1
|
Jin M, Cho HJ, Na YG, Yun TS, Song B, Lee SR, Je S, Oh HG, Park YG, Rho J, Yang MJ, Shin JS, Baek JS, Lee HK, Kim TW, Cho CW. Practical approach to development of GS-445124-loaded PLGA nanoparticles for the long-term treatment of feline infectious peritonitis caused by feline coronavirus infection. Int J Pharm 2025; 674:125468. [PMID: 40107469 DOI: 10.1016/j.ijpharm.2025.125468] [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: 10/24/2024] [Revised: 02/13/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
Feline infectious peritonitis virus (FIPV) caused by feline coronavirus (FCoV) infection leads to a high mortality rate when untreated. GS-441524, an antiviral agent effective against FIPV, is orally administered twice daily or through daily subcutaneous injections for approximately 12 weeks. While the short treatment period recuses concerns about adherence, frequent administrations may cause handling-related stress in cats. Therefore, it is essential to develop a long-acting formulation that requires only a single administration. In this study, we used polylactide-co-glycolide (PLGA) as a carrier, which was used to effectively encapsulate GS-441524 with sustained-release functionality and GS-441524-loaded PLGA nanoparticles (GS-PLGA NP) were prepared. The particle size of GS-PLGA NP was 216 nm, the encapsulation efficiency was 78 %, and the 7-day release was 92 %. When GS-PLGA NP was injected at 22 mg/kg in cats, higher systemic exposure can be expected compared to injecting GS-441524 at 4 mg/kg for one week (relative bioavailability, 152 %). As well as GS-PLGA NP showed lower toxicity, improved cellular uptake, and enhanced antiviral efficacy against FCoV compared to the pure GS-441524.
Collapse
Affiliation(s)
- Minki Jin
- College of Pharmacy and Institute of Drug Research and Development Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea; Center for Large Animals Convergence Research, Korea Institute of Toxicology, Jeongeup, 56212, Republic of Korea
| | - Hyun-Jin Cho
- Center for Large Animals Convergence Research, Korea Institute of Toxicology, Jeongeup, 56212, Republic of Korea; College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Young-Guk Na
- College of Pharmacy and Institute of Drug Research and Development Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Taek-Seon Yun
- College of Pharmacy and Institute of Drug Research and Development Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Bomin Song
- College of Pharmacy and Institute of Drug Research and Development Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Sang-Rae Lee
- College of Pharmacy and Institute of Drug Research and Development Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Seonho Je
- College of Pharmacy and Institute of Drug Research and Development Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Hong-Geun Oh
- R&D Division, Huvet Co., Ltd, Iksan-si 54541, Republic of Korea
| | - Yang-Gyu Park
- R&D Division, Huvet Co., Ltd, Iksan-si 54541, Republic of Korea
| | - Jinhyung Rho
- Center for Translational Toxicologic Research, Korea Institute of Toxicology, Jeongeup, 56212, Republic of Korea
| | - Mi-Jin Yang
- Center for Translational Toxicologic Research, Korea Institute of Toxicology, Jeongeup, 56212, Republic of Korea
| | - Jin Soo Shin
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Jong-Suep Baek
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Hong-Ki Lee
- College of Veterinary Medicine, Chungbuk National University, 1 Chungdae-ro, Seowon-Gu, Cheongju, Chungbuk 28644, Republic of Korea.
| | - Tae-Won Kim
- College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea.
| | - Cheong-Weon Cho
- College of Pharmacy and Institute of Drug Research and Development Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea.
| |
Collapse
|
2
|
Wang R, Zhou P, Xu W, Li D, Xue S, Guo Z, Li J, Jin L, Zuo C, Chen H, Li R, Li X, Lou J. An Auger electron-loaded theranostic biosensor triggered by the ACE2-mediated virus/host endocytosis. Talanta 2025; 285:127288. [PMID: 39632316 DOI: 10.1016/j.talanta.2024.127288] [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: 06/03/2024] [Revised: 11/21/2024] [Accepted: 11/24/2024] [Indexed: 12/07/2024]
Abstract
Accurate diagnosis and effective antiviral strategies are critical to combat acute infection and to avoid damage to the host. Due to their restricted radiation range and energy, Auger electron emitters have shown potential as a RNA-destructing radionuclide therapy in oncology and infection. Focusing on the process of angiotensin-converting enzyme 2 (ACE2)-mediated endocytosis, Technetium-99m-labeled DX600 (99mTc-DX600) was synthesized as an Auger electron vector to specifically bind to surface-expressed ACE2 proteins on 293T-hACE2 cells (293T cells stably expressing human ACE2), and Technetium-99m-loaded microvesicles (99mTc-MVs) served as an antiviral tracer and effector in pseudovirus infection. The whole-body ACE2 expression evaluation was non-invasive, meanwhile, the enhanced green fluorescent protein expression of pseudoviruses was substantially inhibited as a result of the 99mTc-DX600 loading of microvesicles, though the mitochondrial and DNA stabilities of the host cells were not affected. Furthermore, the in vivo distribution of 99mTc-DX600 in humanized ACE2 mice was demonstrated to be both ACE2-specific and long-lasting, and an antiviral effect was fully exhibited with two cycles of intravenous injection at a dosage of 37 MBq. Taking advantage of the ACE2-mediated interaction and natural trigger mechanism of virus-induced endocytosis, 99mTc-MV represents a theranostic biosensor of Auger electrons that can expose viral RNA to lethal amounts of radiation, with the host cells receiving no detrimental radiation.
Collapse
Affiliation(s)
- Ruizhi Wang
- Department of Radiology, Huadong Hospital, Fudan University, Shanghai 200040, China
| | - Pan Zhou
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China; School of Chemistry and Bioengineering, Yichun University, Yichun, Jiangxi 336000, China
| | - Wen Xu
- Department of Radiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei 441000, China
| | - Danni Li
- Department of Nuclear Medicine, Shanghai Changhai Hospital, Shanghai 200433, China
| | - Shuai Xue
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China; School of Chemistry and Bioengineering, Yichun University, Yichun, Jiangxi 336000, China
| | - Zhongqiu Guo
- Department of Nuclear Medicine, Shanghai Changhai Hospital, Shanghai 200433, China
| | - Jie Li
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
| | - Liang Jin
- Department of Radiology, Huadong Hospital, Fudan University, Shanghai 200040, China
| | - Changjing Zuo
- Department of Nuclear Medicine, Shanghai Changhai Hospital, Shanghai 200433, China
| | - Hui Chen
- Department of Radiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei 441000, China.
| | - Rou Li
- Department of Nuclear Medicine, Shanghai Changhai Hospital, Shanghai 200433, China.
| | - Xiao Li
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China; Department of Nuclear Medicine, Shanghai Changhai Hospital, Shanghai 200433, China; Department of Nuclear Medicine, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China.
| | - Jingjing Lou
- Department of Nuclear Medicine, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China.
| |
Collapse
|
3
|
Mendes S, Guimarães LC, de Oliveira LC, Costa PAC, da Silva NJA, Pereira GSAP, Fernandez CC, Figueiredo MM, Dos Santos RAS, Teixeira MM, Costa VV, Guimarães PPG, Frézard F. Intranasal liposomal remdesivir induces SARS-CoV-2 clearance in K18-hACE2 mice and ensures survival. J Control Release 2025; 379:558-573. [PMID: 39837387 DOI: 10.1016/j.jconrel.2025.01.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 12/31/2024] [Accepted: 01/16/2025] [Indexed: 01/23/2025]
Abstract
A huge challenge after the emergence of COVID-19 has been the discovery of effective antiviral drugs. Although remdesivir (RDV) emerged as one of the most promising drugs, its pharmaceutical formulation Veklury® is limited by moderate efficacy, high toxicity and need for parenteral administration. The aim of the present work was to develop a liposomal formulation of RDV for pulmonary administration and evaluate its efficacy in models of COVID-19. Liposomal RDV nanoformulation (LRDV) was selected based on high drug encapsulation efficiency, sustained drug release property and high in vitro selectivity index. A pharmacokinetic study of intranasal LRDV in mice demonstrated effective delivery of the drug to the lungs. LRDV was then evaluated for its efficacy in SARS-CoV-2-infected K18-hACE2 mice after repeated intranasal administration at 10 mg/kg/bid for 5 days. Veklury® given intraperitoneally at 20 mg/kg/bid was used for comparison. Mice receiving LRDV remained alive up to 15 days post-infection (dpi). On the other hand, the control groups receiving PBS and empty liposomes showed 100 % death at 6 dpi and the Veklury® group had 62.5 % death at 8 dpi. Intranasal LRDV also promoted a strong reduction in viral loads in the brain and lungs of mice and prevented the inflammatory response induced by SARS-CoV-2 in the lungs. This is in contrast with Veklury®, which did not significantly reduce the viral titer in the brain and was poorly effective in preventing the inflammatory response in the lungs. Intranasal LRDV emerges as a promising therapeutic strategy for COVID-19, including "Long COVID".
