Aptamer Sandwich Assay for the Detection of SARS-CoV-2 Spike Protein Antigen
- Marketa SvobodovaMarketa SvobodovaINTERFIBIO Research Group, Departament d’Enginyeria Química, Universitat Rovira i Virgili, Avinguda Països Catalans 26, Tarragona 43007, SpainMore by Marketa Svobodova
- ,
- Vasso SkouridouVasso SkouridouINTERFIBIO Research Group, Departament d’Enginyeria Química, Universitat Rovira i Virgili, Avinguda Països Catalans 26, Tarragona 43007, SpainMore by Vasso Skouridou
- ,
- Miriam Jauset-RubioMiriam Jauset-RubioINTERFIBIO Research Group, Departament d’Enginyeria Química, Universitat Rovira i Virgili, Avinguda Països Catalans 26, Tarragona 43007, SpainMore by Miriam Jauset-Rubio
- ,
- Irene ViéitezIrene ViéitezRare Diseases & Pediatric Medicine Research Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-Uvigo, Vigo 36213, SpainMore by Irene Viéitez
- ,
- Alberto Fernández-VillarAlberto Fernández-VillarPneumology Service, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-Uvigo, Vigo 36213, SpainMore by Alberto Fernández-Villar
- ,
- Jorge Julio Cabrera AlvargonzalezJorge Julio Cabrera AlvargonzalezMicrobiology Service, Galicia Sur Health Research Institute (IIS Galicia Sur). SERGAS-UVigo, Vigo 36213, Spain
- ,
- Eva PovedaEva PovedaGroup of Virology and Pathogenesis, Galicia Sur Health Research Institute (IIS Galicia Sur)-Complexo Hospitalario Universitario de Vigo, SERGAS-UVigo, Vigo 36213, SpainMore by Eva Poveda
- ,
- Clara Benavent BofillClara Benavent BofillLaboratori Clinic ICS Camp de Tarragona, Hospital Universitari de Tarragona Joan XXIII, Avda. Dr. Mallafré Guasch, 4, Tarragona 43007, SpainMore by Clara Benavent Bofill
- ,
- Teresa SansTeresa SansLaboratori Clinic ICS Camp de Tarragona, Hospital Universitari de Tarragona Joan XXIII, Avda. Dr. Mallafré Guasch, 4, Tarragona 43007, SpainMore by Teresa Sans
- ,
- Abdulaziz BashammakhAbdulaziz BashammakhDepartment of Chemistry, Faculty of Science, King Abdulaziz University, Jeddah 80215, Kingdom of Saudi ArabiaMore by Abdulaziz Bashammakh
- ,
- Abdulrahman O. AlyoubiAbdulrahman O. AlyoubiDepartment of Chemistry, Faculty of Science, King Abdulaziz University, Jeddah 80215, Kingdom of Saudi ArabiaMore by Abdulrahman O. Alyoubi
- , and
- Ciara K. O’Sullivan*Ciara K. O’Sullivan*Email: [email protected]INTERFIBIO Research Group, Departament d’Enginyeria Química, Universitat Rovira i Virgili, Avinguda Països Catalans 26, Tarragona 43007, SpainInstitució Catalana de Recerca i Estudis Avancats (ICREA), Passeig Lluís Companys 23, Barcelona 08010, SpainMore by Ciara K. O’Sullivan
Abstract

The novel severe acute respiratory syndrome coronavirus (SARS-CoV-2) emerged at the end of 2019, resulting in the ongoing COVID-19 pandemic. The high transmissibility of the virus and the substantial number of asymptomatic individuals have led to an exponential rise in infections worldwide, urgently requiring global containment strategies. Reverse transcription-polymerase chain reaction is the gold standard for the detection of SARS-CoV-2 infections. Antigen tests, targeting the spike (S) or nucleocapsid (N) viral proteins, are considered as complementary tools. Despite their shortcomings in terms of sensitivity and specificity, antigen tests could be deployed for the detection of potentially contagious individuals with high viral loads. In this work, we sought to develop a sandwich aptamer-based assay for the detection of the S protein of SARS-CoV-2. A detailed study on the binding properties of aptamers to the receptor-binding domain of the S protein in search of aptamer pairs forming a sandwich is presented. Screening of aptamer pairs and optimization of assay conditions led to the development of a laboratory-based sandwich assay able to detect 21 ng/mL (270 pM) of the protein with negligible cross-reactivity with the other known human coronaviruses. The detection of 375 pg of the protein in viral transport medium demonstrates the compatibility of the assay with clinical specimens. Finally, successful detection of the S antigen in nasopharyngeal swab samples collected from suspected patients further establishes the suitability of the assay for screening purposes as a complementary tool to assist in the control of the pandemic.
