1
|
Cheng J, Jian L, Chen Z, Li Z, Yu Y, Wu Y. In Vivo Delivery Processes and Development Strategies of Lipid Nanoparticles. Chembiochem 2024; 25:e202400481. [PMID: 39101874 DOI: 10.1002/cbic.202400481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/29/2024] [Accepted: 08/05/2024] [Indexed: 08/06/2024]
Abstract
Lipid nanoparticles (LNPs) represent an advanced and highly efficient delivery system for RNA molecules, demonstrating exceptional biocompatibility and remarkable delivery efficiency. This is evidenced by the clinical authorization of three LNP formulations: Patisiran, BNT162b2, and mRNA-1273. To further maximize the efficacy of RNA-based therapy, it is imperative to develop more potent LNP delivery systems that can effectively protect inherently unstable and negatively charged RNA molecules from degradation by nucleases, while facilitating their cellular uptake into target cells. Therefore, this review presents feasible strategies commonly employed for the development of efficient LNP delivery systems. The strategies encompass combinatorial chemistry for large-scale synthesis of ionizable lipids, rational design strategy of ionizable lipids, functional molecules-derived lipid molecules, the optimization of LNP formulations, and the adjustment of particle size and charge property of LNPs. Prior to introducing these developing strategies, in vivo delivery processes of LNPs, a crucial determinant influencing the clinical translation of LNP formulations, is described to better understand how to develop LNP delivery systems.
Collapse
Affiliation(s)
- Jiashun Cheng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Lina Jian
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Zhaolin Chen
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Zhuoyuan Li
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yaobang Yu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yihang Wu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| |
Collapse
|
2
|
Sisto M, Lisi S. Epigenetic Modulations of Non-Coding RNAs: A Novel Therapeutic Perspective in Sjӧgren's Syndrome. FRONT BIOSCI-LANDMRK 2024; 29:403. [PMID: 39735974 DOI: 10.31083/j.fbl2912403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/02/2024] [Accepted: 07/10/2024] [Indexed: 12/31/2024]
Abstract
Sjögren's syndrome (SS) is an autoimmune disease that can be classified as an epithelitis based on the immune-mediated attack directed specifically at epithelial cells. SS predominantly affects women, is characterized by the production of highly specific circulating autoantibodies, and the major targets are the salivary and lachrymal glands. Although a genetic predisposition has been amply demonstrated for SS, the etiology remains unclear. The recent integration of epigenetic data relating to autoimmune diseases opens new therapeutic perspectives based on a better understanding of the molecular processes implicated. In the autoimmune field, non-coding RNA molecules (nc-RNA), which regulate gene expression by binding to mRNAs and could have a therapeutic value, have aroused great interest. The focus of this review is to summarize the biological functions of nc-RNAs in the pathogenesis of SS and decode molecular pathways implicated in the disease, in order to identify new therapeutic strategies.
Collapse
Affiliation(s)
- Margherita Sisto
- Department of Translational Biomedicine and Neuroscience (DiBraiN), Section of Human Anatomy and Histology, University of Bari "Aldo Moro", 70124 Bari, Italy
| | - Sabrina Lisi
- Department of Translational Biomedicine and Neuroscience (DiBraiN), Section of Human Anatomy and Histology, University of Bari "Aldo Moro", 70124 Bari, Italy
| |
Collapse
|
3
|
Mrksich K, Padilla MS, Mitchell MJ. Breaking the final barrier: Evolution of cationic and ionizable lipid structure in lipid nanoparticles to escape the endosome. Adv Drug Deliv Rev 2024; 214:115446. [PMID: 39293650 DOI: 10.1016/j.addr.2024.115446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/18/2024] [Accepted: 09/07/2024] [Indexed: 09/20/2024]
Abstract
In the past decade, nucleic acid therapies have seen a boon in development and clinical translation largely due to advances in nanotechnology that have enabled their safe and targeted delivery. Nanoparticles can protect nucleic acids from degradation by serum enzymes and can facilitate entry into cells. Still, achieving endosomal escape to allow nucleic acids to enter the cytoplasm has remained a significant barrier, where less than 5% of nanoparticles within the endo-lysosomal pathway are able to transfer their cargo to the cytosol. Lipid-based drug delivery vehicles, particularly lipid nanoparticles (LNPs), have been optimized to achieve potent endosomal escape, and thus have been the vector of choice in the clinic as demonstrated by their utilization in the COVID-19 mRNA vaccines. The success of LNPs is in large part due to the rational design of lipids that can specifically overcome endosomal barriers. In this review, we chart the evolution of lipid structure from cationic lipids to ionizable lipids, focusing on structure-function relationships, with a focus on how they relate to endosomal escape. Additionally, we examine recent advancements in ionizable lipid structure as well as discuss the future of lipid design.
Collapse
Affiliation(s)
- Kaitlin Mrksich
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marshall S Padilla
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael J Mitchell
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
4
|
Fajardo C, De Donato M, Macedo M, Charoonnart P, Saksmerprome V, Yang L, Purton S, Mancera JM, Costas B. RNA Interference Applied to Crustacean Aquaculture. Biomolecules 2024; 14:1358. [PMID: 39595535 PMCID: PMC11592254 DOI: 10.3390/biom14111358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/28/2024] Open
Abstract
RNA interference (RNAi) is a powerful tool that can be used to specifically knock-down gene expression using double-stranded RNA (dsRNA) effector molecules. This approach can be used in aquaculture as an investigation instrument and to improve the immune responses against viral pathogens, among other applications. Although this method was first described in shrimp in the mid-2000s, at present, no practical approach has been developed for the use of dsRNA in shrimp farms, as the limiting factor for farm-scale usage in the aquaculture sector is the lack of cost-effective and simple dsRNA synthesis and administration procedures. Despite these limitations, different RNAi-based approaches have been successfully tested at the laboratory level, with a particular focus on shrimp. The use of RNAi technology is particularly attractive for the shrimp industry because crustaceans do not have an adaptive immune system, making traditional vaccination methods unfeasible. This review summarizes recent studies and the state-of-the-art on the mechanism of action, design, use, and administration methods of dsRNA, as applied to shrimp. In addition, potential constraints that may hinder the deployment of RNAi-based methods in the crustacean aquaculture sector are considered.
Collapse
Affiliation(s)
- Carlos Fajardo
- Department of Biology, Faculty of Marine and Environmental Sciences, Instituto Universitario de Investigación Marina (INMAR), Campus de Excelencia Internacional del Mar (CEI-MAR), University of Cadiz (UCA), 11510 Puerto Real, Spain;
- Interdisciplinary Centre of Marine and Environmental Research, The University of Porto (CIIMAR), 4450-208 Matosinhos, Portugal; (M.M.); (B.C.)
| | - Marcos De Donato
- Center for Aquaculture Technologies (CAT), San Diego, CA 92121, USA;
- Escuela de Medicina y Ciencias de la Salud, Tecnológico de Monterrey, Querétaro 76130, Mexico
| | - Marta Macedo
- Interdisciplinary Centre of Marine and Environmental Research, The University of Porto (CIIMAR), 4450-208 Matosinhos, Portugal; (M.M.); (B.C.)
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto (UP), 4050-313 Porto, Portugal
| | - Patai Charoonnart
- Center of Excellence for Shrimp Molecular Biology and Biotechnology (Centex Shrimp), Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (P.C.); (V.S.)
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Bangkok 12120, Thailand
| | - Vanvimon Saksmerprome
- Center of Excellence for Shrimp Molecular Biology and Biotechnology (Centex Shrimp), Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (P.C.); (V.S.)
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Bangkok 12120, Thailand
| | - Luyao Yang
- Department of Structural and Molecular Biology, University College London (UCL), London WC1E 6BT, UK; (L.Y.); (S.P.)
| | - Saul Purton
- Department of Structural and Molecular Biology, University College London (UCL), London WC1E 6BT, UK; (L.Y.); (S.P.)
| | - Juan Miguel Mancera
- Department of Biology, Faculty of Marine and Environmental Sciences, Instituto Universitario de Investigación Marina (INMAR), Campus de Excelencia Internacional del Mar (CEI-MAR), University of Cadiz (UCA), 11510 Puerto Real, Spain;
| | - Benjamin Costas
- Interdisciplinary Centre of Marine and Environmental Research, The University of Porto (CIIMAR), 4450-208 Matosinhos, Portugal; (M.M.); (B.C.)
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto (UP), 4050-313 Porto, Portugal
| |
Collapse
|
5
|
Ebrahimi N, Manavi MS, Nazari A, Momayezi A, Faghihkhorasani F, Rasool Riyadh Abdulwahid AH, Rezaei-Tazangi F, Kavei M, Rezaei R, Mobarak H, Aref AR, Fang W. Nano-scale delivery systems for siRNA delivery in cancer therapy: New era of gene therapy empowered by nanotechnology. ENVIRONMENTAL RESEARCH 2023; 239:117263. [PMID: 37797672 DOI: 10.1016/j.envres.2023.117263] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 09/17/2023] [Accepted: 09/27/2023] [Indexed: 10/07/2023]
Abstract
RNA interference (RNAi) is a unique treatment approach used to decrease a disease's excessive gene expression, including cancer. SiRNAs may find and destroy homologous mRNA sequences within the cell thanks to RNAi processes. However, difficulties such poor cellular uptake, off-target effects, and susceptibility to destruction by serum nucleases in the bloodstream restrict the therapeutic potential of siRNAs. Since some years ago, siRNA-based therapies have been in the process of being translated into the clinic. Therefore, the primary emphasis of this work is on sophisticated nanocarriers that aid in the transport of siRNA payloads, their administration in combination with anticancer medications, and their use in the treatment of cancer. The research looks into molecular manifestations, difficulties with siRNA transport, the design and development of siRNA-based delivery methods, and the benefits and drawbacks of various nanocarriers. The trapping of siRNA in endosomes is a challenge for the majority of delivery methods, which affects the therapeutic effectiveness. Numerous techniques for siRNA release, including as pH-responsive release, membrane fusion, the proton sponge effect, and photochemical disruption, have been studied to overcome this problem. The present state of siRNA treatments in clinical trials is also looked at in order to give a thorough and systematic evaluation of siRNA-based medicines for efficient cancer therapy.
Collapse
Affiliation(s)
- Nasim Ebrahimi
- Genetics Division, Department of Cell and Molecular Biology and Microbiology, Faculty of Science and Technology, University of Isfahan, Iran
| | | | - Ahmad Nazari
- Tehran University of Medical Science, Tehran, Iran
| | - Amirali Momayezi
- School of Chemical Engineering, Iran University of Science, and Technology, Tehran, Iran
| | | | | | - Fatemeh Rezaei-Tazangi
- Department of Anatomy, School of Medicine, Fasa University of Medical Science, Fasa, Iran
| | - Mohammed Kavei
- Department of Biology, Faculty of Science, Arak University, Arak, Iran
| | - Roya Rezaei
- Department of Microbiology, College of Science, Agriculture and Modern Technology, Shiraz Branch, Islamic Azad University, Shiraz, Iran
| | - Halimeh Mobarak
- Clinical Pathologist, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Reza Aref
- Xsphera Biosciences, Translational Medicine Group, 6 Tide Street, Boston, MA, 02210, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA.
| | - Wei Fang
- Department of Laser and Aesthetic Medicine, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China.
| |
Collapse
|
6
|
Hartl N, Jürgens DC, Carneiro S, König AC, Xiao X, Liu R, Hauck SM, Merkel OM. Protein corona investigations of polyplexes with varying hydrophobicity - From method development to in vitro studies. Int J Pharm 2023; 643:123257. [PMID: 37482228 DOI: 10.1016/j.ijpharm.2023.123257] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/11/2023] [Accepted: 07/18/2023] [Indexed: 07/25/2023]
Abstract
In the field of non-viral drug delivery, polyplexes (PXs) represent an advanced investigated and highly promising tool for the delivery of nucleic acids. Upon encountering physiological fluids, they adsorb biological molecules to form a protein corona (PC), that influence PXs biodistribution, transfection efficiencies and targeting abilities. In an effort to understand protein - PX interactions and the effect of PX material on corona composition, we utilized cationic branched 10 kDa polyethyleneimine (b-PEI) and a hydrophobically modified nylon-3 polymer (NM0.2/CP0.8) within this study to develop appropriate methods for PC investigations. A centrifugation procedure for isolating hard corona - PX complexes (PCPXs) from soft corona proteins after incubating the PXs in fetal bovine serum (FBS) for PC formation was successfully optimized and the identification of proteins by a liquid chromatography-tandem mass spectrometry (LC-MS-MS) method clearly demonstrated that the PC composition is affected by the underlying PXs material. With regard to especially interesting functional proteins, which might be able to induce active targeting effects, several candidates could be detected on b-PEI and NM0.2/CP0.8 PXs. These results are of high interest to better understand how the design of PXs impacts the PC composition and subsequently PCPXs-cell interactions to enable precise adjustment of PXs for targeted drug delivery.
