Role of Nanomaterials in COVID-19 Prevention, Diagnostics, Therapeutics, and Vaccine Development
Abstract
:1. Introduction
2. Role of Nanomaterials in the Disinfection of SARS-CoV-2
3. Role of Nanomaterials in Diagnostics of SARS-CoV-2 (COVID-19)
4. Therapeutic Strategies in COVID-19 Treatment
Nanoparticle-Based Therapeutic Applications
5. Role of Nanomaterials in Vaccine Development
5.1. Antigen Delivery via Nanoparticle Vaccine
5.2. Adjuvant Delivery via Nanoparticle Vaccine
5.3. Traditional Vaccines
5.3.1. Inactivated Vaccines
5.3.2. Live Attenuated Vaccines
5.3.3. Subunit Vaccines
5.4. Next-Generation Vaccines through Advances in Nanotechnology
5.4.1. Viral Vector Vaccines
5.4.2. Nucleic Acid-Based Vaccines
5.4.3. Peptide-Based Vaccines
6. Conclusions
Author Contributions
Funding
Data Availability Statement
Conflicts of Interest
References
- World Health Organization. WHO COVID-19 Dashboard. Available online: https://covid19.who.int (accessed on 25 August 2022).
- Zhu, N.; Zhang, D.; Wang, W.; Li, X.; Yang, B.; Song, J.; Zhao, X.; Huang, B.; Shi, W.; Lu, R.; et al. A Novel Coronavirus from Patients with Pneumonia in China, 2019. N. Engl. J. Med. 2020, 382, 727–733. [Google Scholar] [CrossRef] [PubMed]
- Yesudhas, D.; Srivastava, A.; Gromiha, M.M. COVID-19 outbreak: History, mechanism, transmission, structural studies and therapeutics. Infection 2020, 49, 199–213. [Google Scholar] [CrossRef] [PubMed]
- Li, F. Structure, Function, and Evolution of Coronavirus Spike Proteins. Annu. Rev. Virol. 2016, 3, 237–261. [Google Scholar] [CrossRef] [PubMed]
- Inoue, Y.; Tanaka, N.; Tanaka, Y.; Inoue, S.; Morita, K.; Zhuang, M.; Hattori, T.; Sugamura, K. Clathrin-Dependent Entry of Severe Acute Respiratory Syndrome Coronavirus into Target Cells Expressing ACE2 with the Cytoplasmic Tail Deleted. J. Virol. 2007, 81, 8722–8729. [Google Scholar] [CrossRef] [PubMed]
- Yang, Z.-Y.; Huang, Y.; Ganesh, L.; Leung, K.; Kong, W.P.; Schwartz, O.; Subbarao, K.; Gary, J.N. Ph-Dependent Entry of Severe Acute Respiratory Syndrome Coronavirus Is Mediated by the Spike Glycoprotein and Enhanced by Dendritic Cell Transfer through Dc-Sign. J. Virol. 2004, 78, 5642–5650. [Google Scholar] [CrossRef] [PubMed]
- Yang, N.; Shen, H.-M. Targeting the Endocytic Pathway and Autophagy Process as a Novel Therapeutic Strategy in COVID-19. Int. J. Biol. Sci. 2020, 16, 1724–1731. [Google Scholar] [CrossRef] [PubMed]
- Hofmann, H.; Pöhlmann, S. Cellular entry of the SARS coronavirus. Trends Microbiol. 2004, 12, 466–472. [Google Scholar] [CrossRef] [PubMed]
- Wang, H.; Yang, P.; Liu, K.; Guo, F.; Zhang, Y.; Zhang, G.; Jiang, C. SARS coronavirus entry into host cells through a novel clathrin- and caveolae-independent endocytic pathway. Cell Res. 2008, 18, 290–301. [Google Scholar] [CrossRef] [PubMed]
- Simmons, G.; Jacqueline, D.R.; Andrew, J.R.; Sean, M.A.; Andrew, J.P.; Bates, P. Characterization of Severe Acute Respiratory Syndrome-Associated Coronavirus (SARS-CoV) Spike Glycoprotein-Mediated Viral Entry. Proc. Natl. Acad. Sci. USA 2004, 101, 4240–4245. [Google Scholar] [CrossRef] [PubMed]
- Xiao, X.; Chakraborti, S.; Dimitrov, A.S.; Gramatikoff, K.; Dimitrov, D.S. The SARS-CoV S Glycoprotein: Expression and Functional Characterization. Biochem. Biophys. Res. Commun. 2003, 312, 1159–1164. [Google Scholar] [CrossRef] [PubMed]
- Chen, L.; Liang, J. An overview of functional nanoparticles as novel emerging antiviral therapeutic agents. Mater. Sci. Eng. C 2020, 112, 110924. [Google Scholar] [CrossRef] [PubMed]
- Balagna, C.; Perero, S.; Percivalle, E.; Nepita, E.V.; Ferraris, M. Virucidal Effect against Coronavirus SARS-CoV-2 of a Silver Nanocluster/Silica Composite Sputtered Coating. Open Ceram. 2020, 1, 100006. [Google Scholar] [CrossRef]
- Jeremiah, S.S.; Miyakawa, K.; Morita, T.; Yamaoka, Y.; Ryo, A. Potent Antiviral Effect of Silver Nanoparticles on Sars-Cov-2. Biochem. Biophys. Res. Commun. 2020, 533, 195–200. [Google Scholar] [CrossRef] [PubMed]
- van Doremalen, N.; Bushmaker, T.; Morris, D.H.; Holbrook, M.G.; Gamble, A.; Williamson, B.N.; Tamin, A.; Harcourt, J.L.; Thornburg, N.J.; Gerber, S.I.; et al. Aerosol and Surface Stability of SARS-CoV-2 as Compared with SARS-CoV-1. N. Engl. J. Med. 2020, 382, 1564–1567. [Google Scholar] [CrossRef]
- Foster, H.A.; Ditta, I.B.; Varghese, S.; Steele, A. Photocatalytic disinfection using titanium dioxide: Spectrum and mechanism of antimicrobial activity. Appl. Microbiol. Biotechnol. 2011, 90, 1847–1868. [Google Scholar] [CrossRef]
- Ye, S.; Shao, K.; Li, Z.; Guo, N.; Zuo, Y.; Li, Q.; Lu, Z.; Chen, L.; He, Q.; Han, H.-Y. Antiviral Activity of Graphene Oxide: How Sharp Edged Structure and Charge Matter. ACS Appl. Mater. Interfaces 2015, 7, 21571–21579. [Google Scholar] [CrossRef]
- Dey, P.; Bergmann, T.; Cuellar-Camacho, J.L.; Ehrmann, S.; Chowdhury, M.S.; Zhang, M.; Dahmani, I.; Haag, R.; Azab, W. Multivalent Flexible Nanogels Exhibit Broad-Spectrum Antiviral Activity by Blocking Virus Entry. ACS Nano 2018, 12, 6429–6442. [Google Scholar] [CrossRef]
- Leung, W.W.-F.; Sun, Q. Charged PVDF multilayer nanofiber filter in filtering simulated airborne novel coronavirus (COVID-19) using ambient nano-aerosols. Sep. Purif. Technol. 2020, 245, 116887. [Google Scholar] [CrossRef]
- Donskyi, I.S.; Azab, W.; Cuellar-Camacho, J.L.; Guday, G.; Lippitz, A.; Unger, W.E.S.; Osterrieder, K.; Adeli, M.; Haag, R. Functionalized nanographene sheets with high antiviral activity through synergistic electrostatic and hydrophobic interactions. Nanoscale 2019, 11, 15804–15809. [Google Scholar] [CrossRef]
- El-Atab, N.; Qaiser, N.; Badghaish, H.S.; Shaikh, S.F.; Hussain, M.M. Flexible Nanoporous Template for the Design and Development of Reusable Anti-COVID-19 Hydrophobic Face Masks. ACS Nano 2020, 14, 7659–7665. [Google Scholar] [CrossRef]
- Zhong, H.; Zhu, Z.; Lin, J.; Cheung, C.; Lu, V.L.; Yan, F.; Chan, C.-Y.; Li, G. Reusable and Recyclable Graphene Masks with Outstanding Superhydrophobic and Photothermal Performances. ACS Nano 2020, 14, 6213–6221. [Google Scholar] [CrossRef] [PubMed]
- De Maio, F.; Palmieri, V.; Babini, G.; Augello, A.; Palucci, I.; Perini, G.; Salustri, A.; Spilman, P.; De Spirito, M.; Sanguinetti, M.; et al. Graphene Nanoplatelet and Graphene Oxide Functionalization of Face Mask Materials Inhibits Infectivity of Trapped SARS-CoV-2. Iscience 2021, 24, 102788. [Google Scholar] [CrossRef] [PubMed]
- Caygill, R.L.; Blair, G.E.; Millner, P.A. A review on viral biosensors to detect human pathogens. Anal. Chim. Acta 2010, 681, 8–15. [Google Scholar] [CrossRef]
- Cajigas, S.; Alzate, D.; Orozco, J. Gold nanoparticle/DNA-based nanobioconjugate for electrochemical detection of Zika virus. Mikrochim. Acta 2020, 187, 594. [Google Scholar] [CrossRef] [PubMed]
- Huang, J.; Xie, Z.; Xie, Z.; Luo, S.; Xie, L.; Huang, L.; Fan, Q.; Zhang, Y.; Wang, S.; Zeng, T. Silver nanoparticles coated graphene electrochemical sensor for the ultrasensitive analysis of avian influenza virus H7. Anal. Chim. Acta 2016, 913, 121–127. [Google Scholar] [CrossRef]
