1
|
Yao Z, Liu T, Wang J, Fu Y, Zhao J, Wang X, Li Y, Yang X, He Z. Targeted delivery systems of siRNA based on ionizable lipid nanoparticles and cationic polymer vectors. Biotechnol Adv 2025; 81:108546. [PMID: 40015385 DOI: 10.1016/j.biotechadv.2025.108546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 02/04/2025] [Accepted: 02/23/2025] [Indexed: 03/01/2025]
Abstract
As an emerging therapeutic tool, small interfering RNA (siRNA) had the capability to down-regulate nearly all human mRNAs via sequence-specific gene silencing. Numerous studies have demonstrated the substantial potential of siRNA in the treatment of broad classes of diseases. With the discovery and development of various delivery systems and chemical modifications, six siRNA-based drugs have been approved by 2024. The utilization of siRNA-based therapeutics has significantly propelled efforts to combat a wide array of previously incurable diseases and advanced at a rapid pace, particularly with the help of potent targeted delivery systems. Despite encountering several extracellular and intracellular challenges, the efficiency of siRNA delivery has been gradually enhanced. Currently, targeted strategies aimed at improving potency and reducing toxicity played a crucial role in the druggability of siRNA. This review focused on recent advancements on ionizable lipid nanoparticles (LNPs) and cationic polymer (CP) vectors applied for targeted siRNA delivery. Based on various types of targeted modifications, we primarily described delivery systems modified with receptor ligands, peptides, antibodies, aptamers and amino acids. Finally, we discussed the challenges and opportunities associated with siRNA delivery systems based on ionizable LNPs and CPs vectors.
Collapse
Affiliation(s)
- Ziying Yao
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Taiqing Liu
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jingwen Wang
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yunhai Fu
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jinhua Zhao
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaoyu Wang
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yinqi Li
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaodong Yang
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhiyao He
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
2
|
Lin MH, Maniam P, Li D, Tang B, Bishop CR, Suhrbier A, Earl LW, Tayyar Y, McMillan NA, Li L, Harrich D. Harnessing defective interfering particles and lipid nanoparticles for effective delivery of an anti-dengue virus RNA therapy. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102424. [PMID: 39817192 PMCID: PMC11733052 DOI: 10.1016/j.omtn.2024.102424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 12/10/2024] [Indexed: 01/18/2025]
Abstract
Currently, no approved antiviral drugs target dengue virus (DENV) infection, leaving treatment reliant on supportive care. DENV vaccine efficacy varies depending on the vaccine type, the circulating serotype, and vaccine coverage. We investigated defective interfering particles (DIPs) and lipid nanoparticles (LNPs) to deliver DI290, an anti-DENV DI RNA. Both DIPs and DI290-loaded LNPs (LNP-290) effectively suppressed DENV infection in human primary monocyte-derived macrophages (MDMs), THP-1 macrophages, and fibroblasts-natural DENV targets. Inhibiting interferon (IFN) signaling with a Janus kinase 1/2 inhibitor or an IFN-α/β receptor 1 (IFNAR1)-binding antibody blocked DIP and LNP-290 antiviral activity. LNP-290 demonstrated a greater than log10 inhibition of DENV viral loads in IFNAR-deficient (Ifnar -/- ) and IFN regulatory factor (IRF) 3 and 7 double knockout (Irf3/7 -/- ) mice. Pathway analysis of RNA sequencing data from LNP-treated C57BL/6J mice, Ifnar -/- mice, and human MDMs treated with LNPs or DENV DIPs indicated DI290 treatment enhanced IFN responses, suggesting IFN-λ and IFN-γ provided antiviral activity when IFN-α/β responses were diminished. While viral interference by DI290 is possible, results did not support RNA replication competition as an inhibition mechanism. These findings suggest that DI290 may be a promising DENV therapeutic by activating the innate immune system.
Collapse
Affiliation(s)
- Min-Hsuan Lin
- Program of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Pramila Maniam
- Program of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Dongsheng Li
- Program of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Bing Tang
- Program of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Cameron R. Bishop
- Program of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Andreas Suhrbier
- Program of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
- Global Virus Network (GVN) Center of Excellence, Australian Infectious Disease Research Centre, Brisbane, QLD 4072, Australia
| | - Lucy Wales- Earl
- Menzies Health Institute Queensland and School of Pharmacy and Medical Science, Griffith University, Gold Coast, QLD 4222, Australia
| | - Yaman Tayyar
- Menzies Health Institute Queensland and School of Pharmacy and Medical Science, Griffith University, Gold Coast, QLD 4222, Australia
- Prorenata Biotech, Molendinar, QLD 4214, Australia
| | - Nigel A.J. McMillan
- Menzies Health Institute Queensland and School of Pharmacy and Medical Science, Griffith University, Gold Coast, QLD 4222, Australia
| | - Li Li
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, St Lucia, QLD 4072, Australia
| | - David Harrich
- Program of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
- Global Virus Network (GVN) Center of Excellence, Australian Infectious Disease Research Centre, Brisbane, QLD 4072, Australia
| |
Collapse
|
3
|
Ban E, Kim A. PicoGreen assay for nucleic acid quantification - Applications, challenges, and solutions. Anal Biochem 2024; 692:115577. [PMID: 38789006 DOI: 10.1016/j.ab.2024.115577] [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: 01/18/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 05/26/2024]
Abstract
Various analytical methods and reagents have been employed for nucleic acid analysis in cells, biological fluids, and formulations. Standard techniques like gel electrophoresis and qRT-PCR are widely used for qualitative and quantitative nucleic acid analysis. However, these methods can be time-consuming and labor-intensive, with limitations such as inapplicability to small RNA at low concentrations and high costs associated with qRT-PCR reagents and instruments. As an alternative, PicoGreen (PG) has emerged as a valuable method for the quantitative analysis of nucleic acids. PG, a fluorescent dye, enables the quantitation of double-stranded DNA (dsDNA) or double-stranded RNA, including miRNA mimic and siRNA, in solution. It is also applicable to DNA and RNA analysis within cells using techniques like FACS and fluorescence microscopy. Despite its advantages, PG's fluorescence intensity is affected by various experimental conditions, such as pH, salts, and chemical reagents. This review explores the recent applications of PG as a rapid, cost-effective, robust, and accurate assay tool for nucleic acid quantification. We also address the limitations of PG and discuss approaches to overcome these challenges, recognizing the expanding range of its applications.
Collapse
Affiliation(s)
- Eunmi Ban
- College of Pharmacy, CHA University, Seongnam, 13488, South Korea
| | - Aeri Kim
- College of Pharmacy, CHA University, Seongnam, 13488, South Korea.
| |
Collapse
|
4
|
Wilkinson AN, Chen R, Coleborn E, Neilson T, Le K, Bhavsar C, Wang Y, Atluri S, Irgam G, Wong K, Yang D, Steptoe R, Wu SY. Let-7i enhances anti-tumour immunity and suppresses ovarian tumour growth. Cancer Immunol Immunother 2024; 73:80. [PMID: 38554167 PMCID: PMC10981620 DOI: 10.1007/s00262-024-03674-w] [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: 01/31/2024] [Accepted: 03/08/2024] [Indexed: 04/01/2024]
Abstract
Cancer immunotherapy has seen significant success in the last decade for cancer management by enhancing endogenous cancer immunity. However, immunotherapies developed thus far have seen limited success in the majority of high-grade serous carcinoma (HGSC) ovarian cancer patients. This is largely due to the highly immunosuppressive tumour microenvironment of HGSC and late-stage identification. Thus, novel treatment interventions are needed to overcome this immunosuppression and complement existing immunotherapies. Here, we have identified through analysis of > 600 human HGSC tumours a critical role for Let-7i in modulating the tumoural immune network. Tumoural expression of Let-7i had high positive correlation with anti-cancer immune signatures in HGSC patients. Confirming this role, enforced Let-7i expression in murine HGSC tumours resulted in a significant decrease in tumour burden with a significant increase in tumour T cell numbers in tumours. In concert with the improved tumoural immunity, Let-7i treatment also significantly increased CD86 expression in antigen presenting cells (APCs) in the draining lymph nodes, indicating enhanced APC activity. Collectively, our findings highlight an important role of Let-7i in anti-tumour immunity and its potential use for inducing an anti-tumour effect in HGSC.
Collapse
Affiliation(s)
- Andrew N Wilkinson
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Rui Chen
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Elaina Coleborn
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Trent Neilson
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Khang Le
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Chintan Bhavsar
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Yue Wang
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Sharat Atluri
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Gowri Irgam
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Kiefer Wong
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Da Yang
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Raymond Steptoe
- Frazer Institute, University of Queensland, Brisbane, QLD, 4102, Australia
| | - Sherry Y Wu
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia.
| |
Collapse
|
5
|
Chen R, Coleborn E, Bhavsar C, Wang Y, Alim L, Wilkinson AN, Tran MA, Irgam G, Atluri S, Wong K, Shim JJ, Adityan S, Lee JS, Overwijk WW, Steptoe R, Yang D, Wu SY. miR-146a inhibits ovarian tumor growth in vivo via targeting immunosuppressive neutrophils and enhancing CD8 + T cell infiltration. Mol Ther Oncolytics 2023; 31:100725. [PMID: 37781339 PMCID: PMC10539880 DOI: 10.1016/j.omto.2023.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 09/08/2023] [Indexed: 10/03/2023] Open
Abstract
Immunotherapies have emerged as promising strategies for cancer treatment. However, existing immunotherapies have poor activity in high-grade serous ovarian cancer (HGSC) due to the immunosuppressive tumor microenvironment and the associated low tumoral CD8+ T cell (CTL) infiltration. Through multiple lines of evidence, including integrative analyses of human HGSC tumors, we have identified miR-146a as a master regulator of CTL infiltration in HGSC. Tumoral miR-146a expression is positively correlated with anti-cancer immune signatures in human HGSC tumors, and delivery of miR-146a to tumors resulted in significant reduction in tumor growth in both ID8-p53-/- and IG10 murine HGSC models. Increasing miR-146a expression in tumors improved anti-tumor immune responses by decreasing immune suppressive neutrophils and increasing CTL infiltration. Mechanistically, miR-146a targets IL-1 receptor-associated kinase 1 and tumor necrosis factor receptor-associated factor 6 adaptor molecules of the transcription factor nuclear factor κB signaling pathway in ID8-p53-/- cells and decreases production of the downstream neutrophil chemoattractant, C-X-C motif chemokine ligand 1. In addition to HGSC, tumoral miR-146a expression also correlates strongly with CTL infiltration in other cancer types including thyroid, prostate, breast, and adrenocortical cancers. Altogether, our findings highlight the ability of miR-146a to overcome immune suppression and improve CTL infiltration in tumors.
Collapse
Affiliation(s)
- Rui Chen
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Elaina Coleborn
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Chintan Bhavsar
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Yue Wang
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Louisa Alim
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Andrew N. Wilkinson
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | | | - Gowri Irgam
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Sharat Atluri
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Kiefer Wong
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jae-Jun Shim
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Siddharth Adityan
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Ju-Seog Lee
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Willem W. Overwijk
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Raymond Steptoe
- Frazer Institute, University of Queensland, Brisbane, QLD 4102, Australia
| | - Da Yang
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Sherry Y. Wu
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
6
|
Kafetzis KN, Papalamprou N, McNulty E, Thong KX, Sato Y, Mironov A, Purohit A, Welsby PJ, Harashima H, Yu‐Wai‐Man C, Tagalakis AD. The Effect of Cryoprotectants and Storage Conditions on the Transfection Efficiency, Stability, and Safety of Lipid-Based Nanoparticles for mRNA and DNA Delivery. Adv Healthc Mater 2023; 12:e2203022. [PMID: 36906918 PMCID: PMC11468535 DOI: 10.1002/adhm.202203022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/16/2023] [Indexed: 03/13/2023]
Abstract
Lipid-based nanoparticles have recently shown great promise, establishing themselves as the gold standard in delivering novel RNA therapeutics. However, research on the effects of storage on their efficacy, safety, and stability is still lacking. Herein, the impact of storage temperature on two types of lipid-based nanocarriers, lipid nanoparticles (LNPs) and receptor-targeted nanoparticles (RTNs), loaded with either DNA or messenger RNA (mRNA), is explored and the effects of different cryoprotectants on the stability and efficacy of the formulations are investigated. The medium-term stability of the nanoparticles was evaluated by monitoring their physicochemical characteristics, entrapment and transfection efficiency, every two weeks over one month. It is demonstrated, that the use of cryoprotectants protects nanoparticles against loss of function and degradation in all storage conditions. Moreover, it is shown that the addition of sucrose enables all nanoparticles to remain stable and maintain their efficacy for up to a month when stored at -80 °C, regardless of cargo or type of nanoparticle. DNA-loaded nanoparticles also remain stable in a wider variety of storage conditions than mRNA-loaded ones. Importantly, these novel LNPs show increased GFP expression that can signify their future use in gene therapies, beyond the established role of LNPs in RNA therapeutics.
