Anti-COVID-19 Nanomaterials: Directions to Improve Prevention, Diagnosis, and Treatment
Abstract
:1. Introduction
2. Prevention
2.1. Sterilization and Disinfection of Inanimate Surfaces
2.1.1. Metal Nanoparticle-Based Disinfectant
2.1.2. Naturally Nanomaterials-Based Disinfectant
2.1.3. Nanopolymer-Based Disinfectants
2.1.4. Light-Activated Nanocoating Disinfecting Viruses
2.2. Nano-Based Disinfectant Personal Protective Equipment (PPE)
2.2.1. Nanoparticles-Based PPE
2.2.2. Nanofibrous Membrane-Based PPE
2.3. Vaccination
2.3.1. Nano-Based Approach for COVID-19 Vaccine
2.3.2. Design and Administration Considerations of Nanomedicine-Based Vaccines
3. Diagnosis
Nano-Based COVID-19 Detection
Detection Platform | Pathogens | Assay Time | Limitations | Sample Matrix | Ref. |
---|---|---|---|---|---|
Rapid detection systems | SARS-CoV-2, Influenza virus | 13 min | ~48% false negatives | Nasal or throat Swab | [187] |
RT-PCR | SARS-CoV-2, SARS-CoV, MERS-CoV | 48 h | Laboratory equipment, trained personnel, limited to the detection of nucleic acids | Serum, nasal or throat swabs | [188] |
Magnetic nanosensors | SARS-CoV-2, Influenza virus | 30 min | Laboratory equipment, trained personnel | PBS, serum, water, nasal swab, milk | [125] |
CRISPER-Cas method | SARS-CoV-2, Influenza virus | 1 h | Laboratory equipment, trained personnel | PBS, serum | [189] |
Nanopore Target sequencing | SARS-CoV-2 | 10 h | Laboratory equipment, trained personnel | PBS, serum | [184,190] |
4. Treatment
4.1. Nano-Based Approach for COVID-19 Treatment
4.1.1. Targeting ACE2
4.1.2. Targeting the Immune System
4.1.3. Photodynamic Inactivation of SARS-CoV-2
4.1.4. Nano-Based Delivery of Therapeutic Agents
4.1.5. Gene Editing
5. What Is the Role of In Silico Models?
6. Concluding Remark
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
Abbreviations
WHO | World Health Organization |
CT | Computed tomography |
DIAT | Defence Institute of Advanced Technologies |
ACNSMM | Amrita Centre for Nanosciences and Molecular Medicine |
APCs | Antigen-presenting cells |
DAMPs | Damage-associated molecular patterns |
PRRs | Pattern recognition receptors |
RdRp | RNA-dependent RNA polymerase |
IgM | Immunoglobulin M |
CBC | Complete blood count |
CRP | C-Reactive protein |
ELISA | Enzyme-linked immunosorbent assay |
RT-PCR | Real-time polymerase chain reaction |
STEM | Size-tunable enrichment microdevice |
DPP4 | Dipeptidyl-peptidase 4 |
ARDS | Acute respiratory distress syndrome |
SFV | Semliki Forest virus |
IAV | Influenza A virus |
CQ | Chloroquine |
OTV | Oseltamivir |
SpCas9 | Streptococcus pyogenes Cas9 |
Ttr | Transthyretin |
ACE2 | Angiotensin-converting enzyme 2 |
PPE | Personal protection equipment |
HKUST | Hong Kong University of Science and Technology |
RBD | Receptor-binding domain |
PLGA | Poly(lactide-co-glycolide) |
PAMPs | Pathogen-associated molecular patterns |
NALT | Nasal-associated lymphoid tissue |
IgG | Immunoglobulin G |
IgA | Immunoglobulin A |
LDH | lactate dehydrogenase |
AuNPs | Gold nanoparticles |
PPT | Plasmonic photothermal |
CNT | Carbon nanotubes |
SWCNT | Single-wall carbon nanotube |
RAS | Renin-angiotensin system |
ROS | Reactive oxygen species |
VSV | Vesicular stomatitis virus |
HCQ | Hydroxychloroquine |
RBV | Antiviral drugs ribavirin |
EV71 | Enterovirus 71 |
sgRN | Single-guide RNA |
Pcsk9 | Proprotein convertase subtilisin/kexin type 9 |
References
- WHO. Pandemic. Available online: https://covid19.who.int/ (accessed on 1 January 2022).
- Petersen, E.; Koopmans, M.; Go, U.; Hamer, D.H.; Petrosillo, N.; Castelli, F.; Storgaard, M.; Al Khalili, S.; Simonsen, L. Comparing SARS-CoV-2 with SARS-CoV and influenza pandemics. Lancet Infect. Dis. 2020, 20, e238–e244. [Google Scholar] [CrossRef]
- Chertow, D.S.; Kleine, C.; Edwards, J.K.; Scaini, R.; Giuliani, R.; Sprecher, A. Ebola virus disease in West Africa—Clinical manifestations and management. N. Engl. J. Med. 2014, 371, 2054–2057. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shrestha, S.S.; Swerdlow, D.L.; Borse, R.H.; Prabhu, V.S.; Finelli, L.; Atkins, C.Y.; Owusu-Edusei, K.; Bell, B.; Mead, P.S.; Biggerstaff, M. Estimating the burden of 2009 pandemic influenza A (H1N1) in the United States (April 2009–April 2010). Clin. Infect. Dis. 2011, 52, S75–S82. [Google Scholar] [CrossRef] [Green Version]
- Vahedifard, F.; Chakravarthy, K. Nanomedicine for COVID-19: The role of nanotechnology in the treatment and diagnosis of COVID-19. Emergent Mater. 2021, 4, 75–99. [Google Scholar] [CrossRef]
- Kusumoputro, S.; Tseng, S.; Tse, J.; Au, C.; Lau, C.; Wang, X.; Xia, T. Potential nanoparticle applications for prevention, diagnosis, and treatment of COVID-19. View 2020, 1, 20200105. [Google Scholar] [CrossRef]
- Shereen, M.A.; Khan, S.; Kazmi, A.; Bashir, N.; Siddique, R. COVID-19 infection: Origin, transmission, and characteristics of human coronaviruses. J. Adv. Res. 2020, 24, 91. [Google Scholar] [CrossRef] [PubMed]
- Chaudhary, V.; Royal, A.; Chavali, M.; Yadav, S. Advancements in research and development to combat COVID-19 using nanotechnology. Nanotechnol. Environ. Eng. 2021, 6, 1–15. [Google Scholar] [CrossRef]
- WHO. Modes of Transmission of Virus Causing COVID-19: Implications for IPC Precaution Recommendations. Available online: https://www.who.int/news-room/commentaries/detail/modes-of-transmission-of-virus-causing-covid-19-implications-for-ipc-precaution-recommendations (accessed on 1 January 2022).
- Wu, Y.; Xu, X.; Chen, Z.; Duan, J.; Hashimoto, K.; Yang, L.; Liu, C.; Yang, C. Nervous system involvement after infection with COVID-19 and other coronaviruses. Brain Behav. Immun. 2020, 87, 18–22. [Google Scholar] [CrossRef]
- Baig, A.M.; Khaleeq, A.; Ali, U.; Syeda, H. Evidence of the COVID-19 virus targeting the CNS: Tissue distribution, host–virus interaction, and proposed neurotropic mechanisms. ACS Chem. Neurosci. 2020, 11, 995–998. [Google Scholar] [CrossRef] [Green Version]
- Zheng, Y.-Y.; Ma, Y.-T.; Zhang, J.-Y.; Xie, X. COVID-19 and the cardiovascular system. Nat. Rev. Cardiol. 2020, 17, 259–260. [Google Scholar] [CrossRef] [Green Version]
- Cheng, Y.; Luo, R.; Wang, K.; Zhang, M.; Wang, Z.; Dong, L.; Li, J.; Yao, Y.; Ge, S.; Xu, G. Kidney impairment is associated with in-hospital death of COVID-19 patients. medRxiv 2020. [Google Scholar] [CrossRef] [Green Version]
- Wong, S.H.; Lui, R.N.; Sung, J.J. Covid-19 and the digestive system. J. Gastroenterol. Hepatol. 2020, 35, 744–748. [Google Scholar] [CrossRef] [PubMed]
- Zhang, C.; Shi, L.; Wang, F.-S. Liver injury in COVID-19: Management and challenges. Lancet Gastroenterol. Hepatol. 2020, 5, 428–430. [Google Scholar] [CrossRef]
- Bangash, M.N.; Patel, J.; Parekh, D. COVID-19 and the liver: Little cause for concern. Lancet. Gastroenterol. Hepatol. 2020, 5, 529. [Google Scholar] [CrossRef] [Green Version]
- Hasanzadeh, A.; Alamdaran, M.; Ahmadi, S.; Nourizadeh, H.; Bagherzadeh, M.A.; Jahromi, M.A.M.; Simon, P.; Karimi, M.; Hamblin, M.R. Nanotechnology against COVID-19: Immunization, diagnostic and therapeutic studies. J. Control. Release 2021, 336, 354–374. [Google Scholar] [CrossRef]
- Hamming, I.; Timens, W.; Bulthuis, M.; Lely, A.; Navis, G.V.; van Goor, H. Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis. J. Pathol. A J. Pathol. Soc. Great Br. Irel. 2004, 203, 631–637. [Google Scholar] [CrossRef]
- Salamanna, F.; Maglio, M.; Landini, M.P.; Fini, M. Body localization of ACE-2: On the trail of the keyhole of SARS-CoV-2. Front. Med. 2020, 7, 935. [Google Scholar] [CrossRef]
- FDA Approves First COVID-19 Vaccine. Available online: https://www.fda.gov/news-events/press-announcements/fda-approves-first-covid-19-vaccine (accessed on 1 January 2022).
- Shapiro, R.S. COVID-19 vaccines and nanomedicine. Int. J. Dermatol. 2021, 60, 1047–1052. [Google Scholar] [CrossRef]
- Doagooyan, M.; Alavizadeh, S.H.; Gheibihayat, S.M.; Boustan, A.; Gheybi, F. COVID-19 therapy approaches and vaccine development: The role of nanotechnology. Nanomed. J. 2021, 8, 89–97. [Google Scholar]
- Study to Evaluate the Safety and Antiviral Activity of Remdesivir (GS-5734™) in Participants With Severe Coronavirus Disease (COVID-19). Available online: https://clinicaltrials.gov/ct2/show/NCT04292899 (accessed on 1 January 2022).
- Multiple Versus Single Dose of Ivermectin for the Treatment of Strongyloidiasis (STRONGTREAT). Available online: https://clinicaltrials.gov/ct2/show/NCT01570504 (accessed on 1 January 2022).
- Hydroxychloroquine for Treatment of Non-Severe COVID-19 (HONEST). Available online: https://clinicaltrials.gov/ct2/show/NCT04860284 (accessed on 1 January 2022).
