids. A quite extensive description of the most promising strategies leading to improvement of the bioactivity of PLA, through modification with these biomolecules, is presented in this review. Thus, this article deals mainly with a presentation of the major developments and research results concerning PLA-based materials modified with different biomolecules (described in the world literature during the last decades), with a focus on such methods as blending, copolymerization, or composites fabrication. The biomedical and unique biological applications of PLA-based materials, especially modified with polysaccharides and proteins, are reviewed, taking into account the growing interest and great practical potential of these new biodegradable biomaterials.
Collapse
Affiliation(s)
- Małgorzata Świerczyńska
- Łukasiewicz Research Network—Lodz Institute of Technology (ŁIT), 19/27 Marii Skłodowskiej-Curie Str., 90-570 Łódź, Poland; (M.Ś.); (M.H.K.)
- Circular Economy Center (BCG), Environmental Protection Engineering Research Group, Łukasiewicz Research Network—Lodz Institute of Technology (ŁIT), Brzezińska 5/15, 92-103 Łódź, Poland
- Institute of Polymer and Dye Technology, Faculty of Chemistry, Lodz University of Technology, Stefanowskiego 16, 90-537 Łódź, Poland
| | - Marcin H. Kudzin
- Łukasiewicz Research Network—Lodz Institute of Technology (ŁIT), 19/27 Marii Skłodowskiej-Curie Str., 90-570 Łódź, Poland; (M.Ś.); (M.H.K.)
- Circular Economy Center (BCG), Environmental Protection Engineering Research Group, Łukasiewicz Research Network—Lodz Institute of Technology (ŁIT), Brzezińska 5/15, 92-103 Łódź, Poland
| | - Jerzy J. Chruściel
- Łukasiewicz Research Network—Lodz Institute of Technology (ŁIT), 19/27 Marii Skłodowskiej-Curie Str., 90-570 Łódź, Poland; (M.Ś.); (M.H.K.)
- Circular Economy Center (BCG), Environmental Protection Engineering Research Group, Łukasiewicz Research Network—Lodz Institute of Technology (ŁIT), Brzezińska 5/15, 92-103 Łódź, Poland
| |
Collapse
|
3
|
Li Y, Xin X, Zhou X, Liu J, Liu H, Yuan S, Liu H, Hao W, Sun J, Wang Y, Gong W, Yang M, Li Z, Han Y, Gao C, Yang Y. ROS-responsive biomimetic nanosystem camouflaged by hybrid membranes of platelet-exosomes engineered with neuronal targeting peptide for TBI therapy. J Control Release 2024; 372:531-550. [PMID: 38851535 DOI: 10.1016/j.jconrel.2024.06.018] [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: 01/03/2024] [Revised: 05/30/2024] [Accepted: 06/05/2024] [Indexed: 06/10/2024]
Abstract
Recovery and survival following traumatic brain injury (TBI) depends on optimal amelioration of secondary injuries at lesion site. Delivering mitochondria-protecting drugs to neurons may revive damaged neurons at sites secondarily traumatized by TBI. Pioglitazone (PGZ) is a promising candidate for TBI treatment, limited by its low brain accumulation and poor targetability to neurons. Herein, we report a ROS-responsive nanosystem, camouflaged by hybrid membranes of platelets and engineered extracellular vesicles (EVs) (C3-EPm-|TKNPs|), that can be used for targeted delivery of PGZ for TBI therapy. Inspired by intrinsic ability of macrophages for inflammatory chemotaxis, engineered M2-like macrophage-derived EVs were constructed by fusing C3 peptide to EVs membrane integrator protein, Lamp2b, to confer them with ability to target neurons in inflamed lesions. Platelets provided hybridized EPm with capabilities to target hemorrhagic area caused by trauma via surface proteins. Consequently, C3-EPm-|PGZ-TKNPs| were orientedly delivered to neurons located in the traumatized hemisphere after intravenous administration, and triggered the release of PGZ from TKNPs via oxidative stress. The current work demonstrate that C3-EPm-|TKNPs| can effectively deliver PGZ to alleviate mitochondrial damage via mitoNEET for neuroprotection, further reversing behavioral deficits in TBI mice. Our findings provide proof-of-concept evidence of C3-EPm-|TKNPs|-derived nanodrugs as potential clinical approaches against neuroinflammation-related intracranial diseases.
Collapse
Affiliation(s)
- Yi Li
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China
| | - Xin Xin
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China
| | - Xun Zhou
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China; College of Pharmacy, Henan University, Kaifeng 475000, People's Republic of China
| | - Jingzhou Liu
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China
| | - Hangbing Liu
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China; School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Shuo Yuan
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China; School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Hanhan Liu
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China
| | - Wenyan Hao
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China
| | - Jiejie Sun
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China
| | - Yuli Wang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China
| | - Wei Gong
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China
| | - Meiyan Yang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China
| | - Zhiping Li
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China
| | - Yang Han
- School of Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China.
| | - Chunsheng Gao
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China.
| | - Yang Yang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China.
| |
Collapse
|
4
|
Yang C, Lin ZI, Zhang X, Xu Z, Xu G, Wang YM, Tsai TH, Cheng PW, Law WC, Yong KT, Chen CK. Recent Advances in Engineering Carriers for siRNA Delivery. Macromol Biosci 2024; 24:e2300362. [PMID: 38150293 DOI: 10.1002/mabi.202300362] [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: 08/09/2023] [Revised: 11/29/2023] [Indexed: 12/28/2023]
Abstract
RNA interference (RNAi) technology has been a promising treatment strategy for combating intractable diseases. However, the applications of RNAi in clinical are hampered by extracellular and intracellular barriers. To overcome these barriers, various siRNA delivery systems have been developed in the past two decades. The first approved RNAi therapeutic, Patisiran (ONPATTRO) using lipids as the carrier, for the treatment of amyloidosis is one of the most important milestones. This has greatly encouraged researchers to work on creating new functional siRNA carriers. In this review, the recent advances in siRNA carriers consisting of lipids, polymers, and polymer-modified inorganic particles for cancer therapy are summarized. Representative examples are presented to show the structural design of the carriers in order to overcome the delivery hurdles associated with RNAi therapies. Finally, the existing challenges and future perspective for developing RNAi as a clinical modality will be discussed and proposed. It is believed that the addressed contributions in this review will promote the development of siRNA delivery systems for future clinical applications.
Collapse
Affiliation(s)
- Chengbin Yang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Zheng-Ian Lin
- Polymeric Biomaterials Laboratory, Department of Materials and Optoelectronic Science, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan
| | - Xinmeng Zhang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Zhourui Xu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Gaixia Xu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Yu-Min Wang
- Polymeric Biomaterials Laboratory, Department of Materials and Optoelectronic Science, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan
| | - Tzu-Hsien Tsai
- Division of Cardiology and Department of Internal Medicine, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi, 60002, Taiwan
| | - Pei-Wen Cheng
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, 81362, Taiwan
- Department of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan
| | - Wing-Cheung Law
- Department of Industrial and Systems Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, 999077, P. R. China
| | - Ken-Tye Yong
- School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales, 2006, Australia
| | - Chih-Kuang Chen
- Polymeric Biomaterials Laboratory, Department of Materials and Optoelectronic Science, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan
| |
Collapse
|
5
|
Feng S, Xie X, Liu J, Li A, Wang Q, Guo D, Li S, Li Y, Wang Z, Guo T, Zhou J, Tang DYY, Show PL. A potential paradigm in CRISPR/Cas systems delivery: at the crossroad of microalgal gene editing and algal-mediated nanoparticles. J Nanobiotechnology 2023; 21:370. [PMID: 37817254 PMCID: PMC10563294 DOI: 10.1186/s12951-023-02139-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 10/03/2023] [Indexed: 10/12/2023] Open
Abstract
Microalgae as the photosynthetic organisms offer enormous promise in a variety of industries, such as the generation of high-value byproducts, biofuels, pharmaceuticals, environmental remediation, and others. With the rapid advancement of gene editing technology, CRISPR/Cas system has evolved into an effective tool that revolutionised the genetic engineering of microalgae due to its robustness, high target specificity, and programmability. However, due to the lack of robust delivery system, the efficacy of gene editing is significantly impaired, limiting its application in microalgae. Nanomaterials have become a potential delivery platform for CRISPR/Cas systems due to their advantages of precise targeting, high stability, safety, and improved immune system. Notably, algal-mediated nanoparticles (AMNPs), especially the microalgae-derived nanoparticles, are appealing as a sustainable delivery platform because of their biocompatibility and low toxicity in a homologous relationship. In addition, living microalgae demonstrated effective and regulated distribution into specified areas as the biohybrid microrobots. This review extensively summarised the uses of CRISPR/Cas systems in microalgae and the recent developments of nanoparticle-based CRISPR/Cas delivery systems. A systematic description of the properties and uses of AMNPs, microalgae-derived nanoparticles, and microalgae microrobots has also been discussed. Finally, this review highlights the challenges and future research directions for the development of gene-edited microalgae.
Collapse
Affiliation(s)
- Shuying Feng
- Medical College, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China.
| | - Xin Xie
- Medical College, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China
| | - Junjie Liu
- Medical College, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China
| | - Aifang Li
- Medical College, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China
| | - Qianqian Wang
- Medical College, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China
| | - Dandan Guo
- Medical College, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China
| | - Shuxuan Li
- Medical College, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China
| | - Yalan Li
- Medical College, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China
| | - Zilong Wang
- Medical College, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China
| | - Tao Guo
- Department of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China.
| | - Jin Zhou
- Institute for Ocean Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, Guangdong, China.
| | - Doris Ying Ying Tang
- Department of Chemical and Environmental Engineering, Faculty of Science and Engineering, University of Nottingham Malaysia, 43500, Semenyih, Malaysia
| | - Pau Loke Show
- Department of Chemical Engineering, Khalifa University, P.O. Box 127788, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
6
|
Pires D, Mandal M, Matos AI, Peres C, Catalão MJ, Azevedo-Pereira JM, Satchi-Fainaro R, Florindo HF, Anes E. Development of Chitosan Particles Loaded with siRNA for Cystatin C to Control Intracellular Drug-Resistant Mycobacterium tuberculosis. Antibiotics (Basel) 2023; 12:729. [PMID: 37107091 PMCID: PMC10135320 DOI: 10.3390/antibiotics12040729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
The golden age of antibiotics for tuberculosis (TB) is marked by its success in the 1950s of the last century. However, TB is not under control, and the rise in antibiotic resistance worldwide is a major threat to global health care. Understanding the complex interactions between TB bacilli and their host can inform the rational design of better TB therapeutics, including vaccines, new antibiotics, and host-directed therapies. We recently demonstrated that the modulation of cystatin C in human macrophages via RNA silencing improved the anti-mycobacterial immune responses to Mycobacterium tuberculosis infection. Available in vitro transfection methods are not suitable for the clinical translation of host-cell RNA silencing. To overcome this limitation, we developed different RNA delivery systems (DSs) that target human macrophages. Human peripheral blood-derived macrophages and THP1 cells are difficult to transfect using available methods. In this work, a new potential nanomedicine based on chitosan (CS-DS) was efficiently developed to carry a siRNA-targeting cystatin C to the infected macrophage models. Consequently, an effective impact on the intracellular survival/replication of TB bacilli, including drug-resistant clinical strains, was observed. Altogether, these results suggest the potential use of CS-DS in adjunctive therapy for TB in combination or not with antibiotics.