Collapse
Affiliation(s)
| | | | - Elisha McNulty
- Department of BiologyEdge Hill UniversityOrmskirkL39 4QPUK
| | - Kai X. Thong
- Faculty of Life Sciences & MedicineKing's College LondonLondonSE1 7EHUK
| | - Yusuke Sato
- Faculty of Pharmaceutical SciencesHokkaido UniversityKita‐12, Nishi‐6, Kita‐kuSapporo060–0812Japan
| | - Aleksandr Mironov
- Electron Microscopy Core Facility (RRID: SCR_021147)Faculty of BiologyMedicine and HealthUniversity of ManchesterManchesterM13 9PTUK
| | - Atul Purohit
- Oncology Drug Discovery & Women's Health GroupDepartment of MetabolismDigestion & ReproductionImperial College LondonLondonW12 0HSUK
| | | | - Hideyoshi Harashima
- Faculty of Pharmaceutical SciencesHokkaido UniversityKita‐12, Nishi‐6, Kita‐kuSapporo060–0812Japan
| | | | | |
Collapse
|
7
|
Supramaniam A, Tayyar Y, Clarke DTW, Kelly G, Acharya D, Morris KV, McMillan NAJ, Idris A. Prophylactic intranasal administration of lipid nanoparticle formulated siRNAs reduce SARS-CoV-2 and RSV lung infection. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2023:S1684-1182(23)00068-3. [PMID: 36934064 PMCID: PMC9991324 DOI: 10.1016/j.jmii.2023.02.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/17/2022] [Accepted: 02/28/2023] [Indexed: 03/09/2023]
Abstract
RNA interference (RNAi) is an emerging and promising therapy for a wide range of respiratory viral infections. This highly specific suppression can be achieved by the introduction of short-interfering RNA (siRNA) into mammalian systems, resulting in the effective reduction of viral load. Unfortunately, this has been hindered by the lack of a good delivery system, especially via the intranasal (IN) route. Here, we have developed an IN siRNA encapsulated lipid nanoparticle (LNP) in vivo delivery system that is highly efficient at targeting severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and respiratory syncytial virus (RSV) lung infection in vivo. Importantly, IN siRNA delivery without the aid of LNPs abolishes anti-SARS-CoV-2 activity in vivo. Our approach using LNPs as the delivery vehicle overcomes the significant barriers seen with IN delivery of siRNA therapeutics and is a significant advancement in our ability to delivery siRNAs. The study presented here demonstrates an attractive alternate delivery strategy for the prophylactic treatment of both future and emerging respiratory viral diseases.
Collapse
Affiliation(s)
- Aroon Supramaniam
- Menzies Health Institute Queensland, Griffith University, Southport, Queensland, Australia; School of Pharmacy and Medical Science, Griffith University, Southport, Queensland, Australia
| | - Yaman Tayyar
- Menzies Health Institute Queensland, Griffith University, Southport, Queensland, Australia; School of Pharmacy and Medical Science, Griffith University, Southport, Queensland, Australia; Prorenata Biotech, Molendinar, Queensland, Australia
| | - Daniel T W Clarke
- School of Pharmacy and Medical Science, Griffith University, Southport, Queensland, Australia
| | - Gabrielle Kelly
- Menzies Health Institute Queensland, Griffith University, Southport, Queensland, Australia; School of Pharmacy and Medical Science, Griffith University, Southport, Queensland, Australia
| | - Dhruba Acharya
- Menzies Health Institute Queensland, Griffith University, Southport, Queensland, Australia
| | - Kevin V Morris
- Menzies Health Institute Queensland, Griffith University, Southport, Queensland, Australia; School of Pharmacy and Medical Science, Griffith University, Southport, Queensland, Australia
| | - Nigel A J McMillan
- Menzies Health Institute Queensland, Griffith University, Southport, Queensland, Australia; School of Pharmacy and Medical Science, Griffith University, Southport, Queensland, Australia
| | - Adi Idris
- Menzies Health Institute Queensland, Griffith University, Southport, Queensland, Australia; School of Pharmacy and Medical Science, Griffith University, Southport, Queensland, Australia; School of Biomedical Sciences, Queensland University of Technology, Gardens Point, Queensland, Australia.
| |
Collapse
|
8
|
Bai M, Liu ZL, Zhou YY, Xu QX, Liu TX, Tian HG. Influence of diverse storage conditions of double-stranded RNA in vitro on the RNA interference efficiency in vivo insect Tribolium castaneum. PEST MANAGEMENT SCIENCE 2023; 79:45-54. [PMID: 36086883 DOI: 10.1002/ps.7171] [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: 05/20/2022] [Revised: 08/10/2022] [Accepted: 09/10/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND A significant variation in RNA interference (RNAi) efficiency hinders further functional gene studies and pest control application in many insects. The available double-stranded RNA (dsRNA) molecules introduced into the target cells are regarded as the crucial factor for efficient RNAi response. However, numerous studies have only focused on dsRNA stability in vivo; it is uncertain whether different dsRNA storage conditions in vitro play a role in variable RNAi efficiency among insects. RESULTS A marker gene cardinal, which leads to white eyes when knocked-down in the red flour beetle Tribolium castaneum, was used to evaluate the effects of RNAi efficiency under different dsRNA storage conditions. We demonstrated that the dsRNA molecule is very stable under typical cryopreservation temperatures (-80 and -20 °C) within 180 days, and RNAi efficiency shows no significant differences under either low temperature. Unexpectedly, while dsRNA molecules were treated with multiple freeze-thaw cycles up to 50 times between -80/-20 °C and room temperature, we discovered that dsRNA integrity and RNAi efficiency were comparable with fresh dsRNA. Finally, when the stability of dsRNA was further measured under refrigerated storage conditions (4 °C), we surprisingly found that dsRNA is still stable within 180 days and can induce an efficient RNAi response as that of initial dsRNA. CONCLUSION Our results indicate that dsRNA is extraordinarily stable under various temperature storage conditions that did not significantly impact RNAi efficiency in vivo insects. © 2022 Society of Chemical Industry.
Collapse
Affiliation(s)
- Mei Bai
- State Key Laboratory of Crop Stress Biology for Arid Areas, Northwest A&F University, Yangling, China
- Key Laboratory of Integrated Pest Management on the Loess Plateau of Ministry of Agriculture, College of Plant Protection, Northwest A&F University, Yangling, China
| | - Zi-Ling Liu
- State Key Laboratory of Crop Stress Biology for Arid Areas, Northwest A&F University, Yangling, China
- Key Laboratory of Integrated Pest Management on the Loess Plateau of Ministry of Agriculture, College of Plant Protection, Northwest A&F University, Yangling, China
| | - Yu-Yu Zhou
- State Key Laboratory of Crop Stress Biology for Arid Areas, Northwest A&F University, Yangling, China
- Key Laboratory of Integrated Pest Management on the Loess Plateau of Ministry of Agriculture, College of Plant Protection, Northwest A&F University, Yangling, China
| | - Qiu-Xuan Xu
- State Key Laboratory of Crop Stress Biology for Arid Areas, Northwest A&F University, Yangling, China
- Key Laboratory of Integrated Pest Management on the Loess Plateau of Ministry of Agriculture, College of Plant Protection, Northwest A&F University, Yangling, China
| | - Tong-Xian Liu
- State Key Laboratory of Crop Stress Biology for Arid Areas, Northwest A&F University, Yangling, China
- Key Laboratory of Integrated Pest Management on the Loess Plateau of Ministry of Agriculture, College of Plant Protection, Northwest A&F University, Yangling, China
| | - Hong-Gang Tian
- State Key Laboratory of Crop Stress Biology for Arid Areas, Northwest A&F University, Yangling, China
- Key Laboratory of Integrated Pest Management on the Loess Plateau of Ministry of Agriculture, College of Plant Protection, Northwest A&F University, Yangling, China
| |
Collapse
|
9
|
Schürz M, Danmayr J, Jaritsch M, Klinglmayr E, Benirschke HM, Matea C, Zimmerebner P, Rauter J, Wolf M, Gomes FG, Kratochvil Z, Heger Z, Miller A, Heuser T, Stanojlovic V, Kiefer J, Plank T, Johnson L, Himly M, Blöchl C, Huber CG, Hintersteiner M, Meisner‐Kober N. EVAnalyzer: High content imaging for rigorous characterisation of single extracellular vesicles using standard laboratory equipment and a new open-source ImageJ/Fiji plugin. J Extracell Vesicles 2022; 11:e12282. [PMID: 36437554 PMCID: PMC9702573 DOI: 10.1002/jev2.12282] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 11/03/2022] [Accepted: 11/07/2022] [Indexed: 11/30/2022] Open
Abstract
Extracellular vesicle (EV) research increasingly demands for quantitative characterisation at the single vesicle level to address heterogeneity and complexity of EV subpopulations. Emerging, commercialised technologies for single EV analysis based on, for example, imaging flow cytometry or imaging after capture on chips generally require dedicated instrumentation and proprietary software not readily accessible to every lab. This limits their implementation for routine EV characterisation in the rapidly growing EV field. We and others have shown that single vesicles can be detected as light diffraction limited fluorescent spots using standard confocal and widefield fluorescence microscopes. Advancing this simple strategy into a process for routine EV quantitation, we developed 'EVAnalyzer', an ImageJ/Fiji (Fiji is just ImageJ) plugin for automated, quantitative single vesicle analysis from imaging data. Using EVAnalyzer, we established a robust protocol for capture, (immuno-)labelling and fluorescent imaging of EVs. To exemplify the application scope, the process was optimised and systematically tested for (i) quantification of EV subpopulations, (ii) validation of EV labelling reagents, (iii) in situ determination of antibody specificity, sensitivity and species cross-reactivity for EV markers and (iv) optimisation of genetic EV engineering. Additionally, we show that the process can be applied to synthetic nanoparticles, allowing to determine siRNA encapsulation efficiencies of lipid-based nanoparticles (LNPs) and protein loading of SiO2 nanoparticles. EVAnalyzer further provides a pipeline for automated quantification of cell uptake at the single cell-single vesicle level, thereby enabling high content EV cell uptake assays and plate-based screens. Notably, the entire procedure from sample preparation to the final data output is entirely based on standard reagents, materials, laboratory equipment and open access software. In summary, we show that EVAnalyzer enables rigorous characterisation of EVs with generally accessible tools. Since we further provide the plugin as open-source code, we expect EVAnalyzer to not only be a resource of immediate impact, but an open innovation platform for the EV and nanoparticle research communities.
Collapse
Affiliation(s)
- Melanie Schürz
- Department of Biosciences and Medical BiologyParis Lodron University SalzburgSalzburgAustria
| | - Joachim Danmayr
- Department of Informatics and MathematicsFernuniversität HagenHagenGermany
| | - Maria Jaritsch
- Department of Biosciences and Medical BiologyParis Lodron University SalzburgSalzburgAustria
| | - Eva Klinglmayr
- Department of Biosciences and Medical BiologyParis Lodron University SalzburgSalzburgAustria
| | - Heloisa Melo Benirschke
- Department of Biosciences and Medical BiologyParis Lodron University SalzburgSalzburgAustria
| | - Cristian‐Tudor Matea
- Department of Biosciences and Medical BiologyParis Lodron University SalzburgSalzburgAustria
| | - Patrick Zimmerebner
- Department of Biosciences and Medical BiologyParis Lodron University SalzburgSalzburgAustria
| | - Jakob Rauter
- Department of Biosciences and Medical BiologyParis Lodron University SalzburgSalzburgAustria
| | - Martin Wolf
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical University (PMU)SalzburgAustria
| | - Fausto Gueths Gomes
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Centre Salzburg (SCI‐TReCS)Paracelsus Medical University (PMU)SalzburgAustria
| | - Zdenek Kratochvil
- Department of Chemistry and BiochemistryMendel University in BrnoBrnoCzech Republic
| | - Zbynek Heger
- Department of Chemistry and BiochemistryMendel University in BrnoBrnoCzech Republic
| | - Andrew Miller
- Department of Chemistry and BiochemistryMendel University in BrnoBrnoCzech Republic
- Veterinary Research InstituteBrnoCzech Republic
- KP Therapeutics (Europe) sro.BrnoCzech Republic
| | | | - Vesna Stanojlovic
- Department of Biosciences and Medical BiologyParis Lodron University SalzburgSalzburgAustria
| | - Jana Kiefer
- Department of Biosciences and Medical BiologyParis Lodron University SalzburgSalzburgAustria
| | - Tanja Plank
- Department of Biosciences and Medical BiologyParis Lodron University SalzburgSalzburgAustria
| | - Litty Johnson
- Department of Biosciences and Medical BiologyParis Lodron University SalzburgSalzburgAustria
| | - Martin Himly
- Department of Biosciences and Medical BiologyParis Lodron University SalzburgSalzburgAustria
| | - Constantin Blöchl
- Department of Biosciences and Medical BiologyParis Lodron University SalzburgSalzburgAustria
| | - Christian G. Huber
- Department of Biosciences and Medical BiologyParis Lodron University SalzburgSalzburgAustria
| | | | - Nicole Meisner‐Kober
- Department of Biosciences and Medical BiologyParis Lodron University SalzburgSalzburgAustria
| |
Collapse
|
10
|
Arriaga-Canon C, Contreras-Espinosa L, Rebollar-Vega R, Montiel-Manríquez R, Cedro-Tanda A, García-Gordillo JA, Álvarez-Gómez RM, Jiménez-Trejo F, Castro-Hernández C, Herrera LA. Transcriptomics and RNA-Based Therapeutics as Potential Approaches to Manage SARS-CoV-2 Infection. Int J Mol Sci 2022; 23:11058. [PMID: 36232363 PMCID: PMC9570475 DOI: 10.3390/ijms231911058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 11/24/2022] Open
Abstract
SARS-CoV-2 is a coronavirus family member that appeared in China in December 2019 and caused the disease called COVID-19, which was declared a pandemic in 2020 by the World Health Organization. In recent months, great efforts have been made in the field of basic and clinical research to understand the biology and infection processes of SARS-CoV-2. In particular, transcriptome analysis has contributed to generating new knowledge of the viral sequences and intracellular signaling pathways that regulate the infection and pathogenesis of SARS-CoV-2, generating new information about its biology. Furthermore, transcriptomics approaches including spatial transcriptomics, single-cell transcriptomics and direct RNA sequencing have been used for clinical applications in monitoring, detection, diagnosis, and treatment to generate new clinical predictive models for SARS-CoV-2. Consequently, RNA-based therapeutics and their relationship with SARS-CoV-2 have emerged as promising strategies to battle the SARS-CoV-2 pandemic with the assistance of novel approaches such as CRISPR-CAS, ASOs, and siRNA systems. Lastly, we discuss the importance of precision public health in the management of patients infected with SARS-CoV-2 and establish that the fusion of transcriptomics, RNA-based therapeutics, and precision public health will allow a linkage for developing health systems that facilitate the acquisition of relevant clinical strategies for rapid decision making to assist in the management and treatment of the SARS-CoV-2-infected population to combat this global public health problem.