Collapse
Affiliation(s)
- Natascha Hartl
- Ludwig-Maximilians-University, Pharmaceutical Technology and Biopharmaceutics, Butenandtstr. 5-13, 81377 Munich, Germany
| | - David C Jürgens
- Ludwig-Maximilians-University, Pharmaceutical Technology and Biopharmaceutics, Butenandtstr. 5-13, 81377 Munich, Germany
| | - Simone Carneiro
- Ludwig-Maximilians-University, Pharmaceutical Technology and Biopharmaceutics, Butenandtstr. 5-13, 81377 Munich, Germany
| | - Ann-Christine König
- Metbolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Heidemannsstr. 1, 80939 Munich, Germany
| | - Ximian Xiao
- East China University of Science and Technology, 30 Meilong Rd, Shanghai, China
| | - Runhui Liu
- East China University of Science and Technology, 30 Meilong Rd, Shanghai, China
| | - Stefanie M Hauck
- Metbolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Heidemannsstr. 1, 80939 Munich, Germany
| | - Olivia M Merkel
- Ludwig-Maximilians-University, Pharmaceutical Technology and Biopharmaceutics, Butenandtstr. 5-13, 81377 Munich, Germany.
| |
Collapse
|
7
|
Huang LA, Lin C, Yang L. Plumbing mysterious RNAs in "dark genome" for the conquest of human diseases. Mol Ther 2023; 31:1577-1595. [PMID: 37165619 PMCID: PMC10278048 DOI: 10.1016/j.ymthe.2023.05.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 04/11/2023] [Accepted: 05/05/2023] [Indexed: 05/12/2023] Open
Abstract
Next-generation sequencing has revealed that less than 2% of transcribed genes are translated into proteins, with a large portion transcribed into noncoding RNAs (ncRNAs). Among these, long noncoding RNAs (lncRNAs) represent the largest group and are pervasively transcribed throughout the genome. Dysfunctions in lncRNAs have been found in various diseases, highlighting their potential as therapeutic, diagnostic, and prognostic targets. However, challenges, such as unknown molecular mechanisms and nonspecific immune responses, and issues of drug specificity and delivery present obstacles in translating lncRNAs into clinical applications. In this review, we summarize recent publications that have explored lncRNA functions in human diseases. We also discuss challenges and future directions for developing lncRNA treatments, aiming to bridge the gap between functional studies and clinical potential and inspire further exploration in the field.
Collapse
Affiliation(s)
- Lisa A Huang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chunru Lin
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Liuqing Yang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
8
|
Shui M, Chen Z, Chen Y, Yuan Q, Li H, Vong CT, Farag MA, Wang S. Engineering polyphenol-based carriers for nucleic acid delivery. Theranostics 2023; 13:3204-3223. [PMID: 37351161 PMCID: PMC10283064 DOI: 10.7150/thno.81604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 05/10/2023] [Indexed: 06/24/2023] Open
Abstract
Gene therapy, an effective medical intervention strategy, is increasingly employed in basic research and clinical practice for promising and unique therapeutic effects for diseases treatment, such as cardiovascular disorders, cancer, neurological pathologies, infectious diseases, and wound healing. However, naked DNA/RNA is readily hydrolyzed by nucleic acid degrading enzymes in the extracellular environment and degraded by lysosomes during intracellular physiological conditions, thus gene transfer must cross complex cellular and tissue barriers to deliver genetic materials into targeted cells and drive efficient activation or inhibition of the proteins. At present, the lack of safe, highly efficient, and non-immunogenic drug carriers is the main drawback of gene therapy. Considering the dense hydroxyl groups on the benzene rings in natural polyphenols that exert a strong affinity to various nucleic acids via hydrogen bonding and hydrophobic interactions, polyphenol-based carriers are promising anchors for gene delivery in which polyphenols serve as the primary building blocks. In this review, the recent progress in polyphenol-assisted gene delivery was summarized, which provided an easily accessible reference for the design of future polyphenol-based gene delivery vectors. Nucleic acids discussed in this review include DNA, short interfering RNAs (siRNA), microRNA (miRNA), double-strand RNA (dsRNA), and messenger RNA (mRNA).
Collapse
Affiliation(s)
- Mingju Shui
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
- Macao Centre for Research and Development in Chinese Medicine, University of Macau, Macao SAR 999078, China
| | - Zhejie Chen
- Institute of Molecular Medicine (IMM), Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yi Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
- Macao Centre for Research and Development in Chinese Medicine, University of Macau, Macao SAR 999078, China
| | - Qin Yuan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
| | - Hongyi Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
| | - Chi Teng Vong
- Macao Centre for Research and Development in Chinese Medicine, University of Macau, Macao SAR 999078, China
| | - Mohamed A. Farag
- Pharmacognosy Department, College of Pharmacy, Cairo University, Kasr el Aini St., Cairo 11562, Egypt
| | - Shengpeng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
- Macao Centre for Research and Development in Chinese Medicine, University of Macau, Macao SAR 999078, China
| |
Collapse
|
9
|
Ray L, Ray S. Enhanced anticancer activity of siRNA and drug codelivered by anionic biopolymer: overcoming electrostatic repulsion. Nanomedicine (Lond) 2023; 18:855-874. [PMID: 37503814 DOI: 10.2217/nnm-2022-0225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/29/2023] Open
Abstract
Aim: To codeliver an anticancer drug (doxorubicin) and siRNA in the form of nanoparticles into CD44-overexpressing colon cancer cells (HT-29) using an anionic, amphiphilic biopolymer comprising modified hyaluronic acid (6-O-[3-hexadecyloxy-2-hydroxypropyl]-hyaluronic acid). Materials & methods: Characterization of nanoparticles was performed using dynamic light scattering, scanning electron microscopy, transmission electron microscopy, molecular docking, in vitro drug release and gel mobility assays. Detailed in vitro experiments, including a gene silencing study and western blot, were also performed. Results: A 69% knockdown of the target gene was observed, and western blot showed 5.7-fold downregulation of the target protein. The repulsive forces between siRNA and 6-O-(3-hexadecyloxy-2-hydroxypropyl)-hyaluronic acid were overcome by hydrogen bonding and hydrophobic interactions. Conclusion: The authors successfully codelivered a drug and siRNA by anionic vector.
Collapse
Affiliation(s)
- Lipika Ray
- Pharmaceutics & Pharmacokinetics Division, Council of Scientific & Industrial Research-Central Drug Research Institute, Sector 10, Jankipuram Extension, Lucknow, Uttar Pradesh, 226031, India
| | - Sutapa Ray
- Department of Chemistry, Indian Institute of Technology, Kharagpur, 721302, India
| |
Collapse
|
10
|
Ancheta LR, Shramm PA, Bouajram R, Higgins D, Lappi DA. Streptavidin-Saporin: Converting Biotinylated Materials into Targeted Toxins. Toxins (Basel) 2023; 15:toxins15030181. [PMID: 36977072 PMCID: PMC10059012 DOI: 10.3390/toxins15030181] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/12/2023] [Accepted: 02/19/2023] [Indexed: 03/02/2023] Open
Abstract
Streptavidin-Saporin can be considered a type of ‘secondary’ targeted toxin. The scientific community has taken advantage of this conjugate in clever and fruitful ways using many kinds of biotinylated targeting agents to send saporin into a cell selected for elimination. Saporin is a ribosome-inactivating protein that causes inhibition of protein synthesis and cell death when delivered inside a cell. Streptavidin-Saporin, mixed with biotinylated molecules to cell surface markers, results in powerful conjugates that are used both in vitro and in vivo for behavior and disease research. Streptavidin-Saporin harnesses the ‘Molecular Surgery’ capability of saporin, creating a modular arsenal of targeted toxins used in applications ranging from the screening of potential therapeutics to behavioral studies and animal models. The reagent has become a well-published and validated resource in academia and industry. The ease of use and diverse functionality of Streptavidin-Saporin continues to have a significant impact on the life science industry.
Collapse
|
11
|
Messenger RNA Nanovaccine in Cancer Immunotherapy. Cancer Nanotechnol 2023. [DOI: 10.1007/978-3-031-17831-3_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
12
|
Jai J, Shirleen D, Hanbali C, Wijaya P, Anginan TB, Husada W, Pratama MY. Multiplexed shRNA-miRs as a candidate for anti HIV-1 therapy: strategies, challenges, and future potential. J Genet Eng Biotechnol 2022; 20:172. [PMID: 36576612 PMCID: PMC9797628 DOI: 10.1186/s43141-022-00451-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 12/04/2022] [Indexed: 12/29/2022]
Abstract
The spread of HIV is on the rise and has become a global issue, especially for underdeveloped and developing countries. This is due to the fact that HIV majorly occurs asymptomatically and is implausible for early diagnosis. Recent advances in research and science have enabled the investigation of a new potential treatment involving gene-based therapy, known as RNA interference (RNAi) that will direct gene silencing and further compensate for natural variants and viral mutants. Several types of small regulatory RNA are discussed in this present study, including microRNA (miRNA), small interfering RNA (siRNA), and short hairpin RNA (shRNA).This paper examines the mechanism of RNAi as a viable HIV therapy, using a minimum of four shRNAs to target both dispensable host components (CCR5) and viral genes (Gag, Env, Tat, Pol I, Pol II and Vif). Moreover, a multiplexed mechanism of shRNAs and miRNA is known to be effective in preventing viral escape due to mutation as the miRNA develops a general polycistronic platform for the expression of a large amount of shRNA-miRs. Several administration methods as well as the advantages of this RNAi treatment are also discussed in this study. The administration methods include (1) ex vivo delivery with the help of viral vectors, nanoparticles, and electroporation, (2) nonspecific in vivo delivery using non-viral carriers including liposomes, dendrimers and aptamers, as well as (3) targeted delivery that uses antibodies, modified nanoparticles, nucleic acid aptamers, and tissue-specific serotypes of AAV. Moreover, the advantages of this treatment are related to the effectiveness in silencing the HIV gene, which is more compatible compared to other gene therapy treatments, such as ZFN, TALEN, and CRISPR/Cas9.
Collapse
Affiliation(s)
- Jyotsna Jai
- grid.504251.70000 0004 7706 8927Department of Biotechnology, Indonesia International Institute for Life-Sciences (i3L), Jakarta, Indonesia
| | - Deborah Shirleen
- grid.504251.70000 0004 7706 8927Department of Biotechnology, Indonesia International Institute for Life-Sciences (i3L), Jakarta, Indonesia
| | - Christian Hanbali
- grid.504251.70000 0004 7706 8927Department of Biomedicine, Indonesia International Institute for Life-Sciences (i3L), Jakarta, Indonesia
| | - Pamela Wijaya
- grid.504251.70000 0004 7706 8927Department of Biomedicine, Indonesia International Institute for Life-Sciences (i3L), Jakarta, Indonesia
| | - Theresia Brigita Anginan
- grid.504251.70000 0004 7706 8927Department of Biomedicine, Indonesia International Institute for Life-Sciences (i3L), Jakarta, Indonesia
| | - William Husada
- grid.504251.70000 0004 7706 8927Department of Biotechnology, Indonesia International Institute for Life-Sciences (i3L), Jakarta, Indonesia
| | - Muhammad Yogi Pratama
- grid.504251.70000 0004 7706 8927Department of Biomedicine, Indonesia International Institute for Life-Sciences (i3L), Jakarta, Indonesia ,grid.240324.30000 0001 2109 4251Division of Vascular Surgery, Department of Surgery, New York University Medical Center, New York, USA
| |
Collapse
|
13
|
Basha G, Cottle AG, Pretheeban T, Chan KY, Witzigmann D, Young RN, Rossi FM, Cullis PR. Lipid nanoparticle-mediated silencing of osteogenic suppressor GNAS leads to osteogenic differentiation of mesenchymal stem cells in vivo. Mol Ther 2022; 30:3034-3051. [PMID: 35733339 PMCID: PMC9481989 DOI: 10.1016/j.ymthe.2022.06.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 05/09/2022] [Accepted: 06/17/2022] [Indexed: 11/21/2022] Open
Abstract
Approved drugs for the treatment of osteoporosis can prevent further bone loss but do not stimulate bone formation. Approaches that improve bone density in metabolic diseases are needed. Therapies that take advantage of the ability of mesenchymal stem cells (MSCs) to differentiate into various osteogenic lineages to treat bone disorders are of particular interest. Here we examine the ability of small interfering RNA (siRNA) to enhance osteoblast differentiation and bone formation by silencing the negative suppressor gene GNAS in bone MSCs. Using clinically validated lipid nanoparticle (LNP) siRNA delivery systems, we show that silencing the suppressor gene GNAS in vitro in MSCs leads to molecular and phenotypic changes similar to those seen in osteoblasts. Further, we demonstrate that these LNP-siRNAs can transfect a large proportion of mice MSCs in the compact bone following intravenous injection. Transfection of MSCs in various animal models led to silencing of GNAS and enhanced differentiation of MSCs into osteoblasts. These data demonstrate the potential for LNP delivery of siRNA to enhance the differentiation of MSCs into osteoblasts, and suggests that they are a promising approach for the treatment of osteoporosis and other bone diseases.
