1
|
Yang C, Lin ZI, Zhang X, Xu Z, Xu G, Wang YM, Tsai TH, Cheng PW, Law WC, Yong KT, Chen CK. Recent Advances in Engineering Carriers for siRNA Delivery. Macromol Biosci 2024; 24:e2300362. [PMID: 38150293 DOI: 10.1002/mabi.202300362] [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: 08/09/2023] [Revised: 11/29/2023] [Indexed: 12/28/2023]
Abstract
RNA interference (RNAi) technology has been a promising treatment strategy for combating intractable diseases. However, the applications of RNAi in clinical are hampered by extracellular and intracellular barriers. To overcome these barriers, various siRNA delivery systems have been developed in the past two decades. The first approved RNAi therapeutic, Patisiran (ONPATTRO) using lipids as the carrier, for the treatment of amyloidosis is one of the most important milestones. This has greatly encouraged researchers to work on creating new functional siRNA carriers. In this review, the recent advances in siRNA carriers consisting of lipids, polymers, and polymer-modified inorganic particles for cancer therapy are summarized. Representative examples are presented to show the structural design of the carriers in order to overcome the delivery hurdles associated with RNAi therapies. Finally, the existing challenges and future perspective for developing RNAi as a clinical modality will be discussed and proposed. It is believed that the addressed contributions in this review will promote the development of siRNA delivery systems for future clinical applications.
Collapse
Affiliation(s)
- Chengbin Yang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Zheng-Ian Lin
- Polymeric Biomaterials Laboratory, Department of Materials and Optoelectronic Science, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan
| | - Xinmeng Zhang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Zhourui Xu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Gaixia Xu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Yu-Min Wang
- Polymeric Biomaterials Laboratory, Department of Materials and Optoelectronic Science, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan
| | - Tzu-Hsien Tsai
- Division of Cardiology and Department of Internal Medicine, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi, 60002, Taiwan
| | - Pei-Wen Cheng
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, 81362, Taiwan
- Department of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan
| | - Wing-Cheung Law
- Department of Industrial and Systems Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, 999077, P. R. China
| | - Ken-Tye Yong
- School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales, 2006, Australia
| | - Chih-Kuang Chen
- Polymeric Biomaterials Laboratory, Department of Materials and Optoelectronic Science, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan
| |
Collapse
|
2
|
Moazzam M, Zhang M, Hussain A, Yu X, Huang J, Huang Y. The landscape of nanoparticle-based siRNA delivery and therapeutic development. Mol Ther 2024; 32:284-312. [PMID: 38204162 PMCID: PMC10861989 DOI: 10.1016/j.ymthe.2024.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 10/01/2023] [Accepted: 01/05/2024] [Indexed: 01/12/2024] Open
Abstract
Five small interfering RNA (siRNA)-based therapeutics have been approved by the Food and Drug Administration (FDA), namely patisiran, givosiran, lumasiran, inclisiran, and vutrisiran. Besides, siRNA delivery to the target site without toxicity is a big challenge for researchers, and naked-siRNA delivery possesses several challenges, including membrane impermeability, enzymatic degradation, mononuclear phagocyte system (MPS) entrapment, fast renal excretion, endosomal escape, and off-target effects. The siRNA therapeutics can silence any disease-specific gene, but their intracellular and extracellular barriers limit their clinical applications. For this purpose, several modifications have been employed to siRNA for better transfection efficiency. Still, there is a quest for better delivery systems for siRNA delivery to the target site. In recent years, nanoparticles have shown promising results in siRNA delivery with minimum toxicity and off-target effects. Patisiran is a lipid nanoparticle (LNP)-based siRNA formulation for treating hereditary transthyretin-mediated amyloidosis that ultimately warrants the use of nanoparticles from different classes, especially lipid-based nanoparticles. These nanoparticles may belong to different categories, including lipid-based, polymer-based, and inorganic nanoparticles. This review briefly discusses the lipid, polymer, and inorganic nanoparticles and their sub-types for siRNA delivery. Finally, several clinical trials related to siRNA therapeutics are addressed, followed by the future prospects and conclusions.
Collapse
Affiliation(s)
- Muhammad Moazzam
- Faculty of Engineering and Science, University of Greenwich, Medway Campus, Chatham Maritime, Kent ME4 4TB, UK
| | - Mengjie Zhang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Abid Hussain
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Xiaotong Yu
- Department of Immunology, School of Basic Medical Sciences, Key Laboratory of Medical Immunology of Ministry of Health, Peking University, Beijing 100191, China.
| | - Jia Huang
- Department of Hepatobiliary Surgery, China-Japan Friendship Hospital, Beijing 100029, China.
| | - Yuanyu Huang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing 100081, China; Rigerna Therapeutics Co. Ltd., Suzhou 215127, China.
| |
Collapse
|
3
|
Ebrahimi N, Manavi MS, Nazari A, Momayezi A, Faghihkhorasani F, Rasool Riyadh Abdulwahid AH, Rezaei-Tazangi F, Kavei M, Rezaei R, Mobarak H, Aref AR, Fang W. Nano-scale delivery systems for siRNA delivery in cancer therapy: New era of gene therapy empowered by nanotechnology. ENVIRONMENTAL RESEARCH 2023; 239:117263. [PMID: 37797672 DOI: 10.1016/j.envres.2023.117263] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 09/17/2023] [Accepted: 09/27/2023] [Indexed: 10/07/2023]
Abstract
RNA interference (RNAi) is a unique treatment approach used to decrease a disease's excessive gene expression, including cancer. SiRNAs may find and destroy homologous mRNA sequences within the cell thanks to RNAi processes. However, difficulties such poor cellular uptake, off-target effects, and susceptibility to destruction by serum nucleases in the bloodstream restrict the therapeutic potential of siRNAs. Since some years ago, siRNA-based therapies have been in the process of being translated into the clinic. Therefore, the primary emphasis of this work is on sophisticated nanocarriers that aid in the transport of siRNA payloads, their administration in combination with anticancer medications, and their use in the treatment of cancer. The research looks into molecular manifestations, difficulties with siRNA transport, the design and development of siRNA-based delivery methods, and the benefits and drawbacks of various nanocarriers. The trapping of siRNA in endosomes is a challenge for the majority of delivery methods, which affects the therapeutic effectiveness. Numerous techniques for siRNA release, including as pH-responsive release, membrane fusion, the proton sponge effect, and photochemical disruption, have been studied to overcome this problem. The present state of siRNA treatments in clinical trials is also looked at in order to give a thorough and systematic evaluation of siRNA-based medicines for efficient cancer therapy.
Collapse
Affiliation(s)
- Nasim Ebrahimi
- Genetics Division, Department of Cell and Molecular Biology and Microbiology, Faculty of Science and Technology, University of Isfahan, Iran
| | | | - Ahmad Nazari
- Tehran University of Medical Science, Tehran, Iran
| | - Amirali Momayezi
- School of Chemical Engineering, Iran University of Science, and Technology, Tehran, Iran
| | | | | | - Fatemeh Rezaei-Tazangi
- Department of Anatomy, School of Medicine, Fasa University of Medical Science, Fasa, Iran
| | - Mohammed Kavei
- Department of Biology, Faculty of Science, Arak University, Arak, Iran
| | - Roya Rezaei
- Department of Microbiology, College of Science, Agriculture and Modern Technology, Shiraz Branch, Islamic Azad University, Shiraz, Iran
| | - Halimeh Mobarak
- Clinical Pathologist, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Reza Aref
- Xsphera Biosciences, Translational Medicine Group, 6 Tide Street, Boston, MA, 02210, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA.
| | - Wei Fang
- Department of Laser and Aesthetic Medicine, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China.
| |
Collapse
|
4
|
Zhu X, Chen Y, Yu D, Fang W, Liao W, Pan W. Progress in the application of nanoparticles for the treatment of fungal infections: A review. Mycology 2023; 15:1-16. [PMID: 38558835 PMCID: PMC10977003 DOI: 10.1080/21501203.2023.2285764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 10/28/2023] [Indexed: 04/04/2024] Open
Abstract
The burden of fungal infections on human health is increasing worldwide. Aspergillus, Candida, and Cryptococcus are the top three human pathogenic fungi that are responsible for over 90% of infection-related deaths. Moreover, effective antifungal therapeutics are lacking, primarily due to host toxicity, pathogen resistance, and immunodeficiency. In recent years, nanomaterials have proved not only to be more efficient antifungal therapeutic agents but also to overcome resistance against fungal medication. This review will examine the limitations of standard antifungal therapy as well as focus on the development of nanomaterials.
Collapse
Affiliation(s)
- Xinlin Zhu
- Department of Dermatology, Shanghai Key Laboratory of Molecular Medical Mycology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Youming Chen
- Department of Infectious Diseases and Immunology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Dan Yu
- Department of General Practice, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Wenjie Fang
- Department of Dermatology, Shanghai Key Laboratory of Molecular Medical Mycology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Wanqing Liao
- Department of Dermatology, Shanghai Key Laboratory of Molecular Medical Mycology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Weihua Pan
- Department of Dermatology, Shanghai Key Laboratory of Molecular Medical Mycology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| |
Collapse
|
5
|
Pourali P, Dzmitruk V, Benada O, Svoboda M, Benson V. Conjugation of microbial-derived gold nanoparticles to different types of nucleic acids: evaluation of transfection efficiency. Sci Rep 2023; 13:14669. [PMID: 37674013 PMCID: PMC10482973 DOI: 10.1038/s41598-023-41567-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 08/29/2023] [Indexed: 09/08/2023] Open
Abstract
In this study, gold nanoparticles produced by eukaryotic cell waste (AuNP), were analyzed as a transfection tool. AuNP were produced by Fusarium oxysporum and analyzed by spectrophotometry, transmission electron microscopy (TEM), scanning electron microscopy (SEM), and energy dispersive X-ray spectroscopy (EDS). Fourier transform infrared spectroscopy (FTIR) and dynamic light scattering (DLS) were used before and after conjugation with different nucleic acid (NA) types. Graphite furnace atomic absorption spectroscopy (GF-AAS) was used to determine the AuNP concentration. Conjugation was detected by electrophoresis. Confocal microscopy and quantitative real-time PCR (qPCR) were used to assess transfection. TEM, SEM, and EDS showed 25 nm AuNP with round shape. The amount of AuNP was 3.75 ± 0.2 µg/µL and FTIR proved conjugation of all NA types to AuNP. All the samples had a negative charge of - 36 to - 46 mV. Confocal microscopy confirmed internalization of the ssRNA-AuNP into eukaryotic cells and qPCR confirmed release and activity of carried RNA.