- Heinrich, M.A.; Martina, B.; Prakash, J. Nanomedicine strategies to target coronavirus. Nano Today 2020, 35, 100961. [Google Scholar] [CrossRef]
- Talebian, S.; Conde, J. Why go NANO on COVID-19 pandemic? Matter 2020, 3, 598–601. [Google Scholar] [CrossRef]
- Kashkooli, F.M.; Soltani, M.; Souri, M. Controlled anti-cancer drug release through advanced nano-drug delivery systems: Static and dynamic targeting strategies. J. Control. Release 2020, 327, 316–349. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Jagaran, K.; Singh, M. Nanomedicine for COVID-19: Potential of copper nanoparticles. Biointerface Res. Appl. Chem. 2020, 11, 10716–10728. [Google Scholar]
- Rashidzadeh, H.; Danafar, H.; Rahimi, H.; Mozafari, F.; Salehiabar, M.; Rahmati, M.A.; Rahamooz-Haghighi, S.; Mousazadeh, N.; Mohammadi, A.; Ertas, Y.N. Nanotechnology against the novel coronavirus (severe acute respiratory syndrome coronavirus 2): Diagnosis, treatment, therapy and future perspectives. Nanomedicine 2021, 16, 497–516. [Google Scholar] [CrossRef]
- Tabish, T.A.; Hamblin, M.R. Multivalent nanomedicines to treat COVID-19: A slow train coming. Nano Today 2020, 35, 100962. [Google Scholar] [CrossRef] [PubMed]
- Bidram, E.; Esmaeili, Y.; Amini, A.; Sartorius, R.; Tay, F.R.; Shariati, L.; Makvandi, P. Nanobased Platforms for Diagnosis and Treatment of COVID-19: From Benchtop to Bedside. ACS Biomater. Sci. Eng. 2021, 7, 2150–2176. [Google Scholar] [CrossRef]
- Elkodous, M.A.; Olojede, S.; Morsi, M.; El-Sayyad, G.S. Nanomaterial-based drug delivery systems as promising carriers for patients with COVID-19. RSC Advances 2021, 11, 26463–26480. [Google Scholar] [CrossRef]
- Kampf, G.; Todt, D.; Pfaender, S.; Steinmann, E. Persistence of coronaviruses on inanimate surfaces and their inactivation with biocidal agents. J. Hosp. Infect. 2020, 104, 246–251. [Google Scholar] [CrossRef] [Green Version]
- Habib, A.; Habib, L.; Habib, K. Fluid mechanics of facial masks as personal protection equipment (PPE) of COVID-19 virus. Rev. Sci. Instrum. 2021, 92, 074101. [Google Scholar] [CrossRef] [PubMed]
- Mosallanezhad, B.; Chouhan, V.K.; Paydar, M.M.; Hajiaghaei-Keshteli, M. Disaster relief supply chain design for personal protection equipment during the COVID-19 pandemic. Appl. Soft Comput. 2021, 112, 107809. [Google Scholar] [CrossRef]
- Rodríguez, N.B.; Formentini, G.; Favi, C.; Marconi, M. Environmental implication of personal protection equipment in the pandemic era: LCA comparison of face masks typologies. Procedia Cirp 2021, 98, 306–311. [Google Scholar] [CrossRef] [PubMed]
- Akhbarizadeh, R.; Dobaradaran, S.; Nabipour, I.; Tangestani, M.; Abedi, D.; Javanfekr, F.; Jeddi, F.; Zendehboodi, A. Abandoned Covid-19 personal protective equipment along the Bushehr shores, the Persian Gulf: An emerging source of secondary microplastics in coastlines. Mar. Pollut. Bull. 2021, 168, 112386. [Google Scholar] [CrossRef] [PubMed]
- Machingaidze, S.; Wiysonge, C.S. Understanding COVID-19 vaccine hesitancy. Nat. Med. 2021, 27, 1338–1339. [Google Scholar] [CrossRef] [PubMed]
- Chemaitelly, H.; Yassine, H.M.; Benslimane, F.M.; Khatib, H.A.A.; Tang, P.; Hasan, M.R.; Malek, J.A.; Coyle, P.; Ayoub, H.H.; Kanaani, Z.A. mRNA-1273 COVID-19 vaccine effectiveness against the B. 1.1. 7 and B. 1.351 variants and severe COVID-19 disease in Qatar. Nat. Med. 2021, 27, 1–8. [Google Scholar] [CrossRef] [PubMed]
- Kim, J.H.; Marks, F.; Clemens, J.D. Looking beyond COVID-19 vaccine phase 3 trials. Nat. Med. 2021, 27, 205–211. [Google Scholar] [CrossRef]
- Solís Arce, J.S.; Warren, S.S.; Meriggi, N.F.; Scacco, A.; McMurry, N.; Voors, M.; Syunyaev, G.; Malik, A.A.; Aboutajdine, S.; Adeojo, O. COVID-19 vaccine acceptance and hesitancy in low-and middle-income countries. Nat. Med. 2021, 27, 1385–1394. [Google Scholar] [CrossRef]
- Knoll, M.D.; Wonodi, C. Oxford–AstraZeneca COVID-19 vaccine efficacy. Lancet 2021, 397, 72–74. [Google Scholar] [CrossRef]
- Kisby, T.; Yilmazer, A.; Kostarelos, K. Reasons for success and lessons learnt from nanoscale vaccines against COVID-19. Nat. Nanotechnol. 2021, 16, 1–8. [Google Scholar] [CrossRef]
- Rasmi, Y.; Saloua, K.S.; Nemati, M.; Choi, J.R. Recent Progress in Nanotechnology for COVID-19 Prevention, Diagnostics and Treatment. Nanomaterials 2021, 11, 1788. [Google Scholar] [CrossRef]
- Singh, P.; Singh, D.; Sa, P.; Mohapatra, P.; Khuntia, A.; Sahoo, S.K. Insights from nanotechnology in COVID-19: Prevention, detection, therapy and immunomodulation. Nanomedicine 2021, 16, 1219–1235. [Google Scholar] [CrossRef] [PubMed]
- Khoshnevisan, K.; Maleki, H.; Baharifar, H. Nanobiocide Based-Silver Nanomaterials Upon Coronaviruses: Approaches for Preventing Viral Infections. Nanoscale Res. Lett. 2021, 16, 1–9. [Google Scholar] [CrossRef] [PubMed]
- Hosseini, M.; Behzadinasab, S.; Benmamoun, Z.; Ducker, W.A. The Viability of SARS-COV-2 on Solid Surfaces. Curr. Opin. Colloid Interface Sci. 2021, 55, 101481. [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. 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]
- Chan, W.C. Nano research for COVID-19. ACS Publ. 2020, 14, 3719–3720. [Google Scholar] [CrossRef] [Green Version]
- Kalantar-Zadeh, K.; Ward, S.A.; Kalantar-Zadeh, K.; El-Omar, E.M. Considering the effects of microbiome and diet on SARS-CoV-2 infection: Nanotechnology roles. ACS Nano 2020, 14, 5179–5182. [Google Scholar] [CrossRef]
- Morones, J.R.; Elechiguerra, J.L.; Camacho, A.; Holt, K.; Kouri, J.B.; Ramírez, J.T.; Yacaman, M.J. The bactericidal effect of silver nanoparticles. Nanotechnology 2005, 16, 2346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yougbare, S.; Chang, T.-K.; Tan, S.-H.; Kuo, J.-C.; Hsu, P.-H.; Su, C.-Y.; Kuo, T.-R. Antimicrobial gold nanoclusters: Recent developments and future perspectives. Int. J. Mol. Sci. 2019, 20, 2924. [Google Scholar] [CrossRef] [Green Version]
- Yougbaré, S.; Mutalik, C.; Krisnawati, D.I.; Kristanto, H.; Jazidie, A.; Nuh, M.; Cheng, T.-M.; Kuo, T.-R. Nanomaterials for the photothermal killing of bacteria. Nanomaterials 2020, 10, 1123. [Google Scholar] [CrossRef]
- Look, M.; Bandyopadhyay, A.; Blum, J.S.; Fahmy, T.M. Application of nanotechnologies for improved immune response against infectious diseases in the developing world. Adv. Drug Deliv. Rev. 2010, 62, 378–393. [Google Scholar] [CrossRef] [Green Version]
- Technology against COVID-19: Nano Insights into Prevention, Diagnosis, and Treatment. Available online: https://statnano.com/technology-against-covid-19-nano-insights#ixzz75lv13n98S (accessed on 8 June 2020).
- Dizaj, S.M.; Lotfipour, F.; Barzegar-Jalali, M.; Zarrintan, M.H.; Adibkia, K. Antimicrobial activity of the metals and metal oxide nanoparticles. Mater. Sci. Eng. C 2014, 44, 278–284. [Google Scholar] [CrossRef] [PubMed]
- Mumbai, P. Available online: https://pib.gov.in/PressReleasePage.aspx?PRID=1630197 (accessed on 8 June 2020).
- Prelynx Portal, Corona Virus (Covid-19) First Response Scanner + Sanitizer. Available online: https://blog.123.design/health-2/prelynx-portal-covid-19-corona-virus-covid-19-first-response-scanner-sanitizer/ (accessed on 1 January 2022).
- HKUST Develops New Smart Anti-Microbial Coating in the Fight Against COVID-19. Available online: https://hkust.edu.hk/news/research-and-innovation/hkust-develops-new-smart-anti-microbial-coating-fight-against-covid-19 (accessed on 1 January 2022).
- Guelph Researchers: Self-Sterilizing Nano-Coating and No More Coronavirusinfected Surfaces. Available online: https://statnano.com/news/67675/Guelph-Researchers-Self-sterilizing-Nano-coating-and-No-More-Coronavirus-infected-Surfaces (accessed on 1 January 2022).
