1
|
Jiao L, Sun Z, Sun Z, Liu J, Deng G, Wang X. Nanotechnology-based non-viral vectors for gene delivery in cardiovascular diseases. Front Bioeng Biotechnol 2024; 12:1349077. [PMID: 38303912 PMCID: PMC10830866 DOI: 10.3389/fbioe.2024.1349077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 01/08/2024] [Indexed: 02/03/2024] Open
Abstract
Gene therapy is a technique that rectifies defective or abnormal genes by introducing exogenous genes into target cells to cure the disease. Although gene therapy has gained some accomplishment for the diagnosis and therapy of inherited or acquired cardiovascular diseases, how to efficiently and specifically deliver targeted genes to the lesion sites without being cleared by the blood system remains challenging. Based on nanotechnology development, the non-viral vectors provide a promising strategy for overcoming the difficulties in gene therapy. At present, according to the physicochemical properties, nanotechnology-based non-viral vectors include polymers, liposomes, lipid nanoparticles, and inorganic nanoparticles. Non-viral vectors have an advantage in safety, efficiency, and easy production, possessing potential clinical application value when compared with viral vectors. Therefore, we summarized recent research progress of gene therapy for cardiovascular diseases based on commonly used non-viral vectors, hopefully providing guidance and orientation for future relevant research.
Collapse
Affiliation(s)
- Liping Jiao
- The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Zhuokai Sun
- Queen Mary School, Nanchang University, Nanchang, China
| | - Zhihong Sun
- The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Jie Liu
- The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Guanjun Deng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Shenzhen, China
| | - Xiaozhong Wang
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
- School of Public Health, Nanchang University, Nanchang, China
| |
Collapse
|
2
|
Abstract
This Review examines the state-of-the-art in the delivery of nucleic acid therapies that are directed to the vascular endothelium. First, we review the most important homeostatic functions and properties of the vascular endothelium and summarize the nucleic acid tools that are currently available for gene therapy and nucleic acid delivery. Second, we consider the opportunities available with the endothelium as a therapeutic target and the experimental models that exist to evaluate the potential of those opportunities. Finally, we review the progress to date from investigations that are directly targeting the vascular endothelium: for vascular disease, for peri-transplant therapy, for angiogenic therapies, for pulmonary endothelial disease, and for the blood-brain barrier, ending with a summary of the future outlook in this field.
Collapse
Affiliation(s)
| | | | | | - W. Mark Saltzman
- Department of Biomedical Engineering
- Department of Chemical & Environmental Engineering
- Department of Cellular & Molecular Physiology
- Department of Dermatology, Yale School of Medicine, New Haven, CT 06510
| |
Collapse
|
3
|
Chopra H, Bibi S, Mishra AK, Tirth V, Yerramsetty SV, Murali SV, Ahmad SU, Mohanta YK, Attia MS, Algahtani A, Islam F, Hayee A, Islam S, Baig AA, Emran TB. Nanomaterials: A Promising Therapeutic Approach for Cardiovascular Diseases. JOURNAL OF NANOMATERIALS 2022; 2022. [DOI: 10.1155/2022/4155729] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 02/05/2022] [Indexed: 09/01/2023]
Abstract
Cardiovascular diseases (CVDs) are a primary cause of death globally. A few classic and hybrid treatments exist to treat CVDs. However, they lack in both safety and effectiveness. Thus, innovative nanomaterials for disease diagnosis and treatment are urgently required. The tiny size of nanomaterials allows them to reach more areas of the heart and arteries, making them ideal for CVDs. Atherosclerosis causes arterial stenosis and reduced blood flow. The most common treatment is medication and surgery to stabilize the disease. Nanotechnologies are crucial in treating vascular disease. Nanomaterials may be able to deliver medications to lesion sites after being infused into the circulation. Newer point‐of‐care devices have also been considered together with nanomaterials. For example, this study will look at the use of nanomaterials in imaging, diagnosing, and treating CVDs.
Collapse
|
4
|
Gissler MC, Scherrer P, Anto-Michel N, Pennig J, Hoppe N, Füner L, Härdtner C, Stachon P, Li X, Mitre LS, Marchini T, Madl J, Wadle C, Hilgendorf I, von Zur Mühlen C, Bode C, Weber C, Lutgens E, Wolf D, Gerdes N, Zirlik A, Willecke F. Deficiency of Endothelial CD40 Induces a Stable Plaque Phenotype and Limits Inflammatory Cell Recruitment to Atherosclerotic Lesions in Mice. Thromb Haemost 2021; 121:1530-1540. [PMID: 33618394 DOI: 10.1055/a-1397-1858] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
OBJECTIVES The co-stimulatory CD40L-CD40 dyad exerts a critical role in atherosclerosis by modulating leukocyte accumulation into developing atherosclerotic plaques. The requirement for cell-type specific expression of both molecules, however, remains elusive. Here, we evaluate the contribution of CD40 expressed on endothelial cells (ECs) in a mouse model of atherosclerosis. METHODS AND RESULTS Atherosclerotic plaques of apolipoprotein E-deficient (Apoe -/- ) mice and humans displayed increased expression of CD40 on ECs compared with controls. To interrogate the role of CD40 on ECs in atherosclerosis, we induced EC-specific (BmxCreERT2-driven) deficiency of CD40 in Apoe -/- mice. After feeding a chow diet for 25 weeks, EC-specific deletion of CD40 (iEC-CD40) ameliorated plaque lipid deposition and lesional macrophage accumulation but increased intimal smooth muscle cell and collagen content, while atherosclerotic lesion size did not change. Leukocyte adhesion to the vessel wall was impaired in iEC-CD40-deficient mice as demonstrated by intravital microscopy. In accord, expression of vascular cell adhesion molecule 1 (VCAM-1) and intercellular adhesion molecule 1 (ICAM-1) in the vascular endothelium declined after deletion of CD40. In vitro, antibody-mediated inhibition of human endothelial CD40 significantly abated monocyte adhesion on ECs. CONCLUSION Endothelial deficiency of CD40 in mice promotes structural features associated with a stable plaque phenotype in humans and decreases leukocyte adhesion. These results suggest that endothelial-expressed CD40 contributes to inflammatory cell migration and consecutive plaque formation in atherogenesis.