Collapse
Affiliation(s)
- David Pires
- Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
- Center for Interdisciplinary Research in Health, Católica Medical School, Universidade Católica Portuguesa, Estrada Octávio Pato, 2635-631 Rio de Mouro, Portugal
| | - Manoj Mandal
- Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Ana I. Matos
- Drug Delivery and Immunoengineering Unit, Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Carina Peres
- Drug Delivery and Immunoengineering Unit, Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Maria João Catalão
- Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - José Miguel Azevedo-Pereira
- Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv P.O. Box 39040, Israel
| | - Helena F. Florindo
- Drug Delivery and Immunoengineering Unit, Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Elsa Anes
- Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| |
Collapse
|
7
|
Wen L, Liu H, Hu C, Wei Z, Meng Y, Lu C, Su Y, Lu L, Liang H, Xu Q, Zhan M. Thermoacoustic Imaging-Guided Thermo-Chemotherapy for Hepatocellular Carcinoma Sensitized by a Microwave-Responsive Nitric Oxide Nanogenerator. ACS APPLIED MATERIALS & INTERFACES 2023; 15:10477-10491. [PMID: 36790347 DOI: 10.1021/acsami.2c22523] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Imaging-guided percutaneous microwave thermotherapy has been regarded as an important alternative nonsurgical therapeutic strategy for hepatocellular carcinoma (HCC) that provides excellent local tumor control and favorable survival benefit. However, providing a high-resolution, real-time, and noninvasive imaging technique for intraoperative guidance and controlling postoperative residual tumor recurrence are urgent needs for the clinical setting. In this study, a cisplatin (CDDP)-loaded nanocapsule (NPs@CDDP) with microwave responsive property was prepared to simultaneously serve as a contrast agent of emerging thermoacoustic imaging and a sensitizing agent of microwave thermo-chemotherapy. Accompanying the enzymolysis in the tumor microenvironment, the NPs@CDDP responsively release l-arginine (l-Arg) and CDDP. l-Arg with excellent microwave-absorbing property allowed it to serve as a thermoacoustic imaging contrast agent for accurately delineating the tumor and remarkably increasing tumor temperature under ultralow power microwave irradiation. Apart from the chemotherapeutic effect, CDDP elevated the intracellular H2O2 level through cascade reactions and further accelerated the continuous transformation of l-Arg to nitric oxide (NO), which endowed the NPs@CDDP with NO-generation capability. Notably, the high concentration of intracellular NO was proved to aggravate lipid peroxidation and greatly improved the efficacy of microwave thermo-chemotherapy. Thereby, NPs@CDDP was expected to serve as a theranostic agent integrating the functions of tumor microenvironment-responsive drug delivery system, contrast agent of thermoacoustic imaging, thermal sensitizing agent, and NO nanogenerator, which was promising to provide a potential imaging-guided therapeutic strategy for HCC.
Collapse
Affiliation(s)
- Liewei Wen
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai, Guangdong 519000, P.R. China
| | - Hongyi Liu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai, Guangdong 519000, P.R. China
| | - Cong Hu
- Department of General Surgery, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai 519000, China
| | - Zixuan Wei
- Medical College, Guangxi University, Nanning 530004, China
| | - Ya Meng
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai, Guangdong 519000, P.R. China
- Faculty of Health Sciences, University of Macau, Taipa, Macau SAR China
| | - Cuixia Lu
- Medical College, Guangxi University, Nanning 530004, China
| | - Yanhong Su
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai, Guangdong 519000, P.R. China
- Faculty of Health Sciences, University of Macau, Taipa, Macau SAR China
| | - Ligong Lu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai, Guangdong 519000, P.R. China
| | - Hui Liang
- Department of General Surgery, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai 519000, China
| | - Qingbo Xu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai, Guangdong 519000, P.R. China
| | - Meixiao Zhan
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai, Guangdong 519000, P.R. China
| |
Collapse
|
8
|
Yang J, Wang P, Jiang X, Xu J, Zhang M, Liu F, Lin Y, Tao J, He J, Zhou X, Zhang M. A Nanotherapy of Octanoic Acid Ameliorates Cardiac Arrest/Cardiopulmonary Resuscitation-Induced Brain Injury via RVG29- and Neutrophil Membrane-Mediated Injury Relay Targeting. ACS NANO 2023; 17:3528-3548. [PMID: 36758159 DOI: 10.1021/acsnano.2c09931] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Treatment of cardiac arrest/cardiopulmonary resuscitation (CA/CPR)-induced brain injury remains a challenging issue without viable therapeutic options. Octanoic acid (OA), a lipid oil that is mainly metabolized in the astrocytes of the brain, is a promising treatment for this type of injury owing to its potential functions against oxidative stress, apoptosis, inflammation, and ability to stabilize mitochondria. However, the application of OA is strictly limited by its short half-life and low available concentration in the target organ. Herein, based on our previous research, an OA-based nanotherapy coated with a neutrophil membrane highly expressing RVG29, RVG29-H-NPOA, was successfully constructed by computer simulation-guided supramolecular assembly of polyethylenimine and OA. The in vitro and in vivo experiments showed that RVG29-H-NPOA could target and be distributed in the injured brain focus via the relay-targeted delivery mediated by RVG29-induced blood-brain barrier (BBB) penetration and neutrophil membrane protein-induced BBB binding and injury targeting. This results in enhancements of the antioxidant, antiapoptotic, mitochondrial stability-promoting and anti-inflammatory effects of OA and exhibited systematic alleviation of astrocyte injury, neuronal damage, and inflammatory response in the brain. Due to their systematic intervention in multiple pathological processes, RVG29-H-NPOA significantly increased the 24 h survival rate of CA/CPR model rats from 40% to 100% and significantly improved their neurological functions. Thus, RVG29-H-NPOA are expected to be a promising therapeutic for the treatment of CA/CPR-induced brain injury.
Collapse
Affiliation(s)
- Jingyuan Yang
- Department of Emergency Medicine, Second Affiliated Hospital of Zhejiang University, Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burns of Zhejiang Province, Clinical Research Center for Emergency and Critical Care Medicine of Zhejiang Province, Hangzhou 310009, China
| | - Pan Wang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Xiangkang Jiang
- Department of Emergency Medicine, Second Affiliated Hospital of Zhejiang University, Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burns of Zhejiang Province, Clinical Research Center for Emergency and Critical Care Medicine of Zhejiang Province, Hangzhou 310009, China
| | - Jiefeng Xu
- Department of Emergency Medicine, Second Affiliated Hospital of Zhejiang University, Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burns of Zhejiang Province, Clinical Research Center for Emergency and Critical Care Medicine of Zhejiang Province, Hangzhou 310009, China
| | - Minhai Zhang
- Department of Emergency Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Fei Liu
- Department of Emergency Medicine, Second Affiliated Hospital of Zhejiang University, Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burns of Zhejiang Province, Clinical Research Center for Emergency and Critical Care Medicine of Zhejiang Province, Hangzhou 310009, China
| | - Yao Lin
- Department of Emergency Medicine, Second Affiliated Hospital of Zhejiang University, Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burns of Zhejiang Province, Clinical Research Center for Emergency and Critical Care Medicine of Zhejiang Province, Hangzhou 310009, China
| | - Jiawei Tao
- Department of Emergency Medicine, Second Affiliated Hospital of Zhejiang University, Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burns of Zhejiang Province, Clinical Research Center for Emergency and Critical Care Medicine of Zhejiang Province, Hangzhou 310009, China
| | - Jiantao He
- Department of Emergency Medicine, Second Affiliated Hospital of Zhejiang University, Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burns of Zhejiang Province, Clinical Research Center for Emergency and Critical Care Medicine of Zhejiang Province, Hangzhou 310009, China
| | - Xing Zhou
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Mao Zhang
- Department of Emergency Medicine, Second Affiliated Hospital of Zhejiang University, Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burns of Zhejiang Province, Clinical Research Center for Emergency and Critical Care Medicine of Zhejiang Province, Hangzhou 310009, China
| |
Collapse
|
9
|
Wang H, Qin L, Zhang X, Guan J, Mao S. Mechanisms and challenges of nanocarriers as non-viral vectors of therapeutic genes for enhanced pulmonary delivery. J Control Release 2022; 352:970-993. [PMID: 36372386 PMCID: PMC9671523 DOI: 10.1016/j.jconrel.2022.10.061] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/31/2022] [Accepted: 10/31/2022] [Indexed: 11/17/2022]
Abstract
With the rapid development of biopharmaceuticals and the outbreak of COVID-19, the world has ushered in a frenzy to develop gene therapy. Therefore, therapeutic genes have received enormous attention. However, due to the extreme instability and low intracellular gene expression of naked genes, specific vectors are required. Viral vectors are widely used attributed to their high transfection efficiency. However, due to the safety concerns of viral vectors, nanotechnology-based non-viral vectors have attracted extensive investigation. Still, issues of low transfection efficiency and poor tissue targeting of non-viral vectors need to be addressed. Especially, pulmonary gene delivery has obvious advantages for the treatment of inherited lung diseases, lung cancer, and viral pneumonia, which can not only enhance lung targeting and but also reduce enzymatic degradation. For systemic diseases therapy, pulmonary gene delivery can enhance vaccine efficacy via inducing not only cellular, humoral immunity but also mucosal immunity. This review provides a comprehensive overview of nanocarriers as non-viral vectors of therapeutic genes for enhanced pulmonary delivery. First of all, the characteristics and therapeutic mechanism of DNA, mRNA, and siRNA are provided. Thereafter, the advantages and challenges of pulmonary gene delivery in exerting local and systemic effects are discussed. Then, the inhalation dosage forms for nanoparticle-based drug delivery systems are introduced. Moreover, a series of materials used as nanocarriers for pulmonary gene delivery are presented, and the endosomal escape mechanisms of nanocarriers based on different materials are explored. The application of various non-viral vectors for pulmonary gene delivery are summarized in detail, with the perspectives of nano-vectors for pulmonary gene delivery.
Collapse
Affiliation(s)
| | | | - Xin Zhang
- Corresponding authors at: School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, 110016 Shenyang, China
| | | | - Shirui Mao
- Corresponding authors at: School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, 110016 Shenyang, China
| |
Collapse
|
10
|
Shinn J, Kwon N, Lee SA, Lee Y. Smart pH-responsive nanomedicines for disease therapy. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2022; 52:427-441. [PMID: 35573320 PMCID: PMC9083479 DOI: 10.1007/s40005-022-00573-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 04/20/2022] [Indexed: 02/06/2023]
Abstract
Background Currently nanomedicines are the focus of attention from researchers and clinicians because of the successes of lipid-nanoparticles-based COVID-19 vaccines. Nanoparticles improve existing treatments by providing a number of advantages including protection of cargo molecules from external stresses, delivery of drugs to target tissues, and sustained drug release. To prevent premature release-related side effects, stable drug loading in nanoformulations is required, but the increased stability of the formulation could also lead to a poor drug-release profile at the target sites. Thus, researchers have exploited differences in a range of properties (e.g., enzyme levels, pH, levels of reduced glutathione, and reactive oxygen species) between non-target and target sites for site-specific release of drugs. Among these environmental stimuli, pH gradients have been widely used to design novel, responsive nanoparticles. Area covered In this review, we assess drug delivery based on pH-responsive nanoparticles at the levels of tissues (tumor microenvironment, pH ~ 6.5) and of intracellular compartments (endosome and lysosome, pH 4.5-6.5). Upon exposure to these pH stimuli, pH-responsive nanoparticles respond with physicochemical changes to their material structure and surface characteristics. These changes include swelling, dissociation, or surface charge switching, in a manner that favors drug release at the target site (the tumor microenvironment region and the cytosol followed by endosomal escape) rather than the surrounding tissues. Expert opinion Lastly, we consider the challenges involved in the development of pH-responsive nanomedicines.
