1
|
Neill B, Romero AR, Fenton OS. Advances in Nonviral mRNA Delivery Materials and Their Application as Vaccines for Melanoma Therapy. ACS APPLIED BIO MATERIALS 2024; 7:4894-4913. [PMID: 37930174 PMCID: PMC11220486 DOI: 10.1021/acsabm.3c00721] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
Messenger RNA (mRNA) vaccines are promising platforms for cancer immunotherapy because of their potential to encode for a variety of tumor antigens, high tolerability, and capacity to induce strong antitumor immune responses. However, the clinical translation of mRNA cancer vaccines can be hindered by the inefficient delivery of mRNA in vivo. In this review, we provide an overview of mRNA cancer vaccines by discussing their utility in treating melanoma. Specifically, we begin our review by describing the barriers that can impede mRNA delivery to target cells. We then review native mRNA structure and discuss various modification methods shown to enhance mRNA stability and transfection. Next, we outline the advantages and challenges of three nonviral carrier platforms (lipid nanoparticles, polymeric nanoparticles, and lipopolyplexes) frequently used for mRNA delivery. Last, we summarize preclinical and clinical studies that have investigated nonviral mRNA vaccines for the treatment of melanoma. In writing this review, we aim to highlight innovative nonviral strategies designed to address mRNA delivery challenges while emphasizing the exciting potential of mRNA vaccines as next-generation therapies for the treatment of cancers.
Collapse
Affiliation(s)
- Bevin Neill
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Adriana Retamales Romero
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Owen S. Fenton
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| |
Collapse
|
2
|
Wu Y, Wang Y, Chen F, Wang B. Loading rutin on surfaces by the layer-by-layer assembly technique to improve the oxidation resistance and osteogenesis of titanium implants in osteoporotic rats. Biomed Mater 2024; 19:045011. [PMID: 38740037 DOI: 10.1088/1748-605x/ad4aa8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 05/13/2024] [Indexed: 05/16/2024]
Abstract
The purpose of this study was to construct a rutin-controlled release system on the surface of Ti substrates and investigate its effects on osteogenesis and osseointegration on the surface of implants. The base layer, polyethylenimine (PEI), was immobilised on a titanium substrate. Then, hyaluronic acid (HA)/chitosan (CS)-rutin (RT) multilayer films were assembled on the PEI using layer-by-layer (LBL) assembly technology. We used scanning electron microscopy (SEM), Fourier transform infrared (FTIR) spectroscopy and contact angle measurements to examine all Ti samples. The drug release test of rutin was also carried out to detect the slow-release performance. The osteogenic abilities of the samples were evaluated by experiments on an osteoporosis rat model and MC3T3-E1 cells. The results (SEM, FTIR and contact angle measurements) all confirmed that the PEI substrate layer and HA/CS-RT multilayer film were effectively immobilised on titanium. The drug release test revealed that a rutin controlled release mechanism had been successfully established. Furthermore, thein vitrodata revealed that osteoblasts on the coated titanium matrix had greater adhesion, proliferation, and differentiation capacity than the osteoblasts on the pure titanium surface. When MC3T3-E1 cells were exposed to H2O2-induced oxidative stressin vitro, cell-based tests revealed great tolerance and increased osteogenic potential on HA/CS-RT substrates. We also found that the HA/CS-RT coating significantly increased the new bone mass around the implant. The LBL-deposited HA/CS-RT multilayer coating on the titanium base surface established an excellent rutin-controlled release system, which significantly improved osseointegration and promoted osteogenesis under oxidative stress conditions, suggesting a new implant therapy strategy for patients with osteoporosis.
Collapse
Affiliation(s)
- Yinsheng Wu
- Department of Orthopedics, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, No. 75, JinXiu Road, Lucheng District, Wenzhou 325000, Zhejiang Province, People's Republic of China
| | - Yong Wang
- Department of Orthopedics, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, No. 75, JinXiu Road, Lucheng District, Wenzhou 325000, Zhejiang Province, People's Republic of China
| | - Fengyan Chen
- Department of Orthopedics, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, No. 75, JinXiu Road, Lucheng District, Wenzhou 325000, Zhejiang Province, People's Republic of China
| | - Bingzhang Wang
- Department of Orthopedics, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, No. 75, JinXiu Road, Lucheng District, Wenzhou 325000, Zhejiang Province, People's Republic of China
| |
Collapse
|
3
|
Khanam A, Singh G, Narwal S, Chopra B, Dhingra AK. A Review on Novel Applications of Nanotechnology in the Management of Prostate Cancer. Curr Drug Deliv 2024; 21:1161-1179. [PMID: 37888818 DOI: 10.2174/0115672018180695230925113521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/09/2023] [Accepted: 07/26/2023] [Indexed: 10/28/2023]
Abstract
BACKGROUND Prostate cancer continues to be a serious danger to men's health, despite advances in the field of cancer nanotechnology. Although different types of cancer have been studied using nanomaterials and theranostic systems derived from nanomaterials, they have not yet reached their full potential for prostate cancer due to issues with in vivo biologic compatibility, immune reaction responses, accurate targetability, as well as a therapeutic outcome related to the nano-structured mechanism. METHOD The ultimate motive of this article is to understand the theranostic nanotechnology-based scheme for treating prostate cancer. The categorization of diverse nanomaterials in accordance with biofunctionalization tactics and biomolecule sources has been emphasized in this review so that they might potentially be used in clinical contexts and future advances. These opportunities can enhance the direct visualization of prostate tumors, early identification of prostate cancer-associated biomarkers at extremely low detection limits, and finally, the therapy for prostate cancer. RESULT In December 2022, a thorough examination of the scientific literature was carried out utilizing the Web of Science, PubMed, and Medline databases. The goal was to analyze novel applications of nanotechnology in the treatment of prostate cancer, together with their structural layouts and functionalities. CONCLUSION The various treatments and the reported revolutionary nanotechnology-based systems appear to be precise, safe, and generally successful; as a result, this might open up a new avenue for the detection and eradication of prostate cancer.
Collapse
Affiliation(s)
- Arshi Khanam
- Institute of Pharmaceutical Sciences, Kurukshetra University Kurukshetra-136119, Haryana, India
| | - Gurvirender Singh
- Institute of Pharmaceutical Sciences, Kurukshetra University Kurukshetra-136119, Haryana, India
| | - Smita Narwal
- Global Research Institute of Pharmacy, Radaur, Yamunanagar-135133, Haryana, India
| | - Bhawna Chopra
- Guru Gobind Singh College of Pharmacy, Yamuna Nagar-135001, Haryana, India
| | - Ashwani K Dhingra
- Guru Gobind Singh College of Pharmacy, Yamuna Nagar-135001, Haryana, India
| |
Collapse
|
4
|
Qin Y, Ou L, Zha L, Zeng Y, Li L. Delivery of nucleic acids using nanomaterials. MOLECULAR BIOMEDICINE 2023; 4:48. [PMID: 38092998 PMCID: PMC10719232 DOI: 10.1186/s43556-023-00160-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 11/21/2023] [Indexed: 12/17/2023] Open
Abstract
The increasing number of approved nucleic acid therapeutics demonstrates the potential for the prevention and treatment of a broad spectrum of diseases. This trend underscores the significant impact and promise of nucleic acid-based treatments in the field of medicine. Nevertheless, employing nucleic acids as therapeutics is challenging due to their susceptibility to degradation by nucleases and their unfavorable physicochemical characteristics that hinder delivery into cells. Appropriate vectors play a pivotal role in improving nucleic acid stability and delivering nucleic acids into specific cells. The maturation of delivery systems has led to breakthroughs in the development of therapeutics based on nucleic acids such as DNA, siRNA, and mRNA. Non-viral vectors have gained prominence among the myriad of nanomaterials due to low immunogenicity, ease of manufacturing, and simplicity of cost-effective, large-scale production. Here, we provide an overview of the recent advancements in nanomaterials for nucleic acid delivery. Specifically, we give a detailed introduction to the characteristics of polymers, lipids, and polymer-lipid hybrids, and provide comprehensive descriptions of their applications in nucleic acid delivery. Also, biological barriers, administration routes, and strategies for organ-selective delivery of nucleic acids are discussed. In summary, this review offers insights into the rational design of next-generation delivery vectors for nucleic acid delivery.
Collapse
Affiliation(s)
- Yuyang Qin
- West China School of Public Health and West China Fourth Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, China
| | - Liyuan Ou
- West China School of Public Health and West China Fourth Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, China
| | - Lili Zha
- West China School of Public Health and West China Fourth Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, China
| | - Yue Zeng
- West China School of Public Health and West China Fourth Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, China
| | - Ling Li
- West China School of Public Health and West China Fourth Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
5
|
Yihunie W, Nibret G, Aschale Y. Recent Advances in Messenger Ribonucleic Acid (mRNA) Vaccines and Their Delivery Systems: A Review. Clin Pharmacol 2023; 15:77-98. [PMID: 37554660 PMCID: PMC10405914 DOI: 10.2147/cpaa.s418314] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/28/2023] [Indexed: 08/10/2023] Open
Abstract
Messenger ribonucleic acid (mRNA) was found as the intermediary that transfers genetic information from DNA to ribosomes for protein synthesis in 1961. The emergency use authorization of the two covid-19 mRNA vaccines, BNT162b2 and mRNA-1273, is a significant achievement in the history of vaccine development. Because they are generated in a cell-free environment using the in vitro transcription (IVT) process, mRNA vaccines are risk-free. Moreover, chemical modifications to the mRNA molecule, such as cap structures and changed nucleosides, have proved critical in overcoming immunogenicity concerns, achieving sustained stability, and achieving effective, accurate protein production in vivo. Several vaccine delivery strategies (including protamine, lipid nanoparticles (LNPs), polymers, nanoemulsions, and cell-based administration) were also optimized to load and transport RNA into the cytosol. LNPs, which are composed of a cationic or a pH-dependent ionizable lipid layer, a polyethylene glycol (PEG) component, phospholipids, and cholesterol, are the most advanced systems for delivering mRNA vaccines. Moreover, modifications of the four components that make up the LNPs showed to increase vaccine effectiveness and reduce side effects. Furthermore, the introduction of biodegradable lipids improved LNP biocompatibility. Furthermore, mRNA-based therapies are expected to be effective treatments for a variety of refractory conditions, including infectious diseases, metabolic genetic diseases, cancer, cardiovascular and cerebrovascular diseases. Therefore, the present review aims to provide the scientific community with up-to-date information on mRNA vaccines and their delivery systems.
Collapse
Affiliation(s)
- Wubetu Yihunie
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Getinet Nibret
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Yibeltal Aschale
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| |
Collapse
|
6
|
Li Y, Wang M, Peng X, Yang Y, Chen Q, Liu J, She Q, Tan J, Lou C, Liao Z, Li X. mRNA vaccine in cancer therapy: Current advance and future outlook. Clin Transl Med 2023; 13:e1384. [PMID: 37612832 PMCID: PMC10447885 DOI: 10.1002/ctm2.1384] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 08/10/2023] [Accepted: 08/14/2023] [Indexed: 08/25/2023] Open
Abstract
Messenger ribonucleic acid (mRNA) vaccines are a relatively new class of vaccines that have shown great promise in the immunotherapy of a wide variety of infectious diseases and cancer. In the past 2 years, SARS-CoV-2 mRNA vaccines have contributed tremendously against SARS-CoV2, which has prompted the arrival of the mRNA vaccine research boom, especially in the research of cancer vaccines. Compared with conventional cancer vaccines, mRNA vaccines have significant advantages, including efficient production of protective immune responses, relatively low side effects and lower cost of acquisition. In this review, we elaborated on the development of cancer vaccines and mRNA cancer vaccines, as well as the potential biological mechanisms of mRNA cancer vaccines and the latest progress in various tumour treatments, and discussed the challenges and future directions for the field.