Collapse
Affiliation(s)
- Cristian Arriaga-Canon
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Avenida San Fernando No. 22 ColC. Sección XVI, Tlalpan. C.P., Mexico City 14080, Mexico
| | - Laura Contreras-Espinosa
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Avenida San Fernando No. 22 ColC. Sección XVI, Tlalpan. C.P., Mexico City 14080, Mexico
| | - Rosa Rebollar-Vega
- Genomics Laboratory, Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México, Vasco de Quiroga 15, Belisario Domínguez Secc 16, Tlalpan, Mexico City 14080, Mexico
| | - Rogelio Montiel-Manríquez
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Avenida San Fernando No. 22 ColC. Sección XVI, Tlalpan. C.P., Mexico City 14080, Mexico
| | - Alberto Cedro-Tanda
- Instituto Nacional de Medicina Genómica, Periférico Sur 4809, Arenal Tepepan, Tlalpan. C.P., Mexico City 14610, Mexico
| | - José Antonio García-Gordillo
- Oncología Médica, Instituto Nacional de Cancerología, Avenida San Fernando No. 22 Col. Sección XVI, Tlalpan. C.P., Mexico City 14080, Mexico
| | - Rosa María Álvarez-Gómez
- Clínica de Cáncer Hereditario, Instituto Nacional de Cancerología, Avenida San Fernando No. 22 Col. Sección XVI, Tlalpan. C.P., Mexico City 14080, Mexico
| | - Francisco Jiménez-Trejo
- Instituto Nacional de Pediatría, Insurgentes Sur No. 3700-C, Coyoacán. C.P., Mexico City 04530, Mexico
| | - Clementina Castro-Hernández
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Avenida San Fernando No. 22 ColC. Sección XVI, Tlalpan. C.P., Mexico City 14080, Mexico
| | - Luis A. Herrera
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Avenida San Fernando No. 22 ColC. Sección XVI, Tlalpan. C.P., Mexico City 14080, Mexico
- Instituto Nacional de Medicina Genómica, Periférico Sur 4809, Arenal Tepepan, Tlalpan. C.P., Mexico City 14610, Mexico
| |
Collapse
|
11
|
Iraci N, Corsaro C, Giofrè SV, Neri G, Mezzasalma AM, Vacalebre M, Speciale A, Saija A, Cimino F, Fazio E. Nanoscale Technologies in the Fight against COVID-19: From Innovative Nanomaterials to Computer-Aided Discovery of Potential Antiviral Plant-Derived Drugs. Biomolecules 2022; 12:1060. [PMID: 36008954 PMCID: PMC9405735 DOI: 10.3390/biom12081060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 12/04/2022] Open
Abstract
The last few years have increasingly emphasized the need to develop new active antiviral products obtained from artificial synthesis processes using nanomaterials, but also derived from natural matrices. At the same time, advanced computational approaches have found themselves fundamental in the repurposing of active therapeutics or for reducing the very long developing phases of new drugs discovery, which represents a real limitation, especially in the case of pandemics. The first part of the review is focused on the most innovative nanomaterials promising both in the field of therapeutic agents, as well as measures to control virus spread (i.e., innovative antiviral textiles). The second part of the review aims to show how computer-aided technologies can allow us to identify, in a rapid and therefore constantly updated way, plant-derived molecules (i.e., those included in terpenoids) potentially able to efficiently interact with SARS-CoV-2 cell penetration pathways.
Collapse
Affiliation(s)
- Nunzio Iraci
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno D’Alcontres 31, I-98166 Messina, Italy; (N.I.); (S.V.G.); (G.N.); (A.S.); (A.S.)
| | - Carmelo Corsaro
- Department of Mathematical and Computational Sciences, Physics Science and Earth Science, University of Messina, Viale F. Stagno D’Alcontres 31, I-98166 Messina, Italy; (A.M.M.); (M.V.); (E.F.)
| | - Salvatore V. Giofrè
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno D’Alcontres 31, I-98166 Messina, Italy; (N.I.); (S.V.G.); (G.N.); (A.S.); (A.S.)
| | - Giulia Neri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno D’Alcontres 31, I-98166 Messina, Italy; (N.I.); (S.V.G.); (G.N.); (A.S.); (A.S.)
| | - Angela Maria Mezzasalma
- Department of Mathematical and Computational Sciences, Physics Science and Earth Science, University of Messina, Viale F. Stagno D’Alcontres 31, I-98166 Messina, Italy; (A.M.M.); (M.V.); (E.F.)
| | - Martina Vacalebre
- Department of Mathematical and Computational Sciences, Physics Science and Earth Science, University of Messina, Viale F. Stagno D’Alcontres 31, I-98166 Messina, Italy; (A.M.M.); (M.V.); (E.F.)
| | - Antonio Speciale
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno D’Alcontres 31, I-98166 Messina, Italy; (N.I.); (S.V.G.); (G.N.); (A.S.); (A.S.)
| | - Antonina Saija
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno D’Alcontres 31, I-98166 Messina, Italy; (N.I.); (S.V.G.); (G.N.); (A.S.); (A.S.)
| | - Francesco Cimino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno D’Alcontres 31, I-98166 Messina, Italy; (N.I.); (S.V.G.); (G.N.); (A.S.); (A.S.)
| | - Enza Fazio
- Department of Mathematical and Computational Sciences, Physics Science and Earth Science, University of Messina, Viale F. Stagno D’Alcontres 31, I-98166 Messina, Italy; (A.M.M.); (M.V.); (E.F.)
| |
Collapse
|
12
|
Alavizadeh SH, Doagooyan M, Zahedipour F, Torghabe SY, Baharieh B, Soleymani F, Gheybi F. Antisense technology as a potential strategy for the treatment of coronaviruses infection: With focus on COVID-19. IET Nanobiotechnol 2022; 16:67-77. [PMID: 35274474 PMCID: PMC9007150 DOI: 10.1049/nbt2.12079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 01/25/2022] [Accepted: 02/14/2022] [Indexed: 11/20/2022] Open
Abstract
After the outbreak of coronavirus disease 2019 (COVID-19) in December 2019 and the increasing number of SARS-CoV-2 infections all over the world, researchers are struggling to investigate effective therapeutic strategies for the treatment of this infection. Targeting viral small molecules that are involved in the process of infection is a promising strategy. Since many host factors are also used by SARS-CoV-2 during various stages of infection, down-regulating or silencing these factors can serve as an effective therapeutic tool. Several nucleic acid-based technologies including short interfering RNAs, antisense oligonucleotides, aptamers, DNAzymes, and ribozymes have been suggested for the control of SARS-CoV-2 as well as other respiratory viruses. The antisense technology also plays an indispensable role in the treatment of many other diseases including cancer, influenza, and acquired immunodeficiency syndrome. In this review, we summarised the potential applications of antisense technology for the treatment of coronaviruses and specifically COVID-19 infection.
Collapse
Affiliation(s)
- Seyedeh Hoda Alavizadeh
- Nanotechnology Research CenterPharmaceutical Technology InstituteMashhad University of Medical SciencesMashhadIran
- Department of Pharmaceutical NanotechnologySchool of PharmacyMashhad University of Medical SciencesMashhadIran
| | - Maham Doagooyan
- Department of Medical Biotechnology and NanotechnologyFaculty of MedicineMashhad University of Medical SciencesMashhadIran
- Department of Molecular MedicineBiotechnology Research CenterPasteur Institute of IranTehranIran
| | - Fatemeh Zahedipour
- Department of Medical Biotechnology and NanotechnologyFaculty of MedicineMashhad University of Medical SciencesMashhadIran
- Student Research CommitteeFaculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - Shima Yahoo Torghabe
- Department of Basic SciencesSari Agricultural Sciences and Natural Resources UniversitySariIran
| | - Bahare Baharieh
- Department of Medical Biotechnology and NanotechnologyFaculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - Firooze Soleymani
- Department of Medical Biotechnology and NanotechnologyFaculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - Fatemeh Gheybi
- Nanotechnology Research CenterPharmaceutical Technology InstituteMashhad University of Medical SciencesMashhadIran
- Department of Medical Biotechnology and NanotechnologyFaculty of MedicineMashhad University of Medical SciencesMashhadIran
| |
Collapse
|
13
|
Islam F, Zhou Y, Lam AK. Liposomal siRNA Delivery in Papillary Thyroid Carcinoma Cells. Methods Mol Biol 2022; 2534:121-133. [PMID: 35670972 DOI: 10.1007/978-1-0716-2505-7_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The discovery of RNA interference (RNAi) has opened a new strategy in cancer therapy, especially by silencing target genes. Pharmacologically it can be achieved by introducing of small (19-21 base pairs) dsRNA molecules known as small interfering RNA (siRNA) targeting interested genes. siRNA mediated gene has been widely investigated for its utility in treating various diseases including cancer. However, the systemic delivery of interested siRNA via non-viral methods remains a major challenge with large numbers of polymeric and liposomal systems being tested. The most effective methods involving cationic liposomes delivery to cells. Nonetheless, systemic delivery of siRNA via cationic lipid particles is often poor due to rapid uptake by reticuloendothelial organs, resulting in decreased delivery of these particles to the site of interest. Polyethylene glycol (PEG) has been used in siRNA-liposomes formulation to minimize reticuloendothelial uptake. Also, PEGylation permits the accumulation of the liposomes-loaded siRNA at the tumor sites with defective vasculatures such as enhanced permeability and retention phenomena. Thus, a simple method to prepare stable PEGylated siRNA-loaded lipid particles could provide better systemic delivery system in treating various cancers, including papillary thyroid carcinoma. Here we illustrate a simple protocol for the formulation of siRNA-loaded lipid particles by hydration of freeze-dried matrix (HFDM) method for effective delivery of target specific siRNA to papillary thyroid carcinoma cells.
Collapse
Affiliation(s)
- Farhadul Islam
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, Bangladesh.
- Cancer Molecular Pathology of School of Medicine and Dentistry, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia.
| | - Yaoqi Zhou
- Institute for Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, China
| | - Alfred K Lam
- Cancer Molecular Pathology of School of Medicine and Dentistry, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia.
- Pathology Queensland, Gold Coast University Hospital, Southport, QLD, Australia.
- Faculty of Medicine, University of Queensland, Herston, QLD, Australia.
| |
Collapse
|
14
|
Naqvi RA, Shukla D, Naqvi AR. SARS-CoV-2 targeting by RNAi and host complement inhibition: A two-pronged subterfuge for COVID-19 treatment. Immun Inflamm Dis 2022; 10:22-25. [PMID: 34644457 PMCID: PMC8652782 DOI: 10.1002/iid3.549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/30/2021] [Accepted: 10/04/2021] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND The lack of knowledge about the specific preventive measures and limited scientific information on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) led to an excruciating onset and progression of coronavirus disease 2019 (COVID-19). Swift development of various successful vaccines around the globe is striving to contain the exponential surges of COVID-19 cases. However, the ongoing struggle to vaccinate the global population and alarming spread of highly transmissible variants may thwart global initiatives to contain SARS-CoV-2 as observed by less robust protective immunity. METHODS In this perspective, we propose a thought-provoking, two-pronged strategy involving RNA interference approach to degrade essential SARS-CoV-2 ORFs required for replication and entry in conjunction with a complement inhibitor (compstatin) to stymie the detrimental proinflammatory cytokine storm that exacerbate disease progression and severity. RESULTS We provide supporting evidence suggesting that concurrent targeting of viral and host components will be a superior strategy to effectively suppress viral spread and clinical manifestations of COVID-19. CONCLUSION SARS-CoV-2 specific RNAi in conjunction with systemic delivery of compstatin will be an effective two-pronged strategy to combat local and systemic immune responses in both symptomatic and asymptomatic COVID-19 patients.