Collapse
Affiliation(s)
- Sabrina Mendes
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Lays Cordeiro Guimarães
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Leonardo Camilo de Oliveira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Pedro Augusto Carvalho Costa
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Natália Jordana Alves da Silva
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Gabriel Silva Alves Pessim Pereira
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Clara Couto Fernandez
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | | | - Robson Augusto Souza Dos Santos
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Vivian Vasconcelos Costa
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Pedro Pires Goulart Guimarães
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Frédéric Frézard
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil.
| |
Collapse
|
4
|
Fernández-García R, Fraguas-Sánchez AI. Nanomedicines for Pulmonary Drug Delivery: Overcoming Barriers in the Treatment of Respiratory Infections and Lung Cancer. Pharmaceutics 2024; 16:1584. [PMID: 39771562 PMCID: PMC11677881 DOI: 10.3390/pharmaceutics16121584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/02/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
The pulmonary route for drug administration has garnered a great deal of attention in therapeutics for treating respiratory disorders. It allows for the delivery of drugs directly to the lungs and, consequently, the maintenance of high concentrations at the action site and a reduction in systemic adverse effects compared to other routes, such as oral or intravenous. Nevertheless, the pulmonary administration of drugs is challenging, as the respiratory system tries to eliminate inhaled particles, being the main responsible mucociliary escalator. Nanomedicines represent a primary strategy to overcome the limitations of this route as they can be engineered to prolong pulmonary retention and avoid their clearance while reducing drug systemic distribution and, consequently, systemic adverse effects. This review analyses the use of pulmonary-administered nanomedicines to treat infectious diseases affecting the respiratory system and lung carcinoma, two pathologies that represent major health threats.
Collapse
Affiliation(s)
| | - Ana I. Fraguas-Sánchez
- Department of Pharmaceutics and Food Technology, School of Pharmacy, Complutense University, 28040 Madrid, Spain
- Institute of Industrial Pharmacy, Complutense University, 28040 Madrid, Spain
| |
Collapse
|
5
|
Yu Y, Silva-Ayala D, Zhou Z, Peng Y, Fang RH, Gao W, Griffiths A, Zhang L. Cellular Nanoparticles Treat Coronavirus Infection in Vivo. NANO LETTERS 2024; 24:15136-15141. [PMID: 39535430 PMCID: PMC11613681 DOI: 10.1021/acs.nanolett.4c04653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/08/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
Cellular nanoparticles (CNPs), which refer to nanoparticles coated with natural cell membranes, are promising for neutralizing pathological agents. Here, we use CNPs as a medical countermeasure against the infection of SARS-CoV-2 variants in an animal model. CNPs comprise polymeric cores coated with the plasma membranes of human macrophages. The resulting nanoparticles (MΦ-NPs) act as host cell decoys to intercept SARS-CoV-2 and block its cellular entry, thus inhibiting subsequent viral infection. Our findings indicate that MΦ-NPs bind to the spike proteins of SARS-CoV-2 variants in a dose-dependent manner and inhibit the infectivity of live viruses. In hamsters infected with SARS-CoV-2 variants, MΦ-NPs significantly reduce the viral burden in the lungs, demonstrating their effectiveness in inhibiting viral infectivity in vivo. Furthermore, MΦ-NPs are primarily taken up by alveolar macrophages without inducing noticeable adverse effects. Given the crucial role of macrophages in viral infections, MΦ-NPs present a promising approach to combating emerging viral threats.
Collapse
Affiliation(s)
- Yiyan Yu
- Aiiso Yufeng
Li Family Department of Chemical and Nano Engineering, Shu and K.C.
Chien and Peter Farrell Collaboratory, University
of California San Diego, La Jolla, California 92093, United States
| | - Daniela Silva-Ayala
- Department
of Microbiology and National Emerging Infectious Diseases Laboratories,
School of Medicine, Boston University, Boston, Massachusetts 02115, United States
| | - Zhidong Zhou
- Aiiso Yufeng
Li Family Department of Chemical and Nano Engineering, Shu and K.C.
Chien and Peter Farrell Collaboratory, University
of California San Diego, La Jolla, California 92093, United States
| | - Yifei Peng
- Aiiso Yufeng
Li Family Department of Chemical and Nano Engineering, Shu and K.C.
Chien and Peter Farrell Collaboratory, University
of California San Diego, La Jolla, California 92093, United States
| | - Ronnie H. Fang
- Aiiso Yufeng
Li Family Department of Chemical and Nano Engineering, Shu and K.C.
Chien and Peter Farrell Collaboratory, University
of California San Diego, La Jolla, California 92093, United States
| | - Weiwei Gao
- Aiiso Yufeng
Li Family Department of Chemical and Nano Engineering, Shu and K.C.
Chien and Peter Farrell Collaboratory, University
of California San Diego, La Jolla, California 92093, United States
| | - Anthony Griffiths
- Department
of Microbiology and National Emerging Infectious Diseases Laboratories,
School of Medicine, Boston University, Boston, Massachusetts 02115, United States
- Laboratory
for Infectious Disease Research and Department of Molecular Microbiology
and Immunology, School of Medicine, University
of Missouri, Columbia, Missouri 65211, United States
| | - Liangfang Zhang
- Aiiso Yufeng
Li Family Department of Chemical and Nano Engineering, Shu and K.C.
Chien and Peter Farrell Collaboratory, University
of California San Diego, La Jolla, California 92093, United States
| |
Collapse
|
6
|
Rejinold NS, Jin GW, Choy JH. Insight into Preventing Global Dengue Spread: Nanoengineered Niclosamide for Viral Infections. NANO LETTERS 2024; 24:14541-14551. [PMID: 39194045 PMCID: PMC11583367 DOI: 10.1021/acs.nanolett.4c02845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/25/2024] [Accepted: 08/26/2024] [Indexed: 08/29/2024]
Abstract
Millions of cases of dengue virus (DENV) infection yearly from Aedes mosquitoes stress the need for effective antivirals. No current drug effectively combats dengue efficiently. Transient immunity and severe risks highlight the need for broad-spectrum antivirals targeting all serotypes of DENV. Niclosamide, an antiparasitic, shows promising antiviral activity against the dengue virus, but enhancing its bioavailability is challenging. To overcome this issue and enable niclosamide to address the global dengue problem, nanoengineered niclosamides can be the solution. Not only does it address cost issues but also with its broad-spectrum antiviral effects nanoengineered niclosamide offers hope in addressing the current health crisis associated with DENV and will play a crucial role in combating other arboviruses as well.
Collapse
Affiliation(s)
- N. Sanoj Rejinold
- Intelligent
Nanohybrid Materials Laboratory (INML), College of Medicine, Dankook University, Cheonan 31116, Republic of Korea
- Institute
of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic
of Korea
| | - Geun-woo Jin
- R&D
Center, Hyundai Bioscience Co. LTD., Seoul 03759, Republic
of Korea
| | - Jin-Ho Choy
- Intelligent
Nanohybrid Materials Laboratory (INML), College of Medicine, Dankook University, Cheonan 31116, Republic of Korea
- Division
of Natural Sciences, The National Academy
of Sciences, Seoul 06579, Republic of Korea
- Tokyo
Tech World Research Hub Initiative (WRHI), Institute of Innovative
Research, Tokyo Institute of Technology, Yokohama 226-8503, Japan
| |
Collapse
|
7
|
Shang J, Li H, Liu X, Sun S, Huan S, Xiong B. Single-particle rotational sensing for analyzing the neutralization activity of antiviral antibodies. Talanta 2024; 279:126606. [PMID: 39089080 DOI: 10.1016/j.talanta.2024.126606] [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: 03/30/2024] [Revised: 07/13/2024] [Accepted: 07/22/2024] [Indexed: 08/03/2024]
Abstract
Due to the pathogen-specific targeting, neutralization capabilities, and enduring efficacy, neutralizing antibodies (NAs) have received widespread attentions as a critical immunotherapeutic strategy against infectious viruses. However, because of the high variability and complexity of pathogens, rapid determination of neutralization activity of antiviral antibodies remains a challenge. Here, we report a new method, named as out-of-plane polarization imaging based single-particle rotational sensing, for rapid analysis of neutralization activity of antiviral antibody against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Using the spike protein functionalized gold nanorods (AuNRs) and angiotensin-converting enzyme 2 (ACE2) coated gold nanoparticles (AuNPs) as the rotational sensors and chaperone probes, we demonstrated the single-particle rotational sensing strategy for the measurement of rotational diffusion coefficient of the chaperone-bound rotational sensors caused by the specific spike protein-ACE2 interactions. This enables us to measure the neutralizing activity of neutralizing antibody from the analysis of dose-dependent changes in rotational diffusion coefficient (Dr) of the rotational sensors upon the treatment of SARS-CoV-2 antibody. With this technique, we achieved the quantitative determination of neutralization activity of a commercially available SARS-CoV-2 antibody (IC50, 294.1 ng/mL) with satisfying accuracy and anti-interference ability. This simple and robust method holds the potential for rapid and accurate evaluation of neutralization activity against different pathogenic viruses.
Collapse
Affiliation(s)
- Jinhui Shang
- College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
| | - Huiwen Li
- College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
| | - Xixuan Liu
- College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
| | - Shijie Sun
- College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
| | - Shuangyan Huan
- College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China.
| | - Bin Xiong
- College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China.
| |
Collapse
|
8
|
Chintapula U, Karim SU, Iyer PR, Asokan-Sheeja H, Neupane B, Nazneen F, Dong H, Bai F, Nguyen KT. A novel nanocomposite drug delivery system for SARS-CoV-2 infections. NANOSCALE ADVANCES 2024; 6:3747-3758. [PMID: 39050946 PMCID: PMC11265598 DOI: 10.1039/d4na00361f] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 05/19/2024] [Indexed: 07/27/2024]
Abstract
To develop an inhalable drug delivery system, we synthesized poly (lactic-co-glycolic acid) nanoparticles with Remdesivir (RDV NPs) as an antiviral agent against SARS-CoV-2 replication and formulated Remdesivir-loaded nanocomposites (RDV NCs) via coating of RDV NPs with novel supramolecular cell-penetrating peptide nanofibers (NFs) to enhance cellular uptake and intracellular drug delivery. RDV NPs and RDV NCs were characterized using variou techniques, including Transmission Electron Microscopy (TEM), Dynamic Light Scattering (DLS), and fluorescent microscopy. The cytotoxicity of RDV NCs was assessed in Vero E6 cells and primary human lung epithelial cells, with no significant cytotoxicity observed up to 1000 μg mL-1 and 48 h. RDV NCs were spherically shaped with a size range of 200-300 nm and a zeta potential of ∼+31 mV as well as indicating the presence of coated nanofibers. Reverse Transcription-quantitative Polymerase Chain Reaction (RT-qPCR), immunofluorescence and plaque assays of SARS-CoV-2 infected Vero E6 treated with RDV NCs showed significantly higher antiviral activities compared to those of free drug and uncoated RDV NPs. RDV NCs exhibited high antiviral activity against SARS-CoV-2, and the nanocomposite platform has the potential to be developed into an inhalable drug delivery system for other viral infections in the lungs.