Collapse
Affiliation(s)
- Jongyoon Shinn
- College of Pharmacy, Ewha Womans University, Seoul, 03760 South Korea
| | - Nuri Kwon
- College of Pharmacy, Ewha Womans University, Seoul, 03760 South Korea
| | - Seon Ah Lee
- College of Pharmacy, Ewha Womans University, Seoul, 03760 South Korea
| | - Yonghyun Lee
- College of Pharmacy, Ewha Womans University, Seoul, 03760 South Korea
| |
Collapse
|
11
|
Yi-Bin W, Xiang L, Bing Y, Qi Z, Fei-Tong J, Minghong W, Xiangxiang Z, Le K, Yan L, Ping S, Yufei G, Ye X, Chun-Yan W. Inhibition of the CEBPβ-NFκB interaction by nanocarrier-packaged Carnosic acid ameliorates glia-mediated neuroinflammation and improves cognitive function in an Alzheimer's disease model. Cell Death Dis 2022; 13:318. [PMID: 35393391 PMCID: PMC8989877 DOI: 10.1038/s41419-022-04765-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 03/10/2022] [Accepted: 03/25/2022] [Indexed: 01/09/2023]
Abstract
Neuroinflammation occurs early in Alzheimer’s disease (AD). The initial stage of AD is related to glial dysfunction, which contributes to impairment of Aβ clearance and disruption of synaptic connection. CEBPβ, a member of the CCAAT-enhancer-binding protein (CEBP) family, modulates the expression of inflammation-associated genes, and its expression is elevated in brains undergoing degeneration and injured brains. However, the mechanism underlying CEBPβ-mediated chronic inflammation in AD is unclear. In this study, we observed that increases in the levels of nuclear CEBPβ facilitated the interaction of CEBPβ with the NFκB p65 subunit, increasing the transcription of proinflammatory cytokines in the APP/PS1 mouse brain. Oral administration of nanocarrier-packaged carnosic acid (CA) reduced the aberrant activation of microglia and astrocytes and diminished mature IL-1β, TNFα and IL-6 production in the APP/PS1 mouse brain. CA administration reduced β-amyloid (Aβ) deposition and ameliorated cognitive impairment in APP/PS1 mice. We observed that CA blocked the interaction of CEBPβ with NFκB p65, and chromatin immunoprecipitation revealed that CA reduced the transcription of the NFκB target genes TNFα and IL-6. We confirmed that CA alleviated inflammatory mediator-induced neuronal degeneration and reduced Aβ secretion by inhibiting the CEBPβ-NFκB signalling pathway in vitro. Sulfobutyl ether-beta-cyclodextrin (SBEβCD) was used as the encapsulation agent for the CA-loaded nanocarrier to overcome the poor water solubility and enhance the brain bioavailability of CA. The CA nanoparticles (NPs) had no obvious toxicity. We demonstrated a feasible SBEβCD-based nanodelivery system targeting the brain. Our data provide experimental evidence that CA-loaded NPs are potential therapeutic agents for AD treatment.
Collapse
Affiliation(s)
- Wang Yi-Bin
- Institute of Health Sciences, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang, 110122, China
| | - Li Xiang
- Institute of Health Sciences, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang, 110122, China
| | - Yang Bing
- Institute of Health Sciences, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang, 110122, China
| | - Zhang Qi
- Institute of Health Sciences, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang, 110122, China
| | - Jiao Fei-Tong
- Translational Medicine Laboratory, School of Basic Medical Sciences, Jilin Medical University, Jilin, 132013, China
| | - Wang Minghong
- Translational Medicine Laboratory, School of Basic Medical Sciences, Jilin Medical University, Jilin, 132013, China
| | - Zhang Xiangxiang
- Institute of Health Sciences, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang, 110122, China
| | - Kang Le
- Institute of Health Sciences, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang, 110122, China
| | - Li Yan
- Institute of Health Sciences, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang, 110122, China
| | - Sui Ping
- Institute of Health Sciences, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang, 110122, China
| | - Gao Yufei
- Institute of Health Sciences, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang, 110122, China
| | - Xu Ye
- Translational Medicine Laboratory, School of Basic Medical Sciences, Jilin Medical University, Jilin, 132013, China.
| | - Wang Chun-Yan
- Institute of Health Sciences, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang, 110122, China. .,Translational Medicine Laboratory, School of Basic Medical Sciences, Jilin Medical University, Jilin, 132013, China.
| |
Collapse
|
12
|
Samec T, Boulos J, Gilmore S, Hazelton A, Alexander-Bryant A. Peptide-based delivery of therapeutics in cancer treatment. Mater Today Bio 2022; 14:100248. [PMID: 35434595 PMCID: PMC9010702 DOI: 10.1016/j.mtbio.2022.100248] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 03/14/2022] [Accepted: 03/27/2022] [Indexed: 11/09/2022] Open
Abstract
Current delivery strategies for cancer therapeutics commonly cause significant systemic side effects due to required high doses of therapeutic, inefficient cellular uptake of drug, and poor cell selectivity. Peptide-based delivery systems have shown the ability to alleviate these issues and can significantly enhance therapeutic loading, delivery, and cancer targetability. Peptide systems can be tailor-made for specific cancer applications. This review describes three peptide classes, targeting, cell penetrating, and fusogenic peptides, as stand-alone nanoparticle systems, conjugations to nanoparticle systems, or as the therapeutic modality. Peptide nanoparticle design, characteristics, and applications are discussed as well as peptide applications in the clinical space.
Collapse
Affiliation(s)
- Timothy Samec
- Nanobiotechnology Laboratory, Clemson University, Department of Bioengineering, Clemson, SC, USA
| | - Jessica Boulos
- Nanobiotechnology Laboratory, Clemson University, Department of Bioengineering, Clemson, SC, USA
| | - Serena Gilmore
- Nanobiotechnology Laboratory, Clemson University, Department of Bioengineering, Clemson, SC, USA
| | - Anthony Hazelton
- Nanobiotechnology Laboratory, Clemson University, Department of Bioengineering, Clemson, SC, USA
| | - Angela Alexander-Bryant
- Nanobiotechnology Laboratory, Clemson University, Department of Bioengineering, Clemson, SC, USA
| |
Collapse
|
13
|
Hao W, Cui Y, Fan Y, Chen M, Yang G, Wang Y, Yang M, Li Z, Gong W, Yang Y, Gao C. Hybrid membrane-coated nanosuspensions for multi-modal anti-glioma therapy via drug and antigen delivery. J Nanobiotechnology 2021; 19:378. [PMID: 34801032 PMCID: PMC8606100 DOI: 10.1186/s12951-021-01110-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/02/2021] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Glioma is one of the deadliest human cancers. Although many therapeutic strategies for glioma have been explored, these strategies are seldom used in the clinic. The challenges facing the treatment of glioma not only involve the development of chemotherapeutic drugs and immunotherapeutic agents, but also the lack of a powerful platform that could deliver these two moieties to the targeted sites. Herein, we developed chemoimmunotherapy delivery vehicles based on C6 cell membranes and DC membranes to create hybrid membrane-coated DTX nanosuspensions (DNS-[C6&DC]m). RESULTS Results demonstrated successful hybrid membrane fusion and nanosuspension functionalization, and DNS-[C6&DC]m could be used for different modes of anti-glioma therapy. For drug delivery, membrane coating could be applied to target the source cancer cells via a homotypic-targeting mechanism of the C6 cell membrane. For cancer immunotherapy, biomimetic nanosuspension enabled an immune response based on the professional antigen-presenting characteristic of the dendritic cell membrane (DCm), which carry the full array of cancer cell membrane antigens and facilitate the uptake of membrane-bound tumor antigens for efficient presentation and downstream immune n. CONCLUSION DNS-[C6&DC]m is a multifunctional biomimetic nano-drug delivery system with the potential to treat gliomas through tumor-targeted drug delivery combined with immunotherapy, thereby presenting a promising approach that may be utilized for multiple modes of cancer therapy.
Collapse
Affiliation(s)
- Wenyan Hao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China
| | - Yuexin Cui
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China
| | - Yueyue Fan
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China
| | - Mengyu Chen
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China
| | - Guobao Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China
| | - Yuli Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China.
| | - Meiyan Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China
| | - Zhiping Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China
| | - Wei Gong
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China
| | - Yang Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China.
| | - Chunsheng Gao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China.
| |
Collapse
|
14
|
Kumar R, Santa Chalarca CF, Bockman MR, Bruggen CV, Grimme CJ, Dalal RJ, Hanson MG, Hexum JK, Reineke TM. Polymeric Delivery of Therapeutic Nucleic Acids. Chem Rev 2021; 121:11527-11652. [PMID: 33939409 DOI: 10.1021/acs.chemrev.0c00997] [Citation(s) in RCA: 181] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The advent of genome editing has transformed the therapeutic landscape for several debilitating diseases, and the clinical outlook for gene therapeutics has never been more promising. The therapeutic potential of nucleic acids has been limited by a reliance on engineered viral vectors for delivery. Chemically defined polymers can remediate technological, regulatory, and clinical challenges associated with viral modes of gene delivery. Because of their scalability, versatility, and exquisite tunability, polymers are ideal biomaterial platforms for delivering nucleic acid payloads efficiently while minimizing immune response and cellular toxicity. While polymeric gene delivery has progressed significantly in the past four decades, clinical translation of polymeric vehicles faces several formidable challenges. The aim of our Account is to illustrate diverse concepts in designing polymeric vectors towards meeting therapeutic goals of in vivo and ex vivo gene therapy. Here, we highlight several classes of polymers employed in gene delivery and summarize the recent work on understanding the contributions of chemical and architectural design parameters. We touch upon characterization methods used to visualize and understand events transpiring at the interfaces between polymer, nucleic acids, and the physiological environment. We conclude that interdisciplinary approaches and methodologies motivated by fundamental questions are key to designing high-performing polymeric vehicles for gene therapy.
Collapse
Affiliation(s)
- Ramya Kumar
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | | | - Matthew R Bockman
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Craig Van Bruggen
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Christian J Grimme
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Rishad J Dalal
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Mckenna G Hanson
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Joseph K Hexum
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
15
|
Zhao J, Ullah I, Gao B, Guo J, Ren XK, Xia S, Zhang W, Feng Y. Agmatine-grafted bioreducible poly(l-lysine) for gene delivery with low cytotoxicity and high efficiency. J Mater Chem B 2021; 8:2418-2430. [PMID: 32115589 DOI: 10.1039/c9tb02641j] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Bioreducible cationic polymers have gained considerable attention in gene delivery due to their low cytotoxicity and high efficiency. In the present work, we reported a cationic polymer, poly(disulfide-l-lysine)-g-agmatine (denoted as SSL-AG), and evaluated its ability to transfer pEGFP-ZNF580 plasmid (pZNF580) into human umbilical vein endothelial cells (HUVECs). This SSL-AG polymeric carrier efficiently condensed pZNF580 into positively charged particles (<200 nm) through electrostatic interaction. This carrier also exhibited excellent buffering capacity in the physiological environment, good pDNA protection against enzymatic degradation and rapid pDNA release in a highly reducing environment mainly because of the responsive cleavage of disulfide bonds in the polymer backbone. The hemolysis assay and in vitro cytotoxicity assay suggested that the SSL-AG carrier and corresponding gene complexes possessed both good hemocompatibility and great cell viability in HUVECs. The cellular uptake of the SSL-AG/Cy5-oligonucleotide group was 3.6 times that of the poly(l-lysine)/Cy5-oligonucleotide group, and its mean fluorescence intensity value was even higher than that of the PEI 25 kDa/Cy5-oligonucleotide group. Further, the intracellular trafficking results demonstrated that the SSL-AG/Cy5-oligonucleotide complexes exhibited a high nucleus co-localization rate (CLR) value (36.0 ± 2.8%, 3.4 times that of the poly (l-lysine)/Cy5-oligonucleotide group, 1.6 times that of the poly(disulfide-l-lysine)-g-butylenediamine/Cy5-oligonucleotide group) at 24 h, while the endo/lysosomal CLR value was relatively low. This suggested that SSL-AG successfully delivered plasmid into HUVECs with high cellular uptake, rapid endosomal escape and efficient nuclear accumulation owing to the structural advantages of the bioreducible and agmatine groups. In vitro transfection assay also verified the enhanced transfection efficiency in the SSL-AG/pZNF580 group. Furthermore, the results of CCK-8, cell migration and in vitro/vivo angiogenesis assays revealed that pZNF580 delivered by SSL-AG could effectively enhance the proliferation, migration and vascularization of HUVECs. In a word, the SSL-AG polymer has great potential as a safe and efficient gene carrier for gene therapy.