- Nikaeen, G.; Abbaszadeh, S.; Yousefinejad, S. Application of nanomaterials in treatment, anti-infection and detection of coronaviruses. Nanomedicine 2020, 15, 1501–1512. [Google Scholar] [CrossRef]
- Jones, G.W.; Monopoli, M.P.; Campagnolo, L.; Pietroiusti, A.; Tran, L.; Fadeel, B. No small matter: A perspective on nanotechnology-enabled solutions to fight COVID-19. Nanomedicine 2020, 15, 2411–2427. [Google Scholar] [CrossRef] [PubMed]
- Abo-Zeid, Y.; Ismail, N.S.; 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]
- Vaze, N.; Pyrgiotakis, G.; McDevitt, J.; Mena, L.; Melo, A.; Bedugnis, A.; Kobzik, L.; Eleftheriadou, M.; Demokritou, P. Inactivation of common hospital acquired pathogens on surfaces and in air utilizing engineered water nanostructures (EWNS) based nano-sanitizers. Nanomed. Nanotechnol. Biol. Med. 2019, 18, 234–242. [Google Scholar] [CrossRef] [PubMed]
- Deshmukh, S.P.; Patil, S.; Mullani, S.; Delekar, S. Silver nanoparticles as an effective disinfectant: A review. Mater. Sci. Eng. C 2019, 97, 954–965. [Google Scholar] [CrossRef]
- Nakamura, S.; Sato, M.; Sato, Y.; Ando, N.; Takayama, T.; Fujita, M.; Ishihara, M. Synthesis and application of silver nanoparticles (Ag NPs) for the prevention of infection in healthcare workers. Int. J. Mol. Sci. 2019, 20, 3620. [Google Scholar] [CrossRef] [Green Version]
- Sportelli, M.C.; Izzi, M.; Kukushkina, E.A.; Hossain, S.I.; Picca, R.A.; Ditaranto, N.; Cioffi, N. Can nanotechnology and materials science help the fight against SARS-CoV-2? Nanomaterials 2020, 10, 802. [Google Scholar] [CrossRef] [Green Version]
- Park, Y.J.; Jeong, M.H.; Bang, I.J.; Kim, H.R.; Chung, K.H. Guanidine-based disinfectants, polyhexamethylene guanidine-phosphate (PHMG-P), polyhexamethylene biguanide (PHMB), and oligo (2-(2-ethoxy) ethoxyethyl guanidinium chloride (PGH) induced epithelial-mesenchymal transition in A549 alveolar epithelial cells. Inhal. Toxicol. 2019, 31, 161–166. [Google Scholar] [CrossRef]
- Makvandi, P.; Wang, C.Y.; Zare, E.N.; Borzacchiello, A.; Niu, L.N.; Tay, F.R. Metal-based nanomaterials in biomedical applications: Antimicrobial activity and cytotoxicity aspects. Adv. Funct. Mater. 2020, 30, 1910021. [Google Scholar] [CrossRef]
- Rajendran, S.; Mukherjee, A.; Nguyen, T.A.; Godugu, C.; Shukla, R.K. (Eds.) Nanotoxicity: Prevention and Antibacterial Applications of Nanomaterials; Elsevier: Amsterdam, The Netherlands, 2020; pp. 107–141. [Google Scholar]
- Ray, P.C.; Khan, S.A.; Singh, A.K.; Senapati, D.; Fan, Z. Nanomaterials for targeted detection and photothermal killing of bacteria. Chem. Soc. Rev. 2012, 41, 3193–3209. [Google Scholar] [CrossRef]
- Bucharskaya, A.; Maslyakova, G.; Terentyuk, G.; Yakunin, A.; Avetisyan, Y.; Bibikova, O.; Tuchina, E.; Khlebtsov, B.; Khlebtsov, N.; Tuchin, V. Towards effective photothermal/photodynamic treatment using plasmonic gold nanoparticles. Int. J. Mol. Sci. 2016, 17, 1295. [Google Scholar] [CrossRef]
- Feng, Y.; Liu, L.; Zhang, J.; Aslan, H.; Dong, M. Photoactive antimicrobial nanomaterials. J. Mater. Chem. B 2017, 5, 8631–8652. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Jin, Y.; Chen, W.; Wang, J.; Chen, H.; Sun, L.; Li, X.; Ji, J.; Yu, Q.; Shen, L. Construction of nanomaterials with targeting phototherapy properties to inhibit resistant bacteria and biofilm infections. Chem. Eng. J. 2019, 358, 74–90. [Google Scholar] [CrossRef]
- Sarkar, S.; Bhattacharjee, C.; Sarkar, S. Smart Polymeric Coatings to Enhance the Antibacterial, Anti-fogging and Self-Healing Nature of a Coated Surface. Smart Polym. Compos. 2018, 21, 64. [Google Scholar]
- Howard, J.; Huang, A.; Li, Z.; Tufekci, Z.; Zdimal, V.; van der Westhuizen, H.-M.; von Delft, A.; Price, A.; Fridman, L.; Tang, L.-H. An evidence review of face masks against COVID-19. Proc. Natl. Acad. Sci. USA 2021, 118, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Ju, J.T.; Boisvert, L.; Zuo, Y.Y. Face masks against COVID-19: Standards, efficacy, testing and decontamination methods. Adv. Colloid Interface Sci. 2021, 292, 102435. [Google Scholar] [CrossRef]
- Catching, A.; Capponi, S.; Yeh, M.T.; Bianco, S.; Andino, R. Examining the interplay between face mask usage, asymptomatic transmission, and social distancing on the spread of COVID-19. Sci. Rep. 2021, 11, 1–11. [Google Scholar] [CrossRef]
- Pezzini, A.; Padovani, A. Lifting the mask on neurological manifestations of COVID-19. Nat. Rev. Neurol. 2020, 16, 636–644. [Google Scholar] [CrossRef]
- Rohit, A.; Rajasekaran, S.; Shenoy, S.; Rai, S.; Iddya, K.; Dorairajan, S.K. Reprocessing of N95 masks: Experience from a resource-limited setting in India. Int. J. Infect. Dis. 2021, 104, 41–44. [Google Scholar] [CrossRef]
- Bartoszko, J.J.; Farooqi, M.A.M.; Alhazzani, W.; Loeb, M. Medical masks vs N95 respirators for preventing COVID-19 in healthcare workers: A systematic review and meta-analysis of randomized trials. Influenza Other Respir. Viruses 2020, 14, 365–373. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Karim, N.; Afroj, S.; Lloyd, K.; Oaten, L.C.; Andreeva, D.V.; Carr, C.; Farmery, A.D.; Kim, I.-D.; Novoselov, K.S. Sustainable personal protective clothing for healthcare applications: A review. ACS Nano 2020, 14, 12313–12340. [Google Scholar] [CrossRef]
- Promethean Particles Puts Its Nano-Copper to the Test in the Fight against Viruses. Available online: https://prometheanparticles.co.uk/promethean-particles-puts-its-nano-copper-to-the-test-in-the-fight-against-viruses/ (accessed on 1 January 2022).
- Here’s How Nanotechnologies Tailor Face Masks for Coronavirus Era. Available online: https://statnano.com/news/67662/Here%E2%80%99s%20HowNanotechnologies-Tailor-Face-Masks-for-Coronavirus-Era,%20StatNano%20Publication (accessed on 1 January 2022).
- Sonovia SonoMask. Available online: https://sonoviastore.com/ (accessed on 1 January 2022).
- VIRUS KILLER Membrane. Available online: https://www.respilon.com/products/nanofiber-membranes (accessed on 1 January 2022).
- Announcing the Launch of the “Zexeed™” YamASHIN Filter Mask. Available online: http://www.yamashin-filter.co.jp/nano_mask/eng/index.html (accessed on 1 January 2022).
- Why Is Ultimate Yamashin Filter Mask™ So Special? Available online: https://www.aerasmedical.com/yamashin-nano (accessed on 1 January 2022).
- 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]
- Nickels, L. Antiviral boost for nanoparticles. Met. Powder Rep. 2020, 75, 330–333. [Google Scholar] [CrossRef]
- Konda, A.; Prakash, A.; Moss, G.A.; Schmoldt, M.; Grant, G.D.; Guha, S. Aerosol filtration efficiency of common fabrics used in respiratory cloth masks. ACS Nano 2020, 14, 6339–6347. [Google Scholar] [CrossRef] [PubMed]
- Mahapatra, P.S.; Chatterjee, S.; Tiwari, M.K.; Ganguly, R.; Megaridis, C.M. Surface treatments to enhance the functionality of PPEs. Trans. Indian Natl. Acad. Eng. 2020, 5, 333–336. [Google Scholar] [CrossRef]
- Ramaseshan, R.; Sundarrajan, S.; Liu, Y.; Barhate, R.; Lala, N.L.; Ramakrishna, S. Functionalized polymer nanofibre membranes for protection from chemical warfare stimulants. Nanotechnology 2006, 17, 2947. [Google Scholar] [CrossRef]
- Zhu, M.; Han, J.; Wang, F.; Shao, W.; Xiong, R.; Zhang, Q.; Pan, H.; Yang, Y.; Samal, S.K.; Zhang, F. Electrospun nanofibers membranes for effective air filtration. Macromol. Mater. Eng. 2017, 302, 1600353. [Google Scholar] [CrossRef]
- Tebyetekerwa, M.; Xu, Z.; Yang, S.; Ramakrishna, S. Electrospun nanofibers-based face masks. Adv. Fiber Mater. 2020, 2, 161–166. [Google Scholar] [CrossRef]
- Skaria, S.D.; Smaldone, G.C. Respiratory source control using surgical masks with nanofiber media. Ann. Occup. Hyg. 2014, 58, 771–781. [Google Scholar]
- Recyclable Nano-Filter for Face Masks Developed. Available online: https://www.labonline.com.au/content/consumables/news/recyclable-nano-filter-for-face-masks-developed-341058879 (accessed on 8 June 2020).
- Amrita Nanomask–COVID-19 Ready Face Masks That Can Protect from Bacterial and Viral Droplets. Available online: https://www.amrita.edu/news/amrita-nanomask-covid-19-ready-face-masks-can-protect-bacterial-and-viral-droplets (accessed on 8 June 2020).
- Florindo, H.F.; Kleiner, R.; Vaskovich-Koubi, D.; Acúrcio, R.C.; Carreira, B.; Yeini, E.; Tiram, G.; Liubomirski, Y.; Satchi-Fainaro, R. Immune-mediated approaches against COVID-19. Nat. Nanotechnol. 2020, 15, 630–645. [Google Scholar] [CrossRef] [PubMed]
- Schoeman, D.; Fielding, B.C. Coronavirus envelope protein: Current knowledge. Virol. J. 2019, 16, 1–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xia, S.; Zhu, Y.; Liu, M.; Lan, Q.; Xu, W.; Wu, Y.; Ying, T.; Liu, S.; Shi, Z.; Jiang, S. Fusion mechanism of 2019-nCoV and fusion inhibitors targeting HR1 domain in spike protein. Cell. Mol. Immunol. 2020, 17, 765–767. [Google Scholar] [CrossRef] [PubMed]
- Benvenuto, D.; Giovanetti, 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] [Green Version]
- Wu, F.; Wang, A.; Liu, M.; Wang, Q.; Chen, J.; Xia, S.; Ling, Y.; Zhang, Y.; Xun, J.; Lu, L. Neutralizing antibody responses to SARS-CoV-2 in a COVID-19 recovered patient cohort and their implications. medRxiv 2020. [Google Scholar] [CrossRef]
- Watanabe, Y.; Allen, J.D.; Wrapp, D.; McLellan, J.S.; Crispin, M. Site-specific analysis of the SARS-CoV-2 glycan shield. bioRxiv 2020. [Google Scholar] [CrossRef] [Green Version]
- Banerjee, A.; Santra, D.; Maiti, S. Energetics based epitope screening in SARS CoV-2 (COVID 19) spike glycoprotein by Immuno-informatic analysis aiming to a suitable vaccine development. bioRxiv 2020. [Google Scholar] [CrossRef] [Green Version]
- Vu, M.N.; Kelly, H.G.; Kent, S.J.; Wheatley, A.K. Current and future nanoparticle vaccines for COVID-19. EBioMedicine 2021, 74, 103699. [Google Scholar] [CrossRef]
- NIH. Health, NIH Clinical Trial of Investigational Vaccine for COVID-19 Begins. Available online: https://www.nih.gov/news-events/newsreleases/nih-clinical-trial-investigational-vaccine-covid-19-begins (accessed on 8 June 2020).