This publication is licensed under
License Summary*
You are free to share (copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share (copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share (copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share (copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share (copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
Introduction
Figure 1

Figure 1. Detection of the SARS-CoV-2 S antigen with the aptamer sandwich assay.
Results and Discussion
Affinity and Specificity of the Aptamers
S1 | RBD | |||
---|---|---|---|---|
KD (nM) | R2 | KD (nM) | R2 | |
Apt1 | 10.2 ± 2.7 | 0.9875 | 39.0 ± 11.2 | 0.9903 |
Apt2 | 0.2 ± 0.1 | 0.9768 | 7.7 ± 1.4 | 0.9887 |
Apt3 | 5.3 ± 0.5 | 0.9976 | 10.9 ± 2.9 | 0.9706 |
Apt4 | 3.3 ± 0.7 | 0.9869 | 34.6 ± 17.8 | 0.9851 |
Apt5 | 11.3 ± 6.1 | 0.9810 | 29.8 ± 17.8 | 0.9713 |
Apt6 | 3.4 ± 1.1 | 0.9879 | 32.2 ± 7.3 | 0.9701 |
Apt1C | 8.1 ± 1.0 | 0.9939 | 21.0 ± 2.7 | 0.9916 |
Apt4C | 11.1 ± 2.7 | 0.9623 | 20.8 ± 5.4 | 0.9792 |
Figure 2

Figure 2. Specificity of the aptamers evaluated by direct ELAA. The relative binding of each aptamer was calculated using constant concentrations of aptamers (200 nM) and S1 proteins from the different HCoV’s (2 μg/mL). The error bars correspond to the standard deviation from duplicate measurements.
Screening of Aptamer Pairs
Figure 3

Figure 3. Screening of aptamer pairs for the detection of SARS-CoV-2 S1 protein. Each thiolated capture aptamer (AptxT) was analyzed in combination with each of the biotinylated reporter aptamers (AptxB) using a constant concentration of S1. No protein was added for the control samples. The number refers to the number of the aptamer tested, T indicates the thiolated aptamer and B indicates the biotinylated aptamer.
Figure 4

Figure 4. Sensitivity of the sandwich assays for SARS-CoV-2 S1 detection using different combinations of aptamer pairs.
Figure 5

Figure 5. Specificity of the sandwich assays using different aptamer pairs and S1 from different HCoV’s. Each protein was used at 2 μg/mL, and the error bars indicate the standard deviations from duplicate measurements.
Sensitivity of the Apt1T/Apt5B Pair for Sandwich Detection of SARS-CoV-2 S1
Figure 6

Figure 6. Sensitivity of the sandwich assay based on the Apt1T/Apt5B aptamer pair in aptamer binding buffer and in VTM.
Application of the Assay to Clinical Samples
test (target) | result | number of samples |
---|---|---|
PCR (viral RNA) | positive | 45 |
negative | 5 | |
aptamer sandwich assay (S antigen) | true positive | 36 |
false-positive | 3 | |
true negative | 1 | |
false-negative | 9 |
Conclusions
Experimental Section
Materials
Evaluation of the Binding Properties of the Aptamers with a Direct Enzyme-Linked Aptamer Assay
Screening of Aptamer Pairs for Sandwich ELAA Development
Optimization of Sandwich Assay Conditions for S1 Detection with the Apt1T/Apt5B Aptamer Pair
Detection of the S Antigen in NP Swab Samples
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsomega.1c05521.
Recombinant proteins and aptamers used in this work; biosensors and assays developed for the detection of the SARS-CoV-2 S antigen; binding studies by direct ELAA to monitor S proteins interactions with aptamers; checkerboard titration of concentrations of capture and detection aptamers to optimize aptamer sandwich assay; sensitivity of the sandwich aptamer assay at different concentrations of reporter aptamer; and SARS-CoV-2 RNA detection in NP swab samples by droplet digital PCR analysis (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgments
We would like to thank all the members of COHVID-GS and IISGS Biobank. Members of COHVID-GS (Galicia Sur Health Research Institute): Alejandro Araujo, Jorge Julio Cabrera, Víctor del Campo, Manuel Crespo, Alberto Fernández, Beatriz Gil de Araujo, Carlos Gómez, Virginia Leiro, María Rebeca Longueira, Ana López-Domínguez, José Ramón Lorenzo, María Marcos, Alexandre Pérez, María Teresa Pérez, Lucia Patiño, Sonia Pérez, Silvia Pérez-Fernández, E.P., Cristina Ramos, Benito Regueiro, Cristina Retresas, Tania Rivera, Olga Souto, Isabel Taboada, Susana Teijeira, María Torres, Vanesa Val, and I.V.é
References
This article references 67 other publications.