Collapse
Affiliation(s)
- Jing Zhao
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin 300350, China. and Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin), Weijin Road 92, Tianjin 300072, China
| | - Ihsan Ullah
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin 300350, China.
| | - Bin Gao
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin 300350, China. and Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin), Weijin Road 92, Tianjin 300072, China
| | - Jintang Guo
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin 300350, China. and Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin), Weijin Road 92, Tianjin 300072, China
| | - Xiang-Kui Ren
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin 300350, China. and Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin), Weijin Road 92, Tianjin 300072, China and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
| | - Shihai Xia
- Department of Hepatopancreatobiliary and Splenic Medicine, Affiliated Hospital, Logistics University of People's Armed Police Force, Chenglin Road 220, Tianjin 300162, China
| | - Wencheng Zhang
- Department of Physiology and Pathophysiology, Logistics University of People's Armed Police Force, Chenglin Road 220, Tianjin 300162, China
| | - Yakai Feng
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin 300350, China. and Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin), Weijin Road 92, Tianjin 300072, China and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
| |
Collapse
|
16
|
Han Y, Chu X, Cui L, Fu S, Gao C, Li Y, Sun B. Neuronal mitochondria-targeted therapy for Alzheimer's disease by systemic delivery of resveratrol using dual-modified novel biomimetic nanosystems. Drug Deliv 2021; 27:502-518. [PMID: 32228100 PMCID: PMC7170363 DOI: 10.1080/10717544.2020.1745328] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Reactive oxygen species (ROS)-induced neuronal mitochondrial dysfunction is a key pathologic factor in sporadic Alzheimer’s disease (AD). Neuronal mitochondria have been proposed to be a promising therapeutic target for AD, especially for the failures of phase III clinical trials on conventional amyloid-β (Aβ) targeted therapy. However, the efficient intravenous delivery of therapeutic agents to neuronal mitochondria in the brain remains a major challenge due to the complicated physiological environment. Recently, biomaterials-based nanomedicine has been widely investigated for the treatment of AD. Herein, we devised a strategy for functional antioxidant delivery to neuronal mitochondria by loading antioxidants into red blood cell (RBC) membrane-coated nanostructured lipid carriers (NLC) bearing rabies virus glycoprotein (RVG29) and triphenylphosphine cation (TPP) molecules attached to the RBC membrane surface (RVG/TPP NPs@RBCm). With the advantage of suitable physicochemical properties of NLC and unique biological functions of the RBC membrane, RVG/TPP NPs@RBCm are stabilized and enabled sustained drug release, providing improved biocompatibility and long-term circulation. Under the synergistic effects of RVG29 and TPP, RVG/TPP NPs@RBCm can not only penetrate the blood–brain barrier (BBB) but also target neuron cells and further localize in the mitochondria. After encapsulating Resveratrol (RSV) as the model antioxidant, the data demonstrated that RVG/TPP-RSV NPs@RBCm can relieve AD symptoms by mitigating Aβ-related mitochondrial oxidative stress both in vitro and in vivo. The memory impairment in APP/PS1 mice is significantly improved following the systemic administration of RVG/TPP-RSV NPs@RBCm. In conclusion, intravenous neuronal mitochondria-targeted dual-modified novel biomimetic nanosystems are a promising therapeutic candidate for ROS-induced mitochondrial dysfunction in AD.
Collapse
Affiliation(s)
- Yang Han
- School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Xiaoyang Chu
- Department of stomatology, The Fifth Medical Center of PLA General Hospital, Beijing, PR China
| | - Lin Cui
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, PR China
| | - Shiyao Fu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, PR China
| | - Chunsheng Gao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, PR China
| | - Yi Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, PR China
| | - Baoshan Sun
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, PR China.,Instituto National de Investigação Agrária e Veterinária, I.P., Pólo Dois Portos, Dois Portos, Portugal
| |
Collapse
|
17
|
Jiang X, Abedi K, Shi J. Polymeric nanoparticles for RNA delivery. REFERENCE MODULE IN MATERIALS SCIENCE AND MATERIALS ENGINEERING 2021. [PMCID: PMC8568333 DOI: 10.1016/b978-0-12-822425-0.00017-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
As exemplified by recent clinical approval of RNA drugs including the latest COVID-19 mRNA vaccines, RNA therapy has demonstrated great promise as an emerging medicine. Central to the success of RNA therapy is the delivery of RNA molecules into the right cells at the right location. While the clinical success of nanotechnology in RNA therapy has been limited to lipid-based nanoparticles currently, polymers, due to their tunability and robustness, have also evolved as a class of promising material for the delivery of various therapeutics including RNAs. This article overviews different types of polymers used in RNA delivery and the methods for the formulation of polymeric nanoparticles and highlights recent progress of polymeric nanoparticle-based RNA therapy.
Collapse
|
18
|
Targeted nanomedicine with anti-EGFR scFv for siRNA delivery into triple negative breast cancer cells. Eur J Pharm Biopharm 2020; 157:74-84. [DOI: 10.1016/j.ejpb.2020.10.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/04/2020] [Accepted: 10/08/2020] [Indexed: 01/01/2023]
|
19
|
Wong S, Kemp JA, Shim MS, Kwon YJ. Solvent-driven, self-assembled acid-responsive poly(ketalized serine)/siRNA complexes for RNA interference. Biomater Sci 2020; 8:6718-6729. [PMID: 33111729 DOI: 10.1039/d0bm01478h] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Advances in bionanotechnology aim to develop smart nucleic acid delivery carriers with stimuli-responsive features to overcome challenges such as non-biodegradability, rapid clearance, immune response, and reaching intracellular targets. Peptide-based nanomaterials have become widely used in the field of gene and drug delivery due to their structural versatility and biomimetic properties. Particularly, polypeptide gene vectors that respond to biological stimuli, such as acidic intracellular environments, have promising applications in mediating efficient endosomal escape and drug release. Unfortunately, synthesis strategies for efficient polymerization of acid-labile peptides have been limited due to conditions that fail to preserve acid-degradable functional groups. Stable urethane derivatives of the acid-labile amino acid ketalized serine (kSer) were synthesized and polymerized to a high molecular weight under permissive conditions independent of elevated temperature, restrictive solvents, or an inert atmosphere. A new formulation strategy utilizing solvent-driven self-assembly of poly(kSer) peptides with small interfering RNA (siRNA) was developed, and the resulting poly(kSer)/siRNA complexes were further cross-linked for reinforced stability under physiological conditions. The complexes were highly monodisperse and precisely spherical in morphology, which has significant clinical implications in definitive biodistribution, cellular internalization, and intracellular trafficking patterns. Self-assembled, cross-linked poly(kSer)/siRNA complexes demonstrated efficient nucleic acid encapsulation, internalization, endosomal escape, and acid-triggered cargo release, tackling multiple hurdles in siRNA delivery. The acid-responsive polypeptides and solvent-driven self-assembly strategies demonstrated in this study could be applicable to developing other efficient and safe delivery systems for gene and drug delivery.
Collapse
Affiliation(s)
- Shirley Wong
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, CA 92697, USA.
| | | | | | | |
Collapse
|
20
|
Cui Y, Sun J, Hao W, Chen M, Wang Y, Xu F, Gao C. Dual-Target Peptide-Modified Erythrocyte Membrane-Enveloped PLGA Nanoparticles for the Treatment of Glioma. Front Oncol 2020; 10:563938. [PMID: 33194638 PMCID: PMC7609867 DOI: 10.3389/fonc.2020.563938] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 09/30/2020] [Indexed: 12/14/2022] Open
Abstract
Penetration of the blood–brain barrier (BBB) and the blood–brain tumor barrier (BBTB) remains a significant challenge for the delivery of drugs in the treatment of glioma. Therefore, the development of targeted preparations with the ability to penetrate the BBB and BBTB, and target gliomas, is an important approach if we are to improve the efficacy of glioma treatment. In the current study, an active targeting preparation based on PLGA nanoparticles coated with erythrocyte membranes (RBCNPs) and dual-modified with DWSW and NGR peptide ligands (DWSW/NGR-RBCNPs). Euphorbia factor L1 (EFL1) extracted from euphorbiae semen was used as the model drug. The final nanoparticles were characterized by in vivo and in vitro tests. In vitro results showed that EFL1-loaded DWSW/NGR-RBCNPs were taken up by cells and had the ability to penetrate the BBB and BBTB and produce cytotoxic effects. Furthermore, in vivo studies in mice showed that when injected intravenously, these specialized NPs could enter the brain, target tumor tissue, and significantly extend life span. The results showed that dual-targeting EFL1-loaded DWSW/NGR-RBCNPs have significant potential as a nanotherapeutic tool for the treatment of brain glioma.
Collapse
Affiliation(s)
- Yuexin Cui
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Jiejie Sun
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Wenyan Hao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Mengyu Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Yingzi Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Fenghua Xu
- Department of Pharmacy, People's Liberation Army of China (PLA) General Hospital, Beijing, China
| | - Chunsheng Gao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| |
Collapse
|
21
|
Pereira-Silva M, Jarak I, Santos AC, Veiga F, Figueiras A. Micelleplex-based nucleic acid therapeutics: From targeted stimuli-responsiveness to nanotoxicity and regulation. Eur J Pharm Sci 2020; 153:105461. [DOI: 10.1016/j.ejps.2020.105461] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 07/07/2020] [Accepted: 07/08/2020] [Indexed: 12/11/2022]
|
22
|
Han Y, Gao C, Wang H, Sun J, Liang M, Feng Y, Liu Q, Fu S, Cui L, Gao C, Li Y, Yang Y, Sun B. Macrophage membrane-coated nanocarriers Co-Modified by RVG29 and TPP improve brain neuronal mitochondria-targeting and therapeutic efficacy in Alzheimer's disease mice. Bioact Mater 2020; 6:529-542. [PMID: 32995678 PMCID: PMC7492821 DOI: 10.1016/j.bioactmat.2020.08.017] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 08/21/2020] [Accepted: 08/23/2020] [Indexed: 12/30/2022] Open
Abstract
Neuronal mitochondrial dysfunction caused by excessive reactive oxygen species (ROS) is an early event of sporadic Alzheimer's disease (AD), and considered to be a key pathologic factor in the progression of AD. The targeted delivery of the antioxidants to mitochondria of injured neurons in brain is a promising therapeutic strategy for AD. A safe and effective drug delivery system (DDS) which is able to cross the blood-brain barrier (BBB) and target neuronal mitochondria is necessary. Recently, bioactive materials-based DDS has been widely investigated for the treatment of AD. Herein, we developed macrophage (MA) membrane-coated solid lipid nanoparticles (SLNs) by attaching rabies virus glycoprotein (RVG29) and triphenylphosphine cation (TPP) molecules to the surface of MA membrane (RVG/TPP-MASLNs) for functional antioxidant delivery to neuronal mitochondria. According to the results, MA membranes camouflaged the SLNs from being eliminated by RES-rich organs by inheriting the immunological characteristics of macrophages. The unique properties of the DDS after decoration with RVG29 on the surface was demonstrated by the ability to cross the BBB and the selective targeting to neurons. After entering the neurons in CNS, TPP further lead the DDS to mitochondria driven by electric charge. The Genistein (GS)- encapsulated DDS (RVG/TPP-MASLNs-GS) exhibited the most favorable effects on reliveing AD symptoms in vitro and in vivo by the synergies gained from the combination of MA membranes, RVG29 and TPP. These results demonstrated a promising therapeutic candidate for delaying the progression of AD via neuronal mitochondria-targeted delivery by the designed biomimetic nanosystems. MA membranes inherited the immunological properties of macrophages, providing RVG/TPP-MASLNs with enhanced RES evasion. RVG/TPP-MASLNs combined the advantages of RVG29, TPP and MA, greatly improving the efficiency for brain targeting delivery. The biomimetic nanosystems effectively improve the curative effect of genistein on the symptoms of AD mice with biosafety.