Collapse
Affiliation(s)
- P Pourali
- Institute of Microbiology, Czech Academy of Sciences, Prague, Czech Republic
| | - V Dzmitruk
- Center of Molecular Structure, Institute of Biotechnology, Czech Academy of Sciences, Vesec, Czech Republic
| | - O Benada
- Institute of Microbiology, Czech Academy of Sciences, Prague, Czech Republic
| | - M Svoboda
- Institute of Analytical Chemistry, Czech Academy of Sciences, Brno, Czech Republic
| | - V Benson
- Institute of Microbiology, Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
6
|
Zhou Z, Li X. Research progress in mRNA drug modification and delivery systems. Zhejiang Da Xue Xue Bao Yi Xue Ban 2023; 52:439-450. [PMID: 37643978 PMCID: PMC10495253 DOI: 10.3724/zdxbyxb-2023-0101] [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: 03/02/2023] [Accepted: 08/09/2023] [Indexed: 08/18/2023]
Abstract
Messenger RNA (mRNA) has shown tremendous potential in disease prevention and therapy. The clinical application requires mRNA with enhanced stability and high translation efficiency, ensuring it not to be degraded by nucleases and targeting to specific tissues and cells. mRNA immunogenicity can be reduced by nucleotide modification, and translation efficiency can be enhanced by codon optimization. The 5´ capping structure and 3´ poly A increase mRNA stability, and the addition of 5' and 3' non-translational regions regulate mRNA translation initiation and protein production. Nanoparticle delivery system protects mRNA from degradation by ubiquitous nucleases, enhances mRNA concentration in circulation and assists it cytoplasmic entrance for the purpose of treatment and prevention. Here, we review the recent advances of mRNA technology, discuss the methods and principles to enhance mRNA stability and translation efficiency; summarize the requirements involved in designing mRNA delivery systems with the potential for industrial translation and biomedical application. Furthermore, we provide insights into future directions of mRNA therapeutics to meet the needs for personalized precision medicine.
Collapse
Affiliation(s)
- Zhengjie Zhou
- Department of Medicine, Pritzker School of Molecular Engineering, The University of Chicago, Chicago 60637, USA.
| | - Xin Li
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Center for RNA Medicine, International Institutes of Medicine, Zhejiang University, Jinhua 322000, Zhejiang Province, China
| |
Collapse
|
7
|
Pourali P, Dzmitruk V, Pátek M, Neuhöferová E, Svoboda M, Benson V. Fate of the capping agent of biologically produced gold nanoparticles and adsorption of enzymes onto their surface. Sci Rep 2023; 13:4916. [PMID: 36966192 PMCID: PMC10039949 DOI: 10.1038/s41598-023-31792-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 03/17/2023] [Indexed: 03/27/2023] Open
Abstract
Enzymotherapy based on DNase I or RNase A has often been suggested as an optional strategy for cancer treatment. The efficacy of such procedures is limited e.g. by a short half-time of the enzymes or a low rate of their internalization. The use of nanoparticles, such as gold nanoparticles (AuNPs), helps to overcome these limits. Specifically, biologically produced AuNPs represent an interesting variant here due to naturally occurring capping agents (CA) on their surface. The composition of the CA depends on the producing microorganism. CAs are responsible for the stabilization of the nanoparticles, and promote the direct linking of targeting and therapeutic molecules. This study provided proof of enzyme adsorption onto gold nanoparticles and digestion efficacy of AuNPs-adsorbed enzymes. We employed Fusarium oxysporum extract to produce AuNPs. These nanoparticles were round or polygonal with a size of about 5 nm, negative surface charge of about - 33 mV, and maximum absorption peak at 530 nm. After the adsorption of DNAse I, RNase A, or Proteinase K onto the AuNPs surface, the nanoparticles exhibited shifts in surface charge (values between - 22 and - 13 mV) and maximum absorption peak (values between 513 and 534 nm). The ability of AuNP-enzyme complexes to digest different targets was compared to enzymes alone. We found a remarkable degradation of ssDNA, and dsDNA by AuNP-DNAse I, and a modest degradation of ssRNA by AuNP-RNase A. The presence of particular enzymes on the AuNP surface was proved by liquid chromatography-mass spectrometry (LC-MS). Using SDS-PAGE electrophoresis, we detected a remarkable digestion of collagen type I and fibrinogen by AuNP-proteinase K complexes. We concluded that the biologically produced AuNPs directly bound DNase I, RNase A, and proteinase K while preserving their ability to digest specific targets. Therefore, according to our results, AuNPs can be used as effective enzyme carriers and the AuNP-enzyme conjugates can be effective tools for enzymotherapy.
Collapse
Affiliation(s)
- Parastoo Pourali
- Institute of Microbiology, Czech Academy of Sciences, 142 20, Prague, Czech Republic
| | - Volha Dzmitruk
- Center of Molecular Structure, Institute of Biotechnology, Czech Academy of Sciences, 252 50, Prague, Czech Republic
| | - Miroslav Pátek
- Institute of Microbiology, Czech Academy of Sciences, 142 20, Prague, Czech Republic
| | - Eva Neuhöferová
- Institute of Microbiology, Czech Academy of Sciences, 142 20, Prague, Czech Republic
| | - Milan Svoboda
- Institute of Analytical Chemistry, Czech Academy of Sciences, 602 00, Brno, Czech Republic
| | - Veronika Benson
- Institute of Microbiology, Czech Academy of Sciences, 142 20, Prague, Czech Republic.
| |
Collapse
|
8
|
Patel V, Parekh P, Khimani M, Yusa SI, Bahadur P. Pluronics® based Penta Block Copolymer micelles as a precursor of smart aggregates for various applications: A review. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2022.121140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
9
|
Targeted Nanocarrier Delivery of RNA Therapeutics to Control HIV Infection. Pharmaceutics 2022; 14:pharmaceutics14071352. [PMID: 35890248 PMCID: PMC9324444 DOI: 10.3390/pharmaceutics14071352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 02/04/2023] Open
Abstract
Our understanding of HIV infection has greatly advanced since the discovery of the virus in 1983. Treatment options have improved the quality of life of people living with HIV/AIDS, turning it from a fatal disease into a chronic, manageable infection. Despite all this progress, a cure remains elusive. A major barrier to attaining an HIV cure is the presence of the latent viral reservoir, which is established early in infection and persists for the lifetime of the host, even during prolonged anti-viral therapy. Different cure strategies are currently being explored to eliminate or suppress this reservoir. Several studies have shown that a functional cure may be achieved by preventing infection and also inhibiting reactivation of the virus from the latent reservoir. Here, we briefly describe the main HIV cure strategies, focussing on the use of RNA therapeutics, including small interfering RNA (siRNA) to maintain HIV permanently in a state of super latency, and CRISPR gRNA to excise the latent reservoir. A challenge with progressing RNA therapeutics to the clinic is achieving effective delivery into the host cell. This review covers recent nanotechnological strategies for siRNA delivery using liposomes, N-acetylgalactosamine conjugation, inorganic nanoparticles and polymer-based nanocapsules. We further discuss the opportunities and challenges of those strategies for HIV treatment.
Collapse
|
10
|
Zhang Y, Almazi JG, Ong HX, Johansen MD, Ledger S, Traini D, Hansbro PM, Kelleher AD, Ahlenstiel CL. Nanoparticle Delivery Platforms for RNAi Therapeutics Targeting COVID-19 Disease in the Respiratory Tract. Int J Mol Sci 2022; 23:2408. [PMID: 35269550 PMCID: PMC8909959 DOI: 10.3390/ijms23052408] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/14/2022] [Accepted: 02/18/2022] [Indexed: 02/06/2023] Open
Abstract
Since December 2019, a pandemic of COVID-19 disease, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has rapidly spread across the globe. At present, the Food and Drug Administration (FDA) has issued emergency approval for the use of some antiviral drugs. However, these drugs still have limitations in the specific treatment of COVID-19, and as such, new treatment strategies urgently need to be developed. RNA-interference-based gene therapy provides a tractable target for antiviral treatment. Ensuring cell-specific targeted delivery is important to the success of gene therapy. The use of nanoparticles (NPs) as carriers for the delivery of small interfering RNA (siRNAs) to specific tissues or organs of the human body could play a crucial role in the specific therapy of severe respiratory infections, such as COVID-19. In this review, we describe a variety of novel nanocarriers, such as lipid NPs, star polymer NPs, and glycogen NPs, and summarize the pre-clinical/clinical progress of these nanoparticle platforms in siRNA delivery. We also discuss the application of various NP-capsulated siRNA as therapeutics for SARS-CoV-2 infection, the challenges with targeting these therapeutics to local delivery in the lung, and various inhalation devices used for therapeutic administration. We also discuss currently available animal models that are used for preclinical assessment of RNA-interference-based gene therapy. Advances in this field have the potential for antiviral treatments of COVID-19 disease and could be adapted to treat a range of respiratory diseases.