- Layqah, L.A.; Eissa, S. An electrochemical immunosensor for the corona virus associated with the Middle East respiratory syndrome using an array of gold nanoparticle-modified carbon electrodes. Mikrochim. Acta 2019, 186, 224. [Google Scholar] [CrossRef]
- Wang, S.; Li, L.; Jin, H.; Yang, T.; Bao, W.; Huang, S.; Wang, J. Electrochemical detection of hepatitis B and papilloma virus DNAs using SWCNT array coated with gold nanoparticles. Biosens. Bioelectron. 2013, 41, 205–210. [Google Scholar] [CrossRef]
- Chou, T.-C.; Hsu, W.; Wang, C.-H.; Chen, Y.-J.; Fang, J.-M. Rapid and specific influenza virus detection by functionalized magnetic nanoparticles and mass spectrometry. J. Nanobiotechnology 2011, 9, 52. [Google Scholar] [CrossRef]
- Driskell, J.D.; Jones, C.A.; Tompkins, S.M.; Tripp, R.A. One-step assay for detecting influenza virus using dynamic light scattering and gold nanoparticles. Analyst 2011, 136, 3083–3090. [Google Scholar] [CrossRef]
- Liu, Y.; Zhang, L.; Wei, W.; Zhao, H.; Zhou, Z.; Zhang, Y.; Liu, S. Colorimetric detection of influenza A virus using antibody-functionalized gold nanoparticles. Analyst 2015, 140, 3989–3995. [Google Scholar] [CrossRef]
- Perez, J.M.; Simeone, F.J.; Saeki, Y.; Josephson, L.; Weissleder, R. Viral-Induced Self-Assembly of Magnetic Nanoparticles Allows the Detection of Viral Particles in Biological Media. J. Am. Chem. Soc. 2003, 125, 10192–10193. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Drelich, A.J.; Hopkins, C.M.; Mecozzi, S.; Li, L.; Kwon, G.; Hong, S. Gold Nanoparticles in Virus Detection: Recent Advances and Potential Considerations for SARS-CoV-2 Testing Development. WIREs Nanomed. Nanobiotechnology 2022, 14, e1754. [Google Scholar] [CrossRef] [PubMed]
- Chen, R.; Ren, C.; Liu, M.; Ge, X.; Qu, M.; Zhou, X.; Liang, M.; Liu, Y.; Li, F. Early Detection of SARS-CoV-2 Seroconversion in Humans with Aggregation-Induced near-Infrared Emission Nanoparticle-Labeled Lateral Flow Immunoassay. ACS Nano 2021, 15, 8996–9004. [Google Scholar] [CrossRef] [PubMed]
- Lew, T.T.S.; Aung, K.M.M.; Ow, S.Y.; Amrun, S.N.; Sutarlie, L.; Ng, L.F.P.; Su, X. Epitope-Functionalized Gold Nanoparticles for Rapid and Selective Detection of SARS-CoV-2 Igg Antibodies. ACS Nano 2021, 15, 12286–12297. [Google Scholar] [CrossRef] [PubMed]
- Moitra, P.; Alafeef, M.; Dighe, K.; Frieman, M.B.; Pan, D. Selective Naked-Eye Detection of SARS-CoV-2 Mediated by N Gene Targeted Antisense Oligonucleotide Capped Plasmonic Nanoparticles. ACS Nano 2020, 14, 7617–7627. [Google Scholar] [CrossRef]
- Wang, Z.; Zheng, Z.; Hu, H.; Zhou, Q.; Liu, W.; Li, X.; Liu, Z.; Wang, Y.; Ma, Y. A Point-of-Care Selenium Nanoparticle-Based Test for the Combined Detection of Anti-SARS-CoV-2 Igm and Igg in Human Serum and Blood. Lab Chip 2020, 20, 4255–4261. [Google Scholar] [CrossRef] [PubMed]
- Campos, E.V.R.; Pereira, A.E.S.; De Oliveira, J.L.; Carvalho, L.B.; Guilger-Casagrande, M.; De Lima, R.; Fraceto, L.F. How can nanotechnology help to combat COVID-19? Opportunities and urgent need. J. Nanobiotechnology 2020, 18, 125. [Google Scholar] [CrossRef] [PubMed]
- Medhi, R.; Srinoi, P.; Ngo, N.; Tran, H.-V.; Lee, T.R. Nanoparticle-Based Strategies to Combat COVID-19. ACS Appl. Nano Mater. 2020, 3, 8557–8580. [Google Scholar] [CrossRef]
- LaBauve, A.E.; Rinker, T.E.; Noureddine, A.; Serda, R.E.; Howe, J.Y.; Sherman, M.B.; Rasley, A.; Brinker, C.J.; Sasaki, D.Y.; Negrete, O.A. Lipid-Coated Mesoporous Silica Nanoparticles for the Delivery of the ML336 Antiviral to Inhibit Encephalitic Alphavirus Infection. Sci. Rep. 2018, 8, 13990. [Google Scholar] [CrossRef]
- Yang, X.X.; Li, C.M.; Huang, C.Z. Curcumin modified silver nanoparticles for highly efficient inhibition of respiratory syncytial virus infection. Nanoscale 2016, 8, 3040–3048. [Google Scholar] [CrossRef]
- Idris, A.; Davis, A.; Supramaniam, A.; Acharya, D.; Kelly, G.; Tayyar, Y.; West, N.; Zhang, P.; McMillan, C.L.D.; Soemardy, C.; et al. A SARS-CoV-2 Targeted Sirna-Nanoparticle Therapy for COVID-19. Mol. Ther. 2021, 29, 2219–2226. [Google Scholar] [CrossRef] [PubMed]
- Sanna, V.; Satta, S.; Hsiai, T.; Sechi, M. Development of Targeted Nanoparticles Loaded with Antiviral Drugs for SARS-CoV-2 Inhibition. Eur. J. Med. Chem. 2022, 231, 114121. [Google Scholar] [CrossRef] [PubMed]
- Ullah, A.; Qazi, J.; Rahman, L.; Kanaras, A.G.; Khan, W.S.; Hussain, I.; Rehman, A. Nanoparticles-Assisted Delivery of Antiviral-Sirna as Inhalable Treatment for Human Respiratory Viruses: A Candidate Approach against SARS-CoV-2. Nano Sel. 2020, 1, 612–621. [Google Scholar] [CrossRef] [PubMed]
- Wang, C.; Wang, S.; Chen, Y.; Zhao, J.; Han, S.; Zhao, G.; Kang, J.; Liu, Y.; Wang, L.; Wang, X.; et al. Membrane Nanoparticles Derived from Ace2-Rich Cells Block SARS-CoV-2 Infection. ACS Nano 2021, 15, 6340–6351. [Google Scholar] [CrossRef]
- Tseng, Y.-C.; Mozumdar, S.; Huang, L. Lipid-based systemic delivery of siRNA. Adv. Drug Deliv. Rev. 2009, 61, 721–731. [Google Scholar] [CrossRef]
- Mahase, E. COVID-19: Moderna Vaccine Is Nearly 95% Effective, Trial Involving High Risk and Elderly People Shows. BMJ Br. Med. J. 2020, 371, m4471. [Google Scholar] [CrossRef]
- Polack, F.P.; Thomas, S.J.; Kitchin, N.; Absalon, J.; Gurtman, A.; Lockhart, S.; Perez, J.L.; Pérez Marc, G.; Moreira, E.D.; Zerbini, C.; et al. Safety and efficacy of the BNT162b2 mRNA COVID-19 vaccine. N. Engl. J. Med. 2020, 383, 2603–2615. [Google Scholar] [CrossRef]
- Murphy, F.; Tchetchik, A.; Furxhi, I. Reduction of Health Care-Associated Infections (HAIs) with Antimicrobial Inorganic Nanoparticles Incorporated in Medical Textiles: An Economic Assessment. Nanomaterials 2020, 10, 999. [Google Scholar] [CrossRef]
- Weiss, C.; Carriere, M.; Fusco, L.; Capua, I.; Regla-Nava, J.A.; Pasquali, M.; Scott, J.A.; Vitale, F.; Unal, M.A.; Mattevi, C.; et al. Toward Nanotechnology-Enabled Approaches against the COVID-19 Pandemic. ACS Nano 2020, 14, 6383–6406. [Google Scholar] [CrossRef]
- Murray, J.P.; Laband, S.J. Degradation of poliovirus by adsorption on inorganic surfaces. Appl. Environ. Microbiol. 1979, 37, 480–486. [Google Scholar] [CrossRef] [Green Version]
- Han, J.; Chen, L.; Duan, S.-M.; Yang, Q.-X.; Yang, M.; Gao, C.; Zhang, B.-Y.; He, H.; Dong, X.-P. Efficient and quick inactivation of SARS coronavirus and other microbes exposed to the surfaces of some metal catalysts. Biomed. Environ. Sci. 2005, 18, 176–180. [Google Scholar] [PubMed]
- Warnes, S.L.; Little, Z.R.; Keevil, C.W. Human Coronavirus 229E Remains Infectious on Common Touch Surface Materials. mBio 2015, 6, e01697-15. [Google Scholar] [CrossRef] [PubMed]
- Borkow, G.; Zhou, S.S.; Page, T.; Gabbay, J. A Novel Anti-Influenza Copper Oxide Containing Respiratory Face Mask. PLoS ONE 2010, 5, e11295. [Google Scholar] [CrossRef] [PubMed]
- Matsunaga, T.; Tomoda, R.; Nakajima, T.; Wake, H. Photoelectrochemical Sterilization of Microbial Cells by Semiconductor Powders. FEMS Microbiol. Lett. 1985, 29, 211–214. [Google Scholar] [CrossRef]
- Kato, T.; Tohma, H.; Miki, O.; Shibata, T.; Tamura, M. Degradation of Norovirus in Sewage Treatment Water by Photocatalytic Ultraviolet Disinfection. Nippon. Steel Tech. Rep. 2005, 92, 41–44. [Google Scholar]