Collapse
Affiliation(s)
- Yang Han
- School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang, 110016, PR China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, PR China
| | - Chunhong Gao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, PR China
| | - Hao Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, PR China
| | - Jiejie Sun
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, PR China
| | - Meng Liang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, PR China
| | - Ye Feng
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, PR China
| | - Qianqian Liu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, PR China
| | - Shiyao Fu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, PR China
| | - Lin Cui
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, PR China
| | - Chunsheng Gao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, PR China
| | - Yi Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, PR China
| | - Yang Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, PR China
| | - Baoshan Sun
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, 10016, PR China.,Instituto National de Investigação Agrária e Veterinária, I.P., Pólo Dois Portos, Quinta da Almoinha, Dois Portos, 2565-191, Portugal
| |
Collapse
|
23
|
Zhang X, Dai Y, Dai G, Deng C. Advances in PEG-based ABC terpolymers and their applications. RSC Adv 2020; 10:21602-21614. [PMID: 35518773 PMCID: PMC9054495 DOI: 10.1039/d0ra03478a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 05/18/2020] [Indexed: 12/16/2022] Open
Abstract
ABC terpolymers are a class of very important polymers because of their expansive molecular topologies and extensive architectures. As block A, poly(ethylene glycol) (PEG) is one of the most principal categories owing to good biocompatibility and wide commercial availability. More importantly, the synthetic approaches of ABC terpolymers using PEG as a macroinitiator are facile and varied. PEG-based ABC terpolymers from design and synthesis to applications are highlighted in this review. Linear, 3-miktoarm, and cyclic polymers as the architecture are separated. The synthetic approaches of PEG-based ABC terpolymers mainly include the sequential polymerization or coupling of polymers. PEG-based ABC terpolymers have wide applications in the fields of drug carriers, gene vectors, templates for the fabrication of inorganic hollow nanospheres, and stabilizers of metal nanoparticles.
Collapse
Affiliation(s)
- Xiaojin Zhang
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences Wuhan 430074 China
| | - Yu Dai
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences Wuhan 430074 China
| | - Guofei Dai
- Jiangxi Provincial Key Laboratory of Water Resources and Environment of Poyang Lake, Jiangxi Institute of Water Sciences Nanchang 330029 China
| | - Chunhui Deng
- Department of Chemistry, Shanghai Key Laboratory of Molecular Catalysis, Advanced Materials Laboratory, Fudan University Shanghai 200433 China
| |
Collapse
|
24
|
Gao C, Wang Y, Sun J, Han Y, Gong W, Li Y, Feng Y, Wang H, Yang M, Li Z, Yang Y, Gao C. Neuronal mitochondria-targeted delivery of curcumin by biomimetic engineered nanosystems in Alzheimer's disease mice. Acta Biomater 2020; 108:285-299. [PMID: 32251785 DOI: 10.1016/j.actbio.2020.03.029] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 03/18/2020] [Accepted: 03/19/2020] [Indexed: 02/07/2023]
Abstract
Biomimetic nanotechnology represents a promising approach for the delivery of therapeutic agents for the treatment of complex diseases. Recently, neuronal mitochondria have been proposed to serve as a promising therapeutic target for sporadic Alzheimer's disease (AD). However, the efficient intravenous delivery of therapeutic agents to neuronal mitochondria in the brain remains a major challenge due to the complicated physiological and pathological environment. Herein, we devised and tested a strategy for functional antioxidant delivery to neuronal mitochondria by loading antioxidants into red blood cell (RBC) membrane-camouflaged human serum albumin nanoparticles bearing T807 and triphenylphosphine (TPP) molecules attached to the RBC membrane surface (T807/TPP-RBC-NPs). With the advantage of the suitable physicochemical properties of the nanoparticles and the unique biological functions of the RBC membrane, the T807/TPP-RBC-NPs are stabilized and promote sustained drug release, providing improved biocompatibility and long-term circulation. Under the synergistic effects of T807 and TPP, T807/TPP-RBC-NPs can not only penetrate the blood-brain barrier (BBB) but also target nerve cells and further localize in the mitochondria. After encapsulating curcumin (CUR) as the model antioxidant, the research data demonstrated that CUR-loaded T807/TPP-RBC-NPs can relieve AD symptoms by mitigating mitochondrial oxidative stress and suppressing neuronal death both in vitro and in vivo. In conclusion, the intravenous neuronal mitochondria-targeted biomimetic engineered delivery nanosystems provides an effective drug delivery platform for brain diseases. STATEMENT OF SIGNIFICANCE: The efficient intravenous delivery of therapeutic agents to neuronal mitochondria in the brain remains a major challenge for drug delivery due to the complicated physiological and pathological environment. To address this need, various types of nanovessels have been fabricated using a variety of materials in the last few decades. However, problems with the synthetic materials still exist and even cause toxicology issues. New findings in nanomedicine are promoting the development of biomaterials. Herein, we designed a red blood cell (RBC) membrane-coated human serum albumin nanoparticle dual-modified with T807 and TPP (T807/TPP-RBC-NPs) to accomplish these objectives. After encapsulating curcumin as the model drug, the research data demonstrated that the intravenous neuronal mitochondria-targeted biomimetic engineered delivery nanosystems are a promising therapeutic candidate for mitochondrial dysfunction in Alzheimer's disease (AD).
Collapse
|
25
|
Li Y, Wang S, Song FX, Zhang L, Yang W, Wang HX, Chen QL. A pH-sensitive drug delivery system based on folic acid-targeted HBP-modified mesoporous silica nanoparticles for cancer therapy. Colloids Surf A Physicochem Eng Asp 2020. [DOI: 10.1016/j.colsurfa.2020.124470] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
26
|
Zhang HT, Yu M, Niu YJ, Liu WZ, Pang WH, Ding J, Wang JC. Polyarginine-Mediated siRNA Delivery: A Mechanistic Study of Intracellular Trafficking of PCL-R15/siRNA Nanoplexes. Mol Pharm 2020; 17:1685-1696. [PMID: 32191042 DOI: 10.1021/acs.molpharmaceut.0c00120] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
As a cell-penetrating peptide, polyarginine is widely used in drug delivery systems based on its membrane permeation ability. Previously, we developed the mPEG-PLA-b-polyarginine(R15) triblock copolymer, which exhibited a high siRNA delivery efficiency both in vitro and in vivo. As a continued effort, here the amphiphilic diblock polymer PCL-R15 was synthesized as a simplified model to further elucidate the structure-activity relationship of arginine-based amphiphilic polymers as siRNA delivery systems, and the cellular trafficking mechanisms of the PCL-R15/siRNA nanoplexes were investigated to understand the interaction patterns between the nanoplexes and cells. Compared to the R15/siRNA complexes, the introduction of PCL moiety was found to result in the stronger interactions with cells and the enhanced transfection efficiency after the formation of condensed nanoplexes. Caveolae-mediated endocytosis and clathrin-mediated endocytosis were major routes for the internalization of PCL-R15/siRNA nanoplexes. The intracellular release of siRNA from nanoplexes was confirmed by fluorescence resonance energy transfer assay. It was also noticed that the internalized PCL-R15/siRNA nanoplexes were transported through digestive routes and trapped in lysosomes, which may be the bottleneck for efficient siRNA delivery of PCL-R15/siRNA nanoplexes. This study investigated the relationship between the polymer structure of PCL-R15 and the cellular interaction patterns, which may render implications on the rational design of polyarginine-based siRNA delivery systems.
Collapse
Affiliation(s)
- Hai-Tao Zhang
- Xiangya School of Pharmaceutical Sciences, Central South University, 172 Tongzipo Road, Changsha, Hunan 410013, P. R. China.,Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, XueYuan Rd 38, Haidian Dist, Beijing 100191, P. R. China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, University of South China, 28 Western Changshen Road, Hengyang, Hunan 421001, P. R. China
| | - Minzhi Yu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, XueYuan Rd 38, Haidian Dist, Beijing 100191, P. R. China
| | - Yu-Jie Niu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, XueYuan Rd 38, Haidian Dist, Beijing 100191, P. R. China
| | - Wei-Zhong Liu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, XueYuan Rd 38, Haidian Dist, Beijing 100191, P. R. China
| | - Wen-Hao Pang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, XueYuan Rd 38, Haidian Dist, Beijing 100191, P. R. China
| | - Jinsong Ding
- Xiangya School of Pharmaceutical Sciences, Central South University, 172 Tongzipo Road, Changsha, Hunan 410013, P. R. China
| | - Jian-Cheng Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, XueYuan Rd 38, Haidian Dist, Beijing 100191, P. R. China
| |
Collapse
|
27
|
Kamalov MI, Sadrieva GR, Pavlyuk AM, Salakhieva DV, Petrova NV, Abdullin TI. Synthesis and Characterization of Polyaspartic Acid-Histidine Conjugate as an Analog of Antioxidant Enzymes. APPL BIOCHEM MICRO+ 2019. [DOI: 10.1134/s0003683819050065] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
28
|
Augustine R, Kalva N, Kim HA, Zhang Y, Kim I. pH-Responsive Polypeptide-Based Smart Nano-Carriers for Theranostic Applications. Molecules 2019; 24:E2961. [PMID: 31443287 PMCID: PMC6719039 DOI: 10.3390/molecules24162961] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 08/12/2019] [Accepted: 08/13/2019] [Indexed: 02/07/2023] Open
Abstract
Smart nano-carriers have attained great significance in the biomedical field due to their versatile and interesting designs with different functionalities. The initial stages of the development of nanocarriers mainly focused on the guest loading efficiency, biocompatibility of the host and the circulation time. Later the requirements of less side effects with more efficacy arose by attributing targetability and stimuli-responsive characteristics to nano-carriers along with their bio- compatibility. Researchers are utilizing many stimuli-responsive polymers for the better release of the guest molecules at the targeted sites. Among these, pH-triggered release achieves increasing importance because of the pH variation in different organ and cancer cells of acidic pH. This specific feature is utilized to release the guest molecules more precisely in the targeted site by designing polymers having specific functionality with the pH dependent morphology change characteristics. In this review, we mainly concert on the pH-responsive polypeptides and some interesting nano-carrier designs for the effective theranostic applications. Also, emphasis is made on pharmaceutical application of the different nano-carriers with respect to the organ, tissue and cellular level pH environment.
Collapse
Affiliation(s)
- Rimesh Augustine
- BK 21 PLUS Center for Advanced Chemical Technology, Department of Polymer Science and Engineering, Pusan National University, Geumjeong-gu, Busan 46241, Korea
| | - Nagendra Kalva
- BK 21 PLUS Center for Advanced Chemical Technology, Department of Polymer Science and Engineering, Pusan National University, Geumjeong-gu, Busan 46241, Korea
| | - Ho An Kim
- BK 21 PLUS Center for Advanced Chemical Technology, Department of Polymer Science and Engineering, Pusan National University, Geumjeong-gu, Busan 46241, Korea
| | - Yu Zhang
- BK 21 PLUS Center for Advanced Chemical Technology, Department of Polymer Science and Engineering, Pusan National University, Geumjeong-gu, Busan 46241, Korea
| | - Il Kim
- BK 21 PLUS Center for Advanced Chemical Technology, Department of Polymer Science and Engineering, Pusan National University, Geumjeong-gu, Busan 46241, Korea.
| |
Collapse
|
29
|
Wang H, Ding S, Zhang Z, Wang L, You Y. Cationic micelle: A promising nanocarrier for gene delivery with high transfection efficiency. J Gene Med 2019; 21:e3101. [PMID: 31170324 DOI: 10.1002/jgm.3101] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 05/25/2019] [Accepted: 05/29/2019] [Indexed: 12/15/2022] Open
Abstract
Micelles have demonstrated an excellent ability to deliver several different types of therapeutic agents, including chemotherapy drugs, proteins, small-interfering RNA and DNA, into tumor cells. Cationic micelles, comprising self-assemblies of amphiphilic cationic polymers, have exhibited tremendous promise with respect to the delivery of therapy genes and gene transfection. To date, research in the field has focused on achieving an enhanced stability of the micellar assembly, prolonged circulation times and controlled release of the gene. This review focuses on the micelles as a nanosized carrier system for gene delivery, the system-related modifications for cytoplasm release, stability and biocompatibility, and clinic trials. In accordance with the development of synthetic chemistry and self-assembly technology, the structures and functionalities of micelles can be precisely controlled, and hence the synthetic micelles not only efficiently condense DNA, but also facilitate DNA endocytosis, endosomal escape, DNA uptake and nuclear transport, resulting in a comparable gene transfection of virus.