Collapse
Affiliation(s)
- Youhuai Li
- Department of Breast SurgeryBaoji Municipal Central HospitalWeibin DistrictBaojiShaanxiChina
| | - Mina Wang
- Graduate SchoolBeijing University of Chinese MedicineBeijingChina
- Department of Acupuncture and MoxibustionBeijing Hospital of Traditional Chinese MedicineCapital Medical UniversityBeijing Key Laboratory of Acupuncture NeuromodulationBeijingChina
| | - Xueqiang Peng
- Department of General SurgeryThe Fourth Affiliated HospitalChina Medical UniversityShenyangChina
| | - Yingying Yang
- Clinical Research CenterShanghai Key Laboratory of Maternal Fetal MedicineShanghai Institute of Maternal‐Fetal Medicine and Gynecologic OncologyShanghai First Maternity and Infant HospitalSchool of MedicineTongji UniversityShanghaiChina
| | - Qishuang Chen
- Graduate SchoolBeijing University of Chinese MedicineBeijingChina
| | - Jiaxing Liu
- Department of General SurgeryThe Fourth Affiliated HospitalChina Medical UniversityShenyangChina
| | - Qing She
- Department of Breast SurgeryBaoji Municipal Central HospitalWeibin DistrictBaojiShaanxiChina
| | - Jichao Tan
- Department of Breast SurgeryBaoji Municipal Central HospitalWeibin DistrictBaojiShaanxiChina
| | - Chuyuan Lou
- Department of OphthalmologyXi'an People's Hospital (Xi'an Fourth Hospital)Xi'anShaanxiChina
| | - Zehuan Liao
- School of Biological SciencesNanyang Technological UniversitySingaporeSingapore
- Department of Microbiology, Tumor and Cell Biology (MTC)Karolinska InstitutetSweden
| | - Xuexin Li
- Department of Medical Biochemistry and Biophysics (MBB)Karolinska InstitutetBiomedicumStockholmSweden
| |
Collapse
|
7
|
Pereira GC. Nanotechnology-Driven Delivery Systems in Inoculation Therapies. Methods Mol Biol 2023; 2575:39-57. [PMID: 36301470 DOI: 10.1007/978-1-0716-2716-7_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Nanotechnology and genomics are the newest allies of inoculation design. In recent years, nucleic acids have been targeted as sources of therapeutics to stimulate immune responses, to both fight disease and create memory to trigger further responses to threat. A myriad of promising findings in cancer research and virology has been reported in the current literature. Nanosystems are demonstrating their capabilities as efficient carriers, improving the efficacy of drug delivery, including nucleic acids as therapeutics, at focal sites, in living systems. This chapter approaches major elements involved in the successful use of nanotechnology as delivery platforms to optimise the efficacy of nucleic acids-driven therapeutics, particularly mRNA vectors as coding engines for targeted viral proteins. Latest findings in nanotechnological design are highlighted, key discoveries associated with the success of nanodelivery platforms are presented, and key characteristics of nanodelivery systems in nucleic acids-based vaccine technology are discussed, to illustrate their distinct advantages and disadvantages.
Collapse
|
8
|
Kai Zhang, Liu P, Bai X, Gao X, Liu K, Li A, Lyu Z. The pH-Responsive CS-g-PEI-g-PEG Graft Copolymer as PolyI:C/OVA Drug Carrier. POLYMER SCIENCE SERIES A 2022. [DOI: 10.1134/s0965545x2370061x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
|
9
|
Yang L, Gong L, Wang P, Zhao X, Zhao F, Zhang Z, Li Y, Huang W. Recent Advances in Lipid Nanoparticles for Delivery of mRNA. Pharmaceutics 2022; 14:2682. [PMID: 36559175 PMCID: PMC9787894 DOI: 10.3390/pharmaceutics14122682] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/23/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022] Open
Abstract
Messenger RNA (mRNA), which is composed of ribonucleotides that carry genetic information and direct protein synthesis, is transcribed from a strand of DNA as a template. On this basis, mRNA technology can take advantage of the body's own translation system to express proteins with multiple functions for the treatment of various diseases. Due to the advancement of mRNA synthesis and purification, modification and sequence optimization technologies, and the emerging lipid nanomaterials and other delivery systems, mRNA therapeutic regimens are becoming clinically feasible and exhibit significant reliability in mRNA stability, translation efficiency, and controlled immunogenicity. Lipid nanoparticles (LNPs), currently the leading non-viral delivery vehicles, have made many exciting advances in clinical translation as part of the COVID-19 vaccines and therefore have the potential to accelerate the clinical translation of gene drugs. Additionally, due to their small size, biocompatibility, and biodegradability, LNPs can effectively deliver nucleic acids into cells, which is particularly important for the current mRNA regimens. Therefore, the cutting-edge LNP@mRNA regimens hold great promise for cancer vaccines, infectious disease prevention, protein replacement therapy, gene editing, and rare disease treatment. To shed more lights on LNP@mRNA, this paper mainly discusses the rational of choosing LNPs as the non-viral vectors to deliver mRNA, the general rules for mRNA optimization and LNP preparation, and the various parameters affecting the delivery efficiency of LNP@mRNA, and finally summarizes the current research status as well as the current challenges. The latest research progress of LNPs in the treatment of other diseases such as oncological, cardiovascular, and infectious diseases is also given. Finally, the future applications and perspectives for LNP@mRNA are generally introduced.
Collapse
Affiliation(s)
- Lei Yang
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Liming Gong
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Ping Wang
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xinghui Zhao
- Beijing Bio-Bank Co., Ltd., Beijing 100107, China
| | - Feng Zhao
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhijie Zhang
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yunfei Li
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Wei Huang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
10
|
Gómez-Aguado I, Rodríguez-Castejón J, Beraza-Millor M, Rodríguez-Gascón A, Del Pozo-Rodríguez A, Solinís MÁ. mRNA delivery technologies: Toward clinical translation. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 372:207-293. [PMID: 36064265 DOI: 10.1016/bs.ircmb.2022.04.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Messenger RNA (mRNA)-therapies have recently taken a huge step toward clinic thanks to the first mRNA-based medicinal products marketed. mRNA features for clinical purposes are improved by chemical modifications, but the inclusion in a delivery system is a regular requirement. mRNA nanomedicines must be designed for the specific therapeutic purpose, protecting the nucleic acid and facilitating the overcoming of biological barriers. Polymers, polypeptides, and cationic lipids are the main used materials to design mRNA delivery systems. Among them, lipid nanoparticles (LNPs) are the most advanced ones, and currently they are at the forefront of preclinical and clinical evaluation in several fields, including immunotherapy (against infectious diseases and cancer), protein replacement, gene editing and regenerative medicine. This chapter includes an overview on mRNA delivery technologies, with special interest in LNPs, and the most recent advances in their clinical application. Liposomes are the mRNA delivery technology with the highest clinical translation among LNPs, whereas the first clinical trial of a therapeutic mRNA formulated in exosomes has been recently approved for protein replacement therapy. The first mRNA products approved by the regulatory agencies worldwide are LNP-based mRNA vaccines against viral infections, specifically against the 2019 coronavirus disease (COVID-19). The clinical translation of mRNA-therapies for cancer is mainly focused on three strategies: anti-cancer vaccination by means of delivering cancer antigens or acting as an adjuvant, mRNA-engineered chimeric antigen receptors (CARs) and T-cell receptors (TCRs), and expression of antibodies and immunomodulators. Cancer immunotherapy and, more recently, COVID-19 vaccines spearhead the advance of mRNA clinical use.
Collapse
Affiliation(s)
- Itziar Gómez-Aguado
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain; Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, Vitoria-Gasteiz, Spain
| | - Julen Rodríguez-Castejón
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain; Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, Vitoria-Gasteiz, Spain
| | - Marina Beraza-Millor
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain; Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, Vitoria-Gasteiz, Spain
| | - Alicia Rodríguez-Gascón
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain; Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, Vitoria-Gasteiz, Spain
| | - Ana Del Pozo-Rodríguez
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain; Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, Vitoria-Gasteiz, Spain
| | - María Ángeles Solinís
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain; Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, Vitoria-Gasteiz, Spain.
| |
Collapse
|
11
|
Kubczak M, Michlewska S, Karimov M, Ewe A, Noske S, Aigner A, Bryszewska M, Ionov M. Unmodified and tyrosine-modified polyethylenimines as potential carriers for siRNA: Biophysical characterization and toxicity. Int J Pharm 2022; 614:121468. [PMID: 35031413 DOI: 10.1016/j.ijpharm.2022.121468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 12/20/2021] [Accepted: 01/09/2022] [Indexed: 11/18/2022]
Abstract
Polyethylenimines (PEIs) are being explored as efficient non-viral nanocarriers for nucleic acid delivery in vitro and in vivo. To address limitations regarding PEI efficacy and biocompatibility, modifications of the chemical structure of linear and branched PEIs have been introduced, including grafting with tyrosine. The aim has been to compare linear and branched polyethylenimines of a wider range of different molecular mass with their tyrosine-modified derivatives. To do so, physico-chemical and biological properties of the polymers were investigated. Even in the absence of a negatively charged nucleic acid counterpart, PEIs form particle structures with defined size and surface potential. Tyrosine modification of PEI led to significantly reduced toxicity, while simultaneously increasing interaction with cellular membranes. All the effects were also dependent on the PEI molecular weight and structure (i.e., linear vs. branched). Especially in the case of linear PEIs, the improved membrane interaction also translated into slightly enhanced hemolysis, whereas their genotoxic potential was essentially abolished. Due to the improvement of properties critical for nano-vector efficacy and biocompatibility, our data demonstrate that tyrosine-modified PEIs are very promising and safe nanocarriers for the delivery of small RNAs, like siRNAs and miRNAs.
Collapse
Affiliation(s)
- Małgorzata Kubczak
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Poland.
| | - Sylwia Michlewska
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Poland; Laboratory of Microscopic Imaging and Specialized Biological Techniques, Faculty of Biology and Environmental Protection, University of Lodz, Poland
| | - Michael Karimov
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, Faculty of Medicine, Leipzig University, Germany
| | - Alexander Ewe
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, Faculty of Medicine, Leipzig University, Germany
| | - Sandra Noske
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, Faculty of Medicine, Leipzig University, Germany
| | - Achim Aigner
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, Faculty of Medicine, Leipzig University, Germany
| | - Maria Bryszewska
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Poland
| | - Maksim Ionov
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Poland
| |
Collapse
|
12
|
Sirianni QEA, Wang TD, Borecki A, Deng Z, Ronald J, Gillies ER. Self-immolative Polyplexes for DNA Delivery. Biomater Sci 2022; 10:2557-2567. [DOI: 10.1039/d1bm01684a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Nucleic acids have immense potential for the treatment and prevention of a wide range of diseases, but delivery vehicles are needed to assist with their entry into cells. Polycations can...