- Kozlova, E.A.; Safatov, A.S.; Kiselev, S.A.; Marchenko, V.Y.; Sergeev, A.A.; Skarnovich, M.O.; Emelyanova, E.K.; Smetannikova, M.A.; Buryak, G.A.; Vorontsov, A.V. Inactivation and Mineralization of Aerosol Deposited Model Pathogenic Microorganisms on Tio2 and Pt/Tio2. Environ. Sci. Technol. 2010, 44, 5121–5126. [Google Scholar] [CrossRef] [PubMed]
- Watts, R.J.; Kong, S.; Orr, M.P.; Miller, G.C.; Henry, B.E. Photocatalytic inactivation of coliform bacteria and viruses in secondary wastewater effluent. Water Res. 1995, 29, 95–100. [Google Scholar] [CrossRef]
- Zan, L.; Fa, W.; Peng, T.; Gong, Z.-K. Photocatalysis effect of nanometer TiO2 and TiO2-coated ceramic plate on Hepatitis B virus. J. Photochem. Photobiol. B Biol. 2007, 86, 165–169. [Google Scholar] [CrossRef] [PubMed]
- Han, W.; Zhang, B.; Cao, W.; Yang, D.; Okamoto, I.T.Y.; Arai, J.I.; Yan, X. The Inactivation Effect of Photocatalytic Titanium Apatite Filter on Sars Virus. Prog. Biochem. Biophys. 2004, 31, 982–985. [Google Scholar]
- Khaiboullina, S.; Uppal, T.; Dhabarde, N.; Subramanian, V.R.; Verma, S.C. Inactivation of Human Coronavirus by Titania Nanoparticle Coatings and Uvc Radiation: Throwing Light on SARS-CoV-2. Viruses 2020, 13, 19. [Google Scholar] [CrossRef]
- Palmieri, V.; Papi, M. Can Graphene Take Part in the Fight against COVID-19? Nano Today 2020, 33, 100883. [Google Scholar] [CrossRef] [PubMed]
- Chen, Y.-N.; Hsueh, Y.-H.; Hsieh, C.-T.; Tzou, D.-Y.; Chang, P.-L. Antiviral Activity of Graphene–Silver Nanocomposites against Non-Enveloped and Enveloped Viruses. Int. J. Environ. Res. Public Health 2016, 13, 430. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Du, T.; Lu, J.; Liu, L.; Dong, N.; Fang, L.; Xiao, S.; Han, H. Antiviral Activity of Graphene Oxide–Silver Nanocomposites by Preventing Viral Entry and Activation of the Antiviral Innate Immune Response. ACS Appl. Bio Mater. 2018, 1, 1286–1293. [Google Scholar] [CrossRef] [PubMed]
- Iannazzo, D.; Pistone, A.; Ferro, S.; De Luca, L.; Monforte, A.M.; Romeo, R.; Buemi, M.R.; Pannecouque, C. Graphene Quantum Dots Based Systems as HIV Inhibitors. Bioconjugate Chem. 2018, 29, 3084–3093. [Google Scholar] [CrossRef]
- Sametband, M.; Kalt, I.; Gedanken, A.; Sarid, R. Herpes Simplex Virus Type-1 Attachment Inhibition by Functionalized Graphene Oxide. ACS Appl. Mater. Interfaces 2014, 6, 1228–1235. [Google Scholar] [CrossRef]
- Song, Z.; Wang, X.; Zhu, G.; Nian, Q.; Zhou, H.; Yang, D.; Qin, C.; Tang, R. Virus Capture and Destruction by Label-Free Graphene Oxide for Detection and Disinfection Applications. Small 2014, 11, 1171–1176. [Google Scholar] [CrossRef] [PubMed]
- Raghav, P.K.; Mohanty, S. Are Graphene and Graphene-Derived Products Capable of Preventing COVID-19 Infection? Med. Hypotheses 2020, 144, 110031. [Google Scholar] [CrossRef] [PubMed]
- Stanford, M.G.; Li, J.T.; Chen, Y.; McHugh, E.A.; Liopo, A.; Xiao, H.; Tour, J.M. Self-Sterilizing Laser-Induced Graphene Bacterial Air Filter. ACS Nano 2019, 13, 11912–11920. [Google Scholar] [CrossRef] [PubMed]
- Steinberg, R.S.; Cruz, M.; Mahfouz, N.G.A.; Qiu, Y.; Hurt, R.H. Breathable Vapor Toxicant Barriers Based on Multilayer Graphene Oxide. ACS Nano 2017, 11, 5670–5679. [Google Scholar] [CrossRef] [PubMed]
- Roner, M.R.; Carraher, C.E., Jr.; Shahi, K.; Barot, G. Antiviral Activity of Metal-Containing Polymers—Organotin and Cisplatin-Like Polymers. Materials 2011, 4, 991–1012. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Canady, T.D.; Zhou, Z.; Tang, Y.; Price, D.N.; Bear, D.G.; Chi, E.Y.; Schanze, K.S.; Whitten, D.G. Cationic Phenylene Ethynylene Polymers and Oligomers Exhibit Efficient Antiviral Activity. ACS Appl. Mater. Interfaces 2011, 3, 2209–2214. [Google Scholar] [CrossRef] [PubMed]
- Cagno, V.; Andreozzi, P.; D’Alicarnasso, M.; Silva, P.J.; Mueller, M.; Galloux, M.; Le Goffic, R.; Jones, S.T.; Vallino, M.; Hodek, J.; et al. Broad-spectrum non-toxic antiviral nanoparticles with a virucidal inhibition mechanism. Nat. Mater. 2017, 17, 195–203. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, Q.; Honko, A.; Zhou, J.; Gong, H.; Downs, S.N.; Vasquez, J.H.; Fang, R.H.; Gao, W.; Griffiths, A.; Zhang, L. Cellular Nanosponges Inhibit SARS-CoV-2 Infectivity. Nano Lett. 2020, 20, 5570–5574. [Google Scholar] [CrossRef] [PubMed]
- Talebian, S.; Wallace, G.G.; Schroeder, A.; Stellacci, F.; Conde, J. Nanotechnology-Based Disinfectants and Sensors for SARS-CoV-2. Nat. Nanotechnol. 2020, 15, 618–621. [Google Scholar] [CrossRef]
- Clem, A.L.; Sims, J.; Telang, S.; Eaton, J.W.; Chesney, J. Virus detection and identification using random multiplex (RT)-PCR with 3′-locked random primers. Virol. J. 2007, 4, 65. [Google Scholar] [CrossRef] [PubMed]
- Jung, J.Y.; Yoon, H.K.; An, S.; Lee, J.W.; Ahn, E.-R.; Kim, Y.-J.; Park, H.-C.; Lee, K.; Hwang, J.H.; Lim, S.-K. Rapid oral bacteria detection based on real-time PCR for the forensic identification of saliva. Sci. Rep. 2018, 8, 10852. [Google Scholar] [CrossRef] [PubMed]
- Corman, V.M.; Landt, O.; Kaiser, M.; Molenkamp, R.; Meijer, A.; Chu, D.K.W.; Bleicker, T.; Brünink, S.; Schneider, J.; Schmidt, M.L.; et al. Detection of 2019 novel coronavirus (2019-nCoV) by real-time RT-PCR. Eurosurveillance 2020, 25, 2000045. [Google Scholar] [CrossRef]
- Shen, M.; Zhou, Y.; Ye, J.; Al-Maskri, A.A.A.; Kang, Y.; Zeng, S.; Cai, S. Recent advances and perspectives of nucleic acid detection for coronavirus. J. Pharm. Anal. 2020, 10, 97–101. [Google Scholar] [CrossRef] [PubMed]
- Xiang, J.; Yan, H.; Li, H.; Liu, T.; Lin, C.; Huang, S.; Shen, C. Evaluation of Enzyme-Linked Immunoassay and Colloidal Gold-Immunochromatographic Assay Kit for Detection of Novel Coronavirus (SARS-CoV-2) Causing an Outbreak of Pneumonia (COVID-19). medRxiv 2020. [Google Scholar] [CrossRef]
- Santiago, I. Trends and Innovations in Biosensors for COVID-19 Mass Testing. ChemBioChem 2020, 21, 2880–2889. [Google Scholar] [CrossRef] [PubMed]
- Carter, L.J.; Garner, L.V.; Smoot, J.W.; Li, Y.; Zhou, Q.; Saveson, C.J.; Sasso, J.M.; Gregg, A.C.; Soares, D.J.; Beskid, T.R.; et al. Assay Techniques and Test Development for COVID-19 Diagnosis. ACS Cent. Sci. 2020, 6, 591–605. [Google Scholar] [CrossRef]
- Jayawardena, H.S.N.; Liyanage, S.H.; Rathnayake, K.; Patel, U.; Yan, M. Analytical Methods for Characterization of Nanomaterial Surfaces. Anal. Chem. 2021, 93, 1889–1911. [Google Scholar] [CrossRef] [PubMed]
- Huang, S.-H.; Yang, T.-C.; Tsai, M.-H.; Tsai, I.-S.; Lu, H.-C.; Chuang, P.-H.; Wan, L.; Lin, Y.-J.; Lai, C.-H.; Lin, C.-W. Gold nanoparticle-based RT-PCR and real-time quantitative RT-PCR assays for detection of Japanese encephalitis virus. Nanotechnology 2008, 19, 405101. [Google Scholar] [CrossRef] [PubMed]
- Liu, Z.; Li, J.; Liu, Z.; Li, J.; Li, Z.; Wang, C.; Wang, J.; Guo, L. Development of a nanoparticle-assisted PCR assay for detection of bovine respiratory syncytial virus. BMC Veter- Res. 2019, 15, 110. [Google Scholar] [CrossRef]
- Rabiee, N.; Bagherzadeh, M.; Ghasemi, A.; Zare, H.; Ahmadi, S.; Fatahi, Y.; Dinarvand, R.; Rabiee, M.; Ramakrishna, S.; Shokouhimehr, S.; et al. Point-of-Use Rapid Detection of SARS-CoV-2: Nanotechnology-Enabled Solutions for the COVID-19 Pandemic. Int. J. Mol. Sci. 2020, 21, 5126. [Google Scholar] [CrossRef] [PubMed]