Collapse
Affiliation(s)
- Haili Wang
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui, China
| | - Shenggang Ding
- Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ze Zhang
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui, China
| | - Longhai Wang
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui, China
| | - Yezi You
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
30
|
Daniels AN, Singh M. Sterically stabilized siRNA:gold nanocomplexes enhance c-MYC silencing in a breast cancer cell model. Nanomedicine (Lond) 2019; 14:1387-1401. [PMID: 31166141 DOI: 10.2217/nnm-2018-0462] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To produce sterically stabilized and functionalized gold nanoparticles (AuNPs) for efficient delivery of siRNA for c-MYC silencing in vitro. Materials & methods: Synthesized AuNPs were functionalized with chitosan and PEG400 and PEG2000, morphologically and chemically characterized, and assessed for cytotoxicity and gene silencing in vitro. Results & discussion: AuNPs presented as spherical particles in the nanometer size range and successfully bound and protected the siRNA against degradation and were well tolerated in the breast adenocarcinoma (MCF-7) cell line. Nanoparticle-mediated gene knockdown studies revealed enhanced levels of c-MYC gene silencing with more than 90% reduction of MYC protein levels. Conclusion: These nanoformulations show enhanced potential for siRNA-mediated gene silencing in human breast cancer cells in vitro.
Collapse
Affiliation(s)
- Aliscia N Daniels
- Department of Biochemistry, Nano-Gene & Drug Delivery Laboratory, School of Life Sciences, College of Agriculture, Engineering & Science, University of KwaZulu-Natal (Westville Campus), Private Bag X54001, Durban 4000, Kwa-Zulu Natal, South Africa
| | - Moganavelli Singh
- Department of Biochemistry, Nano-Gene & Drug Delivery Laboratory, School of Life Sciences, College of Agriculture, Engineering & Science, University of KwaZulu-Natal (Westville Campus), Private Bag X54001, Durban 4000, Kwa-Zulu Natal, South Africa
| |
Collapse
|
31
|
Biocompatibility, biodegradation and biomedical applications of poly(lactic acid)/poly(lactic-co-glycolic acid) micro and nanoparticles. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2019. [DOI: 10.1007/s40005-019-00439-x] [Citation(s) in RCA: 187] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
32
|
Cui L, Wang Y, Liang M, Chu X, Fu S, Gao C, Liu Q, Gong W, Yang M, Li Z, Yu L, Yang C, Su Z, Xie X, Yang Y, Gao C. Dual-modified natural high density lipoprotein particles for systemic glioma-targeting drug delivery. Drug Deliv 2019; 25:1865-1876. [PMID: 30474437 PMCID: PMC6263114 DOI: 10.1080/10717544.2018.1519002] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Therapeutic outcome for the treatment of glioma was often limited due to the two barriers involved: the blood-brain barrier (BBB) and blood-brain tumor barrier (BBTB). Therefore, the development of nanocarriers that possess both BBB and BBTB permeability and glioma-targeting ability is of great importance for the chemotherapy of glioma. New frontiers in nanomedicine are advancing the research of new biomaterials. Here we constructed a natural high-density lipoprotein particle (HDL)-based drug delivery system with the dual-modification of T7 and dA7R peptide ligand (T7/dA7R-HDL) to achieve the above goals. HDL, the smallest lipoprotein, plays a biological role and is highly suitable as a platform for delivering imaging and therapeutic agents. T7 is a seven-peptide ligand of transferrin receptors (TfR) capable of circumventing the BBB and then targeting glioma. dA7R is a d-peptide ligand of vascular endothelial growth factor receptor 2 (VEGFR 2) overexpressed on angiogenesis, presenting excellent glioma-homing property. 10-Hydroxycamptothecin (HCPT), a hydrophobic anti-cancer drug, was used as the model drug in this study. By combining the dual-targeting delivery effect, the dual-modified HDL displayed higher glioma localization than that of single ligand-modified HDL or free HCPT. After loading with HCPT, T7/dA7R-HDL showed the most favorable anti-glioma effect in vivo. These results demonstrated that the dual-targeting natural nanocarriers strategy provides a potential method for improving brain drug delivery and anti-glioma treatment efficacy.
Collapse
Affiliation(s)
- Lin Cui
- a Jiamusi University , Jiamusi , China.,b State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Yuli Wang
- b State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Meng Liang
- b State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | | | - Shiyao Fu
- a Jiamusi University , Jiamusi , China.,b State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Chunsheng Gao
- b State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Qianqian Liu
- b State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Wei Gong
- b State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Meiyan Yang
- b State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Zhiping Li
- b State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Lian Yu
- a Jiamusi University , Jiamusi , China
| | | | - Zhide Su
- d Weifang People's Hospital , Weifang , China
| | - Xiangyang Xie
- e Department of Pharmacy , Wuhan General Hospital of the PLA , Wuhan , China
| | - Yang Yang
- b State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Chunsheng Gao
- b State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing , China
| |
Collapse
|
33
|
Liang M, Gao C, Wang Y, Gong W, Fu S, Cui L, Zhou Z, Chu X, Zhang Y, Liu Q, Zhao X, Zhao B, Yang M, Li Z, Yang C, Xie X, Yang Y, Gao C. Enhanced blood-brain barrier penetration and glioma therapy mediated by T7 peptide-modified low-density lipoprotein particles. Drug Deliv 2019; 25:1652-1663. [PMID: 30394123 PMCID: PMC6225487 DOI: 10.1080/10717544.2018.1494223] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Therapeutic outcome for the treatment of glioma was often limited due to the non-targeted nature and low permeability of drugs across the blood-brain barrier (BBB). An ideal glioma-targeted delivery system need to traverse the BBB and then target glioma cells with adequate optimized physiochemical properties and biocompatibility. However, it is an enormous challenge to the researchers to engineer the above-mentioned features into a single nanocarrier particle. New frontiers in nanomedicine are advancing the research of new biomaterials. In this study, we demonstrate a strategy for glioma targeting by encapsulating vincristine sulfate (VCR) into a naturally available low-density lipoprotein particles (LDL)-based drug delivery system with the modification of T7 peptide ligand (T7-LDL). LDL, endogenous lipid transporters, can specifically bind to brain endothelial cells and glioma cells via interacting with the low-density lipoprotein receptors (LDLR). T7 is a seven-peptide ligand of transferrin receptors (TfR) capable of circumventing the BBB and then targeting glioma. By combining the dual-targeting delivery effect of T7 peptide and parent LDL, T7-LDL displayed higher glioma localization than that of parent LDL. After loading with VCR, T7-LDL showed the most favorable antiglioma effect in vitro and in vivo. These results demonstrated that T7-LDL is an important potential drug delivery system for glioma-targeted therapy.
Collapse
Affiliation(s)
- Meng Liang
- a State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Chunhong Gao
- a State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Yuli Wang
- a State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Wei Gong
- a State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Shiyao Fu
- a State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing , China.,b Department of Pharmacy , Wuhan General Hospital of the PLA , Wuhan , China
| | - Lin Cui
- a State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing , China.,c Department of Pharmacy, Jiamusi University , Jiamusi , China
| | - Zhenhan Zhou
- a State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | | | - Yue Zhang
- a State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Qianqian Liu
- a State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Xiong Zhao
- e Beijing Institute of Health Service and Transfusion Medicine , Beijing , China
| | - Baoquan Zhao
- a State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Meiyan Yang
- a State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Zhiping Li
- a State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Chunrong Yang
- c Department of Pharmacy, Jiamusi University , Jiamusi , China
| | - Xiangyang Xie
- e Beijing Institute of Health Service and Transfusion Medicine , Beijing , China
| | - Yang Yang
- a State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Chunsheng Gao
- a State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing , China
| |
Collapse
|
34
|
Fu S, Liang M, Wang Y, Cui L, Gao C, Chu X, Liu Q, Feng Y, Gong W, Yang M, Li Z, Yang C, Xie X, Yang Y, Gao C. Dual-Modified Novel Biomimetic Nanocarriers Improve Targeting and Therapeutic Efficacy in Glioma. ACS APPLIED MATERIALS & INTERFACES 2019; 11:1841-1854. [PMID: 30582685 DOI: 10.1021/acsami.8b18664] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Glioma is a fatal disease with limited treatment options and very short survival. Although chemotherapy is one of the most important strategies in glioma treatment, it remains extremely clinically challenging largely due to the blood-brain barrier (BBB) and the blood-brain tumor barrier (BBTB). Thus, the development of nanoparticles with both BBB and BBTB penetrability, as well as glioma-targeting feature, is extremely important for the therapy of glioma. New findings in nanomedicine are promoting the development of novel biomaterials. Herein, we designed a red blood cell membrane-coated solid lipid nanoparticle (RBCSLN)-based nanocarrier dual-modified with T7 and NGR peptide (T7/NGR-RBCSLNs) to accomplish these objectives. As a new kind of biomimetic nanovessels, RBCSLNs preserve the complex biological functions of natural cell membranes while possessing physicochemical properties that are needed for efficient drug delivery. T7 is a ligand of transferrin receptors with seven peptides that is able to circumvent the BBB and target to glioma. NGR is a peptide ligand of CD13 that is overexpressed during angiogenesis, representing an excellent glioma-homing property. After encapsulating vinca alkaloid vincristine as the model drug, T7/NGR-RBCSLNs exhibited the most favorable antiglioma effects in vitro and in vivo by combining the dual-targeting delivery effect. The results demonstrate that dual-modified biomimetic nanoparticles provide a potential method to improve drug delivery to the brain, hence increasing glioma therapy efficacy.
Collapse
Affiliation(s)
- Shiyao Fu
- State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing 100850 , China
- Jiamusi University , Jiamusi 154002 , China
| | - Meng Liang
- State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing 100850 , China
| | - Yuli Wang
- State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing 100850 , China
| | - Lin Cui
- State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing 100850 , China
- Jiamusi University , Jiamusi 154002 , China
| | - Chunhong Gao
- State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing 100850 , China
| | - Xiaoyang Chu
- 307 Hospital of the PLA , Beijing 100071 , China
| | - Qianqian Liu
- State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing 100850 , China
| | - Ye Feng
- State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing 100850 , China
| | - Wei Gong
- State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing 100850 , China
| | - Meiyan Yang
- State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing 100850 , China
| | - Zhiping Li
- State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing 100850 , China
| | | | - Xiangyang Xie
- General Hospital of Central Theater of the PLA , Wuhan 430070 , China
| | - Yang Yang
- State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing 100850 , China
| | - Chunsheng Gao
- State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing 100850 , China
| |
Collapse
|
35
|
Lin G, Li L, Panwar N, Wang J, Tjin SC, Wang X, Yong KT. Non-viral gene therapy using multifunctional nanoparticles: Status, challenges, and opportunities. Coord Chem Rev 2018. [DOI: 10.1016/j.ccr.2018.07.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
36
|
Polyester based nanovehicles for siRNA delivery. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 92:1006-1015. [DOI: 10.1016/j.msec.2018.05.031] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 02/12/2018] [Accepted: 05/07/2018] [Indexed: 12/18/2022]
|
37
|
Polyester-based nanoparticles for nucleic acid delivery. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 92:983-994. [DOI: 10.1016/j.msec.2018.07.027] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 07/05/2018] [Accepted: 07/11/2018] [Indexed: 12/14/2022]
|
38
|
Zhou S, Sun W, Zhai Y. Amphiphilic block copolymer NPs obtained by coupling ricinoleic acid/sebacic acids and mPEG: Synthesis, characterization, and controlled release of paclitaxel. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2018; 29:2201-2217. [PMID: 30285542 DOI: 10.1080/09205063.2018.1532136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Currently, nanoparticles (NPs) made of amphiphilic block copolymer are still an essential part of drug delivery system. Here, we report a novel amphiphilic block copolymer and paclitaxel (PTX)-loaded copolymer NPs for the controlled delivery of PTX. The block copolymer was synthesized via melt polycondensation method of methoxy poly(ethylene glycol) (mPEG), sebacic acid (SA) and ricinoleic acid (RA). A series of characterization approaches such as Fourier Transform Infrared Spectroscopy (FTIR), 1Hydrogen-Nuclear Magnetic Resonance (1H-NMR), Differential Scanning Calorimetry (DSC), X-Ray Diffraction (XRD) and Gel Permeation Chromatography (GPC) applied have shown that the amphiphilic block copolymer was prepared as designed. NPs prepared by nanoprecipitation method consist of mPEG segments as the hydrophilic shell and RA-SA segments as the hydrophobic core, hydrophobic PTX was encapsulated as model drug. Subsequently, Transmission Electron Microscopy (TEM) analysis indicated that the spherical NPs have effective mean diameters ranging from 100 to 400 nm. Dynamic Light Scattering (DLS) analysis also revealed the controllable NPs diameter by modulating the mass ratio of RA to SA and drug loading amount (DLA). Besides, biphasic profile with zero order drug release was observed in general in vitro release behaviors of PTX from NPs. Further investigation confirmed that the release behaviors depend on the crystallinity of hydrophobic RA-SA segments. Results above suggest that NPs with amphiphlic block copolymer mPEG-b-P(RA-SA)-b-mPEG have a remarkable potential as a carrier for hydrophobic drug delivery in cancer therapy.