Collapse
Affiliation(s)
- Yuan Zhang
- Kirby Institute, UNSW, Sydney, NSW 2052, Australia; (Y.Z.); (S.L.); (A.D.K.)
| | - Juhura G. Almazi
- Respiratory Technology, Woolcock Institute of Medical Research, Sydney, NSW 2037, Australia; (J.G.A.); (H.X.O.); (D.T.)
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Ryde, NSW 2109, Australia
| | - Hui Xin Ong
- Respiratory Technology, Woolcock Institute of Medical Research, Sydney, NSW 2037, Australia; (J.G.A.); (H.X.O.); (D.T.)
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Ryde, NSW 2109, Australia
| | - Matt D. Johansen
- Centre for Inflammation, Faculty of Science, Centenary Institute and University of Technology Sydney, Sydney, NSW 2050, Australia; (M.D.J.); (P.M.H.)
| | - Scott Ledger
- Kirby Institute, UNSW, Sydney, NSW 2052, Australia; (Y.Z.); (S.L.); (A.D.K.)
| | - Daniela Traini
- Respiratory Technology, Woolcock Institute of Medical Research, Sydney, NSW 2037, Australia; (J.G.A.); (H.X.O.); (D.T.)
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Ryde, NSW 2109, Australia
| | - Philip M. Hansbro
- Centre for Inflammation, Faculty of Science, Centenary Institute and University of Technology Sydney, Sydney, NSW 2050, Australia; (M.D.J.); (P.M.H.)
| | - Anthony D. Kelleher
- Kirby Institute, UNSW, Sydney, NSW 2052, Australia; (Y.Z.); (S.L.); (A.D.K.)
| | | |
Collapse
|
11
|
Rahman MM, Ahmed M, Islam MT, Khan MR, Sultana S, Maeesa SK, Hasan S, Hossain MA, Ferdous KS, Mathew B, Rauf A, Uddin MS. Nanotechnology-Based Approaches and Investigational Therapeutics against COVID-19. Curr Pharm Des 2021; 28:948-968. [PMID: 34218774 DOI: 10.2174/1381612827666210701150315] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 04/30/2021] [Indexed: 01/08/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a novel coronavirus which is currently responsible for the global pandemic since December 2019. This class of coronavirus has affected 217 countries around the world. Most of the countries have taken some non-remedial preventive actions like country lockdown, work from home, travel bans, and the most significant one is social isolation. Pharmacists, doctors, nurses, technologists, and all other healthcare professionals are playing a pivotal role during this pandemic. Unluckily, there is no specific drug that can treat patients who are confirmed with COVID-19, though favipiravir and remdesivir have appeared as favorable antiviral drugs. Some vaccines have already developed, and vaccination has started worldwide. Different nanotechnologies are in the developing stage in many countries for preventing SARS-COV-2 and treating COVID-19 conditions. In this article, we review the COVID-19 pandemic situation as well as the nanotechnology-based approaches and investigational therapeutics against COVID-19.
Collapse
Affiliation(s)
- Md Mominur Rahman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka. Bangladesh
| | - Muniruddin Ahmed
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka. Bangladesh
| | - Mohammad Touhidul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka. Bangladesh
| | - Md Robin Khan
- Bangladesh Reference Institute for Chemical Measurements, Dhaka. Bangladesh
| | - Sharifa Sultana
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka. Bangladesh
| | - Saila Kabir Maeesa
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka. Bangladesh
| | - Sakib Hasan
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka. Bangladesh
| | - Md Abid Hossain
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka. Bangladesh
| | - Kazi Sayma Ferdous
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka. Bangladesh
| | - Bijo Mathew
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi 682041, India
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Swabi, Anbar, Khyber Pakhtunkhwa. Bangladesh
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka. Bangladesh
| |
Collapse
|
12
|
Wang L, Yan Y. A Review of pH-Responsive Organic-Inorganic Hybrid Nanoparticles for RNAi-Based Therapeutics. Macromol Biosci 2021; 21:e2100183. [PMID: 34160896 DOI: 10.1002/mabi.202100183] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/04/2021] [Indexed: 12/13/2022]
Abstract
RNA interference (RNAi) shows great potential in the treatment of varying cancer and genetic disorders. The lack of safe and effective delivery methods is an ongoing challenge to realize the full potential of RNAi-based therapeutics. pH-responsive hybrid nanoparticle is a promising non-virus platform for small interfering RNA (siRNA) delivery with unique properties including the robust response to the acidic microenvironment and the capability of theranostic and combined therapeutics. The mechanism of RNAi and the delivery barriers for RNAi-based therapeutics are first discussed. Then, the general patterns of pH-response and the typical construction of hybrid nanoparticles are demonstrated. The recent advances in pH-responsive organic-inorganic hybrid nanoparticles for siRNA delivery are highlighted, in particular, how pH-response of ionizable groups, acid-labile bonds, and decomposition of inorganic components affect the physicochemical properties of hybrid nanoparticles and benefit the cellular uptake and intracellular trafficking of siRNA payloads are discussed. At last, the remaining problems and the prospects for pH-responsive hybrid nanoparticles for siRNA delivery are analyzed.
Collapse
Affiliation(s)
- Lu Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Yunfeng Yan
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| |
Collapse
|
13
|
Torres-Vanegas JD, Cruz JC, Reyes LH. Delivery Systems for Nucleic Acids and Proteins: Barriers, Cell Capture Pathways and Nanocarriers. Pharmaceutics 2021; 13:428. [PMID: 33809969 PMCID: PMC8004853 DOI: 10.3390/pharmaceutics13030428] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/09/2021] [Accepted: 03/12/2021] [Indexed: 12/27/2022] Open
Abstract
Gene therapy has been used as a potential approach to address the diagnosis and treatment of genetic diseases and inherited disorders. In this line, non-viral systems have been exploited as promising alternatives for delivering therapeutic transgenes and proteins. In this review, we explored how biological barriers are effectively overcome by non-viral systems, usually nanoparticles, to reach an efficient delivery of cargoes. Furthermore, this review contributes to the understanding of several mechanisms of cellular internalization taken by nanoparticles. Because a critical factor for nanoparticles to do this relies on the ability to escape endosomes, researchers have dedicated much effort to address this issue using different nanocarriers. Here, we present an overview of the diversity of nanovehicles explored to reach an efficient and effective delivery of both nucleic acids and proteins. Finally, we introduced recent advances in the development of successful strategies to deliver cargoes.
Collapse
Affiliation(s)
- Julian D. Torres-Vanegas
- Grupo de Diseño de Productos y Procesos (GDPP), Department of Chemical and Food Engineering, Universidad de los Andes, Bogotá 111711, Colombia
| | - Juan C. Cruz
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia
| | - Luis H. Reyes
- Grupo de Diseño de Productos y Procesos (GDPP), Department of Chemical and Food Engineering, Universidad de los Andes, Bogotá 111711, Colombia
| |
Collapse
|
14
|
Chow MYT, Qiu Y, Lam JKW. Inhaled RNA Therapy: From Promise to Reality. Trends Pharmacol Sci 2020; 41:715-729. [PMID: 32893004 PMCID: PMC7471058 DOI: 10.1016/j.tips.2020.08.002] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 08/02/2020] [Accepted: 08/03/2020] [Indexed: 12/11/2022]
Abstract
RNA-based medicine is receiving growing attention for its diverse roles and potential therapeutic capacity. The largest obstacle in its clinical translation remains identifying a safe and effective delivery system. Studies investigating RNA therapeutics in pulmonary diseases have rapidly expanded and drug administration by inhalation allows the direct delivery of RNA therapeutics to the target site of action while minimizing systemic exposure. In this review, we highlight recent developments in pulmonary RNA delivery systems with the use of nonviral vectors. We also discuss the major knowledge gaps that require thorough investigation and provide insights that will help advance this exciting field towards the bedside.
Collapse
Affiliation(s)
- Michael Y T Chow
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR; Advanced Drug Delivery Group, Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Yingshan Qiu
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR
| | - Jenny K W Lam
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR.
| |
Collapse
|
15
|
Luther DC, Huang R, Jeon T, Zhang X, Lee YW, Nagaraj H, Rotello VM. Delivery of drugs, proteins, and nucleic acids using inorganic nanoparticles. Adv Drug Deliv Rev 2020; 156:188-213. [PMID: 32610061 PMCID: PMC8559718 DOI: 10.1016/j.addr.2020.06.020] [Citation(s) in RCA: 154] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 01/03/2023]
Abstract
Inorganic nanoparticles provide multipurpose platforms for a broad range of delivery applications. Intrinsic nanoscopic properties provide access to unique magnetic and optical properties. Equally importantly, the structural and functional diversity of gold, silica, iron oxide, and lanthanide-based nanocarriers provide unrivalled control of nanostructural properties for effective transport of therapeutic cargos, overcoming biobarriers on the cellular and organismal level. Taken together, inorganic nanoparticles provide a key addition to the arsenal of delivery vectors for fighting disease and improving human health.