Collapse
Affiliation(s)
- Uday Chintapula
- Department of Bioengineering, University of Texas at Arlington Arlington TX 76010 USA
| | - Shazeed-Ul Karim
- Department of Cell and Molecular Biology, University of Southern Mississippi Hattiesburg MS 39406 USA
| | | | - Haritha Asokan-Sheeja
- Department of Chemistry and Biochemistry, University of Texas at Arlington Arlington TX 76010 USA
| | - Biswas Neupane
- Department of Cell and Molecular Biology, University of Southern Mississippi Hattiesburg MS 39406 USA
| | - Farzana Nazneen
- Department of Cell and Molecular Biology, University of Southern Mississippi Hattiesburg MS 39406 USA
| | - He Dong
- Department of Chemistry and Biochemistry, University of Texas at Arlington Arlington TX 76010 USA
| | - Fengwei Bai
- Department of Cell and Molecular Biology, University of Southern Mississippi Hattiesburg MS 39406 USA
| | - Kytai T Nguyen
- Department of Bioengineering, University of Texas at Arlington Arlington TX 76010 USA
| |
Collapse
|
9
|
Santos JF, del Rocío Silva-Calpa L, de Souza FG, Pal K. Central Countries' and Brazil's Contributions to Nanotechnology. CURRENT NANOMATERIALS 2024; 9:109-147. [DOI: 10.2174/2405461508666230525124138] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 02/09/2023] [Accepted: 03/14/2023] [Indexed: 01/05/2025]
Abstract
Abstract:
Nanotechnology is a cornerstone of the scientific advances witnessed over the past few
years. Nanotechnology applications are extensively broad, and an overview of the main trends
worldwide can give an insight into the most researched areas and gaps to be covered. This document
presents an overview of the trend topics of the three leading countries studying in this area, as
well as Brazil for comparison. The data mining was made from the Scopus database and analyzed
using the VOSviewer and Voyant Tools software. More than 44.000 indexed articles published
from 2010 to 2020 revealed that the countries responsible for the highest number of published articles
are The United States, China, and India, while Brazil is in the fifteenth position. Thematic
global networks revealed that the standing-out research topics are health science, energy,
wastewater treatment, and electronics. In a temporal observation, the primary topics of research are:
India (2020), which was devoted to facing SARS-COV 2; Brazil (2019), which is developing promising
strategies to combat cancer; China (2018), whit research on nanomedicine and triboelectric
nanogenerators; the United States (2017) and the Global tendencies (2018) are also related to the
development of triboelectric nanogenerators. The collected data are available on GitHub. This study
demonstrates the innovative use of data-mining technologies to gain a comprehensive understanding
of nanotechnology's contributions and trends and highlights the diverse priorities of nations in
this cutting-edge field.
Collapse
Affiliation(s)
- Jonas Farias Santos
- Programa de Engenharia da Nanotecnologia, COPPE, Centro de Tecnologia-Cidade Universitária, Universidade
Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leydi del Rocío Silva-Calpa
- Programa de Engenharia da Nanotecnologia, COPPE, Centro de Tecnologia-Cidade Universitária, Universidade
Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernando Gomes de Souza
- Programa de Engenharia da Nanotecnologia, COPPE, Centro de Tecnologia-Cidade Universitária, Universidade
Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto de Macromoléculas Professora Eloisa Mano, Centro de
Tecnologia-Cidade Universitária, Universidade Federal de Rio de Janeiro, Rio de Janeiro, Brazil
| | - Kaushik Pal
- University Center
for Research and Development (UCRD), Department of Physics, Chandigarh University, Ludhiana - Chandigarh State
Hwy, Mohali, Gharuan, 140413 Punjab, India
| |
Collapse
|
10
|
Wang H, Luo S, Xie M, Chen Z, Zhang Y, Xie Z, Zhang Y, Zhang Y, Yang L, Wu F, Chen X, Du G, Zhao J, Sun X. ACE2 Receptor-Targeted Inhaled Nanoemulsions Inhibit SARS-CoV-2 and Attenuate Inflammatory Responses. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2311537. [PMID: 38174591 DOI: 10.1002/adma.202311537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/12/2023] [Indexed: 01/05/2024]
Abstract
Three kinds of coronaviruses are highly pathogenic to humans, and two of them mainly infect humans through Angiotensin-converting enzyme 2 (ACE2)receptors. Therefore, specifically blocking ACE2 binding at the interface with the receptor-binding domain is promising to achieve both preventive and therapeutic effects of coronaviruses. Alternatively, drug-targeted delivery based on ACE2 receptors can further improve the efficacy and safety of inhalation drugs. Here, these two approaches are innovatively combined by designing a nanoemulsion (NE) drug delivery system (termed NE-AYQ) for inhalation that targets binding to ACE2 receptors. This inhalation-delivered remdesivir nanoemulsion (termed RDSV-NE-AYQ) effectively inhibits the infection of target cells by both wild-type and mutant viruses. The RDSV-NE-AYQ strongly inhibits Severe acute respiratory syndrome coronavirus 2 at two dimensions: they not only block the binding of the virus to host cells at the cell surface but also restrict virus replication intracellularly. Furthermore, in the mouse model of acute lung injury, the inhaled drug delivery system loaded with anti-inflammatory drugs (TPCA-1-NE-AYQ) can significantly alleviate the lung tissue injury of mice. This smart combination provides a new choice for dealing with possible emergencies in the future and for the rapid development of inhaled drugs for the treatment of respiratory diseases.
Collapse
Affiliation(s)
- Hairui Wang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Shuang Luo
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Mingxin Xie
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Zhao Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510182, P. R. China
| | - Yunming Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Zhiqiang Xie
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Yongshun Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Yu Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Lan Yang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Fuhua Wu
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Xiaoyan Chen
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Guangsheng Du
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Jincun Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510182, P. R. China
| | - Xun Sun
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| |
Collapse
|
11
|
Xing F, Xu J, Zhou Y, Yu P, Zhe M, Xiang Z, Duan X, Ritz U. Recent advances in metal-organic frameworks for stimuli-responsive drug delivery. NANOSCALE 2024; 16:4434-4483. [PMID: 38305732 DOI: 10.1039/d3nr05776c] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
After entering the human body, drugs for treating diseases, which are prone to delivery and release in an uncontrolled manner, are affected by various factors. Based on this, many researchers utilize various microenvironmental changes encountered during drug delivery to trigger drug release and have proposed stimuli-responsive drug delivery systems. In recent years, metal-organic frameworks (MOFs) have become promising stimuli-responsive agents to release the loaded therapeutic agents at the target site to achieve more precise drug delivery due to their high drug loading, excellent biocompatibility, and high stimuli-responsiveness. The MOF-based stimuli-responsive systems can respond to various stimuli under pathological conditions at the site of the lesion, releasing the loaded therapeutic agent in a controlled manner, and improving the accuracy and safety of drug delivery. Due to the changes in different physical and chemical factors in the pathological process of diseases, the construction of stimuli-responsive systems based on MOFs has become a new direction in drug delivery and controlled release. Based on the background of the rapidly increasing attention to MOFs applied in drug delivery, we aim to review various MOF-based stimuli-responsive drug delivery systems and their response mechanisms to various stimuli. In addition, the current challenges and future perspectives of MOF-based stimuli-responsive drug delivery systems are also discussed in this review.
Collapse
Affiliation(s)
- Fei Xing
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| | - Jiawei Xu
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| | - Yuxi Zhou
- Department of Periodontology, Justus-Liebig-University of Giessen, Germany
| | - Peiyun Yu
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Man Zhe
- Animal Experiment Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Zhou Xiang
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| | - Xin Duan
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
- Department of Orthopedic Surgery, The Fifth People's Hospital of Sichuan Province, Chengdu, China
| | - Ulrike Ritz
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany.
| |
Collapse
|
12
|
Chen H, Xiong X, Huang Y, Huang B, Luo X, Ke Q, Wu P, Wang S. SARS-CoV-2 Neutralization by Cell Membrane-Coated Antifouling Nanoparticles. ACS APPLIED BIO MATERIALS 2024; 7:909-917. [PMID: 38273679 DOI: 10.1021/acsabm.3c00936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2024]
Abstract
The global outbreak of the COVID-19 pandemic has indisputably wreaked havoc on societies worldwide, compelling the scientific community to seek urgently needed therapeutic agents with low-cost and low-side effect profiles. Numerous approaches have been investigated in the quest to prevent or treat COVID-19, but many of them exhibit unwelcome side effects, such as dysfunctional viral immune responses and inflammation. Herein, we present the preparation of solid natural human pulmonary alveolar epithelial cell (ATII) membrane-coated PLGA NPs (PLGA NPs@ATII-M), which demonstrate remarkable affinity and competitiveness to neutralize the SARS-CoV-2 S1 protein-coated NPs (SCMMA NPs-S1), which are employed as a surrogate for coronavirus particles. In addition, we first considered the antifouling properties of these types of NPs, and we found that this membrane-coated NP formulation boasts excellent antifouling capabilities, which serve to protect their neutralization properties out of shielding by protein coronas in blood circulation. Moreover, this formulation is easily prepared and stored with a low-cost profile and exhibits good specificity, high targeting efficiency, and potentially side effect avoiding, thus making it a highly promising candidate for COVID-19 treatment.