- WHO. Draft Landscape of COVID-19 Candidate Vaccines; World Health Organisation: Geneve, Switzerland, 2020; pp. 10–12. [Google Scholar]
- Kamat, S.; Kumari, M.; Jayabaskaran, C. Nano-engineered tools in the diagnosis, therapeutics, prevention, and mitigation of SARS-CoV-2. J. Control. Release 2021, 338, 813–836. [Google Scholar] [CrossRef]
- Chauhan, G.; Madou, M.J.; Kalra, S.; Chopra, V.; Ghosh, D.; Martinez-Chapa, S.O. Nanotechnology for COVID-19: Therapeutics and vaccine research. ACS Nano 2020, 14, 7760–7782. [Google Scholar] [CrossRef]
- Zhao, L.; Seth, A.; Wibowo, N.; Zhao, C.-X.; Mitter, N.; Yu, C.; Middelberg, A.P. Nanoparticle vaccines. Vaccine 2014, 32, 327–337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Apostolopoulos, V.; Thalhammer, T.; Tzakos, A.G.; Stojanovska, L. Targeting antigens to dendritic cell receptors for vaccine development. J. Drug Deliv. 2013, 2013, 1–23. [Google Scholar] [CrossRef]
- Formiga, F.R.; Leblanc, R.; de Souza Rebouças, J.; Farias, L.P.; de Oliveira, R.N.; Pena, L. Ivermectin: An award-winning drug with expected antiviral activity against COVID-19. J. Control. Release 2021, 329, 758–761. [Google Scholar] [CrossRef] [PubMed]
- Shin, M.D.; Shukla, S.; Chung, Y.H.; Beiss, V.; Chan, S.K.; Ortega-Rivera, O.A.; Wirth, D.M.; Chen, A.; Sack, M.; Pokorski, J.K. COVID-19 vaccine development and a potential nanomaterial path forward. Nat. Nanotechnol. 2020, 15, 646–655. [Google Scholar] [CrossRef]
- Coleman, C.M.; Liu, Y.V.; Mu, H.; Taylor, J.K.; Massare, M.; Flyer, D.C.; Glenn, G.M.; Smith, G.E.; Frieman, M.B. Purified coronavirus spike protein nanoparticles induce coronavirus neutralizing antibodies in mice. Vaccine 2014, 32, 3169–3174. [Google Scholar] [CrossRef]
- Sekimukai, H.; Iwata-Yoshikawa, N.; Fukushi, S.; Tani, H.; Kataoka, M.; Suzuki, T.; Hasegawa, H.; Niikura, K.; Arai, K.; Nagata, N. Gold nanoparticle-adjuvanted S protein induces a strong antigen-specific IgG response against severe acute respiratory syndrome-related coronavirus infection, but fails to induce protective antibodies and limit eosinophilic infiltration in lungs. Microbiol. Immunol. 2020, 64, 33–51. [Google Scholar] [CrossRef] [Green Version]
- Pimentel, T.A.; Yan, Z.; Jeffers, S.A.; Holmes, K.V.; Hodges, R.S.; Burkhard, P. Peptide nanoparticles as novel immunogens: Design and analysis of a prototypic severe acute respiratory syndrome vaccine. Chem. Biol. Drug Des. 2009, 73, 53–61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, F.; Kream, R.M.; Stefano, G.B. An evidence based perspective on mRNA-SARS-CoV-2 vaccine development. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2020, 26, e924700–e924701. [Google Scholar] [CrossRef] [Green Version]
- Tai, W.; Zhang, X.; Drelich, A.; Shi, J.; Hsu, J.C.; Luchsinger, L.; Hillyer, C.D.; Tseng, C.-T.K.; Jiang, S.; Du, L. A novel receptor-binding domain (RBD)-based mRNA vaccine against SARS-CoV-2. Cell Res. 2020, 30, 932–935. [Google Scholar] [CrossRef]
- McKay, P.F.; Hu, K.; Blakney, A.K.; Samnuan, K.; Brown, J.C.; Penn, R.; Zhou, J.; Bouton, C.R.; Rogers, P.; Polra, K. Self-amplifying RNA SARS-CoV-2 lipid nanoparticle vaccine candidate induces high neutralizing antibody titers in mice. Nat. Commun. 2020, 11, 1–7. [Google Scholar] [CrossRef]
- Arcturus Reports Additional Supportive Preclinical Data for Its COVID-19 Vaccine Candidate (LUNAR-COV19). Available online: https://www.biospace.com/article/releases/arcturus-reports-additional-supportive-preclinical-data-for-its-covid-19-vaccine-candidate-lunar-cov19-/ (accessed on 8 June 2020).
- Lu, J.; Lu, G.; Tan, S.; Xia, J.; Xiong, H.; Yu, X.; Qi, Q.; Yu, X.; Li, L.; Yu, H. A COVID-19 mRNA vaccine encoding SARS-CoV-2 virus-like particles induces a strong antiviral-like immune response in mice. Cell Res. 2020, 30, 936–939. [Google Scholar] [CrossRef] [PubMed]
- Gubernatorova, E.O.; Gorshkova, E.A.; Polinova, A.I.; Drutskaya, M.S. IL-6: Relevance for immunopathology of SARS-CoV-2. Cytokine Growth Factor Rev. 2020, 53, 13–24. [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] [Green Version]
- 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] [PubMed]
- Kanekiyo, M.; Wei, C.-J.; Yassine, H.M.; McTamney, P.M.; Boyington, J.C.; Whittle, J.R.; Rao, S.S.; Kong, W.-P.; Wang, L.; Nabel, G. Self-assembling influenza nanoparticle vaccines elicit broadly neutralizing H1N1 antibodies. Nature 2013, 499, 102–106. [Google Scholar] [CrossRef]
- Irvine, D.; Hanson, M.C.; Rakhra, K.; Tokatlian, T. Synthetic nanoparticles for vaccines and immunotherapy. Chem. Rev. 2015, 115, 11109–11146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Salazar-González, J.A.; Gonzalez-Ortega, O.; Rosales-Mendoza, S. Gold nanoparticles and vaccine development. Expert Rev. Vaccines 2015, 14, 1197–1211. [Google Scholar] [CrossRef]
- Pati, R.; Shevtsov, M.; Sonawane, A. Nanoparticle vaccines against infectious diseases. Front. Immunol. 2018, 9, 2224. [Google Scholar] [CrossRef] [Green Version]
- 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, 28–51. [Google Scholar] [CrossRef] [Green Version]
- Hamdy, S.; Haddadi, A.; Hung, R.W.; Lavasanifar, A. Targeting dendritic cells with nano-particulate PLGA cancer vaccine formulations. Adv. Drug Deliv. Rev. 2011, 63, 943–955. [Google Scholar] [CrossRef]
- Kranz, L.M.; Diken, M.; Haas, H.; Kreiter, S.; Loquai, C.; Reuter, K.C.; Meng, M.; Fritz, D.; Vascotto, F.; Hefesha, H. Systemic RNA delivery to dendritic cells exploits antiviral defence for cancer immunotherapy. Nature 2016, 534, 396–401. [Google Scholar] [CrossRef]
- Lung, P.; Yang, J.; Li, Q. Nanoparticle formulated vaccines: Opportunities and challenges. Nanoscale 2020, 12, 5746–5763. [Google Scholar] [CrossRef]
- Geall, A.J.; Verma, A.; Otten, G.R.; Shaw, C.A.; Hekele, A.; Banerjee, K.; Cu, Y.; Beard, C.W.; Brito, L.A.; Krucker, T. Nonviral delivery of self-amplifying RNA vaccines. Proc. Natl. Acad. Sci. USA 2012, 109, 14604–14609. [Google Scholar] [CrossRef] [Green Version]
- Reichmuth, A.M.; Oberli, M.A.; Jaklenec, A.; Langer, R.; Blankschtein, D. mRNA vaccine delivery using lipid nanoparticles. Ther. Deliv. 2016, 7, 319–334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kanasty, R.; Dorkin, J.R.; Vegas, A.; Anderson, D. Delivery materials for siRNA therapeutics. Nat. Mater. 2013, 12, 967–977. [Google Scholar] [CrossRef]
- Pardi, N.; Tuyishime, S.; Muramatsu, H.; Kariko, K.; Mui, B.L.; Tam, Y.K.; Madden, T.D.; Hope, M.J.; Weissman, D. Expression kinetics of nucleoside-modified mRNA delivered in lipid nanoparticles to mice by various routes. J. Control. Release 2015, 217, 345–351. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brito, L.A.; Chan, M.; Shaw, C.A.; Hekele, A.; Carsillo, T.; Schaefer, M.; Archer, J.; Seubert, A.; Otten, G.R.; Beard, C.W. A cationic nanoemulsion for the delivery of next-generation RNA vaccines. Mol. Ther. 2014, 22, 2118–2129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kaczmarek, J.C.; Patel, A.K.; Kauffman, K.J.; Fenton, O.S.; Webber, M.J.; Heartlein, M.W.; DeRosa, F.; Anderson, D.G. Polymer–lipid nanoparticles for systemic delivery of mRNA to the lungs. Angew. Chem. 2016, 128, 14012–14016. [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] [PubMed] [Green Version]
- Wang, G.; Pan, L.; Zhang, Y.; Wang, Y.; Zhang, Z.; Lü, J.; Zhou, P.; Fang, Y.; Jiang, S. Intranasal delivery of cationic PLGA nano/microparticles-loaded FMDV DNA vaccine encoding IL-6 elicited protective immunity against FMDV challenge. PLoS ONE 2011, 6, e27605. [Google Scholar] [CrossRef]
- Lim, M.; Badruddoza, A.Z.M.; Firdous, J.; Azad, M.; Mannan, A.; Al-Hilal, T.A.; Cho, C.-S.; Islam, M.A. Engineered nanodelivery systems to improve DNA vaccine technologies. Pharmaceutics 2020, 12, 30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhao, K.; Li, W.; Huang, T.; Luo, X.; Chen, G.; Zhang, Y.; Guo, C.; Dai, C.; Jin, Z.; Zhao, Y. Preparation and efficacy of Newcastle disease virus DNA vaccine encapsulated in PLGA nanoparticles. PLoS ONE 2013, 8, e82648. [Google Scholar] [CrossRef]
- Pack, D.W.; Hoffman, A.S.; Pun, S.; Stayton, P.S. Design and development of polymers for gene delivery. Nat. Rev. Drug Discov. 2005, 4, 581–593. [Google Scholar] [CrossRef] [PubMed]
- Steitz, J.; Britten, C.M.; Wölfel, T.; Tüting, T. Effective induction of anti-melanoma immunity following genetic vaccination with synthetic mRNA coding for the fusion protein EGFP. TRP2. Cancer Immunol. Immunother. 2006, 55, 246–253. [Google Scholar] [CrossRef] [PubMed]
- Hutnick, N.A.; Myles, D.J.; Ferraro, B.; Lucke, C.; Lin, F.; Yan, J.; Broderick, K.E.; Khan, A.S.; Sardesai, N.Y.; Weiner, D.B. Intradermal DNA vaccination enhanced by low-current electroporation improves antigen expression and induces robust cellular and humoral immune responses. Hum. Gene Ther. 2012, 23, 943–950. [Google Scholar] [CrossRef] [Green Version]
- Reed, S.G.; Bertholet, S.; Coler, R.N.; Friede, M. New horizons in adjuvants for vaccine development. Trends Immunol. 2009, 30, 23–32. [Google Scholar] [CrossRef]
- Gallucci, S.; Matzinger, P. Danger signals: SOS to the immune system. Curr. Opin. Immunol. 2001, 13, 114–119. [Google Scholar] [CrossRef]
- Akira, S.; Uematsu, S.; Takeuchi, O. Pathogen recognition and innate immunity. Cell 2006, 124, 783–801. [Google Scholar] [CrossRef] [Green Version]
- Iwasaki, A.; Medzhitov, R. Toll-like receptor control of the adaptive immune responses. Nat. Immunol. 2004, 5, 987–995. [Google Scholar] [CrossRef]
- Platanias, L.C. Mechanisms of type-I-and type-II-interferon-mediated signalling. Nat. Rev. Immunol. 2005, 5, 375–386. [Google Scholar] [CrossRef]
