1
|
Jamil MU, Waheed NK. Gene therapy for geographic atrophy in age-related macular degeneration: current insights. Eye (Lond) 2025; 39:274-283. [PMID: 39578546 DOI: 10.1038/s41433-024-03463-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 10/07/2024] [Accepted: 11/04/2024] [Indexed: 11/24/2024] Open
Abstract
Geographic atrophy (GA) is the advanced stage of non-neovascular (dry) age-related macular degeneration, defined by the presence of sharply demarcated atrophic lesions of the outer retina. The complement system is integral to the body's natural immune response, and hence its overactivation can lead to tissue damage and inflammation. It has been shown to play a significant role in GA lesion development and progression, and therefore, complement inhibition is emerging as a promising avenue for therapeutic intervention. With the recent approval by the Food and Drug Administration of drugs like SYFOVRE™ (pegcetacoplan injection) and IZERVAY™ (avacincaptad pegol intravitreal solution), there is hope for the development of interventions capable of slowing down or arresting the progression of GA. In particular, gene therapy intervention is gaining traction for halting GA atrophy at the source of our genes. The concept is to insert a gene into the eye that will act as an ocular "bio-factory," producing a desired protein. This can either lead to overproduction of an already available protein or produce a substance not typically generated in the eye. This review aims to provide an overview of the present understanding of GA, encompassing risk factors, prevalence, pathophysiology, and genetic associations. It will also highlight the current landscape of GA treatment, with particular emphasis on gene therapy intervention.
Collapse
Affiliation(s)
| | - Nadia K Waheed
- New England Eye Center, Tufts Medical Center, Boston, MA, 02116, USA.
| |
Collapse
|
2
|
Yan W, Cao Y, Yin Q, Li Y. Biomimetic Nucleic Acid Drug Delivery Systems for Relieving Tumor Immunosuppressive Microenvironment. Pharmaceutics 2024; 16:1028. [PMID: 39204373 PMCID: PMC11360391 DOI: 10.3390/pharmaceutics16081028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/19/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Immunotherapy combats tumors by enhancing the body's immune surveillance and clearance of tumor cells. Various nucleic acid drugs can be used in immunotherapy, such as DNA expressing cytokines, mRNA tumor vaccines, small interfering RNAs (siRNA) knocking down immunosuppressive molecules, and oligonucleotides that can be used as immune adjuvants. Nucleic acid drugs, which are prone to nuclease degradation in the circulation and find it difficult to enter the target cells, typically necessitate developing appropriate vectors for effective in vivo delivery. Biomimetic drug delivery systems, derived from viruses, bacteria, and cells, can protect the cargos from degradation and clearance, and deliver them to the target cells to ensure safety. Moreover, they can activate the immune system through their endogenous activities and active components, thereby improving the efficacy of antitumor immunotherapeutic nucleic acid drugs. In this review, biomimetic nucleic acid delivery systems for relieving a tumor immunosuppressive microenvironment are introduced. Their immune activation mechanisms, including upregulating the proinflammatory cytokines, serving as tumor vaccines, inhibiting immune checkpoints, and modulating intratumoral immune cells, are elaborated. The advantages and disadvantages, as well as possible directions for their clinical translation, are summarized at last.
Collapse
Affiliation(s)
- Wenlu Yan
- State Key Laboratory of Drug Research and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; (W.Y.); (Y.C.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ying Cao
- State Key Laboratory of Drug Research and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; (W.Y.); (Y.C.)
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Qi Yin
- State Key Laboratory of Drug Research and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; (W.Y.); (Y.C.)
- University of Chinese Academy of Sciences, Beijing 100049, China
- Yantai Key Laboratory of Nanomedicine and Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
| | - Yaping Li
- State Key Laboratory of Drug Research and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; (W.Y.); (Y.C.)
- University of Chinese Academy of Sciences, Beijing 100049, China
- Yantai Key Laboratory of Nanomedicine and Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264000, China
| |
Collapse
|
3
|
Prasher P, Sharma M, Agarwal V, Singh SK, Gupta G, Dureja H, Dua K. Cationic cycloamylose based nucleic acid nanocarriers. Chem Biol Interact 2024; 395:111000. [PMID: 38614318 DOI: 10.1016/j.cbi.2024.111000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 04/02/2024] [Accepted: 04/07/2024] [Indexed: 04/15/2024]
Abstract
Nucleic acid delivery by viral and non-viral methods has been a cornerstone for the contemporary gene therapy aimed at correcting the defective genes, replacing of the missing genes, or downregulating the expression of anomalous genes is highly desirable for the management of various diseases. Ostensibly, it becomes paramount for the delivery vectors to intersect the biological barriers for accessing their destined site within the cellular environment. However, the lipophilic nature of biological membranes and their potential to limit the entry of large sized, charged, hydrophilic molecules thus presenting a sizeable challenge for the cellular integration of negatively charged nucleic acids. Furthermore, the susceptibility of nucleic acids towards the degrading enzymes (nucleases) in the lysosomes present in cytoplasm is another matter of concern for their cellular and nuclear delivery. Hence, there is a pressing need for the identification and development of cationic delivery systems which encapsulate the cargo nucleic acids where the charge facilitates their cellular entry by evading the membrane barriers, and the encapsulation shields them from the enzymatic attack in cytoplasm. Cycloamylose bearing a closed loop conformation presents a robust candidature in this regard owing to its remarkable encapsulating tendency towards nucleic acids including siRNA, CpG DNA, and siRNA. The presence of numerous hydroxyl groups on the cycloamylose periphery provides sites for its chemical modification for the introduction of cationic groups, including spermine, (3-Chloro-2 hydroxypropyl) trimethylammonium chloride (Q188), and diethyl aminoethane (DEAE). The resulting cationic cycloamylose possesses a remarkable transfection efficiency and provides stability to cargo oligonucleotides against endonucleases, in addition to modulating the undesirable side effects such as unwanted immune stimulation. Cycloamylose is known to interact with the cell membranes where they release certain membrane components such as phospholipids and cholesterol thereby resulting in membrane destabilization and permeabilization. Furthermore, cycloamylose derivatives also serve as formulation excipients for improving the efficiency of other gene delivery systems. This review delves into the various vector and non-vector-based gene delivery systems, their advantages, and limitations, eventually leading to the identification of cycloamylose as an ideal candidate for nucleic acid delivery. The synthesis of cationic cycloamylose is briefly discussed in each section followed by its application for specific delivery/transfection of a particular nucleic acid.
Collapse
Affiliation(s)
- Parteek Prasher
- Department of Chemistry, University of Petroleum & Energy Studies, Energy Acres, Dehradun, 248007, India.
| | - Mousmee Sharma
- Department of Chemistry, Uttaranchal University, Dehradun, 248007, India
| | - Vipul Agarwal
- Cluster for Advanced Macromolecular Design (CAMD), School of Chemical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India; Faculty of Health, Australian Research Center in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia; School of Medical and Life Sciences, Sunway University, 47500 Sunway City, Malaysia
| | - Gaurav Gupta
- School of Pharmacy, Graphic Era Hill University, Dehradun, 248007, India; Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman 346, United Arab Emirates
| | - Harish Dureja
- Department of Pharmaceutical Sciences, Maharishi Dayanand University, Rohtak, 124001, India
| | - Kamal Dua
- Faculty of Health, Australian Research Center in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
| |
Collapse
|
4
|
Wu J, Zheng X, Lin W, Chen L, Wu ZS. Persistent Targeting DNA Nanocarrier Made of 3D Structural Unit Assembled from Only One Basic Multi-Palindromic Oligonucleotide for Precise Gene Cancer Therapy. Adv Healthc Mater 2024; 13:e2303865. [PMID: 38289018 DOI: 10.1002/adhm.202303865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/19/2024] [Indexed: 02/13/2024]
Abstract
Construction of a simple, reconfigurable, and stimuli-responsive DNA nanocarrier remains a technical challenge. In this contribution, by designing three palindromic fragments, a simplest four-sticky end-contained 3D structural unit (PS-unit) made of two same DNA components is proposed. Via regulating the rotation angle of central longitudinal axis of PS-unit, the oriented assembly of one-component spherical architecture is accomplished with high efficiency. Introduction of an aptamer and sticky tail warehouse into one component creates a size-change-reversible targeted siRNA delivery nanovehicle. Volume swelling of 20 nm allows one carrier to load 1987 siPLK1s. Once entering cancer cells and responding to glutathione (GSH) stimuli, siPLK1s are almost 100% released and original size of nanovehicle is restored, inhibiting the expression of PLK1 protein and substantially suppressing tumor growth (superior to commercial transfection agents) in tumor-bearing mice without systemic toxicity.
Collapse
Affiliation(s)
- Jingting Wu
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Xiaoqi Zheng
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Wenqing Lin
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Linhuan Chen
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Zai-Sheng Wu
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| |
Collapse
|
5
|
Zhu Y, Arkin G, Zeng W, Huang Y, Su L, Guo F, Ye J, Wen G, Xu J, Liu Y. Ultrasound image-guided cancer gene therapy using iRGD dual-targeted magnetic cationic microbubbles. Biomed Pharmacother 2024; 172:116221. [PMID: 38306843 DOI: 10.1016/j.biopha.2024.116221] [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: 08/31/2023] [Revised: 01/25/2024] [Accepted: 01/25/2024] [Indexed: 02/04/2024] Open
Abstract
The gene therapy attracted more and more attention for the tumor therapy. To obtain a safe gene therapy system, the new gene vectors beyond the virus were developed for a high gene therapy efficiency. The ultrasound mediated gene therapy was safer and the plasmid DNA could be delivered by the microbubbles and combined with the ultrasound to increase the gene transfection efficiency. In this work, the cationic microbubbles decorated with Cyclo(Cys-Arg-Gly-Asp-Lys-Gly-Pro-AspCys) (iRGD peptides) and magnetic Fe3O4 nanoparticles (MBiM) was designed for targeted ultrasound contrast imaging guided gene therapy of tumors. The ultrasound image intensity was dramatically enhanced at the tumor site that received MBiM with the magnet applied, compared to those administrated the non-targeted microbubbles (MBb) or the microbubbles with only one target material on the surface (MBM and MBbi). The pGPU6/GFP/Neo-shAKT2 was used as a sample gene, which down regulate the AKT2 protein expression for the cancer therapy. It illustrated that MBiM/AKT2 had the highest gene transfection efficiency in the studied microbubbles mediated by the ultrasound, leading to the AKT2 protein expression downregulation and the strongest tumor killing effect in vitro and in vivo. In summary, a novel and biocompatible gene delivery platform via MBiM with both the endogenous and external targeting effects for breast cancer theranostics was developed.