Collapse
Affiliation(s)
- Genc Basha
- NanoMedicines Research Group, Department of Biochemistry and Molecular Biology, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada.
| | - Andrew G Cottle
- NanoMedicines Research Group, Department of Biochemistry and Molecular Biology, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Thavaneetharajah Pretheeban
- School of Biomedical Engineering and Department of Medical Genetics, Biomedical Research Centre University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Karen Yt Chan
- NanoMedicines Research Group, Department of Biochemistry and Molecular Biology, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Dominik Witzigmann
- NanoMedicines Research Group, Department of Biochemistry and Molecular Biology, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; NanoMedicines Innovation Network (NMIN), University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Robert N Young
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6, Canada
| | - Fabio Mv Rossi
- School of Biomedical Engineering and Department of Medical Genetics, Biomedical Research Centre University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Pieter R Cullis
- NanoMedicines Research Group, Department of Biochemistry and Molecular Biology, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; NanoMedicines Innovation Network (NMIN), University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
14
|
Eptaminitaki GC, Stellas D, Bonavida B, Baritaki S. Long Non-coding RNAs (lncRNAs) signaling in Cancer Chemoresistance: From Prediction to Druggability. Drug Resist Updat 2022; 65:100866. [DOI: 10.1016/j.drup.2022.100866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/15/2022] [Accepted: 09/19/2022] [Indexed: 11/03/2022]
|
15
|
Valiunas V, Gordon C, Valiuniene L, Devine D, Lin RZ, Cohen IS, Brink PR. Intercellular delivery of therapeutic oligonucleotides. J Drug Deliv Sci Technol 2022; 72:103404. [PMID: 36721641 PMCID: PMC9886232 DOI: 10.1016/j.jddst.2022.103404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
One promising approach to cancer therapeutics is to induce changes in gene expression that either reduce cancer cell proliferation or induce cancer cell death. Therefore, delivering oligonucleotides (siRNA/miRNA) that target specific genes or gene programs might have a potential therapeutic benefit. The aim of this study was to examine the potential of cell-based delivery of oligonucleotides to cancer cells via two naturally occurring intercellular pathways: gap junctions and vesicular/exosomal traffic. We utilized human mesenchymal stem cells (hMSCs) as delivery cells and chose to deliver in vitro two synthetic oligonucleotides, AllStars HS Cell Death siRNA and miR-16 mimic, as toxic (therapeutic) oligonucleotides targeting three cancer cell lines: prostate (PC3), pancreatic (PANC1) and cervical (HeLa). Both oligonucleotides dramatically reduced cell proliferation and/or induced cell death when transfected directly into target cells and delivery hMSCs. The delivery and target cells we chose express gap junction connexin 43 (Cx43) endogenously (PC3, PANC1, hMSC) or via stable transfection (HeLaCx43). Co-culture of hMSCs (transfected with either toxic oligonucleotide) with any of Cx43 expressing cancer cells induced target cell death (~20% surviving) or senescence (~85% proliferation reduction) over 96 hours. We eliminated gap junction-mediated delivery by using connexin deficient HeLaWT cells or knocking out endogenous Cx43 in PANC1 and PC3 cells via CRISPR/Cas9. Subsequently, all Cx43 deficient target cells co-cultured with the same toxic oligonucleotide loaded hMSCs proliferated, albeit at significantly slower rates, with cell number increasing on average ~2.2-fold (30% of control cells) over 96 hours. Our results show that both gap junction and vesicular/exosomal intercellular delivery pathways from hMSCs to target cancer cells deliver oligonucleotides and function to either induce cell death or significantly reduce their proliferation. Thus, hMSC-based cellular delivery is an effective method of delivering synthetic oligonucleotides that can significantly reduce tumor cell growth and should be further investigated as a possible approach to cancer therapy.
Collapse
Affiliation(s)
- Virginijus Valiunas
- Department of Physiology and Biophysics and the Institute for Molecular Cardiology Stony Brook University, Stony Brook, NY 11794-8661, USA
| | - Chris Gordon
- Department of Physiology and Biophysics and the Institute for Molecular Cardiology Stony Brook University, Stony Brook, NY 11794-8661, USA
| | - Laima Valiuniene
- Department of Physiology and Biophysics and the Institute for Molecular Cardiology Stony Brook University, Stony Brook, NY 11794-8661, USA
| | - Daniel Devine
- Department of Physiology and Biophysics and the Institute for Molecular Cardiology Stony Brook University, Stony Brook, NY 11794-8661, USA
| | - Richard Z Lin
- Department of Physiology and Biophysics and the Institute for Molecular Cardiology Stony Brook University, Stony Brook, NY 11794-8661, USA
| | - Ira S Cohen
- Department of Physiology and Biophysics and the Institute for Molecular Cardiology Stony Brook University, Stony Brook, NY 11794-8661, USA
| | - Peter R Brink
- Department of Physiology and Biophysics and the Institute for Molecular Cardiology Stony Brook University, Stony Brook, NY 11794-8661, USA
| |
Collapse
|
16
|
Wei Y, Yang J, Feng X, Akhavan-Sigari R. Cellular and Molecular Mechanism of Cell Proliferation in Human Gastric Cancer Drug-Resistant Cells After Hyperthermia and Cisplatin: Role of mRNAs and Long-Non-coding RNAs. THE TURKISH JOURNAL OF GASTROENTEROLOGY : THE OFFICIAL JOURNAL OF TURKISH SOCIETY OF GASTROENTEROLOGY 2022; 33:377-386. [PMID: 35678795 PMCID: PMC11158417 DOI: 10.5152/tjg.2022.20845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 07/27/2021] [Indexed: 06/15/2023]
Abstract
BACKGROUND Since thermo-chemotherapy was suggested as an effective treatment for gastric cancer, we aimed to evaluate the effects of hyperthermia combined with cisplatin (DDP) on the inhibition of human gastric cancer drug-resistant cells in vitro and explore its possible mechanisms. METHODS SGC-7901/DDP cells were cultured and divided into control, cisplatin, hyperthermia, and hyperthermia combined with cispla- tin groups. Hyperthermia was done at 42°C, 44°C, 46°C, 48°C, and 50°C for 12 h, 24 h, 36 h; 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl- 2H-tetrazolium bromide (MTT) assay detected the proliferation of SGC-7901/DDP at different time and temperature, and the apoptotic rate of SGC-7901/DDP cells was evaluated by using Annexin staining assay. High-throughput Chromatin immunoprecipitation (ChIP)- seq was applied to test long non-coding RNA expression in SGC-7901/DDP cells. Then, real-time fluorescence quantitative polymerase chain reaction was used to verify the expression of long non-coding RNA in all groups. RESULTS Double staining showed that hyperthermia combined with cisplatin increased the rate of early apoptosis of SGC-7901/DDP cells. Long non-coding RNA high-throughput ChIP-seq showed a significantly larger amount of long non-coding RNAs and mRNAs in the cells treated with hyperthermia combined cisplatin group in comparison with the control group. We observed that the upregulated mRNAs and long non-coding RNAs were highly related to immune system response and CD95 signaling pathway in nucleus, and down- regulated mRNAs and long non-coding RNA were highly related to Mammalian target of rapamycin (mTOR) and Tumor necrosis factor (TNF) receptor signaling pathway in cytoplasm. CONCLUSION Hyperthermia combined with cisplatin reversed the expression of a large number of mRNAs and long non-coding RNAs in human gastric cancer drug-resistant cells. The molecular mechanism of inhibiting the proliferation of human gastric cancer drug- resistant cells may be related to the upregulation of long non-coding RNAs and mRNAs contributed in CD95, mTOR, and TNF receptor signaling pathway.
Collapse
Affiliation(s)
- Ying Wei
- Department of Internal Medicine, Mashhad University of Medical Sciences Faculty of Medicine, Mashhad, Iran
- Department of Gastroenterology, Shangrao People’s Hospital, Jiangxi Province, China
| | - Jing Yang
- Department of Pathology, Shangrao People’s Hospital, Jiangxi Province, China
- Department of Gastroenterology, Shangrao People’s Hospital, Jiangxi Province, China
| | - Xiaoke Feng
- Department of Gastroenterology, Shangrao People’s Hospital, Jiangxi Province, China
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing City, China
| | - Reza Akhavan-Sigari
- Department of Pathology, Shangrao People’s Hospital, Jiangxi Province, China
| |
Collapse
|
17
|
Maishi N, Sakurai Y, Hatakeyama H, Umeyama Y, Nakamura T, Endo R, Alam MT, Li C, Annan DAM, Kikuchi H, Morimoto H, Morimoto M, Akiyama K, Ohga N, Hida Y, Harashima H, Hida K. Novel antiangiogenic therapy targeting biglycan using tumor endothelial cell-specific liposomal siRNA delivery system. Cancer Sci 2022; 113:1855-1867. [PMID: 35266253 PMCID: PMC9128192 DOI: 10.1111/cas.15323] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 02/21/2022] [Accepted: 03/02/2022] [Indexed: 12/01/2022] Open
Abstract
Tumor blood vessels play important roles in tumor progression and metastasis. Targeting tumor endothelial cells (TECs) is one of the strategies for cancer therapy. We previously reported that biglycan, a small leucine‐rich proteoglycan, is highly expressed in TECs. TECs utilize biglycan in an autocrine manner for migration and angiogenesis. Furthermore, TEC‐derived biglycan stimulates tumor cell migration in a paracrine manner leading to tumor cell intravasation and metastasis. In this study, we explored the therapeutic effect of biglycan inhibition in the TECs of renal cell carcinoma using an in vivo siRNA delivery system known as a multifunctional envelope‐type nanodevice (MEND), which contains a unique pH‐sensitive cationic lipid. To specifically deliver MEND into TECs, we incorporated cyclo(Arg–Gly–Asp–d–Phe–Lys) (cRGD) into MEND because αVβ3 integrin, a receptor for cRGD, is selective and highly expressed in TECs. We developed RGD‐MEND‐encapsulating siRNA against biglycan. First, we confirmed that MEND was delivered into OS‐RC‐2 tumor‐derived TECs and induced in vitro RNAi‐mediated gene silencing. MEND was then injected intravenously into OS‐RC‐2 tumor‐bearing mice. Flow cytometry analysis demonstrated that MEND was specifically delivered into TECs. Quantitative RT‐PCR indicated that biglycan was knocked down by biglycan siRNA‐containing MEND. Finally, we analyzed the therapeutic effect of biglycan silencing by MEND in TECs. Tumor growth was inhibited by biglycan siRNA‐containing MEND. Tumor microenvironmental factors such as fibrosis were also normalized using biglycan inhibition in TECs. Biglycan in TECs can be a novel target for cancer treatment.