Collapse
Affiliation(s)
- David C Luther
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, MA 01003, USA
| | - Rui Huang
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, MA 01003, USA
| | - Taewon Jeon
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, MA 01003, USA; Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, MA 01003, USA
| | - Xianzhi Zhang
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, MA 01003, USA
| | - Yi-Wei Lee
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, MA 01003, USA
| | - Harini Nagaraj
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, MA 01003, USA
| | - Vincent M Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, MA 01003, USA.
| |
Collapse
|
16
|
Itani R, Tobaiqy M, Al Faraj A. Optimizing use of theranostic nanoparticles as a life-saving strategy for treating COVID-19 patients. Theranostics 2020; 10:5932-5942. [PMID: 32483428 PMCID: PMC7254986 DOI: 10.7150/thno.46691] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 04/17/2020] [Indexed: 01/05/2023] Open
Abstract
On the 30th of January 2020, the World Health Organization fired up the sirens against a fast spreading infectious disease caused by a newly discovered Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) and gave this disease the name COVID-19. While there is currently no specific treatment for COVID-19, several off label drugs approved for other indications are being investigated in clinical trials across the globe. In the last decade, theranostic nanoparticles were reported as promising tool for efficiently and selectively deliver therapeutic moieties (i.e. drugs, vaccines, siRNA, peptide) to target sites of infection. In addition, they allow monitoring infectious sides and treatment responses using noninvasive imaging modalities. While intranasal delivery was proposed as the preferred administration route for therapeutic agents against viral pulmonary diseases, NP-based delivery systems offer numerous benefits to overcome challenges associated with mucosal administration, and ensure that these agents achieve a concentration that is many times higher than expected in the targeted sites of infection while limiting side effects on normal cells. In this article, we have shed light on the promising role of nanoparticles as effective carriers for therapeutics or immune modulators to help in fighting against COVID-19.
Collapse
Affiliation(s)
- Rasha Itani
- Department of Radiologic Sciences, Faculty of Health Sciences, American University of Science and Technology (AUST), Beirut, Lebanon
| | - Mansour Tobaiqy
- Department of Pharmacology, College of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| | - Achraf Al Faraj
- Department of Radiologic Sciences, Faculty of Health Sciences, American University of Science and Technology (AUST), Beirut, Lebanon
| |
Collapse
|
17
|
Recent advances of smart acid‐responsive gold nanoparticles in tumor therapy. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 12:e1619. [DOI: 10.1002/wnan.1619] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/17/2020] [Accepted: 01/21/2020] [Indexed: 12/14/2022]
|
18
|
Tieu T, Dhawan S, Haridas V, Butler LM, Thissen H, Cifuentes-Rius A, Voelcker NH. Maximizing RNA Loading for Gene Silencing Using Porous Silicon Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2019; 11:22993-23005. [PMID: 31252458 DOI: 10.1021/acsami.9b05577] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Gene silencing by RNA interference is a powerful technology with broad applications. However, this technology has been hampered by the instability of small interfering RNA (siRNA) molecules in physiological conditions and their inefficient delivery into the cytoplasm of target cells. Porous silicon nanoparticles have emerged as a potential delivery vehicle to overcome these limitations-being able to encapsulate RNA molecules within the porous matrix and protect them from degradation. Here, key variables were investigated that influence siRNA loading into porous silicon nanoparticles. The effect of modifying the surface of porous silicon nanoparticles with various amino-functional molecules as well as the effects of salt and chaotropic agents in facilitating siRNA loading was examined. Maximum siRNA loading of 413 μg/(mg of porous silicon nanoparticles) was found when the nanoparticles were modified by a fourth generation polyamidoamine dendrimer. Low concentrations of urea or salt increased loading capacity: an increase in RNA loading by 19% at a concentration of 0.05 M NaCl or 21% at a concentration of 0.25 M urea was observed when compared to loading in water. Lastly, it was demonstrated that dendrimer-functionalized nanocarriers are able to deliver siRNA against ELOVL5, a target for the treatment of advanced prostate cancer.
Collapse
Affiliation(s)
- Terence Tieu
- Monash Institute of Pharmaceutical Sciences , Monash University , Parkville Campus, 381 Royal Parade , Parkville , Victoria 3052 , Australia
- CSIRO Manufacturing , Bayview Avenue , Clayton , Victoria 3168 , Australia
| | - Sameer Dhawan
- Department of Chemistry , Indian Institute of Technology Delhi , Hauz Khas, New Delhi 110016 , India
| | - V Haridas
- Department of Chemistry , Indian Institute of Technology Delhi , Hauz Khas, New Delhi 110016 , India
| | - Lisa M Butler
- Adelaide Medical School & Freemasons Foundation Centre for Men's Health , University of Adelaide , Adelaide , South Australia 5005 , Australia
- South Australian Health & Medical Research Institute , Adelaide , South Australia 5001 , Australia
| | - Helmut Thissen
- CSIRO Manufacturing , Bayview Avenue , Clayton , Victoria 3168 , Australia
| | - Anna Cifuentes-Rius
- Monash Institute of Pharmaceutical Sciences , Monash University , Parkville Campus, 381 Royal Parade , Parkville , Victoria 3052 , Australia
| | - Nicolas H Voelcker
- Monash Institute of Pharmaceutical Sciences , Monash University , Parkville Campus, 381 Royal Parade , Parkville , Victoria 3052 , Australia
- CSIRO Manufacturing , Bayview Avenue , Clayton , Victoria 3168 , Australia
- Melbourne Centre for Nanofabrication , Victorian Node of the Australian National Fabrication Facility , 151 Wellington Road , Clayton , Victoria 3168 , Australia
| |
Collapse
|
19
|
Han X, Li Y, Xu Y, Zhao X, Zhang Y, Yang X, Wang Y, Zhao R, Anderson GJ, Zhao Y, Nie G. Reversal of pancreatic desmoplasia by re-educating stellate cells with a tumour microenvironment-activated nanosystem. Nat Commun 2018; 9:3390. [PMID: 30139933 PMCID: PMC6107580 DOI: 10.1038/s41467-018-05906-x] [Citation(s) in RCA: 223] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 07/26/2018] [Indexed: 12/11/2022] Open
Abstract
Pancreatic ductal adenocarcinoma is characterised by a dense desmoplastic stroma composed of stromal cells and extracellular matrix (ECM). This barrier severely impairs drug delivery and penetration. Activated pancreatic stellate cells (PSCs) play a key role in establishing this unique pathological obstacle, but also offer a potential target for anti-tumour therapy. Here, we construct a tumour microenvironment-responsive nanosystem, based on PEGylated polyethylenimine-coated gold nanoparticles, and utilise it to co-deliver all-trans retinoic acid (ATRA, an inducer of PSC quiescence) and siRNA targeting heat shock protein 47 (HSP47, a collagen-specific molecular chaperone) to re-educate PSCs. The nanosystem simultaneously induces PSC quiescence and inhibits ECM hyperplasia, thereby promoting drug delivery to pancreatic tumours and significantly enhancing the anti-tumour efficacy of chemotherapeutics. Our combination strategy to restore homoeostatic stromal function by targeting activated PSCs represents a promising approach to improving the efficacy of chemotherapy and other therapeutic modalities in a wide range of stroma-rich tumours.
Collapse
Affiliation(s)
- Xuexiang Han
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
- Department of Chemistry, Tsinghua University, Beijing, 100084, P.R. China
| | - Yiye Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P.R. China.
- University of Chinese Academy of Sciences, Beijing, 100049, P.R. China.
| | - Ying Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Xiao Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Yinlong Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Xiao Yang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Yongwei Wang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, P.R. China
| | - Ruifang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Gregory J Anderson
- QIMR Berghofer Medical Research Institute, Royal Brisbane Hospital, Herston, QLD 4029, Australia
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P.R. China.
- University of Chinese Academy of Sciences, Beijing, 100049, P.R. China.
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P.R. China.
- University of Chinese Academy of Sciences, Beijing, 100049, P.R. China.
| |
Collapse
|
20
|
Shen J, Zhang W, Qi R, Mao ZW, Shen H. Engineering functional inorganic-organic hybrid systems: advances in siRNA therapeutics. Chem Soc Rev 2018; 47:1969-1995. [PMID: 29417968 PMCID: PMC5861001 DOI: 10.1039/c7cs00479f] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cancer treatment still faces a lot of obstacles such as tumor heterogeneity, drug resistance and systemic toxicities. Beyond the traditional treatment modalities, exploitation of RNA interference (RNAi) as an emerging approach has immense potential for the treatment of various gene-caused diseases including cancer. The last decade has witnessed enormous research and achievements focused on RNAi biotechnology. However, delivery of small interference RNA (siRNA) remains a key challenge in the development of clinical RNAi therapeutics. Indeed, functional nanomaterials play an important role in siRNA delivery, which could overcome a wide range of sequential physiological and biological obstacles. Nanomaterial-formulated siRNA systems have potential applications in protection of siRNA from degradation, improving the accumulation in the target tissues, enhancing the siRNA therapy and reducing the side effects. In this review, we explore and summarize the role of functional inorganic-organic hybrid systems involved in the siRNA therapeutic advancements. Additionally, we gather the surface engineering strategies of hybrid systems to optimize for siRNA delivery. Major progress in the field of inorganic-organic hybrid platforms including metallic/non-metallic cores modified with organic shells or further fabrication as the vectors for siRNA delivery is discussed to give credit to the interdisciplinary cooperation between chemistry, pharmacy, biology and medicine.
Collapse
Affiliation(s)
- Jianliang Shen
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou 510275, China. and School of Ophthalmology & Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325035, China and Wenzhou Institute of Biomaterials and Engineering, Chinese Academy of Science, Wenzhou, 325001, China and Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas 77030, USA.
| | - Wei Zhang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou 510275, China.
| | - Ruogu Qi
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas 77030, USA.
| | - Zong-Wan Mao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou 510275, China. and Department of Applied Chemistry, South China Agricultural University, Guangzhou 510642, China
| | - Haifa Shen
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas 77030, USA. and Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY10065, USA
| |
Collapse
|
21
|
Abstract
Nanomaterials have been widely used in the design of drug delivery platforms. This work computationally explores the vascular dynamics of nanoworms as drug carriers within blood flow by considering the effects of nanoworm length, stiffness, and local physiological conditions such as hematocrit. We found that nanoworms with length of 8 μm and moderate stiffness are the optimal choice as drug carriers for circulating within normal vascular network due to their lower near wall margination. Compared to those of spherical rigid particles, these nanoworms demonstrate significant demargination behaviors at hematocrit 20%, induced by the local hydrodynamic interactions. Specifically, the interactions between nanoworms and red blood cells create asymmetrical local flow fields, resulting in the demargination of nanoworms. In addition, the flexibility of nanoworms enables them to conform to the deformed shape of red blood cells under shear flow, leading to their high concentration within the core region of vessels. Therefore, the long blood circulation time of nanoworms can be partially attributed to their demargination behaviors and intertwinement with red blood cells. According to these simulation results, tuning the length and stiffness of nanoworms is the key to design drug carries with reduced near wall margination within normal vascular networks and extend their blood circulation time.