- Tan, X.; Sun, L.; Chen, J.; Chen, Z.J. Detection of microbial infections through innate immune sensing of nucleic acids. Annu. Rev. Microbiol. 2018, 72, 447–478. [Google Scholar] [CrossRef] [PubMed]
- Hanson, M.C.; Crespo, M.P.; Abraham, W.; Moynihan, K.D.; Szeto, G.L.; Chen, S.H.; Melo, M.B.; Mueller, S.; Irvine, D.J. Nanoparticulate STING agonists are potent lymph node–targeted vaccine adjuvants. J. Clin. Investig. 2015, 125, 2532–2546. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hamdy, S.; Elamanchili, P.; Alshamsan, A.; Molavi, O.; Satou, T.; Samuel, J. Enhanced antigen-specific primary CD4+ and CD8+ responses by codelivery of ovalbumin and toll-like receptor ligand monophosphoryl lipid A in poly (D, L-lactic-co-glycolic acid) nanoparticles. J. Biomed. Mater. Res. Part A 2007, 81, 652–662. [Google Scholar] [CrossRef]
- Sokolova, V.; Knuschke, T.; Kovtun, A.; Buer, J.; Epple, M.; Westendorf, A.M. The use of calcium phosphate nanoparticles encapsulating Toll-like receptor ligands and the antigen hemagglutinin to induce dendritic cell maturation and T cell activation. Biomaterials 2010, 31, 5627–5633. [Google Scholar] [CrossRef]
- Blander, J.M.; Medzhitov, R. Regulation of phagosome maturation by signals from toll-like receptors. Science 2004, 304, 1014–1018. [Google Scholar] [CrossRef] [PubMed]
- Hoffmann, E.; Kotsias, F.; Visentin, G.; Bruhns, P.; Savina, A.; Amigorena, S. Autonomous phagosomal degradation and antigen presentation in dendritic cells. Proc. Natl. Acad. Sci. USA 2012, 109, 14556–14561. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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. Polymeric synthetic nanoparticles for the induction of antigen-specific immunological tolerance. Proc. Natl. Acad. Sci. USA 2015, 112, E156–E165. [Google Scholar] [CrossRef] [Green Version]
- 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] [Green Version]
- Scanlon, V.C.; Sanders, T. Essentials of Anatomy and Physiology; FA Davis: Philadelphia, PA, USA, 2018; pp. 80–112. [Google Scholar]
- Al-Halifa, S.; Gauthier, L.; Arpin, D.; Bourgault, S.; Archambault, D. Nanoparticle-based vaccines against respiratory viruses. Front. Immunol. 2019, 10, 22. [Google Scholar] [CrossRef] [Green Version]
- Zuercher, A.W.; Coffin, S.E.; Thurnheer, M.C.; Fundova, P.; Cebra, J.J. Nasal-associated lymphoid tissue is a mucosal inductive site for virus-specific humoral and cellular immune responses. J. Immunol. 2002, 168, 1796–1803. [Google Scholar] [CrossRef]
- Neuman, B.W.; Adair, B.D.; Yoshioka, C.; Quispe, J.D.; Orca, G.; Kuhn, P.; Milligan, R.A.; Yeager, M.; Buchmeier, M.J. Supramolecular architecture of severe acute respiratory syndrome coronavirus revealed by electron cryomicroscopy. J. Virol. 2006, 80, 7918–7928. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Martin, J.D.; Cabral, H.; Stylianopoulos, T.; Jain, R.K. Improving cancer immunotherapy using nanomedicines: Progress, opportunities and challenges. Nat. Rev. Clin. Oncol. 2020, 17, 251–266. [Google Scholar] [CrossRef] [PubMed]
- DosSantos, M.F.; Devalle, S.; Aran, V.; Capra, D.; Roque, N.R.; Coelho-Aguiar, J.D.M.; Subilhaga, J.G.; Pereira, C.M.; d’Andrea Meira, I.; Filho, P.N.S. Neuromechanisms of SARS-CoV-2: A review. Front. Neuroanat. 2020, 14, 37. [Google Scholar] [CrossRef] [PubMed]
- Galdiero, S.; Falanga, A.; Cantisani, M.; Ingle, A.; Galdiero, M.; Rai, M. Chapter 15: Silver nanoparticles as novel antibacterial and antiviral agents. In Handbook of Nanobiomedical Research: Fundamentals, Applications and Recent Developments: Volume 1. Materials for Nanomedicine; World Scientific: Singapore, 2014; pp. 565–594. [Google Scholar]
- Peek, L.J.; Middaugh, C.R.; Berkland, C. Nanotechnology in vaccine delivery. Adv. Drug Deliv. Rev. 2008, 60, 915–928. [Google Scholar] [CrossRef] [PubMed]
- Berardi, A.; Evans, D.J.; Bombelli, F.B.; Lomonossoff, G.P. Stability of plant virus-based nanocarriers in gastrointestinal fluids. Nanoscale 2018, 10, 1667–1679. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rae, C.S.; Khor, W.; Wang, Q.; Destito, G.; Gonzalez, M.J.; Singh, P.; Thomas, D.M.; Estrada, M.N.; Powell, E.; Finn, M. Systemic trafficking of plant virus nanoparticles in mice via the oral route. Virology 2005, 343, 224–235. [Google Scholar] [CrossRef] [Green Version]
- Tao, W.; Gill, H.S. M2e-immobilized gold nanoparticles as influenza A vaccine: Role of soluble M2e and longevity of protection. Vaccine 2015, 33, 2307–2315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Borges, O.; Cordeiro-da-Silva, A.; Tavares, J.; Santarém, N.; de Sousa, A.; Borchard, G.; Junginger, H.E. Immune response by nasal delivery of hepatitis B surface antigen and codelivery of a CpG ODN in alginate coated chitosan nanoparticles. Eur. J. Pharm. Biopharm. 2008, 69, 405–416. [Google Scholar] [CrossRef] [Green Version]
- Lee, P.W.; Shukla, S.; Wallat, J.D.; Danda, C.; Steinmetz, N.F.; Maia, J.; Pokorski, J.K. Biodegradable viral nanoparticle/polymer implants prepared via melt-processing. ACS Nano 2017, 11, 8777–8789. [Google Scholar] [CrossRef] [PubMed]
- Bajrovic, I.; Schafer, S.C.; Romanovicz, D.K.; Croyle, M.A. Novel technology for storage and distribution of live vaccines and other biological medicines at ambient temperature. Sci. Adv. 2020, 6, eaau4819. [Google Scholar] [CrossRef] [Green Version]
- Wang, Q.; Lin, T.; Tang, L.; Johnson, J.E.; Finn, M. Icosahedral virus particles as addressable nanoscale building blocks. Angew. Chem. 2002, 114, 477–480. [Google Scholar] [CrossRef]
- Poortahmasebi, V.; Zandi, M.; Soltani, S.; Jazayeri, S.M. Clinical performance of RT-PCR and chest CT scan for COVID-19 diagnosis; a systematic review. Adv. J. Emerg. Med. 2020, 4, e57. [Google Scholar]
- Khailany, R.A.; Safdar, M.; Ozaslan, M. Genomic characterization of a novel SARS-CoV-2. Gene Rep. 2020, 19, 100682. [Google Scholar] [CrossRef]
- Huang, C.; Wang, Y.; Li, X.; Ren, L.; Zhao, J.; Hu, Y.; Zhang, L.; Fan, G.; Xu, J.; Gu, X. Clinical features of patients infected with 2019 novel coronavirus in Wuhan. Lancet 2020, 395, 497–506. [Google Scholar] [CrossRef] [Green Version]
- Emery, S.L.; Erdman, D.D.; Bowen, M.D.; Newton, B.R.; Winchell, J.M.; Meyer, R.F.; Tong, S.; Cook, B.T.; Holloway, B.P.; McCaustland, K.A. Real-time reverse transcription–polymerase chain reaction assay for SARS-associated coronavirus. Emerg. Infect. Dis. 2004, 10, 311. [Google Scholar] [CrossRef]
- Pan, Y.; Guan, H.; Zhou, S.; Wang, Y.; Li, Q.; Zhu, T.; Hu, Q.; Xia, L. Initial CT findings and temporal changes in patients with the novel coronavirus pneumonia (2019-nCoV): A study of 63 patients in Wuhan, China. Eur. Radiol. 2020, 30, 3306–3309. [Google Scholar] [CrossRef] [Green Version]
- Tuaillon, E.; Bollore, K.; Pisoni, A.; Debiesse, S.; Renault, C.; Marie, S.; Groc, S.; Niels, C.; Pansu, N.; Dupuy, A.M. Detection of SARS-CoV-2 antibodies using commercial assays and seroconversion patterns in hospitalized patients. J. Infect. 2020, 81, e39–e45. [Google Scholar] [CrossRef]
- Sadiq, I.Z.; Abubakar, F.S.; Dan-Iya, B.I. Role of nanoparticles in tackling COVID-19 pandemic: A bio-nanomedical approach. J. Taibah Univ. Sci. 2021, 15, 198–207. [Google Scholar] [CrossRef]
- Tabish, T.A.; Narayan, R.J.; Edirisinghe, M. Rapid and label-free detection of COVID-19 using coherent anti-Stokes Raman scattering microscopy. Mrs Commun. 2020, 10, 566–572. [Google Scholar] [CrossRef]
- Rangayasami, A.; Kannan, K.; Murugesan, S.; Radhika, D.; Sadasivuni, K.K.; Reddy, K.R.; Raghu, A.V. Influence of nanotechnology to combat against COVID-19 for global health emergency: A review. Sens. Int. 2021, 2, 100079. [Google Scholar] [CrossRef]
- Quesada-González, D.; Merkoçi, A. Nanomaterial-based devices for point-of-care diagnostic applications. Chem. Soc. Rev. 2018, 47, 4697–4709. [Google Scholar] [CrossRef]
- Klostranec, J.M.; Xiang, Q.; Farcas, G.A.; Lee, J.A.; Rhee, A.; Lafferty, E.I.; Perrault, S.D.; Kain, K.C.; Chan, W.C. Convergence of quantum dot barcodes with microfluidics and signal processing for multiplexed high-throughput infectious disease diagnostics. Nano Lett. 2007, 7, 2812–2818. [Google Scholar] [CrossRef]
- Sadeghi, I.; Byrne, J.; Shakur, R.; Langer, R. Engineered drug delivery devices to address Global Health challenges. J. Control. Release 2021, 331, 503–514. [Google Scholar] [CrossRef] [PubMed]
- Qiu, P.-L.; Liu, S.-Y.; Bradshaw, M.; Rooney-Latham, S.; Takamatsu, S.; Bulgakov, T.S.; Tang, S.-R.; Feng, J.; Jin, D.-N.; Aroge, T. Multi-locus phylogeny and taxonomy of an unresolved, heterogeneous species complex within the genus Golovinomyces (Ascomycota, Erysiphales), including G. ambrosiae, G. circumfusus and G. spadiceus. BMC Microbiol. 2020, 20, 1–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wu, K.; Saha, R.; Su, D.; Krishna, V.D.; Liu, J.; Cheeran, M.C.-J.; Wang, J.-P. Magnetic-nanosensor-based virus and pathogen detection strategies before and during COVID-19. ACS Appl. Nano Mater. 2020, 3, 9560–9580. [Google Scholar] [CrossRef]
- Iravani, S. Nano-and biosensors for the detection of SARS-CoV-2: Challenges and opportunities. Mater. Adv. 2020, 1, 3092–3103. [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]
- 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]
- 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]
- Somvanshi, S.B.; Kharat, P.B.; Saraf, T.S.; Somwanshi, S.B.; Shejul, S.B.; Jadhav, K.M. Multifunctional nano-magnetic particles assisted viral RNA-extraction protocol for potential detection of COVID-19. Mater. Res. Innov. 2021, 25, 169–174. [Google Scholar] [CrossRef]
