1
|
Rajeev A, Siby A, Koottungal MJ, George J, John F. Knocking Down Barriers: Advances in siRNA Delivery. ChemistrySelect 2021. [DOI: 10.1002/slct.202103288] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Anjaly Rajeev
- Bioorganic Chemistry Laboratory Department of Chemistry Sacred Heart College (Autonomous) Kochi Kerala India- 682013
| | - Aiswarya Siby
- Bioorganic Chemistry Laboratory Department of Chemistry Sacred Heart College (Autonomous) Kochi Kerala India- 682013
| | - Merin James Koottungal
- Bioorganic Chemistry Laboratory Department of Chemistry Sacred Heart College (Autonomous) Kochi Kerala India- 682013
| | - Jinu George
- Bioorganic Chemistry Laboratory Department of Chemistry Sacred Heart College (Autonomous) Kochi Kerala India- 682013
| | - Franklin John
- Bioorganic Chemistry Laboratory Department of Chemistry Sacred Heart College (Autonomous) Kochi Kerala India- 682013
| |
Collapse
|
2
|
Zhang R, Nie T, Fang Y, Huang H, Wu J. Poly(disulfide)s: From Synthesis to Drug Delivery. Biomacromolecules 2021; 23:1-19. [PMID: 34874705 DOI: 10.1021/acs.biomac.1c01210] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bioresponsive polymers have been widely used in drug delivery because of their degradability. For example, poly(disulfide)s with repeating disulfide bonds in the main chain have attracted considerable research attention. The characteristics of the disulfide bonds, including their dynamic and reversible properties and their responsiveness to stimuli such as reductants, light, heat, and mechanical force, make them ideal platforms for on-demand drug delivery. This review introduces the synthesis methods and applications of poly(disulfide)s. Furthermore, the synthesis methods of poly(disulfide)s are classified on the basis of the monomers used: oxidative step-growth polymerization with dithiols, ring-opening polymerization with cyclic disulfides, and polymerization with linear disulfides. In addition, recent advances in poly(disulfide)s for the delivery of small-molecule or biomacromolecular drugs are discussed. Quantum-dot-loaded poly(disulfide) delivery systems for imaging are also included. This review provides an overview of the various design strategies employed in the construction of poly(disulfide) platforms to inspire new applications in the field of drug delivery.
Collapse
Affiliation(s)
- Ruhe Zhang
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province, School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| | - Tianqi Nie
- The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Yifen Fang
- Department of Cardiology, The Affiliated TCM Hospital of Guangzhou Medical University, Guangzhou 510180, China
| | - Hai Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jun Wu
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province, School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| |
Collapse
|
3
|
Pandey M, Ojha D, Bansal S, Rode AB, Chawla G. From bench side to clinic: Potential and challenges of RNA vaccines and therapeutics in infectious diseases. Mol Aspects Med 2021; 81:101003. [PMID: 34332771 DOI: 10.1016/j.mam.2021.101003] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/27/2021] [Accepted: 07/16/2021] [Indexed: 12/14/2022]
Abstract
The functional and structural versatility of Ribonucleic acids (RNAs) makes them ideal candidates for overcoming the limitations imposed by small molecule-based drugs. Hence, RNA-based biopharmaceuticals such as messenger RNA (mRNA) vaccines, antisense oligonucleotides (ASOs), small interfering RNAs (siRNAs), microRNA mimics, anti-miRNA oligonucleotides (AMOs), aptamers, riboswitches, and CRISPR-Cas9 are emerging as vital tools for the treatment and prophylaxis of many infectious diseases. Some of the major challenges to overcome in the area of RNA-based therapeutics have been the instability of single-stranded RNAs, delivery to the diseased cell, and immunogenicity. However, recent advancements in the delivery systems of in vitro transcribed mRNA and chemical modifications for protection against nucleases and reducing the toxicity of RNA have facilitated the entry of several exogenous RNAs into clinical trials. In this review, we provide an overview of RNA-based vaccines and therapeutics, their production, delivery, current advancements, and future translational potential in treating infectious diseases.
Collapse
Affiliation(s)
- Manish Pandey
- RNA Biology Laboratory, Regional Centre for Biotechnology, Faridabad, 121001, India
| | - Divya Ojha
- Laboratory of Synthetic Biology, Regional Centre for Biotechnology, Faridabad, 121001, India
| | - Sakshi Bansal
- RNA Biology Laboratory, Regional Centre for Biotechnology, Faridabad, 121001, India
| | - Ambadas B Rode
- Laboratory of Synthetic Biology, Regional Centre for Biotechnology, Faridabad, 121001, India.
| | - Geetanjali Chawla
- RNA Biology Laboratory, Regional Centre for Biotechnology, Faridabad, 121001, India.
| |
Collapse
|
4
|
Dutta K, Das R, Medeiros J, Thayumanavan S. Disulfide Bridging Strategies in Viral and Nonviral Platforms for Nucleic Acid Delivery. Biochemistry 2021; 60:966-990. [PMID: 33428850 PMCID: PMC8753971 DOI: 10.1021/acs.biochem.0c00860] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Self-assembled nanostructures that are sensitive to environmental stimuli are promising nanomaterials for drug delivery. In this class, disulfide-containing redox-sensitive strategies have gained enormous attention because of their wide applicability and simplicity of nanoparticle design. In the context of nucleic acid delivery, numerous disulfide-based materials have been designed by relying on covalent or noncovalent interactions. In this review, we highlight major advances in the design of disulfide-containing materials for nucleic acid encapsulation, including covalent nucleic acid conjugates, viral vectors or virus-like particles, dendrimers, peptides, polymers, lipids, hydrogels, inorganic nanoparticles, and nucleic acid nanostructures. Our discussion will focus on the context of the design of materials and their impact on addressing the current shortcomings in the intracellular delivery of nucleic acids.
Collapse
Affiliation(s)
- Kingshuk Dutta
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Ritam Das
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
- The Center for Bioactive Delivery- Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Jewel Medeiros
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
- The Center for Bioactive Delivery- Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - S. Thayumanavan
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Department of Biomedical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, Massachusetts 01003, United States
- The Center for Bioactive Delivery- Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| |
Collapse
|
5
|
Godeshala S, Miryala B, Dutta S, Christensen MD, Nandi P, Chiu PL, Rege K. A library of aminoglycoside-derived lipopolymer nanoparticles for delivery of small molecules and nucleic acids. J Mater Chem B 2020; 8:8558-8572. [PMID: 32830211 DOI: 10.1039/d0tb00924e] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Simultaneous delivery of small molecules and nucleic acids using a single vehicle can lead to novel combination treatments and multifunctional carriers for a variety of diseases. In this study, we report a novel library of aminoglycoside-derived lipopolymers nanoparticles (LPNs) for the simultaneous delivery of different molecular cargoes including nucleic acids and small-molecules. The LPN library was screened for transgene expression efficacy following delivery of plasmid DNA, and lead LPNs that showed high transgene expression efficacies were characterized using hydrodynamic size, zeta potential, 1H NMR and FT-IR spectroscopy, and transmission electron microscopy. LPNs demonstrated significantly higher efficacies for transgene expression than 25 kDa polyethyleneamine (PEI) and lipofectamine, including in presence of serum. Self-assembly of these cationic lipopolymers into nanoparticles also facilitated the delivery of small molecule drugs (e.g. doxorubicin) to cancer cells. LPNs were also employed for the simultaneous delivery of the small-molecule histone deacetylase (HDAC) inhibitor AR-42 together with plasmid DNA to cancer cells as a combination treatment approach for enhancing transgene expression. Taken together, our results indicate that aminoglycoside-derived LPNs are attractive vehicles for simultaneous delivery of imaging agents or chemotherapeutic drugs together with nucleic acids for different applications in medicine and biotechnology.
Collapse
Affiliation(s)
- Sudhakar Godeshala
- Chemical Engineering, Arizona State University, 501 E. Tyler Mall, ECG 303, Tempe, AZ 85287-6106, USA.
| | - Bhavani Miryala
- Chemical Engineering, Arizona State University, 501 E. Tyler Mall, ECG 303, Tempe, AZ 85287-6106, USA.
| | - Subhadeep Dutta
- School of Molecular Sciences, Arizona State University, Tempe, AZ 85287-6106, USA
| | - Matthew D Christensen
- Chemical Engineering, Arizona State University, 501 E. Tyler Mall, ECG 303, Tempe, AZ 85287-6106, USA.
| | - Purbasha Nandi
- School of Molecular Sciences, Arizona State University, Tempe, AZ 85287-6106, USA
| | - Po-Lin Chiu
- School of Molecular Sciences, Arizona State University, Tempe, AZ 85287-6106, USA
| | - Kaushal Rege
- Chemical Engineering, Arizona State University, 501 E. Tyler Mall, ECG 303, Tempe, AZ 85287-6106, USA.
| |
Collapse
|
6
|
Bajan S, Hutvagner G. RNA-Based Therapeutics: From Antisense Oligonucleotides to miRNAs. Cells 2020; 9:E137. [PMID: 31936122 PMCID: PMC7016530 DOI: 10.3390/cells9010137] [Citation(s) in RCA: 243] [Impact Index Per Article: 60.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 12/23/2019] [Accepted: 12/30/2019] [Indexed: 02/07/2023] Open
Abstract
The first therapeutic nucleic acid, a DNA oligonucleotide, was approved for clinical use in 1998. Twenty years later, in 2018, the first therapeutic RNA-based oligonucleotide was United States Food and Drug Administration (FDA) approved. This promises to be a rapidly expanding market, as many emerging biopharmaceutical companies are developing RNA interference (RNAi)-based, and RNA-based antisense oligonucleotide therapies. However, miRNA therapeutics are noticeably absent. miRNAs are regulatory RNAs that regulate gene expression. In disease states, the expression of many miRNAs is measurably altered. The potential of miRNAs as therapies and therapeutic targets has long been discussed and in the context of a wide variety of infections and diseases. Despite the great number of studies identifying miRNAs as potential therapeutic targets, only a handful of miRNA-targeting drugs (mimics or inhibitors) have entered clinical trials. In this review, we will discuss whether the investment in finding potential miRNA therapeutic targets has yielded feasible and practicable results, the benefits and obstacles of miRNAs as therapeutic targets, and the potential future of the field.