Collapse
Affiliation(s)
- Yao Zhu
- Shenzhen Medical Ultrasound Engineering Center, Department of Ultrasonography, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, First Clinical Medical College of Southern University of Science and Technology, Shenzhen 518020, China
| | - Gulzira Arkin
- Shenzhen Medical Ultrasound Engineering Center, Department of Ultrasonography, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, First Clinical Medical College of Southern University of Science and Technology, Shenzhen 518020, China
| | - Wei Zeng
- Shenzhen Medical Ultrasound Engineering Center, Department of Ultrasonography, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, First Clinical Medical College of Southern University of Science and Technology, Shenzhen 518020, China
| | - Yalan Huang
- Shenzhen Medical Ultrasound Engineering Center, Department of Ultrasonography, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, First Clinical Medical College of Southern University of Science and Technology, Shenzhen 518020, China
| | - Lili Su
- Shenzhen Medical Ultrasound Engineering Center, Department of Ultrasonography, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, First Clinical Medical College of Southern University of Science and Technology, Shenzhen 518020, China
| | - Fengjuan Guo
- Shenzhen Medical Ultrasound Engineering Center, Department of Ultrasonography, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, First Clinical Medical College of Southern University of Science and Technology, Shenzhen 518020, China
| | - Jiayu Ye
- Shenzhen Medical Ultrasound Engineering Center, Department of Ultrasonography, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, First Clinical Medical College of Southern University of Science and Technology, Shenzhen 518020, China
| | - Guanxi Wen
- Shenzhen Medical Ultrasound Engineering Center, Department of Ultrasonography, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, First Clinical Medical College of Southern University of Science and Technology, Shenzhen 518020, China
| | - Jinfeng Xu
- Shenzhen Medical Ultrasound Engineering Center, Department of Ultrasonography, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, First Clinical Medical College of Southern University of Science and Technology, Shenzhen 518020, China.
| | - Yingying Liu
- Shenzhen Medical Ultrasound Engineering Center, Department of Ultrasonography, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, First Clinical Medical College of Southern University of Science and Technology, Shenzhen 518020, China.
| |
Collapse
|
6
|
Alkharobi H. Exploring Various Transfection Approaches and Their Applications in Studying the Regenerative Potential of Dental Pulp Stem Cells. Curr Issues Mol Biol 2023; 45:10026-10040. [PMID: 38132472 PMCID: PMC10742526 DOI: 10.3390/cimb45120626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 12/23/2023] Open
Abstract
Transfection is a contemporary approach for introducing foreign genetic material into target cells. The effective transport of genetic materials into cells is mostly influenced by (a) the characteristics of the genetic material (quantity and quality), (b) the transfection procedure (incubation time, ratio of the reagents to the introduced genetic material, and components of cell culture), and (c) targeted cells for transfection (cell origin and cell type). This review summarizes the findings of different studies focusing on various transfection approaches and their applications to explore the regenerative potential of dental pulp stem cells (DPSCs). Several databases, including Scopus, Google Scholar, and PubMed, were searched to obtain the literature for the current review. Different keywords were used as key terms in the search. Approximately 200 articles were retained after removing duplicates from different databases. Articles published in English that discussed different transfection approaches were included. Several sources were excluded because they did not meet the inclusion criteria. Approximately 70 relevant published sources were included in the final stage to achieve the study objectives. This review demonstrated that no single transfection system is applicable to all cases and the various cell types with no side effects. Further studies are needed to focus on optimizing process parameters, decreasing the toxicity and side effects of available transfection techniques, and increasing their efficiencies. Moreover, this review sheds light on the impact of using different valuable transfection approaches to investigate the regenerative potential of DPSCs.
Collapse
Affiliation(s)
- Hanaa Alkharobi
- Department of Oral Biology, College of Dentistry, King Abdul-Aziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
7
|
Zhao Y, Gan L, Ke D, Chen Q, Fu Y. Mechanisms and research advances in mRNA antibody drug-mediated passive immunotherapy. J Transl Med 2023; 21:693. [PMID: 37794448 PMCID: PMC10552228 DOI: 10.1186/s12967-023-04553-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 09/22/2023] [Indexed: 10/06/2023] Open
Abstract
Antibody technology is widely used in the fields of biomedical and clinical therapies. Nonetheless, the complex in vitro expression of recombinant proteins, long production cycles, and harsh storage conditions have limited their applications in medicine, especially in clinical therapies. Recently, this dilemma has been overcome to a certain extent by the development of mRNA delivery systems, in which antibody-encoding mRNAs are enclosed in nanomaterials and delivered to the body. On entering the cytoplasm, the mRNAs immediately bind to ribosomes and undergo translation and post-translational modifications. This process produces monoclonal or bispecific antibodies that act directly on the patient. Additionally, it eliminates the cumbersome process of in vitro protein expression and extends the half-life of short-lived proteins, which significantly reduces the cost and duration of antibody production. This review focuses on the benefits and drawbacks of mRNA antibodies compared with the traditional in vitro expressed antibodies. In addition, it elucidates the progress of mRNA antibodies in the prevention of infectious diseases and oncology therapy.
Collapse
Affiliation(s)
- Yuxiang Zhao
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian, PR China
| | - Linchuan Gan
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian, PR China
| | - Dangjin Ke
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian, PR China
| | - Qi Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian, PR China.
| | - Yajuan Fu
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian, PR China.
| |
Collapse
|
8
|
García Coll J, Ulrich S. Nucleic-Acid-Templated Synthesis of Smart Polymer Vectors for Gene Delivery. Chembiochem 2023; 24:e202300333. [PMID: 37401911 DOI: 10.1002/cbic.202300333] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/12/2023] [Accepted: 07/04/2023] [Indexed: 07/05/2023]
Abstract
Nucleic acids are information-rich and readily available biomolecules, which can be used to template the polymerization of synthetic macromolecules. Here, we highlight the control over the size, composition, and sequence one can nowadays obtain by using this methodology. We also highlight how templated processes exploiting dynamic covalent polymerization can, in return, result in therapeutic nucleic acids fabricating their own dynamic delivery vector - a biomimicking concept that can provide original solutions for gene therapies.
Collapse
Affiliation(s)
- José García Coll
- IBMM, Institut des Biomolécules Max Mousseron, Université de Montpellier, CNRS, ENSCM, 34095, Montpellier, France
| | - Sébastien Ulrich
- IBMM, Institut des Biomolécules Max Mousseron, Université de Montpellier, CNRS, ENSCM, 34095, Montpellier, France
| |
Collapse
|
9
|
Yadav K, Sahu KK, Sucheta, Gnanakani SPE, Sure P, Vijayalakshmi R, Sundar VD, Sharma V, Antil R, Jha M, Minz S, Bagchi A, Pradhan M. Biomedical applications of nanomaterials in the advancement of nucleic acid therapy: Mechanistic challenges, delivery strategies, and therapeutic applications. Int J Biol Macromol 2023; 241:124582. [PMID: 37116843 DOI: 10.1016/j.ijbiomac.2023.124582] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 04/30/2023]
Abstract
In the past few decades, substantial advancement has been made in nucleic acid (NA)-based therapies. Promising treatments include mRNA, siRNA, miRNA, and anti-sense DNA for treating various clinical disorders by modifying the expression of DNA or RNA. However, their effectiveness is limited due to their concentrated negative charge, instability, large size, and host barriers, which make widespread application difficult. The effective delivery of these medicines requires safe vectors that are efficient & selective while having non-pathogenic qualities; thus, nanomaterials have become an attractive option with promising possibilities despite some potential setbacks. Nanomaterials possess ideal characteristics, allowing them to be tuned into functional bio-entity capable of targeted delivery. In this review, current breakthroughs in the non-viral strategy of delivering NAs are discussed with the goal of overcoming challenges that would otherwise be experienced by therapeutics. It offers insight into a wide variety of existing NA-based therapeutic modalities and techniques. In addition to this, it provides a rationale for the use of non-viral vectors and a variety of nanomaterials to accomplish efficient gene therapy. Further, it discusses the potential for biomedical application of nanomaterials-based gene therapy in various conditions, such as cancer therapy, tissue engineering, neurological disorders, and infections.
Collapse
Affiliation(s)
- Krishna Yadav
- Raipur Institute of Pharmaceutical Education and Research, Sarona, Raipur, Chhattisgarh 492010, India
| | - Kantrol Kumar Sahu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh 281406, India
| | - Sucheta
- School of Medical and Allied Sciences, K. R. Mangalam University, Gurugram, Haryana 122103, India
| | | | - Pavani Sure
- Department of Pharmaceutics, Vignan Institute of Pharmaceutical Sciences, Hyderabad, Telangana, India
| | - R Vijayalakshmi
- Department of Pharmaceutical Analysis, GIET School of Pharmacy, Chaitanya Knowledge City, Rajahmundry, AP 533296, India
| | - V D Sundar
- Department of Pharmaceutical Technology, GIET School of Pharmacy, Chaitanya Knowledge City, Rajahmundry, AP 533296, India
| | - Versha Sharma
- Department of Biotechnology, School of Biological Sciences, Dr. Harisingh Gour Central University, Sagar, M.P. 470003, India
| | - Ruchita Antil
- Addenbrookes Hospital, Cambridge University Hospitals NHS Foundation Trust, England, United Kingdom of Great Britain and Northern Ireland
| | - Megha Jha
- Department of Biotechnology, School of Biological Sciences, Dr. Harisingh Gour Central University, Sagar, M.P. 470003, India
| | - Sunita Minz
- Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, M.P., 484887, India
| | - Anindya Bagchi
- Tumor Initiation & Maintenance Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road La Jolla, CA 92037, USA
| | | |
Collapse
|
10
|
Zhu J, Batra H, Ananthaswamy N, Mahalingam M, Tao P, Wu X, Guo W, Fokine A, Rao VB. Design of bacteriophage T4-based artificial viral vectors for human genome remodeling. Nat Commun 2023; 14:2928. [PMID: 37253769 PMCID: PMC10229621 DOI: 10.1038/s41467-023-38364-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 04/27/2023] [Indexed: 06/01/2023] Open
Abstract
Designing artificial viral vectors (AVVs) programmed with biomolecules that can enter human cells and carry out molecular repairs will have broad applications. Here, we describe an assembly-line approach to build AVVs by engineering the well-characterized structural components of bacteriophage T4. Starting with a 120 × 86 nm capsid shell that can accommodate 171-Kbp DNA and thousands of protein copies, various combinations of biomolecules, including DNAs, proteins, RNAs, and ribonucleoproteins, are externally and internally incorporated. The nanoparticles are then coated with cationic lipid to enable efficient entry into human cells. As proof of concept, we assemble a series of AVVs designed to deliver full-length dystrophin gene or perform various molecular operations to remodel human genome, including genome editing, gene recombination, gene replacement, gene expression, and gene silencing. These large capacity, customizable, multiplex, and all-in-one phage-based AVVs represent an additional category of nanomaterial that could potentially transform gene therapies and personalized medicine.
Collapse
Affiliation(s)
- Jingen Zhu
- Bacteriophage Medical Research Center, Department of Biology, The Catholic University of America, Washington, DC, 20064, USA
| | - Himanshu Batra
- Bacteriophage Medical Research Center, Department of Biology, The Catholic University of America, Washington, DC, 20064, USA
| | - Neeti Ananthaswamy
- Bacteriophage Medical Research Center, Department of Biology, The Catholic University of America, Washington, DC, 20064, USA
| | - Marthandan Mahalingam
- Bacteriophage Medical Research Center, Department of Biology, The Catholic University of America, Washington, DC, 20064, USA
| | - Pan Tao
- Bacteriophage Medical Research Center, Department of Biology, The Catholic University of America, Washington, DC, 20064, USA
| | - Xiaorong Wu
- Bacteriophage Medical Research Center, Department of Biology, The Catholic University of America, Washington, DC, 20064, USA
| | - Wenzheng Guo
- Bacteriophage Medical Research Center, Department of Biology, The Catholic University of America, Washington, DC, 20064, USA
| | - Andrei Fokine
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Venigalla B Rao
- Bacteriophage Medical Research Center, Department of Biology, The Catholic University of America, Washington, DC, 20064, USA.
| |
Collapse
|
11
|
Petukhov IA, Puchkov PA, Morozova NG, Zenkova MA, Maslov MA. The Synthesis and Transfection Activity of Disulfide Polycationic Amphiphiles. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2023; 49:41-51. [PMID: 37192981 PMCID: PMC10156423 DOI: 10.1134/s1068162023010235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 07/18/2022] [Accepted: 08/18/2022] [Indexed: 05/18/2023]
Abstract
Some new polycationic amphiphiles containing a disulfide group were synthesized. Cationic liposomes formed from the compounds synthesized and a helper lipid 1,2-dioleoyl-sn-glycero-3-phosphatidylethanolamine were not toxic for HEK293 and HeLa cells and were highly effective when delivering a fluorescently labeled oligodeoxyribonucleotide. The efficacy of plasmid DNA delivery depended on the cell line and the amphiphile structure, liposomes based on tetracationic amphiphiles being the most effective transfectants. These liposomes can be used for in vitro transfection of eukaryotic cells as well as for further in vivo biological studies.