- Radwan, S.H.; Azzazy, H.M. Gold nanoparticles for molecular diagnostics. Expert Rev. Mol. Diagn. 2009, 9, 511–524. [Google Scholar] [CrossRef]
- Laromaine, A.; Koh, L.; Murugesan, M.; Ulijn, R.V.; Stevens, M.M. Protease-triggered dispersion of nanoparticle assemblies. J. Am. Chem. Soc. 2007, 129, 4156–4157. [Google Scholar] [CrossRef] [PubMed]
- Kim, H.; Park, M.; Hwang, J.; Kim, J.H.; Chung, D.-R.; Lee, K.-S.; Kang, M. Development of label-free colorimetric assay for MERS-CoV using gold nanoparticles. ACS Sens. 2019, 4, 1306–1312. [Google Scholar] [CrossRef] [PubMed]
- Li, H.; Rothberg, L. Colorimetric detection of DNA sequences based on electrostatic interactions with unmodified gold nanoparticles. Proc. Natl. Acad. Sci. USA 2004, 101, 14036–14039. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yadavalli, T.; Shukla, D. Role of metal and metal oxide nanoparticles as diagnostic and therapeutic tools for highly prevalent viral infections. Nanomed. Nanotechnol. Biol. Med. 2017, 13, 219–230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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. 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] [Green Version]
- Qiu, G.; Gai, Z.; Tao, Y.; Schmitt, J.; Kullak-Ublick, G.A.; Wang, J. Dual-functional plasmonic photothermal biosensors for highly accurate severe acute respiratory syndrome coronavirus 2 detection. ACS Nano 2020, 14, 5268–5277. [Google Scholar] [CrossRef] [Green Version]
- Nikazar, S.; Sivasankarapillai, V.S.; Rahdar, A.; Gasmi, S.; Anumol, P.; Shanavas, M.S. Revisiting the cytotoxicity of quantum dots: An in-depth overview. Biophys. Rev. 2020, 12, 703–718. [Google Scholar] [CrossRef]
- Peer, D.; Karp, J.M.; Hong, S.; Farokhzad, O.C.; Margalit, R.; Langer, R. Nanocarriers as an emerging platform for cancer therapy. Nat. Nanotechnol. 2007, 2, 751–760. [Google Scholar] [CrossRef]
- Manivannan, S.; Ponnuchamy, K. Quantum dots as a promising agent to combat COVID-19. Appl. Organomet. Chem. 2020, 34, e5887. [Google Scholar] [CrossRef]
- Ashiba, H.; Sugiyama, Y.; Wang, X.; Shirato, H.; Higo-Moriguchi, K.; Taniguchi, K.; Ohki, Y.; Fujimaki, M. Detection of norovirus virus-like particles using a surface plasmon resonance-assisted fluoroimmunosensor optimized for quantum dot fluorescent labels. Biosens. Bioelectron. 2017, 93, 260–266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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]
- Georgakilas, V.; Perman, J.A.; Tucek, J.; Zboril, R. Broad family of carbon nanoallotropes: Classification, chemistry, and applications of fullerenes, carbon dots, nanotubes, graphene, nanodiamonds, and combined superstructures. Chem. Rev. 2015, 115, 4744–4822. [Google Scholar] [CrossRef]
- Hola, K.; Zhang, Y.; Wang, Y.; Giannelis, E.P.; Zboril, R.; Rogach, A.L. Carbon dots—Emerging light emitters for bioimaging, cancer therapy and optoelectronics. Nano Today 2014, 9, 590–603. [Google Scholar] [CrossRef]
- Lim, S.Y.; Shen, W.; Gao, Z. Carbon quantum dots and their applications. Chem. Soc. Rev. 2015, 44, 362–381. [Google Scholar] [CrossRef] [PubMed]
- Yeh, Y.-T.; Tang, Y.; Sebastian, A.; Dasgupta, A.; Perea-Lopez, N.; Albert, I.; Lu, H.; Terrones, M.; Zheng, S.-Y. Tunable and label-free virus enrichment for ultrasensitive virus detection using carbon nanotube arrays. Sci. Adv. 2016, 2, e1601026. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pinals, R.L.; Ledesma, F.; Yang, D.; Navarro, N.; Jeong, S.; Pak, J.E.; Kuo, L.; Chuang, Y.-C.; Cheng, Y.-W.; Sun, H.-Y. Rapid SARS-CoV-2 spike protein detection by carbon nanotube-based near-infrared nanosensors. Nano Lett. 2021, 21, 2272–2280. [Google Scholar] [CrossRef] [PubMed]
- Báez-Santos, Y.M.; John, S.E.S.; Mesecar, A.D. The SARS-coronavirus papain-like protease: Structure, function and inhibition by designed antiviral compounds. Antivir. Res. 2015, 115, 21–38. [Google Scholar] [CrossRef]
- Hoffmann, M.; Kleine-Weber, H.; Schroeder, S.; Krüger, N.; Herrler, T.; Erichsen, S.; Schiergens, T.S.; Herrler, G.; Wu, N.-H.; Nitsche, A. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell 2000, 181, 271–280. [Google Scholar] [CrossRef]
- Iwata-Yoshikawa, N.; Okamura, T.; Shimizu, Y.; Hasegawa, H.; Takeda, M.; Nagata, N. TMPRSS2 contributes to virus spread and immunopathology in the airways of murine models after coronavirus infection. J. Virol. 2019, 93, e01815–e01818. [Google Scholar] [CrossRef] [Green Version]
- Lin, S.; Shen, R.; He, J.; Li, X.; Guo, X. Molecular modeling evaluation of the binding effect of ritonavir, lopinavir and darunavir to severe acute respiratory syndrome coronavirus 2 proteases. bioRxiv 2020. [Google Scholar] [CrossRef]
- Lung, J.; Lin, Y.S.; Yang, Y.H.; Chou, Y.L.; Shu, L.H.; Cheng, Y.C.; Liu, H.T.; Wu, C.Y. The potential chemical structure of anti-SARS-CoV-2 RNA-dependent RNA polymerase. J. Med. Virol. 2020, 92, 693–697. [Google Scholar] [CrossRef]
- Jiang, S.; Hillyer, C.; Du, L. Neutralizing antibodies against SARS-CoV-2 and other human coronaviruses. Trends Immunol. 2020, 41, 355–359. [Google Scholar] [CrossRef] [PubMed]
- Wu, C.-J.; Huang, H.-W.; Liu, C.-Y.; Hong, C.-F.; Chan, Y.-L. Inhibition of SARS-CoV replication by siRNA. Antivir. Res. 2005, 65, 45–48. [Google Scholar] [CrossRef] [PubMed]
- Liu, C.; Zhou, Q.; Li, Y.; Garner, L.V.; Watkins, S.P.; Carter, L.J.; Smoot, J.; Gregg, A.C.; Daniels, A.D.; Jervey, S. Research and development on therapeutic agents and vaccines for COVID-19 and related human coronavirus diseases. ACS Publ. 2020, 6, 315–331. [Google Scholar] [CrossRef] [PubMed]
- Monteil, V.; Kwon, H.; Prado, P.; Hagelkrüys, A.; Wimmer, R.A.; Stahl, M.; Leopoldi, A.; Garreta, E.; del Pozo, C.H.; Prosper, F. Inhibition of SARS-CoV-2 infections in engineered human tissues using clinical-grade soluble human ACE2. Cell 2020, 181, 905–913. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.; Yang, Y.; Zhang, C.; Huang, F.; Wang, F.; Yuan, J.; Wang, Z.; Li, J.; Li, J.; Feng, C. Clinical and biochemical indexes from 2019-nCoV infected patients linked to viral loads and lung injury. Sci. China Life Sci. 2020, 63, 364–374. [Google Scholar] [CrossRef] [Green Version]
- Pearce, A.K.; O’Reilly, R.K. Insights into active targeting of nanoparticles in drug delivery: Advances in clinical studies and design considerations for cancer nanomedicine. Bioconjugate Chem. 2019, 30, 2300–2311. [Google Scholar] [CrossRef]
- Villaverde, G.; Baeza, A. Targeting strategies for improving the efficacy of nanomedicine in oncology. Beilstein J. Nanotechnol. 2019, 10, 168–181. [Google Scholar] [CrossRef] [Green Version]
- Zhang, H.; Penninger, J.M.; Li, Y.; Zhong, N.; Slutsky, A.S. Angiotensin-converting enzyme 2 (ACE2) as a SARS-CoV-2 receptor: Molecular mechanisms and potential therapeutic target. Intensive Care Med. 2020, 46, 586–590. [Google Scholar] [CrossRef] [Green Version]
- Huang, L.; Sexton, D.J.; Skogerson, K.; Devlin, M.; Smith, R.; Sanyal, I.; Parry, T.; Kent, R.; Enright, J.; Wu, Q.-L. Novel peptide inhibitors of angiotensin-converting enzyme 2. J. Biol. Chem. 2003, 278, 15532–15540. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sun, Y.; Guo, F.; Zou, Z.; Li, C.; Hong, X.; Zhao, Y.; Wang, C.; Wang, H.; Liu, H.; Yang, P. Cationic nanoparticles directly bind angiotensin-converting enzyme 2 and induce acute lung injury in mice. Part. Fibre Toxicol. 2015, 12, 1–13. [Google Scholar] [CrossRef] [Green Version]
- Imai, Y.; Kuba, K.; Rao, S.; Huan, Y.; Guo, F.; Guan, B.; Yang, P.; Sarao, R.; Wada, T.; Leong-Poi, H. Angiotensin-converting enzyme 2 protects from severe acute lung failure. Nature 2005, 436, 112–116. [Google Scholar] [CrossRef]
- Huang, F.; Guo, J.; Zou, Z.; Liu, J.; Cao, B.; Zhang, S.; Li, H.; Wang, W.; Sheng, M.; Liu, S. Angiotensin II plasma levels are linked to disease severity and predict fatal outcomes in H7N9-infected patients. Nat. Commun. 2014, 5, 1–7. [Google Scholar] [CrossRef]
- Kuba, K.; Imai, Y.; Rao, S.; Gao, H.; Guo, F.; Guan, B.; Huan, Y.; Yang, P.; Zhang, Y.; Deng, W. A crucial role of angiotensin converting enzyme 2 (ACE2) in SARS coronavirus–induced lung injury. Nat. Med. 2005, 11, 875–879. [Google Scholar] [CrossRef]
- Feuillet, V.; Canard, B.; Trautmann, A. Combining antivirals and immunomodulators to fight COVID-19. Trends Immunol. 2020, 42, 31–44. [Google Scholar] [CrossRef]
- He, W.; Kapate, N.; Shields, C.V., IV; Mitragotri, S. Drug delivery to macrophages: A review of targeting drugs and drug carriers to macrophages for inflammatory diseases. Adv. Drug Deliv. Rev. 2020, 165, 15–40. [Google Scholar]
- Xiang, Q.-Y.; Wang, M.-T.; Chen, F.; Gong, T.; Jian, Y.-L.; Zhang, Z.-R.; Huang, Y. Lung-targeting delivery of dexamethasone acetate loaded solid lipid nanoparticles. Arch. Pharmacal Res. 2007, 30, 519–525. [Google Scholar] [CrossRef] [PubMed]
- Xiao, B.; Laroui, H.; Ayyadurai, S.; Viennois, E.; Charania, M.A.; Zhang, Y.; Merlin, D. Mannosylated bioreducible nanoparticle-mediated macrophage-specific TNF-α RNA interference for IBD therapy. Biomaterials 2013, 34, 7471–7482. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hu, G.; Guo, M.; Xu, J.; Wu, F.; Fan, J.; Huang, Q.; Yang, G.; Lv, Z.; Wang, X.; Jin, Y. Nanoparticles targeting macrophages as potential clinical therapeutic agents against cancer and inflammation. Front. Immunol. 2019, 10, 1998. [Google Scholar] [CrossRef]