- 1Weiss, S. R. Forty years with coronaviruses. J. Exp. Med. 2020, 217, e20200537 DOI: 10.1084/jem.20200537[Crossref], [PubMed], [CAS], Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXovFSgtLY%253D&md5=eb058fb12d70e155c40213f30c36ab66Forty years with coronavirusesWeiss, Susan R.Journal of Experimental Medicine (2020), 217 (5), e20200537CODEN: JEMEAV; ISSN:1540-9538. (Rockefeller University Press)A review. I have been researching coronaviruses for more than forty years. This viewpoint summarizes some of the major findings in corona virus research made before the SARS epidemic and how they inform current research on the newly emerged SARS-CoV-2.
- 2He, F.; Deng, Y.; Li, W. Coronavirus disease 2019: What we know?. J. Med. Virol. 2020, 92, 719– 725, DOI: 10.1002/jmv.25766[Crossref], [PubMed], [CAS], Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXnsVWqtLc%253D&md5=4b071ade1e6bc6b8a70b2d389a93bc22Coronavirus disease 2019: What we know?He, Feng; Deng, Yu; Li, WeinaJournal of Medical Virology (2020), 92 (7), 719-725CODEN: JMVIDB; ISSN:0146-6615. (Wiley-Blackwell)A review. In late Dec. 2019, a cluster of unexplained pneumonia cases has been reported in Wuhan, China. A few days later, the causative agent of this mysterious pneumonia was identified as a novel coronavirus. This causative virus has been temporarily named as severe acute respiratory syndrome coronavirus 2 and the relevant infected disease has been named as coronavirus disease 2019 (COVID-19) by the World Health Organization, resp. The COVID-19 epidemic is spreading in China and all over the world now. The purpose of this review is primarily to review the pathogen, clin. features, diagnosis, and treatment of COVID-19, but also to comment briefly on the epidemiol. and pathol. based on the current evidence.
- 3Zhu, Z.; Lian, X.; Su, X.; Wu, W.; Marraro, G. A.; Zeng, Y. From SARS and MERS to COVID-19: a brief summary and comparison of severe acute respiratory infections caused by three highly pathogenic human coronaviruses. Respir. Res. 2020, 21, 224, DOI: 10.1186/s12931-020-01479-w[Crossref], [PubMed], [CAS], Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhslWqtrfE&md5=7c9a3bd77971d16156df4aadb70a2a72From SARS and MERS to COVID-19: a brief summary and comparison of severe acute respiratory infections caused by three highly pathogenic human coronavirusesZhu, Zhixing; Lian, Xihua; Su, Xiaoshan; Wu, Weijing; Marraro, Giuseppe A.; Zeng, YimingRespiratory Research (2020), 21 (1), 224CODEN: RREEBZ; ISSN:1465-993X. (BioMed Central Ltd.)A review. Within 2 decades, there have emerged 3 highly pathogenic and deadly human coronaviruses, namely SARS-CoV, MERS-CoV, and SARS-CoV-2. The economic burden and health threats caused by these coronaviruses are extremely dreadful and getting more serious as the increasing no. of global infections and attributed deaths of SARS-CoV-2 and MERS-CoV. Unfortunately, specific medical countermeasures for these hCoVs remain absent. Moreover, the fast spread of misinformation about the ongoing SARS-CoV-2 pandemic uniquely places the virus alongside an annoying infodemic and causes unnecessary worldwide panic. SARS-CoV-2 shares many similarities with SARS-CoV and MERS-CoV, certainly, obvious differences exist as well. Lessons learned from SARS-CoV and MERS-CoV, timely updated information of SARS-CoV-2 and MERS-CoV, and summarized specific knowledge of these hCoVs are extremely invaluable for effectively and efficiently contain the outbreak of SARS-CoV-2 and MERS-CoV. By gaining a deeper understanding of hCoVs and the illnesses caused by them, we can bridge knowledge gaps, provide cultural weapons for fighting and controlling the spread of MERS-CoV and SARS-CoV-2, and prep. effective and robust defense lines against hCoVs that may emerge or reemerge in the future. To this end, the state-of-the-art knowledge and comparing the biol. features of these lethal hCoVs and the clin. characteristics of illnesses caused by them are systematically summarized in the review.