Collapse
Affiliation(s)
- Hao Chen
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, PR China
| | - Xilin Xiong
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, PR China
| | - Yuan Huang
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, PR China
| | - Bo Huang
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, PR China
| | - Xinxin Luo
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, PR China
| | - Qi Ke
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, PR China
| | - Pengyu Wu
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, PR China
| | - Suxiao Wang
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, PR China
| |
Collapse
|
13
|
Wang W, Zhong Z, Huang Z, Hiew TN, Huang Y, Wu C, Pan X. Nanomedicines for targeted pulmonary delivery: receptor-mediated strategy and alternatives. NANOSCALE 2024; 16:2820-2833. [PMID: 38289362 DOI: 10.1039/d3nr05487j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Pulmonary drug delivery of nanomedicines is promising for the treatment of lung diseases; however, their lack of specificity required for targeted delivery limit their applications. Recently, a variety of pulmonary delivery targeting nanomedicines (PDTNs) has been developed for enhancing drug accumulation in lung lesions and reducing systemic side effects. Furthermore, with the increasing profound understanding of the specific microenvironment of different local lung diseases, multiple targeting strategies have been employed to promote drug delivery efficiency, which can be divided into the receptor-mediated strategy and alternatives. In this review, the current publication trend on PDTNs is analyzed and discussed, revealing that the research in this area has been attracting much attention. According to the different unique microenvironments of lung lesions, the reported PDTNs based on the receptor-mediated strategy for lung cancer, lung infection, lung inflammation and pulmonary fibrosis are listed and summarized. In addition, several other well-established strategies for the design of these PDTNs, such as charge regulation, mucus delivery enhancement, stimulus-responsive drug delivery and magnetic force-driven targeting, are introduced and discussed. Besides, bottlenecks in the development of PDTNs are discussed. Finally, we highlight the challenges and opportunities in the development of PDTNs. We hope that this review will provide an overview of the available PDTNs for guiding the treatment of lung diseases.
Collapse
Affiliation(s)
- Wenhao Wang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, Guangdong, PR China.
| | - Ziqiao Zhong
- College of Pharmacy, Jinan University, Guangzhou 510632, Guangdong, PR China.
| | - Zhengwei Huang
- College of Pharmacy, Jinan University, Guangzhou 510632, Guangdong, PR China.
| | - Tze Ning Hiew
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa 52242, USA
| | - Ying Huang
- College of Pharmacy, Jinan University, Guangzhou 510632, Guangdong, PR China.
| | - Chuanbin Wu
- College of Pharmacy, Jinan University, Guangzhou 510632, Guangdong, PR China.
| | - Xin Pan
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, Guangdong, PR China.
| |
Collapse
|
14
|
Walter M, Baumann F, Schorr K, Goepferich A. Ectoenzymes as promising cell identification structures for the high avidity targeting of polymeric nanoparticles. Int J Pharm 2023; 647:123453. [PMID: 37783283 DOI: 10.1016/j.ijpharm.2023.123453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/04/2023]
Abstract
Pharmacotherapy is often limited by undesired side effects while insufficient drug reaches the site of action. Active-targeted nanotherapy should provide a solution for this problem, by using ligands in the nanoparticle corona for the identification of receptors on the target-cell surface. However, since receptor binding is directly associated with pharmacological responses, today's targeting concepts must be critically evaluated. We hypothesized that addressing ectoenzymes would help to overcome this problem, but it was not clear if particles would show sufficiently high avidity to provide us with a viable alternative to classical ligand-receptor concepts. We scrutinized this aspect by immobilizing the highly selective angiotensin-converting enzyme 2 (ACE2) inhibitor MLN-4760 in the corona of block-copolymer nanoparticles and investigated enzyme binding via microscale thermophoresis and flow cytometry. Excellent avidities with Kd values as low as 243 pM for soluble ACE2 and 306 pM for ACE2-positive cells were obtained. In addition, the inhibitory activity had an IC50 value of 2.88 nM. Reliable target cell identification could be proven in coculture experiments. High avidity is the basis for minimizing material loss to off-target sites and paves the way for a paradigm shift in nanoparticle targeting which does not trigger unintended side effects following target cell identification.
Collapse
Affiliation(s)
- Melanie Walter
- Department of Pharmaceutical Technology, University of Regensburg, Regensburg, Bavaria 93053, Germany
| | - Felix Baumann
- Department of Pharmaceutical Technology, University of Regensburg, Regensburg, Bavaria 93053, Germany
| | - Kathrin Schorr
- Department of Pharmaceutical Technology, University of Regensburg, Regensburg, Bavaria 93053, Germany
| | - Achim Goepferich
- Department of Pharmaceutical Technology, University of Regensburg, Regensburg, Bavaria 93053, Germany.
| |
Collapse
|
15
|
Haji Ali B, Shirvaliloo M, Fathi-Karkan S, Mirinejad S, Ulucan-Karnak F, Sargazi S, Sargazi S, Sheervalilou R, Rahman MM. Nanotechnology-Based Strategies for Extended-Release Delivery of Angiotensin Receptor Blockers (ARBs): A Comprehensive Review. Chem Biodivers 2023; 20:e202301157. [PMID: 37796134 DOI: 10.1002/cbdv.202301157] [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: 08/26/2023] [Revised: 09/30/2023] [Accepted: 10/05/2023] [Indexed: 10/06/2023]
Abstract
There has been a significant shift in the perception of hypertension as an important contributor to the global disease burden. Approximately 6 % and 8 % of pregnancies are affected by hypertension, which can adversely affect the mother and the fetus. Furthermore, a hypertensive individual is at increased risk of developing kidney disease, arterial hardening, eye damage, and strokes. Using angiotensin receptor blockers (ARBs) is widespread in treating hypertension, heart failure, coronary artery disease, and diabetic nephropathy. Despite this, some ARBs have limited use due to their poor oral bioavailability and water solubility. To tackle this, a variety of nanoparticle (NP)-based systems, such as polymeric NPs (i. e., dendrimers), polymeric micelles, polymer-drug conjugates, lipid NPs, nanoemulsions, self-emulsifying drug delivery systems (SEDDS), solid lipid NPs (SLNs), nanostructured lipid carriers (NLCs), carbon-based nanocarriers, inorganic NPs, and nanocrystals, have been recently developed for efficient delivery of losartan, Valsartan (Val), Olmesartan (OLM), Telmisartan (TEL), Candesartan, Eprosartan, Irbesartan, and Azilsartan to target cells. This review article provides a literature-based comparison of the various classes of ARBs, their mechanisms of action, and an overview of the nanoformulations developed for ARB delivery and successfully applied to managing hypertension, diabetic complications, and other conditions.
Collapse
Affiliation(s)
- Bahareh Haji Ali
- Department of Medical Physics, Iran University of Medical Sciences, Tehran, Iran
| | - Milad Shirvaliloo
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sonia Fathi-Karkan
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, 9453155166, Iran
- Department of Advanced Sciences and Technologies in Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, 9414974877, Iran
| | - Shekoufeh Mirinejad
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Fulden Ulucan-Karnak
- Department of Medical Biochemistry, Institute of Health Sciences, Ege University, İzmir, 35100, Turkey
| | - Saman Sargazi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran, Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Sara Sargazi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Roghayeh Sheervalilou
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, 9816743463, Iran
| | - Mohammed M Rahman
- Center of Excellence for Advanced Materials Research (CEAMR) & Department of Chemistry, Faculty of Science, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| |
Collapse
|
16
|
Downs IL, David Ordonez Luna A, Kota KP, Rubin SK, Shirsekar SS, Ward MD, Panchal RG, Litosh VA. Modification of N-hydroxycytidine yields a novel lead compound exhibiting activity against the Venezuelan equine encephalitis virus. Bioorg Med Chem Lett 2023; 94:129432. [PMID: 37591319 DOI: 10.1016/j.bmcl.2023.129432] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/27/2023] [Accepted: 08/03/2023] [Indexed: 08/19/2023]
Abstract
Nucleoside and nucleobase analogs capable of interfering with nucleic acid synthesis have played essential roles in fighting infectious diseases. However, many of these agents are associated with important and potentially lethal off-target intracellular effects that limit their use. Based on the previous discovery of base-modified 2'-deoxyuridines, which showed high anticancer activity while exhibiting lower toxicity toward rapidly dividing normal human cells compared to antimetabolite chemotherapeutics, we hypothesized that a similar modification of the N4-hydroxycytidine (NHC) molecule would provide novel antiviral compounds with diminished side effects. This presumption is due to the substantial structural difference with natural cytidine leading to less recognizability by host cell enzymes. Among the 42 antimetabolite species that have been synthesized and screened against VEEV, one hit compound was identified. The structural features of the modifying moiety were similar to those of the anticancer lead 2'-deoxyuridine derivative reported previously, providing an opportunity to pursue further structure-activity relationship (SAR) studies directed to lead improvement, and obtain insight into the mechanism of action, which can lead to identifying drug candidates against a broad spectrum of RNA viral infections.