- Shibu, E.S.; Hamada, M.; Murase, N.; Biju, V. Nanomaterials formulations for photothermal and photodynamic therapy of cancer. J. Photochem. Photobiol. C Photochem. Rev. 2013, 15, 53–72. [Google Scholar] [CrossRef]
- Agostinis, P.; Berg, K.; Cengel, K.A.; Foster, T.H.; Girotti, A.W.; Gollnick, S.O.; Hahn, S.M.; Hamblin, M.R.; Juzeniene, A.; Kessel, D. Photodynamic therapy of cancer: An update. CA A Cancer J. Clin. 2011, 61, 250–281. [Google Scholar] [CrossRef] [PubMed]
- Wiehe, A.; O’Brien, J.M.; Senge, M.O. Trends and targets in antiviral phototherapy. Photochem. Photobiol. Sci. 2019, 18, 2565–2612. [Google Scholar] [CrossRef]
- Wainwright, M. Local treatment of viral disease using photodynamic therapy. Int. J. Antimicrob. Agents 2003, 21, 510–520. [Google Scholar] [CrossRef]
- Kharkwal, G.B.; Sharma, S.K.; Huang, Y.Y.; Dai, T.; Hamblin, M.R. Photodynamic therapy for infections: Clinical applications. Lasers Surg. Med. 2011, 43, 755–767. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Costa, L.; Faustino, M.A.F.; Neves, M.G.P.; Cunha, Â.; Almeida, A. Photodynamic inactivation of mammalian viruses and bacteriophages. Viruses 2012, 4, 1034–1074. [Google Scholar] [CrossRef] [Green Version]
- Lim, M.E.; Lee, Y.-L.; Zhang, Y.; Chu, J.J.H. Photodynamic inactivation of viruses using upconversion nanoparticles. Biomaterials 2012, 33, 1912–1920. [Google Scholar] [CrossRef] [PubMed]
- Fekrazad, R. Photobiomodulation and antiviral photodynamic therapy as a possible novel approach in COVID-19 management. Photobiomodul. Photomed. Laser Surg. 2020, 38, 255–257. [Google Scholar] [CrossRef]
- Souri, M.; Soltani, M.; Kashkooli, F.M. Computational modeling of thermal combination therapies by magneto-ultrasonic heating to enhance drug delivery to solid tumors. Sci. Rep. 2021, 11, 1–12. [Google Scholar] [CrossRef]
- Kashkooli, F.M.; Soltani, M.; Momeni, M.M.; Rahmim, A. Enhanced drug delivery to solid tumors via drug-loaded nanocarriers: An image-based computational framework. Front. Oncol. 2021, 11, 1–19. [Google Scholar] [CrossRef]
- Souri, M.; Soltani, M.; Kashkooli, F.M.; Shahvandi, M.K. Engineered strategies to enhance tumor penetration of drug-loaded nanoparticles. J. Control. Release 2022, 341, 227–246. [Google Scholar] [CrossRef]
- Souri, M.; Soltani, M.; Kashkooli, F.M.; Shahvandi, M.K.; Chiani, M.; Shariati, F.S.; Mehrabi, M.R.; Munn, L.L. Towards principled design of cancer nanomedicine to accelerate clinical translation. Mater. Today Bio 2022, 13, 100208. [Google Scholar] [CrossRef] [PubMed]
- Joyce, E.; Fabre, A.; Mahon, N. Hydroxychloroquine cardiotoxicity presenting as a rapidly evolving biventricular cardiomyopathy: Key diagnostic features and literature review. Eur. Heart J. Acute Cardiovasc. Care 2013, 2, 77–83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rezaee, P.; Akbari, M.; Morad, R.; Koochaki, A.; Maaz, M.; Jamshidi, Z. First principle simulation of coated hydroxychloroquine on Ag, Au and Pt nanoparticle as a potential candidate for treatment of SARS-CoV-2 (COVID-19). arXiv 2020, arXiv:2006.02343. [Google Scholar]
- Dormont, F.; Brusini, R.; Cailleau, C.; Reynaud, F.; Peramo, A.; Gendron, A.; Mougin, J.; Gaudin, F.; Varna, M.; Couvreur, P. Squalene-based multidrug nanoparticles for improved mitigation of uncontrolled inflammation in rodents. Sci. Adv. 2020, 6, eaaz5466. [Google Scholar] [CrossRef]
- Li, Y.; Lin, Z.; Guo, M.; Xia, Y.; Zhao, M.; Wang, C.; Xu, T.; Chen, T.; Zhu, B. Inhibitory activity of selenium nanoparticles functionalized with oseltamivir on H1N1 influenza virus. Int. J. Nanomed. 2017, 12, 5733. [Google Scholar] [CrossRef] [Green Version]
- Zhong, J.; Xia, Y.; Hua, L.; Liu, X.; Xiao, M.; Xu, T.; Zhu, B.; Cao, H. Functionalized selenium nanoparticles enhance the anti-EV71 activity of oseltamivir in human astrocytoma cell model. Artif. Cells Nanomed. Biotechnol. 2019, 47, 3485–3491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Beaucourt, S.; Vignuzzi, M. Ribavirin: A drug active against many viruses with multiple effects on virus replication and propagation. Molecular basis of ribavirin resistance. Curr. Opin. Virol. 2014, 8, 10–15. [Google Scholar] [CrossRef]
- Arabi, Y.M.; Shalhoub, S.; Mandourah, Y.; Al-Hameed, F.; Al-Omari, A.; Al Qasim, E.; Jose, J.; Alraddadi, B.; Almotairi, A.; Al Khatib, K. Ribavirin and interferon therapy for critically ill patients with middle east respiratory syndrome: A multicenter observational study. Clin. Infect. Dis. 2020, 70, 1837–1844. [Google Scholar] [CrossRef] [Green Version]
- Lin, Z.; Li, Y.; Gong, G.; Xia, Y.; Wang, C.; Chen, Y.; Hua, L.; Zhong, J.; Tang, Y.; Liu, X. Restriction of H1N1 influenza virus infection by selenium nanoparticles loaded with ribavirin via resisting caspase-3 apoptotic pathway. Int. J. Nanomed. 2018, 13, 5787. [Google Scholar] [CrossRef] [Green Version]
- Klębowski, B.; Depciuch, J.; Parlińska-Wojtan, M.; Baran, J. Applications of noble metal-based nanoparticles in medicine. Int. J. Mol. Sci. 2018, 19, 4031. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ahmed, E.M.; Solyman, S.M.; Mohamed, N.; Boseila, A.A.; Hanora, A. Antiviral activity of Ribavirin nano-particles against measles virus. Cell. Mol. Biol. 2018, 64, 24–32. [Google Scholar] [CrossRef] [PubMed]
- Gurunathan, S.; Qasim, M.; Choi, Y.; Do, J.T.; Park, C.; Hong, K.; Kim, J.-H.; Song, H. Antiviral potential of nanoparticles—Can nanoparticles fight against coronaviruses? Nanomaterials 2020, 10, 1645. [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] [Green Version]
- Has, C.; Sunthar, P. A comprehensive review on recent preparation techniques of liposomes. J. Liposome Res. 2020, 30, 336–365. [Google Scholar] [CrossRef]
- Baldino, L.; Reverchon, E. Niosomes formation using a continuous supercritical CO2 assisted process. J. CO2 Util. 2021, 52, 101669. [Google Scholar] [CrossRef]
- Roces, C.B.; Port, E.C.; Daskalakis, N.N.; Watts, J.A.; Aylott, J.W.; Halbert, G.W.; Perrie, Y. Rapid scale-up and production of active-loaded PEGylated liposomes. Int. J. Pharm. 2020, 586, 119566. [Google Scholar] [CrossRef]
- Chaves, M.A.; Baldino, L.; Pinho, S.C.; Reverchon, E. Co-encapsulation of curcumin and vitamin D3 in mixed phospholipid nanoliposomes using a continuous supercritical CO2 assisted process. J. Taiwan Inst. Chem. Eng. 2022, 132, 104120. [Google Scholar] [CrossRef]
- Hu, C.-M.J.; Chen, Y.-T.; Fang, Z.-S.; Chang, W.-S.; Chen, H.-W. Antiviral efficacy of nanoparticulate vacuolar ATPase inhibitors against influenza virus infection. Int. J. Nanomed. 2018, 13, 8579. [Google Scholar] [CrossRef] [Green Version]
- Hong, S.-S.; Oh, K.T.; Choi, H.-G.; Lim, S.-J. Liposomal formulations for nose-to-brain delivery: Recent advances and future perspectives. Pharmaceutics 2019, 11, 540. [Google Scholar] [CrossRef] [Green Version]
- Alsarra, I.A.; Hamed, A.Y.; Alanazi, F.K. Acyclovir liposomes for intranasal systemic delivery: Development and pharmacokinetics evaluation. Drug Deliv. 2008, 15, 313–321. [Google Scholar] [CrossRef] [PubMed]
- Ghasemiyeh, P.; Mohammadi-Samani, S. Solid lipid nanoparticles and nanostructured lipid carriers as novel drug delivery systems: Applications, advantages and disadvantages. Res. Pharm. Sci. 2018, 13, 288. [Google Scholar]
- Desfarges, S.; Ciuffi, A. Viral Integration and Consequences on Host Gene Expression, Viruses: Essential Agents of Life; Springer: Berlin/Heidelberg, Germany, 2012; pp. 147–175. [Google Scholar]
- Bayat, H.; Naderi, F.; Khan, A.H.; Memarnejadian, A.; Rahimpour, A. The impact of CRISPR-Cas system on antiviral therapy. Adv. Pharm. Bull. 2018, 8, 591. [Google Scholar] [CrossRef]
- Abbott, T.R.; Dhamdhere, G.; Liu, Y.; Lin, X.; Goudy, L.; Zeng, L.; Chemparathy, A.; Chmura, S.; Heaton, N.S.; Debs, R. Development of CRISPR as a prophylactic strategy to combat novel coronavirus and influenza. bioRxiv 2020. [Google Scholar] [CrossRef] [Green Version]
- Li, B.; Clohisey, S.M.; Chia, B.S.; Wang, B.; Cui, A.; Eisenhaure, T.; Schweitzer, L.D.; Hoover, P.; Parkinson, N.J.; Nachshon, A. Genome-wide CRISPR screen identifies host dependency factors for influenza A virus infection. Nat. Commun. 2020, 11, 1–18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sanjuán, R.; Domingo-Calap, P. Mechanisms of viral mutation. Cell. Mol. Life Sci. 2016, 73, 4433–4448. [Google Scholar] [CrossRef] [Green Version]
- Burmistrz, M.; Krakowski, K.; Krawczyk-Balska, A. RNA-targeting CRISPR–Cas systems and their applications. Int. J. Mol. Sci. 2020, 21, 1122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Abudayyeh, O.O.; Gootenberg, J.S.; Essletzbichler, P.; Han, S.; Joung, J.; Belanto, J.J.; Verdine, V.; Cox, D.B.; Kellner, M.J.; Regev, A. RNA targeting with CRISPR–Cas13. Nature 2017, 550, 280–284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Timin, A.S.; Muslimov, A.R.; Lepik, K.V.; Epifanovskaya, O.S.; Shakirova, A.I.; Mock, U.; Riecken, K.; Okilova, M.V.; Sergeev, V.S.; Afanasyev, B.V. Efficient gene editing via non-viral delivery of CRISPR–Cas9 system using polymeric and hybrid microcarriers. Nanomed. Nanotechnol. Biol. Med. 2018, 14, 97–108. [Google Scholar] [CrossRef] [PubMed]
- Blenke, E.O.; Evers, M.J.; Mastrobattista, E.; van der Oost, J. CRISPR-Cas9 gene editing: Delivery aspects and therapeutic potential. J. Control. Release 2016, 244, 139–148. [Google Scholar] [CrossRef] [PubMed]
- Finn, J.D.; Smith, A.R.; Patel, M.C.; Shaw, L.; Youniss, M.R.; van Heteren, J.; Dirstine, T.; Ciullo, C.; Lescarbeau, R.; Seitzer, J. A single administration of CRISPR/Cas9 lipid nanoparticles achieves robust and persistent in vivo genome editing. Cell Rep. 2018, 22, 2227–2235. [Google Scholar] [CrossRef] [Green Version]