Collapse
Affiliation(s)
- I. A. Petukhov
- Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, 119571 Moscow, Russia
| | - P. A. Puchkov
- Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, 119571 Moscow, Russia
| | - N. G. Morozova
- Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, 119571 Moscow, Russia
| | - M. A. Zenkova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - M. A. Maslov
- Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, 119571 Moscow, Russia
| |
Collapse
|
12
|
Gretskaya N, Akimov M, Andreev D, Zalygin A, Belitskaya E, Zinchenko G, Fomina-Ageeva E, Mikhalyov I, Vodovozova E, Bezuglov V. Multicomponent Lipid Nanoparticles for RNA Transfection. Pharmaceutics 2023; 15:pharmaceutics15041289. [PMID: 37111773 PMCID: PMC10141487 DOI: 10.3390/pharmaceutics15041289] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/11/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Despite the wide variety of available cationic lipid platforms for the delivery of nucleic acids into cells, the optimization of their composition has not lost its relevance. The purpose of this work was to develop multi-component cationic lipid nanoparticles (LNPs) with or without a hydrophobic core from natural lipids in order to evaluate the efficiency of LNPs with the widely used cationic lipoid DOTAP (1,2-dioleoyloxy-3-[trimethylammonium]-propane) and the previously unstudied oleoylcholine (Ol-Ch), as well as the ability of LNPs containing GM3 gangliosides to transfect cells with mRNA and siRNA. LNPs containing cationic lipids, phospholipids and cholesterol, and surfactants were prepared according to a three-stage procedure. The average size of the resulting LNPs was 176 nm (PDI 0.18). LNPs with DOTAP mesylate were more effective than those with Ol-Ch. Core LNPs demonstrated low transfection activity compared with bilayer LNPs. The type of phospholipid in LNPs was significant for the transfection of MDA-MB-231 and SW 620 cancer cells but not HEK 293T cells. LNPs with GM3 gangliosides were the most efficient for the delivery of mRNA to MDA-MB-231 cells and siRNA to SW620 cells. Thus, we developed a new lipid platform for the efficient delivery of RNA of various sizes to mammalian cells.
Collapse
Affiliation(s)
- Nataliya Gretskaya
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Mikhail Akimov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Dmitry Andreev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Anton Zalygin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
- Department of Translational Medicine, National Research Nuclear University, Moscow Engineering Physics Institute, Moscow 115409, Russia
| | - Ekaterina Belitskaya
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
- Department of Translational Medicine, National Research Nuclear University, Moscow Engineering Physics Institute, Moscow 115409, Russia
| | - Galina Zinchenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Elena Fomina-Ageeva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Ilya Mikhalyov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Elena Vodovozova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Vladimir Bezuglov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| |
Collapse
|
13
|
Su DD, Ali LMA, Coste M, Laroui N, Bessin Y, Barboiu M, Bettache N, Ulrich S. Structure-Activity Relationships in Nucleic-Acid-Templated Vectors Based on Peptidic Dynamic Covalent Polymers. Chemistry 2023; 29:e202202921. [PMID: 36342312 PMCID: PMC10108046 DOI: 10.1002/chem.202202921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/26/2022] [Accepted: 11/07/2022] [Indexed: 11/09/2022]
Abstract
The use of nucleic acids as templates, which can trigger the self-assembly of their own vectors represent an emerging, simple and versatile, approach toward the self-fabrication of tailored nucleic acids delivery vectors. However, the structure-activity relationships governing this complex templated self-assembly process that accompanies the complexation of nucleic acids remains poorly understood. Herein, the class of arginine-rich dynamic covalent polymers (DCPs) composed of different monomers varying the number and position of arginines were studied. The combinations that lead to nucleic acid complexation, in saline buffer, using different templates, from short siRNA to long DNA, are described. Finally, a successful peptidic DCP featuring six-arginine repeating unit that promote the safe and effective delivery of siRNA in live cancer cells was identified.
Collapse
Affiliation(s)
- Dan-Dan Su
- IBMM, Institut des Biomolécules Max Mousseron, CNRS, Université de Montpellier, ENSCM, 34095, Montpellier, France.,Institut Européen des Membranes, Adaptive Supramolecular Nanosystems Group, Université de Montpellier, ENSCM, CNRS, Place Eugène Bataillon, CC 047, 34095, Montpellier, France
| | - Lamiaa M A Ali
- IBMM, Institut des Biomolécules Max Mousseron, CNRS, Université de Montpellier, ENSCM, 34095, Montpellier, France.,Department of Biochemistry Medical Research Institute, University of Alexandria, 21561, Alexandria, Egypt
| | - Maëva Coste
- IBMM, Institut des Biomolécules Max Mousseron, CNRS, Université de Montpellier, ENSCM, 34095, Montpellier, France
| | - Nabila Laroui
- IBMM, Institut des Biomolécules Max Mousseron, CNRS, Université de Montpellier, ENSCM, 34095, Montpellier, France
| | - Yannick Bessin
- IBMM, Institut des Biomolécules Max Mousseron, CNRS, Université de Montpellier, ENSCM, 34095, Montpellier, France
| | - Mihail Barboiu
- Institut Européen des Membranes, Adaptive Supramolecular Nanosystems Group, Université de Montpellier, ENSCM, CNRS, Place Eugène Bataillon, CC 047, 34095, Montpellier, France
| | - Nadir Bettache
- IBMM, Institut des Biomolécules Max Mousseron, CNRS, Université de Montpellier, ENSCM, 34095, Montpellier, France
| | - Sébastien Ulrich
- IBMM, Institut des Biomolécules Max Mousseron, CNRS, Université de Montpellier, ENSCM, 34095, Montpellier, France
| |
Collapse
|
14
|
Wang Z, Zhang J, Hu J, Yang G. Gene-activated titanium implants for gene delivery to enhance osseointegration. BIOMATERIALS ADVANCES 2022; 143:213176. [PMID: 36327825 DOI: 10.1016/j.bioadv.2022.213176] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 10/22/2022] [Accepted: 10/26/2022] [Indexed: 06/16/2023]
Abstract
Osseointegration is the direct and intimate contact between mineralized tissue and titanium implant at the bone-implant interface. Early establishment and stable maintenance of osseointegration is the key to long-term implant success. However, in patients with compromised conditions such as osteoporosis and patients beginning early load-bearing activities such as walking, lower osseointegration around titanium implants is often observed, which might result in implant early failure. Gene-activated implants show an exciting prospect of combining gene delivery and biomedical implants to solve the problems of poor osseointegration formation, overcoming the shortcomings of protein therapy, including rapid degradation and overdose adverse effects. The conception of gene-activated titanium implants is based on "gene-activated matrix" (GAM), which means scaffolds using non-viral vectors for in situ gene delivery to achieve a long-term and efficient transfection of target cells. Current preclinical studies in animal models have shown that plasmid DNA (pDNA), microRNA (miRNA), and small interference RNA (siRNA) functionalized titanium implants can enhance osseointegration with safety and efficiency, leading to the expectation of applying this technique in dental and orthopedic clinical scenarios. This review aims to comprehensively summarize fabrication strategies, current applications, and futural outlooks of gene-activated implants, emphasizing nucleic acid targets, non-viral vectors, implant surface modification techniques, nucleic acid/vector complexes loading strategies.
Collapse
Affiliation(s)
- Zhikang Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Jing Zhang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Jinxing Hu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Guoli Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China.
| |
Collapse
|
15
|
Ma XY, Hill BD, Hoang T, Wen F. Virus-inspired strategies for cancer therapy. Semin Cancer Biol 2022; 86:1143-1157. [PMID: 34182141 PMCID: PMC8710185 DOI: 10.1016/j.semcancer.2021.06.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/17/2021] [Accepted: 06/23/2021] [Indexed: 01/27/2023]
Abstract
The intentional use of viruses for cancer therapy dates back over a century. As viruses are inherently immunogenic and naturally optimized delivery vehicles, repurposing viruses for drug delivery, tumor antigen presentation, or selective replication in cancer cells represents a simple and elegant approach to cancer treatment. While early virotherapy was fraught with harsh side effects and low response rates, virus-based therapies have recently seen a resurgence due to newfound abilities to engineer and tune oncolytic viruses, virus-like particles, and virus-mimicking nanoparticles for improved safety and efficacy. However, despite their great potential, very few virus-based therapies have made it through clinical trials. In this review, we present an overview of virus-inspired approaches for cancer therapy, discuss engineering strategies to enhance their mechanisms of action, and highlight their application for overcoming the challenges of traditional cancer therapies.
Collapse
Affiliation(s)
- Xiao Yin Ma
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Brett D Hill
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Trang Hoang
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Fei Wen
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, United States.
| |
Collapse
|
16
|
Rao VB, Zhu J. Bacteriophage T4 as a nanovehicle for delivery of genes and therapeutics into human cells. Curr Opin Virol 2022; 55:101255. [PMID: 35952598 PMCID: PMC11736861 DOI: 10.1016/j.coviro.2022.101255] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 07/09/2022] [Indexed: 11/18/2022]
Abstract
The ability to deliver therapeutic genes and biomolecules into a human cell and restore a defective function has been the holy grail of medicine. Adeno-associated viruses and lentiviruses have been extensively used as delivery vehicles, but their capacity is limited to one (or two) gene(s). Bacteriophages are emerging as novel vehicles for gene therapy. The large 120 × 86-nm T4 capsid allows engineering of both its surface and its interior to incorporate combinations of DNAs, RNAs, proteins, and their complexes. In vitro assembly using purified components allows customization for various applications and for individualized therapies. Its large capacity, cell-targeting capability, safety, and inexpensive manufacturing could open unprecedented new possibilities for gene, cancer, and stem cell therapies. However, efficient entry into primary human cells and intracellular trafficking are significant barriers that must be overcome by gene engineering and evolution in order to translate phage-delivery technology from bench to bedside.
Collapse
Affiliation(s)
- Venigalla B Rao
- Bacteriophage Medical Research Center, Department of Biology, The Catholic University of America, Washington, DC 20064, USA.
| | - Jingen Zhu
- Bacteriophage Medical Research Center, Department of Biology, The Catholic University of America, Washington, DC 20064, USA
| |
Collapse
|
17
|
Liang S, Zuo FF, Yin BC, Ye BC. Delivery of siRNA based on engineered exosomes for glioblastoma therapy by targeting STAT3. Biomater Sci 2022; 10:1582-1590. [DOI: 10.1039/d1bm01723c] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Small interfering RNA (siRNA) has been considered as a promising strategy for treatment of glioblastoma (GBM), which is an aggressive brain disease with the poor prognosis. However, siRNA therapy for...
Collapse
|
18
|
A Nanoparticle-Conjugated Anti-TBK1 siRNA Induces Autophagy-Related Apoptosis and Enhances cGAS-STING Pathway in GBM Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6521953. [PMID: 34931127 PMCID: PMC8684524 DOI: 10.1155/2021/6521953] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 11/05/2021] [Indexed: 11/30/2022]
Abstract
Background Gene therapy shows considerable clinical benefit in cancer therapy, in which single-stranded ribonucleic acid (siRNA) is a promising strategy in the treatment of glioblastoma (GBM). TANK-binding kinase 1 (TBK1) is critical in tumorigenesis and development, which lays a foundation for an ideal target for tumor therapy. However, the practical application of free siRNA is limited. It is urgent to develop novel strategies to deliver TBK1 siRNA to activate apoptosis and cGAS-STING pathway as a therapeutic strategy for GBM. Methods The expression and prognostic value of TBK1 were evaluated in the TCGA, CGGA, and GTEx databases. A novel gene delivery system was designed here via PEGylated reduced graphene oxide (rGO-PEG) to targeted delivery of anti-TBK1 siRNA efficiently. The efficacy of TBK1si/rGO-PEG was evaluated in GBM cells. The underlying pathways were explored by Western blot. Results TBK1 was highly expressed in glioma samples, and its high expression indicated poor prognoses in glioma patients. The rGO-PEG presented great efficiency in targeted delivery of TBK1si RNA into GBM cells with up to 97.1% transfection efficiency. TBK1si/rGO-PEG exhibited anti-GBM activities by inhibiting TBK1 and autophagy, as well as activating apoptosis and cGAS-STING pathway. Conclusion The rGO-PEG could be an efficient system facilitating the delivery of specific siRNA. TBK1si/rGO-PEG could be a novel strategy for the treatment of GBM.