- 4Gorbalenya, A. E.; Baker, S. C.; Baric, R. S.; de Groot, R. J.; Drosten, C.; Gulyaeva, A. A.; Haagmans, B. L.; Lauber, C.; Leontovich, A. M.; Neuman, B. W.; Penzar, D.; Perlman, S.; Poon, L. L. M.; Samborskiy, D. V.; Sidorov, I. A.; Sola, I.; Ziebuhr, J. The species Severe Acute Respiratory Syndrome-related coronavirus: classifying 2019-nCoV and naming it SARS-CoV-2. Nat. Microbiol. 2020, 5, 536– 544, DOI: 10.1038/s41564-020-0695-z
- 5Wrapp, 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, DOI: 10.1126/science.abb2507[Crossref], [PubMed], [CAS], Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXkvFemt70%253D&md5=27d08cbb9a43d1da051a8a92a9f68aa5Cryo-EM structure of the 2019-nCoV spike in the prefusion conformationWrapp, Daniel; Wang, Nianshuang; Corbett, Kizzmekia S.; Goldsmith, Jory A.; Hsieh, Ching-Lin; Abiona, Olubukola; Graham, Barney S.; McLellan, Jason S.Science (Washington, DC, United States) (2020), 367 (6483), 1260-1263CODEN: SCIEAS; ISSN:1095-9203. (American Association for the Advancement of Science)The outbreak of a novel coronavirus (2019-nCoV) represents a pandemic threat that has been declared a public health emergency of international concern. The CoV spike (S) glycoprotein is a key target for vaccines, therapeutic antibodies, and diagnostics. To facilitate medical countermeasure development, we detd. a 3.5-angstrom-resoln. cryo-electron microscopy structure of the 2019-nCoV S trimer in the prefusion conformation. The predominant state of the trimer has one of the three receptor-binding domains (RBDs) rotated up in a receptor-accessible conformation. We also provide biophys. and structural evidence that the 2019-nCoV S protein binds angiotensin-converting enzyme 2 (ACE2) with higher affinity than does severe acute respiratory syndrome (SARS)-CoV S. Addnl., we tested several published SARS-CoV RBD-specific monoclonal antibodies and found that they do not have appreciable binding to 2019-nCoV S, suggesting that antibody cross-reactivity may be limited between the two RBDs. The structure of 2019-nCoV S should enable the rapid development and evaluation of medical countermeasures to address the ongoing public health crisis.
- 6Wang, N.; Shang, J.; Jiang, S.; Du, L. Subunit vaccines against emerging pathogenic human coronaviruses. Front. Microbiol. 2020, 11, 298, DOI: 10.3389/fmicb.2020.00298[Crossref], [PubMed], [CAS], Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB38zislyluw%253D%253D&md5=0bc5d7ff9839b3cc6678b0d3bb57a56fSubunit Vaccines Against Emerging Pathogenic Human CoronavirusesWang Ning; Jiang Shibo; Du Lanying; Shang Jian; Jiang ShiboFrontiers in microbiology (2020), 11 (), 298 ISSN:1664-302X.Seven coronaviruses (CoVs) have been isolated from humans so far. Among them, three emerging pathogenic CoVs, including severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome coronavirus (MERS-CoV), and a newly identified CoV (2019-nCoV), once caused or continue to cause severe infections in humans, posing significant threats to global public health. SARS-CoV infection in humans (with about 10% case fatality rate) was first reported from China in 2002, while MERS-CoV infection in humans (with about 34.4% case fatality rate) was first reported from Saudi Arabia in June 2012. 2019-nCoV was first reported from China in December 2019, and is currently infecting more than 70000 people (with about 2.7% case fatality rate). Both SARS-CoV and MERS-CoV are zoonotic viruses, using bats as their natural reservoirs, and then transmitting through intermediate hosts, leading to human infections. Nevertheless, the intermediate host for 2019-nCoV is still under investigation and the vaccines against this new CoV have not been available. Although a variety of vaccines have been developed against infections of SARS-CoV and MERS-CoV, none of them has been approved for use in humans. In this review, we have described the structure and function of key proteins of emerging human CoVs, overviewed the current vaccine types to be developed against SARS-CoV and MERS-CoV, and summarized recent advances in subunit vaccines against these two pathogenic human CoVs. These subunit vaccines are introduced on the basis of full-length spike (S) protein, receptor-binding domain (RBD), non-RBD S protein fragments, and non-S structural proteins, and the potential factors affecting these subunit vaccines are also illustrated. Overall, this review will be helpful for rapid design and development of vaccines against the new 2019-nCoV and any future CoVs with pandemic potential. This review was written for the topic of Antivirals for Emerging Viruses: Vaccines and Therapeutics in the Virology section of Frontiers in Microbiology.