- Ruiz-Hitzky, E.; Darder, M.; Wicklein, B.; Ruiz-Garcia, C.; Martín-Sampedro, R.; del Real, G.; Aranda, P. Nanotechnology Responses to COVID-19. Adv. Healthc. Mater. 2020, 9, 2000979. [Google Scholar] [CrossRef] [PubMed]
- Draz, M.S.; Shafiee, H. Applications of gold nanoparticles in virus detection. Theranostics 2018, 8, 1985–2017. [Google Scholar] [CrossRef]
- Ventura, B.D.; Cennamo, M.; Minopoli, A.; Campanile, R.; Censi, S.B.; Terracciano, D.; Portella, G.; Velotta, R. Colorimetric Test for Fast Detection of SARS-CoV-2 in Nasal and Throat Swabs. ACS Sens. 2020, 5, 3043–3048. [Google Scholar] [CrossRef]
- Li, Z.; Yi, Y.; Luo, X.; Xiong, N.; Liu, Y.; Li, S.; Sun, R.; Wang, Y.; Hu, B.; Chen, W.; et al. Development and Clinical Application of a Rapid Igm-Igg Combined Antibody Test for SARS-CoV-2 Infection Diagnosis. J. Med. Virol. 2020, 92, 1518–1524. [Google Scholar] [CrossRef] [PubMed]
- Pramanik, A.; Gao, Y.; Patibandla, S.; Mitra, D.; McCandless, M.G.; Fassero, L.A.; Gates, K.; Tandon, R.; Ray, P.C. The rapid diagnosis and effective inhibition of coronavirus using spike antibody attached gold nanoparticles. Nanoscale Adv. 2021, 3, 1588–1596. [Google Scholar] [CrossRef]
- Alafeef, M.; Moitra, P.; Dighe, K.; Pan, D. RNA-extraction-free nano-amplified colorimetric test for point-of-care clinical diagnosis of COVID-19. Nat. Protoc. 2021, 16, 3141–3162. [Google Scholar] [CrossRef]
- He, Y.; Hu, C.; Li, Z.; Wu, C.; Zeng, Y.; Peng, C. Multifunctional carbon nanomaterials for diagnostic applications in infectious diseases and tumors. Mater. Today Bio 2022, 14, 100231. [Google Scholar] [CrossRef] [PubMed]
- Abdelhamid, H.N.; Badr, G. Nanobiotechnology as a platform for the diagnosis of COVID-19: A review. Nanotechnol. Environ. Eng. 2021, 6, 19. [Google Scholar] [CrossRef]
- Seo, G.; Lee, G.; Kim, M.J.; Baek, S.-H.; Choi, M.; Ku, K.B.; Lee, C.-S.; Jun, S.; Park, D.; Kim, H.G.; et al. Rapid Detection of COVID-19 Causative Virus (SARS-CoV-2) in Human Nasopharyngeal Swab Specimens Using Field-Effect Transistor-Based Biosensor. ACS Nano 2020, 14, 5135–5142. [Google Scholar] [CrossRef]
- Tang, C.; He, Z.; Liu, H.; Huang, H.; Yang, G.; Xiao, Z.; Li, S.; Liu, H.; Deng, Y.; Chen, Z.; et al. Application of magnetic nanoparticles in nucleic acid detection. J. Nanobiotechnol. 2020, 18, 62. [Google Scholar] [CrossRef] [PubMed]
- Jayawardena, H.S.; Rathnayake, K.M.; Patel, U.; Sentell, A.; Johnson, J.; Mustain, M.; Devarasetty, V.V.N.M. Rapid Diagnostics of Mycobacteria with Lectin Conjugated Particles. U.S. Patent No. 20210311049A1, 7 October 2021. [Google Scholar]
- Khizar, S.; Al-Dossary, A.A.; Zine, N.; Jaffrezic-Renault, N.; Errachid, A.; Elaissari, A. Contribution of magnetic particles in molecular diagnosis of human viruses. Talanta 2022, 241, 123243. [Google Scholar] [CrossRef] [PubMed]
- Gong, P.; He, X.; Wang, K.; Tan, W.; Xie, W.; Wu, P.; Li, H. Combination of Functionalized Nanoparticles and Polymerase Chain Reaction-Based Method for SARS-CoV Gene Detection. J. Nanosci. Nanotechnol. 2008, 8, 293–300. [Google Scholar] [CrossRef] [PubMed]
- Manivannan, S.; Ponnuchamy, K. Quantum dots as a promising agent to combat COVID-19. Appl. Organomet. Chem. 2020, 34, e5887. [Google Scholar] [CrossRef]
- Zhang, Y.; Malekjahani, A.; Udugama, B.N.; Kadhiresan, P.; Chen, H.; Osborne, M.; Franz, M.; Kucera, M.; Plenderleith, S.; Yip, L.; et al. Surveilling and Tracking COVID-19 Patients Using a Portable Quantum Dot Smartphone Device. Nano Lett. 2021, 21, 5209–5216. [Google Scholar] [CrossRef] [PubMed]
- Mahari, S.; Roberts, A.; Shahdeo, D.; Gandhi, S. Ecovsens-Ultrasensitive Novel in-House Built Printed Circuit Board Based Electrochemical Device for Rapid Detection of Ncovid-19 Antigen, a Spike Protein Domain 1 of SARS-CoV-2. bioRxiv 2020. [Google Scholar] [CrossRef]
- Chao, H.; Wen, T.; Shi, F.-J.; Zeng, X.-Y.; Jiao, Y.-J. Rapid Detection of Igm Antibodies against the Sars-Cov-2 Virus Via Colloidal Gold Nanoparticle-Based Lateral-Flow Assay. ACS Omega 2020, 5, 12550–12556. [Google Scholar]
- Chen, Z.; Zhang, Z.; Zhai, X.; Li, Y.; Lin, L.; Zhao, H.; Bian, L.; Li, P.; Yu, L.; Wu, Y.; et al. Rapid and Sensitive Detection of anti-SARS-CoV-2 IgG, Using Lanthanide-Doped Nanoparticles-Based Lateral Flow Immunoassay. Anal. Chem. 2020, 92, 7226–7231. [Google Scholar] [CrossRef] [PubMed]
- Zhao, Z.; Cui, H.; Song, W.; Ru, X.; Zhou, W.; Yu, X. A Simple Magnetic Nanoparticles-Based Viral Rna Extraction Method for Efficient Detection of SARS-CoV-2. bioRxiv 2020. [Google Scholar] [CrossRef]
- Roh, C.; Jo, S.K. Quantitative and sensitive detection of SARS coronavirus nucleocapsid protein using quantum dots-conjugated RNA aptamer on chip. J. Chem. Technol. Biotechnol. 2011, 86, 1475–1479. [Google Scholar] [CrossRef] [PubMed]
- Ali, M.A.; Hu, C.; Jahan, S.; Yuan, B.; Saleh, M.S.; Ju, E.; Gao, S.-J.; Panat, R. Sensing of COVID-19 Antibodies in Seconds Via Aerosol Jet Nanoprinted Reduced-Graphene-Oxide-Coated 3d Electrodes. Adv. Mater. 2021, 33, 2006647. [Google Scholar] [CrossRef]
- Vadlamani, B.; Uppal, S.T.; Verma, S.C.; Misra, M. Functionalized Tio2 Nanotube-Based Electrochemical Biosensor for Rapid Detection of SARS-CoV-2. Sensors 2020, 20, 5871. [Google Scholar] [CrossRef]
- Imani, S.M.; Ladouceur, L.; Marshall, T.; Maclachlan, R.; Soleymani, L.; Didar, T.F. Antimicrobial Nanomaterials and Coatings: Current Mechanisms and Future Perspectives to Control the Spread of Viruses Including SARS-CoV-2. ACS Nano 2020, 14, 12341–12369. [Google Scholar] [CrossRef]
- Lim, J.-W.; Ahn, Y.-R.; Park, G.; Kim, H.-O.; Haam, S. Application of Nanomaterials as an Advanced Strategy for the Diagnosis, Prevention, and Treatment of Viral Diseases. Pharmaceutics 2021, 13, 1570. [Google Scholar] [CrossRef]
- Abo-Zeid, Y.; Urbanowicz, R.A.; Thomson, B.J.; Irving, W.L.; Tarr, A.W.; Garnett, M.C. Enhanced Nanoparticle Uptake into Virus Infected Cells: Could Nanoparticles Be Useful in Antiviral Therapy? Int. J. Pharm. 2018, 547, 572–581. [Google Scholar] [CrossRef]
- Galdiero, S.; Falanga, A.; Vitiello, M.; Cantisani, M.; Marra, V.; Galdiero, M. Silver Nanoparticles as Potential Antiviral Agents. Molecules 2011, 16, 8894–8918. [Google Scholar] [CrossRef]
- Ferdous, Z.; Nemmar, A. Health Impact of Silver Nanoparticles: A Review of the Biodistribution and Toxicity Following Various Routes of Exposure. Int. J. Mol. Sci. 2020, 21, 2375. [Google Scholar] [CrossRef] [PubMed]
- Li, W.; Chen, X. Gold nanoparticles for photoacoustic imaging. Nanomedicine 2015, 10, 299–320. [Google Scholar] [CrossRef] [PubMed]
- Patel, U.S. Nanomaterials in Theranostics: Therapeutics and Diagnosis against Infectious Diseases. Ph.D. Thesis, The University of Alabama in Huntsville, Ann Arbor, MI, USA, 2022. [Google Scholar]
- Patel, U.; Rathnayake, K.; Jani, H.; Jayawardana, K.W.; Dhakal, R.; Duan, L.; Jayawardena, S.N. Near-infrared responsive targeted drug delivery system that offer chemo-photothermal therapy against bacterial infection. Nano Sel. 2021, 2, 1750–1769. [Google Scholar] [CrossRef]
- Du, T.; Zhang, J.; Li, C.; Song, T.; Li, P.; Liu, J.; Du, X.; Wang, S. Gold/Silver Hybrid Nanoparticles with Enduring Inhibition of Coronavirus Multiplication through Multisite Mechanisms. Bioconjugate Chem. 2020, 31, 2553–2563. [Google Scholar] [CrossRef] [PubMed]