Collapse
Affiliation(s)
- Nako Maishi
- Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan.,Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan.,Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Yu Sakurai
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan.,Membrane Transport and Drug Targeting Laboratory, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Hiroto Hatakeyama
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan.,Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Yui Umeyama
- Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Takashi Nakamura
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Rikito Endo
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Mohammad Towfik Alam
- Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan.,Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan.,Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Cong Li
- Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Dorcas Akuba-Muhyia Annan
- Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan.,Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroshi Kikuchi
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan.,Department of Renal and Genitourinary Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Hirofumi Morimoto
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Masahiro Morimoto
- Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan.,Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan.,Department of Oral Diagnosis and Medicine, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Kosuke Akiyama
- Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Noritaka Ohga
- Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan.,Department of Oral Diagnosis and Medicine, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Yasuhiro Hida
- Department of Cardiovascular and Thoracic Surgery, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | | | - Kyoko Hida
- Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan.,Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan.,Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| |
Collapse
|
18
|
Abdel-Bar HM, Walters AA, Lim Y, Rouatbi N, Qin Y, Gheidari F, Han S, Osman R, Wang JTW, Al-Jamal KT. An "eat me" combinatory nano-formulation for systemic immunotherapy of solid tumors. Theranostics 2021; 11:8738-8754. [PMID: 34522209 PMCID: PMC8419059 DOI: 10.7150/thno.56936] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 05/11/2021] [Indexed: 01/08/2023] Open
Abstract
Rational: Tumor immunogenic cell death (ICD), induced by certain chemotherapeutic drugs such as doxorubicin (Dox), is a form of apoptosis potentiating a protective immune response. One of the hallmarks of ICD is the translocation of calreticulin to the cell surface acting as an 'eat me' signal. This manuscript describes the development of a stable nucleic acid-lipid particles (SNALPs) formulation for the simultaneous delivery of ICD inducing drug (Dox) with small interfering RNA (siRNA) knocking down CD47 (siCD47), the dominant 'don't eat me' marker, for synergistic enhancement of ICD. Methods: SNALPs loaded with Dox or siCD47 either mono or combinatory platforms were prepared by ethanol injection method. The proposed systems were characterized for particle size, surface charge, entrapment efficiency and in vitro drug release. The ability of the SNALPs to preserve the siRNA integrity in presence of serum and RNAse were assessed over 48 h. The in vitro cellular uptake and gene silencing of the prepared SNALPs was assessed in CT26 cells. The immunological responses of the SNALPs were defined in vitro in terms of surface calreticulin expression and macrophage-mediated phagocytosis induction. In vivo therapeutic studies were performed in CT26 bearing mice where the therapeutic outcomes were expressed as tumor volume, expression of CD4 and CD8 as well as in vivo silencing. Results: The optimized SNALPs had a particle size 122 ±6 nm and an entrapment efficiency > 65% for both siRNA and Dox with improved serum stability. SNALPs were able to improve siRNA and Dox uptake in CT26 cells with enhanced cytotoxicity. siCD47 SNALPs were able to knockdown CD47 by approximately 70% with no interference from the presence of Dox. The siCD47 and Dox combination SNALPs were able to induce surface calreticulin expression leading to a synergistic effect on macrophage-mediated phagocytosis of treated cells. In a tumor challenge model, 50% of mice receiving siCD47 and Dox containing SNALPs were able to clear the tumor, while the remaining animals showed significantly lower tumor burden as compared to either monotreatment. Conclusion: Therefore, the combination of siCD47 and Dox in a particulate system showed potent anti-tumor activity which merits further investigation in future clinical studies.
Collapse
Affiliation(s)
- Hend Mohamed Abdel-Bar
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
- Department of Pharmaceutics, Faculty of Pharmacy, University of Sadat City, P.O. box: 32958 Egypt
| | - Adam A Walters
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Yau Lim
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Nadia Rouatbi
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Yue Qin
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Fatemeh Gheidari
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Shunping Han
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Rihab Osman
- Faculty of Pharmacy-Ain Shams University, Abbassia, Cairo, P.O. box: 11566 Egypt
| | - Julie Tzu-Wen Wang
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Khuloud T. Al-Jamal
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| |
Collapse
|
19
|
Engineering of Solid Dosage Forms of siRNA-Loaded Lipidoid-Polymer Hybrid Nanoparticles Using a Quality-by-Design Approach. Methods Mol Biol 2021. [PMID: 33928574 DOI: 10.1007/978-1-0716-1298-9_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Therapy based on RNA interference (RNAi), which can be mediated by exogenous small interfering RNA (siRNA), has potential for the management of diseases at the genetic level by silencing gene function(s). In all eukaryotic cells, RNAi is an endogenous regulatory mechanism, where messenger RNA (mRNA) is degraded, preventing its translation into protein. A significant advantage of RNAi therapy is that siRNA is very potent and gene silencing is highly specific, ensuring few off-target effects. However, the delivery of exogenous siRNA to the RNAi pathway in the cytosol is a challenge, and there is a need for development of advanced delivery systems to ensure safe and effective delivery of siRNA to the intracellular target site. Recently, we demonstrated the ability of lipid-polymer hybrid nanoparticles (LPNs) composed of cationic lipidoid 5 (L5) and the biodegradable polymer poly(DL-lactic-co-glycolic acid) to effectively deliver siRNA directed against tumor necrosis factor alpha (TNF-α) intracellularly to macrophages. L5 is a novel lipid-like material consisting of a tetraamine backbone linked to five C12 alkyl chains. Here, we describe a systematic quality-by-design (QbD) approach including risk assessment and design of experiments to investigate the influence of critical formulation parameters (i.e., L5 content and L5:TNF-α siRNA ratio (w/w)) on the physicochemical properties and the TNF-α gene silencing ability of TNF-α siRNA-loaded LPNs, prepared by using a double emulsion solvent evaporation method. We then detail protocols for the manufacturing of more stable solid dosage forms of LPNs using freeze drying and spray drying processes, respectively. We also provide protocols for characterization of the physicochemical properties of the nanocomposite dry powders, including (1) process yield, (2) aerodynamic particle size, (3) surface morphology, (4) moisture content, and (5) solid state properties. General considerations are provided that emphasize the advantages and disadvantages of applying QbD approaches for optimizing nanoparticulate formulations.
Collapse
|
20
|
Forbes TA, Brown BD, Lai C. Therapeutic RNA interference: A novel approach to the treatment of primary hyperoxaluria. Br J Clin Pharmacol 2021; 88:2525-2538. [PMID: 34022071 PMCID: PMC9291495 DOI: 10.1111/bcp.14925] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 04/19/2021] [Accepted: 05/08/2021] [Indexed: 12/13/2022] Open
Abstract
RNA interference (RNAi) is a natural biological pathway that inhibits gene expression by targeted degradation or translational inhibition of cytoplasmic mRNA by the RNA induced silencing complex. RNAi has long been exploited in laboratory research to study the biological consequences of the reduced expression of a gene of interest. More recently RNAi has been demonstrated as a therapeutic avenue for rare metabolic diseases. This review presents an overview of the cellular RNAi machinery as well as therapeutic RNAi design and delivery. As a clinical example we present primary hyperoxaluria, an ultrarare inherited disease of increased hepatic oxalate production which leads to recurrent calcium oxalate kidney stones. In the most common form of the disease (Type 1), end‐stage kidney disease occurs in childhood or young adulthood, often necessitating combined kidney and liver transplantation. In this context we discuss nedosiran (Dicerna Pharmaceuticals, Inc.) and lumasiran (Alnylam Pharmaceuticals), which are both novel RNAi therapies for primary hyperoxaluria that selectively reduce hepatic expression of lactate dehydrogenase and glycolate oxidase respectively, reducing hepatic oxalate production and urinary oxalate levels. Finally, we consider future optimizations advances in RNAi therapies.
Collapse
Affiliation(s)
- Thomas A Forbes
- Royal Children's Hospital, Parkville, Victoria, Australia.,Murdoch Children's Research Institute, Parkville, Victoria, Australia.,University of Melbourne, Parkville, Victoria, Australia
| | | | | |
Collapse
|
21
|
Dyudeeva ES, Pavlova AS, Kupryushkin MS, Pyshnyi DV, Pyshnaya IA. Problems of the Synthesis of Oligonucleotide Derivatives in the Realization of the Anchimeric Effect. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2021. [DOI: 10.1134/s1068162021020096] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
22
|
Abo-Al-Ela HG. RNA Interference in Aquaculture: A Small Tool for Big Potential. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:4343-4355. [PMID: 33835783 DOI: 10.1021/acs.jafc.1c00268] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
For decades, the tight regulatory functions of DNA and RNA have been the focus of extensive research with the goal of harnessing RNA molecules (e.g., microRNA and small interfering RNA) to control gene expression and to study biological functions. RNA interference (RNAi) has shown evidence of mediating gene expression, has been utilized to study functional genomics, and recently has potential in therapeutic agents. RNAi is a natural mechanism and a well-studied tool that can be used to silence specific genes. This method is also used in aquaculture as a research tool and to enhance immune responses. RNAi methods do have their limitations (e.g., immune triggering); efficient and easy-to-use RNAi methods for large-scale applications need further development. Despite these limitations, RNAi methods have been successfully used in aquaculture, in particular shrimp. This review discusses the uses of RNAi in aquaculture, such as immune- and production-related issues and the possible limitations that may hinder the application of RNAi in the aquaculture industry. Our challenge is to develop a highly potent in vivo RNAi delivery platform that could complete the desired action with minimal side effects and which can be applied on a large-scale with relatively little expense in the aquaculture industry.
Collapse
Affiliation(s)
- Haitham G Abo-Al-Ela
- Genetics and Biotechnology, Department of Aquaculture, Faculty of Fish Resources, Suez University, Suez 43518, Egypt
| |
Collapse
|
23
|
Schellinger IN, Dannert AR, Mattern K, Raaz U, Tsao PS. Unresolved Issues in RNA Therapeutics in Vascular Diseases With a Focus on Aneurysm Disease. Front Cardiovasc Med 2021; 8:571076. [PMID: 33937351 PMCID: PMC8081859 DOI: 10.3389/fcvm.2021.571076] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 02/23/2021] [Indexed: 12/20/2022] Open
Abstract
New technologies have greatly shaped the scientific and medical landscape within the last years. The unprecedented expansion of data and information on RNA biology has led to the discovery of new RNA classes with unique functions and unexpected modifications. Today, the biggest challenge is to transfer the large number of findings in basic RNA biology into corresponding clinical RNA-based therapeutics. Lately, this research begins to yield positive outcomes. RNA drugs advance to the final phases of clinical trials or even receive FDA approval. Furthermore, the introduction of the RNA-guided gene-editing technology CRISPR and advances in the delivery of messenger RNAs have triggered a major progression in the field of RNA-therapeutics. Especially short interfering RNAs and antisense oligonucleotides are promising examples for novel categories of therapeutics. However, several issues need to be addressed including intracellular delivery, toxicity, and immune responses before utilizing RNAs in a clinical setting. In this review, we provide an overview on opportunities and challenges for clinical translation of RNA-based therapeutics, with an emphasis on advances in novel delivery technologies and abdominal aortic aneurysm disease where non-coding RNAs have been shown to play a crucial regulatory role.