Collapse
Affiliation(s)
- Shiya Zhou
- a School of Pharmacy , Shenyang Pharmaceutical University , Shenyang , China
| | - Wei Sun
- b School of Medical Devices , Shenyang Pharmaceutical University , Shenyang , China
| | - Yinglei Zhai
- b School of Medical Devices , Shenyang Pharmaceutical University , Shenyang , China
| |
Collapse
|
39
|
Zhai M, Wang Y, Zhang L, Liang M, Fu S, Cui L, Yang M, Gong W, Li Z, Yu L, Xie X, Yang C, Yang Y, Gao C. Glioma targeting peptide modified apoferritin nanocage. Drug Deliv 2018; 25:1013-1024. [PMID: 29726297 PMCID: PMC6058491 DOI: 10.1080/10717544.2018.1464082] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Therapeutic outcome for the treatment of glioma was often limited due to the non-targeted nature of drugs and the physiological barriers, including the blood-brain barrier (BBB) and the blood-brain tumor barrier (BBTB). An ideal glioma-targeted delivery system must be sufficiently potent to cross the BBB and BBTB and then target glioma cells with adequate optimized physiochemical properties and biocompatibility. However, it is an enormous challenge to the researchers to engineer the above-mentioned features into a single nanocarrier particle. New frontiers in nanomedicine are advancing the research of new biomaterials. In this study, we demonstrate a strategy for glioma targeting by encapsulating vincristine sulfate (VCR) into a naturally available apoferritin nanocage-based drug delivery system with the modification of GKRK peptide ligand (GKRK-APO). Apoferritin (APO), an endogenous nanosize spherical protein, can specifically bind to brain endothelial cells and glioma cells via interacting with the transferrin receptor 1 (TfR1). GKRK is a peptide ligand of heparan sulfate proteoglycan (HSPG) over-expressed on angiogenesis and glioma, presenting excellent glioma-homing property. By combining the dual-targeting delivery effect of GKRK peptide and parent APO, GKRK-APO displayed higher glioma localization than that of parent APO. After loading with VCR, GKRK-APO showed the most favorable antiglioma effect in vitro and in vivo. These results demonstrated that GKRK-APO is an important potential drug delivery system for glioma-targeted therapy.
Collapse
Affiliation(s)
- Meifang Zhai
- a College of Pharmacy of Jiamusi University , Jiamusi Heilongjiang , China.,b State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Yuli Wang
- b State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Ligang Zhang
- d Clinical Department, Beijing Huilongguan Hospital , Beijng , China
| | - Meng Liang
- b State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Shiyao Fu
- a College of Pharmacy of Jiamusi University , Jiamusi Heilongjiang , China.,b State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Lin Cui
- a College of Pharmacy of Jiamusi University , Jiamusi Heilongjiang , China.,b State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Meiyan Yang
- b State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Wei Gong
- b State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Zhiping Li
- b State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Lian Yu
- a College of Pharmacy of Jiamusi University , Jiamusi Heilongjiang , China
| | - Xiangyang Xie
- c Department of Pharmacy , Wuhan General Hospital of the PLA , Wuhan , China
| | - Chunrong Yang
- a College of Pharmacy of Jiamusi University , Jiamusi Heilongjiang , China
| | - Yang Yang
- b State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing , China
| | - Chunsheng Gao
- b State Key Laboratory of Toxicology and Medical Countermeasures , Beijing Institute of Pharmacology and Toxicology , Beijing , China
| |
Collapse
|
40
|
Yang Y, Meng Y, Ye J, Xia X, Wang H, Li L, Dong W, Jin D, Liu Y. Sequential delivery of VEGF siRNA and paclitaxel for PVN destruction, anti-angiogenesis, and tumor cell apoptosis procedurally via a multi-functional polymer micelle. J Control Release 2018; 287:103-120. [PMID: 30144476 DOI: 10.1016/j.jconrel.2018.08.028] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 08/02/2018] [Accepted: 08/20/2018] [Indexed: 01/24/2023]
Abstract
Co-delivery of chemotherapy drugs and VEGF siRNA (siVEGF) to control tumor growth has been a research hotspot for improving cancer treatment. Current systems co-deliver siVEGF and chemo drugs into tumor cells simultaneously. Although effective, these systems do not flow to the abnormal blood vessels around tumor cells (vascular niche, PVN), which play an important role in the metastasis and deterioration of the tumor. Thus, we custom-synthesized triblock copolymer poly(ε-caprolactone)-polyethyleneglycol-poly(L-histidine) (PCL-PEG-PHIS) with previously synthesized folate-PEG-PHIS to construct a targeted multifunctional polymer micelle (PTX/siVEGF-CPPs/TMPM) to sequentially deliver siVEGF-CPPs (disulfide bond-linked siVEGF and cell-penetrating peptides) and paclitaxel (PTX). The sequential delivery vesicles showed the anticipated three-layered TEM structure and dual-convertible (surface charge- and particle size-reversible) features in the tumor environment (pH 6.5), which guaranteed the sequential release of siVEGF-CPPs and PTX in the tumor extracellular environment and tumor cells, respectively. To mimic the in vivo tumor environment, a double cell model was employed by co-culturing HUVECs and MCF-7 cells. Improved cell endocytosis efficiency, VEGF gene silence efficacy, and in vitro anti-proliferation activity were achieved. An in vivo study on MCF-7 tumor-bearing female nude mice also indicated that sequential delivery vesicles could lead to significant induction of tumor cell apoptosis, loss of VEGF expression, and destruction of tumor blood vessels (PVN and neovascularization). These sequential delivery vesicles show potential as an effective co-delivery platform for siVEGF and chemo drugs to improve cancer therapy efficacy.
Collapse
Affiliation(s)
- Yanfang Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China; Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Yingying Meng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China; Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Jun Ye
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China; Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Xuejun Xia
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China; Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Hongliang Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China; Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Lin Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China; Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Wujun Dong
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China; Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Dujia Jin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China; Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Yuling Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China; Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China.
| |
Collapse
|
41
|
Ben Djemaa S, David S, Hervé-Aubert K, Falanga A, Galdiero S, Allard-Vannier E, Chourpa I, Munnier E. Formulation and in vitro evaluation of a siRNA delivery nanosystem decorated with gH625 peptide for triple negative breast cancer theranosis. Eur J Pharm Biopharm 2018; 131:99-108. [PMID: 30063968 DOI: 10.1016/j.ejpb.2018.07.024] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 07/24/2018] [Indexed: 12/12/2022]
Abstract
The development of an efficient small interfering RNA (siRNA) delivery system has held scientists interest since the discovery of the RNA interference mechanism (RNAi). This strategy gives hope for the treatment of many severe diseases. Herein, we developed hybrid nanovectors able to deliver siRNA to triple negative breast cancer cells. The nanovectors are based on PEGylated superparamagnetic iron oxide nanoparticles (SPION) functionalized with gH625 peptide, chitosan and poly-l-arginine. Every component has a key role and specific function: SPION is the core scaffolding the nanovector; PEG participates in the colloidal stability and the immune stealthiness; gH625 peptide promotes the nanovector internalization into cancer cells; cationic polymers provide the siRNA protection and favor siRNA endosomal escape and delivery to cytosol. The formulation was optimized by varying the amount of each compound. The efficacy of the siRNA retention and protection were investigated in the presence of high concentration of serum. Optimized nanovectors show a high uptake by MDA-MB-231 cells. The resulting down regulation of GFP expression was 73 ± 3% with our nanovector compared to 59 ± 8% obtained with the siRNA-Oligofectamine™ complex in the same conditions.
Collapse
Affiliation(s)
- Sanaa Ben Djemaa
- EA6295 Nanomédicaments et Nanosondes, Université de Tours, 31 Avenue Monge, 37200 Tours, France
| | - Stephanie David
- EA6295 Nanomédicaments et Nanosondes, Université de Tours, 31 Avenue Monge, 37200 Tours, France.
| | - Katel Hervé-Aubert
- EA6295 Nanomédicaments et Nanosondes, Université de Tours, 31 Avenue Monge, 37200 Tours, France
| | - Annarita Falanga
- Department of Pharmacy, CIRPEB - University of Naples "Federico II", Via Mezzocannone 16, 80134 Napoli, Italy
| | - Stefania Galdiero
- Department of Pharmacy, CIRPEB - University of Naples "Federico II", Via Mezzocannone 16, 80134 Napoli, Italy
| | - Emilie Allard-Vannier
- EA6295 Nanomédicaments et Nanosondes, Université de Tours, 31 Avenue Monge, 37200 Tours, France
| | - Igor Chourpa
- EA6295 Nanomédicaments et Nanosondes, Université de Tours, 31 Avenue Monge, 37200 Tours, France
| | - Emilie Munnier
- EA6295 Nanomédicaments et Nanosondes, Université de Tours, 31 Avenue Monge, 37200 Tours, France
| |
Collapse
|
42
|
Kara A, Ozturk N, Esendagli G, Ozkose UU, Gulyuz S, Yilmaz O, Telci D, Bozkir A, Vural I. Development of novel self-assembled polymeric micelles from partially hydrolysed poly(2-ethyl-2-oxazoline)-co-PEI-b-PCL block copolymer as non-viral vectors for plasmid DNA in vitro transfection. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:S264-S273. [DOI: 10.1080/21691401.2018.1491478] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Asli Kara
- Department of Biology, Faculty of Art and Science, Hitit University, Corum, Turkey
- Department of Nanotechnology and Nanomedicine, Hacettepe University Institute of Science, Ankara, Turkey
| | - Naile Ozturk
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Gunes Esendagli
- Department of Basic Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Umut Ugur Ozkose
- Materials Institute, Marmara Research Center, TUBITAK, Gebze, Turkey
- Department of Chemistry, Faculty of Science and Letters, Istanbul Technical University, Istanbul, Turkey
- Department of Chemistry, Faculty of Science and Letters, Piri Reis University, Istanbul, Turkey
| | - Sevgi Gulyuz
- Materials Institute, Marmara Research Center, TUBITAK, Gebze, Turkey
- Department of Chemistry, Faculty of Science and Letters, Istanbul Technical University, Istanbul, Turkey
| | - Ozgur Yilmaz
- Materials Institute, Marmara Research Center, TUBITAK, Gebze, Turkey
| | - Dilek Telci
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Asuman Bozkir
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| | - Imran Vural
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| |
Collapse
|
43
|
miR-145-loaded micelleplexes as a novel therapeutic strategy to inhibit proliferation and migration of osteosarcoma cells. Eur J Pharm Sci 2018; 123:28-42. [PMID: 30010029 DOI: 10.1016/j.ejps.2018.07.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 06/12/2018] [Accepted: 07/09/2018] [Indexed: 12/20/2022]
Abstract
Osteosarcoma (OS), the main primary malignancy of bone, is the second leading cause of cancer in children and young adults. Despite the advances in modern treatments, the 5-year survival rate is retained in 60-70%, since the conventional treatment options available are associated with relapse, chemoresistance, and development of metastases, which frequently lead to patients death. In this regard, there is an increasing need to search and develop novel and alternative therapeutic approaches. Concerning this, gene therapy appears as an innovative and promising treatment option. This therapeutic option aims to deliver genetic material, through nanosystems, to repress or replace the expression of mutated genes involved in important regulatory pathways. To attain this goal, gene therapy is decidedly dependent on the efficiency of utilized vectors, constituting such a very important parameter to take in consideration. In this work, the main goal was centered on the development and full characterization of an efficient micellar nanosystem, based on the chemical conjugation between the amphiphilic copolymer Pluronic® L64 and the cationic polymer polyethyleneimine (PEI), to deliver the therapeutic miRNA-145 into OS cells leading to inhibition of cell proliferation and migration, and ultimately inducing cell death, crafting a novel anticancer therapeutic approach to OS.