Collapse
Affiliation(s)
- Isaac L Downs
- US Army Medical Research Institute of Infectious Diseases, 1425 Porter St., Fort Detrick, MD 21702, USA
| | - A David Ordonez Luna
- US Army Medical Research Institute of Infectious Diseases, 1425 Porter St., Fort Detrick, MD 21702, USA
| | - Krishna P Kota
- US Army Medical Research Institute of Infectious Diseases, 1425 Porter St., Fort Detrick, MD 21702, USA
| | - Sarah K Rubin
- US Army Medical Research Institute of Infectious Diseases, 1425 Porter St., Fort Detrick, MD 21702, USA
| | - Serena S Shirsekar
- US Army Medical Research Institute of Infectious Diseases, 1425 Porter St., Fort Detrick, MD 21702, USA
| | - Michael D Ward
- US Army Medical Research Institute of Infectious Diseases, 1425 Porter St., Fort Detrick, MD 21702, USA
| | - Rekha G Panchal
- US Army Medical Research Institute of Infectious Diseases, 1425 Porter St., Fort Detrick, MD 21702, USA
| | - Vladislav A Litosh
- US Army Medical Research Institute of Infectious Diseases, 1425 Porter St., Fort Detrick, MD 21702, USA.
| |
Collapse
|
17
|
Bowden-Reid E, Ledger S, Zhang Y, Di Giallonardo F, Aggarwal A, Stella AO, Akerman A, Milogiannakis V, Walker G, Rawlinson W, Turville S, Kelleher AD, Ahlenstiel C. Novel siRNA therapeutics demonstrate multi-variant efficacy against SARS-CoV-2. Antiviral Res 2023; 217:105677. [PMID: 37478918 DOI: 10.1016/j.antiviral.2023.105677] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 07/15/2023] [Accepted: 07/19/2023] [Indexed: 07/23/2023]
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is a respiratory virus that causes COVID-19 disease, with an estimated global mortality of approximately 2%. While global response strategies, which are predominantly reliant on regular vaccinations, have shifted from zero COVID to living with COVID, there is a distinct lack of broad-spectrum direct acting antiviral therapies that maintain efficacy across evolving SARS-CoV-2 variants of concern. This is of most concern for immunocompromised and immunosuppressed individuals who lack robust immune responses following vaccination, and others at risk for severe COVID and long-COVID. RNA interference (RNAi) therapeutics induced by short interfering RNAs (siRNAs) offer a promising antiviral treatment option, with broad-spectrum antiviral capabilities unparalleled by current antiviral therapeutics and a high genetic barrier to antiviral escape. Here we describe novel siRNAs, targeting highly conserved regions of the SARS-CoV-1 and 2 genome of both human and animal species, with multi-variant antiviral potency against eight SARS-CoV-2 lineages - Ancestral VIC01, Alpha, Beta, Gamma, Delta, Zeta, Kappa and Omicron. Treatment with our siRNA resulted in significant protection against virus-mediated cell death in vitro, with >97% cell survival (P < 0.0001), and corresponding reductions of viral nucleocapsid RNA of up to 99.9% (P < 0.0001). When compared to antivirals; Sotrovimab and Remdesivir, the siRNAs demonstrated a more potent antiviral effect and similarly, when multiplexing siRNAs to target different viral regions simultaneously, an increased antiviral effect was observed compared to individual siRNA treatments (P < 0.0001). These results demonstrate the potential for a highly effective broad-spectrum direct acting antiviral against multiple SARS-CoV-2 variants, including variants resistant to antivirals and vaccine generated neutralizing antibodies.
Collapse
Affiliation(s)
| | - Scott Ledger
- Kirby Institute, UNSW Sydney, Sydney, NSW, Australia
| | - Yuan Zhang
- Kirby Institute, UNSW Sydney, Sydney, NSW, Australia
| | | | | | | | | | | | - Gregory Walker
- New South Wales Health Pathology, Sydney, NSW, Australia
| | - William Rawlinson
- New South Wales Health Pathology, Sydney, NSW, Australia; Virology Research Laboratory, Serology and Virology Division (SAViD), Prince of Wales Hospital, Sydney, NSW, Australia
| | - Stuart Turville
- Kirby Institute, UNSW Sydney, Sydney, NSW, Australia; RNA Institute, UNSW Sydney, Sydney, NSW, Australia
| | - Anthony D Kelleher
- Kirby Institute, UNSW Sydney, Sydney, NSW, Australia; RNA Institute, UNSW Sydney, Sydney, NSW, Australia
| | - Chantelle Ahlenstiel
- Kirby Institute, UNSW Sydney, Sydney, NSW, Australia; RNA Institute, UNSW Sydney, Sydney, NSW, Australia.
| |
Collapse
|
18
|
Karsli B, Uras IS, Konuklugil B, Demirbas A. Synthesis of Axinyssa digitata Extract Directed Hybrid Nanoflower and Investigation of Its Antimicrobial Activity. IEEE Trans Nanobioscience 2023; 22:523-528. [PMID: 36269917 DOI: 10.1109/tnb.2022.3216355] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
First time in this study, the antibacterial effects of Axinyssa digitata sponge extracts and Axinyssa digitata-based cupper hybrid nanoflowers (Cu hNFs) were evaluated. Herein, hybrid nanoflowers (Cu hNFs) were produced by combining Axinyssa digitata sponge extract with Cu2+ ions in Phosphate-buffered saline (PBS) (at pH 7.4) at room temperature for three days using green synthesis method. The shape and size of hNFs were evaluated using scanning electron microscope (SEM) images. Energy dispersive X-ray spectroscopy (EDX) mapping was used to determine the presence of Cu metals and other components. X-ray diffraction (XRD) is a non-destructive analysis method that was used to determine of the crystallographic properties of materials and the phases they contain. Fourier-transform infrared spectroscopy (FT-IR) peaks were used to discuss the presence of functional groups that played a key role in the synthesis. The Cu-hNFs had antimicrobial activity against selected microorganisms. This research is expected to provide knowledge on hNFs synthesis and antimicrobial activity application investigations using Axinyssa digitata rather than biomolecules obtained through costly and time-consuming methods.
Collapse
|
19
|
Solanki R, Shankar A, Modi U, Patel S. New insights from nanotechnology in SARS-CoV-2 detection, treatment strategy, and prevention. MATERIALS TODAY. CHEMISTRY 2023; 29:101478. [PMID: 36950312 PMCID: PMC9981536 DOI: 10.1016/j.mtchem.2023.101478] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 02/13/2023] [Accepted: 02/25/2023] [Indexed: 05/14/2023]
Abstract
The recent outbreak of SARS-CoV-2 resulted into the deadly COVID-19 pandemic, which has made a profound impact on mankind and the world health care system. SARS-CoV-2 is mainly transmitted within the population via symptomatic carriers, enters the host cell via ACE2 and TMPSSR2 receptors and damages the organs. The standard diagnostic tests and treatment methods implemented lack required efficiency to beat SARS-CoV-2 in the race of its spreading. The most prominently used diagnostic test,reverse transcription-polymerase chain reaction (a nucleic acid-based method), has limitations including a prolonged time taken to reveal results, limited sensitivity, a high rate of false negative results, and lacking specificity due to a homology with other viruses. Furthermore, as part of the treatment, antiviral drugs such as remdesivir, favipiravir, lopinavir/ritonavir, chloroquine, daclatasvir, atazanavir, and many more have been tested clinically to check their potency for the treatment of SARS-CoV-2 but none of these antiviral drugs are the definitive cure or suitable prophylaxis. Thus, it is always required to combat SARS-CoV-2 spread and infection for a better and precise prognosis. This review answers the above mentioned challenges by employing nanomedicine for the development of improved detection, treatment, and prevention strategies for SARS-CoV-2. In this review, nanotechnology-based detection methods such as colorimetric assays, photothermal biosensors, molecularly imprinted nanoparticles sensors, electrochemical nanoimmunosensors, aptamer-based biosensors have been discussed. Furthermore, nanotechnology-based treatment strategies involving polymeric nanoparticles, metallic nanoparticles, lipid nanoparticles, and nanocarrier-based antiviral siRNA delivery have been depicted. Moreover, SARS-CoV-2 prevention strategies, which include the nanotechnology for upgrading personal protective equipment, facemasks, ocular protection gears, and nanopolymer-based disinfectants, have been also reviewed. This review will provide a one-site informative platform for researchers to explore the crucial role of nanomedicine in managing the COVID-19 curse more effectively.
Collapse
Affiliation(s)
- R Solanki
- School of Life Sciences, Central University of Gujarat, Sector-30, Gandhinagar, 382030, India
| | - A Shankar
- School of Life Sciences, Central University of Gujarat, Sector-30, Gandhinagar, 382030, India
| | - U Modi
- Biomaterials & Biomimetics Laboratory, School of Life Sciences, Central University of Gujarat, Sector-30, Gandhinagar, 382030, India
| | - S Patel
- School of Life Sciences, Central University of Gujarat, Sector-30, Gandhinagar, 382030, India
| |
Collapse
|
20
|
Liu M, Gan H, Liang Z, Liu L, Liu Q, Mai Y, Chen H, Lei B, Yu S, Chen H, Zheng P, Sun B. Review of therapeutic mechanisms and applications based on SARS-CoV-2 neutralizing antibodies. Front Microbiol 2023; 14:1122868. [PMID: 37007494 PMCID: PMC10060843 DOI: 10.3389/fmicb.2023.1122868] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 02/23/2023] [Indexed: 03/18/2023] Open
Abstract
COVID-19 pandemic is a global public health emergency. Despite extensive research, there are still few effective treatment options available today. Neutralizing-antibody-based treatments offer a broad range of applications, including the prevention and treatment of acute infectious diseases. Hundreds of SARS-CoV-2 neutralizing antibody studies are currently underway around the world, with some already in clinical applications. The development of SARS-CoV-2 neutralizing antibody opens up a new therapeutic option for COVID-19. We intend to review our current knowledge about antibodies targeting various regions (i.e., RBD regions, non-RBD regions, host cell targets, and cross-neutralizing antibodies), as well as the current scientific evidence for neutralizing-antibody-based treatments based on convalescent plasma therapy, intravenous immunoglobulin, monoclonal antibodies, and recombinant drugs. The functional evaluation of antibodies (i.e., in vitro or in vivo assays) is also discussed. Finally, some current issues in the field of neutralizing-antibody-based therapies are highlighted.