Collapse
|
19
|
Nie W, Wang B, Mi X, Chen J, Yu T, Miao J, Lin Y, Yang T, Ran M, Hong Z, Liu X, Liang X, Qian Z, Gao X. Co-Delivery of Paclitaxel and shMCL-1 by Folic Acid-Modified Nonviral Vector to Overcome Cancer Chemotherapy Resistance. SMALL METHODS 2021; 5:e2001132. [PMID: 34928100 DOI: 10.1002/smtd.202001132] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/05/2021] [Indexed: 02/05/2023]
Abstract
Acquired chemoresistance presents a major clinical impediment, which is an urgent problem to be solved. Interestingly, myeloma cell leukemia-1 (MCL-1) and folate receptor expression levels are higher in chemotherapy-resistant patients than in pretreatment patients. In this study, a multifunctional folic acid (FA)-targeting core-shell structure is presented for simultaneous delivery of shMCL-1 and paclitaxel (PTX). The transfection efficiency of shMCL-1 with the FA-targeting delivery system is higher than with a nontargeting delivery system in Skov3 and A2780T cells. The FA-targeting system significantly inhibits cell growth, blocks cell cycles, and promotes apoptosis of cancer cells in vitro. The mechanisms involved in inhibiting growth are related to Bcl-2/Bax and cdc2/Cyclin B1 pathways. An analysis of RNA sequencing suggests that shMCL-1 reverses chemoresistance through regulating genes such as regulator of chromosome condensation 2 (RCC2). The synergetic effect of shMCL-1 and PTX effectively inhibits tumor growth in both PTX-resistant and normal cancer models by inducing tumor apoptosis, inhibiting proliferation, and limiting tumor angiogenesis. The study results indicate that a FA-targeting delivery system combining shMCL-1 with PTX can simultaneously target tumor sites and restore the sensitivity of chemotherapy-resistant cancer to PTX. These findings have important implications for patients with normal or PTX-resistant cancer.
Collapse
Affiliation(s)
- Wen Nie
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Bilan Wang
- Department of Pharmacy, West China Second University Hospital of Sichuan University, Chengdu, 610041, P. R. China
| | - Xue Mi
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Jing Chen
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Ting Yu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Junming Miao
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, P. R. China
| | - Yunzhu Lin
- Department of Pharmacy, West China Second University Hospital of Sichuan University, Chengdu, 610041, P. R. China
| | - Tingting Yang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Mengni Ran
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Zehuo Hong
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Xiaoxiao Liu
- Department of Radiation Oncology, Cancer Center, Affiliated Hospital of Xuzhou Medical University, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, 221000, P. R. China
| | - Xiao Liang
- Department of Pharmacy, West China Second University Hospital of Sichuan University, Chengdu, 610041, P. R. China
| | - Zhiyong Qian
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| |
Collapse
|
20
|
Liao Z, Tu L, Li X, Liang XJ, Huo S. Virus-inspired nanosystems for drug delivery. NANOSCALE 2021; 13:18912-18924. [PMID: 34757354 DOI: 10.1039/d1nr05872j] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
With over millions of years of evolution, viruses can infect cells efficiently by utilizing their unique structures. Similarly, the drug delivery process is designed to imitate the viral infection stages for maximizing the therapeutic effect. From drug administration to therapeutic effect, nanocarriers must evade the host's immune system, break through multiple barriers, enter the cell, and release their payload by endosomal escape or nuclear targeting. Inspired by the virus infection process, a number of virus-like nanosystems have been designed and constructed for drug delivery. This review aims to present a comprehensive summary of the current understanding of the drug delivery process inspired by the viral infection stages. The most recent construction of virus-inspired nanosystems (VINs) for drug delivery is sorted, emphasizing their novelty and design principles, as well as highlighting the mechanism of these nanosystems for overcoming each biological barrier during drug delivery. A perspective on the VINs for therapeutic applications is provided in the end.
Collapse
Affiliation(s)
- Zhihuan Liao
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| | - Li Tu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| | - Xuejian Li
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| | - Xing-Jie Liang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuaidong Huo
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
21
|
Ramy S, Ueda Y, Nakajima H, Hiroi M, Hiroi Y, Torisu T, Uchiyama S. Reduction of Recombinant Adeno-Associated Virus Vector Adsorption on Solid Surfaces by Polyionic Hydrophilic Complex Coating. J Pharm Sci 2021; 111:663-671. [PMID: 34706282 DOI: 10.1016/j.xphs.2021.10.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 10/20/2021] [Accepted: 10/20/2021] [Indexed: 12/19/2022]
Abstract
Recombinant adeno-associated virus (rAAV) vectors have proven efficacy as gene therapy vehicles. However, non-specific adsorption of these vectors on solid surfaces is encountered during production, storage, and administration, as well as in quantification processes. Such adsorption has been reported to result in the loss of up to 90% of vector particles and can also result in high variability in vector genome quantification. In this study, we demonstrate the effective decrease of recombinant adeno-associated virus vector adsorption by application of a polyionic hydrophilic complex polymer coating on the surfaces of the tools used in viral vector quantification analyses [i.e., pipette tips, cryotube vials, and quantitative polymerase chain reaction (qPCR) plates]. qPCR analyses showed efficient recovery of vector particles from tools with this coating, with up to 95% of vector particle loss being prevented, leading to a higher transduction efficiency in vitro. Thus, the tested coating has the potential to be widely used in material processing in the gene therapy field.
Collapse
Affiliation(s)
- Salama Ramy
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yuki Ueda
- Nissan Chemical Corporation, Tokyo, Japan
| | | | - Miya Hiroi
- Nissan Chemical Corporation, Tokyo, Japan
| | | | - Tetsuo Torisu
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Susumu Uchiyama
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi, 444-8787, Japan.
| |
Collapse
|
22
|
Mangion M, Robert MA, Slivac I, Gilbert R, Gaillet B. Production and Use of Gesicles for Nucleic Acid Delivery. Mol Biotechnol 2021; 64:278-292. [PMID: 34596870 DOI: 10.1007/s12033-021-00389-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 09/08/2021] [Indexed: 12/29/2022]
Abstract
Over-expression of the vesicular stomatitis virus glycoprotein (VSVG) in mammalian cells can induce the formation of VSVG-pseudotyped vesicles (named "gesicles") from membrane budding. Its use as a nucleic acid delivery tool is still poorly documented. Naked-plasmid DNA can be delivered in animal cells with gesicles in presence of hexadimethrine bromide (polybrene). However, little is known about gesicle manufacturing process and conditions to obtain successful nucleic acid delivery. In this study, gesicles production process using polyethylenimine (PEI)-transfected HEK293 cells was developed by defining the VSVG-plasmid concentration, the DNA:PEI mass ratio, and the time of gesicle harvest. Furthermore, parameters described in the literature relevant for nucleic acid delivery such as (i) component concentrations in assembly mixture, (ii) component addition order, (iii) incubation time, and (iv) polybrene concentration were tested by assessing the transfection capacity of the gesicles complexed with a green fluorescent protein (GFP)-coding plasmid. Interestingly, freezing/thawing cycles and storage at + 4 °C, - 20 °C, and - 80 °C did not reduce gesicles' ability to transfer plasmid DNA. Transfection efficiency of 55% and 22% was obtained for HeLa cells and for hard-to-transfect cells such as human myoblasts, respectively. For the first time, gesicles were used for delivery of a large plasmid (18-kb) with 42% of efficiency and for enhanced green fluorescent protein (eGFP) gene silencing with siRNA (up to 60%). In conclusion, gesicles represent attractive bioreagents with great potential to deliver nucleic acids in mammalian cells.
Collapse
Affiliation(s)
- Mathias Mangion
- Chemical Engineering Department, Laval University, Pouliot Building, 1065 Avenue de la Médecine, Québec, QC, G1V0A6, Canada.,PROTEO: The Quebec Network for Research on Protein Function, Structure, and Engineering, Université Laval, Vachon Building, local 3403, 1045 Avenue de la Médecine, Québec, QC, G1V 0A6, Canada.,ThéCell: FRQS Cell and Tissue Therapy Network, LOEX, Aile R, local R-125, Hôpital de l'Enfant-Jésus, 1401 18e rue, Québec, QC, G1J 1Z4, Canada
| | - Marc-André Robert
- Chemical Engineering Department, Laval University, Pouliot Building, 1065 Avenue de la Médecine, Québec, QC, G1V0A6, Canada.,PROTEO: The Quebec Network for Research on Protein Function, Structure, and Engineering, Université Laval, Vachon Building, local 3403, 1045 Avenue de la Médecine, Québec, QC, G1V 0A6, Canada.,ThéCell: FRQS Cell and Tissue Therapy Network, LOEX, Aile R, local R-125, Hôpital de l'Enfant-Jésus, 1401 18e rue, Québec, QC, G1J 1Z4, Canada.,Human Health Therapeutics Portfolio, National Research Council Canada, 6100 Avenue Royalmount, Montréal, QC, H4P 2R2, Canada
| | - Igor Slivac
- Chemical Engineering Department, Laval University, Pouliot Building, 1065 Avenue de la Médecine, Québec, QC, G1V0A6, Canada.,PROTEO: The Quebec Network for Research on Protein Function, Structure, and Engineering, Université Laval, Vachon Building, local 3403, 1045 Avenue de la Médecine, Québec, QC, G1V 0A6, Canada.,ThéCell: FRQS Cell and Tissue Therapy Network, LOEX, Aile R, local R-125, Hôpital de l'Enfant-Jésus, 1401 18e rue, Québec, QC, G1J 1Z4, Canada
| | - Rénald Gilbert
- ThéCell: FRQS Cell and Tissue Therapy Network, LOEX, Aile R, local R-125, Hôpital de l'Enfant-Jésus, 1401 18e rue, Québec, QC, G1J 1Z4, Canada.,Human Health Therapeutics Portfolio, National Research Council Canada, 6100 Avenue Royalmount, Montréal, QC, H4P 2R2, Canada
| | - Bruno Gaillet
- Chemical Engineering Department, Laval University, Pouliot Building, 1065 Avenue de la Médecine, Québec, QC, G1V0A6, Canada. .,PROTEO: The Quebec Network for Research on Protein Function, Structure, and Engineering, Université Laval, Vachon Building, local 3403, 1045 Avenue de la Médecine, Québec, QC, G1V 0A6, Canada. .,ThéCell: FRQS Cell and Tissue Therapy Network, LOEX, Aile R, local R-125, Hôpital de l'Enfant-Jésus, 1401 18e rue, Québec, QC, G1J 1Z4, Canada.
| |
Collapse
|
23
|
Chen Z, Farag MA, Zhong Z, Zhang C, Yang Y, Wang S, Wang Y. Multifaceted role of phyto-derived polyphenols in nanodrug delivery systems. Adv Drug Deliv Rev 2021; 176:113870. [PMID: 34280511 DOI: 10.1016/j.addr.2021.113870] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/16/2021] [Accepted: 07/11/2021] [Indexed: 12/12/2022]
Abstract
As naturally occurring bioactive products, several lines of evidence have shown the potential of polyphenols in the medical intervention of various diseases, including tumors, inflammatory diseases, and cardiovascular diseases. Notably, owing to the particular molecular structure, polyphenols can combine with proteins, metal ions, polymers, and nucleic acids providing better strategies for polyphenol-delivery strategies. This contributes to the inherent advantages of polyphenols as important functional components for other drug delivery strategies, e.g., protecting nanodrugs from oxidation as a protective layer, improving the physicochemical properties of carbohydrate polymer carriers, or being used to synthesize innovative functional delivery vehicles. Polyphenols have emerged as a multifaceted player in novel drug delivery systems, both as therapeutic agents delivered to intervene in disease progression and as essential components of drug carriers. Although an increasing number of studies have focused on polyphenol-based nanodrug delivery including epigallocatechin-3-gallate, curcumin, resveratrol, tannic acid, and polyphenol-related innovative preparations, these molecules are not without inherent shortcomings. The active biochemical characteristics of polyphenols constitute a prerequisite to their high-frequency use in drug delivery systems and likewise to provoke new challenges for the design and development of novel polyphenol drug delivery systems of improved efficacies. In this review, we focus on both the targeted delivery of polyphenols and the application of polyphenols as components of drug delivery carriers, and comprehensively elaborate on the application of polyphenols in new types of drug delivery systems. According to the different roles played by polyphenols in innovative drug delivery strategies, potential limitations and risks are discussed in detail including the influences on the physical and chemical properties of nanodrug delivery systems, and their influence on normal physiological functions inside the organism.