Collapse
Affiliation(s)
- Sarah Bajan
- Faculty of Science, University of Technology Sydney, Sydney, NSW 2000, Australia
- Health and Sport Science, University of Sunshine Coast, Sunshine Coast, QLD 4556, Australia
| | - Gyorgy Hutvagner
- School of Biomedical Engineering Faculty of Engineering and Information Technology, University of Technology Sydney, Sydney, NSW 2000, Australia
| |
Collapse
|
7
|
Zhou Y, Han S, Liang Z, Zhao M, Liu G, Wu J. Progress in arginine-based gene delivery systems. J Mater Chem B 2020; 8:5564-5577. [DOI: 10.1039/d0tb00498g] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Arginine based gene delivery systems with enhanced membrane penetration and lower cytotoxicity greatly enrich the gene vectors library and outline a new development direction of gene delivery.
Collapse
Affiliation(s)
- Yang Zhou
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province
- School of Biomedical Engineering
- Sun Yat-sen University
- Guangzhou
- China
| | - Shuyan Han
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province
- School of Biomedical Engineering
- Sun Yat-sen University
- Guangzhou
- China
| | - Zhiqing Liang
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province
- School of Biomedical Engineering
- Sun Yat-sen University
- Guangzhou
- China
| | - Meng Zhao
- Shenzhen Lansi Institute of Artificial Intelligence in Medicine
- Shenzhen
- China
| | - Guiting Liu
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province
- School of Biomedical Engineering
- Sun Yat-sen University
- Guangzhou
- China
| | - Jun Wu
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province
- School of Biomedical Engineering
- Sun Yat-sen University
- Guangzhou
- China
| |
Collapse
|
8
|
Xiao Y, Shi K, Qu Y, Chu B, Qian Z. Engineering Nanoparticles for Targeted Delivery of Nucleic Acid Therapeutics in Tumor. Mol Ther Methods Clin Dev 2019; 12:1-18. [PMID: 30364598 PMCID: PMC6197778 DOI: 10.1016/j.omtm.2018.09.002] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
In the past 10 years, with the increase of investment in clinical nano-gene therapy, there are many trials that have been discontinued due to poor efficacy and serious side effects. Therefore, it is particularly important to design a suitable gene delivery system. In this paper, we introduce the application of liposomes, polymers, and inorganics in gene delivery; also, different modifications with some stimuli-responsive systems can effectively improve the efficiency of gene delivery and reduce cytotoxicity and other side effects. Besides, the co-delivery of chemotherapy drugs with a drug tolerance-related gene or oncogene provides a better theoretical basis for clinical cancer gene therapy.
Collapse
Affiliation(s)
- Yao Xiao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Kun Shi
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Ying Qu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Bingyang Chu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Zhiyong Qian
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center, Chengdu, China
| |
Collapse
|
9
|
Xu L, Yang H. Folate-Decorated Polyamidoamine Dendrimer Nanoparticles for Head and Neck Cancer Gene Therapy. Methods Mol Biol 2019; 1974:393-408. [PMID: 31099016 DOI: 10.1007/978-1-4939-9220-1_26] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Gene delivery systems have been developed on the basis of dendrimers and many other types of nanoparticle carriers, but few have been developed for head and neck squamous cell carcinomas (HNSCC). Herein, we describe the design and synthesis of fluorescently labeled, folic acid-decorated polyamidoamine (PAMAM) generation 4 (G4) dendrimer conjugates for HNSCC-targeted gene delivery. This delivery system comprises a dendrimer as the carrier that is conjugated with folic acid (FA) as HNSCC targeting moiety and imaging agents fluorescein isothiocyanate (FITC) or IRDye 800CW (NIR) for in vitro trafficking or bioimaging, respectively. By complexing with plasmid or siRNA, G4-FA/plasmid (or siRNA) significantly enhances gene transfection or knockdown efficiency in HNSCC cells. In a mouse xenograft model of HNSCC, this versatile G4-FA vector shows high biocompatibility, tumor targeting, high uptake, and sustained retention, making it a suitable platform for HNSCC gene therapy.
Collapse
Affiliation(s)
- Leyuan Xu
- Section of Nephrology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Hu Yang
- Department of Chemical and Life Science Engineering, Virginia Commonwealth University, Richmond, VA, USA. .,Department of Pharmaceutics, Virginia Commonwealth University, Richmond, VA, USA. .,Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
10
|
Le TMD, Duong HTT, Thambi T, Giang Phan V, Jeong JH, Lee DS. Bioinspired pH- and Temperature-Responsive Injectable Adhesive Hydrogels with Polyplexes Promotes Skin Wound Healing. Biomacromolecules 2018; 19:3536-3548. [DOI: 10.1021/acs.biomac.8b00819] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Thai Minh Duy Le
- School of Chemical Engineering, Theranostic Macromolecules Research Center, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Huu Thuy Trang Duong
- School of Chemical Engineering, Theranostic Macromolecules Research Center, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Thavasyappan Thambi
- School of Chemical Engineering, Theranostic Macromolecules Research Center, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - V.H. Giang Phan
- Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City 70000, Vietnam
| | - Ji Hoon Jeong
- School of Pharmacy, Theranostic Macromolecules Research Center, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Doo Sung Lee
- School of Chemical Engineering, Theranostic Macromolecules Research Center, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| |
Collapse
|
11
|
Abstract
The phenomenal advances in pharmaceutical sciences over the last few decades have led to the development of new therapeutics like peptides, proteins, RNAs, DNAs and highly potent small molecules. Fruitful applications of these therapeutics have been challenged by several anatomical and physiological barriers that limit adequate drug disposition at the site-of-action and by off-target drug distribution to undesired tissues, which together result in the reduced effectiveness and increased side effects of therapeutic agents. As such, the development of drug delivery and targeting systems has been recognised as a cornerstone for future drug development. Research in pharmaceutical sciences is now devoted to tackling delivery challenges through engineering delivery systems that move beyond conventional dosage forms and regimens into state-of-the-art targeted drug delivery tailored toward specific therapeutic needs. Modern drug delivery systems comprise passive and active targeting approaches. While passive targeting relies on the natural course of distribution of drugs or drug carriers in the body, as governed by their physicochemical properties, active targeting often exploits targeting moieties that home preferentially into target tissues. Here, we provide an overview of theories of and approaches to passive and active drug delivery. As the design of drug delivery is dependent on the unique structure of target tissues and organs, we present our discussion in an organ-specific manner with the aim to inspire the development of new strategies for curing disease with high accuracy and efficiency.
Collapse
Affiliation(s)
- Mohammad Alsaggar
- a Department of Pharmaceutical Technology, College of Pharmacy , Jordon University of Science and Technology , Irbid , Jordan
| | - Dexi Liu
- b Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy , University of Georgia , Athens , GA , USA
| |
Collapse
|
12
|
Ardeleanu R, Dascalu AI, Neamtu A, Peptanariu D, Uritu CM, Maier SS, Nicolescu A, Simionescu BC, Barboiu M, Pinteala M. Multivalent polyrotaxane vectors as adaptive cargo complexes for gene therapy. Polym Chem 2018. [DOI: 10.1039/c7py01256j] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The philosophy to design and construct polyrotaxane carriers, as efficient gene delivery systems.