- Jiang, C.; Mei, M.; Li, B.; Zhu, X.; Zu, W.; Tian, Y.; Wang, Q.; Guo, Y.; Dong, Y.; Tan, X. A non-viral CRISPR/Cas9 delivery system for therapeutically targeting HBV DNA and pcsk9 in vivo. Cell Res. 2017, 27, 440–443. [Google Scholar] [CrossRef] [PubMed]
- Soltani, M.; Moradi Kashkooli, F.; Souri, M.; Zare Harofte, S.; Harati, T.; Khadem, A.; Haeri Pour, M.; Raahemifar, K. Enhancing Clinical Translation of Cancer Using Nanoinformatics. Cancers 2021, 13, 2481. [Google Scholar] [CrossRef] [PubMed]
- Kashkooli, F.M.; Soltani, M.; Souri, M.; Meaney, C.; Kohandel, M. Nexus between in silico and in vivo models to enhance clinical translation of nanomedicine. Nano Today 2021, 36, 101057. [Google Scholar] [CrossRef]
- Kashkooli, F.M.; Soltani, M.; Rezaeian, M.; Meaney, C.; Hamedi, M.-H.; Kohandel, M. Effect of vascular normalization on drug delivery to different stages of tumor progression: In-silico analysis. J. Drug Deliv. Sci. Technol. 2020, 60, 101989. [Google Scholar] [CrossRef]
- Soltani, M.; Souri, M.; Moradi Kashkooli, F. Effects of hypoxia and nanocarrier size on pH-responsive nano-delivery system to solid tumors. Sci. Rep. 2021, 11, 1–12. [Google Scholar]
- Subudhi, S.; Verma, A.; Patel, A.B.; Hardin, C.C.; Khandekar, M.J.; Lee, H.; McEvoy, D.; Stylianopoulos, T.; Munn, L.L.; Dutta, S. Comparing machine learning algorithms for predicting ICU admission and mortality in COVID-19. NPJ Digit. Med. 2021, 4, 1–7. [Google Scholar] [CrossRef]
- Voutouri, C.; Nikmaneshi, M.R.; Hardin, C.C.; Patel, A.B.; Verma, A.; Khandekar, M.J.; Dutta, S.; Stylianopoulos, T.; Munn, L.L.; Jain, R.K. In silico dynamics of COVID-19 phenotypes for optimizing clinical management. Proc. Natl. Acad. Sci. USA 2021, 118, 1–8. [Google Scholar] [CrossRef]
- Ostaszewski, M.; Mazein, A.; Gillespie, M.E.; Kuperstein, I.; Niarakis, A.; Hermjakob, H.; Pico, A.R.; Willighagen, E.L.; Evelo, C.T.; Hasenauer, J. COVID-19 Disease Map, building a computational repository of SARS-CoV-2 virus-host interaction mechanisms. Sci. Data 2020, 7, 1–4. [Google Scholar]
- Weiss, C.; Carriere, M.; Fusco, L.; Capua, I.; Regla-Nava, J.A.; Pasquali, M.; Scott, J.A.; Vitale, F.; Unal, M.A.; Mattevi, C. Toward nanotechnology-enabled approaches against the COVID-19 pandemic. ACS Nano 2020, 14, 6383–6406. [Google Scholar] [CrossRef]
- Ahmadzadeh, M.; Farokhi, E.; Shams, M. Investigating the effect of air conditioning on the distribution and transmission of Covid-19 virus particles. J. Clean. Prod. 2021, 316, 128147. [Google Scholar] [CrossRef] [PubMed]
- Moradi Kashkooli, F.; Sefidgar, M.; Soltani, M.; Anbari, S.; Shahandashti, S.-A.; Zargar, B. Numerical assessment of an air cleaner device under different working conditions in an indoor environment. Sustainability 2021, 13, 369. [Google Scholar] [CrossRef]
- Moradi Kashkooli, F.; Soltani, M.; Zargar, B.; Ijaz, M.K.; Taatizadeh, E.; Sattar, S.A. Analysis of an indoor air decontamination device inside an aerobiology chamber: A numerical-experimental study. Air Qual. Atmos. Health 2020, 13, 281–288. [Google Scholar] [CrossRef]
- Zargar, B.; Kashkooli, F.M.; Soltani, M.; Wright, K.E.; Ijaz, M.K.; Sattar, S.A. Mathematical modeling and simulation of bacterial distribution in an aerobiology chamber using computational fluid dynamics. Am. J. Infect. Control. 2016, 44, S127–S137. [Google Scholar] [CrossRef]
- Hwang, W.; Lei, W.; Katritsis, N.M.; MacMahon, M.; Chapman, K.; Han, N. Current and prospective computational approaches and challenges for developing COVID-19 vaccines. Adv. Drug Deliv. Rev. 2021, 172, 249–274. [Google Scholar] [CrossRef] [PubMed]
- Sohail, M.S.; Ahmed, S.F.; Quadeer, A.A.; McKay, M.R. In silico T cell epitope identification for SARS-CoV-2: Progress and perspectives. Adv. Drug Deliv. Rev. 2021, 171, 29–47. [Google Scholar] [CrossRef] [PubMed]
- Moradi Kashkooli, F.; Soltani, M. Evaluation of solid tumor response to sequential treatment cycles via a new computational hybrid approach. Sci. Rep. 2021, 11, 1–15. [Google Scholar] [CrossRef]
- Han, Y.; Král, P. Computational design of ACE2-based peptide inhibitors of SARS-CoV-2. ACS Nano 2020, 14, 5143–5147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Developer | Structure | Characteristics | Ref. |
---|---|---|---|
SHEPROS SDN BHD | A nano silver-based multipurpose disinfectant using a nano-colloidal technique | Environmentally friendly, non-irritating, and non-foaming properties to fight viruses, germs, and fungi | [56] |
Nano Tech Surface | A nano-sterilizing based on silver ions and titanium dioxide | Environmentally friendly, non-irritating, High disinfection potential | [57] |
Weinnovate Biosolutions | A nano-sterilizing based on silver nanoparticle solution | Low toxic effects, High disinfection potential, non-irritating, | [58] |
Tamil Nadu University | A natural nanomaterial-based disinfectant containing hydrogen peroxide and alcohol molecules | High environmental friendliness, non-corrosive, non-toxic, cost-effective | [8] |
Defence Institute of Advanced Technologies (DIAT) in Pune | A disinfectant called Ananya based on a water spray using nanomaterial | Able to adhere to fabric, plastics, and metal surfaces, and disinfection effects about 6 months. | [59] |
Design.123 | A disinfect based on nanopolymer | High disinfection potential, Fast performance in inactivating viruses | [60] |
Hong Kong University of Science and Technology | A temperature-responsive nanopolymer-based antimicrobial | Slowly releasing disinfectants, high lifespan | [61] |
EnvisionSQ | An antiviral nanocoating | High lifespan, high ability to kill viruses | [62] |
NanoTouch Materials | A nano-coating based on mineral nanomaterials | High disinfection potential, non-corrosive | [8] |
Developer | Structure | Characteristics | Ref. |
---|---|---|---|
Promethean Particles Ltd. | A fabric based on copper nanoparticles embedded in a polymer matrix | Enhancing antiviral and antimicrobial properties | [85] |
ZEN Graphene Solution Ltd. | silver-nanoparticle-modified graphene oxide nanocomposite membranes | Virus capture and killing, virucidal | [86] |
Sonovia Ltd. | A fabric based on zinc oxide nanoparticles | Antiviral properties, and can be washed for reuse | [86,87] |
Master Dynamic Limited | A coating based on nanodiamonds | High anti-virus performance | [86] |
X.TiO2 Inc. (XTI) | TiO2Ag-based facemasks | Able to kill 99.99% of viruses under zero light conditions | [86] |
Verdex Technologies Inc. | Nanocomposite membranes | Virus removal, breathable | [86] |
Respilon | Nanofiber membranes | High virus removal | [88] |
Yamashin-filter Corp. | Polymer-based nanofiber membranes | High virus trapping, virus removal | [89,90] |
LIGC Application Ltd. | Graphene-based technology | Reusable, self-sterilizing, antiviral activity | [86] |
Balagna et al. | Silver nanoclusters/silica composite sputtered coating | Virucidal, increased lifetime of masks and filter media | [91] |
Strain of Coronavirus | Vaccine | Nano Component | Mechanism | Ref. |
---|---|---|---|---|
Influenza virus | Pulmonary surfactant biomimetic nanoparticles | Biomimetic liposomes | Potentiate heterosubtypic influenza immunity | [116] |
MERS-CoV | Purified coronavirus spike protein nanoparticles | Spike nanoparticles | Induce coronavirus neutralizing antibodies in mice | [117] |
SARS-CoV | Gold nanoparticle-adjuvanted S protein | Gold nanoparticles | Induced antigen-specific IgG response | [118] |
SARS subunit vaccine | Peptide nanoparticles | Neutralizing antibody and strong humoral response | [119] | |
SARS-CoV-2 | Novel lipid nanoparticle (LNP)- encapsulated mRNA based vaccine | Lipid nanoparticles | Recombined mRNA of the S protein | [120] |
LNP-encapsulated mRNA encoding RBD | Lipid nanoparticles | RBD mRNA reacted strongly with a SARS-CoV-2 RBD specific antibody | [121] | |
LNP-nCoV-saRNA | Lipid nanoparticles | Robust neutralization of a pseudovirus, proportional to quantity of specific IgG | [122] | |
Self-replicating RNA based therapeutic vaccine (LUNAR-COV19 STARR™) | RNA nanoparticles delivery systems | Enhanced adaptive cellular (CD8+ cells) and balanced (Th1/Th2) immune response | [123] | |
virus-like nanoparticles (VLNP) | Protein nanoparticle scaffold | Promotes B cell immune response | [124] | |
T-COVID | Adenovirus | Decreased cellular inflammation and lower concentrations of IL-6 | [125] |
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
Souri, M.; Chiani, M.; Farhangi, A.; Mehrabi, M.R.; Nourouzian, D.; Raahemifar, K.; Soltani, M. Anti-COVID-19 Nanomaterials: Directions to Improve Prevention, Diagnosis, and Treatment. Nanomaterials 2022, 12, 783. https://doi.org/10.3390/nano12050783
Souri M, Chiani M, Farhangi A, Mehrabi MR, Nourouzian D, Raahemifar K, Soltani M. Anti-COVID-19 Nanomaterials: Directions to Improve Prevention, Diagnosis, and Treatment. Nanomaterials. 2022; 12(5):783. https://doi.org/10.3390/nano12050783
Chicago/Turabian StyleSouri, Mohammad, Mohsen Chiani, Ali Farhangi, Mohammad Reza Mehrabi, Dariush Nourouzian, Kaamran Raahemifar, and M. Soltani. 2022. "Anti-COVID-19 Nanomaterials: Directions to Improve Prevention, Diagnosis, and Treatment" Nanomaterials 12, no. 5: 783. https://doi.org/10.3390/nano12050783
APA StyleSouri, M., Chiani, M., Farhangi, A., Mehrabi, M. R., Nourouzian, D., Raahemifar, K., & Soltani, M. (2022). Anti-COVID-19 Nanomaterials: Directions to Improve Prevention, Diagnosis, and Treatment. Nanomaterials, 12(5), 783. https://doi.org/10.3390/nano12050783