Collapse
Affiliation(s)
- Zhejie Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China; Macau Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Mohamed A Farag
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, Egypt; Chemistry Department, American University in Cairo AUC, Cairo, Egypt
| | - Zhangfeng Zhong
- Macau Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Chen Zhang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu Yang
- Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shengpeng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China.
| | - Yitao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China.
| |
Collapse
|
24
|
Yang Y, Yue S, Qiao Y, Zhang P, Jiang N, Ning Z, Liu C, Hou Y. Activable Multi-Modal Nanoprobes for Imaging Diagnosis and Therapy of Tumors. Front Chem 2021; 8:572471. [PMID: 33912535 PMCID: PMC8075363 DOI: 10.3389/fchem.2020.572471] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 12/18/2020] [Indexed: 01/05/2023] Open
Abstract
Malignant tumors have become one of the major causes of human death, but there remains a lack of effective methods for tiny tumor diagnosis, metastasis warning, clinical efficacy prediction, and effective treatment. In this context, localizing tiny tumors via imaging and non-invasively extracting molecular information related to tumor proliferation, invasion, metastasis, and drug resistance from the tumor microenvironment have become the most fundamental tasks faced by cancer researchers. Tumor-associated microenvironmental physiological parameters, such as hypoxia, acidic extracellular pH, protease, reducing conditions, and so forth, have much to do with prognostic indicators for cancer progression, and impact therapeutic administrations. By combining with various novel nanoparticle-based activatable probes, molecular imaging technologies can provide a feasible approach to visualize tumor-associated microenvironment parameters noninvasively and realize accurate treatment of tumors. This review focuses on the recent achievements in the design of “smart” nanomedicine responding to the tumor microenvironment-related features and highlights state-of- the-art technology in tumor imaging diagnosis and therapy.
Collapse
Affiliation(s)
- Yan Yang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Saisai Yue
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Yuanyuan Qiao
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Peisen Zhang
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China.,School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Ni Jiang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Zhenbo Ning
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Chunyan Liu
- Institute of Atmospheric Physics, Chinese Academy of Sciences, Beijing, China
| | - Yi Hou
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China.,Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
25
|
de la Fuente IF, Sawant SS, Tolentino MQ, Corrigan PM, Rouge JL. Viral Mimicry as a Design Template for Nucleic Acid Nanocarriers. Front Chem 2021; 9:613209. [PMID: 33777893 PMCID: PMC7987652 DOI: 10.3389/fchem.2021.613209] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 01/06/2021] [Indexed: 12/11/2022] Open
Abstract
Therapeutic nucleic acids hold immense potential in combating undruggable, gene-based diseases owing to their high programmability and relative ease of synthesis. While the delivery of this class of therapeutics has successfully entered the clinical setting, extrahepatic targeting, endosomal escape efficiency, and subcellular localization. On the other hand, viruses serve as natural carriers of nucleic acids and have acquired a plethora of structures and mechanisms that confer remarkable transfection efficiency. Thus, understanding the structure and mechanism of viruses can guide the design of synthetic nucleic acid vectors. This review revisits relevant structural and mechanistic features of viruses as design considerations for efficient nucleic acid delivery systems. This article explores how viral ligand display and a metastable structure are central to the molecular mechanisms of attachment, entry, and viral genome release. For comparison, accounted for are details on the design and intracellular fate of existing nucleic acid carriers and nanostructures that share similar and essential features to viruses. The review, thus, highlights unifying themes of viruses and nucleic acid delivery systems such as genome protection, target specificity, and controlled release. Sophisticated viral mechanisms that are yet to be exploited in oligonucleotide delivery are also identified as they could further the development of next-generation nonviral nucleic acid vectors.
Collapse
Affiliation(s)
| | | | | | | | - Jessica L. Rouge
- Department of Chemistry, University of Connecticut, Storrs, CT, United States
| |
Collapse
|
26
|
van den Berg AIS, Yun CO, Schiffelers RM, Hennink WE. Polymeric delivery systems for nucleic acid therapeutics: Approaching the clinic. J Control Release 2021; 331:121-141. [PMID: 33453339 DOI: 10.1016/j.jconrel.2021.01.014] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/26/2020] [Accepted: 01/08/2021] [Indexed: 12/20/2022]
Abstract
Gene therapy using nucleic acids has many clinical applications for the treatment of diseases with a genetic origin as well as for the development of innovative vaccine formulations. Since nucleic acids in their free form are rapidly degraded by nucleases present in extracellular matrices, have poor pharmacokinetics and hardly pass cellular membranes, carrier systems are required. Suitable carriers that protect the nucleic acid payload against enzymatic attack, prolong circulation time after systemic administration and assist in cellular binding and internalization are needed to develop nucleic acid based drug products. Viral vectors have been investigated and are also clinically used as delivery vehicles. However, some major drawbacks are associated with their use. Therefore there has been substantial attention on the use of non-viral carrier systems based on cationic lipids and polymers. This review focuses on the properties of polymer-based nucleic acid formulations, also referred as polyplexes. Different polymeric systems are summarized, and the cellular barriers polyplexes encounter and ways to tackle these are discussed. Finally attention is given to the clinical status of non-viral nucleic acid formulations.
Collapse
Affiliation(s)
- Annette I S van den Berg
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Heidelberglaan 100, 3584, CX, Utrecht, the Netherlands
| | - Chae-Ok Yun
- Institute of Nano Science and Technology, Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Raymond M Schiffelers
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Heidelberglaan 100, 3584, CX, Utrecht, the Netherlands
| | - Wim E Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584, CG, Utrecht, the Netherlands.
| |
Collapse
|
27
|
Fleischmann D, Maslanka Figueroa S, Goepferich A. Steric Shielding of cRGD-Functionalized Nanoparticles from Premature Exposition to Off-Target Endothelial Cells under a Physiological Flow. ACS APPLIED BIO MATERIALS 2020. [DOI: 10.1021/acsabm.0c01193] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Daniel Fleischmann
- Department of Pharmaceutical Technology, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany
| | - Sara Maslanka Figueroa
- Department of Pharmaceutical Technology, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany
| | - Achim Goepferich
- Department of Pharmaceutical Technology, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany
| |
Collapse
|
28
|
Gao X, Ding J, Long Q, Zhan C. Virus-mimetic systems for cancer diagnosis and therapy. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 13:e1692. [PMID: 33354937 DOI: 10.1002/wnan.1692] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/13/2020] [Accepted: 11/30/2020] [Indexed: 01/02/2023]
Abstract
Over past decades, various strategies have been developed to enhance the delivery efficiency of therapeutics and imaging agents to tumor tissues. However, the therapeutic outcome of tumors to date have not been significantly improved, which can be partly attributed to the weak targeting ability, fast elimination, and low stability of conventional delivery systems. Viruses are the most efficient agents for gene transfer, serving as a valuable source of inspiration for designing nanoparticle-based delivery systems. Based on the properties of viruses, including well-defined geometry, precise composition, easy modification, stable construction, and specific infection, researchers attempt to design biocompatible delivery vectors by mimicking virus assembly and using the vector system to selectively concentrate drugs or imaging probes in tumors with mitigated toxicity and improved efficacy. In this review, we introduce common viruses features and provide an overview of various virus-mimetic strategies for cancer therapy and diagnosis. The challenges faced by virus-mimetic systems are also discussed. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Xihui Gao
- School of Basic Medical Sciences & Center of Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University, Shanghai, China
| | - Junqiang Ding
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, Shanghai, China
| | - Qianqian Long
- School of Basic Medical Sciences & Center of Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University, Shanghai, China
| | - Changyou Zhan
- School of Basic Medical Sciences & Center of Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University, Shanghai, China.,State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, China
| |
Collapse
|
29
|
Neva T, Carbajo-Gordillo AI, Benito JM, Lana H, Marcelo G, Ortiz Mellet C, Tros de Ilarduya C, Mendicuti F, García Fernández JM. Tuning the Topological Landscape of DNA-Cyclodextrin Nanocomplexes by Molecular Design. Chemistry 2020; 26:15259-15269. [PMID: 32710799 DOI: 10.1002/chem.202002951] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Indexed: 12/25/2022]
Abstract
Original molecular vectors that ensure broad flexibility to tune the shape and surface properties of plasmid DNA (pDNA) condensates are reported herein. The prototypic design involves a cyclodextrin (CD) platform bearing a polycationic cluster at the primary face and a doubly linked aromatic module bridging two consecutive monosaccharide units at the secondary face that behaves as a topology-encoding element. Subtle differences at the molecular level then translate into disparate morphologies at the nanoscale, including rods, worms, toroids, globules, ellipsoids, and spheroids. In vitro evaluation of the transfection capabilities revealed marked selectivity differences as a function of nanocomplex morphology. Remarkably high transfection efficiencies were associated with ellipsoidal or spherical shapes with a lamellar internal arrangement of pDNA chains and CD bilayers. Computational studies support that the stability of such supramolecular edifices is directly related to the tendency of the molecular vector to form noncovalent dimers upon DNA templating. Because the stability of the dimers depends on the protonation state of the polycationic clusters, the coaggregates display pH responsiveness, which facilitates endosomal escape and timely DNA release, a key step in successful transfection. The results provide a versatile strategy for the construction of fully synthetic and perfectly monodisperse nonviral gene delivery systems uniquely suited for optimization schemes.