Collapse
Affiliation(s)
- Mingtao Liu
- National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Hui Gan
- National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Zhiman Liang
- National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Li Liu
- Guangzhou Medical University, Guangzhou, China
| | - Qiwen Liu
- Guangzhou Medical University, Guangzhou, China
| | - Yiyin Mai
- Guangzhou Medical University, Guangzhou, China
| | | | - Baoying Lei
- Guangzhou Medical University, Guangzhou, China
| | - Shangwei Yu
- Guangzhou Medical University, Guangzhou, China
| | - Huihui Chen
- Guangzhou Medical University, Guangzhou, China
| | - Peiyan Zheng
- National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Baoqing Sun
- National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, China
| |
Collapse
|
21
|
Žigrayová D, Mikušová V, Mikuš P. Advances in Antiviral Delivery Systems and Chitosan-Based Polymeric and Nanoparticulate Antivirals and Antiviral Carriers. Viruses 2023; 15:647. [PMID: 36992356 PMCID: PMC10054433 DOI: 10.3390/v15030647] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 02/24/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023] Open
Abstract
Current antiviral therapy research is focused on developing dosage forms that enable highly effective drug delivery, providing a selective effect in the organism, lower risk of adverse effects, a lower dose of active pharmaceutical ingredients, and minimal toxicity. In this article, antiviral drugs and the mechanisms of their action are summarized at the beginning as a prerequisite background to develop relevant drug delivery/carrier systems for them, classified and briefly discussed subsequently. Many of the recent studies aim at different types of synthetic, semisynthetic, and natural polymers serving as a favorable matrix for the antiviral drug carrier. Besides a wider view of different antiviral delivery systems, this review focuses on advances in antiviral drug delivery systems based on chitosan (CS) and derivatized CS carriers. CS and its derivatives are evaluated concerning methods of their preparation, their basic characteristics and properties, approaches to the incorporation of an antiviral drug in the CS polymer as well as CS nanoparticulate systems, and their recent biomedical applications in the context of actual antiviral therapy. The degree of development (i.e., research study, in vitro/ex vivo/in vivo preclinical testing), as well as benefits and limitations of CS polymer and CS nanoparticulate drug delivery systems, are reported for particular viral diseases and corresponding antivirotics.
Collapse
Affiliation(s)
- Dominika Žigrayová
- Department of Galenic Pharmacy, Faculty of Pharmacy, Comenius University Bratislava, Odbojárov 10, 83232 Bratislava, Slovakia
| | - Veronika Mikušová
- Department of Galenic Pharmacy, Faculty of Pharmacy, Comenius University Bratislava, Odbojárov 10, 83232 Bratislava, Slovakia
| | - Peter Mikuš
- Department of Pharmaceutical Analysis and Nuclear Pharmacy, Faculty of Pharmacy, Comenius University Bratislava, Odbojárov 10, 83232 Bratislava, Slovakia
- Toxicological and Antidoping Center, Faculty of Pharmacy, Comenius University Bratislava, Odbojárov 10, 83232 Bratislava, Slovakia
| |
Collapse
|
22
|
Overview of Antimicrobial Biodegradable Polyester-Based Formulations. Int J Mol Sci 2023; 24:ijms24032945. [PMID: 36769266 PMCID: PMC9917530 DOI: 10.3390/ijms24032945] [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: 11/29/2022] [Revised: 01/18/2023] [Accepted: 01/21/2023] [Indexed: 02/05/2023] Open
Abstract
As the clinical complications induced by microbial infections are known to have life-threatening side effects, conventional anti-infective therapy is necessary, but not sufficient to overcome these issues. Some of their limitations are connected to drug-related inefficiency or resistance and pathogen-related adaptive modifications. Therefore, there is an urgent need for advanced antimicrobials and antimicrobial devices. A challenging, yet successful route has been the development of new biostatic or biocide agents and biomaterials by considering the indisputable advantages of biopolymers. Polymers are attractive materials due to their physical and chemical properties, such as compositional and structural versatility, tunable reactivity, solubility and degradability, and mechanical and chemical tunability, together with their intrinsic biocompatibility and bioactivity, thus enabling the fabrication of effective pharmacologically active antimicrobial formulations. Besides representing protective or potentiating carriers for conventional drugs, biopolymers possess an impressive ability for conjugation or functionalization. These aspects are key for avoiding malicious side effects or providing targeted and triggered drug delivery (specific and selective cellular targeting), and generally to define their pharmacological efficacy. Moreover, biopolymers can be processed in different forms (particles, fibers, films, membranes, or scaffolds), which prove excellent candidates for modern anti-infective applications. This review contains an overview of antimicrobial polyester-based formulations, centered around the effect of the dimensionality over the properties of the material and the effect of the production route or post-processing actions.
Collapse
|
23
|
Qin Z, Lv G, Wang T, Li H, Zhao B, Chen M, Gang H, Tan Y, Jia H. The delivery of nanoparticles improves the pharmacokinetic properties of celecoxib to open a therapeutic window for oral administration of insoluble drugs. Biomed Chromatogr 2023; 37:e5552. [PMID: 36408991 DOI: 10.1002/bmc.5552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/30/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022]
Abstract
A sensitive and reliable LC-MS/MS method is established and validated to determine the concentration of celecoxib, in the serum of cynomolgus monkey, using celecoxib-D7 as an internal standard. The pharmacokinetic process was investigated after giving Celebrex, celecoxib nanoparticles (CXB-NPs) and hyaluronic acid celecoxib nanoparticles (HA-CXB-NPs) by intragastric (i.g.) administration. Chromatographic separation was performed with a C18 column (2.1 × 100 mm, 2.6 μm) at 40°C with a mobile phase of 2‰ HCOOH in water and acetonitrile. The mass spectral acquisition was then performed in the multiple reaction monitoring mode, with negative ESI ion at m/z 380.0 → 316.0 and m/z 387.1 → 323.1 for celecoxib and celecoxib-D7, respectively. Good linearity was observed over the concentration range from 3 to 2,000 ng/ml (R2 = 0.9954). The intra- and inter-day precision and accuracy, matrix effect and extraction recovery, as well as stability, all met the determination requirements of biological samples. The pharmacokinetic parameters of Celebrex, CXB-NPs and HA-CXB-NPs were determined as: area under the curve, 1,855.98 ± 346.59, 1,908.00 ± 1,130.24 and 2,164.48 ± 657.47 h·ng/ml; peak concentration, 261.08 ± 113.26, 261.12 ± 94.67 and 263.34 ± 151.78 μg/L; time to peak concentration, 2.00 ± 1.22, 4.00 ± 0.00 and 3.60 ± 0.89 h; half-life, 4.39 ± 1.26, 2.33 ± 0.94 and 4.92 ± 3.13 h; relative bioavailability, 102.80 ± 49.62 and 116.63 ± 25.55%. The validated method was successfully applied to the pharmacokinetic study of celecoxib in cynomolgus monkey, after i.g. administration. The preparation of the nanoparticles of celecoxib and the modification of hyaluronic acid on the surface of nanoparticles could improve the bioavailability and prolong the circulation of celecoxib in vivo, which could lay the foundation for further development of celecoxib nanoparticles.
Collapse
Affiliation(s)
- Zhenmiao Qin
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, China
| | - Geng Lv
- Guangdong Pharmaceutical University, Guangzhou, China
| | - Tong Wang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, China
| | - Hailong Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, China
| | - Beicheng Zhao
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, China
| | - Meili Chen
- Guangdong Laboratory Animals Monitoring Institute, Guangdong Provincial Key Laboratory of Laboratory Animals, Guangzhou, China
| | - Hou Gang
- Department of Orthopaedics, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yinfeng Tan
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, China
| | - Huanhuan Jia
- Guangdong Laboratory Animals Monitoring Institute, Guangdong Provincial Key Laboratory of Laboratory Animals, Guangzhou, China
| |
Collapse
|
24
|
Chaudhary KR, Kujur S, Singh K. Recent advances of nanotechnology in COVID 19: A critical review and future perspective. OPENNANO 2023; 9. [PMCID: PMC9749399 DOI: 10.1016/j.onano.2022.100118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The global anxiety and economic crisis causes the deadly pandemic coronavirus disease of 2019 (COVID 19) affect millions of people right now. Subsequently, this life threatened viral disease is caused due to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, morbidity and mortality of infected patients are due to cytokines storm syndrome associated with lung injury and multiorgan failure caused by COVID 19. Thereafter, several methodological advances have been approved by WHO and US-FDA for the detection, diagnosis and control of this wide spreadable communicable disease but still facing multi-challenges to control. Herein, we majorly emphasize the current trends and future perspectives of nano-medicinal based approaches for the delivery of anti-COVID 19 therapeutic moieties. Interestingly, Nanoparticles (NPs) loaded with drug molecules or vaccines resemble morphological features of SARS-CoV-2 in their size (60–140 nm) and shape (circular or spherical) that particularly mimics the virus facilitating strong interaction between them. Indeed, the delivery of anti-COVID 19 cargos via a nanoparticle such as Lipidic nanoparticles, Polymeric nanoparticles, Metallic nanoparticles, and Multi-functionalized nanoparticles to overcome the drawbacks of conventional approaches, specifying the site-specific targeting with reduced drug loading and toxicities, exhibit their immense potential. Additionally, nano-technological based drug delivery with their peculiar characteristics of having low immunogenicity, tunable drug release, multidrug delivery, higher selectivity and specificity, higher efficacy and tolerability switch on the novel pathway for the prevention and treatment of COVID 19.