Collapse
Affiliation(s)
- Isabel N Schellinger
- Department of Cardiology and Pneumology, Heart Center at the University Medical Center Göttingen, Göttingen, Germany.,German Center for Cardiovascular Research (DZHK) e.V., Partner Site Göttingen, Göttingen, Germany.,Department for Endocrinology, Nephrology and Rheumatology, University Medical Center Leipzig, University of Leipzig, Leipzig, Germany.,Department for Angiology, University Medical Center Leipzig, University of Leipzig, Leipzig, Germany
| | - Angelika R Dannert
- Department of Cardiology and Pneumology, Heart Center at the University Medical Center Göttingen, Göttingen, Germany
| | - Karin Mattern
- Department of Cardiology and Pneumology, Heart Center at the University Medical Center Göttingen, Göttingen, Germany.,German Center for Cardiovascular Research (DZHK) e.V., Partner Site Göttingen, Göttingen, Germany
| | - Uwe Raaz
- Department of Cardiology and Pneumology, Heart Center at the University Medical Center Göttingen, Göttingen, Germany.,German Center for Cardiovascular Research (DZHK) e.V., Partner Site Göttingen, Göttingen, Germany
| | - Philip S Tsao
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, United States.,Veteran Affairs (VA) Palo Alto Health Care System, Palo Alto, CA, United States
| |
Collapse
|
24
|
RNA Interference Strategies for Future Management of Plant Pathogenic Fungi: Prospects and Challenges. PLANTS 2021; 10:plants10040650. [PMID: 33805521 PMCID: PMC8067263 DOI: 10.3390/plants10040650] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/24/2021] [Accepted: 03/25/2021] [Indexed: 12/11/2022]
Abstract
Plant pathogenic fungi are the largest group of disease-causing agents on crop plants and represent a persistent and significant threat to agriculture worldwide. Conventional approaches based on the use of pesticides raise social concern for the impact on the environment and human health and alternative control methods are urgently needed. The rapid improvement and extensive implementation of RNA interference (RNAi) technology for various model and non-model organisms has provided the initial framework to adapt this post-transcriptional gene silencing technology for the management of fungal pathogens. Recent studies showed that the exogenous application of double-stranded RNA (dsRNA) molecules on plants targeting fungal growth and virulence-related genes provided disease attenuation of pathogens like Botrytis cinerea, Sclerotinia sclerotiorum and Fusarium graminearum in different hosts. Such results highlight that the exogenous RNAi holds great potential for RNAi-mediated plant pathogenic fungal disease control. Production of dsRNA can be possible by using either in-vitro or in-vivo synthesis. In this review, we describe exogenous RNAi involved in plant pathogenic fungi and discuss dsRNA production, formulation, and RNAi delivery methods. Potential challenges that are faced while developing a RNAi strategy for fungal pathogens, such as off-target and epigenetic effects, with their possible solutions are also discussed.
Collapse
|
25
|
Monty MA, Islam MA, Nan X, Tan J, Tuhin IJ, Tang X, Miao M, Wu D, Yu L. Emerging role of RNA interference in immune cells engineering and its therapeutic synergism in immunotherapy. Br J Pharmacol 2021; 178:1741-1755. [PMID: 33608889 DOI: 10.1111/bph.15414] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 02/09/2021] [Accepted: 02/09/2021] [Indexed: 12/12/2022] Open
Abstract
RNAi effectors (e.g. siRNA, shRNA and miRNA) can trigger the silencing of specific genes causing alteration of genomic functions becoming a new therapeutic area for the treatment of infectious diseases, neurodegenerative disorders and cancer. In cancer treatment, RNAi effectors showed potential immunomodulatory actions by down-regulating immuno-suppressive proteins, such as PD-1 and CTLA-4, which restrict immune cell function and present challenges in cancer immunotherapy. Therefore, compared with extracellular targeting by antibodies, RNAi-mediated cell-intrinsic disruption of inhibitory pathways in immune cells could promote an increased anti-tumour immune response. Along with non-viral vectors, DNA-based RNAi strategies might be a more promising method for immunomodulation to silence multiple inhibitory pathways in T cells than immune checkpoint blockade antibodies. Thus, in this review, we discuss diverse RNAi implementation strategies, with recent viral and non-viral mediated RNAi synergism to immunotherapy that augments the anti-tumour immunity. Finally, we provide the current progress of RNAi in clinical pipeline.
Collapse
Affiliation(s)
- Masuma Akter Monty
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Md Ariful Islam
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Xu Nan
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Jingwen Tan
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Israth Jahan Tuhin
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Xiaowen Tang
- The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Miao Miao
- The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Depei Wu
- The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lei Yu
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| |
Collapse
|
26
|
Yang C, Cheng X, Shen P. Silencing of BCSG1 with specific siRNA via nanocarriers for breast cancer treatment. Bull Cancer 2021; 108:323-332. [PMID: 33423781 DOI: 10.1016/j.bulcan.2020.10.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 10/15/2020] [Accepted: 10/29/2020] [Indexed: 01/22/2023]
Abstract
Breast cancer is the most common cancer diagnosed in women worldwide. The current treatments for breast cancer, including surgery, radiotherapy and chemotherapy aim to destroy cancer cells, whereas they also cause damage to normal tissues and cells. Thus, an effective, safe and specific breast cancer treatment is urgently needed. The breast cancer-specific gene 1 (BCSG1) has been shown to be specific for the development of breast cancer and is a target for breast cancer diagnosis and treatment. It is expected to silence the expression of BCSG1 at the gene level for the purpose of treating breast cancer. The effect of RNAi technology on silencing target genes is comparable to gene knockout and has been widely used in animal experiments and plant genetic research. In the field of cancer therapy, numerous investigators have used siRNAs to specifically inhibit target genes, demonstrating that siRNAs can treat cancers at the molecular level. However, the delivery of siRNAs into humans needs to overcome multiple physiological barriers, limiting the clinical applications of siRNAs. This review focuses on the application of BCSG1 gene, siRNAs in cancer treatments, and the nanocarrier delivery system of siRNAs. The potential application and research value of BCSG1-specific siRNA in the treatment of breast cancer are discussed.
Collapse
Affiliation(s)
- Chenbo Yang
- Zhengzhou University, School of Basic Medical Sciences, Zhengzhou, Henan Province 450001, China
| | - Xiaoman Cheng
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Peihong Shen
- The Cancer Hospital Affiliated of Zhengzhou University, Zhengzhou, Henan Province 450008, China.
| |
Collapse
|
27
|
MicroRNAs Regulating Autophagy in Neurodegeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1208:191-264. [PMID: 34260028 DOI: 10.1007/978-981-16-2830-6_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Social and economic impacts of neurodegenerative diseases (NDs) become more prominent in our constantly aging population. Currently, due to the lack of knowledge about the aetiology of most NDs, only symptomatic treatment is available for patients. Hence, researchers and clinicians are in need of solid studies on pathological mechanisms of NDs. Autophagy promotes degradation of pathogenic proteins in NDs, while microRNAs post-transcriptionally regulate multiple signalling networks including autophagy. This chapter will critically discuss current research advancements in the area of microRNAs regulating autophagy in NDs. Moreover, we will introduce basic strategies and techniques used in microRNA research. Delineation of the mechanisms contributing to NDs will result in development of better approaches for their early diagnosis and effective treatment.
Collapse
|
28
|
Zhang S, Gan Y, Shao L, Liu T, Wei D, Yu Y, Guo H, Zhu H. Virus Mimetic Shell-Sheddable Chitosan Micelles for siVEGF Delivery and FRET-Traceable Acid-Triggered Release. ACS APPLIED MATERIALS & INTERFACES 2020; 12:53598-53614. [PMID: 33201664 DOI: 10.1021/acsami.0c13023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Targeting vascular endothelial growth factor (VEGF) using small interfering RNA (siVEGF) has shown great potential in inhibiting the growth, proliferation, and migration of tumors by reducing the proliferation of blood vessels. On the basis of bionic principles, a novel pH-responsive and virus mimetic shell-sheddable chitosan (CS) micelles (CMs) as siRNA delivery system was introduced in this study. The cyclo(Arg-Gly-Asp-d-Phe-Lys) (cRGD) modified poly(enthylene glycol) (PEG) was conjugated to the HA2 modified chitosan via a hydrazone linkage (cRGD-PEG-Hz-CS-HA2). The cRGD-PEG-Hz-CS-HA2 conjugate could form micelles by interacting with the complex of octanal, Boc-l-lysine, and 9-d-arginine (9R) (octyl-Lys-9R) as a hydrophodic core forming agent, termed as cRGD-PEG-Hz-CS-HA2/octyl-Lys-9R (abbreviated as cRGD/HA2/Hz-CMs).The CMs modified with cRGD can accurately target glioma cells (U87MG cells) with high expression of αvβ3. The payloads of siVEGF were packed into the core of cRGD/HA2/Hz-CMs via electrostatic interaction and hydrophobic interaction. The intracellular cargo release was achieved by the pH-responsive lysis of the hydrazone bond in acidic environment of endosome. Moreover, the exposed HA2, as a pH-sensitive membrane-disruptive peptide, assists the escape of the carriers from endosome into cytosol. In addition, cRGD/HA2/Hz-CMs can effectively deliver siVEGF and silence VEGF gene expression in U87MG cells, leading to the significant tumor growth inhibition. This study demonstrates that cRGD/HA2/Hz-CMs can deliver and release siVEGF in a controlled manner, which was traced by the fluorescence resonance energy transfer (FRET) system in order to achieve RNAi-based anti-angiogenic treatment of cancer in vivo.
Collapse
Affiliation(s)
- Shengyu Zhang
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
- Department of Pharmacy, Haimen People's Hospital, Nantong 226100, China
| | - Ye Gan
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Lanlan Shao
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Tianqing Liu
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 1006, Australia
| | - Danyi Wei
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Yanyan Yu
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Hongwei Guo
- College of Pharmacy, Guangxi Medical University, Nanning 530021, China
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education &Center for Translational Medicine, Guangxi Medical University, Nanning 530021, China
| | - Hongyan Zhu
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| |
Collapse
|
29
|
|
30
|
Khan A, Aljarbou AN, Aldebasi YH, Allemailem KS, Alsahli MA, Khan S, Alruwetei AM, Khan MA. Fatty Acid Synthase (FASN) siRNA-Encapsulated-Her-2 Targeted Fab'-Immunoliposomes for Gene Silencing in Breast Cancer Cells. Int J Nanomedicine 2020; 15:5575-5589. [PMID: 32801705 PMCID: PMC7415462 DOI: 10.2147/ijn.s256022] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 06/29/2020] [Indexed: 12/11/2022] Open
Abstract
Purpose The overexpression of Her-2 in 25–30% breast cancer cases and the crosstalk between Her-2 and fatty acid synthase (FASN) establishes Her-2 as a promising target for site-directed delivery. The present study aimed to develop the novel lipid base formulations to target and inhibit the cellular proliferation of Her-2-expressing breast cancer cells through the silencing of FASN. In order to achieve this goal, we prepared DSPC/Chol and DOPE/CHEMS immunoliposomes, conjugated with the anti-Her-2 fab’ and encapsulated FASN siRNA against breast cancer cells. Methods We evaluated the size, stability, cellular uptake and internalization of various formulations of liposomes. The antiproliferative gene silencing potential was investigated by the cell cytotoxicity, crystal violet, wound healing and Western blot analyses in Her-2+ and Her-2¯ breast cancer cells. Results The data revealed that both nanosized FASN-siRNA-encapsulated liposomes showed significantly higher cellular uptake and internalization with enhanced stability. The cell viability of Her-2+ SK-BR3 cells treated with the targeted formulation of DSPC/Chol- and DOPE/CHEMS-encapsulating FASN-siRNA reduced to 30% and 20%, respectively, whereas it was found to be 45% and 36% in MCF-7 cells. The wounds were not only failed to close but they became broader in Her-2+ cells treated with targeted liposomes of siRNA. Consequently, the amount of FASN decreased by 80% in SK-BR3 cells treated with non-targeted liposomes and it was 30% and 60% in the MCF-7 cells treated with DSPC/Chol and DOPE/CHEMS liposomes, respectively. Conclusion In this study, we developed the formulation that targeted Her-2 for the suppression of FASN and, therefore, inhibited the proliferation of breast cancer cells.