Collapse
Affiliation(s)
- Tania Neva
- Institute for Chemical Research, IIQ, CSIC-Univ. Sevilla, C/ Américo Vespucio 49, 41092, Sevilla, Spain
| | - Ana I Carbajo-Gordillo
- Institute for Chemical Research, IIQ, CSIC-Univ. Sevilla, C/ Américo Vespucio 49, 41092, Sevilla, Spain
| | - Juan M Benito
- Institute for Chemical Research, IIQ, CSIC-Univ. Sevilla, C/ Américo Vespucio 49, 41092, Sevilla, Spain
| | - Hugo Lana
- Department of Pharmaceutical Technology and Chemistry, School of Pharmacy and Nutrition, University of Navarra, 31080, Pamplona, Spain
| | - Gema Marcelo
- Department of Analytical Chemistry, Physical Chemistry and Chemical Engineering, Instituto de Investigación Química, "Andrés M. del Rio" (IQAR), University of Alcalá, Campus Universitario Ctra. Madrid-Barcelona, Km 33.600, 28871, Alcalá de Henares, Spain
| | - Carmen Ortiz Mellet
- Department of Organic Chemistry, Faculty of Chemistry, University of Sevilla, C/ Prof García González 1, 41012, Sevilla, Spain
| | - Conchita Tros de Ilarduya
- Department of Pharmaceutical Technology and Chemistry, School of Pharmacy and Nutrition, University of Navarra, 31080, Pamplona, Spain
| | - Francisco Mendicuti
- Department of Analytical Chemistry, Physical Chemistry and Chemical Engineering, Instituto de Investigación Química, "Andrés M. del Rio" (IQAR), University of Alcalá, Campus Universitario Ctra. Madrid-Barcelona, Km 33.600, 28871, Alcalá de Henares, Spain
| | - José M García Fernández
- Institute for Chemical Research, IIQ, CSIC-Univ. Sevilla, C/ Américo Vespucio 49, 41092, Sevilla, Spain
| |
Collapse
|
30
|
Fleischmann D, Maslanka Figueroa S, Beck S, Abstiens K, Witzgall R, Schweda F, Tauber P, Goepferich A. Adenovirus-Mimetic Nanoparticles: Sequential Ligand-Receptor Interplay as a Universal Tool for Enhanced In Vitro/ In Vivo Cell Identification. ACS APPLIED MATERIALS & INTERFACES 2020; 12:34689-34702. [PMID: 32639709 DOI: 10.1021/acsami.0c10057] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Viral infection patterns often rely on precisely coordinated sequences of distinct ligand-receptor interactions, leading in many cases to an outstanding target cell specificity. A successful mimicry of viral targeting strategies to create more site-specific nanoparticles (NPs) would therefore require particle-cell interactions to also be adequately controllable. In the present study, hetero-multivalent block-copolymer NPs present their attached ligands in a sterically controlled manner to create a sequential NP-cell interaction similar to the cell infiltration strategy of human adenovirus type 2. Targeting renal mesangial cells, particles therefore initially bind angiotensin II receptor type 1 (AT1r) on the cell surface via a structurally flexible AT1r antagonist. After a mandatory spatial approach, particle endocytosis is realized via binding of immobile αVβ3 integrins with a previously concealed secondary ligand, thereby creating a stepwise particle-cell interplay of primary NP attachment and subsequent uptake. Manufactured adenovirus-mimetic NPs show great avidity for both target motifs in vitro, leading to a substantial binding as well as subsequent cell uptake into target mesangial cells. Additionally, steric shielding of secondary ligand visibility leads to a highly controllable, sequential ligand-receptor interaction, whereby hetero-functional NPs activate mesangial cell surface integrins only after a successful prior binding to the AT1r. This stepwise cell identification significantly enhances mesangial cell specificity in co-culture assays with different off-target cells. Additionally, described NPs display excellent in vivo robustness by efficiently accumulating in the mesangium upon injection, thereby opening new paths for possible drug delivery applications.
Collapse
Affiliation(s)
- Daniel Fleischmann
- Department of Pharmaceutical Technology, University of Regensburg, Universitaetsstrasse 31, 93053 Regensburg, Germany
| | - Sara Maslanka Figueroa
- Department of Pharmaceutical Technology, University of Regensburg, Universitaetsstrasse 31, 93053 Regensburg, Germany
| | - Sebastian Beck
- Department of Pharmaceutical Technology, University of Regensburg, Universitaetsstrasse 31, 93053 Regensburg, Germany
| | - Kathrin Abstiens
- Department of Pharmaceutical Technology, University of Regensburg, Universitaetsstrasse 31, 93053 Regensburg, Germany
| | - Ralph Witzgall
- Institute for Molecular and Cellular Anatomy, University of Regensburg, 93053 Regensburg, Germany
| | - Frank Schweda
- Department of Physiology II, Institute for Physiology, University of Regensburg, 93053 Regensburg, Germany
| | - Philipp Tauber
- Department of Physiology II, Institute for Physiology, University of Regensburg, 93053 Regensburg, Germany
| | - Achim Goepferich
- Department of Pharmaceutical Technology, University of Regensburg, Universitaetsstrasse 31, 93053 Regensburg, Germany
| |
Collapse
|
31
|
Harisa GI, Sherif AY, Youssof AM, Alanazi FK, Salem-Bekhit MM. Bacteriosomes as a Promising Tool in Biomedical Applications: Immunotherapy and Drug Delivery. AAPS PharmSciTech 2020; 21:168. [PMID: 32514657 DOI: 10.1208/s12249-020-01716-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/18/2020] [Indexed: 12/24/2022] Open
Abstract
Bacteriosomes are a member of cell-derived vesicles that are proposed as promising tools in diagnosis, therapy, and drug delivery. These vesicles could be derived from a virus, bacterial cells, and animal cells. Biotechnology techniques were used in bioengineering of cell-derived vesicles in vitro, and in vivo. Bacterial vesicles such as bacterial cells, bacterial ghost, or bacteriosomes are vesicular structures derived from bacteria produced by manipulation of bacterial cells by chemical agents or gene-mediated lysis. Subsequently, bacterial vesicles (bacteriosomes) are non-living, non-denatured bacterial cell envelopes free of the cytoplasm and genetic materials. Gram-negative and Gram-positive bacteria are exploited in the production of bacteriosomes. Bacteriosomes have instinct organs, tissues, cells, as well as subcellular tropism. Moreover, bacteriosomes might be used as immunotherapy and/or drug delivery shuttles. They could act as cargoes for the delivery of small drugs, large therapeutics, and nanoparticles to the specific location. Furthermore, bacteriosomes have nature endosomal escaping ability, hence they could traffic different bio-membranes by endocytosis mechanisms. Therefore, bacterial-derived vesicles could be used in therapy and development of an innovative drug delivery systems. Consequently, utilizing bacteriosomes as drug cargoes enhances the delivery and efficacy of administered therapeutic agents. This review highlighted bacteriosomes in terms of source, engineering, characterization, applications, and limitations.
Collapse
|
32
|
Zou Y, Sun X, Wang Y, Yan C, Liu Y, Li J, Zhang D, Zheng M, Chung RS, Shi B. Single siRNA Nanocapsules for Effective siRNA Brain Delivery and Glioblastoma Treatment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2000416. [PMID: 32374446 DOI: 10.1002/adma.202000416] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 03/29/2020] [Accepted: 04/08/2020] [Indexed: 06/11/2023]
Abstract
Small interfering RNA (siRNA) has been considered as a highly promising therapeutic agent for human cancer treatment including glioblastoma (GBM), which is a fatal disease without effective therapy methods. However, siRNA-based GBM therapy is seriously hampered by a number of challenges in siRNA brain delivery including poor stability, short blood circulation, low blood-brain barrier (BBB) penetration, and tumor accumulation, as well as inefficient siRNA intracellular release. Herein, an Angiopep-2 (Ang) functionalized intracellular-environment-responsive siRNA nanocapsule (Ang-NCss (siRNA)) is successfully developed as a safe and efficient RNAi agent to boost siRNA-based GBM therapy. The experimental results demonstrate that the developed Ang-NCss (siRNA) displays long circulation in plasma, efficient BBB penetration capability, and GBM accumulation and retention, as well as responsive intracellular siRNA release due to the unique design of small size (25 nm) with polymeric shell for siRNA protection, Ang functionalization for BBB crossing and GBM targeting, and disulfide bond as a linker for intracellular-environment-responsive siRNA release. Such superior properties of Ang-NCss (siRNA) result in outstanding growth inhibition of orthotopic U87MG xenografts without causing adverse effects, achieving remarkably improved survival benefits. The developed siRNA nanocapsules provide a new strategy for RNAi therapy of GBM and beyond.
Collapse
Affiliation(s)
- Yan Zou
- Henan-Macquarie University Joint Centre for Biomedical Innovation School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Xinhong Sun
- Henan-Macquarie University Joint Centre for Biomedical Innovation School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Yibin Wang
- Henan-Macquarie University Joint Centre for Biomedical Innovation School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Chengnan Yan
- Henan-Macquarie University Joint Centre for Biomedical Innovation School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Yanjie Liu
- Henan-Macquarie University Joint Centre for Biomedical Innovation School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Jia Li
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Dongya Zhang
- Henan-Macquarie University Joint Centre for Biomedical Innovation School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Meng Zheng
- Henan-Macquarie University Joint Centre for Biomedical Innovation School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Roger S Chung
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Bingyang Shi
- Henan-Macquarie University Joint Centre for Biomedical Innovation School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| |
Collapse
|
33
|
Zhang Y, Liu Y, Zhang W, Tang Q, Zhou Y, Li Y, Rong T, Wang H, Chen Y. Isolated cell-bound membrane vesicles (CBMVs) as a novel class of drug nanocarriers. J Nanobiotechnology 2020; 18:69. [PMID: 32375799 PMCID: PMC7204042 DOI: 10.1186/s12951-020-00625-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 04/27/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Cell-bound membrane vesicles (CBMVs) are a type of membrane vesicles different from the well-known extracellular vesicles (EVs). In recent years, the applications of EVs as drug delivery systems have been studied widely. A question may arise whether isolated CBMVs also have the possibility of being recruited as a drug delivery system or nanocarrier? METHODS To test the possibility, CBMVs were isolated/purified from the surfaces of cultured endothelial cells, loaded with a putative antitumor drug doxorubicin (Dox), and characterized. Subsequently, cellular experiments and animal experiments using mouse models were performed to determine the in vitro and in vivo antitumor effects of Dox-loaded CBMVs (Dox-CBMVs or Dox@CBMVs), respectively. RESULTS Both Dox-free and Dox-loaded CBMVs were globular-shaped and nanometer-sized with an average diameter of ~ 300-400 nm. Dox-CBMVs could be internalized by cells and could kill multiple types of cancer cells. The in vivo antitumor ability of Dox-CBMVs also was confirmed. Moreover, Quantifications of blood cells (white blood cells and platelets) and specific enzymes (aspartate aminotransferase and creatine kinase isoenzymes) showed that Dox-CBMVs had lower side effects compared with free Dox. CONCLUSIONS The data show that the CBMV-entrapped Doxorubicin has the antitumor efficacy with lower side effects. This study provides evidence supporting the possibility of isolated cell-bound membrane vesicles as a novel drug nanocarrier.
Collapse
Affiliation(s)
- Yang Zhang
- Jiangxi Key Laboratory for Microscale Interdisciplinary Study, Institute for Advanced Study, Nanchang University, 999 Xuefu Ave., Honggutan District, Nanchang, Jiangxi 330031 People’s Republic of China
- School of Materials Science and Engineering, Nanchang University, Nanchang, Jiangxi 330031 People’s Republic of China
| | - Yang Liu
- Jiangxi Key Laboratory for Microscale Interdisciplinary Study, Institute for Advanced Study, Nanchang University, 999 Xuefu Ave., Honggutan District, Nanchang, Jiangxi 330031 People’s Republic of China
| | - Wendiao Zhang
- Jiangxi Key Laboratory for Microscale Interdisciplinary Study, Institute for Advanced Study, Nanchang University, 999 Xuefu Ave., Honggutan District, Nanchang, Jiangxi 330031 People’s Republic of China
| | - Qisheng Tang
- Jiangxi Key Laboratory for Microscale Interdisciplinary Study, Institute for Advanced Study, Nanchang University, 999 Xuefu Ave., Honggutan District, Nanchang, Jiangxi 330031 People’s Republic of China
| | - Yun Zhou
- Jiangxi Key Laboratory for Microscale Interdisciplinary Study, Institute for Advanced Study, Nanchang University, 999 Xuefu Ave., Honggutan District, Nanchang, Jiangxi 330031 People’s Republic of China
| | - Yuanfang Li
- Jiangxi Key Laboratory for Microscale Interdisciplinary Study, Institute for Advanced Study, Nanchang University, 999 Xuefu Ave., Honggutan District, Nanchang, Jiangxi 330031 People’s Republic of China
| | - Tong Rong
- Jiangxi Key Laboratory for Microscale Interdisciplinary Study, Institute for Advanced Study, Nanchang University, 999 Xuefu Ave., Honggutan District, Nanchang, Jiangxi 330031 People’s Republic of China
| | - Huaying Wang
- Jiangxi Key Laboratory for Microscale Interdisciplinary Study, Institute for Advanced Study, Nanchang University, 999 Xuefu Ave., Honggutan District, Nanchang, Jiangxi 330031 People’s Republic of China
| | - Yong Chen
- Jiangxi Key Laboratory for Microscale Interdisciplinary Study, Institute for Advanced Study, Nanchang University, 999 Xuefu Ave., Honggutan District, Nanchang, Jiangxi 330031 People’s Republic of China
| |
Collapse
|
34
|
Surin M, Ulrich S. From Interaction to Function in DNA-Templated Supramolecular Self-Assemblies. ChemistryOpen 2020; 9:480-498. [PMID: 32328404 PMCID: PMC7175023 DOI: 10.1002/open.202000013] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/24/2020] [Indexed: 12/13/2022] Open
Abstract
DNA-templated self-assembly represents a rich and growing subset of supramolecular chemistry where functional self-assemblies are programmed in a versatile manner using nucleic acids as readily-available and readily-tunable templates. In this review, we summarize the different DNA recognition modes and the basic supramolecular interactions at play in this context. We discuss the recent results that report the DNA-templated self-assembly of small molecules into complex yet precise nanoarrays, going from 1D to 3D architectures. Finally, we show their emerging functions as photonic/electronic nanowires, sensors, gene delivery vectors, and supramolecular catalysts, and their growing applications in a wide range of area from materials to biological sciences.