Collapse
Affiliation(s)
- Mark Colin Gissler
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Philipp Scherrer
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Nathaly Anto-Michel
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jan Pennig
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Natalie Hoppe
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lisa Füner
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Carmen Härdtner
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Peter Stachon
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Xiaowei Li
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lucia Sol Mitre
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Timoteo Marchini
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Josef Madl
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Germany
| | - Carolin Wadle
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ingo Hilgendorf
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Constantin von Zur Mühlen
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christoph Bode
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
- German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Esther Lutgens
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
- German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
- Department of Medical Biochemistry, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Dennis Wolf
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Norbert Gerdes
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Andreas Zirlik
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Florian Willecke
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Klinik für Allgemeine und Interventionelle Kardiologie/Angiologie, Herz- und Diabeteszentrum Nordrhein-Westfalen, Universitätsklinik der Ruhr-Universität Bochum, Bochum, Germany
| |
Collapse
|
5
|
Poly (Lactic- co-Glycolic Acid) Nanoparticles and Nanoliposomes for Protein Delivery in Targeted Therapy: A Comparative In Vitro Study. Polymers (Basel) 2020; 12:polym12112566. [PMID: 33139610 PMCID: PMC7692461 DOI: 10.3390/polym12112566] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/27/2020] [Accepted: 10/29/2020] [Indexed: 12/12/2022] Open
Abstract
Over the previous years, the design, development, and potential application of nanocarriers in the medical field have been intensively studied for their ability to preserve drug properties, especially their pharmacological activity, and to improve their bioavailability. This work is a comparative study between two different types of nanocarriers, poly (lactic-co-glycolic acid)-based nanoparticles and phosphatidylcholine-based nanoliposomes, both prepared for the encapsulation of bovine serum albumin as a model protein. Polymeric nanoparticles were produced using the double emulsion water-oil-water evaporation method, whereas nanoliposomes were obtained by the thin-film hydration method. Both nanocarriers were characterized by morphological analysis, particle mean size, particle size distribution, and protein entrapment efficiency. Invitro release studies were performed for 12 days at 37 °C. In order to explore a possible application of these nanocarriers for a targeted therapy in the cardiovascular field, hemolytic activity and biocompatibility, in terms of cell viability, were performed by using human red blood cells and EA.hy926 human endothelial cell line, respectively.
Collapse
|
6
|
Li J, Shen M, Shi X. Poly(amidoamine) Dendrimer-Gold Nanohybrids in Cancer Gene Therapy: A Concise Overview. ACS APPLIED BIO MATERIALS 2020; 3:5590-5605. [DOI: 10.1021/acsabm.0c00863] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Jin Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, International Joint Laboratory for Advanced Fiber and Low-Dimension Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, People’s Republic of China
| | - Mingwu Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, International Joint Laboratory for Advanced Fiber and Low-Dimension Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, People’s Republic of China
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, International Joint Laboratory for Advanced Fiber and Low-Dimension Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, People’s Republic of China
- CQM-Centro de Quimica da Madeira, Universidade da Madeira, Campus da Penteada, Funchal 9020-105, Portugal
| |
Collapse
|
7
|
Zeng M, Alshehri F, Zhou D, Lara-Sáez I, Wang X, Li X, A S, Xu Q, Zhang J, Wang W. Efficient and Robust Highly Branched Poly(β-amino ester)/Minicircle COL7A1 Polymeric Nanoparticles for Gene Delivery to Recessive Dystrophic Epidermolysis Bullosa Keratinocytes. ACS APPLIED MATERIALS & INTERFACES 2019; 11:30661-30672. [PMID: 31390173 DOI: 10.1021/acsami.9b13135] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Recessive dystrophic epidermolysis bullosa (RDEB) is a severe congenital skin fragility disease caused by COL7A1 mutations that result in type VII collagen (C7) deficiency. Herein, we report a synergistic polyplex system that can efficiently restore C7 expression in RDEB keratinocytes. A highly branched multifunctional poly(β-amino ester) (HPAE), termed as HC32-122, was optimized systematically as the high-performance gene delivery vector for keratinocytes, achieving much higher transfection capability than polyethylenimine, SuperFect, and Lipofectamine 2000 without inducing obvious cytotoxicity. Concurrently, a 12 kb length minicircle DNA encoding ∼9 kb full-length COL7A1 (MCC7) devoid of bacterial sequence was biosynthesized as the therapeutic gene. Combining the highly potent polymer and the miniaturized gene structure, HC32-122/MCC7 polyplexes achieve 96.4% cellular uptake efficiency, 4019-fold COL7A1 mRNA enhancement, and robust recombinant C7 expression. Structure-property investigations reveal that HC32-122 can effectively condense MCC7 to form small, uniform, compact, and positively charged spherical nanoparticles with high DNA release flexibility. Moreover, formulation study shows that sucrose is conductive to lyophilized HC32-122/DNA polyplexes for maintaining the transfection capability. Direct frozen polyplexes can maintain full gene transfection capability after one-year storage. High efficiency, biocompatibility, facile manipulation, and long-term stability make the HC32-122/MCC7 system a promising bench-to-bed candidate for treating the debilitating RDEB.
Collapse
Affiliation(s)
- Ming Zeng
- Charles Institute of Dermatology , University College Dublin , Dublin D04 V1W8 , Ireland
- Department of Dermatology , The First Affiliated Hospital of Anhui Medical University , Hefei 230022 , China
| | - Fatma Alshehri
- Charles Institute of Dermatology , University College Dublin , Dublin D04 V1W8 , Ireland
- Princess Nourah bint Abdulrahman University , Riyadh 11671 , Saudi Arabia
| | - Dezhong Zhou
- Charles Institute of Dermatology , University College Dublin , Dublin D04 V1W8 , Ireland
- School of Chemical Engineering and Technology , Xi'an Jiaotong University , Xi'an , Shaanxi 710049 , China
| | - Irene Lara-Sáez
- Charles Institute of Dermatology , University College Dublin , Dublin D04 V1W8 , Ireland
| | - Xi Wang
- Charles Institute of Dermatology , University College Dublin , Dublin D04 V1W8 , Ireland
| | - Xiaolin Li
- Charles Institute of Dermatology , University College Dublin , Dublin D04 V1W8 , Ireland
| | - Sigen A
- Charles Institute of Dermatology , University College Dublin , Dublin D04 V1W8 , Ireland
| | - Qian Xu
- Charles Institute of Dermatology , University College Dublin , Dublin D04 V1W8 , Ireland
| | - Jing Zhang
- Charles Institute of Dermatology , University College Dublin , Dublin D04 V1W8 , Ireland
| | - Wenxin Wang
- Charles Institute of Dermatology , University College Dublin , Dublin D04 V1W8 , Ireland
| |
Collapse
|
8
|
Farahavar G, Abolmaali SS, Gholijani N, Nejatollahi F. Antibody-guided nanomedicines as novel breakthrough therapeutic, diagnostic and theranostic tools. Biomater Sci 2019; 7:4000-4016. [PMID: 31355391 DOI: 10.1039/c9bm00931k] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Recent advances in nanotechnology, such as the development of various types of nanoparticles and hybrid nanomaterials, have revolutionized nanomedicine. The small size, customizable surface, enhanced solubility, and multi-functionality endow the nanoparticles with an ability to interact with complex cellular and biological functions in new ways. Furthermore, these systems can deliver drugs to specific tissues and provide a targeted therapy. For this purpose, different categories of molecules, particularly antibodies, have been used as ligands. Antibody-conjugated nanomaterials can significantly enhance the efficiency of nanomedicines, especially in the field of cancer. This review is focused on three major medical applications of antibody-conjugated nanomaterials, namely, therapeutic, diagnostic and theranostic applications. To provide comprehensive information on the topic and an overview of these hybrid nanomaterials for biomedical applications, a brief summary of nanomaterials and antibodies is given. Moreover, the review has depicted the potential applications of antibody-conjugated nanomaterials in different fields and their capabilities to empower nanomedicine, particularly in relation to the treatment and detection of malignancies.