Collapse
Affiliation(s)
- Raza Ali Naqvi
- Department of Periodontics, College of DentistryUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Deepak Shukla
- Department of Ophthalmology and Visual SciencesUniversity of Illinois Medical CenterChicagoIllinoisUSA
- Department of Microbiology and ImmunologyUniversity of Illinois Medical CenterChicagoIllinoisUSA
| | - Afsar R. Naqvi
- Department of Periodontics, College of DentistryUniversity of Illinois at ChicagoChicagoIllinoisUSA
| |
Collapse
|
15
|
A novel method for the development of plasmid DNA-loaded nanoliposomes for cancer gene therapy. Drug Deliv Transl Res 2021; 12:1508-1520. [PMID: 34322851 DOI: 10.1007/s13346-021-01034-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2021] [Indexed: 10/20/2022]
Abstract
We aimed to develop a simple yet novel method to prepare plasmid DNA-loaded nanoliposomes for cancer gene therapy. Murine interleukin-12 (mIL-12) pDNA-loaded nanoliposomes were prepared via novel freeze-drying of a monophase solution method. The physicochemical characteristics, cytotoxicity, and transfection efficiency of the prepared nanoliposomes in murine CT-26 colon carcinoma cells were evaluated. Furthermore, tumor progression and survival rate in CT-26 colon carcinoma-bearing BALB/c mice subsequent to direct intratumoral injections were investigated over a period of 40 days. Using this preparation method, nanoliposomes with particle size of around 300 nm and zeta potential of 96.5 mV were obtained. The transmission electron microscope results showed that the liposomes were nano-sized and almost spherical. The agarose gel retardation assay revealed the pDNA encapsulation in the nanoliposomes. The nanoliposomes with 72.4% encapsulation efficiency and low cell toxicity could significantly improve mIL-12 expression by approximately 25-fold relative to the naked mIL-12 pDNA. There was a significant tumor growth inhibition after repeated injections of mIL-12 pDNA-loaded nanoliposomes. This is the first study on the freeze-drying of a monophase solution method as a simple yet novel technique for the preparation of pDNA-loaded nanoliposomes. Given the ease of preparation method and promising in vitro and in vivo characteristics, this investigation demonstrates advances in pDNA lipid formulation for cancer gene therapy.
Collapse
|
16
|
Idris A, Davis A, Supramaniam A, Acharya D, Kelly G, Tayyar Y, West N, Zhang P, McMillan CLD, Soemardy C, Ray R, O'Meally D, Scott TA, McMillan NAJ, Morris KV. A SARS-CoV-2 targeted siRNA-nanoparticle therapy for COVID-19. Mol Ther 2021; 29:2219-2226. [PMID: 33992805 PMCID: PMC8118699 DOI: 10.1016/j.ymthe.2021.05.004] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/02/2021] [Accepted: 05/05/2021] [Indexed: 01/16/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in humans. Despite several emerging vaccines, there remains no verifiable therapeutic targeted specifically to the virus. Here we present a highly effective small interfering RNA (siRNA) therapeutic against SARS-CoV-2 infection using a novel lipid nanoparticle (LNP) delivery system. Multiple siRNAs targeting highly conserved regions of the SARS-CoV-2 virus were screened, and three candidate siRNAs emerged that effectively inhibit the virus by greater than 90% either alone or in combination with one another. We simultaneously developed and screened two novel LNP formulations for the delivery of these candidate siRNA therapeutics to the lungs, an organ that incurs immense damage during SARS-CoV-2 infection. Encapsulation of siRNAs in these LNPs followed by in vivo injection demonstrated robust repression of virus in the lungs and a pronounced survival advantage to the treated mice. Our LNP-siRNA approaches are scalable and can be administered upon the first sign of SARS-CoV-2 infection in humans. We suggest that an siRNA-LNP therapeutic approach could prove highly useful in treating COVID-19 disease as an adjunctive therapy to current vaccine strategies.
Collapse
Affiliation(s)
- Adi Idris
- Menzies Health Institute Queensland, School of Medical Science Griffith University, Gold Coast Campus, QLD 4222, Australia
| | - Alicia Davis
- Center for Gene Therapy, Hematological Malignancy and Stem Cell Transplantation Institute at the City of Hope and City of Hope Beckman Research Institute, 1500 E. Duarte Road, Duarte, CA 91010, USA; Irell & Manella Graduate School of Biological Sciences at the City of Hope, Duarte, CA 91010, USA
| | - Aroon Supramaniam
- Menzies Health Institute Queensland, School of Medical Science Griffith University, Gold Coast Campus, QLD 4222, Australia
| | - Dhruba Acharya
- Menzies Health Institute Queensland, School of Medical Science Griffith University, Gold Coast Campus, QLD 4222, Australia
| | - Gabrielle Kelly
- Menzies Health Institute Queensland, School of Medical Science Griffith University, Gold Coast Campus, QLD 4222, Australia
| | - Yaman Tayyar
- Menzies Health Institute Queensland, School of Medical Science Griffith University, Gold Coast Campus, QLD 4222, Australia
| | - Nic West
- Menzies Health Institute Queensland, School of Medical Science Griffith University, Gold Coast Campus, QLD 4222, Australia
| | - Ping Zhang
- Menzies Health Institute Queensland, School of Medical Science Griffith University, Gold Coast Campus, QLD 4222, Australia
| | - Christopher L D McMillan
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Citradewi Soemardy
- Center for Gene Therapy, Hematological Malignancy and Stem Cell Transplantation Institute at the City of Hope and City of Hope Beckman Research Institute, 1500 E. Duarte Road, Duarte, CA 91010, USA
| | - Roslyn Ray
- Center for Gene Therapy, Hematological Malignancy and Stem Cell Transplantation Institute at the City of Hope and City of Hope Beckman Research Institute, 1500 E. Duarte Road, Duarte, CA 91010, USA
| | - Denis O'Meally
- Center for Gene Therapy, Hematological Malignancy and Stem Cell Transplantation Institute at the City of Hope and City of Hope Beckman Research Institute, 1500 E. Duarte Road, Duarte, CA 91010, USA
| | - Tristan A Scott
- Center for Gene Therapy, Hematological Malignancy and Stem Cell Transplantation Institute at the City of Hope and City of Hope Beckman Research Institute, 1500 E. Duarte Road, Duarte, CA 91010, USA
| | - Nigel A J McMillan
- Menzies Health Institute Queensland, School of Medical Science Griffith University, Gold Coast Campus, QLD 4222, Australia.
| | - Kevin V Morris
- Menzies Health Institute Queensland, School of Medical Science Griffith University, Gold Coast Campus, QLD 4222, Australia; Center for Gene Therapy, Hematological Malignancy and Stem Cell Transplantation Institute at the City of Hope and City of Hope Beckman Research Institute, 1500 E. Duarte Road, Duarte, CA 91010, USA.
| |
Collapse
|
17
|
Idris A, Davis A, Supramaniam A, Acharya D, Kelly G, Tayyar Y, West N, Zhang P, McMillan CLD, Soemardy C, Ray R, O'Meally D, Scott TA, McMillan NAJ, Morris KV. A SARS-CoV-2 targeted siRNA-nanoparticle therapy for COVID-19. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021. [PMID: 33907744 DOI: 10.1101/2021.04.19.440531] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in humans. Despite several emerging vaccines, there remains no verifiable therapeutic targeted specifically to the virus. Here we present a highly effective siRNA therapeutic against SARS-CoV-2 infection using a novel lipid nanoparticle delivery system. Multiple small-interfering RNAs (siRNAs) targeting highly conserved regions of the SARS-CoV-2 virus were screened and three candidate siRNAs emerged that effectively inhibit virus by greater than 90% either alone or in combination with one another. We simultaneously developed and screened two novel lipid nanoparticle formulations for the delivery of these candidate siRNA therapeutics to the lungs, an organ that incurs immense damage during SARS-CoV-2 infection. Encapsulation of siRNAs in these LNPs followed by in vivo injection demonstrated robust repression of virus in the lungs and a pronounced survival advantage to the treated mice. Our LNP-siRNA approaches are scalable and can be administered upon the first sign of SARS-CoV-2 infection in humans. We suggest that an siRNA-LNP therapeutic approach could prove highly useful in treating COVID-19 disease as an adjunctive therapy to current vaccine strategies.
Collapse
|
18
|
Scrutinizing the therapeutic and diagnostic potential of nanotechnology in thyroid cancer: Edifying drug targeting by nano-oncotherapeutics. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2020.102221] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
19
|
CRISPR/Cas9-loaded stealth liposomes effectively cleared established HPV16-driven tumours in syngeneic mice. PLoS One 2021; 16:e0223288. [PMID: 33411765 PMCID: PMC7790238 DOI: 10.1371/journal.pone.0223288] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 11/02/2020] [Indexed: 11/19/2022] Open
Abstract
Gene-editing has raised the possibility of being able to treat or cure cancers, but key challenges remain, including efficient delivery, in vivo efficacy, and its safety profile. Ideal targets for cancer therapy are oncogenes, that when edited, cause cell death. Here, we show, using the human papillomavirus (HPV) type 16 cancer cell line TC1, that CRISPR/Cas9 targeting the E7 oncogene and packaged in PEGylated liposomes cleared established tumours in immunocompetent mice. Treatment caused no significant toxicity in the spleen or liver. An ideal therapeutic outcome would be the induction of an immunogenic cell death (ICD), such that recurrent tumours would be eliminated by the host immune system. We show here for the first time that CRISPR/Cas9-mediated cell death via targeting E7 did not result in ICD. Overall, our data show that in vivo CRISPR/Cas targeting of oncogenes is an effective treatment approach for cancer.
Collapse
|
20
|
Delivery of ionizable hydrophilic drugs based on pharmaceutical formulation of ion pairs and ionic liquids. Eur J Pharm Biopharm 2020; 156:203-218. [DOI: 10.1016/j.ejpb.2020.09.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 09/17/2020] [Indexed: 12/12/2022]
|
21
|
Boca S, Gulei D, Zimta AA, Onaciu A, Magdo L, Tigu AB, Ionescu C, Irimie A, Buiga R, Berindan-Neagoe I. Nanoscale delivery systems for microRNAs in cancer therapy. Cell Mol Life Sci 2020; 77:1059-1086. [PMID: 31637450 PMCID: PMC11105078 DOI: 10.1007/s00018-019-03317-9] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 07/26/2019] [Accepted: 09/20/2019] [Indexed: 12/15/2022]
Abstract
Concomitant with advances in research regarding the role of miRNAs in sustaining carcinogenesis, major concerns about their delivery options for anticancer therapies have been raised. The answer to this problem may come from the world of nanoparticles such as liposomes, exosomes, polymers, dendrimers, mesoporous silica nanoparticles, quantum dots and metal-based nanoparticles which have been proved as versatile and valuable vehicles for many biomolecules including miRNAs. In another train of thoughts, the general scheme of miRNA modulation consists in inhibition of oncomiRNA expression and restoration of tumor suppressor ones. The codelivery of two miRNAs or miRNAs in combination with chemotherapeutics or small molecules was also proposed. The present review presents the latest advancements in miRNA delivery based on nanoparticle-related strategies.
Collapse
Affiliation(s)
- Sanda Boca
- Nanobiophotonics and Laser Microspectroscopy Center, Interdisciplinary Research Institute on Bio-Nano-Sciences, Babes-Bolyai University, 42 T. Laurian, 400271, Cluj-Napoca, Romania
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu", University of Medicine and Pharmacy, 23 Marinescu Street, 400337, Cluj-Napoca, Romania
| | - Diana Gulei
- MEDFUTURE-Research Center for Advanced Medicine, "Iuliu-Hatieganu" University of Medicine and Pharmacy, 23 Marinescu Street, Cluj-Napoca, Romania
| | - Alina-Andreea Zimta
- MEDFUTURE-Research Center for Advanced Medicine, "Iuliu-Hatieganu" University of Medicine and Pharmacy, 23 Marinescu Street, Cluj-Napoca, Romania
| | - Anca Onaciu
- MEDFUTURE-Research Center for Advanced Medicine, "Iuliu-Hatieganu" University of Medicine and Pharmacy, 23 Marinescu Street, Cluj-Napoca, Romania
| | - Lorand Magdo
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu", University of Medicine and Pharmacy, 23 Marinescu Street, 400337, Cluj-Napoca, Romania
| | - Adrian Bogdan Tigu
- MEDFUTURE-Research Center for Advanced Medicine, "Iuliu-Hatieganu" University of Medicine and Pharmacy, 23 Marinescu Street, Cluj-Napoca, Romania
| | - Calin Ionescu
- 5th Surgical Department, Municipal Hospital, Cluj-Napoca, Romania
- "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Alexandru Irimie
- Department of Oncological Surgery and Gynecological Oncology, 400015, Cluj-Napoca, Romania
- Department of Surgery, The Oncology Institute "Prof. Dr. Ion Chiricuta", 400015, Cluj-Napoca, Romania
| | - Rares Buiga
- Department of Pathology, "Prof Dr. Ion Chiricuta" Oncology Institute, Cluj-Napoca, Romania.