Collapse
|
44
|
Hu C, Gu F, Tai Z, Yao C, Gong C, Xia Q, Gao Y, Gao S. Synergistic effect of reduced polypeptide micelle for co-delivery of doxorubicin and TRAIL against drug-resistance in breast cancer. Oncotarget 2018; 7:61832-61844. [PMID: 27557520 PMCID: PMC5308694 DOI: 10.18632/oncotarget.11451] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 08/13/2016] [Indexed: 11/25/2022] Open
Abstract
Cationic peptides as a non-viral gene vector have become a hotspot of research because of their high transfection efficcacy and safety. Based on our previous study, we synthesized a cationic reduction-responsive vector based on disulfide cross-linked L-arginine, L-histidine and lipoic acid (LHRss) as the co-carrier of both doxorubicin (DOX) and the necrosis factor-related apoptosis-inducing ligand (pTRAIL). The LHRss/DOX/TRAIL construct has reduction-sensitive behavior and an enhanced endosomal escape ability to increase the cytotoxicity of DOX and the transfection efficiency. Further, the LHRss/DOX/TRAIL construct increased the accumulation of DOX and promoted the expression of pTRAIL, thus increasing cellular apoptosis by 83.7% in MCF-7/ADR cells. In addition, the in vivo biodistribution results showed that the LHRss/DOX/TRAIL construct could target tumors well. The in vivo anti-tumor effect study demonstrated that the LHRss/DOX/TRAIL construct inhibited tumor growth markedly, with a tumor inhibitory rate of 94.0%. The co-delivery system showed a significant synergistic anti-tumor effect. The LHRss/DOX/TRAIL construct may prove to be a promising co-delivery vector for the effective treatment of drug resistant breast cancer.
Collapse
Affiliation(s)
- Chuling Hu
- Department of Pharmaceutics, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Fenfen Gu
- Department of Pharmaceutics, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Zongguang Tai
- Department of Pharmaceutics, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Chong Yao
- Department of Pharmaceutics, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Chunai Gong
- Department of Pharmaceutics, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Qingming Xia
- Department of Pharmaceutics, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Yuan Gao
- Department of Pharmaceutics, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Shen Gao
- Department of Pharmaceutics, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
45
|
Wang J, Li S, Han Y, Guan J, Chung S, Wang C, Li D. Poly(Ethylene Glycol)-Polylactide Micelles for Cancer Therapy. Front Pharmacol 2018; 9:202. [PMID: 29662450 PMCID: PMC5890116 DOI: 10.3389/fphar.2018.00202] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 02/22/2018] [Indexed: 12/21/2022] Open
Abstract
For the treatment of malignancy, many therapeutic agents, including small molecules, photosensitizers, immunomodulators, proteins and genes, and so forth, have been loaded into nanocarriers for controllable cancer therapy. Among these nanocarriers, polymeric micelles have been considered as one of the most promising nanocarriers, some of which have already been applied in different stages of clinical trials. The successful advantages of polymeric micelles from bench to bedside are due to their special core/shell structures, which can carry specific drugs in certain disease conditions. Particularly, poly(ethylene glycol)–polylactide (PEG–PLA) micelles have been considered as one of the most promising platforms for drug delivery. The PEG shell effectively prevents the adsorption of proteins and phagocytes, thereby evidently extending the blood circulation period. Meanwhile, the hydrophobic PLA core can effectively encapsulate many therapeutic agents. This review summarizes recent advances in PEG–PLA micelles for the treatment of malignancy. In addition, future perspectives for the development of PEG–PLA micelles as drug delivery systems are also presented.
Collapse
Affiliation(s)
- Jixue Wang
- Department of Urology, The First Hospital of Jilin University, Changchun, China.,Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Shengxian Li
- Department of Urology, The First Hospital of Jilin University, Changchun, China.,Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Yuping Han
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jingjing Guan
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Shirley Chung
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - Chunxi Wang
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Di Li
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| |
Collapse
|
46
|
Zong L, Li X, Wang H, Cao Y, Yin L, Li M, Wei Z, Chen D, Pu X, Han J. Formulation and characterization of biocompatible and stable I.V. itraconazole nanosuspensions stabilized by a new stabilizer polyethylene glycol-poly(β-Benzyl- l -aspartate) (PEG-PBLA). Int J Pharm 2017; 531:108-117. [DOI: 10.1016/j.ijpharm.2017.08.082] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 07/29/2017] [Accepted: 08/15/2017] [Indexed: 10/19/2022]
|
47
|
Chen Z, Zhai M, Xie X, Zhang Y, Ma S, Li Z, Yu F, Zhao B, Zhang M, Yang Y, Mei X. Apoferritin Nanocage for Brain Targeted Doxorubicin Delivery. Mol Pharm 2017; 14:3087-3097. [PMID: 28728419 DOI: 10.1021/acs.molpharmaceut.7b00341] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
An ideal brain-targeted nanocarrier must be sufficiently potent to penetrate the blood-brain barrier (BBB) and sufficiently competent to target the cells of interest with adequate optimized physiochemical features and biocompatibility. However, it is an enormous challenge to the researchers to organize the above-mentioned properties into a single nanocarrier particle. New frontiers in nanomedicine are advancing the research of new biomaterials. Herein, we demonstrate a straightforward strategy for brain targeting by encapsulating doxorubicin (DOX) into a naturally available and unmodified apoferritin nanocage (DOX-loaded APO). APO can specifically bind to cells expressing transferrin receptor 1 (TfR1). Because of the high expression of TfR1 in both brain endothelial and glioma cells, DOX-loaded APO can cross the BBB and deliver drugs to the glioma with TfR1. Subsequent research demonstrated that the DOX-loaded APO had good physicochemical properties (particle size of 12.03 ± 0.42 nm, drug encapsulation efficiency of 81.8 ± 1.1%) and significant penetrating and targeting effects in the coculture model of bEnd.3 and C6 cells in vitro. In vivo imaging revealed that DOX-loaded APO accumulated specifically in brain tumor tissues. Additionally, in vivo tumor therapy experiments (at a dosage of 1 mg/kg DOX) demonstrated that a longer survival period was observed in mice that had been treated with DOX-loaded APO (30 days) compared with mice receiving free DOX solution (19 days).
Collapse
Affiliation(s)
- Zhijiang Chen
- State Key Laboratory of Toxicology and Medical Countermeasure , Beijing 100850, China.,Beijing Institute of Pharmacology and Toxicology , Beijing 100850, China.,Hubei University of Science and Technology , Xianning 437100, China
| | - Meifang Zhai
- State Key Laboratory of Toxicology and Medical Countermeasure , Beijing 100850, China.,Beijing Institute of Pharmacology and Toxicology , Beijing 100850, China.,Jiamusi University , Jiamusi 154002, China
| | | | - Yue Zhang
- State Key Laboratory of Toxicology and Medical Countermeasure , Beijing 100850, China.,Beijing Institute of Pharmacology and Toxicology , Beijing 100850, China.,Wuhan General Hospital of PLA , Wuhan 430070, China
| | - Siyu Ma
- State Key Laboratory of Toxicology and Medical Countermeasure , Beijing 100850, China.,Beijing Institute of Pharmacology and Toxicology , Beijing 100850, China
| | - Zhiping Li
- State Key Laboratory of Toxicology and Medical Countermeasure , Beijing 100850, China.,Beijing Institute of Pharmacology and Toxicology , Beijing 100850, China
| | - Fanglin Yu
- State Key Laboratory of Toxicology and Medical Countermeasure , Beijing 100850, China.,Beijing Institute of Pharmacology and Toxicology , Beijing 100850, China
| | - Baoquan Zhao
- State Key Laboratory of Toxicology and Medical Countermeasure , Beijing 100850, China.,Beijing Institute of Pharmacology and Toxicology , Beijing 100850, China
| | - Min Zhang
- State Key Laboratory of Toxicology and Medical Countermeasure , Beijing 100850, China.,Beijing Institute of Pharmacology and Toxicology , Beijing 100850, China
| | - Yang Yang
- State Key Laboratory of Toxicology and Medical Countermeasure , Beijing 100850, China.,Beijing Institute of Pharmacology and Toxicology , Beijing 100850, China
| | - Xingguo Mei
- State Key Laboratory of Toxicology and Medical Countermeasure , Beijing 100850, China.,Beijing Institute of Pharmacology and Toxicology , Beijing 100850, China
| |
Collapse
|
48
|
Engineered polymeric nanoparticles to guide the cellular internalization and trafficking of small interfering ribonucleic acids. J Control Release 2017; 259:3-15. [DOI: 10.1016/j.jconrel.2017.02.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 02/15/2017] [Accepted: 02/18/2017] [Indexed: 12/29/2022]
|
49
|
Recent advance of pH-sensitive nanocarriers targeting solid tumors. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2017. [DOI: 10.1007/s40005-017-0349-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
50
|
Omar R, Yang J, Liu H, Davies NM, Gong Y. Hepatic Stellate Cells in Liver Fibrosis and siRNA-Based Therapy. Rev Physiol Biochem Pharmacol 2017; 172:1-37. [PMID: 27534415 DOI: 10.1007/112_2016_6] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Hepatic fibrosis is a reversible wound-healing response to either acute or chronic liver injury caused by hepatitis B or C, alcohol, and toxic agents. Hepatic fibrosis is characterized by excessive accumulation and reduced degradation of extracellular matrix (ECM). Excessive accumulation of ECM alters the hepatic architecture leading to liver fibrosis and cirrhosis. Cirrhosis results in failure of common functions of the liver. Hepatic stellate cells (HSC) play a major role in the development of liver fibrosis as HSC are the main source of the excessive production of ECM in an injured liver. RNA interference (RNAi) is a recently discovered therapeutic tool that may provide a solution to manage multiple diseases including liver fibrosis through silencing of specific gene expression in diseased cells. However, gene silencing using small interfering RNA (siRNA) is encountering many challenges in the body after systemic administration. Efficient and stable siRNA delivery to the target cells is a key issue for the development of siRNA therapeutic. For that reason, various viral and non-viral carriers for liver-targeted siRNA delivery have been developed. This review will cover the current strategies for the treatment of liver fibrosis as well as discussing non-viral approaches such as cationic polymers and lipid-based nanoparticles for targeted delivery of siRNA to the liver.
Collapse
Affiliation(s)
- Refaat Omar
- College of Pharmacy, Faculty of Health Sciences, University of Manitoba, 750 McDermot Avenue, Winnipeg, MB, Canada, R3E 0T5
| | - Jiaqi Yang
- College of Pharmacy, Faculty of Health Sciences, University of Manitoba, 750 McDermot Avenue, Winnipeg, MB, Canada, R3E 0T5
| | - Haoyuan Liu
- College of Pharmacy, Faculty of Health Sciences, University of Manitoba, 750 McDermot Avenue, Winnipeg, MB, Canada, R3E 0T5
| | - Neal M Davies
- College of Pharmacy, Faculty of Health Sciences, University of Manitoba, 750 McDermot Avenue, Winnipeg, MB, Canada, R3E 0T5
- Faculty of Pharmacy & Pharmaceutical Sciences, University of Alberta, 8613-114 Street, Edmonton, AB, Canada, T6G 2H1
| | - Yuewen Gong
- College of Pharmacy, Faculty of Health Sciences, University of Manitoba, 750 McDermot Avenue, Winnipeg, MB, Canada, R3E 0T5.
| |
Collapse
|