Collapse
Affiliation(s)
- Ghazal Farahavar
- Pharmaceutical Nanotechnology Department, Shiraz University of Medical Sciences, Shiraz 71345, Iran.
| | - Samira Sadat Abolmaali
- Pharmaceutical Nanotechnology Department and Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz 71345, Iran.
| | - Nasser Gholijani
- Autoimmune Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Foroogh Nejatollahi
- Shiraz HIV/AIDS research center, Institute of health, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
9
|
Zeng M, Zhou D, Alshehri F, Lara-Sáez I, Lyu Y, Creagh-Flynn J, Xu Q, A S, Zhang J, Wang W. Manipulation of Transgene Expression in Fibroblast Cells by a Multifunctional Linear-Branched Hybrid Poly(β-Amino Ester) Synthesized through an Oligomer Combination Approach. NANO LETTERS 2019; 19:381-391. [PMID: 30565945 DOI: 10.1021/acs.nanolett.8b04098] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Delivery of functional genetic materials into fibroblast cells to manipulate the transgene expression is of great significance in skin gene therapy. Despite numerous polymeric gene delivery systems having been developed, highly safe and efficient fibroblast gene transfection has not yet been achieved. Here, through a new linear oligomer combination strategy, linear poly(β-amino ester) oligomers are connected by the branching units, forming a new type of poly(β-amino ester). This new multifunctional linear-branched hybrid poly(β-amino ester) (LBPAE) shows high-performance fibroblast gene transfection. In human primary dermal fibroblasts (HPDFs) and mouse embryo fibroblasts (3T3s), ultrahigh transgene expression is achieved by LBPAE: up to 3292-fold enhancement in Gaussia luciferase (Gluc) expression and nearly 100% of green fluorescence protein expression are detected. Concurrently, LBPAE is of high in vitro biocompatibility. In depth mechanistic studies reveal that versatile LBPAE can navigate multiple extra- and intracellular barriers involved in the fibroblast gene transfection. More importantly, LBPAE can effectively deliver minicircle DNA encoding COL7A1 gene (a large and functional gene construct) to substantially upregulate the expression of type VII collagen (C7) in HPDFs, demonstrating its great potential in the treatment of C7-deficiency related genodermatoses such as recessive dystrophic epidermolysis bullosa.
Collapse
Affiliation(s)
- Ming Zeng
- Charles Institute of Dermatology, School of Medicine , University College Dublin , Dublin 4 , Ireland
- Department of Dermatology , the First Affiliated Hospital of Anhui Medical University , Hefei 230022 , China
| | - Dezhong Zhou
- Charles Institute of Dermatology, School of Medicine , University College Dublin , Dublin 4 , Ireland
- School of Chemical Engineering and Technology (SCET) , Xi'an Jiaotong University , Xi'an , Shaanxi , China
| | - Fatma Alshehri
- Charles Institute of Dermatology, School of Medicine , University College Dublin , Dublin 4 , Ireland
| | - Irene Lara-Sáez
- Charles Institute of Dermatology, School of Medicine , University College Dublin , Dublin 4 , Ireland
| | - Yuanning Lyu
- Charles Institute of Dermatology, School of Medicine , University College Dublin , Dublin 4 , Ireland
| | - Jack Creagh-Flynn
- Charles Institute of Dermatology, School of Medicine , University College Dublin , Dublin 4 , Ireland
| | - Qian Xu
- Charles Institute of Dermatology, School of Medicine , University College Dublin , Dublin 4 , Ireland
| | - Sigen A
- Charles Institute of Dermatology, School of Medicine , University College Dublin , Dublin 4 , Ireland
| | - Jing Zhang
- Charles Institute of Dermatology, School of Medicine , University College Dublin , Dublin 4 , Ireland
| | - Wenxin Wang
- Charles Institute of Dermatology, School of Medicine , University College Dublin , Dublin 4 , Ireland
| |
Collapse
|
10
|
Lai WF, Rogach AL, Wong WT. Molecular design of upconversion nanoparticles for gene delivery. Chem Sci 2017; 8:7339-7358. [PMID: 29163885 PMCID: PMC5672820 DOI: 10.1039/c7sc02956j] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 08/29/2017] [Indexed: 12/17/2022] Open
Abstract
Due to their large anti-Stokes shifts, sharp emission spectra and long excited-state lifetimes, upconversion nanoparticles (UCNPs) have attracted an increasing amount of research interests, and have shown great potential for enhancing the practical utility of gene therapy, whose versatility has been limited by existing gene delivery technologies that are basically mono-functional in nature. Despite this, up to now in-depth analysis of the development of UCNPs for gene delivery has been scant in the literature, even though there has been an upsurge of reviews on the chemistry of UCNPs and their applications in bioimaging and drug delivery. To fill this gap, this review aims to present the latest advances in the development and applications of UCNPs as gene carriers. Prior to describing the prominent works published in the field, a critical view on the properties, chemistry and molecular design of UCNPs for gene delivery is provided. With a synopsis of the recent advances in UCNP-mediated gene delivery, challenges and opportunities could be illuminated for clinical translation of works in this nascent field of research.