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu", University of Medicine and Pharmacy, 23 Marinescu Street, 400337, Cluj-Napoca, Romania.
- MEDFUTURE-Research Center for Advanced Medicine, "Iuliu-Hatieganu" University of Medicine and Pharmacy, 23 Marinescu Street, Cluj-Napoca, Romania.
- Department of Functional Genomics and Experimental Pathology, The Oncology Institute "Prof. Dr. Ion Chiricuta", 34-36 Republicii Street, Cluj-Napoca, Romania.
| |
Collapse
|
22
|
Villamizar O, Waters SA, Scott T, Saayman S, Grepo N, Urak R, Davis A, Jaffe A, Morris KV. Targeted Activation of Cystic Fibrosis Transmembrane Conductance Regulator. Mol Ther 2019; 27:1737-1748. [PMID: 31383454 PMCID: PMC6822231 DOI: 10.1016/j.ymthe.2019.07.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 06/24/2019] [Accepted: 07/03/2019] [Indexed: 01/05/2023] Open
Abstract
Cystic fibrosis (CF) is caused by mutations in the CF transmembrane conductance regulator (CFTR) gene. The majority of CFTR mutations result in impaired chloride channel function as only a fraction of the mutated CFTR reaches the plasma membrane. The development of a therapeutic approach that facilitates increased cell-surface expression of CFTR could prove clinically relevant. Here, we evaluate and contrast two molecular approaches to activate CFTR expression. We find that an RNA-guided nuclease null Cas9 (dCas9) fused with a tripartite activator, VP64-p65-Rta can activate endogenous CFTR in cultured human nasal epithelial cells from CF patients. We also find that targeting BGas, a long non-coding RNA involved in transcriptionally modulating CFTR expression with a gapmer, induced both strong knockdown of BGas and concordant activation of CFTR. Notably, the gapmer can be delivered to target cells when generated as electrostatic particles with recombinant HIV-Tat cell penetrating peptide (CPP), when packaged into exosomes, or when loaded into lipid nanoparticles (LNPs). Treatment of patient-derived human nasal epithelial cells containing F508del with gapmer-CPP, gapmer-exosomes, or LNPs resulted in increased expression and function of CFTR. Collectively, these observations suggest that CRISPR/dCas-VPR (CRISPR) and BGas-gapmer approaches can target and specifically activate CFTR.
Collapse
Affiliation(s)
- Olga Villamizar
- Center for Gene Therapy, City of Hope-Beckman Research Institute at the City of Hope, Duarte, CA 91010, USA
| | - Shafagh A Waters
- Faculty of Medicine, School of Women's & Children's Health, University of New South Wales (UNSW), Sydney, NSW, Australia; Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), School of Women's & Children's Health, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Tristan Scott
- Center for Gene Therapy, City of Hope-Beckman Research Institute at the City of Hope, Duarte, CA 91010, USA
| | - Sheena Saayman
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Nicole Grepo
- Center for Gene Therapy, City of Hope-Beckman Research Institute at the City of Hope, Duarte, CA 91010, USA
| | - Ryan Urak
- Center for Gene Therapy, City of Hope-Beckman Research Institute at the City of Hope, Duarte, CA 91010, USA
| | - Alicia Davis
- Center for Gene Therapy, City of Hope-Beckman Research Institute at the City of Hope, Duarte, CA 91010, USA
| | - Adam Jaffe
- Faculty of Medicine, School of Women's & Children's Health, University of New South Wales (UNSW), Sydney, NSW, Australia; Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), School of Women's & Children's Health, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia; Department of Respiratory Medicine, Sydney Children's Hospital, Sydney, NSW, Australia
| | - Kevin V Morris
- Center for Gene Therapy, City of Hope-Beckman Research Institute at the City of Hope, Duarte, CA 91010, USA.
| |
Collapse
|
23
|
Jubair L, Fallaha S, McMillan NAJ. Systemic Delivery of CRISPR/Cas9 Targeting HPV Oncogenes Is Effective at Eliminating Established Tumors. Mol Ther 2019; 27:2091-2099. [PMID: 31537455 DOI: 10.1016/j.ymthe.2019.08.012] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 07/30/2019] [Accepted: 08/21/2019] [Indexed: 12/15/2022] Open
Abstract
The recent advancements in CRISPR/Cas9 engineering have resulted in the development of more targeted and potentially safer gene therapies. The challenge in the cancer setting is knowing the driver oncogenes responsible, and the translation of these therapies is hindered by effective and safe delivery methods to target organs with minimal systemic toxicities, on-target specificity of gene editing, and demonstrated lack of long-term adverse events. Using a model system based on cervical cancer, which is driven by the ongoing expression of the human papillomavirus E6 and E7 proteins, we show that CRISPR/Cas9 delivered systemically in vivo using PEGylated liposomes results in tumor elimination and complete survival in treated animals. We compared treatment and editing efficiency of two Cas9 variants, wild-type (WT) Cas9 and the highly specific FokI-dCas9, and showed that the latter was not effective. We also explored high-fidelity repair but found that repair was inefficient, occurring in 6%-8% of cells, whereas non-homologous end joining (NHEJ) was highly efficient, occurring in ∼80% of the cells. Finally, we explored the post gene-editing events in tumors and showed that cell death is induced by apoptosis. Overall, our work demonstrates that in vivo CRISPR/Cas editing treatment of preexisting tumors is completely effective despite the large payloads.
Collapse
Affiliation(s)
- Luqman Jubair
- School of Medical Sciences, Griffith University, Gold Coast, QLD 4222 Australia; Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD 4222 Australia
| | - Sora Fallaha
- School of Medical Sciences, Griffith University, Gold Coast, QLD 4222 Australia; Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD 4222 Australia
| | - Nigel A J McMillan
- School of Medical Sciences, Griffith University, Gold Coast, QLD 4222 Australia; Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD 4222 Australia.
| |
Collapse
|
24
|
Clarke D, Idris A, McMillan NAJ. Development of novel lipidic particles for siRNA delivery that are highly effective after 12 months storage. PLoS One 2019; 14:e0211954. [PMID: 30735545 PMCID: PMC6368384 DOI: 10.1371/journal.pone.0211954] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 01/24/2019] [Indexed: 01/30/2023] Open
Abstract
Liposomes are versatile and well-proven as a means to deliver nucleic acids into cells. Most of the formulation procedures used are labour intensive and result in unstable end products. We have previously reported on the development of a simple, yet efficient, hydration-of-freeze-dried-matrix (HFDM) method to entrap siRNA within lipid particles. Here we show that the particles are stable up to 12 months after storage at room temperature (RT), 4°C or -20°C. While RT storage results in changes in particle size and polydispersity, gene silencing of all particles was similar to freshly prepared particles following storage for 3, 6, 9 or 12 months at all temperatures. This is the first report of such long-term stability in siRNA-loaded liposomes.
Collapse
Affiliation(s)
- Daniel Clarke
- Menzies Health Institute Queensland, School of Medical Science, Griffith University, Southport, Australia
| | - Adi Idris
- Menzies Health Institute Queensland, School of Medical Science, Griffith University, Southport, Australia
- * E-mail:
| | - Nigel A. J. McMillan
- Menzies Health Institute Queensland, School of Medical Science, Griffith University, Southport, Australia
| |
Collapse
|
25
|
McKiernan PJ, Lynch P, Ramsey JM, Cryan SA, Greene CM. Knockdown of Gene Expression in Macrophages by microRNA Mimic-Containing Poly (Lactic- co-glycolic Acid) Microparticles. MEDICINES (BASEL, SWITZERLAND) 2018; 5:E133. [PMID: 30558310 PMCID: PMC6313440 DOI: 10.3390/medicines5040133] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 12/10/2018] [Accepted: 12/14/2018] [Indexed: 01/05/2023]
Abstract
Background: microRNA (miRNA) regulate target gene expression through translational repression and/or mRNA degradation and are involved in the regulation of inflammation. Macrophages are key inflammatory cells that are important in chronic inflammatory lung diseases such as cystic fibrosis (CF). Macrophage-expressed miRNA represent therapeutic drug targets, yet delivery of nucleic acids to macrophages has proved challenging. Methods: miRNAs were encapsulated in poly (lactic-co-glycolic acid) (PLGA)-based microparticles using double emulsion solvent evaporation and characterised for physicochemical features. Phorbol myristic acetate (PMA)-differentiated U937 macrophages were transfected with empty PLGA microparticles or those encapsulating a premiR-19b-3p or scrambled control miRNA mimic. miRNA internalisation and knockdown of a miR-19b-3p target gene, secretory leucoprotease inhibitor (SLPI), were determined by qRT-PCR. Results: Microparticle formulations were consistently found to be 2⁻3μm and all had a negative ζ potential (-5 mV to -14 mV). Encapsulation efficiency of premiR-19b-3p was 37.6 ± 13.4%. Levels of mature miR-19b-3p were higher in macrophages after delivery of premiR-19b-3p microparticles compared to empty or scrambled control miRNA-containing microparticles. Significant SLPI knockdown was achieved 72 hours post-delivery of premiR-19b-3p microparticles compared to controls. Conclusions: miRNA-encapsulating PLGA microparticles offer a new treatment paradigm for delivery to macrophages that could potentially be administered to CF lungs via inhalation.
Collapse
Affiliation(s)
- Paul J McKiernan
- Department of Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland.
| | - Patrick Lynch
- Department of Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland.
- Drug Delivery and Advanced Materials Team, School of Pharmacy, Royal College of Surgeons in Ireland, Dublin 2, Ireland.
| | - Joanne M Ramsey
- Drug Delivery and Advanced Materials Team, School of Pharmacy, Royal College of Surgeons in Ireland, Dublin 2, Ireland.
- Centre for Research in Medical Devices (CURAM), RCSI, Dublin and National University of Ireland, Galway H91 HE94, Ireland.
- Trinity Centre for Bioengineering, Trinity College Dublin, Dublin 2, Ireland.
- Tissue Engineering Research Group, Royal College of Surgeons in Ireland, 123 St Stephens Green, Dublin 2, Ireland.
| | - Sally Ann Cryan
- Drug Delivery and Advanced Materials Team, School of Pharmacy, Royal College of Surgeons in Ireland, Dublin 2, Ireland.
- Centre for Research in Medical Devices (CURAM), RCSI, Dublin and National University of Ireland, Galway H91 HE94, Ireland.
- Trinity Centre for Bioengineering, Trinity College Dublin, Dublin 2, Ireland.
- Tissue Engineering Research Group, Royal College of Surgeons in Ireland, 123 St Stephens Green, Dublin 2, Ireland.
| | - Catherine M Greene
- Lung Biology Group, Department of Clinical Microbiology, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland.
| |
Collapse
|
26
|
Liposomal Delivery of miR-34b-5p Induced Cancer Cell Death in Thyroid Carcinoma. Cells 2018; 7:cells7120265. [PMID: 30544959 PMCID: PMC6315437 DOI: 10.3390/cells7120265] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 11/30/2018] [Accepted: 12/10/2018] [Indexed: 01/03/2023] Open
Abstract
This study aims to determine the functional roles of microRNA-34b-5p (miR-34b) in the suppression of anaplastic thyroid carcinoma. We used hydration-of-freeze-dried-matrix (HFDM) formulated liposomes (liposome-loaded miR-34b) for effective delivery of miR-34b to anaplastic thyroid carcinoma in vitro and in vivo. Real time polymerase chain was used to determine the level of miR-34b. Immunocytochemistry, Western blot and ELISA were carried out to determine the effect of this manipulation on VEGF-A expression. In addition, an in vivo xenotransplantation mouse model was used to investigate the functional roles of overexpression of miR-34b in the carcinoma. In anaplastic thyroid carcinoma cells, miR-34b expression was low and significant overexpression (p < 0.05) was noted following transfection with liposome-loaded miR-34b. The miR-34b overexpressed thyroid carcinoma cell lines showed reduction in VEGF-A protein expression, decreased cell proliferation, decreased wound healing, reduced cell cycle progression and increased apoptosis (p < 0.05). In in vivo experiments, when compared to control groups, smaller tumours formed upon intravenous administration of liposome-loaded miR-34b. To conclude, the current study confirmed the tumour suppressor properties of miR-34b via VEGF-A regulation in anaplastic thyroid carcinoma. In addition, delivery of miR-34b using cationic liposome could be a useful therapeutic strategy for targeting therapy in the carcinoma.