- Bobo, D.; Robinson, K.J.; Islam, J.; Thurecht, K.J.; Corrie, S.R. Nanoparticle-Based Medicines: A Review of Fda-Approved Materials and Clinical Trials to Date. Pharm. Res. 2016, 33, 2373–2387. [Google Scholar] [CrossRef]
- Elahi, S. Hematopoietic Responses to SARS-CoV-2 Infection. Cell. Mol. Life Sci. 2022, 79, 187. [Google Scholar] [CrossRef]
- Kronstein-Wiedemann, R.; Stadtmüller, M.; Traikov, S.; Georgi, M.; Teichert, M.; Yosef, H.; Wallenborn, J.; Karl, A.; Schütze, K.; Wagner, M.; et al. SARS-CoV-2 Infects Red Blood Cell Progenitors and Dysregulates Hemoglobin and Iron Metabolism. Stem Cell Rev. Rep. 2022, 18, 1809–1821. [Google Scholar] [CrossRef]
- Koksharov, Y.A.; Gubin, S.P.; Taranov, I.V.; Khomutov, G.B.; Gulyaev, Y.V. Magnetic Nanoparticles in Medicine: Progress, Problems, and Advances. J. Commun. Technol. Electron. 2022, 67, 101–116. [Google Scholar] [CrossRef]
- Cardoso, V.M.d.O.; Moreira, B.J.; Comparetti, E.J.; Sampaio, I.; Ferreira, L.M.B.; Lins, P.M.P.; Zucolotto, V. Is Nanotechnology Helping in the Fight against COVID-19? Front. Nanotechnol. 2020, 2, 588915. [Google Scholar] [CrossRef]
- Rathnayake, K.; Patel, U.; Pham, C.; McAlpin, A.; Budisalich, T.; Jayawardena, S.N. Targeted Delivery of Antibiotic Therapy to Inhibit Pseudomonas aeruginosa Using Lipid-Coated Mesoporous Silica Core–Shell Nanoassembly. ACS Appl. Bio Mater. 2020, 3, 6708–6721. [Google Scholar] [CrossRef]
- James, N.D.; Coker, R.J.; Tomlinson, D.; Harris, J.R.W.; Gompels, M.; Pinching, A.J.; Stewart, J.S.W. Liposomal Doxorubicin (Doxil): An Effective New Treatment for Kaposi’s Sarcoma in Aids. Clin. Oncol. 1994, 6, 294–296. [Google Scholar] [CrossRef]
- Anselmo, A.C.; Mitragotri, S. Nanoparticles in the clinic: An update. Bioeng. Transl. Med. 2019, 4, e10143. [Google Scholar] [CrossRef] [PubMed]
- Daraee, H.; Etemadi, A.; Kouhi, M.; Alimirzalu, S.; Akbarzadeh, A. Application of liposomes in medicine and drug delivery. Artif. Cells Nanomed. Biotechnol. 2016, 44, 381–391. [Google Scholar] [CrossRef] [PubMed]
- Satta, S.; Meng, Z.; Hernandez, R.; Cavallero, S.; Zhou, T.; Hsiai, T.K.; Zhou, C. An Engineered Nano-Liposome-Human Ace2 Decoy Neutralizes SARS-CoV-2 Spike Protein-Induced Inflammation in Both Murine and Human Macrophages. Theranostics 2022, 12, 2639–2657. [Google Scholar] [CrossRef] [PubMed]
- Lima, A.C.; Ferreira, H.; Reis, R.L.; Neves, N.M. Biodegradable polymers: An update on drug delivery in bone and cartilage diseases. Expert Opin. Drug Deliv. 2019, 16, 795–813. [Google Scholar] [CrossRef] [PubMed]
- Sánchez, A.; Mejía, S.P.; Orozco, J. Recent Advances in Polymeric Nanoparticle-Encapsulated Drugs against Intracellular Infections. Molecules 2020, 25, 3760. [Google Scholar] [CrossRef]
- Begines, B.; Ortiz, T.; Pérez-Aranda, M.; Martínez, G.; Merinero, M.; Argüelles-Arias, F.; Alcudia, A. Polymeric Nanoparticles for Drug Delivery: Recent Developments and Future Prospects. Nanomaterials 2020, 10, 1403. [Google Scholar] [CrossRef]
- Wei, M.; Gao, Y.; Li, X.; Serpe, M.J. Stimuli-responsive polymers and their applications. Polym. Chem. 2017, 8, 127–143. [Google Scholar] [CrossRef]
- Charelli, L.E.; de Mattos, G.C.; Sousa-Batista, A.D.J.; Pinto, J.C.; Balbino, T.A. Polymeric nanoparticles as therapeutic agents against coronavirus disease. J. Nanoparticle Res. 2022, 24, 12. [Google Scholar] [CrossRef]
- Piao, H.; Rejinold, N.S.; Choi, G.; Pei, Y.-R.; Jin, G.-W.; Choy, J.-H. Niclosamide encapsulated in Mesoporous silica and geopolymer: A potential oral formulation for COVID-19. Microporous Mesoporous Mater. 2021, 326, 111394. [Google Scholar] [CrossRef]
- Abo-Zeid, Y.; Ismail, N.S.M.; McLean, G.R.; Hamdy, N.M. A Molecular Docking Study Repurposes Fda Approved Iron Oxide Nanoparticles to Treat and Control Covid-19 Infection. Eur. J. Pharm. Sci. 2020, 153, 105465. [Google Scholar] [CrossRef] [PubMed]
- Abd Elkodous, M.; Olojede, S.O.; Morsi, M.; El-Sayyad, G.S. Nanomaterial-Based Drug Delivery Systems as Promising Carriers for Patients with Covid-19. RSC Adv 2021, 11, 26463–26480. [Google Scholar] [CrossRef] [PubMed]
- Tenchov, R.; Bird, R.; Curtze, A.E.; Zhou, Q. Lipid Nanoparticles─from Liposomes to Mrna Vaccine Delivery, a Landscape of Research Diversity and Advancement. ACS Nano 2021, 15, 16982–17015. [Google Scholar] [CrossRef] [PubMed]
- Loutfy, S.A.; Abdel-Salam, A.I.; Moatasim, Y.; Gomaa, M.R.; Fattah, N.F.A.; Emam, M.H.; Ali, F.; ElShehaby, H.A.; Ragab, E.A.; Alam El-Din, H.M. Antiviral Activity of Chitosan Nanoparticles Encapsulating Silymarin (Sil–Cnps) against SARS-CoV-2 (in Silico and in Vitro Study). RSC Adv. 2022, 12, 15775–15786. [Google Scholar] [CrossRef] [PubMed]
- Almanza-Reyes, H.; Moreno, S.; Plascencia-López, I.; Alvarado-Vera, M.; Patrón-Romero, L.; Borrego, B.; Reyes-Escamilla, A.; Valencia-Manzo, D.; Brun, A.; Pestryakov, A. Evaluation of Silver Nanoparticles for the Prevention of SARS-CoV-2 Infection in Health Workers: In Vitro and in Vivo. PLOS ONE 2021, 16, e0256401. [Google Scholar] [CrossRef] [PubMed]
- Fenner, F.; Henderson, D.A.; Arita, I.; Jezek, Z.; Ladnyi, I.D. Smallpox and Its Eradication; World Health Organization: Geneva, Switzerland, 1988; Volume 6. [Google Scholar]
- Slütter, B.; Bal, S.; Keijzer, C.; Mallants, R.; Hagenaars, N.; Que, I.; Kaijzel, E.; van Eden, W.; Augustijns, P.; Löwik, C.; et al. Nasal vaccination with N-trimethyl chitosan and PLGA based nanoparticles: Nanoparticle characteristics determine quality and strength of the antibody response in mice against the encapsulated antigen. Vaccine 2010, 28, 6282–6291. [Google Scholar] [CrossRef]
- Li, A.V.; Moon, J.J.; Abraham, W.; Suh, H.; Elkhader, J.; Seidman, M.A.; Yen, M.; Im, E.-J.; Foley, M.H.; Barouch, D.H.; et al. Generation of Effector Memory T Cell–Based Mucosal and Systemic Immunity with Pulmonary Nanoparticle Vaccination. Sci. Transl. Med. 2013, 5, 204ra130. [Google Scholar] [CrossRef]
- Lin, L.C.-W.; Chattopadhyay, S.; Lin, J.-C.; Hu, C.-M.J. Advances and Opportunities in Nanoparticle-and Nanomaterial-Based Vaccines against Bacterial Infections. Adv. Healthc. Mater. 2018, 7, 1701395. [Google Scholar] [CrossRef]
- Park, Y.-M.; Lee, S.J.; Kim, Y.S.; Lee, M.H.; Cha, G.S.; Jung, I.D.; Kang, T.H.; Han, H.D. Nanoparticle-Based Vaccine Delivery for Cancer Immunotherapy. Immune Netw. 2013, 13, 177–183. [Google Scholar] [CrossRef]
- Ruiz-Hitzky, E.; Darder, M.; Aranda, P.; del Burgo, M.M.; del Real, G. Bionanocomposites as New Carriers for Influenza Vaccines. Adv. Mater. 2009, 21, 4167–4171. [Google Scholar] [CrossRef]
- Nel, A.E.; Mädler, L.; Velegol, D.; Xia, T.; Hoek, E.M.V.; Somasundaran, P.; Klaessig, F.; Castranova, V.; Thompson, M. Understanding biophysicochemical interactions at the nano–bio interface. Nat. Mater. 2009, 8, 543–557. [Google Scholar] [CrossRef] [PubMed]
- Dykman, L.A.; Sumaroka, M.V.; Staroverov, S.A.; Zaitseva, I.S.; Bogatyrev, V.A. Immunogenic Properties of Colloidal Gold. Biol. Bull. Russ. Acad. Sci. 2004, 31, 75–79. [Google Scholar] [CrossRef]