Collapse
Affiliation(s)
- Kabi Raj Chaudhary
- Department of Pharmaceutics, ISF College of Pharmacy, Ghal Kalan, Ferozpur G.T Road, Moga, Punjab 142001, India,Department of Research and Development, United Biotech (P) Ltd. Bagbania, Nalagarh, Solan, Himachal Pradesh, India,Corresponding author at: Department of Pharmaceutics, ISF College of Pharmacy, Ghal Kalan, Ferozpur G.T Road, MOGA, Punjab 142001, India
| | - Sima Kujur
- Department of Pharmaceutics, ISF College of Pharmacy, Ghal Kalan, Ferozpur G.T Road, Moga, Punjab 142001, India
| | - Karanvir Singh
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Ghal Kalan, Ferozpur G.T Road, Moga, Punjab 142001, India,Department of Research and Development, United Biotech (P) Ltd. Bagbania, Nalagarh, Solan, Himachal Pradesh, India
| |
Collapse
|
25
|
Role of Nanomaterials in COVID-19 Prevention, Diagnostics, Therapeutics, and Vaccine Development. JOURNAL OF NANOTHERANOSTICS 2022. [DOI: 10.3390/jnt3040011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Facing the deadly pandemic caused by the SARS-CoV-2 virus all over the globe, it is crucial to devote efforts to fighting and preventing this infectious virus. Nanomaterials have gained much attention after the approval of lipid nanoparticle-based COVID-19 vaccines by the United States Food and Drug Administration (USFDA). In light of increasing demands for utilizing nanomaterials in the management of COVID-19, this comprehensive review focuses on the role of nanomaterials in the prevention, diagnostics, therapeutics, and vaccine development of COVID-19. First, we highlight the variety of nanomaterials usage in the prevention of COVID-19. We discuss the advantages of nanomaterials as well as their uses in the production of diagnostic tools and treatment methods. Finally, we review the role of nanomaterials in COVID-19 vaccine development. This review offers direction for creating products based on nanomaterials to combat COVID-19.
Collapse
|
26
|
León-Gutiérrez G, Elste JE, Cabello-Gutiérrez C, Millán-Pacheco C, Martínez-Gómez MH, Mejía-Alvarez R, Tiwari V, Mejía A. A potent virucidal activity of functionalized TiO 2 nanoparticles adsorbed with flavonoids against SARS-CoV-2. Appl Microbiol Biotechnol 2022; 106:5987-6002. [PMID: 35951081 PMCID: PMC9366830 DOI: 10.1007/s00253-022-12112-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/22/2022] [Accepted: 07/27/2022] [Indexed: 12/24/2022]
Abstract
Abstract The coronavirus SARS-CoV-2 has caused a pandemic with > 550 millions of cases and > 6 millions of deaths worldwide. Medical management of COVID-19 relies on supportive care as no specific targeted therapies are available yet. Given its devastating effects on the economy and mental health, it is imperative to develop novel antivirals. An ideal candidate will be an agent that blocks the early events of viral attachment and cell entry, thereby preventing viral infection and spread. This work reports functionalized titanium dioxide (TiO2)-based nanoparticles adsorbed with flavonoids that block SARS-CoV-2 entry and fusion. Using molecular docking analysis, two flavonoids were chosen for their specific binding to critical regions of the SARS-CoV-2 spike glycoprotein that interacts with the host cell angiotensin-converting enzyme-2 (ACE-2) receptor. These flavonoids were adsorbed onto TiO2 functionalized nanoparticles (FTNP). This new nanoparticulate compound was assayed in vitro against two different coronaviruses; HCoV 229E and SARS-CoV-2, in both cases a clear antiviral effect was observed. Furthermore, using a reporter-based cell culture model, a potent antiviral activity is demonstrated. The adsorption of flavonoids to functionalized TiO2 nanoparticles induces a ~ threefold increase of that activity. These studies also indicate that FTNP interferes with the SARS-CoV-2 spike, impairing the cell fusion mechanism. Key points/Highlights • Unique TiO2nanoparticles displaying flavonoid showed potent anti-SARS-CoV-2 activity. • The nanoparticles precisely targeting SARS-CoV-2 were quantitatively verified by cell infectivity in vitro. • Flavonoids on nanoparticles impair the interactions between the spike glycoprotein and ACE-2 receptor. Graphical abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1007/s00253-022-12112-9.
Collapse
Affiliation(s)
- Gabriela León-Gutiérrez
- Departamento de Biotecnología, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de Mexico, Mexico
| | - James Edward Elste
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL, USA
| | - Carlos Cabello-Gutiérrez
- Departamento de Virología e Investigación en Micología, Instituto Nacional de Enfermedades Respiratorias, Ciudad de Mexico, Mexico
| | - Cesar Millán-Pacheco
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, Mexico
| | - Mario H Martínez-Gómez
- Departamento de Biotecnología, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de Mexico, Mexico
| | - Rafael Mejía-Alvarez
- Department of Physiology, College of Graduate Studies, Midwestern University, Downers Grove, IL, USA
| | - Vaibhav Tiwari
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL, USA
| | - Armando Mejía
- Departamento de Biotecnología, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de Mexico, Mexico.
| |
Collapse
|
27
|
Lawal B, Kuo YC, Rachmawati Sumitra M, Wu ATH, Huang HS. Identification of a novel immune-inflammatory signature of COVID-19 infections, and evaluation of pharmacokinetics and therapeutic potential of RXn-02, a novel small-molecule derivative of quinolone. Comput Biol Med 2022; 148:105814. [PMID: 35841781 PMCID: PMC9272679 DOI: 10.1016/j.compbiomed.2022.105814] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 06/07/2022] [Accepted: 07/03/2022] [Indexed: 01/18/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is a global pandemic and respiratory infection that has enormous damage to human lives and economies. It is caused by SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2), a non-pair-stranded positive-sense RNA virus. With increasing global threats and few therapeutic options, the discovery of new potential drug targets and the development of new therapy candidates against COVID-19 are urgently needed. Based on these premises, we conducted an analysis of transcriptomic datasets from SARS-CoV-2-infected patients and identified several SARS-CoV-2 infection signatures, among which TNFRSF5/PTPRC/IDO1/MKI67 appeared to be the most pertinent signature. Subsequent integrated bioinformatics analysis identified the signature as an important immunomodulatory and inflammatory signature of SARS-CoV-2 infection. It was suggested that this gene signature mediates the interplay of immune and immunosuppressive cells leading to infiltration-exclusion of effector memory T cells in the lungs, which is of translation relevance for developing novel SARS-CoV-2 drug and vaccine candidates. Consequently, we designed and synthesized a novel small-molecule quinoline derivative (RXn-02) and evaluated its pharmacokinetics in rats, revealing a peak plasma concentration (Cmax) and time to Cmax (Tmax) of 1.756 μg/mL and 0.6 h, respectively. Values of the area under the curve (AUC) (0–24 h) and AUC (0 h∼∞) were 18.90 and 71.20 μg h/mL, respectively. Drug absorption from the various regional segments revealed that the duodenum (49.84%), jejunum (47.885%), cecum (1.82%), and ileum (0.32%) were prime sites of RXn-02 absorption. No absorption was detected from the stomach, and the least was from the colon (0.19%). Interestingly, RXn-02 exhibited in vitro antiproliferative activities against hub gene hyper-expressing cell lines; A549 (IC50 = 48.1 μM), K-562 (IC50 = 100 μM), and MCF7 (IC50 = 0.047 μM) and against five cell lines originating from human lungs (IC50 range of 33.2–69.5 μM). In addition, RXn-02 exhibited high binding efficacies for targeting the TNFRSF5/PTPRC/IDO1/MK signature with binding affinities (ΔG) of −6.6, −6.0, −9.9, −6.9 kcal/mol respectively. In conclusion, our study identified a novel signature of SARS-CoV-2 pathogenesis. RXn-02 is a drug-like candidate with good in vivo pharmacokinetics and hence possesses great translational relevance worthy of further preclinical and clinical investigations for treating SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Bashir Lawal
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan; Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Yu-Cheng Kuo
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan; School of Post-baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan
| | - Maryam Rachmawati Sumitra
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan; Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Alexander T H Wu
- The PhD Program of Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; Clinical Research Center, Taipei Medical University Hospital, Taipei Medical University, Taipei, 11031, Taiwan; TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, 11031, Taiwan; Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 11490, Taiwan.
| | - Hsu-Shan Huang
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan; Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 11490, Taiwan; School of Pharmacy, National Defense Medical Center, Taipei, 11490, Taiwan; PhD Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan.
| |
Collapse
|
28
|
Moreno-Mendieta S, Guillén D, Vasquez-Martínez N, Hernández-Pando R, Sánchez S, Rodríguez-Sanoja R. Understanding the Phagocytosis of Particles: the Key for Rational Design of Vaccines and Therapeutics. Pharm Res 2022; 39:1823-1849. [PMID: 35739369 DOI: 10.1007/s11095-022-03301-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 05/23/2022] [Indexed: 12/17/2022]
Abstract
A robust comprehension of phagocytosis is crucial for understanding its importance in innate immunity. A detailed description of the molecular mechanisms that lead to the uptake and clearance of endogenous and exogenous particles has helped elucidate the role of phagocytosis in health and infectious or autoimmune diseases. Furthermore, knowledge about this cellular process is important for the rational design and development of particulate systems for the administration of vaccines or therapeutics. Depending on these specific applications and the required biological responses, particles must be designed to encourage or avoid their phagocytosis and prolong their circulation time. Functionalization with specific polymers or ligands and changes in the size, shape, or surface of particles have important effects on their recognition and internalization by professional and nonprofessional phagocytes and have a major influence on their fate and safety. Here, we review the phagocytosis of particles intended to be used as carrier or delivery systems for vaccines or therapeutics, the cells involved in this process depending on the route of administration, and the strategies employed to obtain the most desirable particles for each application through the manipulation of their physicochemical characteristics. We also offer a view of the challenges and potential opportunities in the field and give some recommendations that we expect will enable the development of improved approaches for the rational design of these systems.