Collapse
Affiliation(s)
- Wing-Fu Lai
- School of Pharmaceutical Sciences , Health Science Centre , Shenzhen University , Shenzhen , China
- Department of Applied Biology & Chemical Technology , The Hong Kong Polytechnic University , Hong Kong . ;
| | - Andrey L Rogach
- Department of Materials Science and Engineering & Centre for Functional Photonics (CFP) , City University of Hong Kong , Hong Kong
| | - Wing-Tak Wong
- Department of Applied Biology & Chemical Technology , The Hong Kong Polytechnic University , Hong Kong . ;
| |
Collapse
|
11
|
Ayatollahi S, Salmasi Z, Hashemi M, Askarian S, Oskuee RK, Abnous K, Ramezani M. Aptamer-targeted delivery of Bcl-xL shRNA using alkyl modified PAMAM dendrimers into lung cancer cells. Int J Biochem Cell Biol 2017; 92:210-217. [PMID: 29031805 DOI: 10.1016/j.biocel.2017.10.005] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 10/07/2017] [Accepted: 10/10/2017] [Indexed: 11/25/2022]
Abstract
RNAi-based gene therapy has been recently considered as a promising approach against cancer. Targeted delivery of drug, gene or therapeutic RNAi-based systems to tumor cells is one of the important issues in order to reduce side effects on normal cells. Several strategies have been developed to improve the safety and selectivity of cancer treatments including antibodies, peptides and recently aptamers with various attractive characteristics including higher target specificity, affinity and reduced toxicity. Here we described a novel targeted delivery platform comprising modified PAMAM with 10-bromodecanoic acid (10C) and 10C-PEG for improvement of transfection efficiency, AS1411 aptamer for targeting nucleolin ligand on target cancer cells and shRNA plasmid for specific knockdown of Bcl-xL protein. Modified vector could significantly improve the transfection efficiency even after covalent or non-covalent aptamer binding compared to the non-targeted vector in A549 cells. The results of gene silencing and apoptosis assay indicated that our targeted shRNA delivery system could efficiently down-regulate the Bcl-xL expression up to 25% and induce 14% late apoptosis in target cancer cells with strong cell selectivity. This study proposed a novel targeted non-viral system for shRNA-mediated gene-silencing in cancer cells.
Collapse
Affiliation(s)
- Sara Ayatollahi
- Pharmaceutical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Salmasi
- Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Hashemi
- Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saeedeh Askarian
- Research Center of Advanced Technologies in Medicine, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran; Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Kazemi Oskuee
- Targeted drug delivery research center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
12
|
Wodicka JR, Chambers AM, Sangha GS, Goergen CJ, Panitch A. Development of a Glycosaminoglycan Derived, Selectin Targeting Anti-Adhesive Coating to Treat Endothelial Cell Dysfunction. Pharmaceuticals (Basel) 2017; 10:ph10020036. [PMID: 28353658 PMCID: PMC5490393 DOI: 10.3390/ph10020036] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 03/22/2017] [Accepted: 03/24/2017] [Indexed: 12/26/2022] Open
Abstract
Endothelial cell (EC) dysfunction is associated with many disease states including deep vein thrombosis (DVT), chronic kidney disease, sepsis and diabetes. Loss of the glycocalyx, a thin glycosaminoglycan (GAG)-rich layer on the EC surface, is a key feature of endothelial dysfunction and increases exposure of EC adhesion molecules such as selectins, which are involved in platelet binding to ECs. Once bound, platelets cause thrombus formation and an increased inflammatory response. We have developed a GAG derived, selectin targeting anti-adhesive coating (termed EC-SEAL) consisting of a dermatan sulfate backbone and multiple selectin-binding peptides designed to bind to inflamed endothelium and prevent platelet binding to create a more quiescent endothelial state. Multiple EC-SEAL variants were evaluated and the lead variant was found to preferentially bind to selectin-expressing ECs and smooth muscle cells (SMCs) and inhibit platelet binding and activation in a dose-dependent manner. In an in vivo model of DVT, treatment with the lead variant resulted in reduced thrombus formation. These results indicate that EC-SEAL has promise as a potential therapeutic in the treatment of endothelial dysfunction.
Collapse
Affiliation(s)
- James R Wodicka
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA.
- Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Andrea M Chambers
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA.
| | - Gurneet S Sangha
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA.
| | - Craig J Goergen
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA.
| | - Alyssa Panitch
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA.
- Department of Biomedical Engineering, University of California-Davis, Davis, CA 95616, USA.
| |
Collapse
|
13
|
Abd Ellah NH, Abouelmagd SA. Surface functionalization of polymeric nanoparticles for tumor drug delivery: approaches and challenges. Expert Opin Drug Deliv 2016; 14:201-214. [DOI: 10.1080/17425247.2016.1213238] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Noura H. Abd Ellah
- Department of Pharmaceutics, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| | - Sara A. Abouelmagd
- Department of Pharmaceutics, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| |
Collapse
|
14
|
Pyykkö I, Zou J, Schrott-Fischer A, Glueckert R, Kinnunen P. An Overview of Nanoparticle Based Delivery for Treatment of Inner Ear Disorders. Methods Mol Biol 2016; 1427:363-415. [PMID: 27259938 DOI: 10.1007/978-1-4939-3615-1_21] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Nanoparticles offer new possibilities for inner ear treatment as they can carry a variety of drugs, protein, and nucleic acids to inner ear. Nanoparticles are equipped with several functions such as targetability, immuno-transparency, biochemical stability, and ability to be visualized in vivo and in vitro. A group of novel peptides can be attached to the surface of nanoparticles that will enhance the cell entry, endosomal escape, and nuclear targeting. Eight different types of nanoparticles with different payload carrying strategies are available now. The transtympanic delivery of nanoparticles indicates that, depending on the type of nanoparticle, different migration pathways into the inner ear can be employed, and that optimal carriers can be designed according to the intended cargo. The use of nanoparticles as drug/gene carriers is especially attractive in conjunction with cochlear implantation or even as an inclusion in the implant as a drug/gene reservoir.