Collapse
|
13
|
Yin Y, Su W, Zhang J, Huang W, Li X, Ma H, Tan M, Song H, Cao G, Yu S, Yu D, Jeong JH, Zhao X, Li H, Nie G, Wang H. Separable Microneedle Patch to Protect and Deliver DNA Nanovaccines Against COVID-19. ACS NANO 2021; 15:14347-14359. [PMID: 34472328 PMCID: PMC8425335 DOI: 10.1021/acsnano.1c03252] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 08/26/2021] [Indexed: 05/12/2023]
Abstract
The successful control of coronavirus disease 2019 (COVID-19) pandemic is not only relying on the development of vaccines, but also depending on the storage, transportation, and administration of vaccines. Ideally, nucleic acid vaccine should be directly delivered to proper immune cells or tissue (such as lymph nodes). However, current developed vaccines are normally treated through intramuscular injection, where immune cells do not normally reside. Meanwhile, current nucleic acid vaccines must be stored in a frozen state that may hinder their application in developing countries. Here, we report a separable microneedle (SMN) patch to deliver polymer encapsulated spike (or nucleocapsid) protein encoding DNA vaccines and immune adjuvant for efficient immunization. Compared with intramuscular injection, SMN patch can deliver nanovaccines into intradermal for inducing potent and durable adaptive immunity. IFN-γ+CD4/8+ and IL-2+CD4/8+ T cells or virus specific IgG are significantly increased after vaccination. Moreover, in vivo results show the SMN patches can be stored at room temperature for at least 30 days without decreases in immune responses. These features of nanovaccines-laden SMN patch are important for developing advanced COVID-19 vaccines with global accessibility.
Collapse
Affiliation(s)
- Yue Yin
- CAS Key Laboratory for Biomedical Effects of
Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience,
National Center for Nanoscience and Technology, Beijing,
100190, China
| | - Wen Su
- CAS Key Laboratory for Biomedical Effects of
Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience,
National Center for Nanoscience and Technology, Beijing,
100190, China
| | - Jie Zhang
- CAS Key Laboratory for Biomedical Effects of
Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience,
National Center for Nanoscience and Technology, Beijing,
100190, China
| | - Wenping Huang
- CAS Key Laboratory for Biomedical Effects of
Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience,
National Center for Nanoscience and Technology, Beijing,
100190, China
| | - Xiaoyang Li
- CAS Key Laboratory for Biomedical Effects of
Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience,
National Center for Nanoscience and Technology, Beijing,
100190, China
- Department of Orthopedics, National Cancer
Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese
Academy of Medical Sciences and Peking Union Medical College, Beijing,
100021, China
| | - Haixia Ma
- CAS Key Laboratory for Biomedical Effects of
Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience,
National Center for Nanoscience and Technology, Beijing,
100190, China
| | - Mixiao Tan
- CAS Key Laboratory for Biomedical Effects of
Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience,
National Center for Nanoscience and Technology, Beijing,
100190, China
| | - Haohao Song
- CAS Key Laboratory for Biomedical Effects of
Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience,
National Center for Nanoscience and Technology, Beijing,
100190, China
| | - Guoliang Cao
- CAS Key Laboratory for Biomedical Effects of
Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience,
National Center for Nanoscience and Technology, Beijing,
100190, China
| | - Shengji Yu
- Department of Orthopedics, National Cancer
Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese
Academy of Medical Sciences and Peking Union Medical College, Beijing,
100021, China
| | - Di Yu
- Department of Immunology, Genetics and Pathology,
Science for Life Laboratory, Uppsala University, Uppsala,
75185, Sweden
| | - Ji Hoon Jeong
- School of Pharmacy, Sungkyunkwan
University, Suwon 16419, Republic of Korea
| | - Xiao Zhao
- CAS Key Laboratory for Biomedical Effects of
Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience,
National Center for Nanoscience and Technology, Beijing,
100190, China
| | - Hui Li
- Dongfang Hospital, Beijing University of
Chinese Medicine, Beijing, 100078, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of
Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience,
National Center for Nanoscience and Technology, Beijing,
100190, China
- University of Chinese Academy of
Sciences, Beijing, 100049, China
| | - Hai Wang
- CAS Key Laboratory for Biomedical Effects of
Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience,
National Center for Nanoscience and Technology, Beijing,
100190, China
- University of Chinese Academy of
Sciences, Beijing, 100049, China
| |
Collapse
|
14
|
Lü JM, Liang Z, Liu D, Zhan B, Yao Q, Chen C. Two Antibody-Guided Lactic-co-Glycolic Acid-Polyethylenimine (LGA-PEI) Nanoparticle Delivery Systems for Therapeutic Nucleic Acids. Pharmaceuticals (Basel) 2021; 14:841. [PMID: 34577541 PMCID: PMC8470087 DOI: 10.3390/ph14090841] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/14/2021] [Accepted: 08/23/2021] [Indexed: 01/10/2023] Open
Abstract
We previously reported a new polymer, lactic-co-glycolic acid-polyethylenimine (LGA-PEI), as an improved nanoparticle (NP) delivery for therapeutic nucleic acids (TNAs). Here, we further developed two antibody (Ab)-conjugated LGA-PEI NP technologies for active-targeting delivery of TNAs. LGA-PEI was covalently conjugated with a single-chain variable fragment antibody (scFv) against mesothelin (MSLN), a biomarker for pancreatic cancer (PC), or a special Ab fragment crystallizable region-binding peptide (FcBP), which binds to any full Ab (IgG). TNAs used in the current study included tumor suppressor microRNA mimics (miR-198 and miR-520h) and non-coding RNA X-inactive specific transcript (XIST) fragments; green fluorescence protein gene (GFP plasmid DNA) was also used as an example of plasmid DNA. MSLN scFv-LGA-PEI NPs with TNAs significantly improved their binding and internalization in PC cells with high expression of MSLN in vitro and in vivo. Anti-epidermal growth factor receptor (EGFR) monoclonal Ab (Cetuximab) binding to FcBP-LGA-PEI showed active-targeting delivery of TNAs to EGFR-expressing PC cells.
Collapse
Affiliation(s)
- Jian-Ming Lü
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Plaza, Houston, TX 77030, USA; (J.-M.L.); (Z.L.); (D.L.); (Q.Y.)
| | - Zhengdong Liang
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Plaza, Houston, TX 77030, USA; (J.-M.L.); (Z.L.); (D.L.); (Q.Y.)
| | - Dongliang Liu
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Plaza, Houston, TX 77030, USA; (J.-M.L.); (Z.L.); (D.L.); (Q.Y.)
| | - Bin Zhan
- National School of Tropical Medicine and Department of Pediatrics, Section of Tropical Medicine, Baylor College of Medicine, One Plaza, Houston, TX 77030, USA;
| | - Qizhi Yao
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Plaza, Houston, TX 77030, USA; (J.-M.L.); (Z.L.); (D.L.); (Q.Y.)
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center, Houston, TX 77030, USA
| | - Changyi Chen
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Plaza, Houston, TX 77030, USA; (J.-M.L.); (Z.L.); (D.L.); (Q.Y.)
| |
Collapse
|
15
|
Liang Y, Huang L, Liu T. Development and Delivery Systems of mRNA Vaccines. Front Bioeng Biotechnol 2021; 9:718753. [PMID: 34386486 PMCID: PMC8354200 DOI: 10.3389/fbioe.2021.718753] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/02/2021] [Indexed: 01/15/2023] Open
Abstract
Since the outbreak of SARS-CoV-2, mRNA vaccine development has undergone a tremendous drive within the pharmaceutical field. In recent years, great progress has been made into mRNA vaccine development, especially in individualized tumor vaccines. mRNA vaccines are a promising approach as the production process is simple, safety profiles are better than those of DNA vaccines, and mRNA-encoded antigens are readily expressed in cells. However, mRNA vaccines also possess some inherent limitations. While side effects such as allergy, renal failure, heart failure, and infarction remain a risk, the vaccine mRNA may also be degraded quickly after administration or cause cytokine storms. This is a substantial challenge for mRNA delivery. However, appropriate carriers can avoid degradation and enhance immune responses, effector presentation, biocompatibility and biosafety. To understand the development and research status of mRNA vaccines, this review focuses on analysis of molecular design, delivery systems and clinical trials of mRNA vaccines, thus highlighting the route for wider development and further clinical trials of mRNA vaccines.
Collapse
Affiliation(s)
- Yongjun Liang
- Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Liping Huang
- Obstetrics and Gynecology Center, Nanfang Hospital, Guangzhou, China
| | - Tiancai Liu
- Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| |
Collapse
|
16
|
Liu HW, Hu Y, Ren Y, Nam H, Santos JL, Ng S, Gong L, Brummet M, Carrington CA, Ullman CG, Pomper MG, Minn I, Mao HQ. Scalable Purification of Plasmid DNA Nanoparticles by Tangential Flow Filtration for Systemic Delivery. ACS APPLIED MATERIALS & INTERFACES 2021; 13:30326-30336. [PMID: 34162211 PMCID: PMC9701136 DOI: 10.1021/acsami.1c05750] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Plasmid DNA (pDNA) nanoparticles synthesized by complexation with linear polyethylenimine (lPEI) are one of the most effective non-viral gene delivery vehicles. However, the lack of scalable and reproducible production methods and the high toxicity have hindered their clinical translation. Previously, we have developed a scalable flash nanocomplexation (FNC) technique to formulate pDNA/lPEI nanoparticles using a continuous flow process. Here, we report a tangential flow filtration (TFF)-based scalable purification method to reduce the uncomplexed lPEI concentration in the nanoparticle formulation and improve its biocompatibility. The optimized procedures achieved a 60% reduction of the uncomplexed lPEI with preservation of the nanoparticle size and morphology. Both in vitro and in vivo studies showed that the purified nanoparticles significantly reduced toxicity while maintaining transfection efficiency. TFF also allows for gradual exchange of solvents to isotonic solutions and further concentrating the nanoparticles for injection. Combining FNC production and TFF purification, we validated the purified pDNA/lPEI nanoparticles for future clinical translation of this gene nanomedicine.
Collapse
Affiliation(s)
- Heng-Wen Liu
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Yizong Hu
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Yong Ren
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Hwanhee Nam
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jose Luis Santos
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Shirley Ng
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Like Gong
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Mary Brummet
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | | | | | - Martin G. Pomper
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Il Minn
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Hai-Quan Mao
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
17
|
Ansari AS, K C R, Jiang X, Uludaǧ H. Investigation of water-insoluble hydrophobic polyethylenimines as RNAi vehicles in chronic myeloid leukemia therapy. J Biomed Mater Res A 2021; 109:2306-2321. [PMID: 33964112 DOI: 10.1002/jbm.a.37214] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/19/2021] [Accepted: 04/25/2021] [Indexed: 11/10/2022]
Abstract
The discovery of RNA interference (RNAi) more than two decades ago opened avenues for avant-garde cancer treatments that possess the ability to evade issues hampering current chemotherapeutic strategies, owing to its specific gene sequence-driven mechanism of action. A potent short interfering RNA (siRNA) delivery vehicle designed to overcome physiological barriers is imperative for successful RNAi therapy. For this purpose, this study explored the characteristics and therapeutic efficacy of low-molecular weight (MW) polyethylenimine (PEI) with high cholesterol substitution, yielding water-insoluble polymers, in chronic myeloid leukemia (CML) K562 cells. A strong impact of cholesterol grafting on the physicochemical attributes of the resultant polymers and their corresponding complexes with siRNA was observed, with the siRNA binding capacity of polymers increasing and complex dissociation sensitivity decreasing with increase in cholesterol content of the polymers. The modified polymer complexes were significantly smaller in size and possessed higher cationic charge compared to the parent polymer. The interaction with anionic heparan sulfate preoteoglycans present on the cell surface was significant in cellular uptake of the complexes. The therapeutic efficacy of siRNA/polymer complexes was reflected in their ability to effectively silence the reporter green fluorescent protein gene and endogenous CML oncogene BCR-ABL as well as significantly inhibit colony formation by K562 cells post BCR-ABL silencing. The results of this study demonstrated beneficial effects of high levels of hydrophobic substitution on low MW PEI on their functional performance bestowing them the potential to be potent RNAi agents for CML therapy.