Collapse
Affiliation(s)
- Silvia Moreno-Mendieta
- Consejo Nacional de Ciencia y Tecnología (CONACyT), Ciudad de México, Mexico. .,Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), A.P. 70228, Ciudad Universitaria, 04510, Ciudad de México, Mexico.
| | - Daniel Guillén
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), A.P. 70228, Ciudad Universitaria, 04510, Ciudad de México, Mexico
| | - Nathaly Vasquez-Martínez
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), A.P. 70228, Ciudad Universitaria, 04510, Ciudad de México, Mexico.,Doctorado en Ciencias Bioquímicas, Universidad Nacional Autónoma de México (UNAM), A.P. 70228, Ciudad Universitaria, 04510, Ciudad de México, Mexico
| | - Rogelio Hernández-Pando
- Sección de Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Delegación Tlalpan, Ciudad de México, Mexico
| | - Sergio Sánchez
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), A.P. 70228, Ciudad Universitaria, 04510, Ciudad de México, Mexico
| | - Romina Rodríguez-Sanoja
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), A.P. 70228, Ciudad Universitaria, 04510, Ciudad de México, Mexico.
| |
Collapse
|
29
|
Satta S, Shahabipour F, Gao W, Lentz SR, Perlman S, Ashammakhi N, Hsiai T. Engineering viral genomics and nano-liposomes in microfluidic platforms for patient-specific analysis of SARS-CoV-2 variants. Theranostics 2022; 12:4779-4790. [PMID: 35832078 PMCID: PMC9254234 DOI: 10.7150/thno.72339] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/21/2022] [Indexed: 11/15/2022] Open
Abstract
New variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are continuing to spread globally, contributing to the persistence of the COVID-19 pandemic. Increasing resources have been focused on developing vaccines and therapeutics that target the Spike glycoprotein of SARS-CoV-2. Recent advances in microfluidics have the potential to recapitulate viral infection in the organ-specific platforms, known as organ-on-a-chip (OoC), in which binding of SARS-CoV-2 Spike protein to the angiotensin-converting enzyme 2 (ACE2) of the host cells occurs. As the COVID-19 pandemic lingers, there remains an unmet need to screen emerging mutations, to predict viral transmissibility and pathogenicity, and to assess the strength of neutralizing antibodies following vaccination or reinfection. Conventional detection of SARS-CoV-2 variants relies on two-dimensional (2-D) cell culture methods, whereas simulating the micro-environment requires three-dimensional (3-D) systems. To this end, analyzing SARS-CoV-2-mediated pathogenicity via microfluidic platforms minimizes the experimental cost, duration, and optimization needed for animal studies, and obviates the ethical concerns associated with the use of primates. In this context, this review highlights the state-of-the-art strategy to engineer the nano-liposomes that can be conjugated with SARS-CoV-2 Spike mutations or genomic sequences in the microfluidic platforms; thereby, allowing for screening the rising SARS-CoV-2 variants and predicting COVID-19-associated coagulation. Furthermore, introducing viral genomics to the patient-specific blood accelerates the discovery of therapeutic targets in the face of evolving viral variants, including B1.1.7 (Alpha), B.1.351 (Beta), B.1.617.2 (Delta), c.37 (Lambda), and B.1.1.529 (Omicron). Thus, engineering nano-liposomes to encapsulate SARS-CoV-2 viral genomic sequences enables rapid detection of SARS-CoV-2 variants in the long COVID-19 era.
Collapse
Affiliation(s)
- Sandro Satta
- Department of Bioengineering, School of Engineering, University of California, Los Angeles, California, USA
- Department of Medicine, Greater Los Angeles VA Healthcare System, Los Angeles, California, USA
- Division of Cardiology, Department of Medicine, School of Medicine, University of California, Los Angeles, California, USA
| | - Fahimeh Shahabipour
- Skin Research Center, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Wei Gao
- Medical Engineering, California Institute of Technology, California, Pasadena, USA
| | - Steven R. Lentz
- Section of Hematology, Oncology, and Blood & Marrow Transplantation, Department of Medicine, College of Medicine, University of Iowa, Iowa, USA
| | - Stanley Perlman
- Department of Microbiology and Immunology, College of Medicine, University of Iowa, USA
| | - Nureddin Ashammakhi
- Department of Bioengineering, Henry Samueli School of Engineering & Applied Science, University of California, CA, USA
- Institute for Quantitative Health Science & Engineering and Department of Biomedical Engineering, College of Engineering, Michigan State University, MI, USA
| | - Tzung Hsiai
- Department of Bioengineering, School of Engineering, University of California, Los Angeles, California, USA
- Department of Medicine, Greater Los Angeles VA Healthcare System, Los Angeles, California, USA
- Division of Cardiology, Department of Medicine, School of Medicine, University of California, Los Angeles, California, USA
| |
Collapse
|
30
|
Inhalable Mannosylated Rifampicin–Curcumin Co-Loaded Nanomicelles with Enhanced In Vitro Antimicrobial Efficacy for an Optimized Pulmonary Tuberculosis Therapy. Pharmaceutics 2022; 14:pharmaceutics14050959. [PMID: 35631546 PMCID: PMC9145552 DOI: 10.3390/pharmaceutics14050959] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/20/2022] [Accepted: 04/25/2022] [Indexed: 02/04/2023] Open
Abstract
Among respiratory infections, tuberculosis was the second deadliest infectious disease in 2020 behind COVID-19. Inhalable nanocarriers offer the possibility of actively targeting anti-tuberculosis drugs to the lungs, especially to alveolar macrophages (cellular reservoirs of the Mycobacterium tuberculosis). Our strategy was based on the development of a mannose-decorated micellar nanoformulation based in Soluplus® to co-encapsulate rifampicin and curcumin. The former is one of the most effective anti-tuberculosis first-line drugs, while curcumin has demonstrated potential anti-mycobacterial properties. Mannose-coated rifampicin (10 mg/mL)–curcumin (5 mg/mL)-loaded polymeric micelles (10% w/v) demonstrated excellent colloidal properties with micellar size ~108 ± 1 nm after freeze-drying, and they remain stable under dilution in simulated interstitial lung fluid. Drug-loaded polymeric micelles were suitable for drug delivery to the deep lung with lung accumulation, according to the in vitro nebulization studies and the in vivo biodistribution assays of radiolabeled (99mTc) polymeric micelles, respectively. Hence, the nanoformulation did not exhibit hemolytic potential. Interestingly, the addition of mannose significantly improved (5.2-fold) the microbicidal efficacy against Mycobacterium tuberculosis H37Rv of the drug-co-loaded systems in comparison with their counterpart mannose-free polymeric micelles. Thus, this novel inhaled nanoformulation has demonstrated its potential for active drug delivery in pulmonary tuberculosis therapy.
Collapse
|
31
|
Phuna ZX, Panda BP, Hawala Shivashekaregowda NK, Madhavan P. Nanoprotection from SARS-COV-2: would nanotechnology help in Personal Protection Equipment (PPE) to control the transmission of COVID-19? INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2022:1-30. [PMID: 35253535 DOI: 10.1080/09603123.2022.2046710] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 02/21/2022] [Indexed: 06/14/2023]
Abstract
The coronavirus disease 2019 (COVID-19) has caused a worldwide outbreak. The severe acute respiratory syndrome coronavirus 2 virus can be transmitted human-to-human through droplets and close contact where personal protective equipment (PPE) is imperative to protect the individuals. The advancement of nanotechnology with significant nanosized properties can confer a higher form of protection. Incorporation of nanotechnology into facemasks can exhibit antiviral properties. Nanocoating on surfaces can achieve self-disinfecting purposes and be applied in highly populated places. Moreover, nano-based hand sanitizers can confer better sterilizing efficacies with low skin irritation as compared to alcohol-based hand sanitizers. The present review discusses the incorporation of nanotechnology into nano-based materials and coatings in facemasks, self-surface disinfectants and hand sanitizers, in the hope to contribute to the current understanding of PPE to combat COVID-19.
Collapse
Affiliation(s)
- Zhi Xin Phuna
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor, Malaysia
| | - Bibhu Prasad Panda
- Department of Pharmaceutical Technology, Schoolof Pharmacy, Faculty of Health & Medical Sciences, Taylor's University, Subang Jaya, Malaysia
| | | | - Priya Madhavan
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor, Malaysia
| |
Collapse
|