Collapse
Affiliation(s)
- Rodinel Ardeleanu
- Centre of Advanced Research in Bionanoconjugates and Biopolymers
- “Petru Poni” Institute of Macromolecular Chemistry
- 700487 Iasi
- Romania
| | - Andrei I. Dascalu
- Centre of Advanced Research in Bionanoconjugates and Biopolymers
- “Petru Poni” Institute of Macromolecular Chemistry
- 700487 Iasi
- Romania
| | - Andrei Neamtu
- Centre of Advanced Research in Bionanoconjugates and Biopolymers
- “Petru Poni” Institute of Macromolecular Chemistry
- 700487 Iasi
- Romania
- Regional Institute of Oncology (IRO)
| | - Dragos Peptanariu
- Centre of Advanced Research in Bionanoconjugates and Biopolymers
- “Petru Poni” Institute of Macromolecular Chemistry
- 700487 Iasi
- Romania
| | - Cristina M. Uritu
- Centre of Advanced Research in Bionanoconjugates and Biopolymers
- “Petru Poni” Institute of Macromolecular Chemistry
- 700487 Iasi
- Romania
| | - Stelian S. Maier
- Centre of Advanced Research in Bionanoconjugates and Biopolymers
- “Petru Poni” Institute of Macromolecular Chemistry
- 700487 Iasi
- Romania
- Department of Textile and Leather Chemical Engineering
| | - Alina Nicolescu
- Centre of Advanced Research in Bionanoconjugates and Biopolymers
- “Petru Poni” Institute of Macromolecular Chemistry
- 700487 Iasi
- Romania
| | - Bogdan C. Simionescu
- Centre of Advanced Research in Bionanoconjugates and Biopolymers
- “Petru Poni” Institute of Macromolecular Chemistry
- 700487 Iasi
- Romania
- Department of Natural and Synthetic Polymers
| | - Mihail Barboiu
- Adaptive Supramolecular Nanosystems Group
- Institut
- Européen des Membranes
- ENSCM/UMII/UMR-CNRS 5635
- 34095 Montpellier, Cedex 5
| | - Mariana Pinteala
- Centre of Advanced Research in Bionanoconjugates and Biopolymers
- “Petru Poni” Institute of Macromolecular Chemistry
- 700487 Iasi
- Romania
| |
Collapse
|
13
|
Folic acid-decorated polyamidoamine dendrimer exhibits high tumor uptake and sustained highly localized retention in solid tumors: Its utility for local siRNA delivery. Acta Biomater 2017; 57:251-261. [PMID: 28438704 DOI: 10.1016/j.actbio.2017.04.023] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 04/06/2017] [Accepted: 04/20/2017] [Indexed: 12/13/2022]
Abstract
The utility of folic acid (FA)-decorated polyamidoamine dendrimer G4 (G4-FA) as a vector was investigated for local delivery of siRNA. In a xenograft HN12 (or HN12-YFP) tumor mouse model of head and neck squamous cell carcinomas (HNSCC), intratumorally (i.t.) injected G4-FA exhibited high tumor uptake and sustained highly localized retention in the tumors according to near infrared (NIR) imaging assessment. siRNA against vascular endothelial growth factor A (siVEGFA) was chosen as a therapeutic modality. Compared to the nontherapeutic treatment groups (PBS solution or dendrimer complexed with nontherapeutic siRNA against green fluorescent protein (siGFP)), G4-FA/siVEGFA showed tumor inhibition effects in single-dose and two-dose regimen studies. In particular, two doses of G4-FA/siVEGFA i.t. administered eight days apart resulted in a more profound inhibition of tumor growth, accompanied with significant reduction in angiogenesis, as judged by CD31 staining and microvessel counts. Tumor size reduction in the two-dose regimen study was ascertained semi-quantitatively by live fluorescence imaging of YFP tumors and independently supported antitumor effects of G4-FA/siVEGFA. Taken together, G4-FA shows high tumor uptake and sustained retention properties, making it a suitable platform for local delivery of siRNAs to treat cancers that are readily accessible such as HNSCC. STATEMENT OF SIGNIFICANCE Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer worldwide and is difficult to transfect for gene therapy. We developed folate receptor (FR)-targeted polyamidoamine (PAMAM) dendrimer for enhanced delivery of genes to HNSCC and gained in-depth understanding of how gene delivery and transfection in head and neck squamous cancer cells can be enhanced via FR-targeted PAMAM dendrimers. The results we report here are encouraging and present latest advances in using dendrimers for cancer therapies, in particular for HNSCC. Our work has demonstrated that localized delivery of FR-targeted PAMAM dendrimer G4 complexed with siVEGFA resulted in pronounced tumor suppression in an HN12 xenograft tumor model. Tumor suppression was attributed to enhanced tumor uptake of siRNA and prolonged nanoparticle retention in the tumor. Taken together, G4-FA shows high tumor uptake and sustained highly localized retention properties, making it a suitable platform for local delivery of siRNAs to treat cancers that are readily accessible such as HNSCC.
Collapse
|
14
|
Mathew AP, Cho KH, Uthaman S, Cho CS, Park IK. Stimuli-Regulated Smart Polymeric Systems for Gene Therapy. Polymers (Basel) 2017; 9:E152. [PMID: 30970831 PMCID: PMC6432211 DOI: 10.3390/polym9040152] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 04/19/2017] [Accepted: 04/20/2017] [Indexed: 01/02/2023] Open
Abstract
The physiological condition of the human body is a composite of different environments, each with its own parameters that may differ under normal, as well as diseased conditions. These environmental conditions include factors, such as pH, temperature and enzymes that are specific to a type of cell, tissue or organ or a pathological state, such as inflammation, cancer or infection. These conditions can act as specific triggers or stimuli for the efficient release of therapeutics at their destination by overcoming many physiological and biological barriers. The efficacy of conventional treatment modalities can be enhanced, side effects decreased and patient compliance improved by using stimuli-responsive material that respond to these triggers at the target site. These stimuli or triggers can be physical, chemical or biological and can be internal or external in nature. Many smart/intelligent stimuli-responsive therapeutic gene carriers have been developed that can respond to either internal stimuli, which may be normally present, overexpressed or present in decreased levels, owing to a disease, or to stimuli that are applied externally, such as magnetic fields. This review focuses on the effects of various internal stimuli, such as temperature, pH, redox potential, enzymes, osmotic activity and other biomolecules that are present in the body, on modulating gene expression by using stimuli-regulated smart polymeric carriers.
Collapse
Affiliation(s)
- Ansuja Pulickal Mathew
- Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju 61469, Korea.
| | - Ki-Hyun Cho
- Department of Plastic Surgery, Institute of Dermatology and Plastic Surgery, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, USA.
| | - Saji Uthaman
- Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju 61469, Korea.
| | - Chong-Su Cho
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea.
| | - In-Kyu Park
- Department of Biomedical Sciences, BK21 PLUS Center for Creative Biomedical Scientists at Chonnam National University, Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 61469, Korea.
| |
Collapse
|
15
|
Nguyen MK, McMillan A, Huynh CT, Schapira DS, Alsberg E. Photocrosslinkable, biodegradable hydrogels with controlled cell adhesivity for prolonged siRNA delivery to hMSCs to enhance their osteogenic differentiation. J Mater Chem B 2017; 5:485-495. [PMID: 28652917 PMCID: PMC5482539 DOI: 10.1039/c6tb01739h] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Photocrosslinked, biodegradable hydrogels have been extensively investigated for biomedical applications, including drug delivery and tissue engineering. Here, dextran (DEX) was chemically modified with mono(2-acryloyloxyethyl) succinate (MAES) via an esterification reaction, resulting in macromers that could be photocrosslinked to form hydrolytically degradable hydrogels. Hydrogel swelling ratio and degradation rate were controlled by varying the degree of MAES modification. Thiolated cell adhesion peptides (GRGDSPC) were conjugated to acrylated dextran via thiol-acrylate reaction to regulate the interactions of human mesenchymal stem cells (hMSCs) with the photocrosslinkable hydrogels. The hydrogels permitted sustained release of short interfering RNA (siRNA) over 7 weeks and were cytocompatible with hMSCs. Sustained presentation of siRNA from these photocrosslinked DEX hydrogels enhanced the osteogenic differentiation of encapsulated hMSCs. These DEX hydrogels with tunable siRNA delivery and cell adhesive properties may provide an excellent platform for bioactive molecule delivery and tissue regeneration applications.
Collapse
Affiliation(s)
- Minh Khanh Nguyen
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, Ohio 44106
| | - Alexandra McMillan
- Department of Pathology, Case Western Reserve University, 10900 Euclid Ave., Cleveland, Ohio 44106
| | - Cong Truc Huynh
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, Ohio 44106
| | - Daniel S Schapira
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, Ohio 44106
| | - Eben Alsberg
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, Ohio 44106
- Department of Orthopaedic Surgery, Case Western Reserve University, 10900 Euclid Ave., Cleveland, Ohio 44106
- National Center for Regenerative Medicine, Division of General Medical Sciences, Case Western Reserve University, 10900 Euclid Ave., Cleveland, Ohio 44106
| |
Collapse
|
16
|
Yang R, Nam K, Kim SW, Turkson J, Zou Y, Zuo YY, Haware RV, Chougule MB. Factorial Design Based Multivariate Modeling and Optimization of Tunable Bioresponsive Arginine Grafted Poly(cystaminebis(acrylamide)-diaminohexane) Polymeric Matrix Based Nanocarriers. Mol Pharm 2016; 14:252-263. [DOI: 10.1021/acs.molpharmaceut.6b00861] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Rongbing Yang
- Translational Drug
and Gene Delivery Research (TransDGDR) Laboratory, Department of Pharmaceutical
Sciences, Department of Pharmaceutics and Drug Delivery, Research
of Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, Mississippi 38677, United States
- Translational
Drug Delivery Research (TransDDR) Laboratory, Department of Pharmaceutical
Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawai’i at Hilo, Hilo, Hawaii 96720, United States
| | - Kihoon Nam
- Center for Controlled Chemical Delivery
(CCCD), Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, Utah 84112, United States
- School
of Dentistry, University of Utah, Salt Lake City, Utah 84112, United States
| | - Sung Wan Kim
- Center for Controlled Chemical Delivery
(CCCD), Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, Utah 84112, United States
| | - James Turkson
- Natural Products and Experimental Therapeutics Program, University of Hawaii Cancer Center, Honolulu, Hawaii 96813, United States
| | - Ye Zou
- Department
of Mechanical Engineering, University of Hawaii at Manoa, Honolulu, Hawaii 96822, United States
| | - Yi Y. Zuo
- Department
of Mechanical Engineering, University of Hawaii at Manoa, Honolulu, Hawaii 96822, United States
| | - Rahul V. Haware
- College of Pharmacy & Health Sciences, Campbell University, Buies Creek, North Carolina 27506, United States
| | - Mahavir B. Chougule
- Translational Drug
and Gene Delivery Research (TransDGDR) Laboratory, Department of Pharmaceutical
Sciences, Department of Pharmaceutics and Drug Delivery, Research
of Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, Mississippi 38677, United States
- Translational
Drug Delivery Research (TransDDR) Laboratory, Department of Pharmaceutical
Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawai’i at Hilo, Hilo, Hawaii 96720, United States
- Natural Products and Experimental Therapeutics Program, University of Hawaii Cancer Center, Honolulu, Hawaii 96813, United States
- Pii Center
for Pharmaceutical Technology, Research Institute of Pharmaceutical
Sciences, University of Mississippi, University, Mississippi 38677, United States
- National Center for Natural Products Research, Research Institute
of Pharmaceutical Sciences, University of Mississippi, University, Mississippi 38677, United States
| |
Collapse
|
17
|
Caffrey LM, deRonde BM, Minter LM, Tew GN. Mapping Optimal Charge Density and Length of ROMP-Based PTDMs for siRNA Internalization. Biomacromolecules 2016; 17:3205-3212. [PMID: 27599388 PMCID: PMC5094354 DOI: 10.1021/acs.biomac.6b00900] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
A fundamental understanding of how polymer structure impacts internalization and delivery of biologically relevant cargoes, particularly small interfering ribonucleic acid (siRNA), is of critical importance to the successful design of improved delivery reagents. Herein we report the use of ring-opening metathesis polymerization (ROMP) methods to synthesize two series of guanidinium-rich protein transduction domain mimics (PTDMs): one based on an imide scaffold that contains one guanidinium moiety per repeat unit, and another based on a diester scaffold that contains two guanidinium moieties per repeat unit. By varying both the degree of polymerization and, in effect, the relative number of cationic charges in each PTDM, the performances of the two ROMP backbones for siRNA internalization were evaluated and compared. Internalization of fluorescently labeled siRNA into Jurkat T cells demonstrated that fluorescein isothiocyanate (FITC)-siRNA internalization had a charge content dependence, with PTDMs containing approximately 40 to 60 cationic charges facilitating the most internalization. Despite this charge content dependence, the imide scaffold yielded much lower viabilities in Jurkat T cells than the corresponding diester PTDMs with similar numbers of cationic charges, suggesting that the diester scaffold is preferred for siRNA internalization and delivery applications. These developments will not only improve our understanding of the structural factors necessary for optimal siRNA internalization, but will also guide the future development of optimized PTDMs for siRNA internalization and delivery.