Collapse
Affiliation(s)
- Ilmari Pyykkö
- Department of Otolaryngology, University of Tampere and University Hospital of Tampere, Tampere, 33014, Finland. .,Hearing and Balance Research Unit, Field of Otolaryngology, School of Medicine, University of Tampere, Medisiinarinkatu 3, Tampere, 33520, Finland.
| | - Jing Zou
- BECS, Department of Biomedical Engineering and Computational Science, Aalto University, Aalto, 02150, Espoo, Finland
| | - Annelies Schrott-Fischer
- Department of Otolaryngology, Medical University of Innsbruck, Anichstrasse 35, Innsbruck, 6020, Austria
| | - Rudolf Glueckert
- Department of Otolaryngology, Medical University of Innsbruck, Anichstrasse 35, Innsbruck, 6020, Austria
| | - Paavo Kinnunen
- BECS, Department of Biomedical Engineering and Computational Science, Aalto University, Aalto, Finland
| |
Collapse
|
15
|
Uram Ł, Szuster M, Filipowicz A, Gargasz K, Wołowiec S, Wałajtys-Rode E. Different patterns of nuclear and mitochondrial penetration by the G3 PAMAM dendrimer and its biotin-pyridoxal bioconjugate BC-PAMAM in normal and cancer cells in vitro. Int J Nanomedicine 2015; 10:5647-61. [PMID: 26379435 PMCID: PMC4567239 DOI: 10.2147/ijn.s87307] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
The intracellular localization and colocalization of a fluorescently labeled G3 amine-terminated cationic polyamidoamine (PAMAM) dendrimer and its biotin-pyridoxal (BC-PAMAM) bioconjugate were investigated in a concentration-dependent manner in normal human fibroblast (BJ) and squamous epithelial carcinoma (SCC-15) cell lines. After 24 hours treatment, both cell lines revealed different patterns of intracellular dendrimer accumulation depending on their cytotoxic effects. Cancer cells exhibited much higher (20-fold) tolerance for native PAMAM treatment than fibroblasts, whereas BC-PAMAM was significantly toxic only for fibroblasts at 50 µM concentration. Fibroblasts accumulated the native and bioconjugated dendrimers in a concentration-dependent manner at nontoxic range of concentration, with significantly lower bioconjugate loading. After reaching the cytotoxicity level, fluorescein isothiocyanate-PAMAM accumulation remains at high, comparable level. In cancer cells, native PAMAM loading at higher, but not cytotoxic concentrations, was kept at constant level with a sharp increase at toxic concentration. Mander's coefficient calculated for fibroblasts and cancer cells confirmed more efficient native PAMAM penetration as compared to BC-PAMAM. Significant differences in nuclear dendrimer penetration were observed for both cell lines. In cancer cells, PAMAM signals amounted to ~25%-35% of the total nuclei area at all investigated concentrations, with lower level (15%-25%) observed for BC-PAMAM. In fibroblasts, the dendrimer nuclear signal amounted to 15% at nontoxic and up to 70% at toxic concentrations, whereas BC-PAMAM remained at a lower concentration-dependent level (0.3%-20%). Mitochondrial localization of PAMAM and BC-PAMAM revealed similar patterns in both cell lines, depending on the extracellular dendrimer concentration, and presented significantly lower signals from BC-PAMAM, which correlated well with the cytotoxicity.
Collapse
Affiliation(s)
- Łukasz Uram
- Bioorganic Chemistry Laboratory, Faculty of Chemistry, Rzeszow University of Technology, Rzeszow, Poland
| | - Magdalena Szuster
- Bioorganic Chemistry Laboratory, Faculty of Chemistry, Rzeszow University of Technology, Rzeszow, Poland
| | - Aleksandra Filipowicz
- Cosmetology Department, University of Information Technology and Management in Rzeszow, Rzeszow, Poland
| | - Krzysztof Gargasz
- Institute of Nursery and Health Sciences, Faculty of Medicine, University of Rzeszow, Rzeszow, Poland
| | - Stanisław Wołowiec
- Institute of Nursery and Health Sciences, Faculty of Medicine, University of Rzeszow, Rzeszow, Poland
| | - Elżbieta Wałajtys-Rode
- Department of Drug Technology and Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
| |
Collapse
|
16
|
Yu M, Jie X, Xu L, Chen C, Shen W, Cao Y, Lian G, Qi R. Recent Advances in Dendrimer Research for Cardiovascular Diseases. Biomacromolecules 2015; 16:2588-98. [DOI: 10.1021/acs.biomac.5b00979] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Maomao Yu
- Peking
University Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Xu Jie
- School
of Pharmacy, Shihezi University, Shihezi 832000, China
| | - Lu Xu
- Peking
University Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Cong Chen
- Peking
University Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Wanli Shen
- School
of Pharmacy, Shihezi University, Shihezi 832000, China
| | - Yini Cao
- Peking
University Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Guan Lian
- School
of Pharmacy, Shihezi University, Shihezi 832000, China
| | - Rong Qi
- Peking
University Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing 100191, China
- School
of Pharmacy, Shihezi University, Shihezi 832000, China
| |
Collapse
|
17
|
Dehshahri A, Sadeghpour H. Surface decorations of poly(amidoamine) dendrimer by various pendant moieties for improved delivery of nucleic acid materials. Colloids Surf B Biointerfaces 2015; 132:85-102. [PMID: 26022400 DOI: 10.1016/j.colsurfb.2015.05.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Revised: 05/05/2015] [Accepted: 05/07/2015] [Indexed: 12/22/2022]
|
18
|
Yang J, Zhang Q, Chang H, Cheng Y. Surface-Engineered Dendrimers in Gene Delivery. Chem Rev 2015; 115:5274-300. [PMID: 25944558 DOI: 10.1021/cr500542t] [Citation(s) in RCA: 321] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Jiepin Yang
- Shanghai
Key Laboratory of
Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, P. R. China
| | - Qiang Zhang
- Shanghai
Key Laboratory of
Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, P. R. China
| | - Hong Chang
- Shanghai
Key Laboratory of
Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, P. R. China
| | - Yiyun Cheng
- Shanghai
Key Laboratory of
Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, P. R. China
| |
Collapse
|
19
|
Liu X, Hao W, Lok CN, Wang YC, Zhang R, Wong KKY. Dendrimer encapsulation enhances anti-inflammatory efficacy of silver nanoparticles. J Pediatr Surg 2014; 49:1846-51. [PMID: 25487498 DOI: 10.1016/j.jpedsurg.2014.09.033] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 09/06/2014] [Indexed: 01/27/2023]
Abstract
BACKGROUND Our previous studies revealed that silver nanoparticles (AgNPs) promoted wound healing in part through their anti-inflammatory actions. As recent reports also suggested anti-inflammatory effects of dendrimers, we therefore undertook this study using dendrimer as the delivery system for AgNP to explore any potential synergistic anti-inflammatory efficacy. METHODS Lipopolysaccharide (LPS) was added to cultured RAW264.7 and J774.1 cells to mimic in vitro inflammation condition, followed by the addition of either silver dendrimer nanocomposite (Ag-DNC), AgNPs, or dendrimer. The levels of inflammatory markers TNF-alpha and interleukin-6 were assessed using ELISA assay. Furthermore, in vivo effects such of Ag-DNC, AgNPs, or dendrimer were studied in a burn wound model in mice. RESULTS Our results confirmed that both naked dendrimer and AgNPs had anti-inflammatory properties. In in vitro study, Ag-DNC was shown to have the best anti-inflammatory efficacy than AgNPs or dendrimer alone. In-vivo experiments also indicated that animals in the Ag-DNC group had the fastest healing time with the least inflammation. CONCLUSION Our study would suggest that dendrimer could provide additional anti-inflammatory benefits and might be an excellent delivery system for silver nanoparticles for future clinical application.