- Salvador-Morales, C.; Flahaut, E.; Sim, E.; Sloan, J.; Green, M.L.H.; Sim, R. Complement activation and protein adsorption by carbon nanotubes. Mol. Immunol. 2006, 43, 193–201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mahony, D.; Cavallaro, A.S.; Stahr, F.; Mahony, T.J.; Qiao, S.Z.; Mitter, N. Mesoporous silica Nanoparticles Act as a Self-Adjuvant for Ovalbumin Model Antigen in Mice. Small 2013, 9, 3138–3146. [Google Scholar] [CrossRef] [PubMed]
- Mody, K.T.; Popat, A.; Mahony, D.; Cavallaro, A.S.; Yu, C.; Mitter, N. Mesoporous silica nanoparticles as antigen carriers and adjuvants for vaccine delivery. Nanoscale 2013, 5, 5167–5179. [Google Scholar] [CrossRef] [PubMed]
- Castaldello, A.; Brocca-Cofano, E.; Voltan, R.; Triulzi, C.; Altavilla, G.; Laus, M.; Sparnacci, K.; Ballestri, M.; Tondelli, L.; Fortini, C.; et al. DNA prime and protein boost immunization with innovative polymeric cationic core-shell nanoparticles elicits broad immune responses and strongly enhance cellular responses of HIV-1 tat DNA vaccination. Vaccine 2006, 24, 5655–5669. [Google Scholar] [CrossRef] [PubMed]
- Nagamoto, T.; Hattori, Y.; Takayama, K.; Maitani, Y. Novel Chitosan Particles and Chitosan-Coated Emulsions Inducing Immune Response via Intranasal Vaccine Delivery. Pharm. Res. 2004, 21, 671–674. [Google Scholar] [CrossRef] [PubMed]
- Monopoli, M.P.; Åberg, C.; Salvati, A.; Dawson, K.A. Biomolecular Coronas Provide the Biological Identity of Nanosized Materials. Nat. Nanotechnol. 2012, 7, 779–786. [Google Scholar] [CrossRef] [PubMed]
- Sivaraman, B.; Fears, K.P.; Latour, R.A. Investigation of the Effects of Surface Chemistry and Solution Concentration on the Conformation of Adsorbed Proteins Using an Improved Circular Dichroism Method. Langmuir 2009, 25, 3050–3056. [Google Scholar] [CrossRef] [PubMed]
- Salazar-González, J.A.; González-Ortega, O.; Rosales-Mendoza, S. Gold Nanoparticles and Vaccine Development. Expert Rev. Vaccines 2015, 14, 1197–1211. [Google Scholar] [CrossRef] [PubMed]
- Jin, R.; Wu, G.; Li, Z.; Mirkin, C.A.; Schatz, G.C. What Controls the Melting Properties of DNA-Linked Gold Nanoparticle Assemblies? J. Am. Chem. Soc. 2003, 125, 1643–1654. [Google Scholar] [CrossRef] [PubMed]
- Rosi, N.L.; Giljohann, D.A.; Thaxton, C.S.; Lytton-Jean, A.K.R.; Han, M.S.; Mirkin, C.A. Oligonucleotide-Modified Gold Nanoparticles for Intracellular Gene Regulation. Science 2006, 312, 1027–1030. [Google Scholar] [CrossRef] [PubMed]
- Mirkin, C.A.; Letsinger, R.L.; Mucic, R.C.; Storhoff, J.J. A DNA-Based Method for Rationally Assembling Nanoparticles into Macroscopic Materials. Nature 1996, 382, 607–609. [Google Scholar] [CrossRef] [PubMed]
- Totaro, K.A.; Liao, X.; Bhattacharya, K.; Finneman, J.I.; Sperry, J.B.; Massa, M.A.; Thorn, J.; Ho, S.V.; Pentelute, B.L. Systematic Investigation of Edc/Snhs-Mediated Bioconjugation Reactions for Carboxylated Peptide Substrates. Bioconjugate Chem. 2016, 27, 994–1004. [Google Scholar] [CrossRef]
- Handké, N.; Ficheux, D.; Rollet, M.; Delair, T.; Mabrouk, K.; Bertin, D.; Gigmes, D.; Verrier, B.; Trimaille, T. Lysine-tagged peptide coupling onto polylactide nanoparticles coated with activated ester-based amphiphilic copolymer: A route to highly peptide-functionalized biodegradable carriers. Colloids Surf. B Biointerfaces 2013, 103, 298–303. [Google Scholar] [CrossRef] [PubMed]
- Heuking, S.; Iannitelli, A.; Di Stefano, A.; Borchard, G. Toll-like receptor-2 agonist functionalized biopolymer for mucosal vaccination. Int. J. Pharm. 2009, 381, 97–105. [Google Scholar] [CrossRef]
- Chen, W.; Huang, L. Induction of Cytotoxic T-Lymphocytes and Antitumor Activity by a Liposomal Lipopeptide Vaccine. Mol. Pharm. 2008, 5, 464–471. [Google Scholar] [CrossRef]
- Rossman, J.S.; Leser, G.P.; Lamb, R.A. Filamentous Influenza Virus Enters Cells via Macropinocytosis. J. Virol. 2012, 86, 10950–10960. [Google Scholar] [CrossRef]
- Galloway, A.L.; Murphy, A.; DeSimone, J.M.; Di, J.; Herrmann, J.P.; Hunter, M.E.; Kindig, J.P.; Malinoski, F.J.; Rumley, M.A.; Stoltz, D.M.; et al. Development of a nanoparticle-based influenza vaccine using the PRINT® technology. Nanomed. Nanotechnol. Biol. Med. 2013, 9, 523–531. [Google Scholar] [CrossRef] [PubMed]
- Macauley, M.; Pfrengle, F.; Rademacher, C.; Nycholat, C.M.; Gale, A.J.; Von Drygalski, A.; Paulson, J.C. Antigenic liposomes displaying CD22 ligands induce antigen-specific B cell apoptosis. J. Clin. Investig. 2013, 123, 3074–3083. [Google Scholar] [CrossRef] [PubMed]
- Capini, C.; Jaturanpinyo, M.; Chang, H.-I.; Mutalik, S.; McNally, A.; Street, S.; Steptoe, R.; O’Sullivan, B.; Davies, N.; Thomas, R. Antigen-Specific Suppression of Inflammatory Arthritis Using Liposomes. J. Immunol. 2009, 182, 3556–3565. [Google Scholar] [CrossRef] [PubMed]
- Kishimoto, T.K.; Ferrari, J.D.; Lamothe, R.A.; Kolte, P.N.; Griset, A.P.; O’Neil, C.; Chan, V.; Browning, E.; Chalishazar, A.; Kuhlman, W.; et al. Improving the efficacy and safety of biologic drugs with tolerogenic nanoparticles. Nat. Nanotechnol. 2016, 11, 890–899. [Google Scholar] [CrossRef]
- Maldonado, R.A.; LaMothe, R.A.; Ferrari, J.D.; Zhang, A.-H.; Rossi, R.J.; Kolte, P.N.; Griset, A.P.; O’Neil, C.; Altreuter, D.H.; Browning, E.; et al. Polymeric synthetic nanoparticles for the induction of antigen-specific immunological tolerance. Proc. Natl. Acad. Sci. USA 2015, 112, E156–E165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sands, E.; Kivitz, A.; Johnston, L.; Kishimoto, T. THU0422 SEL-212: Enhanced serum uric acid control in hyperuricemic patients through selective mitigation of anti-drug antibodies against pegsiticase. Ann. Rheum. Dis. 2017, 76, 367. [Google Scholar] [CrossRef]
- Yeste, A.; Takenaka, M.C.; Mascanfroni, I.D.; Nadeau, M.; Kenison, J.E.; Patel, B.; Tukpah, A.-M.; Babon, J.A.B.; DeNicola, M.; Kent, S.C.; et al. Tolerogenic nanoparticles inhibit T cell–mediated autoimmunity through SOCS2. Sci. Signal. 2016, 9, ra61. [Google Scholar] [CrossRef] [PubMed]
- Yeste, A.; Nadeau, M.; Burns, E.J.; Weiner, H.L.; Quintana, F.J. Nanoparticle-mediated codelivery of myelin antigen and a tolerogenic small molecule suppresses experimental autoimmune encephalomyelitis. Proc. Natl. Acad. Sci. USA 2012, 109, 11270–11275. [Google Scholar] [CrossRef] [PubMed]
- Suk, J.S.; Xu, Q.; Kim, N.; Hanes, J.; Ensign, L.M. PEGylation as a strategy for improving nanoparticle-based drug and gene delivery. Adv. Drug Deliv. Rev. 2016, 99 Pt A, 28–51. [Google Scholar] [CrossRef]
- Reed, S.G.; Orr, M.T.; Fox, C.B. Key roles of adjuvants in modern vaccines. Nat. Med. 2013, 19, 1597–1608. [Google Scholar] [CrossRef] [PubMed]
- Mbow, M.L.; De Gregorio, E.; Ulmer, J.B. Alum’s Adjuvant Action: Grease Is the Word. Nat. Med. 2011, 17, 415–416. [Google Scholar] [CrossRef] [PubMed]
- Weeratna, R.D.; Makinen, S.R.; McCluskie, M.J.; Davis, H.L. TLR agonists as vaccine adjuvants: Comparison of CpG ODN and Resiquimod (R-848). Vaccine 2005, 23, 5263–5270. [Google Scholar] [CrossRef] [PubMed]
- Novavax Announces Positive Phase 3 Data for Its COVID-19 Vaccine Candidate. Available online: https://ir.novavax.com/news-releases/news-release-details/novavax-announces-positive-phase-1-data-its-covid-19-vaccine (accessed on 25 August 2022).