Collapse
Affiliation(s)
- Aysha S Ansari
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, Canada
| | - Remant K C
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, Canada
| | - Xiaoyan Jiang
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Hasan Uludaǧ
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, Canada.,Department of Biomedical Engineering, Faculty of Medicine, University of Alberta, Edmonton, Canada.,Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| |
Collapse
|
18
|
Controlled post-polymerization modification through modulation of repeating unit reactivity: Proof of concept discussed using linear polyethylenimine example. POLYMER 2021. [DOI: 10.1016/j.polymer.2021.123452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
19
|
Anionic Complex with Efficient Expression and Good Safety Profile for mRNA Delivery. Pharmaceutics 2021; 13:pharmaceutics13010126. [PMID: 33478077 PMCID: PMC7835867 DOI: 10.3390/pharmaceutics13010126] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/14/2021] [Accepted: 01/15/2021] [Indexed: 01/31/2023] Open
Abstract
We previously found that a complex comprising plasmid DNA (pDNA), polyethylenimine (PEI), and γ-polyglutamic acid (γ-PGA) had high transgene efficiency without cytotoxicity in vitro and in vivo. However, messenger RNA (mRNA) remains an attractive alternative to pDNA. In this study, we developed a safe and effective delivery system for mRNA to prevent its degradation and efficiently deliver it into target cells. Various cationic and anionic complexes were produced containing PEI, γ-PGA, and an mRNA encoding firefly luciferase. Their physicochemical properties and cytotoxicities were analyzed and the in vitro and in vivo protein expression were determined. The cationic mRNA/PEI complex showed high in vitro protein expression with strong cytotoxicity. The anionic complex was constructed as mRNA/PEI8/γ-PGA12 complex with a theoretical charge ratio of 1:8:12 based on the phosphate groups of the mRNA, nitrogen groups of PEI, and carboxylate groups of γ-PGA. It was stable and showed high in vitro protein expression without cytotoxicity. After intravenous administration of mRNA/PEI8/γ-PGA12 complex to mice, high protein expression was observed in the spleen and liver and slight expression was observed in the lung over 24 h. Thus, the newly constructed mRNA/PEI8/γ-PGA12 complex provides a safe and effective strategy for the delivery of mRNA.
Collapse
|
20
|
Brandariz-Nuñez A, Robinson SJ, Evilevitch A. Pressurized DNA state inside herpes capsids-A novel antiviral target. PLoS Pathog 2020; 16:e1008604. [PMID: 32702029 PMCID: PMC7377361 DOI: 10.1371/journal.ppat.1008604] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 05/05/2020] [Indexed: 01/25/2023] Open
Abstract
Drug resistance in viruses represents one of the major challenges of healthcare. As part of an effort to provide a treatment that avoids the possibility of drug resistance, we discovered a novel mechanism of action (MOA) and specific compounds to treat all nine human herpesviruses and animal herpesviruses. The novel MOA targets the pressurized genome state in a viral capsid, "turns off" capsid pressure, and blocks viral genome ejection into a cell nucleus, preventing viral replication. This work serves as a proof-of-concept to demonstrate the feasibility of a new antiviral target-suppressing pressure-driven viral genome ejection-that is likely impervious to developing drug resistance. This pivotal finding presents a platform for discovery of a new class of broad-spectrum treatments for herpesviruses and other viral infections with genome-pressure-dependent replication. A biophysical approach to antiviral treatment such as this is also a vital strategy to prevent the spread of emerging viruses where vaccine development is challenged by high mutation rates or other evasion mechanisms.
Collapse
Affiliation(s)
- Alberto Brandariz-Nuñez
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Scott J. Robinson
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Alex Evilevitch
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
21
|
Vaughan HJ, Green JJ, Tzeng SY. Cancer-Targeting Nanoparticles for Combinatorial Nucleic Acid Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1901081. [PMID: 31222852 PMCID: PMC6923623 DOI: 10.1002/adma.201901081] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 04/13/2019] [Indexed: 05/03/2023]
Abstract
Nucleic acids are a promising type of therapeutic for the treatment of a wide range of conditions, including cancer, but they also pose many delivery challenges. For efficient and safe delivery to cancer cells, nucleic acids must generally be packaged into a vehicle, such as a nanoparticle, that will allow them to be taken up by the target cells and then released in the appropriate cellular compartment to function. As with other types of therapeutics, delivery vehicles for nucleic acids must also be designed to avoid unwanted side effects; thus, the ability of such carriers to target their cargo to cancer cells is crucial. Classes of nucleic acids, hurdles that must be overcome for effective intracellular delivery, types of nonviral nanomaterials used as delivery vehicles, and the different strategies that can be employed to target nucleic acid delivery specifically to tumor cells are discussed. Additonally, nanoparticle designs that facilitate multiplexed delivery of combinations of nucleic acids are reviewed.
Collapse
Affiliation(s)
- Hannah J Vaughan
- Department of Biomedical Engineering, Translational Tissue Engineering Center and Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, 400 North Broadway, Smith Building 5001, Baltimore, MD, 21231, USA
| | - Jordan J Green
- Department of Biomedical Engineering, Translational Tissue Engineering Center and Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, 400 North Broadway, Smith Building 5001, Baltimore, MD, 21231, USA
| | - Stephany Y Tzeng
- Department of Biomedical Engineering, Translational Tissue Engineering Center and Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, 400 North Broadway, Smith Building 5001, Baltimore, MD, 21231, USA
| |
Collapse
|
22
|
Tang J, Chen L, Yan D, Shen Z, Wang B, Weng S, Wu Z, Xie Z, Shao J, Yang L, Shen L. Surface Functionalization with Proanthocyanidins Provides an Anti-Oxidant Defense Mechanism That Improves the Long-Term Stability and Osteogenesis of Titanium Implants. Int J Nanomedicine 2020; 15:1643-1659. [PMID: 32210558 PMCID: PMC7073973 DOI: 10.2147/ijn.s231339] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 02/17/2020] [Indexed: 12/17/2022] Open
Abstract
PURPOSE Aseptic loosening is a major complication after total joint replacement. Reactive oxygen species generated by local tissue cells and liberated from implant surfaces have been suggested to cause implant failures. Surface modification of titanium (Ti)-based implants with proanthocyanidins (PAC) is a promising approach for the development of anti-oxidant defense mechanism to supplement the mechanical functions of Ti implants. In this study, a controlled PAC release system was fabricated on the surface of Ti substrates using the layer-by-layer (LBL) assembly. MATERIALS AND METHODS Polyethyleneimine (PEI) base layer was fabricated to enable layer-by-layer (LBL) deposition of hyaluronic acid/chitosan (HA/CS) multi-layers without or with the PAC. Surface topography and wettability of the fabricated HA/CS-PAC substrates were characterized by scanning electron microscopy (SEM), atomic force microscopy (AFM), Fourier-transform infrared spectroscopy (FTIR) and contact angle measurement. PAC release profiles were investigated using drug release assays. MC3T3-E1 pre-osteoblast cells were used to assess the osteo-inductive effects of HA/CS-PAC substrates under conditions H2O2-induced oxidative stress in vitro. A rat model of femoral intramedullary implantation evaluated the osseo-integration and osteo-inductive potential of the HA/CS-PAC coated Ti implants in vivo. RESULTS SEM, AFM, FTIR and contact angle measurements verified the successful fabrication of Ti surfaces with multi-layered HA/CS-PAC coating. Drug release assays revealed controlled and sustained release of PAC over 14 days. In vitro, cell-based assays showed high tolerability and enhanced the osteogenic potential of MC3T3-E1 cells on HA/CS-PAC substrates when under conditions of H2O2-induced oxidative stress. In vivo evaluation of femoral bone 14 days after femoral intramedullary implantation confirmed the enhanced osteo-inductive potential of the HA/CS-PAC coated Ti implants. CONCLUSION Multi-layering of HA/CS-PAC coating onto Ti-based surfaces by the LBL deposition significantly enhances implant osseo-integration and promotes osteogenesis under conditions of oxidative stress. This study provides new insights for future applications in the field of joint arthroplasty.
Collapse
Affiliation(s)
- Jiahao Tang
- The Second School of Medicine Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
- Key Laboratory of Orthopedics of Zhejiang Province, Department of Orthopedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
| | - Liang Chen
- The Second School of Medicine Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
- Key Laboratory of Orthopedics of Zhejiang Province, Department of Orthopedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
| | - Deyi Yan
- The Second School of Medicine Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
- Key Laboratory of Orthopedics of Zhejiang Province, Department of Orthopedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
| | - Zijian Shen
- The Second School of Medicine Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
- Key Laboratory of Orthopedics of Zhejiang Province, Department of Orthopedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
| | - Bingzhang Wang
- The Second School of Medicine Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
- Key Laboratory of Orthopedics of Zhejiang Province, Department of Orthopedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
| | - Sheji Weng
- The Second School of Medicine Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
- Key Laboratory of Orthopedics of Zhejiang Province, Department of Orthopedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
| | - Zongyi Wu
- The Second School of Medicine Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
- Key Laboratory of Orthopedics of Zhejiang Province, Department of Orthopedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
| | - Zhongjie Xie
- The Second School of Medicine Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
- Key Laboratory of Orthopedics of Zhejiang Province, Department of Orthopedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
| | - Jiancan Shao
- The Second School of Medicine Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
- Key Laboratory of Orthopedics of Zhejiang Province, Department of Orthopedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
| | - Lei Yang
- The Second School of Medicine Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
- Key Laboratory of Orthopedics of Zhejiang Province, Department of Orthopedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
| | - Liyan Shen
- The Second School of Medicine Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
- Key Laboratory of Orthopedics of Zhejiang Province, Department of Orthopedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
| |
Collapse
|
23
|
Abstract
Efficient intracellular delivery of small-interfering ribonucleic acid (siRNA) to the target organ or tissues in the body is assumed as the main hurdle for a widespread use of siRNAs in the clinics. Solid lipid-based nanoparticles (SLNs) and derivatives can potentially fit this purpose by enabling to overcome the extracellular and intracellular physiological barriers affecting the delivery. For that, rational formulations and rational process designs are needed. This chapter addresses a comprehensive description and critical appraisal of the main production methods of this particular type of lipid nanoparticles and the leading strategies to prompt a targeted delivery of siRNA.
Collapse
Affiliation(s)
- Andreia Jorge
- Department of Chemistry, Coimbra Chemistry Centre, University of Coimbra, Coimbra, Portugal.
| | - Alberto Pais
- Department of Chemistry, Coimbra Chemistry Centre, University of Coimbra, Coimbra, Portugal
| | - Carla Vitorino
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.
- Center for Neurosciences and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.
- Department of Chemistry, Coimbra Chemistry Centre, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
24
|
Fisher DG, Price RJ. Recent Advances in the Use of Focused Ultrasound for Magnetic Resonance Image-Guided Therapeutic Nanoparticle Delivery to the Central Nervous System. Front Pharmacol 2019; 10:1348. [PMID: 31798453 PMCID: PMC6864822 DOI: 10.3389/fphar.2019.01348] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 10/24/2019] [Indexed: 12/12/2022] Open
Abstract
Targeting systemically-administered drugs and genes to specific regions of the central nervous system (CNS) remains a challenge. With applications extending into numerous disorders and cancers, there is an obvious need for approaches that facilitate the delivery of therapeutics across the impervious blood-brain barrier (BBB). Focused ultrasound (FUS) is an emerging treatment method that leverages acoustic energy to oscillate simultaneously administered contrast agent microbubbles. This FUS-mediated technique temporarily disrupts the BBB, allowing ordinarily impenetrable agents to diffuse and/or convect into the CNS. Under magnetic resonance image guidance, FUS and microbubbles enable regional targeting-limiting the large, and potentially toxic, dosage that is often characteristic of systemically-administered therapies. Subsequent to delivery across the BBB, therapeutics face yet another challenge: penetrating the electrostatically-charged, mesh-like brain parenchyma. Non-bioadhesive, encapsulated nanoparticles can help overcome this additional barrier to promote widespread treatment in selected target areas. Furthermore, nanoparticles offer significant advantages over conventional systemically-administered therapeutics. Surface modifications of nanoparticles can be engineered to enhance targeted cellular uptake, and nanoparticle formulations can be tailored to control many pharmacokinetic properties such as rate of drug liberation, distribution, and excretion. For instance, nanoparticles loaded with gene plasmids foster relatively stable transfection, thus obviating the need for multiple, successive treatments. As the formulations and applications of these nanoparticles can vary greatly, this review article provides an overview of FUS coupled with polymeric or lipid-based nanoparticles currently utilized for drug delivery, diagnosis, and assessment of function in the CNS.