Collapse
Affiliation(s)
- Xuelai Liu
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Wei Hao
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Chun-Nam Lok
- Department of Chemistry, The University of Hong Kong, Hong Kong, China
| | - Yue Chun Wang
- Department of Physiology, Medical College, Ji Nan University, Guangzhou, China
| | - RuiZhong Zhang
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Kenneth K Y Wong
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
20
|
Luo K, He B, Wu Y, Shen Y, Gu Z. Functional and biodegradable dendritic macromolecules with controlled architectures as nontoxic and efficient nanoscale gene vectors. Biotechnol Adv 2014; 32:818-30. [PMID: 24389086 DOI: 10.1016/j.biotechadv.2013.12.008] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 12/13/2013] [Accepted: 12/15/2013] [Indexed: 12/28/2022]
Abstract
Gene therapy has provided great potential to revolutionize the treatment of many diseases. This therapy is strongly relied on whether a delivery vector efficiently and safely directs the therapeutic genes into the target tissue/cells. Nonviral gene delivery vectors have been emerging as a realistic alternative to the use of viral analogs with the potential of a clinically relevant output. Dendritic polymers were employed as nonviral vectors due to their branched and layered architectures, globular shape and multivalent groups on their surface, showing promise in gene delivery. In the present review, we try to bring out the recent trend of studies on functional and biodegradable dendritic polymers as nontoxic and efficient gene delivery vectors. By regulating dendritic polymer design and preparation, together with recent progress in the design of biodegradable polymers, it is possible to precisely manipulate their architectures, molecular weight and chemical composition, resulting in predictable tuning of their biocompatibility as well as gene transfection activities. The multifunctional and biodegradable dendritic polymers possessing the desirable characteristics are expected to overcome extra- and intracellular obstacles, and as efficient and nontoxic gene delivery vectors to move into the clinical arena.
Collapse
Affiliation(s)
- Kui Luo
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Bin He
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Yao Wu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Youqing Shen
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Zhejiang University, Hangzhou 310027, China; Center for Bionanoengineering, Zhejiang University, Hangzhou 310027, China.
| | - Zhongwei Gu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China.
| |
Collapse
|
21
|
Ennen F, Boye S, Lederer A, Cernescu M, Komber H, Brutschy B, Voit B, Appelhans D. Biohybrid structures consisting of biotinylated glycodendrimers and proteins: influence of the biotin ligand's number and chemical nature on the biotin–avidin conjugation. Polym Chem 2014. [DOI: 10.1039/c3py01152f] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
22
|
Injectable nanomaterials for drug delivery: Carriers, targeting moieties, and therapeutics. Eur J Pharm Biopharm 2013; 84:1-20. [DOI: 10.1016/j.ejpb.2012.12.009] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Revised: 12/07/2012] [Accepted: 12/10/2012] [Indexed: 02/07/2023]
|
23
|
Eniola-Adefeso O, Heslinga MJ, Porter TM. Design of nanovectors for therapy and imaging of cardiovascular diseases. Methodist Debakey Cardiovasc J 2012; 8:13-7. [PMID: 22891105 DOI: 10.14797/mdcj-8-1-13] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular diseases are widely prevalent in western societies, and their associated costs number in the billions of dollars and affect millions of patients each year. Nanovectors targeted to tissues involved in cardiovascular diseases offer great opportunities to improve cardiovascular treatment through their imaging and drug delivery capabilities. Vascular-targeted imaging particles may permit the early identification of atherosclerosis, discriminate between stable and vulnerable atherosclerotic plaques, or guide surgeons as they work on fragile vasculature. Tailored therapeutic nanoparticles may provide safer, more efficient and effective intervention through localization and release of encapsulated therapeutics. Nanovector design involves numerous considerations such as fabrication material, particle size, and surface-modification with ligands for targeting and increasing blood circulation times. Complex blood rheology may affect the efficiency with which dissimilarsized particles target ligand receptors associated with disease. Additionally, the intended use of a nanovector is a critical factor in its design as some materials with poor drug-loading qualities or release kinetics may be suitable for imaging purposes only. Overall, vectors targeted to the vasculature will need to be efficient in avoiding blood clearance, honing to the target location, and binding at the desired site.
Collapse
|
24
|
Monaghan M, Greiser U, Wall JG, O’Brien T, Pandit A. Interference: an alteRNAtive therapy following acute myocardial infarction. Trends Pharmacol Sci 2012; 33:635-45. [DOI: 10.1016/j.tips.2012.09.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 09/10/2012] [Accepted: 09/14/2012] [Indexed: 10/27/2022]
|
25
|
Kaminskas LM, Boyd BJ, Porter CJH. Dendrimer pharmacokinetics: the effect of size, structure and surface characteristics on ADME properties. Nanomedicine (Lond) 2012; 6:1063-84. [PMID: 21955077 DOI: 10.2217/nnm.11.67] [Citation(s) in RCA: 149] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Dendrimers show increasing promise as drug-delivery vectors and can be generated with a wide range of scaffold structures, sizes and surface functionalities. To this point, the majority of studies of dendrimer-based drug-delivery systems have detailed pharmacodynamic outcomes, or have followed the pharmacokinetics of a solubilized or conjugated drug. By contrast, detailed commentary on the in vivo fate of the dendrimer carrier is less evident, even though the pharmacokinetics of the carrier will likely dictate both pharmacodynamic and toxicokinetic outcomes. In the current article, the influence of size, structure and surface functionality on the absorption, distribution, metabolism and elimination (ADME) properties of dendrimers have been examined and the implications of these findings for delivery system design are discussed.