Collapse
Affiliation(s)
- Leah M Caffrey
- Department of Polymer Science and Engineering, ‡Department of Veterinary and Animal Sciences, and §Molecular and Cellular Biology Program, University of Massachusetts Amherst , Amherst, Massachusetts 01003, United States
| | - Brittany M deRonde
- Department of Polymer Science and Engineering, ‡Department of Veterinary and Animal Sciences, and §Molecular and Cellular Biology Program, University of Massachusetts Amherst , Amherst, Massachusetts 01003, United States
| | - Lisa M Minter
- Department of Polymer Science and Engineering, ‡Department of Veterinary and Animal Sciences, and §Molecular and Cellular Biology Program, University of Massachusetts Amherst , Amherst, Massachusetts 01003, United States
| | - Gregory N Tew
- Department of Polymer Science and Engineering, ‡Department of Veterinary and Animal Sciences, and §Molecular and Cellular Biology Program, University of Massachusetts Amherst , Amherst, Massachusetts 01003, United States
| |
Collapse
|
18
|
Sarisozen C, Salzano G, Torchilin VP. Recent advances in siRNA delivery. Biomol Concepts 2016; 6:321-41. [PMID: 26609865 DOI: 10.1515/bmc-2015-0019] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 11/04/2015] [Indexed: 01/05/2023] Open
Abstract
In the 1990s an unexpected gene-silencing phenomena in plants, the later called RNA interference (RNAi), perplexed scientists. Following the proof of activity in mammalian cells, small interfering RNAs (siRNAs) have quickly crept into biomedical research as a new powerful tool for the potential treatment of different human diseases based on altered gene expression. In the past decades, several promising data from ongoing clinical trials have been reported. However, despite surprising successes in many pre-clinical studies, concrete obstacles still need to be overcome to translate therapeutic siRNAs into clinical reality. Here, we provide an update on the recent advances of RNAi-based therapeutics and highlight novel synthetic platforms for the intracellular delivery of siRNAs.
Collapse
|
19
|
deRonde BM, Posey ND, Otter R, Minter LM, Tew GN. Optimal Hydrophobicity in Ring-Opening Metathesis Polymerization-Based Protein Mimics Required for siRNA Internalization. Biomacromolecules 2016; 17:1969-77. [PMID: 27103189 PMCID: PMC4964964 DOI: 10.1021/acs.biomac.6b00138] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Exploring the role of polymer structure for the internalization of biologically relevant cargo, specifically siRNA, is of critical importance to the development of improved delivery reagents. Herein, we report guanidinium-rich protein transduction domain mimics (PTDMs) based on a ring-opening metathesis polymerization scaffold containing tunable hydrophobic moieties that promote siRNA internalization. Structure-activity relationships using Jurkat T cells and HeLa cells were explored to determine how the length of the hydrophobic block and the hydrophobic side chain compositions of these PTDMs impacted siRNA internalization. To explore the hydrophobic block length, two different series of diblock copolymers were synthesized: one series with symmetric block lengths and one with asymmetric block lengths. At similar cationic block lengths, asymmetric and symmetric PTDMs promoted siRNA internalization in the same percentages of the cell population regardless of the hydrophobic block length; however, with 20 repeat units of cationic charge, the asymmetric block length had greater siRNA internalization, highlighting the nontrivial relationships between hydrophobicity and overall cationic charge. To further probe how the hydrophobic side chains impacted siRNA internalization, an additional series of asymmetric PTDMs was synthesized that featured a fixed hydrophobic block length of five repeat units that contained either dimethyl (dMe), methyl phenyl (MePh), or diphenyl (dPh) side chains and varied cationic block lengths. This series was further expanded to incorporate hydrophobic blocks consisting of diethyl (dEt), diisobutyl (diBu), and dicyclohexyl (dCy) based repeat units to better define the hydrophobic window for which our PTDMs had optimal activity. High-performance liquid chromatography retention times quantified the relative hydrophobicities of the noncationic building blocks. PTDMs containing the MePh, diBu, and dPh hydrophobic blocks were shown to have superior siRNA internalization capabilities compared to their more and less hydrophobic counterparts, demonstrating a critical window of relative hydrophobicity for optimal internalization. This better understanding of how hydrophobicity impacts PTDM-induced internalization efficiencies will help guide the development of future delivery reagents.
Collapse
Affiliation(s)
- Brittany M. deRonde
- Department of Polymer Science and Engineering, University of Massachusetts Amherst, Amherst, MA 01003
| | - Nicholas D. Posey
- Department of Polymer Science and Engineering, University of Massachusetts Amherst, Amherst, MA 01003
| | - Ronja Otter
- Department of Polymer Science and Engineering, University of Massachusetts Amherst, Amherst, MA 01003
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, MA 01003
| | - Lisa M. Minter
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA 01003
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, MA 01003
| | - Gregory N. Tew
- Department of Polymer Science and Engineering, University of Massachusetts Amherst, Amherst, MA 01003
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA 01003
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, MA 01003
| |
Collapse
|
20
|
Yoo J, Lee D, Gujrati V, Rejinold NS, Lekshmi KM, Uthaman S, Jeong C, Park IK, Jon S, Kim YC. Bioreducible branched poly(modified nona-arginine) cell-penetrating peptide as a novel gene delivery platform. J Control Release 2016; 246:142-154. [PMID: 27170226 DOI: 10.1016/j.jconrel.2016.04.040] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 04/23/2016] [Accepted: 04/27/2016] [Indexed: 11/28/2022]
Abstract
Cell-penetrating peptides (CPPs) have been widely used to deliver nucleic acid molecules. Generally, CPPs consisting of short amino acid sequences have a linear structure, resulting in a weak complexation and low transfection efficacy. To overcome these drawbacks, a novel type of CPP is required to enhance the delivery efficacy while maintaining its safe use at the same time. Herein, we report that a bioreducible branched poly-CPP structure capable of responding to reducing conditions attained both outstanding delivery effectiveness and selective gene release in carcinoma cells. Branched structures provide unusually strong electrostatic attraction between DNA and siRNA molecules, thereby improving the transfection capability through a tightly condensed form. We designed a modified type of nona-arginine (mR9) and synthesized a branched-mR9 (B-mR9) using disulfide bonds. A novel B-mR9/pDNA polyplex exhibited redox-cleavability and high transfection efficacy compared to conventional CPPs, with higher cell viability as well. B-mR9/VEGF siRNA polyplex exhibited significant serum stability and high gene-silencing effects in vitro. Furthermore, the B-mR9 polyplex showed outstanding tumor accumulation and inhibition ability in vivo. The results suggest that the bioreducible branched poly CPP has great potential as a gene delivery platform.
Collapse
Affiliation(s)
- Jisang Yoo
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - DaeYong Lee
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Vipul Gujrati
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - N Sanoj Rejinold
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Kamali Manickavasagam Lekshmi
- Department of Biomedical Science and BK21 PLUS Centre for Creative Biomedical Scientists, Chonnam National University Medical School, 160 Baekseo-ro, Gwangju 501-746, Republic of Korea
| | - Saji Uthaman
- Department of Biomedical Science and BK21 PLUS Centre for Creative Biomedical Scientists, Chonnam National University Medical School, 160 Baekseo-ro, Gwangju 501-746, Republic of Korea
| | - Chanuk Jeong
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - In-Kyu Park
- Department of Biomedical Science and BK21 PLUS Centre for Creative Biomedical Scientists, Chonnam National University Medical School, 160 Baekseo-ro, Gwangju 501-746, Republic of Korea
| | - Sangyong Jon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Yeu-Chun Kim
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea.
| |
Collapse
|
21
|
Kamaly N, Yameen B, Wu J, Farokhzad OC. Degradable Controlled-Release Polymers and Polymeric Nanoparticles: Mechanisms of Controlling Drug Release. Chem Rev 2016; 116:2602-63. [PMID: 26854975 PMCID: PMC5509216 DOI: 10.1021/acs.chemrev.5b00346] [Citation(s) in RCA: 1600] [Impact Index Per Article: 200.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Nazila Kamaly
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Basit Yameen
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Jun Wu
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Omid C. Farokhzad
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
22
|
deRonde BM, Torres JA, Minter LM, Tew GN. Development of Guanidinium-Rich Protein Mimics for Efficient siRNA Delivery into Human T Cells. Biomacromolecules 2015; 16:3172-9. [PMID: 26324222 DOI: 10.1021/acs.biomac.5b00795] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
RNA interference is gaining attention as a means to explore new molecular pathways and for its potential as a therapeutic; however, its application in immortal and primary T cells is limited due to challenges with efficient delivery in these cell types. Herein, we report the development of guanidinium-rich protein transduction domain mimics (PTDMs) based on a ring-opening metathesis polymerization scaffold that delivers siRNA into Jurkat T cells and human peripheral blood mononuclear cells (hPBMCs). Homopolymer and block copolymer PTDMs with varying numbers of guanidinium moieties were designed and tested to assess the effect cationic charge content and the addition of a segregated, hydrophobic block had on siRNA internalization and delivery. Internalization of fluorescently labeled siRNA into Jurkat T cells illustrates that the optimal cationic charge content, 40 charges per polymer, leads to higher efficiencies, with block copolymers outperforming their homopolymer counterparts. PTDMs also outperformed commercial reagents commonly used for siRNA delivery applications. Select PTDM candidates were further screened to assess the role the PTDM structure has on the delivery of biologically active siRNA into primary cells. Specifically, siRNA to hNOTCH1 was delivered to hPBMCs enabling 50-80% knockdown efficiencies, with longer PTDMs showing improved protein reduction. By evaluating the PTDM design parameters for siRNA delivery, more efficient PTDMs were discovered that improved delivery and gene (NOTCH) knockdown in T cells. Given the robust delivery of siRNA by these novel PTDMs, their development should aid in the exploration of T cell molecular pathways leading eventually to new therapeutics.