Collapse
Affiliation(s)
- Mathieu Surin
- Laboratory for Chemistry of Novel MaterialsCenter of Innovation and Research in Materials and Polymers (CIRMAP)University of Mons-UMONS7000MonsBelgium
| | | |
Collapse
|
35
|
Zhou J, Shao Z, Liu J, Duan Q, Wang X, Li J, Yang H. From Endocytosis to Nonendocytosis: The Emerging Era of Gene Delivery. ACS APPLIED BIO MATERIALS 2020; 3:2686-2701. [DOI: 10.1021/acsabm.9b01131] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Jie Zhou
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350116, People’s Republic of China
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, People’s Republic of China
| | - Zhentao Shao
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, People’s Republic of China
| | - Jia Liu
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, People’s Republic of China
| | - Qiao Duan
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, People’s Republic of China
| | - Xiang Wang
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, People’s Republic of China
| | - Juan Li
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350116, People’s Republic of China
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, People’s Republic of China
| | - Huanghao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350116, People’s Republic of China
| |
Collapse
|
36
|
Deviatkin AA, Vakulenko YA, Akhmadishina LV, Tarasov VV, Beloukhova MI, Zamyatnin Jr. AA, Lukashev AN. Emerging Concepts and Challenges in Rheumatoid Arthritis Gene Therapy. Biomedicines 2020; 8:biomedicines8010009. [PMID: 31936504 PMCID: PMC7168286 DOI: 10.3390/biomedicines8010009] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/03/2020] [Accepted: 01/07/2020] [Indexed: 02/07/2023] Open
Abstract
Rheumatoid arthritis (RA) is a systemic inflammatory joint disease affecting about 1% of the population worldwide. Current treatment approaches do not ensure a cure for every patient. Moreover, classical regimens are based on nontargeted systemic immune suppression and have significant side effects. Biological treatment has advanced considerably but efficacy and specificity issues remain. Gene therapy is one of the potential future directions for RA therapy, which is rapidly developing. Several gene therapy trials done so far have been of moderate success, but experimental and genetics studies have yielded novel targets. As a result, the arsenal of gene therapy tools keeps growing. Currently, both viral and nonviral delivery systems are used for RA therapy. Herein, we review recent approaches for RA gene therapy.
Collapse
Affiliation(s)
- Andrei A. Deviatkin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119048 Moscow, Russia; (M.I.B.); (A.A.Z.J.); (A.N.L.)
- Correspondence:
| | - Yulia A. Vakulenko
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector Borne Diseases, Sechenov First Moscow State Medical University, 119435 Moscow, Russia; (Y.A.V.); (L.V.A.)
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Ludmila V. Akhmadishina
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector Borne Diseases, Sechenov First Moscow State Medical University, 119435 Moscow, Russia; (Y.A.V.); (L.V.A.)
| | - Vadim V. Tarasov
- Department of Pharmacology and Pharmacy, Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
| | - Marina I. Beloukhova
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119048 Moscow, Russia; (M.I.B.); (A.A.Z.J.); (A.N.L.)
| | - Andrey A. Zamyatnin Jr.
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119048 Moscow, Russia; (M.I.B.); (A.A.Z.J.); (A.N.L.)
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Alexander N. Lukashev
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119048 Moscow, Russia; (M.I.B.); (A.A.Z.J.); (A.N.L.)
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector Borne Diseases, Sechenov First Moscow State Medical University, 119435 Moscow, Russia; (Y.A.V.); (L.V.A.)
| |
Collapse
|
37
|
Ni R, Chau Y. Nanoassembly of Oligopeptides and DNA Mimics the Sequential Disassembly of a Spherical Virus. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201913611] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Rong Ni
- Department of Chemical and Biological EngineeringThe Hong Kong University of Science and Technology Clearwater Bay Kowloon, Hong Kong China
| | - Ying Chau
- Department of Chemical and Biological EngineeringThe Hong Kong University of Science and Technology Clearwater Bay Kowloon, Hong Kong China
| |
Collapse
|
38
|
Ni R, Chau Y. Nanoassembly of Oligopeptides and DNA Mimics the Sequential Disassembly of a Spherical Virus. Angew Chem Int Ed Engl 2019; 59:3578-3584. [DOI: 10.1002/anie.201913611] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Indexed: 11/05/2022]
Affiliation(s)
- Rong Ni
- Department of Chemical and Biological EngineeringThe Hong Kong University of Science and Technology Clearwater Bay Kowloon, Hong Kong China
| | - Ying Chau
- Department of Chemical and Biological EngineeringThe Hong Kong University of Science and Technology Clearwater Bay Kowloon, Hong Kong China
| |
Collapse
|
39
|
Alonso-Valenteen F, Pacheco S, Srinivas D, Rentsendorj A, Chu D, Lubow J, Sims J, Miao T, Mikhael S, Hwang JY, Abrol R, Medina Kauwe LK. HER3-targeted protein chimera forms endosomolytic capsomeres and self-assembles into stealth nucleocapsids for systemic tumor homing of RNA interference in vivo. Nucleic Acids Res 2019; 47:11020-11043. [PMID: 31617560 PMCID: PMC6868389 DOI: 10.1093/nar/gkz900] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 09/12/2019] [Accepted: 10/09/2019] [Indexed: 12/31/2022] Open
Abstract
RNA interference represents a potent intervention for cancer treatment but requires a robust delivery agent for transporting gene-modulating molecules, such as small interfering RNAs (siRNAs). Although numerous molecular approaches for siRNA delivery are adequate in vitro, delivery to therapeutic targets in vivo is limited by payload integrity, cell targeting, efficient cell uptake, and membrane penetration. We constructed nonviral biomaterials to transport small nucleic acids to cell targets, including tumor cells, on the basis of the self-assembling and cell-penetrating activities of the adenovirus capsid penton base. Our recombinant penton base chimera contains polypeptide domains designed for noncovalent assembly with anionic molecules and tumor homing. Here, structural modeling, molecular dynamics simulations, and functional assays suggest that it forms pentameric units resembling viral capsomeres that assemble into larger capsid-like structures when combined with siRNA cargo. Pentamerization forms a barrel lined with charged residues mediating pH-responsive dissociation and exposing masked domains, providing insight on the endosomolytic mechanism. The therapeutic impact was examined on tumors expressing high levels of HER3/ErbB3 that are resistant to clinical inhibitors. Our findings suggest that our construct may utilize ligand mimicry to avoid host attack and target the siRNA to HER3+ tumors by forming multivalent capsid-like structures.
Collapse
Affiliation(s)
- Felix Alonso-Valenteen
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Sayuri Pacheco
- Department of Chemistry and Biochemistry, California State University, Northridge, CA 91330, USA
| | - Dustin Srinivas
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Altan Rentsendorj
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - David Chu
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jay Lubow
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jessica Sims
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Tianxin Miao
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Simoun Mikhael
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jae Youn Hwang
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Information and Communication Engineering, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea
| | - Ravinder Abrol
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Chemistry and Biochemistry, California State University, Northridge, CA 91330, USA
| | - Lali K Medina Kauwe
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
40
|
Pal Singh P, Vithalapuram V, Metre S, Kodipyaka R. Lipoplex-based therapeutics for effective oligonucleotide delivery: a compendious review. J Liposome Res 2019; 30:313-335. [DOI: 10.1080/08982104.2019.1652645] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Pirthi Pal Singh
- Department of Formulation Research and Development, Custom Pharmaceutical Services, Dr. Reddy’s Laboratories Ltd., Hyderabad, India
| | - Veena Vithalapuram
- Department of Formulation Research and Development, Custom Pharmaceutical Services, Dr. Reddy’s Laboratories Ltd., Hyderabad, India
| | - Sunita Metre
- Department of Formulation Research and Development, Custom Pharmaceutical Services, Dr. Reddy’s Laboratories Ltd., Hyderabad, India
| | - Ravinder Kodipyaka
- Department of Formulation Research and Development, Custom Pharmaceutical Services, Dr. Reddy’s Laboratories Ltd., Hyderabad, India
| |
Collapse
|
41
|
Zhu J, Tao P, Mahalingam M, Sha J, Kilgore P, Chopra AK, Rao V. A prokaryotic-eukaryotic hybrid viral vector for delivery of large cargos of genes and proteins into human cells. SCIENCE ADVANCES 2019; 5:eaax0064. [PMID: 31457098 PMCID: PMC6703872 DOI: 10.1126/sciadv.aax0064] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 07/11/2019] [Indexed: 05/03/2023]
Abstract
Development of safe and efficient nanoscale vehicles that can deliver large molecular cargos into human cells could transform future human therapies and personalized medicine. Here, we design a hybrid viral vector composed of a prokaryotic virus (bacteriophage T4) and a eukaryotic virus [adeno-associated virus (AAV)]. The small 25-nm AAV is attached to the large 120 nm × 86 nm T4 head through avidin-biotin cross-bridges using the phage decoration proteins Soc and Hoc. AAV "piggy-backed" on T4 capsid, by virtue of its natural ability to enter human cells acted as an efficient "driver," delivering the largest payloads of foreign DNA (up to 170 kb) and protein (up to 1025 molecules) reported to date, and elicited robust immune responses in mice against flu and plague pathogens and conferred complete protection against lethal pneumonic plague challenge. The T4-AAV represents a unique platform for assembly of natural building blocks into potential therapeutics against genetic and infectious diseases.
Collapse
Affiliation(s)
- Jingen Zhu
- Department of Biology, The Catholic University of America, Washington, DC 20064, USA
| | - Pan Tao
- Department of Biology, The Catholic University of America, Washington, DC 20064, USA
| | - Marthandan Mahalingam
- Department of Biology, The Catholic University of America, Washington, DC 20064, USA
| | - Jian Sha
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Paul Kilgore
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Ashok K. Chopra
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Venigalla Rao
- Department of Biology, The Catholic University of America, Washington, DC 20064, USA
| |
Collapse
|
42
|
Biomaterial-based delivery systems of nucleic acid for regenerative research and regenerative therapy. Regen Ther 2019; 11:123-130. [PMID: 31338391 PMCID: PMC6626072 DOI: 10.1016/j.reth.2019.06.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/10/2019] [Accepted: 06/25/2019] [Indexed: 12/22/2022] Open
Abstract
Regenerative medicine is a new and promising medical method aiming at treating patients with defective or dysfunctional tissues by maintaining or enhancing the biological activity of cells. The development of biomaterial-based technologies, such as cell scaffolds and carriers for drug delivery system, are highly required to promote the regenerative research and regenerative therapy. Nucleic acids are one of the most feasible factors to efficiently modify the biological activity of cells. The effective and stable delivery of nucleic acids into cells is highly required to succeed in the modification. Biomaterials-based non-viral carriers or biological carriers, like exosomes, play an important role in the efficient delivery of nucleic acids. This review introduces the examples of regenerative research and regenerative therapy based on the delivery of nucleic acids with biomaterials technologies and emphasizes their importance to accomplish regenerative medicine. Modifying the activity of cells is important for regenerative medicine. Various nucleic acids regulate gene expression to modify the activity of cells. Intracellular delivery system is vital to the nucleic acids-based modification. Biomaterials are useful for the intracellular delivery of nucleic acids.