Collapse
Affiliation(s)
- Lisa M Kaminskas
- Drug Delivery Disposition & Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University. 381 Royal Parade, Parkville, VIC, 3052, Australia
| | | | | |
Collapse
|
26
|
Affiliation(s)
- Quanming Lin
- a College of Materials and Textiles, Zhejiang Sci-Tech University, Hangzhou 310018, P. R. China
| | - Guohua Jiang
- b College of Materials and Textiles, Zhejiang Sci-Tech University, Hangzhou 310018, P. R. China, Key Laboratory of Advanced Textile Materials and Manufacturing Technology (ATMT), Zhejiang Sci-Tech University, Ministry of Education, Hangzhou 310018, P. R. China;,
| | - Kangkang Tong
- c College of Materials and Textiles, Zhejiang Sci-Tech University, Hangzhou 310018, P. R. China
| |
Collapse
|
27
|
Khandare J, Calderón M, Dagia NM, Haag R. Multifunctional dendritic polymers in nanomedicine: opportunities and challenges. Chem Soc Rev 2011; 41:2824-48. [PMID: 22158998 DOI: 10.1039/c1cs15242d] [Citation(s) in RCA: 319] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Nanotechnology has resulted in materials that have greatly improved the effectiveness of drug delivery because of their ability to control matter on the nanoscale. Advanced forms of nanomedicine have been synthesized for better pharmacokinetics to obtain higher efficacy, less systemic toxicity, and better targeting. These criteria have long been the goal in nanomedicine, in particular, for systemic applications in oncological disorders. Now, the "holy grail" in nanomedicine is to design and synthesize new advanced macromolecular nanocarriers and to translate them from lab to clinic. This review describes the current and future perspectives of nanomedicine with particular emphasis on the clinical targets in cancer and inflammation. The advanced forms of liposomes and polyethylene glycol (PEG) based nanocarriers, as well as dendritic polymer conjugates will be discussed with particular attention paid to designs, synthetic strategies, and chemical pathways. In this critical review, we also report on the current status and perspective of dendritic polymer nanoconjugate platforms (e.g. polyamidoamine dendrimers and dendritic polyglycerols) for cellular localization and targeting of specific tissues (192 references).
Collapse
Affiliation(s)
- Jayant Khandare
- Piramal Life Sciences Ltd, 1 Nirlon Complex, Off Western Express Highway, Goregaon (E), Mumbai-400063, India
| | | | | | | |
Collapse
|
28
|
Jubeli E, Moine L, Vergnaud-Gauduchon J, Barratt G. E-selectin as a target for drug delivery and molecular imaging. J Control Release 2011; 158:194-206. [PMID: 21983284 DOI: 10.1016/j.jconrel.2011.09.084] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Accepted: 09/22/2011] [Indexed: 01/02/2023]
Abstract
E-selectin, also known as CD62E, is a cell adhesion molecule expressed on endothelial cells activated by cytokines. Like other selectins, it plays an important part in inflammation and in the adhesion of metastatic cancer cells to the endothelium. E-selectin recognizes and binds to sialylated carbohydrates present on the surface proteins of certain leukocytes. E-selectin has been chosen as a target for several therapeutic and medical imaging applications, based on its expression in the vicinity of inflammation, infection or cancer. These systems for drug delivery and molecular imaging include immunoconjugates, liposomes, nanoparticles, and microparticles prepared from a wide range of starting materials including lipids, synthetic polymers, polypeptides and organo-metallic structures. After a brief introduction presenting the selectin family and their implication in physiology and pathology, this review focuses on the formulation of these new delivery systems targeting E-selectin at a molecular level.
Collapse
Affiliation(s)
- Emile Jubeli
- Université Paris-Sud 11, Faculté de Pharmacie 5 rue J.B. Clément Chatenay-Malabry, FR 92296, UMR 8612 CNRS, LabEx LERMIT, France
| | | | | | | |
Collapse
|
29
|
Cyrus T, Wickline SA, Lanza GM. Nanotechnology in interventional cardiology. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2011; 4:82-95. [PMID: 21748858 DOI: 10.1002/wnan.154] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
High-grade atherosclerotic stenoses are reduced to zero or minimal residual stenosis grades by a single or a series of balloon angioplasties. Currently, stents are implanted to prevent immediate vascular recoil and elution of an antimitotic drug from the stent struts minimizes restenosis. An unwanted side-effect of this drug elution is delayed re-endothelialization which requires treatment with two anti-platelet drugs, in many cases for a minimum of 1 year to prevent acute in-stent thrombosis. Advances in stent design and drug elution technology, now in its fourth generation, have not abated this issue. Nanotechnology-based local drug delivery has the potential to achieve restenosis prevention while not impeding endothelial healing. Molecularly targeted drugs can be aimed to specifically bind to epitopes in the injured media and adventitia. Thus, endothelial healing may progress unhindered. To prevent restenosis, this technology may be used with bare metal or biodegradable stents. In this article novel nanoparticulate agents will be compared regarding their potential to deliver drugs to molecular targets within the vascular wall. Potential molecular targets, targeting mechanisms, drug-delivery propensities, and biocompatibility will be reviewed.
Collapse
Affiliation(s)
- Tillmann Cyrus
- Division of Cardiology, University of Missouri, One Hospital Drive, Columbia, MO, USA.
| | | | | |
Collapse
|
30
|
Monaghan M, Pandit A. RNA interference therapy via functionalized scaffolds. Adv Drug Deliv Rev 2011; 63:197-208. [PMID: 21241760 DOI: 10.1016/j.addr.2011.01.006] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2010] [Revised: 12/16/2010] [Accepted: 01/05/2011] [Indexed: 12/27/2022]
Abstract
Tissue engineering aims to provide structural and biomolecular cues to compromised tissues through scaffolds. An emerging biomolecular cue is that of RNA interference by which the expression of genes can be silenced through a potent endogenous pathway. Recombinant viral-based approaches in RNAi delivery exist; however non-viral strategies offer many opportunities to exploit this mechanism of regulation in a safer way. Current RNAi therapies in clinical trials are without a vector (naked) or have slightly modified structures. Modification of these molecules with efficient backbone moieties for improved stability and potency, protecting and buffering them with delivery vehicles, and using scaffolds as reservoirs of delivery is at the frontier of current research. However, to enable an efficient sustained therapeutic effect scaffolds have a potentially significant role to play. This review presents non-viral delivery of RNAi that have been attempted via tissue engineered scaffolds. For RNAi to have a clinical impact, it is imperative to evaluate optimal delivery systems to ensure that the efficacy of this promising technology can be maximized.