Collapse
Affiliation(s)
- Brittany M deRonde
- Department of Polymer Science and Engineering, University of Massachusetts Amherst , Amherst, Massachusetts 01003, United States
| | - Joe A Torres
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst , Amherst, Massachusetts 01003, United States.,Molecular and Cellular Biology Program, University of Massachusetts Amherst , Amherst, Massachusetts 01003, United States
| | - Lisa M Minter
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst , Amherst, Massachusetts 01003, United States.,Molecular and Cellular Biology Program, University of Massachusetts Amherst , Amherst, Massachusetts 01003, United States
| | - Gregory N Tew
- Department of Polymer Science and Engineering, University of Massachusetts Amherst , Amherst, Massachusetts 01003, United States.,Department of Veterinary and Animal Sciences, University of Massachusetts Amherst , Amherst, Massachusetts 01003, United States.,Molecular and Cellular Biology Program, University of Massachusetts Amherst , Amherst, Massachusetts 01003, United States
| |
Collapse
|
23
|
Lee GJ, Ryu K, Kim K, Choi JY, Kim TI. Crosslinked Polypropylenimine Dendrimers With Bioreducible Linkages for Gene Delivery Systems and Their Reductive Degradation Behaviors. Macromol Biosci 2015; 15:1595-604. [DOI: 10.1002/mabi.201500141] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 06/03/2015] [Indexed: 12/23/2022]
Affiliation(s)
- Gyeong Jin Lee
- Department of Biosystems & Biomaterials Science and Engineering, College of Agriculture and Life Sciences; Seoul National University; 1 Gwanak-ro Gwanak-gu Seoul 151-921 Republic of Korea
| | - Kitae Ryu
- Department of Biosystems & Biomaterials Science and Engineering, College of Agriculture and Life Sciences; Seoul National University; 1 Gwanak-ro Gwanak-gu Seoul 151-921 Republic of Korea
| | - Kyunghwan Kim
- Department of Biosystems & Biomaterials Science and Engineering, College of Agriculture and Life Sciences; Seoul National University; 1 Gwanak-ro Gwanak-gu Seoul 151-921 Republic of Korea
| | - Ji-yeong Choi
- Department of Biosystems & Biomaterials Science and Engineering, College of Agriculture and Life Sciences; Seoul National University; 1 Gwanak-ro Gwanak-gu Seoul 151-921 Republic of Korea
| | - Tae-il Kim
- Department of Biosystems & Biomaterials Science and Engineering, College of Agriculture and Life Sciences; Seoul National University; 1 Gwanak-ro Gwanak-gu Seoul 151-921 Republic of Korea
- Research Institute of Agriculture and Life Sciences; Seoul National University; 1 Gwanak-ro Gwanak-gu Seoul 151-921 Republic of Korea
| |
Collapse
|
24
|
deRonde BM, Tew GN. Development of protein mimics for intracellular delivery. Biopolymers 2015; 104:265-80. [PMID: 25858701 PMCID: PMC4516575 DOI: 10.1002/bip.22658] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 03/24/2015] [Accepted: 03/30/2015] [Indexed: 12/19/2022]
Abstract
Designing delivery agents for therapeutics is an ongoing challenge. As treatments and desired cargoes become more complex, the need for improved delivery vehicles becomes critical. Excellent delivery vehicles must ensure the stability of the cargo, maintain the cargo's solubility, and promote efficient delivery and release. In order to address these issues, many research groups have looked to nature for design inspiration. Proteins, such as HIV-1 trans-activator of transcription (TAT) and Antennapedia homeodomain protein, are capable of crossing cellular membranes. However, due to the complexities of their structures, they are synthetically challenging to reproduce in the laboratory setting. Being able to incorporate the key features of these proteins that enable cell entry into simpler scaffolds opens up a wide range of opportunities for the development of new delivery reagents with improved performance. This review charts the development of protein mimics based on cell-penetrating peptides (CPPs) and how structure-activity relationships (SARs) with these molecules and their protein counterparts ultimately led to the use of polymeric scaffolds. These scaffolds deviate from the normal peptide backbone, allowing for simpler, synthetic procedures to make carriers and tune chemical compositions for application specific needs. Successful design of polymeric protein mimics would allow researchers to further understand the key features in proteins and peptides necessary for efficient delivery and to design the next generation of more efficient delivery reagents.
Collapse
Affiliation(s)
- Brittany M deRonde
- Department of Polymer Science and Engineering, University of Massachusetts Amherst, Amherst, MA, 01003
| | - Gregory N Tew
- Department of Polymer Science and Engineering, University of Massachusetts Amherst, Amherst, MA, 01003
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, 01003
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, MA, 01003
| |
Collapse
|
25
|
Beloor J, Ramakrishna S, Nam K, Seon Choi C, Kim J, Kim SH, Cho HJ, Shin H, Kim H, Kim SW, Lee SK, Kumar P. Effective gene delivery into human stem cells with a cell-targeting Peptide-modified bioreducible polymer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2015; 11:2069-2079. [PMID: 25515928 DOI: 10.1002/smll.201402933] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Indexed: 06/04/2023]
Abstract
Stem cells are poorly permissive to non-viral gene transfection reagents. In this study, we explored the possibility of improving gene delivery into human embryonic (hESC) and mesenchymal (hMSC) stem cells by synergizing the activity of a cell-binding ligand with a polymer that releases nucleic acids in a cytoplasm-responsive manner. A 29 amino acid long peptide, RVG, targeting the nicotinic acetylcholine receptor (nAchR) was identified to bind both hMSC and H9-derived hESC. Conjugating RVG to a redox-sensitive biodegradable dendrimer-type arginine-grafted polymer (PAM-ABP) enabled nanoparticle formation with plasmid DNA without altering the environment-sensitive DNA release property and favorable toxicity profile of the parent polymer. Importantly, RVG-PAM-ABP quantitatively enhanced transfection into both hMSC and hESC compared to commercial transfection reagents like Lipofectamine 2000 and Fugene. ∼60% and 50% of hMSC and hESC were respectively transfected, and at increased levels on a per cell basis, without affecting pluripotency marker expression. RVG-PAM-ABP is thus a novel bioreducible, biocompatible, non-toxic, synthetic gene delivery system for nAchR-expressing stem cells. Our data also demonstrates that a cell-binding ligand like RVG can cooperate with a gene delivery system like PAM-ABP to enable transfection of poorly-permissive cells.
Collapse
Affiliation(s)
- Jagadish Beloor
- Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT, 06520, USA; Department of Bioengineering and Institute of Nano Science and Technology, Hanyang University, Seoul, 133-791, Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Foster AA, Greco CT, Green MD, Epps TH, Sullivan MO. Light-mediated activation of siRNA Release in diblock copolymer assemblies for controlled gene silencing. Adv Healthc Mater 2015; 4:760-70. [PMID: 25530259 PMCID: PMC4429132 DOI: 10.1002/adhm.201400671] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 11/24/2014] [Indexed: 11/08/2022]
Abstract
Controllable release is particularly important for the delivery of small interfering RNA (siRNA), as siRNAs have a high susceptibility to enzymatic degradation if release is premature, yet lack silencing activity if they remain inaccessible within the cytoplasm. To overcome these hurdles, novel and tailorable mPEG-b-poly(5-(3-(amino)propoxy)-2-nitrobenzyl methacrylate) (mPEG-b-P(APNBMA)) diblock copolymers containing light-sensitive o-nitrobenzyl moieties and pendant amines are employed to provide both efficient siRNA binding, via electrostatic and hydrophobic interactions, as well as triggered charge reversal and nucleic acid release. In particular, siRNA/mPEG-b-P(APNBMA)23.6 polyplexes show minimal aggregation in physiological salt and serum, and enhanced resistance to polyanion-induced unpackaging compared to polyethylenimine preparations. Cellular delivery of siRNA/mPEG-b-P(APNBMA)23.6 polyplexes reveals greater than 80% cellular transfection, as well as rapid and widespread cytoplasmic distribution. Additionally, UV irradiation indicates ≈70% reduction in targeted gene expression following siRNA/mPEG-b-P(APNBMA)23.6 polyplex treatment, as compared to 0% reduction in polyplex-treated cells without UV irradiation, and only ≈30% reduction for Lipofectamine-treated cells. The results here highlight the potential of these light-sensitive copolymers with a well-defined on/off switch for applications including cellular patterning for guided cell growth and extension, and cellular microarrays for exploring protein and drug interactions that require enhanced spatiotemporal control of gene activation.