Collapse
|
27
|
Mohammadi A, Mansoori B, Savadi P, Khaze V, Minouei M, McMillan NAJ, Hallaj-Nezhadi S, Baradaran B. Targeting of high mobility group A2 by small interfering RNA-loaded nanoliposome-induced apoptosis and migration inhibition in gastrointestinal cancer cells. J Cell Biochem 2018; 120:9203-9212. [PMID: 30507008 DOI: 10.1002/jcb.28196] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 11/08/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Considering the complex nature of gastrointestinal cancer, different methods including surgery, radiotherapy, and chemotherapy are considered for the treatment. Novel strategies including silencing of oncogenes using safe delivery systems could be considered as a novel approach in colorectal cancer treatment. The aim of this study was to investigate the silencing effect of high mobility group A2 (HMGA2) small interfering RNA (siRNA)-loaded nanoliposomes on gastrointestinal cancers. METHODS The siRNA-lipoplexes were prepared using dioleoyl trimethylammonium propane (DOTAP)/cholesterol (Chol)/1, 2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) through the freeze-drying of a monophase solution method. The size, polydispersity index (PDI), and zeta-potential of nanoliposomes were determined using Zetasizer analyzer. The morphology of the nanoliposomes was determined by transmission electron microscopy (TEM). The agarose gel-retardation assay was carried out to confirm the loading of siRNAs into liposome. The silencing of the HMGA2 in cancer cells was evaluated by quantitative reverse-transcription polymerase chain reaction (qRT-PCR). The effect of liposomes on cell cytotoxicity was studied by MTT assay. The inhibitory effect of siRNA-loaded liposomes was evaluated by a wound-healing assay. The apoptosis induction was investigated via the annexin V/propidium iodide assay. RESULTS The size, PDI, and zeta-potential of the prepared liposomes were found to be 350 nm, 0.67, and 86.3 mV, respectively. They were spherical in shape and could efficiently associate with siRNA. The results of gene silencing showed that the optimum condition of HMGA2 silencing was 80 pmol HMGA2 and 24 hours after treatment in each cancer cell lines. MTT assays indicated that silencing of HMGA2 in optimal condition could reduce the viability of the cancer cells more than 60% in the three cell lines. The result of the apoptosis assay showed more than 50% of the cell deaths related to the apoptosis in all three cell lines. The gene expression evaluation confirmed that apoptosis was induced via the intrinsic pathway inducing both caspase-3 and -9 expressions. Also, the reduction in Bcl2 expression confirmed the activation apoptosis pathway in the treated cancer cells. The wound-healing assay showed the suppression of cancer cell migration after treatment with the prepared nanoliposomes. CONCLUSION The results of this study showed the HMGA2 siRNA-loaded nanoliposomes could be effective in the treatment of gastrointestinal cancers.
Collapse
Affiliation(s)
- Ali Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pouria Savadi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahid Khaze
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahsa Minouei
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nigel A J McMillan
- School of Medical Sciences and Menzies Health Institute Queensland, Griffith University, Southport, Australia
| | | | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
28
|
Shaikh MH, Idris A, Johnson NW, Fallaha S, Clarke DTW, Martin D, Morgan IM, Gabrielli B, McMillan NAJ. Aurora kinases are a novel therapeutic target for HPV-positive head and neck cancers. Oral Oncol 2018; 86:105-112. [PMID: 30409290 DOI: 10.1016/j.oraloncology.2018.09.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 08/30/2018] [Accepted: 09/08/2018] [Indexed: 01/22/2023]
Abstract
OBJECTIVES Human papilloma virus (HPV) is the main culprit in cancers of the cervix, penis, anus, skin, eye and head and neck. Current treatments for HPV cancers have not altered survival outcomes for 30 years and there is a significant lack of targeted therapeutic agents in the management of advanced HPV-related HNSCC. Here we show that survival and maintenance of HPV-positive HNC cells relies on the continuous expression of the major HPV oncogene, E7, and that Aurora kinases are critical for survival of high-risk HPV-positive HNC cells. MATERIALS AND METHODS To assess the role of HPV E7 on HNC cell survival, RNA interference (RNAi) of the E7 gene was initially performed. Using an Aurora kinase inhibitor, Alisertib, the role of Aurora kinases in the carcinogenesis of HPV E7 positive HNC tumour lines was then investigated. An in vivo HNC xenograft model was also utilised to assess loss of tumour volume in response to RNAi E7 gene silencing and Alisertib treatment. RESULTS RNAi silencing of the HPV E7 gene inhibited the growth of HPV-positive HNC cells and in vivo tumour load. We show that HPV E7 oncogene expression confers sensitivity to Alisertib on HNC cells where Alisertib-mediated loss in in vitro cell viability and in vivo tumour load is dependent on E7 expression. Moreover, Aurora kinase inhibition induced degradation of MCL-1 in HPV E7-expressing HNC cells. CONCLUSION Overall, we show that Aurora kinases are a novel therapeutic target for HPV-positive HNCs. It might be feasible to combine Aurora kinase and MCL-1 inhibitors for future HNC therapies.
Collapse
Affiliation(s)
- Mushfiq H Shaikh
- Menzies Health Institute Queensland, School of Dentistry and Oral Health, Griffith University, Southport, Queensland, Australia; Menzies Health Institute Queensland, School of Medical Science, Griffith University, Southport, Queensland, Australia
| | - Adi Idris
- Menzies Health Institute Queensland, School of Medical Science, Griffith University, Southport, Queensland, Australia; Department of Clinical Laboratory, Kaifeng Central Hospital, Kaifeng City, China
| | - Newell W Johnson
- Menzies Health Institute Queensland, School of Dentistry and Oral Health, Griffith University, Southport, Queensland, Australia; Dental Institute, King's College London, London, United Kingdom
| | - Sora Fallaha
- Menzies Health Institute Queensland, School of Medical Science, Griffith University, Southport, Queensland, Australia
| | - Daniel T W Clarke
- Menzies Health Institute Queensland, School of Medical Science, Griffith University, Southport, Queensland, Australia
| | - David Martin
- Mater Research Institute, University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia; Diamantina Institute, University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Iain M Morgan
- Department of Oral and Craniofacial Molecular Biology, VCU Philips Institute for Oral Health Research, Virginia Commonwealth University School of Dentistry, Richmond, VA, USA
| | - Brian Gabrielli
- Mater Research Institute, University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Nigel A J McMillan
- Menzies Health Institute Queensland, School of Medical Science, Griffith University, Southport, Queensland, Australia.
| |
Collapse
|
29
|
Chen C, Yang Z, Tang X. Chemical modifications of nucleic acid drugs and their delivery systems for gene-based therapy. Med Res Rev 2018; 38:829-869. [PMID: 29315675 DOI: 10.1002/med.21479] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Revised: 11/27/2017] [Accepted: 11/27/2017] [Indexed: 12/12/2022]
Abstract
Gene-based therapy is one of essential therapeutic strategies for precision medicine through targeting specific genes in specific cells of target tissues. However, there still exist many problems that need to be solved, such as safety, stability, selectivity, delivery, as well as immunity. Currently, the key challenges of gene-based therapy for clinical potential applications are the safe and effective nucleic acid drugs as well as their safe and efficient gene delivery systems. In this review, we first focus on current nucleic acid drugs and their formulation in clinical trials and on the market, including antisense oligonucleotide, siRNA, aptamer, and plasmid nucleic acid drugs. Subsequently, we summarize different chemical modifications of nucleic acid drugs as well as their delivery systems for gene-based therapeutics in vivo based on nucleic acid chemistry and nanotechnology methods.
Collapse
Affiliation(s)
- Changmai Chen
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Zhenjun Yang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Xinjing Tang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| |
Collapse
|
30
|
Okuda T. [Development of Inhalable Dry Powder Formulations Loaded with Nanoparticles Maintaining Their Original Physical Properties and Functions]. YAKUGAKU ZASSHI 2017; 137:1339-1348. [PMID: 29093369 DOI: 10.1248/yakushi.17-00155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Functional nanoparticles, such as liposomes and polymeric micelles, are attractive drug delivery systems for solubilization, stabilization, sustained release, prolonged tissue retention, and tissue targeting of various encapsulated drugs. For their clinical application in therapy for pulmonary diseases, the development of dry powder inhalation (DPI) formulations is considered practical due to such advantages as: (1) it is noninvasive and can be directly delivered into the lungs; (2) there are few biocomponents in the lungs that interact with nanoparticles; and (3) it shows high storage stability in the solid state against aggregation or precipitation of nanoparticles in water. However, in order to produce effective nanoparticle-loaded dry powders for inhalation, it is essential to pursue an innovative and comprehensive formulation strategy in relation to composition and powderization which can achieve (1) the particle design of dry powders with physical properties suitable for pulmonary delivery through inhalation, and (2) the effective reconstitution of nanoparticles that will maintain their original physical properties and functions after dissolution of the powders. Spray-freeze drying (SFD) is a relatively new powderization technique combining atomization and lyophilization, which can easily produce highly porous dry powders from an aqueous sample solution. Previously, we advanced the optimization of components and process conditions for the production of SFD powders suitable to DPI application. This review describes our recent results in the development of novel DPI formulations effectively loaded with various nanoparticles (electrostatic nanocomplexes for gene therapy, liposomes, and self-assembled lipid nanoparticles), based on SFD.
Collapse
Affiliation(s)
- Tomoyuki Okuda
- Drug Delivery Research, Faculty of Pharmacy, Meijo University
| |
Collapse
|
31
|
Zununi Vahed S, Salehi R, Davaran S, Sharifi S. Liposome-based drug co-delivery systems in cancer cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 71:1327-1341. [DOI: 10.1016/j.msec.2016.11.073] [Citation(s) in RCA: 190] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 11/10/2016] [Accepted: 11/21/2016] [Indexed: 02/07/2023]
|
32
|
Ball RL, Bajaj P, Whitehead KA. Achieving long-term stability of lipid nanoparticles: examining the effect of pH, temperature, and lyophilization. Int J Nanomedicine 2016; 12:305-315. [PMID: 28115848 PMCID: PMC5221800 DOI: 10.2147/ijn.s123062] [Citation(s) in RCA: 168] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The broadest clinical application of siRNA therapeutics will be facilitated by drug-loaded delivery systems that maintain stability and potency for long times under ambient conditions. In the present study, we seek to better understand the stability and effect of storage conditions on lipidoid nanoparticles (LNPs), which have been previously shown by our group and others to potently deliver RNA to various cell and organ targets both in vitro and in vivo. Specifically, this study evaluates the influence of pH, temperature, and lyophilization on LNP efficacy in HeLa cells. When stored under aqueous conditions, we found that refrigeration (2°C) kept LNPs the most stable over 150 days compared to storage in the −20°C freezer or at room temperature. Because the pH of the storage buffer was not found to influence stability, it is suggested that the LNPs be stored under physiologically appropriate conditions (pH 7) for ease of use. Although aggregation and loss of efficacy were observed when LNPs were subjected to freeze–thaw cycles, their stability was retained with the use of the cryoprotectants, trehalose, and sucrose. Initially, lyophilization of the LNPs followed by reconstitution in aqueous buffer also led to reductions in efficacy, most likely due to aggregation upon reconstitution. Although the addition of ethanol to the reconstitution buffer restored efficacy, this approach is not ideal, as LNP solutions would require dialysis prior to use. Fortunately, we found that the addition of trehalose or sucrose to LNP solutions prior to lyophilization facilitated room temperature storage and reconstitution in aqueous buffer without diminishing delivery potency.
Collapse
Affiliation(s)
| | - Palak Bajaj
- Department of Chemical Engineering; Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Kathryn A Whitehead
- Department of Chemical Engineering; Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| |
Collapse
|
33
|
Sheng YH, He Y, Hasnain SZ, Wang R, Tong H, Clarke DT, Lourie R, Oancea I, Wong KY, Lumley JW, Florin TH, Sutton P, Hooper JD, McMillan NA, McGuckin MA. MUC13 protects colorectal cancer cells from death by activating the NF-κB pathway and is a potential therapeutic target. Oncogene 2016; 36:700-713. [PMID: 27399336 PMCID: PMC5541270 DOI: 10.1038/onc.2016.241] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Revised: 05/24/2016] [Accepted: 06/01/2016] [Indexed: 02/07/2023]
Abstract
MUC13 is a transmembrane mucin glycoprotein that is over produced by many cancers, although its functions are not fully understood. Nuclear factor-κB (NF-κB) is a key transcription factor promoting cancer cell survival, but therapeutically targeting this pathway has proved difficult because NF-κB has pleiotropic functions. Here, we report that MUC13 prevents colorectal cancer cell death by promoting two distinct pathways of NF-kB activation, consequently upregulating BCL-XL. MUC13 promoted tumor necrosis factor (TNF)-induced NF-κB activation by interacting with TNFR1 and the E3 ligase, cIAP1, to increase ubiquitination of RIPK1. MUC13 also promoted genotoxin-induced NF-κB activation by increasing phosphorylation of ATM and SUMOylation of NF-κB essential modulator. Moreover, elevated expression of cytoplasmic MUC13 and NF-κB correlated with colorectal cancer progression and metastases. Our demonstration that MUC13 enhances NF-κB signaling in response to both TNF and DNA-damaging agents provides a new molecular target for specific inhibition of NF-κB activation. As proof of principle, silencing MUC13 sensitized colorectal cancer cells to killing by cytotoxic drugs and inflammatory signals and abolished chemotherapy-induced enrichment of CD133+ CD44+ cancer stem cells, slowed xenograft growth in mice, and synergized with 5-fluourouracil to induce tumor regression. Therefore, these data indicate that combining chemotherapy and MUC13 antagonism could improve the treatment of metastatic cancers.