Collapse
Affiliation(s)
- Michael Monaghan
- Network of Excellence for Functional Biomaterials, National University of Ireland-Galway, Ireland
| | | |
Collapse
|
31
|
Bachtarzi H, Stevenson M, Šubr V, Ulbrich K, Seymour LW, Fisher KD. Targeting adenovirus gene delivery to activated tumour-associated vasculature via endothelial selectins. J Control Release 2010; 150:196-203. [PMID: 20965218 PMCID: PMC3071491 DOI: 10.1016/j.jconrel.2010.10.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2010] [Revised: 10/06/2010] [Accepted: 10/08/2010] [Indexed: 02/04/2023]
Abstract
Clinical experience with adenovirus vectors has highlighted the need for improved delivery and targeting. Tumour-associated endothelium offers an additional mechanism for enhanced viral uptake into tumours which is accessible for systemic gene delivery. Building on expertise in using polymer ‘stealthed’ viruses for targeting in vivo, adenovirus expressing luciferase (Adluc) was coated with an amino-reactive polymer based on poly [N-(2-hydroxypropyl) methacrylamide] to ablate normal infection pathways. Direct linkage of a monoclonal antibody against E-selectin (MHES) demonstrated E-selectin-specific transduction of tumour necrosis factor-α (TNF-α)-activated endothelial cells. A two-component targeting system using protein G was developed, to provide optimal antibody orientation. We report an enhancement in transduction of TNF-α-activated endothelium in vitro and ex vivo in a human umbilical vein cord model using the MHES antibody. Similarly a virus retargeted using a chimeric P-selectin Glycoprotein Ligand-1-Fc fusion (PSGL-1) protein showed better circulation kinetics and significant uptake into HepG2 xenografts following systemic administration in mice, with 36-fold higher genome copies, compared with non-modified virus. Immunohistochemistry staining of tumour sections from mice treated with PSGL-1-retargeted virus showed a co-localisation of firefly luciferase with CD31 suggesting selective endothelial targeting. Employment of optimal viral modification using protein G will enable exploration and comparison of alternative targeting ligands targeting tumour-associated endothelium.
Collapse
Affiliation(s)
- Houria Bachtarzi
- Department of Clinical Pharmacology, University of Oxford, Old Road Campus, Headington, Oxford OX3 7DQ, UK.
| | | | | | | | | | | |
Collapse
|
32
|
Tang M, Zhao XG, Gu YJ, Chen CZ. An in vitro model for studying neutrophil activation during cardiopulmonary bypass by using a polymerase chain reaction thermocycler. Altern Lab Anim 2010; 38:213-9. [PMID: 20602537 DOI: 10.1177/026119291003800307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The accurate temperature control of a polymerase chain reaction (PCR) thermocycler was exploited in developing an in vitro model to study neutrophil activation during cardiopulmonary bypass. Neutrophils from 12 volunteers underwent temperature changes in a PCR thermocycler (37 degrees C for 30 minutes, 28 degrees C for 60 minutes, and then 37 degrees C for 90 minutes). Different co-incubates were applied to neutrophils, as follows: Group A: phosphate-buffered saline solution; Group B: platelet activating factor (PAF) ; Group C: platelet-depleted plasma; Group D: platelet-depleted plasma + PAF; and Group E: platelet-rich plasma. Membrane-bound elastase (MBE) activity was measured every 30 minutes throughout the experiment. MBE activity decreased significantly after hypothermia, compared with the baseline level (p < 0.001), and it resumed an increase after re-warming. Among all co-incubates, platelet-rich plasma was the most potent pro-inflammatory stimulus to neutrophils. A linear correlation was found between MBE and platelet count in platelet-rich plasma (p = 0.004). A novel in vitro model involving a PCR thermocycler has been proved to be reliable in the study of neutrophil activation during cardiopulmonary bypass. The model could possibly be used as an alternative to animals in the development of new drugs to combat neutrophil damage to tissues and organs during cardiopulmonary bypass in cardiac surgery.
Collapse
Affiliation(s)
- Min Tang
- Department of Cardiothoracic Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | | | | | | |
Collapse
|
33
|
Wang P, Zhao XH, Wang ZY, Meng M, Li X, Ning Q. Generation 4 polyamidoamine dendrimers is a novel candidate of nano-carrier for gene delivery agents in breast cancer treatment. Cancer Lett 2010; 298:34-49. [PMID: 20594639 DOI: 10.1016/j.canlet.2010.06.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2010] [Revised: 06/01/2010] [Accepted: 06/02/2010] [Indexed: 10/19/2022]
Abstract
Polyamidoamine dendrimer (PAMAM-D) is a new gene vector developed in recent years. In this study, we successfully prepared G4PAMAM and detected its unique structure by NMR, FITR and TEM. We revealed that G4PAMAM could bind to human erythrocytes and BSA through electrostatic interaction respectively, and caused haemolysis and reduced bioavailability. However, G4PAMAM-VEGF-ASODN (antisense oligodeoxynucleotides) complex could prevent G4PAMAM from binding to the erythrocytes and BSA and remained stable as a conjugate, therefore the toxicity of the complex was reduced. Meanwhile, we showed that G4PAMAM could be used as a gene vector to deliver AODNs into breast cancer MDA-MB-231 cells without significant cell toxicity, and it enhanced cellular uptake of ODNs. In vivo experiment of human breast tumor xenograft mice model, G4PAMAM also showed more efficiency of accumulating VEGF-ASODN to inhibit the tumor vascularization of breast tumor tissue than naked AODN. Furthermore, G4PAMAM could protect DNA in cytoplasm from digestion of restriction enzymes, which was important to become an effective tool in gene research and therapy.
Collapse
Affiliation(s)
- Pei Wang
- Department of Oncology, The First Hospital Affiliated to School of Medicine of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China
| | | | | | | | | | | |
Collapse
|
34
|
Shcharbin DG, Klajnert B, Bryszewska M. Dendrimers in gene transfection. BIOCHEMISTRY (MOSCOW) 2009; 74:1070-9. [DOI: 10.1134/s0006297909100022] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|