Collapse
Affiliation(s)
- Huilin Ye
- Department of Mechanical Engineering, University of Connecticut, 191 Auditorium Road, Unit 3139, Storrs, Connecticut 06269, United States
| | - Zhiqiang Shen
- Department of Mechanical Engineering, University of Connecticut, 191 Auditorium Road, Unit 3139, Storrs, Connecticut 06269, United States
| | - Le Yu
- Department of Materials Science and Engineering, University of Connecticut, 97 North Eagleville Road, Unit 3136, Storrs, Connecticut 06269, United States
| | - Mei Wei
- Department of Materials Science and Engineering, University of Connecticut, 97 North Eagleville Road, Unit 3136, Storrs, Connecticut 06269, United States.,Institute of Materials Science, University of Connecticut, 97 North Eagleville Road, Unit 3136, Storrs, Connecticut 06269, United States
| | - Ying Li
- Department of Mechanical Engineering, University of Connecticut, 191 Auditorium Road, Unit 3139, Storrs, Connecticut 06269, United States.,Institute of Materials Science, University of Connecticut, 97 North Eagleville Road, Unit 3136, Storrs, Connecticut 06269, United States
| |
Collapse
|
22
|
Yue J, Feliciano TJ, Li W, Lee A, Odom TW. Gold Nanoparticle Size and Shape Effects on Cellular Uptake and Intracellular Distribution of siRNA Nanoconstructs. Bioconjug Chem 2017; 28:1791-1800. [PMID: 28574255 PMCID: PMC5737752 DOI: 10.1021/acs.bioconjchem.7b00252] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Gold nanoparticles (AuNPs) show potential for transfecting target cells with small interfering RNA (siRNA), but the influence of key design parameters such as the size and shape of the particle core is incomplete. This paper describes a side-by-side comparison of the in vitro response of U87 glioblastoma cells to different formulations of siRNA-conjugated gold nanoconstructs targeting the expression of isocitrate dehydrogenase 1 (IDH1) based on 13 nm spheres, 50 nm spheres, and 40 nm stars. 50 nm spheres and 40 nm stars showed much higher uptake efficiency compared to 13 nm spheres. Confocal fluorescence microscopy showed that all three formulations were localized in the endosomes at early incubation times (2 h), but after 24 h, 50 nm spheres and 40 nm stars were neither in endosomes nor in lysosomes while 13 nm spheres remained in endosomes. Transmission electron microscopy images revealed that the 13 nm spheres were enclosed and dispersed within endocytic vesicles while 50 nm spheres and 40 nm stars were aggregated, and some of these NPs were outside of endocytic vesicles. In our comparison of nanoconstructs with different sizes and shapes, while holding siRNA surface density and nanoparticle concentration constant, we found that larger particles (50 nm spheres and 40 nm stars) showed higher potential as carriers for the delivery of siRNA.
Collapse
Affiliation(s)
- Jun Yue
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Timothy Joel Feliciano
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Wenlong Li
- School of Materials Science and Engineering, Nanyang Technological University, 639798, Singapore
| | - Andrew Lee
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Teri W. Odom
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
- Department of Materials Science and Engineering, Northwestern University, Evanston, Illinois 60208, United States
| |
Collapse
|
23
|
Tang Y, Zeng Z, He X, Wang T, Ning X, Feng X. SiRNA Crosslinked Nanoparticles for the Treatment of Inflammation-induced Liver Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2017; 4:1600228. [PMID: 28251047 PMCID: PMC5323819 DOI: 10.1002/advs.201600228] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 07/29/2016] [Indexed: 05/20/2023]
Abstract
RNA interference mediated by small interfering RNA (siRNA) provides a powerful tool for gene regulation, and has a broad potential as a promising therapeutic strategy. However, therapeutics based on siRNA have had limited clinical success due to their undesirable pharmacokinetic properties. This study presents pH-sensitive nanoparticles-based siRNA delivery systems (PNSDS), which are positive-charge-free nanocarriers, composed of siRNA chemically crosslinked with multi-armed poly(ethylene glycol) carriers via acid-labile acetal linkers. The unique siRNA crosslinked structure of PNSDS allows it to have minimal cytotoxicity, high siRNA loading efficiency, and a stimulus-responsive property that enables the selective intracellular release of siRNA in response to pH conditions. This study demonstrates that PNSDS can deliver tumor necrosis factor alpha (TNF-α) siRNA into macrophages and induce the efficient down regulation of the targeted gene in complete cell culture media. Moreover, PNSDS with mannose targeting moieties can selectively accumulate in mice liver, induce specific inhibition of macrophage TNF-α expression in vivo, and consequently protect mice from inflammation-induced liver damages. Therefore, this novel siRNA delivering platform would greatly improve the therapeutic potential of RNAi based therapies.
Collapse
Affiliation(s)
- Yaqin Tang
- Innovative Drug Research CenterChongqing UniversityChongqing401331P. R. China
| | - Ziying Zeng
- Innovative Drug Research CenterChongqing UniversityChongqing401331P. R. China
| | - Xiao He
- Innovative Drug Research CenterChongqing UniversityChongqing401331P. R. China
| | - Tingting Wang
- Innovative Drug Research CenterChongqing UniversityChongqing401331P. R. China
| | - Xinghai Ning
- Department of Biomedical EngineeringNanjing UniversityNanjing210093P. R. China
| | - Xuli Feng
- Innovative Drug Research CenterChongqing UniversityChongqing401331P. R. China
| |
Collapse
|
24
|
Yi Y, Kim HJ, Mi P, Zheng M, Takemoto H, Toh K, Kim BS, Hayashi K, Naito M, Matsumoto Y, Miyata K, Kataoka K. Targeted systemic delivery of siRNA to cervical cancer model using cyclic RGD-installed unimer polyion complex-assembled gold nanoparticles. J Control Release 2016; 244:247-256. [DOI: 10.1016/j.jconrel.2016.08.041] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 08/22/2016] [Accepted: 08/28/2016] [Indexed: 11/29/2022]
|
25
|
Uz M, Bulmus V, Alsoy Altinkaya S. Effect of PEG Grafting Density and Hydrodynamic Volume on Gold Nanoparticle-Cell Interactions: An Investigation on Cell Cycle, Apoptosis, and DNA Damage. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2016; 32:5997-6009. [PMID: 27206138 DOI: 10.1021/acs.langmuir.6b01289] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
In this study, interactions of polyethylene glycol (PEG)-coated gold nanoparticles (AuNPs) with cells were investigated with particular focus on the relationship between the PEG layer properties (conformation, grafting density, and hydrodynamic volume) and cell cycle arrest, apoptosis, and DNA damage. Steric hindrance and PEG hydrodynamic volume controlled the protein adsorption, whereas the AuNP core size and PEG hydrodynamic volume were primary factors for cell uptake and viability. At all PEG grafting densities, the particles caused significant cell cycle arrest and DNA damage against CaCo2 and PC3 cells without apoptosis. However, at a particular PEG grafting density (∼0.65 chains/nm(2)), none of these severe damages were observed on 3T3 cells indicating discriminating behavior of the healthy (3T3) and cancer (PC3 and CaCo2) cells. It was concluded that the PEG grafting density and hydrodynamic volume, tuned with the PEG concentration and AuNP size, played an important role in particle-cell interactions.
Collapse
Affiliation(s)
- Metin Uz
- Department of Chemical Engineering, ‡Department of Bioengineering, and §Biotechnology and Bioengineering Graduate Program, Izmir Institute of Technology , Gulbahce Koyu, Urla, Izmir 35430, Turkey
| | - Volga Bulmus
- Department of Chemical Engineering, ‡Department of Bioengineering, and §Biotechnology and Bioengineering Graduate Program, Izmir Institute of Technology , Gulbahce Koyu, Urla, Izmir 35430, Turkey
| | - Sacide Alsoy Altinkaya
- Department of Chemical Engineering, ‡Department of Bioengineering, and §Biotechnology and Bioengineering Graduate Program, Izmir Institute of Technology , Gulbahce Koyu, Urla, Izmir 35430, Turkey
| |
Collapse
|
26
|
Esteban-Fernández de Ávila B, Angell C, Soto F, Lopez-Ramirez MA, Báez DF, Xie S, Wang J, Chen Y. Acoustically Propelled Nanomotors for Intracellular siRNA Delivery. ACS NANO 2016; 10:4997-5005. [PMID: 27022755 DOI: 10.1021/acsnano.6b01415] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
An effective intracellular gene silencing strategy based on acoustically propelled nanowires modified with an interfering RNA's (siRNA) payload is described. The gold nanowires (AuNW) are wrapped with a Rolling Circle Amplification (RCA) DNA strand, which serves to anchor the siRNA therapy. The ultrasound (US)-powered propulsion of the AuNW leads to fast internalization and rapid intracellular movement and hence to an accelerated siRNA delivery and silencing response. To optimize the micromotor gene silencing procedure, the influence of motion, time, and siRNA dosage was investigated, leading up to a 94% silencing after few minutes treatment with US-propelled siRNA-AuNWs, and to a dramatic (∼13-fold) improvement in the silencing response compared to the static modified nanowires. The ability of the nanomotor-based method for gene silencing has been demonstrated by measuring the GFP silencing response in two different cell lines (HEK-293 and MCF-7) and using detailed control experiments. The viability of the cells after the nanomotors treatment was examined using the MCF-7 cancer cell line. The use of DNA structures carried by the US-propelled nanomotors for gene silencing represents an efficient tool that addresses the challenges associated with RNA transportation and intracellular delivery. Future implementation of nanomachines in gene therapy applications can be expanded into a co-delivery platform for therapeutics.