Collapse
Affiliation(s)
- Abbygail A. Foster
- Department of Chemical and Biomolecular Engineering, Newark, DE 19716, USA
| | - Chad T. Greco
- Department of Chemical and Biomolecular Engineering, Newark, DE 19716, USA
| | - Matthew D. Green
- Department of Chemical and Biomolecular Engineering, Newark, DE 19716, USA
| | - Thomas H. Epps
- Department of Chemical and Biomolecular Engineering, Newark, DE 19716, USA
| | | |
Collapse
|
27
|
Targeted siRNA therapy using cytoplasm-responsive nanocarriers and cell-penetrating peptides. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2014. [DOI: 10.1007/s40005-014-0155-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
28
|
Islam MA, Park T, Singh B, Maharjan S, Firdous J, Cho MH, Kang SK, Yun CH, Choi Y, Cho CS. Major degradable polycations as carriers for DNA and siRNA. J Control Release 2014; 193:74-89. [DOI: 10.1016/j.jconrel.2014.05.055] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 05/24/2014] [Accepted: 05/27/2014] [Indexed: 12/17/2022]
|
29
|
Multifunctional, stimuli-sensitive nanoparticulate systems for drug delivery. Nat Rev Drug Discov 2014; 13:813-27. [PMID: 25287120 DOI: 10.1038/nrd4333] [Citation(s) in RCA: 1009] [Impact Index Per Article: 100.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The use of nanoparticulate pharmaceutical drug delivery systems (NDDSs) to enhance the in vivo effectiveness of drugs is now well established. The development of multifunctional and stimulus-sensitive NDDSs is an active area of current research. Such NDDSs can have long circulation times, target the site of the disease and enhance the intracellular delivery of a drug. This type of NDDS can also respond to local stimuli that are characteristic of the pathological site by, for example, releasing an entrapped drug or shedding a protective coating, thus facilitating the interaction between drug-loaded nanocarriers and target cells or tissues. In addition, imaging contrast moieties can be attached to these carriers to track their real-time biodistribution and accumulation in target cells or tissues. Here, I highlight recent developments with multifunctional and stimuli-sensitive NDDSs and their therapeutic potential for diseases including cancer, cardiovascular diseases and infectious diseases.
Collapse
|
30
|
Abstract
The use of nanoparticulate pharmaceutical drug delivery systems (NDDSs) to enhance the in vivo effectiveness of drugs is now well established. The development of multifunctional and stimulus-sensitive NDDSs is an active area of current research. Such NDDSs can have long circulation times, target the site of the disease and enhance the intracellular delivery of a drug. This type of NDDS can also respond to local stimuli that are characteristic of the pathological site by, for example, releasing an entrapped drug or shedding a protective coating, thus facilitating the interaction between drug-loaded nanocarriers and target cells or tissues. In addition, imaging contrast moieties can be attached to these carriers to track their real-time biodistribution and accumulation in target cells or tissues. Here, I highlight recent developments with multifunctional and stimuli-sensitive NDDSs and their therapeutic potential for diseases including cancer, cardiovascular diseases and infectious diseases.
Collapse
|
31
|
Oupický D, Li J. Bioreducible polycations in nucleic acid delivery: past, present, and future trends. Macromol Biosci 2014; 14:908-22. [PMID: 24678057 PMCID: PMC4410047 DOI: 10.1002/mabi.201400061] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 02/19/2014] [Indexed: 12/16/2022]
Abstract
Polycations that are degradable by reduction of disulfide bonds are developed for applications in delivery of nucleic acids. This Feature Article surveys methods of synthesis of bioreducible polycations and discusses current understanding of the mechanism of action of bioreducible polyplexes. Emphasis is placed on the relationship between the biological redox environment and toxicity, trafficking, transfection activity, and in vivo behavior of bioreducible polycations and polyplexes.
Collapse
Affiliation(s)
- David Oupický
- Department of Pharmaceutical Sciences, Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Durham Research Center, 985830 Nebraska Medical Center, Omaha, NE 68198-5830, USA.
| | | |
Collapse
|
32
|
Draz MS, Fang BA, Zhang P, Hu Z, Gu S, Weng KC, Gray JW, Chen FF. Nanoparticle-mediated systemic delivery of siRNA for treatment of cancers and viral infections. Am J Cancer Res 2014; 4:872-92. [PMID: 25057313 PMCID: PMC4107289 DOI: 10.7150/thno.9404] [Citation(s) in RCA: 150] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 05/27/2014] [Indexed: 12/17/2022] Open
Abstract
RNA interference (RNAi) is an endogenous post-transcriptional gene regulatory mechanism, where non-coding, double-stranded RNA molecules interfere with the expression of certain genes in order to silence it. Since its discovery, this phenomenon has evolved as powerful technology to diagnose and treat diseases at cellular and molecular levels. With a lot of attention, short interfering RNA (siRNA) therapeutics has brought a great hope for treatment of various undruggable diseases, including genetic diseases, cancer, and resistant viral infections. However, the challenge of their systemic delivery and on how they are integrated to exhibit the desired properties and functions remains a key bottleneck for realizing its full potential. Nanoparticles are currently well known to exhibit a number of unique properties that could be strategically tailored into new advanced siRNA delivery systems. This review summarizes the various nanoparticulate systems developed so far in the literature for systemic delivery of siRNA, which include silica and silicon-based nanoparticles, metal and metal oxides nanoparticles, carbon nanotubes, graphene, dendrimers, polymers, cyclodextrins, lipids, hydrogels, and semiconductor nanocrystals. Challenges and barriers to the delivery of siRNA and the role of different nanoparticles to surmount these challenges are also included in the review.
Collapse
|
33
|
Lee YS, Kim SW. Bioreducible polymers for therapeutic gene delivery. J Control Release 2014; 190:424-39. [PMID: 24746626 DOI: 10.1016/j.jconrel.2014.04.012] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 04/09/2014] [Accepted: 04/09/2014] [Indexed: 01/18/2023]
Abstract
Most currently available cationic polymers have significant acute toxicity concerns such as cellular toxicity, aggregation of erythrocytes, and entrapment in the lung capillary bed, largely due to their poor biocompatibility and non-degradability under physiological conditions. To develop more intelligent polymers, disulfide bonds are introduced in the design of biodegradable polymers. Herein, the sustained innovations of biomimetic nano-sized constructs with bioreducible poly(disulfide amine)s demonstrate a viable clinical tool for the treatment of cardiovascular disease, anemia, diabetes, and cancer.
Collapse
Affiliation(s)
- Young Sook Lee
- Center for Controlled Chemical Delivery, Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, USA.
| | - Sung Wan Kim
- Center for Controlled Chemical Delivery, Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, USA; Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Republic of Korea.
| |
Collapse
|
34
|
Li J, Oupický D. Effect of biodegradability on CXCR4 antagonism, transfection efficacy and antimetastatic activity of polymeric Plerixafor. Biomaterials 2014; 35:5572-9. [PMID: 24726746 DOI: 10.1016/j.biomaterials.2014.03.047] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 03/18/2014] [Indexed: 02/02/2023]
Abstract
Chemokine receptor CXCR4 and its sole ligand SDF-1 are key players in regulating cancer cell invasion and metastasis. Plerixafor (AMD3100) is a small-molecule CXCR4 antagonist that prevents binding of SDF-1 to CXCR4 and has potential in prevention of cancer metastasis. This study investigates the influence of biodegradability of a recently reported polymeric Plerixafor (PAMD) on CXCR4 antagonism, antimetastatic activity, and transfection efficacy of PAMD polyplexes with plasmid DNA. We show that PAMD exhibits CXCR4 antagonism and inhibition of cancer cell invasion in vitro regardless of its biodegradability. Biodegradable PAMD showed considerably enhanced transfection efficiency and decreased cytotoxicity when compared with the non-degradable PAMD. Despite similar CXCR4 antagonism in vitro, only biodegradable PAMD displayed antimetastatic activity in experimental lung metastasis model in vivo.
Collapse
Affiliation(s)
- Jing Li
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA; Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI 48202, USA
| | - David Oupický
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA; Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI 48202, USA.
| |
Collapse
|
35
|
Florinas S, Kim J, Nam K, Janát-Amsbury MM, Kim SW. Ultrasound-assisted siRNA delivery via arginine-grafted bioreducible polymer and microbubbles targeting VEGF for ovarian cancer treatment. J Control Release 2014; 183:1-8. [PMID: 24657947 DOI: 10.1016/j.jconrel.2014.03.025] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Revised: 03/03/2014] [Accepted: 03/14/2014] [Indexed: 12/17/2022]
Abstract
The major drawback hampering siRNA therapies from being more widely accepted in clinical practice is its insufficient accumulation at the target site mainly due to poor cellular uptake and rapid degradation in serum. Therefore, we designed a novel polymeric siRNA carrier system, which would withstand serum-containing environments and tested its performance in vitro as well as in vivo. Delivering siRNA with a system combining an arginine-grafted bioreducible polymer (ABP), microbubbles (MBs), and ultrasound technology (US) we were able to synergize the advantages each delivery system owns individually, and created our innovative siRNA-ABP-MB (SAM) complexes. SAM complexes show significantly higher siRNA uptake and VEGF protein knockdown in vitro with serum-containing media when compared to naked siRNA, and 25k-branched-polyethylenimine (bPEI) representing the current standard in nonviral gene therapy. SAM complexes activated by US are also able to improve siRNA uptake in tumor tissue resulting in decelerating tumor growth in vivo.
Collapse
Affiliation(s)
- Stelios Florinas
- Center for Controlled Chemical Delivery (CCCD), Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, USA
| | - Jaesung Kim
- Center for Controlled Chemical Delivery (CCCD), Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, USA
| | - Kihoon Nam
- Center for Controlled Chemical Delivery (CCCD), Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, USA
| | - Margit M Janát-Amsbury
- Center for Controlled Chemical Delivery (CCCD), Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, USA; Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University of Utah, Salt Lake City 84132, USA
| | - Sung Wan Kim
- Center for Controlled Chemical Delivery (CCCD), Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, USA.
| |
Collapse
|
36
|
Beloor J, Nam HY, Lee SK, Kumar P. Arginine-grafted biodegradable polymer: a versatile transfection reagent for both DNA and siRNA. Methods Mol Biol 2014; 1176:115-26. [PMID: 25030923 DOI: 10.1007/978-1-4939-0992-6_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Effective delivery of DNA or siRNA into primary cells demands an efficient delivery system. However, the significant differences in physical and molecular characteristics of the two molecules generally necessitate distinct delivery systems or considerable differences in carrier formulation protocols for effective transfection. Arginine-grafted bioreducible poly (disulfide amine) (ABP) is a redox-sensitive, bioreducible, positively charged polymer which complexes with siRNA and DNA via charge interactions to form nanoplexes. ABP effectively mediates cytoplasmic delivery of both DNA and siRNA into multiple cell types, including primary cells like myoblast, human umbilical vein endothelial cells (HUVECs), and primary rat aorta vascular smooth muscle cells (SMCs) eliciting functional activity. In this chapter, we provide the detailed protocols for the synthesis of ABP as well as transfection of both siRNA and DNA using ABP.