Collapse
Affiliation(s)
| | - Richard J. Price
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
25
|
Danilevich VN, Kozlov SA, Shevchuk TV, Oleinikov VA, Sizova SV, Khodarovich YM, Mulyukin AL. Ribonucleic acid (RNA) condensation by thermal cycling with metal cations: yield of nanoparticles and their applicability for transfection. J Biomol Struct Dyn 2019; 38:3959-3971. [PMID: 31543001 DOI: 10.1080/07391102.2019.1671228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
To the present, different efficient but expensive, multistage, and time-consuming technologies have been developed to deliver ribonucleic acids (RNA) into eukaryotic cells. Here, we report a simple and feasible solution to design RNA nanocarriers based on nucleic acid condensation by bi- and trivalent metal ions during thermal cycling. Efficient RNA conversion to nanoparticles with small size (10-50 nm) suitable for transfection was achieved using cations Ni2+, Co2+ or Cu2+ alone or in combination with Ca2+ at the specially selected concentrations (2.0 mM-3.5 mM), low ionic strength, and narrow pH range (8.0-8.5). Other ions - Mn2+, Zn2+, Tb3+, or Gd3+ - caused RNA-cleaving effect that was abolished in the presence of Ni2+, Co2+, Zn2+, or Cu2+. Naked RNA-metal ion nanoparticles were extremely unstable in phosphate buffer and sensitive to serum ribonucleases (RNases), and this problem was solved by treatment with polyarginines-16 and 8. Polyarginine-stabilized nanoparticles, containing malachite green (MG) aptamer RNA and metal cations, crossed the cell membrane, dissociated in the cytoplasm, and preserved the functionality of transported RNA, as judged from efficient transfection of human embryonic kidney 293 cells. The technology, involving RNA condensation by metal cations, can be used as a cheap alternative to produce nanoscale carriers to deliver various RNAs into cells in vitro and in vivo.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Vasily N Danilevich
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Science, Moscow, Russia
| | - Sergey A Kozlov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Science, Moscow, Russia
| | - Taras V Shevchuk
- Branch of the M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Pushchino, Moscow, Russia
| | - Vladimir A Oleinikov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Science, Moscow, Russia
| | - Svetlana V Sizova
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Science, Moscow, Russia
| | - Yuriy M Khodarovich
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Science, Moscow, Russia
| | - Andrey L Mulyukin
- Winogradsky Institute of Microbiology, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
26
|
Schwarzenbach H, Gahan PB. Circulating non-coding RNAs in recurrent and metastatic ovarian cancer. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2019; 2:399-418. [PMID: 35582568 PMCID: PMC8992516 DOI: 10.20517/cdr.2019.51] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/15/2019] [Accepted: 08/21/2019] [Indexed: 12/25/2022]
Abstract
Ovarian cancer has a poor outcome because it is usually detected at advanced tumor stages, and the majority of the patients develop disease relapse as a result of chemotherapy resistance. This most lethal gynecological malignancy metastasizes within the peritoneal fluid or ascites to pelvic and distal organs. In ovarian cancer progression and metastasis, small non-coding RNAs (ncRNAs), including long noncoding RNAs and microRNAs have been recognized as important regulators. Their dysregulation modulates gene expression and cellular signal pathways and can be detected in liquid biopsies. In this review, we provide an overview on circulating plasma and serum ncRNAs participating in tumor cell migration and invasion, and contributing to recurrence and metastasis of ovarian cancer. We will also discuss the development of potential, novel therapies using ncRNAs as target molecules or tumor markers for ovarian cancer.
Collapse
Affiliation(s)
- Heidi Schwarzenbach
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Peter B Gahan
- Fondazione "Enrico Puccinelli" Onlus, Perugia 06123, Italy
| |
Collapse
|
27
|
Zhao Z, Li Y, Shukla R, Liu H, Jain A, Barve A, Cheng K. Development of a Biocompatible Copolymer Nanocomplex to Deliver VEGF siRNA for Triple Negative Breast Cancer. Theranostics 2019; 9:4508-4524. [PMID: 31285776 PMCID: PMC6599648 DOI: 10.7150/thno.34314] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 05/20/2019] [Indexed: 12/17/2022] Open
Abstract
Triple negative breast cancer (TNBC) is the most difficult breast cancer subtype to treat. TNBC patients have significantly higher expression of vascular endothelial growth factor (VEGF) in tumors compared to non-TNBC patients. VEGF not only exerts its pro-angiogenic effects on endothelial cells but also acts as a survival and autocrine growth factor for VEGF receptor (VEGFR) expressing cancer cells. Silencing the expression of VEGF is therefore a potential therapy for TNBC. Methods: A novel biocompatible linear copolymer poly[bis(ε-Lys-PEI)Glut-PEG] (PLEGP) was developed to deliver VEGF siRNA for TNBC therapy. The copolymer is composed of lysine and glutaric acid, a natural metabolite of amino acids in the body. Low-molecular weight polyethyleneimine (PEI) was grafted to the copolymer to efficiently condense siRNA into nanocomplex without inducing cytotoxicity. Various in vitro studies were performed to evaluate the stability, cellular uptake, tumor penetration, and biological activities of the VEGF siRNA nanocomplex. The anti-tumor activities of the nanocomplex was also evaluated in an orthotopic TNBC mouse model. Results: PEIs with different molecular weights were evaluated, and the copolymer PLEGP1800 was able to easily form a stable nanocomplex with siRNAs and protect them from serum degradation. The siRNA/PLEGP1800 nanocomplex exhibited negligible cytotoxicity but showed high cellular uptake, high transfection efficiency, and high tumor penetration. In vitro activity studies showed that the siRNA nanocomplex significantly inhibited migration and invasion of TNBC cells. Moreover, the VEGF siRNA nanocomplex efficiently inhibited tumor growth in an orthotopic TNBC mouse model and down-regulated VEGF expression in the tumor. Conclusion: PLEGP1800 is a safe and efficient copolymer to deliver siRNAs for TNBC therapy. It could potentially be applied to other cancers by changing the cargo and incorporating tumor-specific ligands.
Collapse
|
28
|
Su Y, Quan X, Li L, Zhou J. Computer Simulation of DNA Condensation by PAMAM Dendrimer. MACROMOL THEOR SIMUL 2018. [DOI: 10.1002/mats.201700070] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Yunxiang Su
- School of Chemistry and Chemical Engineering; South China University of Technology; Guangzhou 510460 China
| | - Xuebo Quan
- School of Chemistry and Chemical Engineering; South China University of Technology; Guangzhou 510460 China
| | - Libo Li
- School of Chemistry and Chemical Engineering; South China University of Technology; Guangzhou 510460 China
| | - Jian Zhou
- School of Chemistry and Chemical Engineering; South China University of Technology; Guangzhou 510460 China
| |
Collapse
|
29
|
Gao T, Bi A, Yang S, Liu Y, Kong X, Zeng W. Applications of Nanoparticles Probes for Prostate Cancer Imaging and Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1096:99-115. [PMID: 30324350 DOI: 10.1007/978-3-319-99286-0_6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Prostate cancer (PCa) is the most common type of cancer in men with high morbidity and mortality. However, the current treatment with drugs often leads to chemotherapy resistance. It is known that the multi-disciplines research on molecular imaging is very helpful for early diagnosing, staging, restaging and precise treatment of PCa. In the past decades, the tumor-specific targeted drugs were developed for the clinic to treat prostate cancer. Among them, the emerging nanotechnology has brought about many exciting novel diagnosis and treatments systems for PCa. Nanotechnology can greatly enhance the treatment activity of PCa and provide novel theranostics platform by utilizing the unique physical/chemical properties, targeting strategy, or by loading with imaging/therapeutic agents. Herein, this chapter focuses on state-of-art advances in imaging and diagnosing PCa with nanomaterials and highlights the approaches used for functionalization of the targeted biomolecules, and in the treatment for various aspects of PCa with multifunctional nanoparticles, nanoplatforms and nanodelivery system.
Collapse
Affiliation(s)
- Tang Gao
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China.,Molecular Imaging Research Center, Central South University, Changsha, China
| | - Anyao Bi
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China.,Molecular Imaging Research Center, Central South University, Changsha, China
| | - Shuiqi Yang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China.,Molecular Imaging Research Center, Central South University, Changsha, China
| | - Yi Liu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China.,Molecular Imaging Research Center, Central South University, Changsha, China
| | - Xiangqi Kong
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China.,Molecular Imaging Research Center, Central South University, Changsha, China
| | - Wenbin Zeng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China. .,Molecular Imaging Research Center, Central South University, Changsha, China.
| |
Collapse
|
30
|
Neamtu I, Rusu AG, Diaconu A, Nita LE, Chiriac AP. Basic concepts and recent advances in nanogels as carriers for medical applications. Drug Deliv 2017; 24:539-557. [PMID: 28181831 PMCID: PMC8240973 DOI: 10.1080/10717544.2016.1276232] [Citation(s) in RCA: 236] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 12/13/2016] [Accepted: 12/20/2016] [Indexed: 01/18/2023] Open
Abstract
Nanogels in biomedical field are promising and innovative materials as dispersions of hydrogel nanoparticles based on crosslinked polymeric networks that have been called as next generation drug delivery systems due to their relatively high drug encapsulation capacity, uniformity, tunable size, ease of preparation, minimal toxicity, stability in the presence of serum, and stimuli responsiveness. Nanogels show a great potential in chemotherapy, diagnosis, organ targeting and delivery of bioactive substances. The main subjects reviewed in this article concentrates on: (i) Nanogel assimilation in the nanomedicine domain; (ii) Features and advantages of nanogels, the main characteristics, such as: swelling capacity, stimuli sensitivity, the great surface area, functionalization, bioconjugation and encapsulation of bioactive substances, which are taken into account in designing the structures according to the application; some data on the advantages and limitations of the preparation techniques; (iii) Recent progress in nanogels as a carrier of genetic material, protein and vaccine. The majority of the scientific literature presents the multivalency potential of bioconjugated nanogels in various conditions. Today's research focuses over the overcoming of the restrictions imposed by cost, some medical requirements and technological issues, for nanogels' commercial scale production and their integration as a new platform in biomedicine.