Collapse
Affiliation(s)
| | - Chava Angell
- Department of Nanoengineering, University of California, San Diego , La Jolla, California 92093, United States
| | - Fernando Soto
- Department of Nanoengineering, University of California, San Diego , La Jolla, California 92093, United States
| | - Miguel Angel Lopez-Ramirez
- Department of Nanoengineering, University of California, San Diego , La Jolla, California 92093, United States
| | - Daniela F Báez
- Department of Nanoengineering, University of California, San Diego , La Jolla, California 92093, United States
| | - Sibai Xie
- Department of Nanoengineering, University of California, San Diego , La Jolla, California 92093, United States
| | - Joseph Wang
- Department of Nanoengineering, University of California, San Diego , La Jolla, California 92093, United States
| | - Yi Chen
- Department of Nanoengineering, University of California, San Diego , La Jolla, California 92093, United States
| |
Collapse
|
27
|
Rizk N, Christoforou N, Lee S. Optimization of anti-cancer drugs and a targeting molecule on multifunctional gold nanoparticles. NANOTECHNOLOGY 2016; 27:185704. [PMID: 27004512 DOI: 10.1088/0957-4484/27/18/185704] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Breast cancer is the most common and deadly cancer among women worldwide. Currently, nanotechnology-based drug delivery systems are useful for cancer treatment; however, strategic planning is critical in order to enhance the anti-cancer properties and reduce the side effects of cancer therapy. Here, we designed multifunctional gold nanoparticles (AuNPs) conjugated with two anti-cancer drugs, TGF-β1 antibody and methotrexate, and a cancer-targeting molecule, folic acid. First, optimum size and shape of AuNPs was selected by the highest uptake of AuNPs by MDA-MB-231, a metastatic human breast cancer cell line. It was 100 nm spherical AuNPs (S-AuNPs) that were used for further studies. A fixed amount (900 μl) of S-AuNP (3.8 × 10(8) particles/ml) was conjugated with folic acid-BSA or methotrexate-BSA. Methotrexate on S-AuNP induced cellular toxicity and the optimum amount of methotrexate-BSA (2.83 mM) was 500 μl. Uptake of S-AuNPs was enhanced by folate conjugation that binds to folate receptors overexpressed by MDA-MB-231 and the optimum uptake was at 500 μl of folic acid-BSA (2.83 mM). TGF-β1 antibody on S-AuNP reduced extracellular TGF-β1 of cancer cells by 30%. Due to their efficacy and tunable properties, we anticipate numerous clinical applications of multifunctional gold nanospheres in treating breast cancer.
Collapse
Affiliation(s)
- Nahla Rizk
- Department of Biomedical Engineering, Khalifa University of Science, Technology, and Research, PO Box 127788, Abu Dhabi, United Arab Emirates
| | | | | |
Collapse
|
28
|
Mehrotra N, Tripathi RM. Short interfering RNA therapeutics: nanocarriers, prospects and limitations. IET Nanobiotechnol 2016; 9:386-95. [PMID: 26647816 DOI: 10.1049/iet-nbt.2015.0018] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Since the first experiment depicting gene inhibition using RNA interference mechanism, extensive research has been carried out to design targeted delivery systems that use short interfering RNAs (siRNAs) for gene expression regulation. Although several siRNAs loaded nanoparticle systems have reached clinical trial stage, cellular uptake, reticuloendothelial entrapment and endosomal escape still limit the efficacy of these drugs considerably. This review discusses about the RNA interference mechanism, nanostructures being used as non-viral vectors for targeted delivery, limitations of the common delivery systems and the current siRNA-loaded nanoparticle formulations undergoing clinical testing.
Collapse
Affiliation(s)
- Neha Mehrotra
- Amity Institute of Nanotechnology, Amity University, Sector 125, Noida 201303, India
| | - Ravi Mani Tripathi
- Amity Institute of Nanotechnology, Amity University, Sector 125, Noida 201303, India.
| |
Collapse
|
29
|
Gasparini G, Bang EK, Montenegro J, Matile S. Cellular uptake: lessons from supramolecular organic chemistry. Chem Commun (Camb) 2016; 51:10389-402. [PMID: 26030211 DOI: 10.1039/c5cc03472h] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The objective of this Feature Article is to reflect on the importance of established and emerging principles of supramolecular organic chemistry to address one of the most persistent problems in life sciences. The main topic is dynamic covalent chemistry on cell surfaces, particularly disulfide exchange for thiol-mediated uptake. Examples of boronate and hydrazone exchange are added for contrast, comparison and completion. Of equal importance are the discussions of proximity effects in polyions and counterion hopping, and more recent highlights on ring tension and ion pair-π interactions. These lessons from supramolecular organic chemistry apply to cell-penetrating peptides, particularly the origin of "arginine magic" and the "pyrenebutyrate trick," and the currently emerging complementary "disulfide magic" with cell-penetrating poly(disulfide)s. They further extend to the voltage gating of neuronal potassium channels, gene transfection, and the delivery of siRNA. The collected examples illustrate that the input from conceptually innovative chemistry is essential to address the true challenges in biology beyond incremental progress and random screening.
Collapse
Affiliation(s)
- Giulio Gasparini
- School of Chemistry and Biochemistry, University of Geneva, Geneva, Switzerland.
| | | | | | | |
Collapse
|
30
|
DNA Nanotechnology Mediated Gold Nanoparticle Conjugates and Their Applications in Biomedicine. CHINESE J CHEM 2016. [DOI: 10.1002/cjoc.201500839] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
31
|
Misra SK, Ray T, Ostadhossein F, Kim B, Ray PS, Pan D. Carotenoid Nanovector for Efficient Therapeutic Gene Knockdown of Transcription Factor FOXC1 in Liver Cancer. Bioconjug Chem 2016; 27:594-603. [PMID: 26720420 DOI: 10.1021/acs.bioconjchem.5b00601] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Transcription factor FOXC1 has been implicated to play a critical role in hepatocellular carcinoma (HCC) progression, but targeting FOXC1 for therapeutic benefit remains a challenge owing to its location inside the cell nucleus. Herein we report successful therapeutic gene knockdown of transcription factor FOXC1 in liver cancer cells through efficient delivery of siFOXC1 using novel carotenoid functionalized dendritic nanoparticles (CDN). This delivery system also displayed a markedly reduced toxicity profile compared to a standard siRNA transfection agent. We were able to achieve ∼90% FOXC1 knockdown using the CDN-siFOXC1 complex. Additionally, it was found to have ∼18% greater delivery efficiency compared to treatments with particles which have no carotenoid tagging, thereby emphasizing the role of carotenoid mediated cell internalization in the efficient delivery of CDN-siFOXC1 complex in liver cancer cells.
Collapse
Affiliation(s)
- Santosh K Misra
- Department of Bioengineering and Beckman Institute, Materials Science and Engineering, University of Illinois at Urbana-Champaign and Carle Cancer Center , 502 North Busey, Urbana, Illinois 61801, United States
| | - Tania Ray
- Department of Surgery, University of Illinois College of Medicine, Division of Surgical Oncology, Carle Cancer Center , 509 West University Avenue, Urbana, Illinois 61801, United States
| | - Fatemeh Ostadhossein
- Department of Bioengineering and Beckman Institute, Materials Science and Engineering, University of Illinois at Urbana-Champaign and Carle Cancer Center , 502 North Busey, Urbana, Illinois 61801, United States
| | - Bomy Kim
- Department of Surgery, University of Illinois College of Medicine, Division of Surgical Oncology, Carle Cancer Center , 509 West University Avenue, Urbana, Illinois 61801, United States
| | - Partha S Ray
- Department of Surgery, University of Illinois College of Medicine, Division of Surgical Oncology, Carle Cancer Center , 509 West University Avenue, Urbana, Illinois 61801, United States
| | - Dipanjan Pan
- Department of Bioengineering and Beckman Institute, Materials Science and Engineering, University of Illinois at Urbana-Champaign and Carle Cancer Center , 502 North Busey, Urbana, Illinois 61801, United States
| |
Collapse
|
32
|
Oligonucleotide therapeutics: chemistry, delivery and clinical progress. Future Med Chem 2015; 7:2221-42. [PMID: 26510815 DOI: 10.4155/fmc.15.144] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Oligonucleotide therapeutics have the potential to become a third pillar of drug development after small molecules and protein therapeutics. However, the three approved oligonucleotide drugs over the past 17 years have not proven to be highly successful in a commercial sense. These trailblazer drugs have nonetheless laid the foundations for entire classes of drug candidates to follow. This review will examine further advances in chemistry that are earlier in the pipeline of oligonucleotide drug candidates. Finally, we consider the possible effect of delivery systems that may provide extra footholds to improve the potency and specificity of oligonucleotide drugs. Our overview focuses on strategies to imbue antisense oligonucleotides with more drug-like properties and their applicability to other nucleic acid therapeutics.