Collapse
Affiliation(s)
- Arif Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Al-Qassim, Buraydah, Saudi Arabia
| | - Ahmed N Aljarbou
- Department of Pharmaceutics, College of Pharmacy, Qassim University, Al-Qassim, Buraydah, Saudi Arabia
| | - Yousef H Aldebasi
- Department of Optometry, College of Applied Medical Sciences, Qassim University, Al-Qassim, Buraydah, Saudi Arabia
| | - Khaled S Allemailem
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Al-Qassim, Buraydah, Saudi Arabia
| | - Mohammed A Alsahli
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Al-Qassim, Buraydah, Saudi Arabia
| | - Shamshir Khan
- Dentistry and Pharmacy College, Buraydah Private Colleges, Al-Qassim, Buraydah, Saudi Arabia
| | - Abdulmohsen M Alruwetei
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Al-Qassim, Buraydah, Saudi Arabia
| | - Masood A Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Al-Qassim, Buraydah, Saudi Arabia
| |
Collapse
|
31
|
Chuang CC, Chen YN, Wang YY, Huang YC, Lin SY, Huang RY, Jang YY, Yang CC, Huang YF, Chang CW. Stem Cell-Based Delivery of Gold/Chlorin e6 Nanocomplexes for Combined Photothermal and Photodynamic Therapy. ACS APPLIED MATERIALS & INTERFACES 2020; 12:30021-30030. [PMID: 32594734 DOI: 10.1021/acsami.0c03446] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Combining photothermal and photodynamic modalities has shown encouraging therapeutic efficacy against various malignant cancers. Developing a delivery method for targeting and penetrating tumors is still a major focus for advancing this therapeutic approach. Herein, we report a novel strategy involving the utilization of stem cells as a live carrier to codeliver photothermal and photodynamic agents for cancer therapy. To this end, a novel gold nanorod (AuNR)-PEG-PEI (APP)/chlorin e6 (Ce6)-loaded adipose-derived stem cell (ADSC) system is proposed in which AuNRs and Ce6 act as the photothermal and photodynamic agents, respectively. To integrate with stem cells, the APP/Ce6 nanocomplexes exhibit advantages of low drug leakage, low cytotoxicity, efficient cellular uptake, and redox-responsive release. After loading of APP/Ce6 nanocomplexes, the ADSCs still maintained good tumor tropism and were capable of penetrating into the tumor spheroids. The photothermal effect induced by exposure to near-infrared light irradiation at 808 nm promoted the release of Ce6 from the stem cells into the surroundings and hence increased its availability to treat cancer cells. APP/Ce6-loaded ADSCs exerted effective dose-dependent in vitro anticancer activities via anticipated photothermal and photodynamic effects. In a murine CT26 colon cancer model, APP/Ce6 delivered by ADSCs resulted in superior tumor suppression compared to other delivery strategies. It was also noted that in vivo applications of APP/Ce6-loaded ADSCs did not induce noticeable detrimental effects on normal tissues/organs.
Collapse
Affiliation(s)
- Chun-Chiao Chuang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan R.O.C
| | - Yi-Ning Chen
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan R.O.C
| | - Yi-Ya Wang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan R.O.C
| | - Yu-Chen Huang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan R.O.C
| | - Ssu-Yu Lin
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan R.O.C
| | - Rih-Yang Huang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan R.O.C
| | - Yu-Yun Jang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan R.O.C
| | - Chun-Chi Yang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan R.O.C
| | - Yu-Fen Huang
- Institute of Analytical and Environmental Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan R.O.C
| | - Chien-Wen Chang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan R.O.C
| |
Collapse
|
32
|
Pereira-Silva M, Jarak I, Alvarez-Lorenzo C, Concheiro A, Santos AC, Veiga F, Figueiras A. Micelleplexes as nucleic acid delivery systems for cancer-targeted therapies. J Control Release 2020; 323:442-462. [DOI: 10.1016/j.jconrel.2020.04.041] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 02/09/2023]
|
33
|
Yin L, Zhao F, Sun H, Wang Z, Huang Y, Zhu W, Xu F, Mei S, Liu X, Zhang D, Wei L, Cen S, Hu S, Liang C, Guo F. CRISPR-Cas13a Inhibits HIV-1 Infection. MOLECULAR THERAPY-NUCLEIC ACIDS 2020; 21:147-155. [PMID: 32585623 PMCID: PMC7321785 DOI: 10.1016/j.omtn.2020.05.030] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/15/2020] [Accepted: 05/26/2020] [Indexed: 12/26/2022]
Abstract
CRISPR-Cas provides bacteria and archaea with immunity against invading phages and foreign plasmid DNA and has been successfully adapted for gene editing in a variety of species. The class 2 type VI CRISPR-Cas effector Cas13a targets and cleaves RNA, providing protection against RNA phages. Here we report the repurposing of CRISPR-Cas13a to inhibit human immunodeficiency virus type 1 (HIV-1) infection through targeting HIV-1 RNA and diminishing viral gene expression. We observed strong inhibition of HIV-1 infection by CRISPR-Cas13a in human cells. We showed that CRISPR-Cas13a not only diminishes the level of newly synthesized viral RNA, either from the transfected plasmid DNA or from the viral DNA, which is integrated into cellular DNA, but it also targets and destroys the viral RNA that enters cells within viral capsid, leading to strong inhibition of HIV-1 infection. Together, our results suggest that CRISPR-Cas13a provides a potential novel tool to treat viral diseases in humans.
Collapse
Affiliation(s)
- Lijuan Yin
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, and Center for AIDS Research, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, P.R. China
| | - Fei Zhao
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, and Center for AIDS Research, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, P.R. China
| | - Hong Sun
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, and Center for AIDS Research, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, P.R. China
| | - Zhen Wang
- McGill University AIDS Centre, Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - Yu Huang
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, and Center for AIDS Research, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, P.R. China
| | - Weijun Zhu
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, and Center for AIDS Research, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, P.R. China
| | - Fengwen Xu
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, and Center for AIDS Research, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, P.R. China
| | - Shan Mei
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, and Center for AIDS Research, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, P.R. China
| | - Xiaoman Liu
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, and Center for AIDS Research, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, P.R. China
| | - Di Zhang
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, and Center for AIDS Research, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, P.R. China
| | - Liang Wei
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, and Center for AIDS Research, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, P.R. China
| | - Shan Cen
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, P.R. China
| | - Siqi Hu
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, and Center for AIDS Research, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, P.R. China.
| | - Chen Liang
- McGill University AIDS Centre, Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada.
| | - Fei Guo
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, and Center for AIDS Research, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, P.R. China.
| |
Collapse
|
34
|
Feldmann DP, Heyza J, Zimmermann CM, Patrick SM, Merkel OM. Nanoparticle-Mediated Gene Silencing for Sensitization of Lung Cancer to Cisplatin Therapy. Molecules 2020; 25:molecules25081994. [PMID: 32344513 PMCID: PMC7221615 DOI: 10.3390/molecules25081994] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 04/16/2020] [Accepted: 04/17/2020] [Indexed: 12/17/2022] Open
Abstract
Platinum-based chemotherapy remains a mainstay treatment for the management of advanced non-small cell lung cancer. A key cellular factor that contributes to sensitivity to platinums is the 5'-3' structure-specific endonuclease excision repair cross-complementation group 1 (ERCC1)/ xeroderma pigmentosum group F (XPF). ERCC1/XPF is critical for the repair of platinum-induced DNA damage and has been the subject of intense research efforts to identify small molecule inhibitors of its nuclease activity for the purpose of enhancing patient response to platinum-based chemotherapy. As an alternative to small molecule inhibitors, small interfering RNA (siRNA) has often been described to be more efficient in interrupting protein-protein interactions. The goal of this study was therefore to determine whether biocompatible nanoparticles consisting of an amphiphilic triblock copolymer (polyethylenimine-polycaprolactone-polyethylene glycol (PEI-PCL-PEG)) and carrying siRNA targeted to ERCC1 and XPF made by microfluidic assembly are capable of efficient gene silencing and able to sensitize lung cancer cells to cisplatin. First, we show that our PEI-PCL-PEG micelleplexes carrying ERCC1 and XPF siRNA efficiently knocked down ERCC1/XPF protein expression to the same extent as the standard siRNA transfection reagent, Lipofectamine. Second, we show that our siRNA-carrying nanoparticles enhanced platinum sensitivity in a p53 wildtype model of non-small cell lung cancer in vitro. Our results suggest that nanoparticle-mediated targeting of ERCC1/XPF is feasible and could represent a novel therapeutic strategy for targeting ERCC1/XPF in vivo.
Collapse
Affiliation(s)
- Daniel P. Feldmann
- Department of Oncology, School of Medicine and Barbara Ann Karmanos Institute, Wayne State University, Detroit, MI 48201, USA; (D.P.F.); (J.H.); (S.M.P.)
- Department of Pharmaceutical Sciences, School of Pharmacy, Wayne State University, Detroit, MI 48201, USA
| | - Joshua Heyza
- Department of Oncology, School of Medicine and Barbara Ann Karmanos Institute, Wayne State University, Detroit, MI 48201, USA; (D.P.F.); (J.H.); (S.M.P.)
| | | | - Steve M. Patrick
- Department of Oncology, School of Medicine and Barbara Ann Karmanos Institute, Wayne State University, Detroit, MI 48201, USA; (D.P.F.); (J.H.); (S.M.P.)
| | - Olivia M. Merkel
- Department of Oncology, School of Medicine and Barbara Ann Karmanos Institute, Wayne State University, Detroit, MI 48201, USA; (D.P.F.); (J.H.); (S.M.P.)
- Department of Pharmaceutical Sciences, School of Pharmacy, Wayne State University, Detroit, MI 48201, USA
- Department of Pharmacy, Ludwig-Maximilians-Universität München, 81377 München, Germany;
- Correspondence:
| |
Collapse
|
35
|
Cao M, Gao Y, Qiu N, Shen Y, Shen P. Folic acid directly modified low molecular weight of polyethyleneimine for targeted pDNA delivery. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101522] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
36
|
Liu S, Jaouannet M, Dempsey DA, Imani J, Coustau C, Kogel KH. RNA-based technologies for insect control in plant production. Biotechnol Adv 2020; 39:107463. [DOI: 10.1016/j.biotechadv.2019.107463] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 10/24/2019] [Accepted: 10/26/2019] [Indexed: 12/23/2022]
|
37
|
Park JH, Seo JH, Jeon HY, Seo SM, Lee HK, Park JI, Kim JY, Choi YK. Lentivirus-Mediated VEGF Knockdown Suppresses Gastric Cancer Cell Proliferation and Tumor Growth in vitro and in vivo. Onco Targets Ther 2020; 13:1331-1341. [PMID: 32104000 PMCID: PMC7025738 DOI: 10.2147/ott.s234344] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 02/02/2020] [Indexed: 01/17/2023] Open
Abstract
Purpose Gastric cancer has a high mortality rate worldwide. Although treatments, such as molecular-targeted therapy, have been introduced, the resulting long-term survival and prognosis remain unsatisfactory. Downregulation of the target genes using lentivirus-mediated short hairpin RNA (shRNA) can be an effective therapeutic strategy for patients with gastric cancer. Overexpressed vascular endothelial growth factor A (VEGF) in human gastric cancer cells can be an effective novel therapeutic target for human gastric cancer. Thus, this study aimed to evaluate the therapeutic effects of lentivirus-mediated knockdown of VEGF gene expression in human gastric cancer growth. Materials and Methods Specific shRNA sequences targeting VEGF were designed to construct a lentiviral expression vector. After human gastric carcinoma cells (cell line NCI-N87) were infected with the lentiviral vector, the therapeutic effects of the lentivirus-mediated shRNA targeting VEGF were analyzed both in vitro and in vivo. Results Stable suppression of VEGF gene expression in NCI-N87 cells using shRNA (ShVEGF) showed significant inhibition of cell proliferation, clonogenicity, and cell motility. ShVEGF also showed increased G0/G1 cell cycle arrest and apoptosis. In addition, in vivo results from nude mice xenografted ShVEGF showed significant inhibition of tumor growth. Assessing the therapeutic effects of intratumoral injection of lentivirus-targeting VEGF (Virus_VEGF) revealed that it significantly inhibited tumor growth compared to that in the Virus_Scramble or saline injection control groups. Conclusion The constructed ShVEGF showed significant inhibition of NCI-N87 gastric cancer cell growth both in vitro and in vivo. These experimental results suggest a novel therapeutic strategy for patients with gastric cancer using lentivirus-mediated shRNA targeting VEGF.