Collapse
Affiliation(s)
- Y H Sheng
- Inflammatory Disease Biology and Therapeutics Group-Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Y He
- Cancer Biology Group, Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - S Z Hasnain
- Inflammatory Disease Biology and Therapeutics Group-Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - R Wang
- Inflammatory Disease Biology and Therapeutics Group-Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - H Tong
- Inflammatory Disease Biology and Therapeutics Group-Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - D T Clarke
- Molecular Basis of Disease Program, School of Medical Sciences, Griffith University, Gold Coast Campus, Southport, Queensland, Australia
| | - R Lourie
- Inflammatory Disease Biology and Therapeutics Group-Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia.,Inflammatory Bowel Diseases Group, Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - I Oancea
- Inflammatory Disease Biology and Therapeutics Group-Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia.,Inflammatory Bowel Diseases Group, Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - K Y Wong
- Inflammatory Disease Biology and Therapeutics Group-Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - J W Lumley
- Wesley Hospital, Auchenflower, Australia
| | - T H Florin
- Inflammatory Bowel Diseases Group, Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - P Sutton
- Mucosal Immunology, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Melbourne, Victoria, Australia.,Centre for Animal Biotechnology, School of Veterinary and Agricultural Science, University of Melbourne, Melbourne, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Victoria, Australia
| | - J D Hooper
- Cancer Biology Group, Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - N A McMillan
- Molecular Basis of Disease Program, School of Medical Sciences, Griffith University, Gold Coast Campus, Southport, Queensland, Australia
| | - M A McGuckin
- Inflammatory Disease Biology and Therapeutics Group-Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
34
|
Effect of surface properties on liposomal siRNA delivery. Biomaterials 2015; 79:56-68. [PMID: 26695117 DOI: 10.1016/j.biomaterials.2015.11.056] [Citation(s) in RCA: 168] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 11/11/2015] [Accepted: 11/29/2015] [Indexed: 12/18/2022]
Abstract
Liposomes are one of the most widely investigated carriers for siRNA delivery. The surface properties of liposomal carriers, including the surface charge, PEGylation, and ligand modification can significantly affect the gene silencing efficiency. Three barriers of systemic siRNA delivery (long blood circulation, efficient tumor penetration and efficient cellular uptake/endosomal escape) are analyzed on liposomal carriers with different surface charges, PEGylations and ligand modifications. Cationic formulations dominate siRNA delivery and neutral formulations also have good performance while anionic formulations are generally not proper for siRNA delivery. The PEG dilemma (prolonged blood circulation vs. reduced cellular uptake/endosomal escape) and the side effect of repeated PEGylated formulation (accelerated blood clearance) were discussed. Effects of ligand modification on cationic and neutral formulations were analyzed. Finally, we summarized the achievements in liposomal siRNA delivery, outlined existing problems and provided some future perspectives.
Collapse
|
35
|
Nascimento TL, Hillaireau H, Noiray M, Bourgaux C, Arpicco S, Pehau-Arnaudet G, Taverna M, Cosco D, Tsapis N, Fattal E. Supramolecular Organization and siRNA Binding of Hyaluronic Acid-Coated Lipoplexes for Targeted Delivery to the CD44 Receptor. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2015; 31:11186-11194. [PMID: 26375384 DOI: 10.1021/acs.langmuir.5b01979] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The dynamics of the formation of siRNA-lipoplexes coated with hyaluronic acid (HA) and the parameters influencing their supramolecular organization were studied. The insertion of a HA-dioleylphosphatidylethanolamine (DOPE) conjugate in the liposome structure as well as subsequent complexation with siRNA increased the liposome size. Lipoplexes were around 110 nm at high ± charge ratios with a zeta potential around +50 mV and around 230 nm at low ± ratios, with a zeta potential that decreased to negative values, reaching -45 mV. The addition of the conjugate did not compromise siRNA binding to liposomes, although these nucleic acids induced a displacement of part of the HA-DOPE conjugate upon lipoplex formation, as confirmed by capillary electrophoresis. Isothermal titration calorimetry, X-ray diffraction studies, and cryo-TEM microscopy demonstrated that in addition to electrostatic interactions with siRNA a rearrangement of the lipid bilayers takes place, resulting in condensed oligolamellar vesicles. This phenomenon is dependent on the number of siRNA molecules and the degree of modification with HA. Finally, the suitable positioning of HA on the lipoplex surface and its ability to bind specifically to the CD44 receptors in a concentration-dependent manner was demonstrated by surface plasmon resonance analysis.
Collapse
Affiliation(s)
- Thais L Nascimento
- Faculté de Pharmacie, Institut Galien Paris-Sud. Université Paris-Sud , LabEx LERMIT, 5 rue JB Clément, 92296 Châtenay-Malabry Cedex, France
- CNRS, UMR 8612, 5 rue JB Clément, 92296 Châtenay-Malabry Cedex, France
- CAPES Foundation, Ministry of Education of Brazil, Brasília DF 70040-020, Brazil
| | - Hervé Hillaireau
- Faculté de Pharmacie, Institut Galien Paris-Sud. Université Paris-Sud , LabEx LERMIT, 5 rue JB Clément, 92296 Châtenay-Malabry Cedex, France
- CNRS, UMR 8612, 5 rue JB Clément, 92296 Châtenay-Malabry Cedex, France
| | - Magali Noiray
- Faculté de Pharmacie, Institut Galien Paris-Sud. Université Paris-Sud , LabEx LERMIT, 5 rue JB Clément, 92296 Châtenay-Malabry Cedex, France
- CNRS, UMR 8612, 5 rue JB Clément, 92296 Châtenay-Malabry Cedex, France
| | - Claudie Bourgaux
- Faculté de Pharmacie, Institut Galien Paris-Sud. Université Paris-Sud , LabEx LERMIT, 5 rue JB Clément, 92296 Châtenay-Malabry Cedex, France
- CNRS, UMR 8612, 5 rue JB Clément, 92296 Châtenay-Malabry Cedex, France
| | - Silvia Arpicco
- Dipartemento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, Facoltà di Farmacia , Via Pietro Giuria 9, 10125 Torino, Italy
| | - Gérard Pehau-Arnaudet
- Institut Pasteur, Plate-Forme de Microscopie Ultrastructurale, 25-28 rue du Docteur Roux, 75015 Paris, France
| | - Myriam Taverna
- Faculté de Pharmacie, Institut Galien Paris-Sud. Université Paris-Sud , LabEx LERMIT, 5 rue JB Clément, 92296 Châtenay-Malabry Cedex, France
- CNRS, UMR 8612, 5 rue JB Clément, 92296 Châtenay-Malabry Cedex, France
| | - Donato Cosco
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta" , Viale S. Venuta, Germaneto, I-88100 Catanzaro, Italy
| | - Nicolas Tsapis
- Faculté de Pharmacie, Institut Galien Paris-Sud. Université Paris-Sud , LabEx LERMIT, 5 rue JB Clément, 92296 Châtenay-Malabry Cedex, France
- CNRS, UMR 8612, 5 rue JB Clément, 92296 Châtenay-Malabry Cedex, France
| | - Elias Fattal
- Faculté de Pharmacie, Institut Galien Paris-Sud. Université Paris-Sud , LabEx LERMIT, 5 rue JB Clément, 92296 Châtenay-Malabry Cedex, France
- CNRS, UMR 8612, 5 rue JB Clément, 92296 Châtenay-Malabry Cedex, France
| |
Collapse
|
36
|
Dong H, Parekh HS, Xu ZP. Enhanced cellular delivery and biocompatibility of a small layered double hydroxide-liposome composite system. Pharmaceutics 2014; 6:584-98. [PMID: 25431895 PMCID: PMC4279134 DOI: 10.3390/pharmaceutics6040584] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 11/10/2014] [Accepted: 11/11/2014] [Indexed: 01/13/2023] Open
Abstract
The various classes of gene delivery vectors possess distinct advantages and disadvantages, each of which impacts on cargo loading, delivery and, ultimately, its function. With this in mind, herein we report on a small layered double hydroxide (sLDH)–liposome composite system, drawing upon the salient features of LDH and liposome classes of vectors, while avoiding their inherent shortfalls when used independently. sLDH–liposome composites were prepared by the hydration of freeze-dried matrix method. These composite systems, with a Z-average size of ≈200 nm, exhibited low cytotoxicity and demonstrated good suspension stability, both in water and cell culture medium after rehydration. Our studies demonstrate that short dsDNAs/ssDNAs were completely bound and protected in the composite system at an sLDH:DNA mass ratio of 20:1, regardless of the approach to DNA loading. This composite system delivered DNA to HCT-116 cells with ≈3-fold greater efficiency, when compared to sLDH alone. Our findings point towards the sLDH-liposome composite system being an effective and biocompatible gene delivery system.
Collapse
Affiliation(s)
- Haiyan Dong
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Harendra S Parekh
- Pharmacy Australia Centre of Excellence, School of Pharmacy, The University of Queensland, Brisbane, QLD 4102, Australia.
| | - Zhi Ping Xu
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
37
|
Haigh O, Depelsenaire AC, Meliga SC, Yukiko SR, McMillan NA, Frazer IH, Kendall MA. CXCL1 gene silencing in skin using liposome-encapsulated siRNA delivered by microprojection array. J Control Release 2014; 194:148-56. [DOI: 10.1016/j.jconrel.2014.08.021] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 05/09/2014] [Accepted: 08/23/2014] [Indexed: 11/26/2022]
|
38
|
Kelly C, Yadav AB, Lawlor C, Nolan K, O’Dwyer J, Greene CM, McElvaney NG, Sivadas N, Ramsey JM, Cryan SA. Therapeutic Aerosol Bioengineering of siRNA for the Treatment of Inflammatory Lung Disease by TNFα Gene Silencing in Macrophages. Mol Pharm 2014; 11:4270-9. [DOI: 10.1021/mp500473d] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
| | | | | | | | | | - Catherine M. Greene
- Department
of Medicine, Respiratory Research Division, RCSI Education and Research
Centre, Beaumont Hospital, Dublin 9, Ireland
| | - Noel G. McElvaney
- Department
of Medicine, Respiratory Research Division, RCSI Education and Research
Centre, Beaumont Hospital, Dublin 9, Ireland
| | | | - Joanne M. Ramsey
- Tissue Engineering
Research Group, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Sally-Ann Cryan
- Trinity
Centre for Bioengineering, Trinity College Dublin, Dublin 2, Ireland
- Tissue Engineering
Research Group, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| |
Collapse
|
39
|
Hong CA, Nam YS. Functional nanostructures for effective delivery of small interfering RNA therapeutics. Am J Cancer Res 2014; 4:1211-32. [PMID: 25285170 PMCID: PMC4183999 DOI: 10.7150/thno.8491] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Accepted: 02/23/2014] [Indexed: 02/04/2023] Open
Abstract
Small interfering RNA (siRNA) has proved to be a powerful tool for target-specific gene silencing via RNA interference (RNAi). Its ability to control targeted gene expression gives new hope to gene therapy as a treatment for cancers and genetic diseases. However, siRNA shows poor pharmacological properties, such as low serum stability, off-targeting, and innate immune responses, which present a significant challenge for clinical applications. In addition, siRNA cannot cross the cell membrane for RNAi activity because of its anionic property and stiff structure. Therefore, the development of a safe, stable, and efficient system for the delivery of siRNA therapeutics into the cytoplasm of targeted cells is crucial. Several nanoparticle platforms for siRNA delivery have been developed to overcome the major hurdles facing the therapeutic uses of siRNA. This review covers a broad spectrum of non-viral siRNA delivery systems developed for enhanced cellular uptake and targeted gene silencing in vitro and in vivo and discusses their characteristics and opportunities for clinical applications of therapeutic siRNA.