- Hayashi, M.; Aoshi, T.; Haseda, Y.; Kobiyama, K.; Wijaya, E.; Nakatsu, N.; Igarashi, Y.; Standley, D.M.; Yamada, H.; Honda-Okubo, Y.; et al. Advax, a Delta Inulin Microparticle, Potentiates In-built Adjuvant Property of Co-administered Vaccines. eBioMedicine 2016, 15, 127–136. [Google Scholar] [CrossRef] [PubMed]
- Dalpke, A.H.; Helm, M. RNA mediated toll-like receptor stimulation in health and disease. RNA Biol. 2012, 9, 828–842. [Google Scholar] [CrossRef] [PubMed]
- Perrie, Y.; Crofts, F.; Devitt, A.; Griffiths, H.R.; Kastner, E.; Nadella, V. Designing liposomal adjuvants for the next generation of vaccines. Adv. Drug Deliv. Rev. 2016, 99 Pt A, 85–96. [Google Scholar] [CrossRef] [Green Version]
- Wang, Z.-B.; Jing, X. Better Adjuvants for Better Vaccines: Progress in Adjuvant Delivery Systems, Modifications, and Adjuvant-Antigen Codelivery. Vaccines 2020, 8, 128. [Google Scholar] [CrossRef] [PubMed]
- Kreutz, M.; Giquel, B.; Hu, Q.; Abuknesha, R.; Uematsu, S.; Akira, S.; Nestle, F.O.; Diebold, S.S. Antibody-Antigen-Adjuvant Conjugates Enable Co-Delivery of Antigen and Adjuvant to Dendritic Cells in Cis but Only Have Partial Targeting Specificity. PLoS ONE 2012, 7, e40208. [Google Scholar] [CrossRef] [PubMed]
- WHO. COVID-19 Vaccine Tracker and Landscape. Available online: https://www.who.int/publications/m/item/draft-landscape-of-covid-19-candidate-vaccines (accessed on 26 August 2022).
- Wrapp, D.; Wang, N.; Corbett, K.S.; Goldsmith, J.A.; Hsieh, C.-L.; Abiona, O.; Graham, B.S.; McLellan, J.S. Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science 2020, 367, 1260–1263. [Google Scholar] [CrossRef]
- Benvenuto, D.; Giovannetti, M.; Ciccozzi, A.; Spoto, S.; Angeletti, S.; Ciccozzi, M. The 2019-New Coronavirus Epidemic: Evidence for Virus Evolution. J. Med. Virol. 2020, 92, 455–459. [Google Scholar] [CrossRef] [PubMed]
- Yuan, M.; Wu, N.C.; Zhu, X.; Lee, C.-C.D.; So, R.T.Y.; Lv, H.; Mok, C.K.P.; Wilson, I.A. A Highly Conserved Cryptic Epitope in the Receptor Binding Domains of SARS-CoV-2 and SARS-CoV. Science 2020, 368, 630–633. [Google Scholar] [CrossRef] [PubMed]
- Clinicaltrials.gov. Clinical Trial of Efficacy and Safety of Sinovac’s Adsorbed Covid-19 (Inactivated) Vaccine in Healthcare Professionals (Profiscov). Available online: https://clinicaltrials.gov/ct2/show/NCT04456595 (accessed on 26 August 2022).
- Xia, S.; Duan, K.; Zhang, Y.; Zhao, D.; Zhang, H.; Xie, Z.; Li, X.; Peng, C.; Zhang, Y.; Zhang, W.; et al. Effect of an Inactivated Vaccine against SARS-CoV-2 on Safety and Immunogenicity Outcomes: Interim Analysis of 2 Randomized Clinical Trials. Jama 2020, 324, 951–960. [Google Scholar] [CrossRef] [PubMed]
- Clinicaltrials.gov. An Efficacy and Safety Clinical Trial of an Investigational COVID-19 Vaccine (Bbv152) in Adult Volunteers. Available online: https://clinicaltrials.gov/ct2/show/NCT04641481 (accessed on 26 August 2022).
- Zhou, B.; Meliopoulos, V.A.; Wang, W.; Lin, X.; Stucker, K.M.; Halpin, R.A.; Stockwell, T.B.; Schultz-Cherry, S.; Wentworth, D.E. Reversion of Cold-Adapted Live Attenuated Influenza Vaccine into a Pathogenic Virus. J. Virol. 2016, 90, 8454–8463. [Google Scholar] [CrossRef] [PubMed]
- Prnewswire.com. Codagenix and Serum Institute of India Announce Commencement of First-in-Human Trial of Covi-Vac, a Single Dose, Intranasal Live Attenuated Vaccine for COVID-19. Available online: https://www.prnewswire.com/news-releases/codagenix-and-serum-institute-of-india-announce-commencement-of-first-in-human-trial-of-covi-vac-a-single-dose-intranasal-live-attenuated-vaccine-for-covid-19-301191756.html (accessed on 26 August 2022).
- Sanchez-Felipe, L.; Vercruysse, T.; Sharma, S.; Ma, J.; Lemmens, V.; Van Looveren, D.; Javarappa, M.P.A.; Boudewijns, R.; Malengier-Devlies, B.; Liesenborghs, L.; et al. A Single-Dose Live-Attenuated Yf17d-Vectored SARS-CoV-2 Vaccine Candidate. Nature 2020, 590, 320–325. [Google Scholar] [CrossRef] [PubMed]
- Herzog, C.; Hartmann, K.; Künzi, V.; Kürsteiner, O.; Mischler, R.; Lazar, H.; Glück, R. Eleven years of Inflexal® V—A virosomal adjuvanted influenza vaccine. Vaccine 2009, 27, 4381–4387. [Google Scholar] [CrossRef] [PubMed]
- Altmann, D.M.; Boyton, R.J. COVID-19 vaccination: The road ahead. Science 2022, 375, 1127–1132. [Google Scholar] [CrossRef] [PubMed]
- Henao-Restrepo, A.M.; Camacho, A.; Longini, I.M.; Watson, C.H.; Edmunds, W.J.; Egger, M.; Carroll, M.W.; Dean, N.E.; Diatta, I.; Doumbia, M.; et al. Efficacy and Effectiveness of an Rvsv-Vectored Vaccine in Preventing Ebola Virus Disease: Final Results from the Guinea Ring Vaccination, Open-Label, Cluster-Randomised Trial (Ebola Ça Suffit!). Lancet 2017, 389, 505–518. [Google Scholar] [CrossRef]
- Lázaro-Frías, A.; Gómez-Medina, S.; Sánchez-Sampedro, L.; Ljungberg, K.; Ustav, M.; Liljeström, P.; Muñoz-Fontela, C.; Esteban, M.; García-Arriaza, J. Distinct Immunogenicity and Efficacy of Poxvirus-Based Vaccine Candidates against Ebola Virus Expressing GP and VP40 Proteins. J. Virol. 2018, 92, e00363-18. [Google Scholar] [CrossRef] [PubMed]
- Koch, T.; Dahlke, C.; Fathi, A.; Kupke, A.; Krähling, V.; Okba, N.M.A.; Halwe, S.; Rohde, C.; Eickmann, M.; Volz, A.; et al. Safety and immunogenicity of a modified vaccinia virus Ankara vector vaccine candidate for Middle East respiratory syndrome: An open-label, phase 1 trial. Lancet Infect. Dis. 2020, 20, 827–838. [Google Scholar] [CrossRef]
- Zhu, F.-C.; Li, Y.-H.; Guan, X.-H.; Hou, L.-H.; Wang, W.-J.; Li, J.-X.; Wu, S.-P.; Wang, B.-S.; Wang, Z.; Wang, L.; et al. Safety, tolerability, and immunogenicity of a recombinant adenovirus type-5 vectored COVID-19 vaccine: A dose-escalation, open-label, non-randomised, first-in-human trial. Lancet 2020, 395, 1845–1854. [Google Scholar] [CrossRef]
- Alberer, M.; Gnad-Vogt, U.; Hong, H.S.; Mehr, K.T.; Backert, L.; Finak, G.; Gottardo, R.; Bica, M.A.; Garofano, A.; Koch, S.D.; et al. Safety and Immunogenicity of a Mrna Rabies Vaccine in Healthy Adults: An Open-Label, Non-Randomised, Prospective, First-in-Human Phase 1 Clinical Trial. Lancet 2017, 390, 1511–1520. [Google Scholar] [CrossRef]
- Zeng, C.; Hou, X.; Yan, J.; Zhang, C.; Li, W.; Zhao, W.; Du, S.; Dong, Y. Leveraging Mrnas Sequences to Express SARS-CoV-2 Antigens in Vivo. bioRxiv 2020. [Google Scholar] [CrossRef]
- Pardi, N.; Hogan, M.J.; Porter, F.W.; Weissman, D. Mrna Vaccines—A New Era in Vaccinology. Nat. Rev. Drug Discov. 2018, 17, 261–279. [Google Scholar] [CrossRef]
- FDA News Release: FDA Takes Key Action in Fight against COVID-19 By Issuing Emergency Use Authorization for First COVID-19 Vaccine. Available online: https://www.fda.gov/news-events/press-announcements/fda-takes-key-action-fight-against-covid-19-issuing-emergency-use-authorization-first-covid-19 (accessed on 26 August 2022).
- Clinicaltrials.gov. Dose-Confirmation Study to Evaluate the Safety, Reactogenicity, and Immunogenicity of Mrna-1273 COVID-19 Vaccine in Adults Aged 18 Years and Older. Available online: https://clinicaltrials.gov/ct2/show/NCT04405076 (accessed on 26 August 2022).
- Kowalski, P.S.; Rudra, A.; Miao, L.; Anderson, D.G. Delivering the Messenger: Advances in Technologies for Therapeutic Mrna Delivery. Mol. Ther. 2019, 27, 710–728. [Google Scholar] [CrossRef]
- Clinicaltrials.gov. A Trial Investigating the Safety and Effects of Four Bnt162 Vaccines against Covid-2019 in Healthy Adults. Available online: https://clinicaltrials.gov/ct2/show/NCT04380701 (accessed on 26 August 2022).