Collapse
Affiliation(s)
- Jong-Hyung Park
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea.,Helixmith Co. Ltd., Seoul 08826, Republic of Korea
| | - Jin-Hee Seo
- Laboratory Animal Center, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea
| | - Hee-Yeon Jeon
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea.,Department of Core Research Laboratory, Clinical Research Institute, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea
| | - Sun-Min Seo
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Han-Kyul Lee
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Jin-Il Park
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea.,Helixmith Co. Ltd., Seoul 08826, Republic of Korea
| | - Jun-Young Kim
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Yang-Kyu Choi
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
38
|
Lennox KA, Behlke MA. Chemical Modifications in RNA Interference and CRISPR/Cas Genome Editing Reagents. Methods Mol Biol 2020; 2115:23-55. [PMID: 32006393 DOI: 10.1007/978-1-0716-0290-4_2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Chemically modified oligonucleotides (ONs) are routinely used in the laboratory to assess gene function, and clinical advances are rapidly progressing as continual efforts are being made to optimize ON efficacy. Over the years, RNA interference (RNAi) has become one of the main tools used to inhibit RNA expression across a wide variety of species. Efforts have been made to improve the exogenous delivery of the double-stranded RNA components to the endogenous intracellular RNAi machinery to direct efficacious degradation of a user-defined RNA target. More recently, synthetic RNA ONs are being used to mimic the bacterial-derived CRISPR/Cas system to direct specific editing of the mammalian genome. Both of these techniques rely on the use of various chemical modifications to the RNA phosphate backbone or sugar in specific positions throughout the ONs to improve the desired biological outcome. Relevant chemical modifications also include conjugated targeting ligands to assist ON delivery to specific cell types. Chemical modifications are most beneficial for therapeutically relevant ONs, as they serve to enhance target binding, increase drug longevity, facilitate cell-specific targeting, improve internalization into productive intracellular compartments, and mitigate both sequence-specific as well as immune-related off-target effects (OTEs). The knowledge gained from years of optimizing RNAi reagents and characterizing the biochemical and biophysical properties of each chemical modification will hopefully accelerate the CRISPR/Cas technology into the clinic, as well as further expand the use of RNAi to treat currently undruggable diseases. This review discusses the most commonly employed chemical modifications in RNAi reagents and CRISPR/Cas guide RNAs and provides an overview of select publications that have demonstrated success in improving ON efficacy and/or mitigating undesired OTEs.
Collapse
Affiliation(s)
- Kim A Lennox
- Integrated DNA Technologies, Inc., Coralville, IA, USA.
| | - Mark A Behlke
- Integrated DNA Technologies, Inc., Coralville, IA, USA
| |
Collapse
|
39
|
Abstract
RNA interference (RNAi), a natural gene silencing process, is a widely used technique in basic research, preclinical studies, and drug development strategies. Although the technique has great potential to generate new human therapies and treat undruggable diseases, the clinical application of RNAi is still challenging primarily because of the delivery problem and potential off-target effects. Over the past two decades, great efforts have been undertaken to develop delivery agents and chemical modifications to overcome these challenges. Such advances in RNA delivery and chemical modifications have benefited researchers who are developing gene-editing therapies based on CRISPR-Cas9, an RNA-guided endonuclease, which is already having a major impact on biology and medicine. Here, I review the discovery of these two interference tools, identify the technical challenges yet to be overcome and provide some perspectives on how these two RNA-based technologies can be harnessed to treat human diseases.
Collapse
Affiliation(s)
- Mouldy Sioud
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway.
| |
Collapse
|
40
|
Zins K, Abraham D. Cancer Immunotherapy: Targeting Tumor-Associated Macrophages by Gene Silencing. Methods Mol Biol 2020; 2115:289-325. [PMID: 32006408 DOI: 10.1007/978-1-0716-0290-4_17] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Tumor-associated macrophages (TAMs) are representing a major leukocyte population in solid tumors. Macrophages are very heterogeneous and plastic cells and can acquire distinct functional phenotypes ranging from antitumorigenic to immunosuppressive tumor-promoting M2-like TAMs, depending on the local tissue microenvironment (TME). TAMs express cytokines, chemokines, growth factors, and extracellular matrix (ECM) modifying factors, and the cross talk with the TME regulates pathways involved in the recruitment, polarization, and metabolism of TAMs during tumor progression. Due to their crucial role in tumor growth and metastasis, selective targeting of TAM for the treatment of cancer with therapeutic agents that promote phagocytosis or suppress survival, proliferation, trafficking, or polarization of TAMs may prove to be beneficial in cancer therapy. In this chapter, we will discuss TAM biology and current strategies for the targeting of TAMs using small interfering RNA (siRNA)-based drugs. In the past few years, advances in the field of nanomedicine pave the way for the development of siRNA-based drugs as an additional class of personalized cancer immuno-nanomedicines. Fundamental challenges associated with this group of therapeutics include the development process, delivery system, and clinical translation for siRNA-based drugs.
Collapse
Affiliation(s)
- Karin Zins
- Division of Cell and Developmental Biology, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Dietmar Abraham
- Division of Cell and Developmental Biology, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
41
|
Improving the therapeutic efficiency of noncoding RNAs in cancers using targeted drug delivery systems. Drug Discov Today 2019; 25:718-730. [PMID: 31758914 DOI: 10.1016/j.drudis.2019.11.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 11/01/2019] [Accepted: 11/13/2019] [Indexed: 12/11/2022]
Abstract
The delivery of noncoding (nc)RNA to target cancer stem cells and metastatic tumors has shown many positive outcomes, resulting in improved and more efficient treatment strategies. The success of therapeutic RNA depends solely on passing cellular barriers to reach the target site, where it binds to the mRNA of the interest. By 2018, 20 clinical trials had been initiated, most focusing on cancer and diabetes, with some progressing to Phase II clinical trials testing the safety and efficacy of small interfering (si)RNA. Many challenges limit RNA interference (RNAi) and miRNA usage in vivo; therefore, various approaches have been developed to promote ncRNA efficiency and stability. In this review, we focus on targeting the tumor microenvironment (TME) via the modification of delivery systems utilizing nanotechnology-based delivery approaches.
Collapse
|
42
|
Hossian AKMN, Mackenzie GG, Mattheolabakis G. miRNAs in gastrointestinal diseases: can we effectively deliver RNA-based therapeutics orally? Nanomedicine (Lond) 2019; 14:2873-2889. [PMID: 31735124 DOI: 10.2217/nnm-2019-0180] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Nucleic acid-based therapeutics are evaluated for their potential of treating a plethora of diseases, including cancer and inflammation. Short nucleic acids, such as miRNAs, have emerged as versatile regulators for gene expression and are studied for therapeutic purposes. However, their inherent instability in vivo following enteral and parenteral administration has prompted the development of novel methodologies for their delivery. Although research on the oral delivery of siRNAs is progressing, with the development and utilization of promising carrier-based methodologies for the treatment of a plethora of gastrointestinal diseases, research on miRNA-based oral therapeutics is lagging behind. In this review, we present the potential role of miRNAs in diseases of the GI tract, and analyze current research and the cardinal features of the novel carrier systems used for nucleic acid oral delivery that can be expanded for oral miRNA administration.
Collapse
Affiliation(s)
- A K M Nawshad Hossian
- School of Basic Pharmaceutical & Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA 71201, USA
| | | | - George Mattheolabakis
- School of Basic Pharmaceutical & Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA 71201, USA
| |
Collapse
|
43
|
Cao M, Gao Y, Zhan M, Qiu N, Piao Y, Zhou Z, Shen Y. Glycyrrhizin Acid and Glycyrrhetinic Acid Modified Polyethyleneimine for Targeted DNA Delivery to Hepatocellular Carcinoma. Int J Mol Sci 2019; 20:E5074. [PMID: 31614879 PMCID: PMC6829341 DOI: 10.3390/ijms20205074] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/09/2019] [Accepted: 10/10/2019] [Indexed: 12/22/2022] Open
Abstract
In the last 2-3 decades, gene therapy represented a promising option for hepatocellular carcinoma (HCC) treatment. However, the design of safe and efficient gene delivery systems is still one of the major challenges that require solutions. In this study, we demonstrate a versatile method for covalent conjugation of glycyrrhizin acid (GL) or glycyrrhetinic acid (GA) to increase the transfection efficiency of Polyethyleneimine (PEI, Mw 1.8K) and improve their targeting abilities of hepatoma carcinoma cells. GA and GL targeting ligands were grafted to PEI via N-acylation, and we systematically investigated their biophysical properties, cytotoxicity, liver targeting and transfection efficiency, and endocytosis pathway trafficking. PEI-GA0.75, PEI-GL10.62 and PEI-GL20.65 conjugates caused significant increases in gene transfection efficiency and superior selectivity for HepG2 cells, with all three conjugates showing specific recognition of HepG2 cells by the free GA competition assay. The endocytosis inhibition and intracellular trafficking results indicated that PEI-GA0.75 and GL10.62 conjugates behaved similarly to SV40 virus, by proceeding via the caveolae- and clathrin-independent mediated endocytosis pathway and bypassing entry into lysosomes, with an energy independent manner, achieving their high transfection efficiencies. In the HepG2 intraperitoneal tumor model, PEI-GA0.75 and PEI-GL10.62 carrying the luciferase reporter gene gained high gene expression, suggesting potential use for in vivo application.
Collapse
Affiliation(s)
- Mingzhuo Cao
- Center for Bio-nanoengineering and Key Laboratory of Biomass Chemical Engineering, Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China.
- Scientific Research and Experimental Center, Henan University of Chinese Medicine, Zhengzhou 450058, China.
| | - Yong Gao
- Henan province food and drug Administration, Food and Drug Evaluation and Inspection Center, Zhengzhou 450018, China.
| | - Mengling Zhan
- Scientific Research and Experimental Center, Henan University of Chinese Medicine, Zhengzhou 450058, China.
| | - Nasha Qiu
- Center for Bio-nanoengineering and Key Laboratory of Biomass Chemical Engineering, Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China.
| | - Ying Piao
- Center for Bio-nanoengineering and Key Laboratory of Biomass Chemical Engineering, Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China.
| | - Zhuxian Zhou
- Center for Bio-nanoengineering and Key Laboratory of Biomass Chemical Engineering, Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China.
| | - Youqing Shen
- Center for Bio-nanoengineering and Key Laboratory of Biomass Chemical Engineering, Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China.
| |
Collapse
|
44
|
Dua K, Wadhwa R, Singhvi G, Rapalli V, Shukla SD, Shastri MD, Gupta G, Satija S, Mehta M, Khurana N, Awasthi R, Maurya PK, Thangavelu L, S R, Tambuwala MM, Collet T, Hansbro PM, Chellappan DK. The potential of siRNA based drug delivery in respiratory disorders: Recent advances and progress. Drug Dev Res 2019; 80:714-730. [DOI: 10.1002/ddr.21571] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/11/2019] [Accepted: 05/21/2019] [Indexed: 12/24/2022]
Affiliation(s)
- Kamal Dua
- Discipline of Pharmacy, Graduate School of HealthUniversity of Technology Sydney Ultimo New South Wales Australia
- Centenary InstituteRoyal Prince Alfred Hospital Camperdown New South Wales Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI) and School of Biomedical Sciences and PharmacyUniversity of Newcastle Callaghan New South Wales Australia
| | - Ridhima Wadhwa
- Faculty of Life Sciences and BiotechnologySouth Asian University New Delhi India
| | - Gautam Singhvi
- Department of PharmacyBirla Institute of Technology and Science (BITS) Pilani India
| | | | - Shakti Dhar Shukla
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI) and School of Biomedical Sciences and PharmacyUniversity of Newcastle Callaghan New South Wales Australia
| | - Madhur D. Shastri
- School of Health Sciences, College of Health and MedicineUniversity of Tasmania Launceston Australia
| | - Gaurav Gupta
- School of PharmacySuresh Gyan Vihar University Jaipur India
| | - Saurabh Satija
- School of Pharmaceutical SciencesLovely Professional University Phagwara Punjab India
| | - Meenu Mehta
- School of Pharmaceutical SciencesLovely Professional University Phagwara Punjab India
| | - Navneet Khurana
- School of Pharmaceutical SciencesLovely Professional University Phagwara Punjab India
| | - Rajendra Awasthi
- Amity Institute of PharmacyAmity University Noida Uttar Pradesh India
| | - Pawan Kumar Maurya
- Department of BiochemistryCentral University of Haryana Mahendergarh Haryana India
| | - Lakshmi Thangavelu
- Nanobiomedicine Lab, Department of Pharmacology, Saveetha Dental CollegeSaveetha Institute of Medical and Technical Sciences Chennai Tamil Nadu India
| | - Rajeshkumar S
- Nanobiomedicine Lab, Department of Pharmacology, Saveetha Dental CollegeSaveetha Institute of Medical and Technical Sciences Chennai Tamil Nadu India
| | - Murtaza M. Tambuwala
- School of Pharmacy and Pharmaceutical SciencesUlster University, Coleraine London United Kingdom of Great Britain and Northern Ireland
| | - Trudi Collet
- Inovative Medicines Group, Institute of Health and Biomedical InnovationQueensland University of Technology Brisbane Queensland Australia
| | - Philip M. Hansbro
- Centenary InstituteRoyal Prince Alfred Hospital Camperdown New South Wales Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI) and School of Biomedical Sciences and PharmacyUniversity of Newcastle Callaghan New South Wales Australia
- School of Life SciencesUniversity of Technology Sydney Sydney New South Wales Australia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of PharmacyInternational Medical University Kuala Lumpur Malaysia
| |
Collapse
|
45
|
The Impact of Nylon-3 Copolymer Composition on the Efficiency of siRNA Delivery to Glioblastoma Cells. NANOMATERIALS 2019; 9:nano9070986. [PMID: 31288448 PMCID: PMC6669510 DOI: 10.3390/nano9070986] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 06/28/2019] [Accepted: 07/01/2019] [Indexed: 02/06/2023]
Abstract
Glioblastoma multiforme is a devastating disease that has attracted enormous attention due to poor prognosis and high recurrence. Small interfering RNA (siRNA) in principle offers a promising therapeutic approach by the downregulation of disease-related genes via RNA interference. For efficient siRNA delivery to target sites, cationic polymers are often used in preclinical studies for the protection of siRNA and complex formation based on electrostatic interactions. In an effort to develop biocompatible and efficient nanocarriers with a translational outlook for optimal gene silencing at reduced toxicity, we synthesized two sets of nylon-3 copolymers with variable cationic content (DM or NM monomer) and hydrophobic subunits (CP monomer) and evaluated their suitability for in vitro siRNA delivery into glioblastoma cells. DM0.4/CP0.6 and NM0.4/CP0.6 polymers with similar subunit ratios were synthesized to compare the effect of different cationic subunits. Additionally, we utilized NM0.2/CP0.8 polymers to evaluate the impact of the different hydrophobic content in the polymer chain. The siRNA condensation ability and polymer–siRNA complex stability was evaluated by unmodified and modified SYBR gold assays, respectively. Further physicochemical characteristics, e.g., particle size and surface charge, were evaluated by dynamic light scattering and laser Doppler anemometry, whereas a relatively new method for polyplex size distribution analysis—tunable resistive pulse sensing—was additionally developed and compared to DLS measurements. Transfection efficiencies, the route of cell internalization, and protein knockdown abilities in glioblastoma cells were investigated by flow cytometry. Furthermore, cellular tolerability was evaluated by MTT and LDH assays. All the polymers efficiently condensed siRNA at N/P ratios of three, whereas polymers with NM cationic subunits demonstrated smaller particle size and lower polyplex stability. Furthermore, NM0.2/CP0.8 polyplexes with the highest hydrophobic content displayed significantly higher cellular internalization in comparison to more cationic formulations and successful knockdown capabilities. Detailed investigations of the cellular uptake route demonstrated that these polyplexes mainly follow clathrin-mediated endocytotic uptake mechanisms, implying high interaction capacity with cellular membranes. Taken together with conducive toxicity profiles, highly hydrophobic nylon-3 polymers provide an appropriate siRNA delivery agent for the potential treatment of glioblastoma.