Collapse
Affiliation(s)
- Iordana Neamtu
- “Petru Poni” Institute of Macromolecular Chemistry, Iasi, Romania
| | | | - Alina Diaconu
- “Petru Poni” Institute of Macromolecular Chemistry, Iasi, Romania
| | | | | |
Collapse
|
31
|
Grasso G, Deriu MA, Patrulea V, Borchard G, Möller M, Danani A. Free energy landscape of siRNA-polycation complexation: Elucidating the effect of molecular geometry, polymer flexibility, and charge neutralization. PLoS One 2017; 12:e0186816. [PMID: 29088239 PMCID: PMC5663398 DOI: 10.1371/journal.pone.0186816] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 10/09/2017] [Indexed: 12/16/2022] Open
Abstract
The success of medical threatments with DNA and silencing interference RNA is strongly related to the design of efficient delivery technologies. Cationic polymers represent an attractive strategy to serve as nucleic-acid carriers with the envisioned advantages of efficient complexation, low cost, ease of production, well-defined size, and low polydispersity index. However, the balance between efficacy and toxicity (safety) of these polymers is a challenge and in need of improvement. With the aim of designing more effective polycationic-based gene carriers, many parameters such as carrier morphology, size, molecular weight, surface chemistry, and flexibility/rigidity ratio need to be taken into consideration. In the present work, the binding mechanism of three cationic polymers (polyarginine, polylysine and polyethyleneimine) to a model siRNA target is computationally investigated at the atomistic level. In order to better understand the polycationic carrier-siRNA interactions, replica exchange molecular dynamic simulations were carried out to provide an exhaustive exploration of all the possible binding sites, taking fully into account the siRNA flexibility together with the presence of explicit solvent and ions. Moreover, well-tempered metadynamics simulations were employed to elucidate how molecular geometry, polycation flexibility, and charge neutralization affect the siRNA-polycations free energy landscape in term of low-energy binding modes and unbinding free energy barriers. Significant differences among polymer binding modes have been detected, revealing the advantageous binding properties of polyarginine and polylysine compared to polyethyleneimine.
Collapse
Affiliation(s)
- Gianvito Grasso
- Istituto Dalle Molle di Studi Sull'Intelligenza Artificiale (IDSIA), Scuola Universitaria Professionale della Svizzera Italiana (SUPSI), Università della Svizzera Italiana (USI), Centro Galleria 2, Manno, Switzerland
| | - Marco Agostino Deriu
- Istituto Dalle Molle di Studi Sull'Intelligenza Artificiale (IDSIA), Scuola Universitaria Professionale della Svizzera Italiana (SUPSI), Università della Svizzera Italiana (USI), Centro Galleria 2, Manno, Switzerland
| | - Viorica Patrulea
- School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, Geneva, Switzerland
| | - Gerrit Borchard
- School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, Geneva, Switzerland
| | - Michael Möller
- School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, Geneva, Switzerland
| | - Andrea Danani
- Istituto Dalle Molle di Studi Sull'Intelligenza Artificiale (IDSIA), Scuola Universitaria Professionale della Svizzera Italiana (SUPSI), Università della Svizzera Italiana (USI), Centro Galleria 2, Manno, Switzerland
| |
Collapse
|
32
|
Disassembly of micelle-like polyethylenimine nanocomplexes for siRNA delivery: High transfection efficiency and reduced toxicity achieved by simple reducible lipid modification. J Colloid Interface Sci 2017; 504:633-644. [PMID: 28618383 DOI: 10.1016/j.jcis.2017.05.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 04/30/2017] [Accepted: 05/03/2017] [Indexed: 12/18/2022]
Abstract
Amphiphilic compounds consisting of polycations and lipid segments are well established as building blocks for the construction of siRNA carriers. They are capable of forming nanoparticles with high-affinity positive charges for siRNA in aqueous media due to their intra- and/or intermolecular hydrophobic and electrostatic interactions. Unfortunately, safety and efficiency of lipid-modified polycations as the two great challenges to the clinical application need to be improved. Beyond that, the role of the hydrophobic segment in the process of siRNA delivery is elusive. Herein, in this study, branched polyethylenimine with a molecular weight of 600 (bPEI600) was grafted with reducible lipids via Michael addition reaction between amines and alkyl acrylates. Reducible amphiphilic polyethylenimines (PEIs) were able to condense siRNA into nanoparticles and disassemble under the reductive environment. Investigations with these materials in vitro revealed that the polymers with higher grafting degree provided high luciferase knockdown efficacies even at lower N/P ratios and the polymers with longer lipid chain displayed greater cellular uptake rate. Interestingly, the polymers with lower grafting degree had efficient cellular uptake than native bPEI600, although their in luciferase knockdown assays were most likely inefficient. The inconsistency between the cellular uptake profile and silencing efficacy proved that the intracellular trafficking of siRNA was a bottleneck for siRNA delivery with some polymers prepared in this study. As expected, reducible lipid-modified PEIs were equally efficient and much less toxic compared to non-reducible counterparts and might provide broader therapeutic windows. These findings showed the feasibility of reducible lipid-modified PEIs as carriers for therapeutic siRNA.
Collapse
|
33
|
Hashem Nia A, Behnam B, Taghavi S, Oroojalian F, Eshghi H, Shier WT, Abnous K, Ramezani M. Evaluation of chemical modification effects on DNA plasmid transfection efficiency of single-walled carbon nanotube-succinate- polyethylenimine conjugates as non-viral gene carriers. MEDCHEMCOMM 2017; 8:364-375. [PMID: 30108752 PMCID: PMC6072421 DOI: 10.1039/c6md00481d] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 11/28/2016] [Indexed: 11/21/2022]
Abstract
Polyethylenimine (PEI) is a widely used non-viral vector for DNA delivery. One major obstacle of higher molecular weight PEIs is the increased cytotoxicity despite the improved transfection efficiency and numerous chemical modifications that have been reported to overcome this problem. Carbon nanotubes (CNT) are carbon nanomaterials capable of penetrating into cell membranes with no cytotoxic effects. Covalent and noncovalent functionalization methods have been used to improve their solubility in aqueous media. The idea of conjugating PEIs and CNT through different chemical bonds and linkers seems promising as it may result in highly effective carriers due to combination of the transfection ability of PEI with cell internalization of CNT. In this study, six different water-soluble PEI conjugates of single-walled carbon nanotubes (SWNTs) were prepared by grafting PEI with one of three molecular weights (1.8, 10 and 25 kDa) through succinate as a linker which refers to "an organic moiety through which a SWNT is conjugated to PEI." The succinate linker was introduced to the surface of SWNTs through two different chemical strategies: a) ester and b) acyl linkages. The resulting SWNT-PEI vectors were characterized by IR spectroscopy, thermogravimetric analysis (TGA) and SEM imaging. All synthesized carriers were evaluated and compared for their cytotoxicity and transfection efficiency in murine neuroblastoma cells as polyplexes with plasmid DNA for luciferase and green fluorescent protein (GFP). The most efficient carriers were prepared by attaching PEI with the lowest molecular weight (1.8 kDa) through acyl linkage, which gave a transfection efficiency 190-fold greater than that of the corresponding free PEI. Transfection efficiency was the highest in polyplexes prepared with acyl-linked conjugates in all the plasmid/vector ratios studied.
Collapse
Affiliation(s)
- Azadeh Hashem Nia
- Pharmaceutical Research Center , School of Pharmacy , Mashhad University of Medical Sciences , Mashhad , Iran . ; ; Tel: +98513 7112470
- Department of Chemistry , Faculty of Sciences , Ferdowsi University of Mashhad , Mashhad , Iran
| | - Behzad Behnam
- Pharmaceutics Research Center , Institute of Neuropharmacology , Kerman University of Medical Sciences , Kerman , Iran
| | - Sahar Taghavi
- Pharmaceutical Research Center , School of Pharmacy , Mashhad University of Medical Sciences , Mashhad , Iran . ; ; Tel: +98513 7112470
| | - Fatemeh Oroojalian
- Nanobiotechnology Group , Department of Life Science Engineering , Faculty of New Sciences and Technologies , University of Tehran , Tehran , Iran
| | - Hossein Eshghi
- Department of Chemistry , Faculty of Sciences , Ferdowsi University of Mashhad , Mashhad , Iran
| | - Wayne T Shier
- Department of Medicinal Chemistry , University of Minnesota-Twin Cities , Minneapolis , MN 55455 , USA
| | - Khalil Abnous
- Pharmaceutical Research Center , School of Pharmacy , Mashhad University of Medical Sciences , Mashhad , Iran . ; ; Tel: +98513 7112470
| | - Mohammad Ramezani
- Pharmaceutical Research Center , School of Pharmacy , Mashhad University of Medical Sciences , Mashhad , Iran . ; ; Tel: +98513 7112470
| |
Collapse
|
34
|
Biodegradable lipid nanoparticles induce a prolonged RNA interference-mediated protein knockdown and show rapid hepatic clearance in mice and nonhuman primates. Int J Pharm 2017; 519:34-43. [PMID: 28089936 DOI: 10.1016/j.ijpharm.2017.01.016] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 12/18/2016] [Accepted: 01/09/2017] [Indexed: 01/02/2023]
Abstract
Lipid nanoparticles based on ionizable lipids have been clinically validated as a means of delivery for RNA interference (RNAi) therapeutics. The ideal properties of RNAi carriers are efficient delivery of oligonucleotides into target cells and rapid elimination after the function is performed. Here, we report that degradable lipid nanoparticles are effective carriers of small interfering RNA (siRNA) and have a high therapeutic index. The newly developed degradable lipid nanoparticles carrying siRNA showed potent gene-silencing activity in mouse hepatocytes (ED50≈0.02mg/kg siRNA). The ester bond in the lipid tail was hydrolyzed in the liver, resulting in rapid metabolism of the lipid. Toxicity assays showed that the degradable lipid was well-tolerated at siRNA doses of up to 16mg/kg in rats (over 800-fold higher than ED50). A single intravenous injection of siRNA targeting proprotein convertase subtilisin/kexin type 9 (PCSK9) in cynomolgus monkeys resulted in more than 90% protein silencing, and a 50% decrease in plasma low-density lipoprotein (LDL) cholesterol, with a measurable reduction for 2 months. Moreover, quantification of lipids in liver biopsies revealed rapid hepatic clearance of the degradable lipid in nonhuman primates. These degradable lipid nanoparticles with a high therapeutic index hold promise for RNA-based treatments.
Collapse
|
35
|
Chen Y, Yue Q, De G, Wang J, Li Z, Xiao S, Yu H, Ma H, Sui F, Zhao Q. Inhibition of breast cancer metastasis by paclitaxel-loaded pH responsive poly(β-amino ester) copolymer micelles. Nanomedicine (Lond) 2017; 12:147-164. [DOI: 10.2217/nnm-2016-0335] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: Tumor metastasis is one of the leading causes of insufficient chemotherapy during cancer treatment. In this study, a poly(β-amino ester) derivate was developed to fabricate paclitaxel (PTX) entrapped pH-responsive copolymer micelles for inhibition of breast cancer metastasis. Materials & methods: PTX-loaded micelles were fabricated by thin film hydration method. The inhibition efficacy of the as-prepared micelles was evaluated on MDA-MB-231 cells and tumor bearing mice. Results: PTX-loaded micelles were successfully prepared. Such micelles could promote drug uptake and MDA-MB-231 cell deaths, and suppress tumor metastasis. Conclusion: The pH-responsive PTX-loaded micelles are promising candidates in developing stimuli triggered drug delivery systems in acidic tumor microenvironments with improved inhibitory effects on tumor metastasis.