Collapse
|
33
|
Loczenski Rose V, Shubber S, Sajeesh S, Spain SG, Puri S, Allen S, Lee DK, Winkler GS, Mantovani G. Phosphonium Polymethacrylates for Short Interfering RNA Delivery: Effect of Polymer and RNA Structural Parameters on Polyplex Assembly and Gene Knockdown. Biomacromolecules 2015; 16:3480-90. [DOI: 10.1021/acs.biomac.5b00898] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Vanessa Loczenski Rose
- School
of Pharmacy, University of Nottingham, Boots Science Building, University
Park, Nottingham NG7 2RD, U.K
| | - Saif Shubber
- School
of Pharmacy, University of Nottingham, Boots Science Building, University
Park, Nottingham NG7 2RD, U.K
| | - S. Sajeesh
- Global
Research Laboratory for RNAi Medicine, Department of Chemistry, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Sebastian G. Spain
- Department
of Chemistry, University of Sheffield, Dainton Building, Sheffield S3 7HF, U.K
| | - Sanyogitta Puri
- Pharmaceutical
Development, Astrazeneca UK Ltd., Alderley Park, Macclesfield SK10 2NA, U.K
| | - Stephanie Allen
- School
of Pharmacy, University of Nottingham, Boots Science Building, University
Park, Nottingham NG7 2RD, U.K
| | - Dong-Ki Lee
- Global
Research Laboratory for RNAi Medicine, Department of Chemistry, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - G. Sebastiaan Winkler
- School
of Pharmacy, University of Nottingham, Boots Science Building, University
Park, Nottingham NG7 2RD, U.K
| | - Giuseppe Mantovani
- School
of Pharmacy, University of Nottingham, Boots Science Building, University
Park, Nottingham NG7 2RD, U.K
| |
Collapse
|
34
|
Child HW, Hernandez Y, Conde J, Mullin M, Baptista P, de la Fuente JM, Berry CC. Gold nanoparticle-siRNA mediated oncogene knockdown at RNA and protein level, with associated gene effects. Nanomedicine (Lond) 2015; 10:2513-25. [DOI: 10.2217/nnm.15.95] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Aims: RNAi is a powerful tool for gene silencing that can be used to reduce undesirable overexpression of oncogenes as a novel form of cancer treatment. However, when using RNAi as a therapeutic tool there is potential for associated gene effects. This study aimed to utilize gold nanoparticles to deliver siRNA into HeLa cells. Results: Knockdown of the c-myc oncogene by RNAi, at the RNA, protein and cell proliferation level was achieved, while also identifying associated gene responses. Discussion: The gold nanoparticles used in this study present an excellent delivery platform for siRNA, but do note associated gene changes. Conclusion: The study highlights the need to more widely assess the cell physiological response to RNAi treatment, rather than focus on the immediate RNA levels.
Collapse
Affiliation(s)
| | - Yulán Hernandez
- Instituto de Nanociencia de Aragón, University of Zaragoza, C/Mariano Esquillor s/n Zaragoza, Spain
| | - João Conde
- Massachusetts Institute of Technology, Institute for Medical Engineering & Science, Harvard-MIT Division for Health Sciences & Technology, Cambridge, MA 02139-4307, USA
| | - Margaret Mullin
- Integrated Microscopy Facility, Joseph Black Building, Glasgow University, G12 8QQ, UK
| | - Pedro Baptista
- UCIBIO, CIGMH, Departamento de Ciencias da Vida, Faculdade de Ciencias e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, Portugal
| | - Jesus Maria de la Fuente
- Instituto de Nanociencia de Aragón, University of Zaragoza, C/Mariano Esquillor s/n Zaragoza, Spain
- Instituto de Ciencia de Materiales de Aragón (ICMA), CSIC-Universidad de Zaragoza, c/Pedro Cerbuna 12, 50009 Zaragoza, Spain
| | | |
Collapse
|
35
|
Lee MS, Kim NW, Lee JE, Lim DW, Suh W, Kim HT, Park JW, Jeong JH. Micelle-templated dendritic gold nanoparticles for enhanced cellular delivery of siRNA. Macromol Res 2015. [DOI: 10.1007/s13233-015-3091-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
36
|
Friberg S, Nyström AM. Nanotechnology in the war against cancer: new arms against an old enemy – a clinical view. Future Oncol 2015; 11:1961-75. [DOI: 10.2217/fon.15.91] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
ABSTRACT Clinical oncology is facing a paradigm shift. A new treatment philosophy is emerging and new targets are appearing that require new active agents. The medical use of nanotechnology – nanomedicine – holds several promising possibilities in the war against cancer. Some of these include: new formats for old drugs, that is, increasing efficacy while diminishing side effects; and new administration routes – that is, dermal, oral and pulmonary. In this overview, we describe some nanoparticles and their medical uses as well as highlight advantages of nanoparticles compared with conventional pharmaceuticals. We also point to some of the many technical challenges and potential risks with using nanotechnology for oncological applications.
Collapse
Affiliation(s)
- Sten Friberg
- Swedish Medical Nanoscience Center, Department of Neuroscience, Retzius väg 8, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Andreas M Nyström
- Institute of Environmental Medicine, Nobels väg 13, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| |
Collapse
|
37
|
Jagani H, Kasinathan N, Meka SR, Josyula VR. Antiapoptotic Bcl-2 protein as a potential target for cancer therapy: A mini review. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2015; 44:1212-21. [DOI: 10.3109/21691401.2015.1019668] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Hitesh Jagani
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal University, Manipal, Karnataka, India
| | - Narayanan Kasinathan
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal University, Manipal, Karnataka, India
| | - Sreenivasa Reddy Meka
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal University, Manipal, Karnataka, India
| | - Venkata Rao Josyula
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal University, Manipal, Karnataka, India
| |
Collapse
|
38
|
Jebali A, Anvari-Tafti MH. Hybridization of different antisense oligonucleotides on the surface of gold nanoparticles to silence zinc metalloproteinase gene after uptake by Leishmania major. Colloids Surf B Biointerfaces 2015; 129:107-13. [PMID: 25835145 DOI: 10.1016/j.colsurfb.2015.03.029] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 03/06/2015] [Accepted: 03/10/2015] [Indexed: 01/22/2023]
Abstract
The use of antisense oligonucleotides is a novel strategy to treat infectious diseases. In this approach, vital mRNAs are targeted by antisense oligonucleotides. The aim of this study was to evaluate the effects of gold nanoparticles hybridized with different antisense oligonucleotides on Leishmania (L) major. In this project, gold nanoparticles were first synthesized, and then conjugated with primary oligonucleotides, 3'-AAA-5'. Next, conjugated gold nanoparticles (NP1) were separately hybridized with three types of antisense oligonucleotide from coding reign of GP63 gene (NP2), non-coding reign of GP63 gene (NP3), and both coding and non-coding reigns of GP63 (NP4). Then, 1mL of L. major suspension was separately added to 1mL of different hybridized gold nanoparticles at serial concentrations (1-200μg/mL), and incubated for 24, 48, and 72h at 37°C. Next, the uptake of each nanoparticle was separately measured by atomic absorption spectroscopy. After incubation, the cell viability was separately evaluated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide assay. Also, the expression of GP63 gene was read out by quantitative-real-time PCR. This study showed that NP2 and NP3 had higher (5-fold) uptake than NP1 and NP4. Moreover, NP2 and NP3 led to less cell viability and gene expression, compared with NP1 and NP4. It could be concluded that both sequence and size of antisense oligonucleotide were important for transfection of L. major. Importantly, these antisense oligonucleotides can be obtained from both coding and non-coding reign of GP63 gene. Moreover, hybridized gold nanoparticles not only could silence GP63 gene, but also could kill L. major.
Collapse
Affiliation(s)
- Ali Jebali
- Department of Laboratory Sciences, School of Paramedicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | |
Collapse
|
39
|
Calabrese CM, Merkel TJ, Briley WE, Randeria PS, Narayan SP, Rouge JL, Walker DA, Scott AW, Mirkin CA. Biocompatible infinite-coordination-polymer nanoparticle-nucleic-acid conjugates for antisense gene regulation. Angew Chem Int Ed Engl 2015; 54:476-480. [PMID: 25393766 PMCID: PMC4314394 DOI: 10.1002/anie.201407946] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 10/20/2014] [Indexed: 01/15/2023]
Abstract
Herein, we report the synthesis of DNA-functionalized infinite-coordination-polymer (ICP) nanoparticles as biocompatible gene-regulation agents. ICP nanoparticles were synthesized from ferric nitrate and a ditopic 3-hydroxy-4-pyridinone (HOPO) ligand bearing a pendant azide. Addition of Fe(III) to a solution of the ligand produced nanoparticles, which were colloidally unstable in the presence of salts. Conjugation of DNA to the Fe(III)-HOPO ICP particles by copper-free click chemistry afforded colloidally stable nucleic-acid nanoconstructs. The DNA-ICP particles, when cross-linked through sequence-specific hybridization, exhibited narrow, highly cooperative melting transitions consistent with dense DNA surface loading. The ability of the DNA-ICP particles to enter cells and alter protein expression was also evaluated. Our results indicate that these novel particles carry nucleic acids into mammalian cells without the need for transfection agents and are capable of efficient gene knockdown.
Collapse
Affiliation(s)
| | - Timothy J. Merkel
- International Institute for Nanotechnology Northwestern University, Evanston, IL (USA)
| | - William E. Briley
- Interdepartmental Biological Sciences Northwestern University, Evanston, IL (USA)
| | - Pratik S. Randeria
- Department of Biomedical Engineering Northwestern University, Evanston, IL (USA)
| | - Suguna P. Narayan
- Department of Biomedical Engineering Northwestern University, Evanston, IL (USA)
| | - Jessica L. Rouge
- International Institute for Nanotechnology Northwestern University, Evanston, IL (USA)
| | - David A. Walker
- International Institute for Nanotechnology Northwestern University, Evanston, IL (USA)
- Department of Chemical and Biological Engineering Northwestern University, Evanston, IL (USA)
| | - Alexander W. Scott
- Department of Biomedical Engineering Northwestern University, Evanston, IL (USA)
| | - Chad A. Mirkin
- Department of Chemistry Northwestern University, Evanston, IL (USA)
- International Institute for Nanotechnology Northwestern University, Evanston, IL (USA)
- Department of Chemical and Biological Engineering Northwestern University, Evanston, IL (USA)
| |
Collapse
|
40
|
Abstract
Temperature and pH responsive pentablock copolymers were used for the first time in developing polyplex and multicomponent siRNA delivery systems.