Collapse
|
46
|
Mahmoodi Chalbatani G, Dana H, Gharagouzloo E, Grijalvo S, Eritja R, Logsdon CD, Memari F, Miri SR, Rad MR, Marmari V. Small interfering RNAs (siRNAs) in cancer therapy: a nano-based approach. Int J Nanomedicine 2019; 14:3111-3128. [PMID: 31118626 PMCID: PMC6504672 DOI: 10.2147/ijn.s200253] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 03/23/2019] [Indexed: 12/31/2022] Open
Abstract
Cancer is one of the most complex diseases that has resulted in multiple genetic disorders and cellular abnormalities. Globally, cancer is the most common health concern disease that is affecting human beings. Great efforts have been made over the past decades in biology with the aim of searching novel and more efficient tools in therapy. Thus, small interfering RNAs (siRNAs) have been considered one of the most noteworthy developments which are able to regulate gene expression following a process known as RNA interference (RNAi). RNAi is a post-transcriptional mechanism that involves the inhibition of gene expression through promoting cleavage on a specific area of a target messenger RNA (mRNA). This technology has shown promising therapeutic results for a good number of diseases, especially in cancer. However, siRNA therapeutics have to face important drawbacks in therapy including stability and successful siRNA delivery in vivo. In this regard, the development of effective siRNA delivery systems has helped addressing these issues by opening novel therapeutic windows which have allowed to build up important advances in Nanomedicine. In this review, we discuss the progress of siRNA therapy as well as its medical application via nanoparticle-mediated delivery for cancer treatment.
Collapse
Affiliation(s)
| | - Hassan Dana
- Cancer Research Center, Cancer Institute of Iran, Tehran University of Medical Science, Tehran, Iran
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Elahe Gharagouzloo
- Cancer Research Center, Cancer Institute of Iran, Tehran University of Medical Science, Tehran, Iran
| | - Santiago Grijalvo
- Institute of Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona08034, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER BBN), Madrid, Spain
| | - Ramon Eritja
- Institute of Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona08034, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER BBN), Madrid, Spain
| | - Craig D Logsdon
- Department of Cancer Biology, University of Texas, M.D. Anderson Cancer Center, Houston, TX, USA
- Department of GI Medical Oncology, University of Texas, M.D. Anderson Cancer Center, Houston, TX, USA
| | - Fereidoon Memari
- Cancer Research Center, Cancer Institute of Iran, Tehran University of Medical Science, Tehran, Iran
| | - Seyed Rouhollah Miri
- Cancer Research Center, Cancer Institute of Iran, Tehran University of Medical Science, Tehran, Iran
| | | | - Vahid Marmari
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| |
Collapse
|
47
|
Singh S, Maurya PK. Nanomaterials-Based siRNA Delivery: Routes of Administration, Hurdles and Role of Nanocarriers. NANOTECHNOLOGY IN MODERN ANIMAL BIOTECHNOLOGY 2019. [PMCID: PMC7121101 DOI: 10.1007/978-981-13-6004-6_3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
Abstract
Ribonucleic acid interference (RNAi) is a potential alternative therapeutic approach to knock down the overexpression of genes in several disorders especially cancers with underlying genetic dysfunctions. For silencing of specific genes involved in cell cycle, small/short interfering ribonucleic acids (siRNAs) are being used clinically. The siRNA-based RNAi is more efficient, specific and safe antisense technology than other RNAi approaches. The route of siRNA administration for siRNA therapy depends on the targeted site. However, certain hurdles like poor stability of siRNA, saturation, off-target effect, immunogenicity, anatomical barriers and non-targeted delivery restrict the successful siRNA therapy. Thus, advancement of an effective, secure, and long-term delivery system is prerequisite to the medical utilization of siRNA. Polycationic nanocarriers mediated targeted delivery system is an ideal system to remove these hurdles and to increase the blood retention time and rate of intracellular permeability. In this chapter, we will mainly discuss the different biocompatible, biodegradable, non-toxic (organic, inorganic and hybrid) nanocarriers that encapsulate and shield the siRNA from the different harsh environment and provides the increased systemic siRNA delivery.
Collapse
Affiliation(s)
- Sanjay Singh
- Division of Biological and Life Sciences, Ahmedabad University, Ahmedabad, Gujarat India
| | | |
Collapse
|
48
|
Raja MAG, Katas H, Amjad MW. Design, mechanism, delivery and therapeutics of canonical and Dicer-substrate siRNA. Asian J Pharm Sci 2019; 14:497-510. [PMID: 32104477 PMCID: PMC7032099 DOI: 10.1016/j.ajps.2018.12.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 12/07/2018] [Accepted: 12/24/2018] [Indexed: 12/12/2022] Open
Abstract
Upon the discovery of RNA interference (RNAi), canonical small interfering RNA (siRNA) has been recognized to trigger sequence-specific gene silencing. Despite the benefits of siRNAs as potential new drugs, there are obstacles still to be overcome, including off-target effects and immune stimulation. More recently, Dicer substrate siRNA (DsiRNA) has been introduced as an alternative to siRNA. Similarly, it also is proving to be potent and target-specific, while rendering less immune stimulation. DsiRNA is 25–30 nucleotides in length, and is further cleaved and processed by the Dicer enzyme. As with siRNA, it is crucial to design and develop a stable, safe, and efficient system for the delivery of DsiRNA into the cytoplasm of targeted cells. Several polymeric nanoparticle systems have been well established to load DsiRNA for in vitro and in vivo delivery, thereby overcoming a major hurdle in the therapeutic uses of DsiRNA. The present review focuses on a comparison of siRNA and DsiRNA on the basis of their design, mechanism, in vitro and in vivo delivery, and therapeutics.
Collapse
Affiliation(s)
- Maria Abdul Ghafoor Raja
- Department of Pharmaceutics, Faculty of Pharmacy, Northern Border University, Rafha 73211, Saudi Arabia
| | - Haliza Katas
- Centre for Drug Delivery Research, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia
| | - Muhammad Wahab Amjad
- Department of Pharmaceutics, Faculty of Pharmacy, Northern Border University, Rafha 73211, Saudi Arabia
| |
Collapse
|
49
|
Sioud M. Releasing the Immune System Brakes Using siRNAs Enhances Cancer Immunotherapy. Cancers (Basel) 2019; 11:cancers11020176. [PMID: 30717461 PMCID: PMC6406640 DOI: 10.3390/cancers11020176] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 01/29/2019] [Accepted: 01/31/2019] [Indexed: 12/12/2022] Open
Abstract
Therapeutic dendritic cell (DC) cancer vaccines rely on the immune system to eradicate tumour cells. Although tumour antigen-specific T cell responses have been observed in most studies, clinical responses are fairly low, arguing for the need to improve the design of DC-based vaccines. The incorporation of small interfering RNAs (siRNAs) against immunosuppressive factors in the manufacturing process of DCs can turn the vaccine into potent immune stimulators. Additionally, siRNA modification of ex vivo-expanded T cells for adoptive immunotherapy enhanced their killing potency. Most of the siRNA-targeted immune inhibitory factors have been successful in that their blockade produced the strongest cytotoxic T cell responses in preclinical and clinical studies. Cancer patients treated with the siRNA-modified DC vaccines showed promising clinical benefits providing a strong rationale for further development of these immunogenic vaccine formulations. This review covers the progress in combining siRNAs with DC vaccines or T cell therapy to boost anti-tumour immunity.
Collapse
Affiliation(s)
- Mouldy Sioud
- Department of Immunology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Montebello, N-0310 Oslo, Norway.
| |
Collapse
|
50
|
Maeda Y, Iwata Hara R, Nishina K, Yoshida-Tanaka K, Sakamoto T, Yokota T, Wada T. Artificial cationic peptides that increase nuclease resistance of siRNA without disturbing RNAi activity. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2019; 38:338-348. [PMID: 30663497 DOI: 10.1080/15257770.2018.1543890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Properties of cationic peptides bearing amino or guanidino groups with various side chain lengths that bind to double stranded RNAs (dsRNAs) were investigated. Peptides with shorter side chain lengths effectively bound to dsRNAs (12mers) increasing their thermal stability. NMR measurements suggested that the cationic peptide binds to the inner side of the major groove of dsRNA. These peptides also increased the thermal stability of siRNA and effectively protected from RNase A digestion. On the other hand, both peptides containing amino groups and guanidine groups did not disturb RNAi activity.
Collapse
Affiliation(s)
- Yusuke Maeda
- a Faculty of Pharmaceutical Sciences , Tokyo University of Science , Noda , Chiba , Japan
| | - Rintaro Iwata Hara
- a Faculty of Pharmaceutical Sciences , Tokyo University of Science , Noda , Chiba , Japan.,b Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences , Tokyo Medical and Dental University , Bunkyo-ku , Tokyo , Japan
| | - Kazutaka Nishina
- b Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences , Tokyo Medical and Dental University , Bunkyo-ku , Tokyo , Japan
| | - Kie Yoshida-Tanaka
- b Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences , Tokyo Medical and Dental University , Bunkyo-ku , Tokyo , Japan
| | - Taiichi Sakamoto
- c Faculty of Advanced Engineering , Chiba Institute of Technology , Narashino , Chiba , Japan
| | - Takanori Yokota
- b Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences , Tokyo Medical and Dental University , Bunkyo-ku , Tokyo , Japan
| | - Takeshi Wada
- a Faculty of Pharmaceutical Sciences , Tokyo University of Science , Noda , Chiba , Japan
| |
Collapse
|