Collapse
Affiliation(s)
- Yanjun Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Bejing 100700, China
| | - Qiaoxin Yue
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Bejing 100700, China
| | - Gejing De
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Bejing 100700, China
| | - Jie Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Bejing 100700, China
| | - Zhenzhen Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Bejing 100700, China
| | - Shuiming Xiao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Bejing 100700, China
| | - Huatao Yu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Bejing 100700, China
| | - Hai Ma
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Bejing 100700, China
| | - Feng Sui
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Bejing 100700, China
| | - Qinghe Zhao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Bejing 100700, China
| |
Collapse
|
36
|
Santos JL, Ren Y, Vandermark J, Archang MM, Williford JM, Liu HW, Lee J, Wang TH, Mao HQ. Continuous Production of Discrete Plasmid DNA-Polycation Nanoparticles Using Flash Nanocomplexation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:6214-6222. [PMID: 27717227 PMCID: PMC5149445 DOI: 10.1002/smll.201601425] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 06/27/2016] [Indexed: 05/27/2023]
Abstract
Despite successful demonstration of linear polyethyleneimine (lPEI) as an effective carrier for a wide range of gene medicine, including DNA plasmids, small interfering RNAs, mRNAs, etc., and continuous improvement of the physical properties and biological performance of the polyelectrolyte complex nanoparticles prepared from lPEI and nucleic acids, there still exist major challenges to produce these nanocomplexes in a scalable manner, particularly for lPEI/DNA nanoparticles. This has significantly hindered the progress toward clinical translation of these nanoparticle-based gene medicine. Here the authors report a flash nanocomplexation (FNC) method that achieves continuous production of lPEI/plasmid DNA nanoparticles with narrow size distribution using a confined impinging jet device. The method involves the complex coacervation of negatively charged DNA plasmid and positive charged lPEI under rapid, highly dynamic, and homogeneous mixing conditions, producing polyelectrolyte complex nanoparticles with narrow distribution of particle size and shape. The average number of plasmid DNA packaged per nanoparticles and its distribution are similar between the FNC method and the small-scale batch mixing method. In addition, the nanoparticles prepared by these two methods exhibit similar cell transfection efficiency. These results confirm that FNC is an effective and scalable method that can produce well-controlled lPEI/plasmid DNA nanoparticles.
Collapse
Affiliation(s)
- Jose Luis Santos
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Yong Ren
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - John Vandermark
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Maani M. Archang
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - John-Michael Williford
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Heng-wen Liu
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Jason Lee
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Tza-Huei Wang
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Mechanical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hai-Quan Mao
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Translational Tissue Engineering Center and Whitaker Biomedical Engineering Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
37
|
Wu Z, Zhan S, Fan W, Ding X, Wu X, Zhang W, Fu Y, Huang Y, Huang X, Chen R, Li M, Xu N, Zheng Y, Ding B. Peptide-Mediated Tumor Targeting by a Degradable Nano Gene Delivery Vector Based on Pluronic-Modified Polyethylenimine. NANOSCALE RESEARCH LETTERS 2016; 11:122. [PMID: 26932761 PMCID: PMC4773318 DOI: 10.1186/s11671-016-1337-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 02/23/2016] [Indexed: 05/29/2023]
Abstract
Polyethylenimine (PEI) is considered to be a promising non-viral gene delivery vector. To solve the toxicity versus efficacy and tumor-targeting challenges of PEI used as gene delivery vector, we constructed a novel non-viral vector DR5-TAT-modified Pluronic-PEI (Pluronic-PEI-DR5-TAT), which was based on the attachment of low-molecular-weight polyethylenimine (LMW-PEI) to the amphiphilic polymer Pluronic to prepare Pluronic-modified LMW-PEI (Pluronic-PEI). This was then conjugated to a multifunctional peptide containing a cell-penetrating peptide (TAT) and a synthetic peptide that would bind to DR5-a receptor that is overexpressed in cancer cells. The vector showed controlled degradation, favorable DNA condensation and protection performance. The Pluronic-PEI-DR5-TAT/DNA complexes at an N/P ratio of 15:1 were spherical nanoparticles of 122 ± 11.6 nm and a zeta potential of about 22 ± 2.8 mV. In vitro biological characterization results indicated that Pluronic-PEI-DR5-TAT/DNA complexes had a higher specificity for the DR5 receptor and were taken up more efficiently by tumor cells than normal cells, compared to complexes formed with PEI 25 kDa or Pluronic-PEI. Thus, the novel complexes showed much lower cytotoxicity to normal cells and higher gene transfection efficiency in tumor cells than that exhibited by PEI 25 kDa and Pluronic-PEI. In summary, our novel, degradable non-viral tumor-targeting vector is a promising candidate for use in gene therapy.
Collapse
Affiliation(s)
- Zhaoyong Wu
- Department of Pharmacy, Jiaxing Maternal and Child Health Care Hospital, Affiliated Hospital of Jiaxing University, Jiaxing, People's Republic of China
| | - Shuyu Zhan
- Department of Pharmaceutics, Medical College of Jiaxing University, Jiaxing, People's Republic of China
| | - Wei Fan
- Department of Pharmacy, The 425th Hospital of PLA, Sanya, People's Republic of China
| | - Xueying Ding
- Department of Pharmaceutics, Shanghai First People's Hospital, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Xin Wu
- Department of Pharmaceutics, Shanghai First People's Hospital, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Wei Zhang
- Department of Pharmacy, Shanghai Pulmonary Hospital, Shanghai, People's Republic of China
| | - Yinghua Fu
- Department of Pharmaceutics, Medical College of Jiaxing University, Jiaxing, People's Republic of China
| | - Yueyan Huang
- Department of Pharmaceutics, Medical College of Jiaxing University, Jiaxing, People's Republic of China
| | - Xuan Huang
- Department of Pharmaceutics, Medical College of Jiaxing University, Jiaxing, People's Republic of China
| | - Rubing Chen
- Department of Pharmaceutics, Medical College of Jiaxing University, Jiaxing, People's Republic of China
| | - Mingjuan Li
- Department of Pharmaceutics, Medical College of Jiaxing University, Jiaxing, People's Republic of China
| | - Ningyin Xu
- Department of Pharmaceutics, Medical College of Jiaxing University, Jiaxing, People's Republic of China
| | - Yongxia Zheng
- Department of Pharmaceutics, Medical College of Jiaxing University, Jiaxing, People's Republic of China.
| | - Baoyue Ding
- Department of Pharmaceutics, Medical College of Jiaxing University, Jiaxing, People's Republic of China.
| |
Collapse
|
38
|
Pandey AP, Sawant KK. Polyethylenimine: A versatile, multifunctional non-viral vector for nucleic acid delivery. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 68:904-918. [DOI: 10.1016/j.msec.2016.07.066] [Citation(s) in RCA: 147] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 07/16/2016] [Accepted: 07/24/2016] [Indexed: 12/21/2022]
|
39
|
Ho W, Zhang XQ, Xu X. Biomaterials in siRNA Delivery: A Comprehensive Review. Adv Healthc Mater 2016; 5:2715-2731. [PMID: 27700013 DOI: 10.1002/adhm.201600418] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 06/07/2016] [Indexed: 01/31/2023]
Abstract
With the dearth of effective treatment options for prominent diseases including Ebola and cancer, RNA interference (RNAi), a sequence-specific mechanism for genetic regulation that can silence nearly any gene, holds the promise of unlimited potential in treating illness ever since its discovery in 1999. Given the large size, unstable tertiary structure in physiological conditions and negative charge of small interfering RNAs (siRNAs), the development of safe and effective delivery vehicles is of critical importance in order to drive the widespread use of RNAi therapeutics into clinical settings. Immense amounts of time and billions of dollars have been devoted into the design of novel and diverse delivery strategies, and there are a handful of delivery systems that have been successfully translated into clinic. This review provides an introduction to the in vivo barriers that need to be addressed by siRNA delivery systems. We also discuss the progress up to the most effective and clinically advanced siRNA delivery systems including liposomal, polymeric and siRNA conjugate delivery systems, as well as their design to overcome the challenges.
Collapse
Affiliation(s)
- William Ho
- Department of Chemical, Biological and Pharmaceutical Engineering; Newark School of Engineering; New Jersey Institute of Technology; Newark NJ 07102 USA
| | - Xue-Qing Zhang
- Department of Chemical, Biological and Pharmaceutical Engineering; Newark School of Engineering; New Jersey Institute of Technology; Newark NJ 07102 USA
| | - Xiaoyang Xu
- Department of Chemical, Biological and Pharmaceutical Engineering; Newark School of Engineering; New Jersey Institute of Technology; Newark NJ 07102 USA
| |
Collapse
|
40
|
Wei Z, Luijten E. Systematic coarse-grained modeling of complexation between small interfering RNA and polycations. J Chem Phys 2016; 143:243146. [PMID: 26723631 DOI: 10.1063/1.4937384] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
All-atom molecular dynamics simulations can provide insight into the properties of polymeric gene-delivery carriers by elucidating their interactions and detailed binding patterns with nucleic acids. However, to explore nanoparticle formation through complexation of these polymers and nucleic acids and study their behavior at experimentally relevant time and length scales, a reliable coarse-grained model is needed. Here, we systematically develop such a model for the complexation of small interfering RNA (siRNA) and grafted polyethyleneimine copolymers, a promising candidate for siRNA delivery. We compare the predictions of this model with all-atom simulations and demonstrate that it is capable of reproducing detailed binding patterns, charge characteristics, and water release kinetics. Since the coarse-grained model accelerates the simulations by one to two orders of magnitude, it will make it possible to quantitatively investigate nanoparticle formation involving multiple siRNA molecules and cationic copolymers.
Collapse
Affiliation(s)
- Zonghui Wei
- Graduate Program in Applied Physics, Northwestern University, Evanston, Illinois 60208, USA
| | - Erik Luijten
- Graduate Program in Applied Physics, Northwestern University, Evanston, Illinois 60208, USA
| |
Collapse
|
41
|
Lü JM, Liang Z, Wang X, Gu J, Yao Q, Chen C. New polymer of lactic-co-glycolic acid-modified polyethylenimine for nucleic acid delivery. Nanomedicine (Lond) 2016; 11:1971-91. [PMID: 27456396 DOI: 10.2217/nnm-2016-0128] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM To develop an improved delivery system for nucleic acids. MATERIALS & METHODS We designed, synthesized and characterized a new polymer of lactic-co-glycolic acid-modified polyethylenimine (LGA-PEI). Functions of LGA-PEI polymer were determined. RESULTS The new LGA-PEI polymer spontaneously formed nanoparticles (NPs) with DNA or RNA, and showed higher DNA or RNA loading efficiency, higher or comparable transfection efficacy, and lower cytotoxicity in several cell types including PANC-1, Jurkat and HEK293 cells, when compared with lipofectamine 2000, branched or linear PEI (25 kDa). In nude mouse models, LGA-PEI showed higher delivery efficiency of plasmid DNA or miRNA mimic into pancreatic and ovarian xenograft tumors. LGA-PEI/DNA NPs showed much lower toxicity than control PEI NPs in mouse models. CONCLUSION The new LGA-PEI polymer is a safer and more effective system to deliver DNA or RNA than PEI.