Collapse
Affiliation(s)
- Jagadish Beloor
- Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, 25 York Street, New Haven, CT, 06520, USA
| | | | | | | |
Collapse
|
37
|
Wang JG, Zheng XX, Zeng GY, Zhou YJ, Yuan H. Purified vitexin compound 1 induces apoptosis through activation of FOXO3a in hepatocellular carcinoma. Oncol Rep 2013; 31:488-96. [PMID: 24247909 DOI: 10.3892/or.2013.2855] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 10/29/2013] [Indexed: 11/06/2022] Open
Abstract
We previously reported that purified vitexin compound 1 (VB1, a neolignan from the seed of Chinese herb Vitex negundo) exhibited antitumor activity in cancer cell lines and xenograft models. In the present study, we examined the molecular mechanisms by which activation of the FOXO3a transcription factor mediated VB1-induced apoptosis in hepatocellular carcinoma (HCC) cells. The effects of VB1 on the proliferation of HCC cell lines HepG2, Hep3B, Huh-7 and human embryo liver L-02 cells were investigated using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Apoptotic death in HepG2 cells was examined using an enzyme-linked immunosorbent assay (ELISA) detection kit, flow cytometry after propidium iodide (PI) staining, and by DNA agarose gel electrophoresis. Caspase activity was measured using ELISA. The AKT/FOXO3a and ERK/FOXO3a pathways were analyzed using western blotting. VB1 inhibited human HCC cell proliferation in a concentration-dependent manner and increased the percentage of sub-G1 population HepG2 cells. Histone/DNA fragmentation and active caspase-3, -8 and -9 levels increased in a concentration-dependent manner and a DNA ladder was formed. The phosphorylation of AKT and ERK1/2 were inhibited and FOXO3a transcription factor was activated, resulting in apoptotic death. Knockdown of AKT1 by small interfering RNA (siRNA) and the MEK1/2 inhibitor, PD98059, enhanced VB1-induced apoptosis and FOXO3a transcriptional activity. Suppression of FOXO3a expression by siRNA inhibited VB1-induced apoptosis. VB1 induced expression of Bim, TRAIL, DR4 and DR5. Activation of the FOXO3a transcription factor appears to mediate pro-apoptotic effects of VB1 by inhibiting the AKT and ERK pathways.
Collapse
Affiliation(s)
- Jian-Gang Wang
- Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | | | | | | | | |
Collapse
|
38
|
Meng Q, Yin Q, Li Y. Nanocarriers for siRNA delivery to overcome cancer multidrug resistance. ACTA ACUST UNITED AC 2013. [DOI: 10.1007/s11434-013-6030-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
39
|
Ryu K, Kim TI. Therapeutic gene delivery using bioreducible polymers. Arch Pharm Res 2013; 37:31-42. [DOI: 10.1007/s12272-013-0275-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2013] [Accepted: 10/22/2013] [Indexed: 12/14/2022]
|
40
|
Mura S, Nicolas J, Couvreur P. Stimuli-responsive nanocarriers for drug delivery. NATURE MATERIALS 2013; 12:991-1003. [PMID: 24150417 DOI: 10.1038/nmat3776] [Citation(s) in RCA: 4072] [Impact Index Per Article: 370.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Accepted: 09/10/2013] [Indexed: 05/17/2023]
Abstract
Spurred by recent progress in materials chemistry and drug delivery, stimuli-responsive devices that deliver a drug in spatial-, temporal- and dosage-controlled fashions have become possible. Implementation of such devices requires the use of biocompatible materials that are susceptible to a specific physical incitement or that, in response to a specific stimulus, undergo a protonation, a hydrolytic cleavage or a (supra)molecular conformational change. In this Review, we discuss recent advances in the design of nanoscale stimuli-responsive systems that are able to control drug biodistribution in response to specific stimuli, either exogenous (variations in temperature, magnetic field, ultrasound intensity, light or electric pulses) or endogenous (changes in pH, enzyme concentration or redox gradients).
Collapse
Affiliation(s)
- Simona Mura
- Institut Galien Paris-Sud, Université Paris-Sud, UMR CNRS 8612, Faculté de Pharmacie, 5 rue Jean-Baptiste Clément, F-92296 Châtenay-Malabry Cedex, France
| | | | | |
Collapse
|
41
|
Shukla RS, Tai W, Mahato R, Jin W, Cheng K. Development of streptavidin-based nanocomplex for siRNA delivery. Mol Pharm 2013; 10:4534-45. [PMID: 24160908 DOI: 10.1021/mp400355q] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In our previous study, we have identified a PCBP2 siRNA that exhibits antifibrotic activity in rat hepatic stellate cells (HSCs) by inhibition of αCP2, a protein responsible for stabilization of the collagen α1 (I) mRNA in alcoholic liver fibrosis. This study aims to develop a streptavidin-based nanocomplex that can efficiently deliver the PCBP2 siRNA to HSCs. Biotin-siRNA and biotin-cholesterol were mixed with streptavidin to form the streptavidin-biotin complex, which was further condensed electrostatically with positively charged protamine to form the final multicomponent siRNA nanocomplex in the size range of 150-250 nm. The siRNA nanocomplex does not induce cytotoxicity in rat HSCs as compared to commercially available transfection agents. The cellular uptake efficiency of the siRNA nanocomplex is higher in rat HSCs than other cell lines, such as Caco-2 and PC-3, indicating that receptor-mediated endocytosis mainly contributes to the cellular uptake of the siRNA nanocomplex. The siRNA nanocomplex exhibits more than 85% silencing effect on the PCBP2 mRNA in HSCs. Stability study indicates that the nanocomplex can efficiently protect siRNA from degradation in the serum. The streptavidin-based multicomponent siRNA nanocomplex provides a promising strategy to deliver the PCBP2 siRNA to HSCs. Moreover, the nanocomplex can be used as a platform for other diseases by changing the siRNA sequence and targeting ligand.
Collapse
Affiliation(s)
- Ravi S Shukla
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City , Kansas City, Missouri 64108, United States
| | | | | | | | | |
Collapse
|
42
|
Baigude H, Su J, McCarroll J, Rana TM. In Vivo Delivery of RNAi by Reducible Interfering Nanoparticles (iNOPs). ACS Med Chem Lett 2013; 4:720-723. [PMID: 24319542 PMCID: PMC3850243 DOI: 10.1021/ml4001003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Accepted: 06/18/2013] [Indexed: 11/30/2022] Open
Abstract
![]()
RNA interference (RNAi) has considerable
potential as a therapeutic
strategy, but the development of efficient in vivo RNA delivery methods
remains challenging. To this end, we designed and synthesized chemically
modified interfering nanoparticles (iNOPs) composed of functionalized
poly-l-lysine dendrimers modified with reducible spacers
to facilitate release of small interfering RNAs (siRNAs) in vivo.
We show that the novel siRNA–iNOP complexes mediate efficient
gene-specific RNAi in cultured cells and in mice, where they display
enhanced tissue-targeting capabilities. At a clinically feasible dose
of 1 mg kg–1, apolipoprotein B (apoB) siRNA–iNOP
complexes achieved ∼40–45% reduction of liver apoB mRNA
and plasma apoB protein levels within 48 h of administration to mice,
without apparent toxicity. Collectively, these findings demonstrate
that siRNA delivery by the modified reducible iNOPs can provide a
clinically significant and potentially tissue-specific new approach
for RNAi therapy.
Collapse
Affiliation(s)
- Huricha Baigude
- Program for RNA Biology, Sanford−Burnham Medical Research Institute,
10901 North Torrey Pines Road, La Jolla, California 92037, United
States
- Department of Biochemistry
and
Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
| | - Jie Su
- Department of Biochemistry
and
Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
| | - Joshua McCarroll
- Department of Biochemistry
and
Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
| | - Tariq M. Rana
- Program for RNA Biology, Sanford−Burnham Medical Research Institute,
10901 North Torrey Pines Road, La Jolla, California 92037, United
States
- Department of Biochemistry
and
Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
- Department of Pediatrics and Biomedical
Sciences Graduate Program, University of California, San Diego, California 92093, United States
| |
Collapse
|
43
|
Lim KS, Cha MJ, Kim JK, Park EJ, Chae JW, Rhim T, Hwang KC, Kim YH. Protective effects of protein transduction domain-metallothionein fusion proteins against hypoxia- and oxidative stress-induced apoptosis in an ischemia/reperfusion rat model. J Control Release 2013; 169:306-12. [DOI: 10.1016/j.jconrel.2013.01.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 01/10/2013] [Accepted: 01/27/2013] [Indexed: 11/26/2022]
|
44
|
Zhu L, Torchilin VP. Stimulus-responsive nanopreparations for tumor targeting. Integr Biol (Camb) 2013; 5:96-107. [PMID: 22869005 DOI: 10.1039/c2ib20135f] [Citation(s) in RCA: 159] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Nanopreparations such as liposomes, micelles, polymeric and inorganic nanoparticles, and small molecule/nucleic acid/protein conjugates have demonstrated various advantages over "naked" therapeutic molecules. These nanopreparations can be further engineered with functional moieties to improve their performance in terms of circulation longevity, targetability, enhanced intracellular penetration, carrier-mediated enhanced visualization, and stimuli-sensitivity. The idea of application of a stimulus-sensitive drug or imaging agent delivery system for tumor targeting is based on the significant abnormalities in the tumor microenvironment and its cells, such as an acidic pH, altered redox potential, up-regulated proteins and hyperthermia. These internal conditions as well as external stimuli, such as magnetic field, ultrasound and light, can be used to modify the behavior of the nanopreparations that control drug release, improve drug internalization, control the intracellular drug fate and even allow for certain physical interactions, resulting in an enhanced tumor targeting and antitumor effect. This article provides a critical view of current stimulus-sensitive drug delivery strategies and possible future directions in tumor targeting with primary focus on the combined use of stimulus-sensitivity with other strategies in the same nanopreparation, including multifunctional nanopreparations and theranostics.