Collapse
Key Words
- Biomaterials
- CRISPR, clustered regularly interspaced short palindromic repeats
- Cas, CRISPR-associated systems
- Cell scaffold
- DDS, drug delivery system
- Drug delivery system
- ECM, extracellular matrix
- MSC, mesenchymal stem cells
- Nucleic acids
- PEG, polyethylene glycol
- PLGA, poly(d,l-lactic acid-co-glycolic acid)
- RISC, RNA-induced silencing complex
- RNAi, RNA interferince
- Regenerative research
- Regenerative therapy
- TALEN, transcription activator-like effector nuclease
- ZFN, zinc finger nucleases
- lncRNA, long non-coding RNA
- mRNA, messenger RNA
- miRNA, microRNA
- siRNA, small interfering RNA
Collapse
|
43
|
Zhang J, Liu N, Lu Y, Huang Z, Zang Y, Chen J, Zhang J, Ding Z. Phosphorothioated antisense oligodeoxynucleotide suppressing interleukin-10 is a safe and potent vaccine adjuvant. Vaccine 2019; 37:4081-4088. [PMID: 31164303 DOI: 10.1016/j.vaccine.2019.05.076] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/30/2019] [Accepted: 05/26/2019] [Indexed: 12/26/2022]
Abstract
While vaccination is highly effective for the prevention of many infectious diseases, the number of adjuvants licensed for human use is currently very limited. The aim of this study was to evaluate the safety, efficacy, and to clarify the mechanism of a phosphorothioated interleukin (IL)-10-targeted antisense oligonucleotide (ASO) as an immune adjuvant in intradermal vaccination. The cytotoxicity of IL-10 ASO and its ability to promote T cell proliferation were assessed by Cell Counting Kit-8 (CCK-8) assay. The contents of IL-6, IL-8, TNF-α, IL-1β, and IL-10 in inoculated local tissue and the antigen-specific antibody titers in mouse serum samples were determined by ELISA. The target cells of IL-10 ASO were observed using immunofluorescent staining. The results showed that the specific antibody titer of ovalbumin (OVA), a model antigen, was increased 100-fold upon addition of IL-10 ASO as an adjuvant compared to that of OVA alone. IL-10 ASO showed an immunopotentiation efficacy similar to that of Freund's incomplete adjuvant, with no detectable cell or tissue toxicity. In vitro and in vivo experiments confirmed that IL-10 ASO enhances immune responses by temporarily suppressing IL-10 expression from local dendritic cells and consequently promoting T cell proliferation. In conclusion, IL-10 ASO significantly enhances immune responses against co-delivered vaccine antigens with high efficacy and low toxicity. It has the potential to be developed into a safe and efficient immune adjuvant.
Collapse
Affiliation(s)
- Jin Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Ninghua Liu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Yang Lu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Zhen Huang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Yuhui Zang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Jiangning Chen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Junfeng Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Collaborative Innovation Center of Chemistry for Life Sciences, Nanjing University, Nanjing 210023, China.
| | - Zhi Ding
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Changzhou High-Tech Research Institute of Nanjing University, Changzhou 213164, China.
| |
Collapse
|
44
|
Strategies in the design of endosomolytic agents for facilitating endosomal escape in nanoparticles. Biochimie 2019; 160:61-75. [DOI: 10.1016/j.biochi.2019.02.012] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 02/19/2019] [Indexed: 12/23/2022]
|
45
|
Jiang H, Hu X, Mosel S, Knauer SK, Hirschhäuser C, Schmuck C. A Branched Tripeptide with an Anion‐Binding Motif as a New Delivery Carrier for Efficient Gene Transfection. Chembiochem 2019; 20:1410-1416. [DOI: 10.1002/cbic.201800728] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Indexed: 11/08/2022]
Affiliation(s)
- Hao Jiang
- Key Laboratory of Materials Chemistry for Energy Conversion and StorageSchool of Chemistry and Chemical EngineeringHuazhong University of Science and Technology (HUST) Wuhan 430074 P.R. China
- Institute for Organic ChemistryUniversity of Duisburg–Essen Universitätsstrasse 7 45141 Essen Germany
| | - Xiao‐Yu Hu
- Institute for Organic ChemistryUniversity of Duisburg–Essen Universitätsstrasse 7 45141 Essen Germany
- Applied Chemistry DepartmentSchool of Material Science and EngineeringNanjing University of Aeronautics and Astronautics Nanjing 210016 P.R. China
| | - Stefanie Mosel
- Institute for BiologyUniversity of Duisburg–Essen Universitätsstrasse 5 45141 Essen Germany
| | - Shirley K. Knauer
- Institute for BiologyUniversity of Duisburg–Essen Universitätsstrasse 5 45141 Essen Germany
| | - Christoph Hirschhäuser
- Institute for Organic ChemistryUniversity of Duisburg–Essen Universitätsstrasse 7 45141 Essen Germany
| | - Carsten Schmuck
- Institute for Organic ChemistryUniversity of Duisburg–Essen Universitätsstrasse 7 45141 Essen Germany
| |
Collapse
|
46
|
Wang X, Hua Y, Xu G, Deng S, Yang D, Gao X. Targeting EZH2 for glioma therapy with a novel nanoparticle-siRNA complex. Int J Nanomedicine 2019; 14:2637-2653. [PMID: 31043779 PMCID: PMC6472285 DOI: 10.2147/ijn.s189871] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Background For the past few years, gene-therapy has recently shown considerable clinical benefit in cancer therapy, and the applications of gene therapies in cancer treatments continue to increase perennially. EZH2, an ideal candidate for tumor gene therapy, plays an important role in the tumorigenesis. Methods In this study, we developed a novel gene delivery system with a self-assembly method by Methoxy polyethylene glycol-polycaprolactone (MPEG-PCL) and DOTAP(DMC). And EZH2si-DMC was used to research anti-glioma both in vitro and in vivo. Results DMC with zeta-potential value of 36.7 mV and size of 35.6 nm showed good performance in the delivery siRNA to glioma cell in vitro with high 98% transfection efficiency. EZH2si-DMC showed good anti-glioma effect in vitro through inducing cell apoptosis and inhibiting cell growth. What’s more, treatment of tumor-bearing mice with DMC-EZH2si complex had significantly inhibited tumor growth at the subcutaneous model in vivo by inhibiting EZH2 protein expression, promoting apoptosis and reducing proliferation. Conclusion The EZH2 siRNA and DMC complex may be used to treat the glioma in clinical as a new drug.
Collapse
Affiliation(s)
- Xiang Wang
- Department of Neurosurgery, Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China,
| | - Yuanqi Hua
- Department of Neurosurgery, Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China,
| | - Guangya Xu
- Department of Neurosurgery, Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China,
| | - Senyi Deng
- Department of Neurosurgery, Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China,
| | - Daoke Yang
- Tumor Hospital of First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Xiang Gao
- Department of Neurosurgery, Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China,
| |
Collapse
|
47
|
Zhong Y, Wang Y, Luo L, Nurhidayah D, Maruf A, Gregersen H, Wu W, Wang GX. Targeted polyethylenimine/(p53 plasmid) nanocomplexes for potential antitumor applications. NANOTECHNOLOGY 2019; 30:145601. [PMID: 30524021 DOI: 10.1088/1361-6528/aaf41a] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The development of the tumor-targeting ability of nanocarriers is of paramount importance for gene delivery into tumor lesions as well as to avoid biotoxicity. Here we report the synthesis of the polyethyleneimine-fluorescein isothiocyanate-folic acid (PEI-FITC-FA) polymer, which could condense the tumor suppressor pp53 to form nanocomplexes. These targeted nanocomplexes exhibited favorable physical properties including a small size of <100 nm, exploiting the enhanced permeability and retention effect and tumor-targeting ability by binding to the overexpressed FA receptors on tumor cell surfaces. In addition, once the nanocomplexes are accumulating in the tumor tissue, the target functional ligand, FA, can selectively recognize the over-expressed FA receptor and subsequently remain on the tumor cell surface, which can significantly promote the tumor cell uptake because of the time- and concentration-dependent internalization caused by the enhanced interaction between nanocomplex and tumor cell. Our results indicated that PEI-FITC-FA/pp53 nanocomplexes could be efficiently delivered into tumor cells, and subsequently induce tumor cell apoptosis. Thus, the targeted cationic polymer PEI-FITC-FA could be used as an advanced nanocarrier for gene delivery.
Collapse
Affiliation(s)
- Yuan Zhong
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Carbajo-Gordillo AI, Rodríguez-Lavado J, Jiménez Blanco JL, Benito JM, Di Giorgio C, Vélaz I, Tros de Ilarduya C, Ortiz Mellet C, García Fernández JM. Trehalose-based Siamese twin amphiphiles with tunable self-assembling, DNA nanocomplexing and gene delivery properties. Chem Commun (Camb) 2019; 55:8227-8230. [DOI: 10.1039/c9cc04489b] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Trehalose Siamese twin vectors, encompassing gemini and facial amphiphilicity, promote pDNA compaction into core–shell nanocomplexes and selective delivery in the lungs.
Collapse
Affiliation(s)
| | - Julio Rodríguez-Lavado
- Department of Organic Chemistry
- Faculty of Chemistry, University of Seville
- 41012 Seville
- Spain
| | | | - Juan M. Benito
- Instituto de Investigaciones Químicas (IIQ)
- CSIC – Universidad de Sevilla
- 41092 Sevilla
- Spain
| | | | - Itziar Vélaz
- Department of Chemistry
- Faculty of Sciences
- University of Navarra
- Pamplona
- Spain
| | - Concepción Tros de Ilarduya
- Department of Pharmaceutical Technology and Chemistry
- School of Pharmacy and Nutrition
- University of Navarra
- 31080 Pamplona
- Spain
| | - Carmen Ortiz Mellet
- Department of Organic Chemistry
- Faculty of Chemistry, University of Seville
- 41012 Seville
- Spain
| | | |
Collapse
|
49
|
Qiu N, Gao J, Liu Q, Wang J, Shen Y. Enzyme-Responsive Charge-Reversal Polymer-Mediated Effective Gene Therapy for Intraperitoneal Tumors. Biomacromolecules 2018; 19:2308-2319. [DOI: 10.1021/acs.biomac.8b00440] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Nasha Qiu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| | - Jianqing Gao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qi Liu
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Jinqiang Wang
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| | - Youqing Shen
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
50
|
Naso MF, Tomkowicz B, Perry WL, Strohl WR. Adeno-Associated Virus (AAV) as a Vector for Gene Therapy. BioDrugs 2018; 31:317-334. [PMID: 28669112 PMCID: PMC5548848 DOI: 10.1007/s40259-017-0234-5] [Citation(s) in RCA: 779] [Impact Index Per Article: 111.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
There has been a resurgence in gene therapy efforts that is partly fueled by the identification and understanding of new gene delivery vectors. Adeno-associated virus (AAV) is a non-enveloped virus that can be engineered to deliver DNA to target cells, and has attracted a significant amount of attention in the field, especially in clinical-stage experimental therapeutic strategies. The ability to generate recombinant AAV particles lacking any viral genes and containing DNA sequences of interest for various therapeutic applications has thus far proven to be one of the safest strategies for gene therapies. This review will provide an overview of some important factors to consider in the use of AAV as a vector for gene therapy.
Collapse
Affiliation(s)
- Michael F Naso
- Janssen Research and Development, 200 McKean Road, Spring House, PA, 19477, USA.
| | - Brian Tomkowicz
- Janssen Research and Development, 200 McKean Road, Spring House, PA, 19477, USA
| | - William L Perry
- Janssen Research and Development, 200 McKean Road, Spring House, PA, 19477, USA
| | | |
Collapse
|