Collapse
Affiliation(s)
- Jian-Ming Lü
- Division of Surgical Research, Michael E DeBakey Department of Surgery, Baylor College of Medicine, One Plaza, Houston, TX 77030, USA
| | - Zhengdong Liang
- Division of Surgical Research, Michael E DeBakey Department of Surgery, Baylor College of Medicine, One Plaza, Houston, TX 77030, USA
| | - Xiaoxiao Wang
- Division of Surgical Research, Michael E DeBakey Department of Surgery, Baylor College of Medicine, One Plaza, Houston, TX 77030, USA
| | - Jianhua Gu
- AFM/SEM Core Facility, The Methodist Hospital Research Institute, 6670 Bertner Avenue, Houston, TX 77030, USA
| | - Qizhi Yao
- Division of Surgical Research, Michael E DeBakey Department of Surgery, Baylor College of Medicine, One Plaza, Houston, TX 77030, USA.,Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center, Houston, TX 77030, USA
| | - Changyi Chen
- Division of Surgical Research, Michael E DeBakey Department of Surgery, Baylor College of Medicine, One Plaza, Houston, TX 77030, USA
| |
Collapse
|
42
|
Kim YD, Pofali P, Park TE, Singh B, Cho K, Maharjan S, Dandekar P, Jain R, Choi YJ, Arote R, Cho CS. Gene therapy for bone tissue engineering. Tissue Eng Regen Med 2016; 13:111-125. [PMID: 30603391 PMCID: PMC6170855 DOI: 10.1007/s13770-016-9063-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 09/24/2015] [Accepted: 09/29/2015] [Indexed: 02/06/2023] Open
Abstract
Gene therapy holds a great promise and has been extensively investigated to improve bone formation and regeneration therapies in bone tissue engineering. A variety of osteogenic genes can be delivered by combining different vectors (viral or non-viral), scaffolds and delivery methodologies. Ex vivo & in vivo gene enhanced tissue engineering approaches have led to successful osteogenic differentiation and bone formation. In this article, we review recent advances of gene therapy-based bone tissue engineering discussing strengths and weaknesses of various strategies as well as general overview of gene therapy.
Collapse
Affiliation(s)
- Young-Dong Kim
- Department of Molecular Genetics, School of Dentistry, Seoul National University, Seoul, Korea
| | - Prasad Pofali
- Department of Chemical Engineering, Institute of Chemical Technology, Mumbai, India
| | - Tae-Eun Park
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Korea
| | - Bijay Singh
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Korea
| | - Kihyun Cho
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Korea
| | - Sushila Maharjan
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Korea
| | - Prajakta Dandekar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
| | - Ratnesh Jain
- Department of Chemical Engineering, Institute of Chemical Technology, Mumbai, India
| | - Yun-Jaie Choi
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Korea
| | - Rohidas Arote
- Department of Molecular Genetics, School of Dentistry, Seoul National University, Seoul, Korea
| | - Chong-Su Cho
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Korea
| |
Collapse
|
43
|
Williford JM, Archang MM, Minn I, Ren Y, Wo M, Vandermark J, Fisher PB, Pomper MG, Mao HQ. Critical Length of PEG Grafts on lPEI/DNA Nanoparticles for Efficient in Vivo Delivery. ACS Biomater Sci Eng 2016; 2:567-578. [PMID: 27088129 PMCID: PMC4829937 DOI: 10.1021/acsbiomaterials.5b00551] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 03/02/2016] [Indexed: 12/03/2022]
Abstract
![]()
Nanoparticle-mediated
gene delivery is a promising alternative
to viral methods; however, its use in vivo, particularly following
systemic injection, has suffered from poor delivery efficiency. Although
PEGylation of nanoparticles has been successfully demonstrated as
a strategy to enhance colloidal stability, its success in improving
delivery efficiency has been limited, largely due to reduced cell
binding and uptake, leading to poor transfection efficiency. Here
we identified an optimized PEGylation scheme for DNA micellar nanoparticles
that delivers balanced colloidal stability and transfection activity.
Using linear polyethylenimine (lPEI)-g-PEG as a carrier,
we characterized the effect of graft length and density of polyethylene
glycol (PEG) on nanoparticle assembly, micelle stability, and gene
delivery efficiency. Through variation of PEG grafting degree, lPEI
with short PEG grafts (molecular weight, MW 500–700 Da) generated
micellar nanoparticles with various shapes including spherical, rodlike,
and wormlike nanoparticles. DNA micellar nanoparticles prepared with
short PEG grafts showed comparable colloidal stability in salt and
serum-containing media to those prepared with longer PEG grafts (MW
2 kDa). Corresponding to this trend, nanoparticles prepared with short
PEG grafts displayed significantly higher in vitro transfection efficiency
compared to those with longer PEG grafts. More importantly, short
PEG grafts permitted marked increase in transfection efficiency following
ligand conjugation to the PEG terminal in metastatic prostate cancer-bearing
mice. This study identifies that lPEI-g-PEG with
short PEG grafts (MW 500–700 Da) is the most effective to ensure
shape control and deliver high colloidal stability, transfection activity,
and ligand effect for DNA nanoparticles in vitro and in vivo following
intravenous administration.
Collapse
Affiliation(s)
- John-Michael Williford
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, Maryland 21205, United States; Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States
| | - Maani M Archang
- Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States; Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States
| | - Il Minn
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins Medical Institutions , 601 N. Caroline Street, Baltimore, Maryland 21287, United States
| | - Yong Ren
- Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States; Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States
| | - Mark Wo
- Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States; Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States
| | - John Vandermark
- Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States; Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, 1101 East Marshall Street, Richmond, Virginia 23298, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, 1220 East Broad Street, Richmond, Virginia 23298, United States; VCU Massey Cancer Center, Virginia Commonwealth University, 401 College Street, Richmond, Virginia 23298, United States
| | - Martin G Pomper
- Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States; Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States; Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins Medical Institutions, 601 N. Caroline Street, Baltimore, Maryland 21287, United States
| | - Hai-Quan Mao
- Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States; Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States; Translational Tissue Engineering Center and Whitaker Biomedical Engineering Institute, Johns Hopkins University School of Medicine, 400 N. Broadway, Baltimore, Maryland 21287, United States
| |
Collapse
|
44
|
Targeting CXCR4/SDF-1 axis by lipopolymer complexes of siRNA in acute myeloid leukemia. J Control Release 2016; 224:8-21. [DOI: 10.1016/j.jconrel.2015.12.052] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 12/22/2015] [Accepted: 12/28/2015] [Indexed: 02/07/2023]
|
45
|
Englert C, Hartlieb M, Bellstedt P, Kempe K, Yang C, Chu SK, Ke X, Garcı́a JM, Ono RJ, Fevre M, Wojtecki RJ, Schubert US, Yang YY, Hedrick JL. Enhancing the Biocompatibility and Biodegradability of Linear Poly(ethylene imine) through Controlled Oxidation. Macromolecules 2015. [DOI: 10.1021/acs.macromol.5b01940] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Christoph Englert
- Laboratory of Organic and Macromolecular
Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse
10, 07743 Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
- IBM Almaden
Research Center, 650 Harry Road, San Jose, California 95120, United States
| | - Matthias Hartlieb
- Laboratory of Organic and Macromolecular
Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse
10, 07743 Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Peter Bellstedt
- Laboratory of Organic and Macromolecular
Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse
10, 07743 Jena, Germany
| | - Kristian Kempe
- Laboratory of Organic and Macromolecular
Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse
10, 07743 Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Chuan Yang
- Institute of Bioengineering
and Nanotechnology, 31 Biopolis Way,
The Nanos, Singapore 138669, Singapore
| | - Swee Kwang Chu
- Institute of Bioengineering
and Nanotechnology, 31 Biopolis Way,
The Nanos, Singapore 138669, Singapore
| | - Xiyu Ke
- Institute of Bioengineering
and Nanotechnology, 31 Biopolis Way,
The Nanos, Singapore 138669, Singapore
| | - Jeannette M. Garcı́a
- IBM Almaden
Research Center, 650 Harry Road, San Jose, California 95120, United States
| | - Robert J. Ono
- IBM Almaden
Research Center, 650 Harry Road, San Jose, California 95120, United States
| | - Mareva Fevre
- IBM Almaden
Research Center, 650 Harry Road, San Jose, California 95120, United States
| | - Rudy J. Wojtecki
- IBM Almaden
Research Center, 650 Harry Road, San Jose, California 95120, United States
| | - Ulrich S. Schubert
- Laboratory of Organic and Macromolecular
Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse
10, 07743 Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Yi Yan Yang
- Institute of Bioengineering
and Nanotechnology, 31 Biopolis Way,
The Nanos, Singapore 138669, Singapore
| | - James L. Hedrick
- IBM Almaden
Research Center, 650 Harry Road, San Jose, California 95120, United States
| |
Collapse
|
46
|
Zarogoulidis P, Domvri K, Huang H, Zarogoulidis K. Gene therapy for lung cancer malignant pleural effusion: current and future nano-biotechnology. Transl Lung Cancer Res 2015; 1:234-7. [PMID: 25806188 DOI: 10.3978/j.issn.2218-6751.2012.08.01] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Accepted: 08/20/2012] [Indexed: 01/21/2023]
Affiliation(s)
- Paul Zarogoulidis
- Pulmonary Department-Oncology Unit, "G. Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Kalliopi Domvri
- Pulmonary Department-Oncology Unit, "G. Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Haidong Huang
- Department of Respiratory diseases, Changhai hospital, Yangpu District, Shanghai, China
| | - Konstantinos Zarogoulidis
- Pulmonary Department-Oncology Unit, "G. Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
47
|
Navarro G, Pan J, Torchilin VP. Micelle-like nanoparticles as carriers for DNA and siRNA. Mol Pharm 2015; 12:301-13. [PMID: 25557580 DOI: 10.1021/mp5007213] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Gene therapy represents a potential efficient approach of disease prevention and therapy. However, due to their poor in vivo stability, gene molecules need to be associated with delivery systems to overcome extracellular and intracellular barriers and allow access to the site of action. Cationic polymeric nanoparticles are popular carriers for small interfering RNA (siRNA) and DNA-based therapeutics for which efficient and safe delivery are important factors that need to be optimized. Micelle-like nanoparticles (MNP) (half micelles, half polymeric nanoparticles) can overcome some of the disadvantages of such cationic carriers by unifying in one single carrier the best of both delivery systems. In this review, we will discuss how the unique properties of MNP including self-assembly, condensation and protection of nucleic acids, improved cell association and gene transfection, and low toxicity may contribute to the successful application of siRNA- and DNA-based therapeutics into the clinic. Recent developments of MNP involving the addition of stimulus-sensitive functions to respond specifically to pathological or externally applied "triggers" (e.g., temperature, pH or enzymatic catalysis, light, or magnetic fields) will be discussed. Finally, we will overview the use of MNP as two-in-one carriers for the simultaneous delivery of different agents (small molecules, imaging agents) and nucleic acid combinations.
Collapse
Affiliation(s)
- Gemma Navarro
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University , Boston, Massachusetts 02115, United States
| | | | | |
Collapse
|
48
|
Yue P, Zhang Y, Guo ZF, Cao AC, Lu ZL, Zhai YG. Synthesis of bifunctional molecules containing [12]aneN3 and coumarin moieties as effective DNA condensation agents and new non-viral gene vectors. Org Biomol Chem 2015; 13:4494-505. [DOI: 10.1039/c4ob02676d] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Bifunctional molecules with different combinations of [12]aneN3 and coumarin moieties were successfully applied in DNA condensation and gene transfection.
Collapse
Affiliation(s)
- Pan Yue
- College of Chemistry
- Beijing Normal University
- Beijing
- China
| | - Ying Zhang
- College of Chemistry
- Beijing Normal University
- Beijing
- China
| | - Zhi-Fo Guo
- College of Chemistry
- Beijing Normal University
- Beijing
- China
- College of Life Science
| | - Ao-Cheng Cao
- Institute of Plant Protection
- Chinese Academy of Agricultural Sciences
- Beijing
- China
| | - Zhong-Lin Lu
- College of Chemistry
- Beijing Normal University
- Beijing
- China
| | - Yong-Gong Zhai
- College of Life Science
- Beijing Normal University
- Beijing
- China
| |
Collapse
|
49
|
|
50
|
Hong SH, Park SJ, Lee S, Cho CS, Cho MH. Aerosol gene delivery using viral vectors and cationic carriers forin vivolung cancer therapy. Expert Opin Drug Deliv 2014; 12:977-91. [DOI: 10.1517/17425247.2015.986454] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|