Collapse
|
40
|
Khairuddin N, Blake SJ, Firdaus F, Steptoe RJ, Behlke MA, Hertzog PJ, McMillan NAJ. In vivo comparison of local versus systemic delivery of immunostimulating siRNA in HPV-driven tumours. Immunol Cell Biol 2013; 92:156-63. [PMID: 24217808 DOI: 10.1038/icb.2013.75] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 10/16/2013] [Accepted: 10/16/2013] [Indexed: 12/12/2022]
Abstract
Small interfering RNAs (siRNAs) to inhibit oncogene expression and also to activate innate immune responses via Toll-like receptor (TLR) recognition have been shown to be beneficial as anti-cancer therapy in certain cancer models. In this study, we investigated the effects of local versus systemic delivery of such immune-stimulating Dicer-substrate siRNAs (IS-DsiRNAs) on a human papillomavirus (HPV)-driven tumour model. Localized siRNA delivery using intratumour injection of siRNA was able to increase siRNA delivery to the tumour compared with intravenous (IV) delivery and potently activated innate immune responses. However, IV injection remained the more effective delivery route for reducing tumour growth. Although IS-DsiRNAs activated innate immune cells and required interferon-α (IFNα) for full effect on tumour growth, we found that potent silencing siRNA acting independently of IFNα were overall more effective at inhibiting TC-1 tumour growth. Other published work utilising IS-siRNAs have been carried out on tumour models with low levels of major histocompatibility complex (MHC)-class 1, a target of natural killer cells that are potently activated by IS-siRNA. As TC-1 cells used in our study express high levels of MHC-class I, the addition of the immunostimulatory motifs may not be as beneficial in this particular tumour model. Our data suggest that selection of siRNA profile and delivery method based on tumour environment is crucial to developing siRNA-based therapies.
Collapse
Affiliation(s)
- Norliana Khairuddin
- 1] The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia [2] Immunotherapeutics Laboratory (ITL) and Centre of Excellence for Research in AIDS (CERiA), Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Stephen J Blake
- The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia
| | - Farah Firdaus
- The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia
| | - Raymond J Steptoe
- The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia
| | - Mark A Behlke
- Integrated DNA Technologies (IDT), Coralville, IA, USA
| | - Paul J Hertzog
- Monash Institute of Medical Research (MIMR), Clayton, Victoria, Australia
| | - Nigel A J McMillan
- 1] The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia [2] School of Medical Science and Griffith Health Institute, Griffith University, Southport, Queensland, Australia
| |
Collapse
|
41
|
Formulation development of lyophilized, long-term stable siRNA/oligoaminoamide polyplexes. Eur J Pharm Biopharm 2013; 85:294-305. [DOI: 10.1016/j.ejpb.2013.05.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2013] [Revised: 05/12/2013] [Accepted: 05/21/2013] [Indexed: 11/18/2022]
|
42
|
Asymmetric liposome particles with highly efficient encapsulation of siRNA and without nonspecific cell penetration suitable for target-specific delivery. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1818:1633-41. [PMID: 22465072 DOI: 10.1016/j.bbamem.2012.03.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Revised: 03/10/2012] [Accepted: 03/19/2012] [Indexed: 01/05/2023]
Abstract
The discovery of siRNA has been an important step in gene therapy, but the problem of delivering siRNA to a target organ limits its use as a therapeutic drug. Liposomes can be used as a nonviral vector to deliver siRNA to target cells. In this study we developed a novel method of producing asymmetric liposome particles (ALPs) with highly efficient siRNA encapsulation. Two kinds of lipid inverted micelles were prepared for the purpose of obtaining ALPs. The inner one is composed of ionizable cationic 1,2-dioleoyl-3-dimethylammonium-propane (DODAP) and 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), which entrap siRNA, and the outer one is composed of 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), DOPE, polyethylene glycol-1,2-distearoyl-sn-glycero-3-phosphatidylethanolamine (PEG-PE), and cholesterol. After mixing the inverted micelles, ALPs encapsulating siRNA were obtained by solvent evaporation and dialysis. This process allowed more than 90% siRNA encapsulation as well as the negatively charged surface. The ALPs protected siRNA from ribonuclease A degradation. ALPs without any surface modification elicited almost no uptake into cells, while the surface-modified ALPs with a polyarginine peptide (R12) induced nonspecific cell penetration. The conjugation of the anti-human epidermal growth factor receptor antibody (anti-EGFR) to ALPs induces an EGFR-mediated uptake into the non-small cell lung cancer cell lines but not into NIH-3T3 cells without the receptor. The siRNA encapsulated in ALPs showed the R12- or anti-EGFR-dependent target gene silencing in NCI-H322 cells. These properties of ALPs are useful for target-specific delivery of siRNA after modification of ALPs with a target-specific ligand.
Collapse
|
43
|
Efficient Biodistribution and Gene Silencing in the Lung epithelium via Intravenous Liposomal Delivery of siRNA. MOLECULAR THERAPY-NUCLEIC ACIDS 2013; 2:e96. [PMID: 23736774 PMCID: PMC3696903 DOI: 10.1038/mtna.2013.22] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
RNA interference (RNAi) may provide a therapeutic solution to many pulmonary epithelium diseases. However, the main barrier to the clinical use of RNAi remains the lack of efficient delivery vectors. Research has mainly concentrated on the intranasal route of delivery of short interfering RNA (siRNA) effector molecules for the treatment of respiratory diseases. However, this may be complicated in a diseased state due to the increased fluid production and tissue remodeling. Therefore, we investigated our hydration of a freeze-dried matrix (HFDM) formulated liposomes for systemic delivery to the lung epithelium. Here, we show that 45 ± 2% of epithelial murine lung cells receive siRNA delivery upon intravenous (IV) liposomal administration. Furthermore, we demonstrate that liposomal siRNA delivery resulted in targeted gene and protein knockdown throughout the lung, including lung epithelium. Taken together, this is the first description of lung epithelial delivery via cationic liposomes, and provides a proof of concept for the use of IV liposomal RNAi delivery to specifically knockdown targeted genes in the respiratory system. This approach may provide an attractive alternate therapeutic delivery strategy for the treatment of lung epithelium diseases.
Collapse
|
44
|
Resnier P, Montier T, Mathieu V, Benoit JP, Passirani C. A review of the current status of siRNA nanomedicines in the treatment of cancer. Biomaterials 2013; 34:6429-43. [PMID: 23727262 DOI: 10.1016/j.biomaterials.2013.04.060] [Citation(s) in RCA: 162] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 04/27/2013] [Indexed: 12/11/2022]
Abstract
RNA interference currently offers new opportunities for gene therapy by the specific extinction of targeted gene(s) in cancer diseases. However, the main challenge for nucleic acid delivery still remains its efficacy through intravenous administration. Over the last decade, many delivery systems have been developed and optimized to encapsulate siRNA and to specifically promote their delivery into tumor cells and improve their pharmacokinetics for anti-cancer purposes. This review aims to sum up the potential targets in numerous pathways and the properties of recently optimized siRNA synthetic nanomedicines with their preclinical applications and efficacy. Future perspectives in cancer treatment are discussed including promising concomitant treatment with chemotherapies or other siRNA. The outcomes in human clinical trials are also presented.
Collapse
|
45
|
Gindy ME, Leone AM, Cunningham JJ. Challenges in the pharmaceutical development of lipid-based short interfering ribonucleic acid therapeutics. Expert Opin Drug Deliv 2012; 9:171-82. [DOI: 10.1517/17425247.2012.642363] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
46
|
Chang HI, Yeh MK. Clinical development of liposome-based drugs: formulation, characterization, and therapeutic efficacy. Int J Nanomedicine 2011; 7:49-60. [PMID: 22275822 PMCID: PMC3260950 DOI: 10.2147/ijn.s26766] [Citation(s) in RCA: 284] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2011] [Indexed: 01/06/2023] Open
Abstract
Research on liposome formulations has progressed from that on conventional vesicles to new generation liposomes, such as cationic liposomes, temperature sensitive liposomes, and virosomes, by modulating the formulation techniques and lipid composition. Many research papers focus on the correlation of blood circulation time and drug accumulation in target tissues with physicochemical properties of liposomal formulations, including particle size, membrane lamellarity, surface charge, permeability, encapsulation volume, shelf time, and release rate. This review is mainly to compare the therapeutic effect of current clinically approved liposome-based drugs with free drugs, and to also determine the clinical effect via liposomal variations in lipid composition. Furthermore, the major preclinical and clinical data related to the principal liposomal formulations are also summarized.
Collapse
Affiliation(s)
- Hsin-I Chang
- Department of Biochemical Science and Technology, National Chia Yi University, Chiayi City, Taiwan
| | | |
Collapse
|
47
|
Buyens K, De Smedt SC, Braeckmans K, Demeester J, Peeters L, van Grunsven LA, de Mollerat du Jeu X, Sawant R, Torchilin V, Farkasova K, Ogris M, Sanders NN. Liposome based systems for systemic siRNA delivery: stability in blood sets the requirements for optimal carrier design. J Control Release 2011; 158:362-70. [PMID: 22023849 DOI: 10.1016/j.jconrel.2011.10.009] [Citation(s) in RCA: 149] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Revised: 09/29/2011] [Accepted: 10/09/2011] [Indexed: 02/07/2023]
Abstract
siRNA therapeutics are currently regarded as promising candidates to make a leap forward in the search for treatments of various hard to cure diseases. In order to exploit the full potential of siRNA based therapeutics, development of delivery systems that can efficiently guide the siRNA molecules to their target without major side effects will be the key to success. Lipid based delivery systems, originating from earlier research in the fields of gene delivery, are the most studied candidates for siRNA delivery. Here we discuss the requirements that need to be met by these siRNA delivery systems to ensure adequate stability after systemic application and subsequent deposition in the target tissue. The encountered hurdles in the blood stream and the solutions proposed in literature are discussed.
Collapse
Affiliation(s)
- Kevin Buyens
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Harelbekestraat 72, 9000 Ghent, Belgium
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Truong NP, Jia Z, Burgess M, Payne L, McMillan NAJ, Monteiro MJ. Self-catalyzed degradable cationic polymer for release of DNA. Biomacromolecules 2011; 12:3540-8. [PMID: 21838265 DOI: 10.1021/bm2007423] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The controlled release of siRNA or DNA complexes from cationic polymers is an important parameter design in polymer-based delivery carriers. In this work, we use the self-catalyzed degradable poly(2-dimethylaminoethyl acrylate) (PDMAEA) to strongly bind, protect, and then release oligo DNA (a mimic for siRNA) without the need for a cellular or external trigger. This self-catalyzed hydrolysis process of PDMAEA forms poly(acrylic acid) and N,N'-dimethylamino ethyl ethanol, both of which have little or no toxicity to cells, and offers the advantage of little or no toxicity to off-target cells and tissues. We found that PDMAEA makes an ideal component of a delivery carrier by protecting the oligo DNA for a sufficiently long period of time to transfect most cells (80% transfection after 4 h) and then has the capacity to release the DNA inside the cells after ~10 h. The PDMAEA formed large nanoparticle complexes with oligo DNA of ~400 nm that protected the oligo DNA from DNase in serum. The nanoparticle complexes showed no toxicity for all molecular weights at a nitrogen/phosphorus (N/P) ratio of 10. Only the higher molecular weight polymers at very high N/P ratios of 200 showed significant levels of cytotoxicity. These attributes make PDMAEA a promising candidate as a component in the design of a gene delivery carrier without the concern about accumulated toxicity of nanoparticles in the human body after multiadministration, an issue that has become increasingly more important.
Collapse
Affiliation(s)
- Nghia P Truong
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane QLD 4072, Australia
| | | | | | | | | | | |
Collapse
|
49
|
Effective Delivery of PEGylated siRNA-Containing Lipoplexes to Extraperitoneal Tumours following Intraperitoneal Administration. JOURNAL OF DRUG DELIVERY 2011; 2011:192562. [PMID: 21773042 PMCID: PMC3134833 DOI: 10.1155/2011/192562] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Accepted: 04/07/2011] [Indexed: 02/05/2023]
Abstract
Intraperitoneal (i.p.) administration of small interfering RNA (siRNA) has, to date, shown promise in treating tumours located within the peritoneal cavity. The ability of these siRNA molecules to reach extraperitoneal tumours following i.p. administration is, however, yet to be investigated. Here, we examined the impact of PEGylation on the biodistribution of i.p. administered nucleic acids-containing lipoplexes. We showed that in contrast to non-PEGylated liposomes, PEGylated liposomes can deliver siRNA efficiently to extraperitoneal tumours following i.p. administration, resulting in a 45% reduction in tumour size when the oncogene-targeted siRNA was used. This difference was likely contributed by the decreased uptake of PEGylated lipoplexes in the first-pass organs, and, in particular, we observed a 10-fold decrease in the macrophage uptake of these particles compared to non-PEGylated counterparts. Overall, our results indicated the potential of using PEGylated liposomes to deliver siRNA for the treatment of i.p. localized cancer with coexisting extraperitoneal metastasis.
Collapse
|
50
|
Koike H, Morikawa Y, Sekine Y, Matsui H, Shibata Y, Suzuki K. Survivin Is Associated With Cell Proliferation and Has a Role in 1a,25-Dihydroxyvitamin D
3
Induced Cell Growth Inhibition in Prostate Cancer. J Urol 2011; 185:1497-503. [DOI: 10.1016/j.juro.2010.12.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2010] [Indexed: 11/26/2022]
Affiliation(s)
- Hidekazu Koike
- Department of Urology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Yasuyuki Morikawa
- Department of Urology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Yoshitaka Sekine
- Department of Urology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Hiroshi Matsui
- Department of Urology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Yasuhiro Shibata
- Department of Urology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Kazuhiro Suzuki
- Department of Urology, Gunma University Graduate School of Medicine, Gunma, Japan
| |
Collapse
|