Collapse
Affiliation(s)
- Metin Uz
- Izmir Institute of Technology
- Department of Chemical Engineering
- Izmir
- 35430-Turkey
- Iowa State University
| | | | | |
Collapse
|
41
|
Niikura K, Kobayashi K, Takeuchi C, Fujitani N, Takahara S, Ninomiya T, Hagiwara K, Mitomo H, Ito Y, Osada Y, Ijiro K. Amphiphilic gold nanoparticles displaying flexible bifurcated ligands as a carrier for siRNA delivery into the cell cytosol. ACS APPLIED MATERIALS & INTERFACES 2014; 6:22146-54. [PMID: 25466488 DOI: 10.1021/am505577j] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
The nanoparticle-based delivery of siRNA with a noncationic outermost surface at a low particle concentration is greatly desired. We newly synthesized a bifurcated ligand (BL) possessing hydrophobic and hydrophilic arms as a surface ligand for gold nanoparticles (AuNPs) to allow siRNA delivery. The concept underlying the design of this ligand is that amphiphilic property should allow AuNPs to permeate the cell cytosol thorough the endosomal membrane. BLs and quaternary cationic ligands were codisplayed on 40 nm AuNPs, which were subsequently coated with siRNA via electrostatic interaction. The number of siRNAs immobilized on a single nanoparticle was 26, and the conjugate showed a negative zeta potential due to siRNAs on the outermost surface of the AuNPs. Apparent gene silencing of luciferase expression in HeLa cells was achieved at an AuNP concentration as low as 60 pM. Almost no gene silencing was observed for AuNPs not displaying BLs. To reveal the effect of the BL, we compared the number of AuNPs internalized into HeLa cells and the localization in the cytosol between AuNPs displaying and those not displaying BLs. These analyses indicated that the role of BLs is not only the simple promotion of cellular uptake but also involves the enhancement of AuNPs permeation into the cytosol from the endosomes, leading to effective gene silencing.
Collapse
Affiliation(s)
- Kenichi Niikura
- Research Institute for Electronic Science (RIES), Hokkaido University , Kita 21, Nishi 10, Kita-ku, Sapporo, Hokkaido 001-0021, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
|
43
|
Calabrese CM, Merkel TJ, Briley WE, Randeria PS, Narayan SP, Rouge JL, Walker DA, Scott AW, Mirkin CA. Biocompatible Infinite-Coordination-Polymer Nanoparticle-Nucleic-Acid Conjugates for Antisense Gene Regulation. Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201407946] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
44
|
Draz MS, Fang BA, Zhang P, Hu Z, Gu S, Weng KC, Gray JW, Chen FF. Nanoparticle-mediated systemic delivery of siRNA for treatment of cancers and viral infections. Am J Cancer Res 2014; 4:872-92. [PMID: 25057313 PMCID: PMC4107289 DOI: 10.7150/thno.9404] [Citation(s) in RCA: 150] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 05/27/2014] [Indexed: 12/17/2022] Open
Abstract
RNA interference (RNAi) is an endogenous post-transcriptional gene regulatory mechanism, where non-coding, double-stranded RNA molecules interfere with the expression of certain genes in order to silence it. Since its discovery, this phenomenon has evolved as powerful technology to diagnose and treat diseases at cellular and molecular levels. With a lot of attention, short interfering RNA (siRNA) therapeutics has brought a great hope for treatment of various undruggable diseases, including genetic diseases, cancer, and resistant viral infections. However, the challenge of their systemic delivery and on how they are integrated to exhibit the desired properties and functions remains a key bottleneck for realizing its full potential. Nanoparticles are currently well known to exhibit a number of unique properties that could be strategically tailored into new advanced siRNA delivery systems. This review summarizes the various nanoparticulate systems developed so far in the literature for systemic delivery of siRNA, which include silica and silicon-based nanoparticles, metal and metal oxides nanoparticles, carbon nanotubes, graphene, dendrimers, polymers, cyclodextrins, lipids, hydrogels, and semiconductor nanocrystals. Challenges and barriers to the delivery of siRNA and the role of different nanoparticles to surmount these challenges are also included in the review.
Collapse
|
45
|
Wen Y, Meng WS. Recent In Vivo Evidences of Particle-Based Delivery of Small-Interfering RNA (siRNA) into Solid Tumors. J Pharm Innov 2014; 9:158-173. [PMID: 25221632 PMCID: PMC4161233 DOI: 10.1007/s12247-014-9183-4] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Small-interfering RNA (siRNA) is both a powerful tool in research and a promising therapeutic platform to modulate expression of disease-related genes. Malignant tumors are attractive disease targets for nucleic acid-based therapies. siRNA directed against oncogenes, and genes driving metastases or angiogenesis have been evaluated in animal models and in some cases, in humans. The outcomes of these studies indicate that drug delivery is a significant limiting factor. This review provides perspectives on in vivo validated nanoparticle-based siRNA delivery systems. Results of recent advances in liposomes and polymeric and inorganic formulations illustrate the need for mutually optimized attributes for performance in systemic circulation, tumor interstitial space, plasma membrane, and endosomes. Physiochemical properties conducive to efficient siRNA delivery are summarized and directions for future research are discussed.
Collapse
Affiliation(s)
- Yi Wen
- Division of Pharmaceutical Sciences, Duquesne University, 600, Forbes Avenue, Pittsburgh, PA 15282, USA
| | - Wilson S. Meng
- Division of Pharmaceutical Sciences, Duquesne University, 600, Forbes Avenue, Pittsburgh, PA 15282, USA
| |
Collapse
|
46
|
Gold nanoparticles for nucleic acid delivery. Mol Ther 2014; 22:1075-1083. [PMID: 24599278 DOI: 10.1038/mt.2014.30] [Citation(s) in RCA: 331] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2013] [Accepted: 02/21/2014] [Indexed: 12/11/2022] Open
Abstract
Gold nanoparticles provide an attractive and applicable scaffold for delivery of nucleic acids. In this review, we focus on the use of covalent and noncovalent gold nanoparticle conjugates for applications in gene delivery and RNA-interference technologies. We also discuss challenges in nucleic acid delivery, including endosomal entrapment/escape and active delivery/presentation of nucleic acids in the cell.
Collapse
|
47
|
Lee MY, Kong WH, Jung HS, Hahn SK. Hyaluronic acid–siRNA conjugates complexed with cationic solid lipid nanoparticles for target specific gene silencing. RSC Adv 2014. [DOI: 10.1039/c4ra01485e] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The reducible hyaluronic acid (HA)–siRNA conjugates complexed with cationic solid lipid nanoparticles (CSLNs) with low cytotoxicity and high transfection efficiency was successfully developed for effective liver-specific gene silencing by the dual targeting effect of HA and CSLN.
Collapse
Affiliation(s)
- Min-Young Lee
- Department of Materials Science and Engineering
- Pohang University of Science and Technology (POSTECH)
- Pohang, Korea
| | - Won Ho Kong
- Department of Materials Science and Engineering
- Pohang University of Science and Technology (POSTECH)
- Pohang, Korea
| | - Ho Sang Jung
- Department of Materials Science and Engineering
- Pohang University of Science and Technology (POSTECH)
- Pohang, Korea
| | - Sei Kwang Hahn
- Department of Materials Science and Engineering
- Pohang University of Science and Technology (POSTECH)
- Pohang, Korea
| |
Collapse
|
48
|
Meng Q, Yin Q, Li Y. Nanocarriers for siRNA delivery to overcome cancer multidrug resistance. ACTA ACUST UNITED AC 2013. [DOI: 10.1007/s11434-013-6030-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
49
|
Zuckerman JE, Davis ME. Targeting therapeutics to the glomerulus with nanoparticles. Adv Chronic Kidney Dis 2013; 20:500-7. [PMID: 24206602 DOI: 10.1053/j.ackd.2013.06.003] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 06/07/2013] [Accepted: 06/07/2013] [Indexed: 12/19/2022]
Abstract
Nanoparticles are an enabling technology for the creation of tissue-/cell-specific therapeutics that have been investigated extensively as targeted therapeutics for cancer. The kidney, specifically the glomerulus, is another accessible site for nanoparticle delivery that has been relatively overlooked as a target organ. Given the medical need for the development of more potent, kidney-targeted therapies, the use of nanoparticle-based therapeutics may be one such solution to this problem. Here, we review the literature on nanoparticle targeting of the glomerulus. Specifically, we provide a broad overview of nanoparticle-based therapeutics and how the unique structural characteristics of the glomerulus allow for selective, nanoparticle targeting of this area of the kidney. We then summarize literature examples of nanoparticle delivery to the glomerulus and elaborate on the appropriate nanoparticle design criteria for glomerular targeting. Finally, we discuss the behavior of nanoparticles in animal models of diseased glomeruli and review examples of nanoparticle therapeutic approaches that have shown promise in animal models of glomerulonephritic disease.
Collapse
|
50
|
Inorganic nanomaterials as delivery systems for proteins, peptides, DNA, and siRNA. Curr Opin Colloid Interface Sci 2013. [DOI: 10.1016/j.cocis.2013.06.002] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|