- Study to Describe the Safety, Tolerability, Immunogenicity, and Efficacy of Rna Vaccine Candidates against COVID-19 in Healthy Individuals. Available online: https://clinicaltrials.gov/ct2/show/NCT04368728 (accessed on 26 August 2022).
- Karikó, K.; Muramatsu, H.; Welsh, F.A.; Ludwig, J.; Kato, H.; Akira, S.; Weissman, D. Incorporation of Pseudouridine Into mRNA Yields Superior Nonimmunogenic Vector with Increased Translational Capacity and Biological Stability. Mol. Ther. 2008, 16, 1833–1840. [Google Scholar] [CrossRef] [PubMed]
- Technologyreview.com. What Are the Ingredients of Pfizer’s COVID-19 Vaccine? Available online: https://www.technologyreview.com/2020/12/09/1013538/what-are-the-ingredients-of-pfizers-covid-19-vaccine/ (accessed on 26 August 2022).
- Cross, R. Without These Lipid Shells, There Would Be No Mrna Vaccines for COVID-19. Chem. Eng. News Drug Deliv. 2021, 99, 8. [Google Scholar]
- Li, W.; Joshi, M.D.; Singhania, S.; Ramsey, K.H.; Murthy, A.K. Peptide Vaccine: Progress and Challenges. Vaccines 2014, 2, 515–536. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mohsen, M.O.; Zha, L.; Cabral-Miranda, G.; Bachmann, M.F. Major findings and recent advances in virus–like particle (VLP)-based vaccines. Semin. Immunol. 2017, 34, 123–132. [Google Scholar] [CrossRef]
- Bezu, L.; Kepp, O.; Cerrato, G.; Pol, J.; Fucikova, J.; Spisek, R.; Zitvogel, L.; Kroemer, G.; Galluzzi, L. Trial watch: Peptide-based vaccines in anticancer therapy. OncoImmunology 2018, 7, e1511506. [Google Scholar] [CrossRef] [PubMed]
- Heitmann, J.S.; Bilich, T.; Tandler, C.; Nelde, A.; Maringer, Y.; Marconato, M.; Reusch, J.; Jäger, S.; Denk, M.; Richter, M.; et al. A COVID-19 Peptide Vaccine for the Induction of SARS-CoV-2 T Cell Immunity. Nature 2022, 601, 617–622. [Google Scholar] [CrossRef]
Nanoparticles | Conjugates | Detection Technique | Target | Limit of Detection | Ref. |
---|---|---|---|---|---|
AuNP | Fluorine-doped tin oxide electrode (FTO) and nCOVID- monoclonal antibody | Immunosensor | SARS-CoV-2 virus | 90 fM | [102] |
IgM antibody | Lateral flow assay | SARS-CoV-2 virus | N/A | [103] | |
Lanthanide doped polystyrene NPs | Nucleocapsid phosphoprotein of SARS-CoV-2 | Lateral flow immunoassay (LFIA) | IgG in serum | N/A | [104] |
Magnetic NPs (MNPs) | poly (amino ester) with carboxyl groups (PC) | RT-PCR | Viral RNA | 10 copies | [105] |
Quantum dots (QDs) | RNA aptamer | Fluorescent sensor | SARS-CoV N protein | 0.1 pg mL−1 | [106] |
Reduced graphene oxide | Viral antigen | Electrochemical Biosensor | SARS-CoV-2 spike S1 protein | 1 × 10−12 M 1 × 10−15 M | [107] |
Titanium Dioxide nanotubes | Spike Receptor Binding Domain (RBD) | Electrochemical sensor | SARS-CoV-2 | 0.7 Nm | [108] |
Nanoparticles (NPs) | Conjugates | Virus | Mechanism of Action | Study Type | Key Conclusion for COVID-19 Treatment | Ref. |
---|---|---|---|---|---|---|
Mesopore silica nanoparticles | Niclosamide | SARS-CoV2 | Interference with spike protein of virus, hence hinder their adhesion to human cell receptor | In vitro In vivo | Inhibit SARS-CoV-2 Infectivity by targeting and therapeutic effect from Niclosamide | [133] |
Iron oxide NPs | - | SARS-CoV2 | Interference with the virus binding and entry to the host cells. | Molecular Docking | Models were effective to be bonded with viral proteins by causing conformational changes and viral inactivation | [134] |
Nano emulsion | Favipiravir | SARS-CoV2 | Interfere with the virion envelope composition or mask the virus (SARS-CoV-2) building | - | Hinders virus replication through inhibiting the SARS-COV-2 RNA replication | [135] |
Lipid NPs | m-RNA 1273 | SARS-CoV-2 | Viral inactivation | In vitro In vivo | Significant reduction of viral load. | [136] |
Silymarin–chitosan nanoparticles | Silymarin | SARS-CoV2 and ADV-5 | Blocking viral host receptor ACE2 | In vitro | Preventing viral attachment and its entry to the cells | [137] |
Silver NPs | - | SARS-CoV2 | Interfere with the structural proteins of the virus by inhibiting their ability to bind with cell receptors | In vitro In vivo | Inhibitory effect on SARS-CoV-2 infectivity | [138] |
Vaccine Designer | Vaccine Platform | Vaccine Type | ROA | Clinical Trials Registry |
---|---|---|---|---|
Sinovac Research and Development Co., Ltd., Beijing, China | Formalin inactivating whole virus particles combined with an alum adjuvant | SARS-CoV-2 vaccine (inactivated) | IM | NCT04456595 |
Study Report | ||||
NCT04508075 | ||||
NCT04582344 | ||||
NCT04617483 | ||||
Sinopharm + Wuhan Institute of Biological Products, Beijing, China | Inactivated SARS-CoV-2 vaccine (Vero cell) | Inactivated virus | IM | ChiCTR2000034780 |
ChiCTR2000039000 | ||||
NCT04510207 | ||||
NCT04612972 | ||||
Sinopharm + Beijing Institute of Biological Products, Beijing, China | Inactivated SARS-CoV-2 vaccine (Vero cell) | Inactivated virus | IM | ChiCTR2000034780 |
NCT04560881 | ||||
NCT04510207 | ||||
Bharat Biotech International Limited, Hyderabad, India | Whole-Virion Inactivated SARS-CoV-2 Vaccine (BBV152) | Inactivated virus | IM | NCT04641481 |
CTRI/2020/11/028976 | ||||
AstraZeneca + University of Oxford, Cambridge, Oxford, England | ChAdOx1-S—(AZD1222) (Covishield) | Viral vector (non-replicating) | IM | ISRCTN89951424 |
NCT04516746 | ||||
NCT04540393 | ||||
NCT04536051 | ||||
CanSino Biological Inc./Beijing Institute of Biotechnology, Beijing, China | Recombinant novel coronavirus vaccine (Adenovirus type 5 vector) | Viral vector (non-replicating) | IM | NCT04526990 |
NCT04540419 | ||||
Gamaleya Research Institute; Health Ministry of the Russian Federation, Moscow, Russia | Gam-COVID-Vac Adeno-based (rAd26-S+rAd5-S) | Viral vector (non-replicating) | IM | NCT04530396 |
NCT04564716 | ||||
NCT04642339 | ||||
Janssen Pharmaceuticals, Beerse, Belgium | Ad26.COV2.S | Viral vector (non-replicating) | IM | NCT04505722 |
NCT04614948 | ||||
Novavax, Gaithersburg, MD, USA | SARS-CoV-2 Rs/Matrix M1-Adjuvant (Full length recombinant SARS CoV-2 glycoprotein nanoparticle vaccine adjuvanted with Matrix M) | Protein subunit | IM | NCT04611802 |
EUCTR2020-004123-16-GB | ||||
NCT04583995 | ||||
Anhui Zhifei Longcom Biopharmaceutical + Institute of Microbiology, Chinese Academy of Sciences, Anhui, China | Recombinant SARS-CoV-2 vaccine (CHO Cell) | Protein subunit | IM | ChiCTR2000040153 |
NCT04646590 | ||||
Moderna + National Institute of Allergy and Infectious Diseases (NIAID), Massachusetts, USA | Lipid nanoparticle mRNA vaccine (mRNA-1273) | mRNA vaccine | IM | NCT04470427 |
BioNTech + Fosun Pharma; Jiangsu Provincial Center for Disease Prevention and Control + Pfizer, Mainz, Germany, Shanghai, China, Newyork, USA | Lipid nanoparticle mRNA vaccine (BNT162b2) | mRNA vaccine | IM | NCT04368728 |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Patel, U.; Rathnayake, K.; Hunt, E.C.; Singh, N. Role of Nanomaterials in COVID-19 Prevention, Diagnostics, Therapeutics, and Vaccine Development. J. Nanotheranostics 2022, 3, 151-176. https://doi.org/10.3390/jnt3040011
Patel U, Rathnayake K, Hunt EC, Singh N. Role of Nanomaterials in COVID-19 Prevention, Diagnostics, Therapeutics, and Vaccine Development. Journal of Nanotheranostics. 2022; 3(4):151-176. https://doi.org/10.3390/jnt3040011
Chicago/Turabian StylePatel, Unnati, Kavini Rathnayake, Emily C. Hunt, and Nirupama Singh. 2022. "Role of Nanomaterials in COVID-19 Prevention, Diagnostics, Therapeutics, and Vaccine Development" Journal of Nanotheranostics 3, no. 4: 151-176. https://doi.org/10.3390/jnt3040011
APA StylePatel, U., Rathnayake, K., Hunt, E. C., & Singh, N. (2022). Role of Nanomaterials in COVID-19 Prevention, Diagnostics, Therapeutics, and Vaccine Development. Journal of Nanotheranostics, 3(4), 151-176. https://doi.org/10.3390/jnt3040011