Collapse
Affiliation(s)
- Lin Zhu
- Department of Pharmaceutical Sciences and Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, 140 The Fenway, Boston, MA 02115, USA
| | | |
Collapse
|
45
|
Dynamics of PEGylated-dextran-spermine nanoparticles for gene delivery to leukemic cells. Appl Biochem Biotechnol 2013; 170:841-53. [PMID: 23615733 DOI: 10.1007/s12010-013-0224-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Accepted: 04/07/2013] [Indexed: 01/03/2023]
Abstract
Leukemic cells are hard-to-transfect cell lines. Many transfection reagents which can provide high gene transfer efficiency in common adherent cell lines are not effective to transfect established blood cell lines or primary leukemic cells. This study aims to examine a new class of cationic polymer non-viral vector, PEGylated-dextran-spermine (PEG-D-SPM), to determine its ability to transfect the leukemic cells. Here, the optimal conditions of the complex preparation (PEG-D-SPM/plasmid DNA (pDNA)) were examined. Different weight-mixing (w/w) ratios of PEG-D-SPM/pDNA complex were prepared to obtain an ideal mixing ratio to protect encapsulated pDNA from DNase degradation and to determine the optimal transfection efficiency of the complex. Strong complexation between polymer and pDNA in agarose gel electrophoresis and protection of pDNA from DNase were detected at ratios from 25 to 15. Highest gene expression was detected at w/w ratio of 18 in HL60 and K562 cells. However, gene expression from both leukemic cell lines was lower than the control MCF-7 cells. The cytotoxicity of PEG-D-SPM/pDNA complex at the most optimal mixing ratios was tested in HL60 and K562 cells using MTS assay and the results showed that the PEG-D-SPM/pDNA complex had no cytotoxic effect on these cell lines. Spherical shape and nano-nature of PEG-D-SPM/pDNA complex at ratio 18 was observed using transmission electron microscopy. As PEG-D-SPM showed modest transfection efficiency in the leukemic cell lines, we conclude that further work is needed to improve the delivery efficiency of the PEG-D-SPM.
Collapse
|
46
|
Alexander C, Fernandez Trillo F. Bioresponsive Polyplexes and Micelleplexes. SMART MATERIALS FOR DRUG DELIVERY 2013. [DOI: 10.1039/9781849736800-00256] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The delivery of nucleic acids (NAs) is hindered by several factors, such as the size of the biomolecule (micron size for plasmid DNA), the presence of different biological barriers or the degradation of NAs. Most of these limitations are avoided by complexation with polycationic species, which collapse NAs into nanometer-sized polyplexes that can be efficiently internalized into the target cells. Because there are subtle changes in physiological conditions, such as the drop in pH at the endosome, or the increase in temperature in tumor tissue, stimuli responsive synthetic polymers are ideal candidates for the synthesis of efficient gene delivery vehicles. In this chapter, representative examples of “smart” polypexes that exploit these changes in physiological environment for the delivery of NAs are described, and the transfection efficiency of pH-, redox-, temperature- and light-responsive polyplexes is analyzed.
Collapse
|
47
|
Chen CJ, Wang JC, Zhao EY, Gao LY, Feng Q, Liu XY, Zhao ZX, Ma XF, Hou WJ, Zhang LR, Lu WL, Zhang Q. Self-assembly cationic nanoparticles based on cholesterol-grafted bioreducible poly(amidoamine) for siRNA delivery. Biomaterials 2013; 34:5303-16. [PMID: 23570718 DOI: 10.1016/j.biomaterials.2013.03.056] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 03/18/2013] [Indexed: 12/20/2022]
Abstract
In this study, a series of bioreducible poly(amidoamine)s grafting different percentages of cholesterol (rPAA-Ch14: 14%, rPAA-Ch29: 29%, rPAA-Ch57: 57% and rPAA-Ch87: 87%) was synthesized and used for siRNA delivery. These amphiphilic polymers were able to self-assemble into cationic nanoparticles in aqueous solution at low concentrations. The nanoparticle formation was evidenced via cryo-transmission electron microscope (Cryo-TEM) and dynamic light scattering analysis. The average hydrodynamic size of rPAA-Ch blank nanoparticles was about 80-160 nm with zeta potential of 50-60 mV. Also, the effects of different percentages of cholesterol grafted onto rPAA on physicochemical characteristics, in vitro cytotoxicity, cellular uptake, VEGF gene silencing efficacy and translocation mechanism of rPAA-Ch/siRNA complexes were investigated. The results showed that rPAA-Ch57 polymer was not only able to form stable nanocomplexes and possess high cell uptake, but also to exhibit the best in vitro VEGF gene silencing efficacy and the best in vivo tumor growth inhibition effect when it was formulated with VEGF-siRNA. Moreover, the observations of confocal laser scanning microscope (CLSM) and the study of cholesterol competitive inhibition demonstrated that endosomal/lysosomal escape and cytoplasmic dissociation of rPAA-Ch57/siRNA complexes were dependent on the "proton sponge effect" and disulfide cleavage, following internalization with cholesterol-related endocytosis pathway and subsequent transportion into endosomes/lysosomes. These findings indicated that the rPAA-Ch57 polymer should be a promising and potent carrier for siRNA delivery.
Collapse
Affiliation(s)
- Cheng-Jun Chen
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Florinas S, Nam HY, Kim SW. Enhanced siRNA delivery using a combination of an arginine-grafted bioreducible polymer, ultrasound, and microbubbles in cancer cells. Mol Pharm 2013; 10:2021-30. [PMID: 23527953 DOI: 10.1021/mp400048p] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
RNAi-based gene therapy for cancer treatment has not shown significant clinical impact due to poor siRNA delivery to the target site. Here, we design a nonviral siRNA gene carrier using a combination of an arginine-grafted bioreducible polymer (ABP), microbubbles (MB), and ultrasound (US), for targeting vascular endothelial growth factor (VEGF) in a human ovarian cancer cell line. Newly designed MBs with a perfluorocrownether gas core show higher stability compared to controls. Further, MBs in combination with polyplexes show a significant higher loading capacity compared to naked siRNA. Lastly, only siRNA-ABP-MB (SAM) complexes in combination with US show significant VEGF knock down in A2780 human ovarian cancer cell line compared to naked siRNA when incubated for a short time after sonication treatment.
Collapse
Affiliation(s)
- Stelios Florinas
- Center for Controlled Chemical Delivery (CCCD), Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, Utah 84112, United States
| | | | | |
Collapse
|
49
|
Li J, Wang W, Sun D, Chen J, Zhang PH, Zhang JR, Min Q, Zhu JJ. Aptamer-functionalized silver nanoclusters-mediated cell type-specific siRNA delivery and tracking. Chem Sci 2013. [DOI: 10.1039/c3sc51538a] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
50
|
Chen CJ, Zhao ZX, Wang JC, Zhao EY, Gao LY, Zhou SF, Liu XY, Lu WL, Zhang Q. A comparative study of three ternary complexes prepared in different mixing orders of siRNA/redox-responsive hyperbranched poly (amido amine)/hyaluronic acid. Int J Nanomedicine 2012; 7:3837-49. [PMID: 22888238 PMCID: PMC3414221 DOI: 10.2147/ijn.s32676] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
In this study, a novel redox-responsive hyperbranched poly(amido amine) (named PCD) was synthesized and used as a cationic polymer to form a ternary complex with small interfering RNA (siRNA) and hyaluronic acid (HA) for siRNA delivery. Here, it is hypothesized that different mixing orders result in different assembly structures, which may affect the siRNA delivery efficiency. To investigate the effects of mixing orders on siRNA delivery efficiency in two human breast cancer cell lines, three ternary complexes with different mixing orders of siRNA/PCD/HA were prepared and characterized: mixing order I (initially prepared siRNA/PCD binary complex further coated by negatively charged HA), mixing order II ( initially prepared HA/PCD binary complex further incubated with siRNA), and mixing order III ( initially prepared siRNA/HA mixture further electrostatically compacted by positively charged PCD). With an optimized siRNA/PCD/HA charge ratio of 1/20/16, the particle sizes and zeta potentials of these ternary complexes were 124.8 nm and 27.3 mV (mixing order I), 147.5 nm and 29.9 mV (mixing order II), and 128.8 nm and 19.4 mV (mixing order III). Also, the effects on stability, cellular uptake, and gene silencing efficiency of siRNA formulated in ternary complexes with different mixing orders were investigated. The results showed that mixing orders I and III displayed better siRNA transfection and protection than mixing order II in human breast cancer MCF-7 and MDA-MB-231 cells. More interesting, at the siRNA/PCD/HA charge ratio of 1/20/16, the gene silencing effects on vascular endothelial growth factor expression in MDA-MB- 231 cells were as follows: mixing order III > mixing order I > mixing order II. Based on these results, a likely explanation for the difference in functionality dependent on mixing orders is the formation of different assembly structures. These results may help future optimization of siRNA ternary complexes for achieving better delivery efficiencies, especially for target-specific siRNA delivery to cells with HA receptor overexpression.
Collapse
Affiliation(s)
- Cheng-Jun Chen
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmaceutics, School of Pharmaceutical Science, Peking University, Beijing, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|