- 7Yao, H.; Song, Y.; Chen, Y.; Wu, N.; Xu, J.; Sun, C.; Zhang, J.; Weng, T.; Zhang, Z.; Wu, Z.; Cheng, L.; Shi, D.; Lu, X.; Lei, J.; Crispin, M.; Shi, Y.; Li, L.; Li, S. Molecular architecture of the SARS-CoV-2 virus. Cell 2020, 183, 730– 738, e13 DOI: 10.1016/j.cell.2020.09.018[Crossref], [PubMed], [CAS], Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvFKjs7nM&md5=7bd110334cb42931034a4eb5f9d2ff00Molecular Architecture of the SARS-CoV-2 VirusYao, Hangping; Song, Yutong; Chen, Yong; Wu, Nanping; Xu, Jialu; Sun, Chujie; Zhang, Jiaxing; Weng, Tianhao; Zhang, Zheyuan; Wu, Zhigang; Cheng, Linfang; Shi, Danrong; Lu, Xiangyun; Lei, Jianlin; Crispin, Max; Shi, Yigong; Li, Lanjuan; Li, SaiCell (Cambridge, MA, United States) (2020), 183 (3), 730-738.e13CODEN: CELLB5; ISSN:0092-8674. (Cell Press)SARS-CoV-2 is an enveloped virus responsible for the COVID-19 pandemic. Despite recent advances in the structural elucidation of SARS-CoV-2 proteins, the detailed architecture of the intact virus remains to be unveiled. Here we report the mol. assembly of the authentic SARS-CoV-2 virus using cryoelectron tomog. (cryo-ET) and subtomogram averaging (STA). Native structures of the S proteins in pre- and postfusion conformations were detd. to av. resolns. of 8.7-11 Å. Compns. of the N-linked glycans from the native spikes were analyzed by mass spectrometry, which revealed overall processing states of the native glycans highly similar to that of the recombinant glycoprotein glycans. The native conformation of the ribonucleoproteins (RNPs) and their higher-order assemblies were revealed. Overall, these characterizations revealed the architecture of the SARS-CoV-2 virus in exceptional detail and shed light on how the virus packs its ∼30-kb-long single-segmented RNA in the ∼80-nm-diam. lumen.
- 8Zhou, Y.; Jiang, S.; Du, L. Prospects for a MERS-CoV spike vaccine. Expert Rev. Vaccines 2018, 17, 677– 686, DOI: 10.1080/14760584.2018.1506702[Crossref], [PubMed], [CAS], Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhsV2isb3J&md5=f249563636d50985a7a70127ac0db27aProspects for a MERS-CoV spike vaccineZhou, Yusen; Jiang, Shibo; Du, LanyingExpert Review of Vaccines (2018), 17 (8), 677-686CODEN: ERVXAX; ISSN:1476-0584. (Taylor & Francis Ltd.): Six years have passed since Middle East respiratory syndrome (MERS) coronavirus (MERS-CoV), a newly emerging infectious virus, was first reported in 2012. Although MERS-CoV has had a consistently high mortality rate in humans, no vaccines have been approved to prevent MERS-CoV infection in humans. MERS-CoV spike (S) protein is a key target for development of MERS vaccines.: In this review, we illustrate the structure and function of S protein as a vaccine target, describe available animal models for evaluating MERS vaccines, and summarize recent progress on MERS-CoV S-based vaccines, focusing on their ability to elicit antibody and/or cellular immune responses, neutralizing antibodies, and protection against MERS-CoV infection in different models. Prospects for future MERS-CoV S-based vaccines are discussed.: The majority of MERS vaccines under development are based on MERS-CoV S protein, including full-length S, S1, and receptor-binding domain (RBD). While it is essential to evaluate the safety of full-length S and S1-based MERS vaccines, further improvement of the efficacy of RBD-based vaccines using novel strategies would be necessary. Overall, this review provides informative guidance for designing and developing safe and effective MERS vaccines based on viral S protein.
- 9Lan, J.; Ge, J.; Yu, J.; Shan, S.; Zhou, H.; Fan, S.; Zhang, Q.; Shi, X.; Wang, Q.; Zhang, L.; Wang, X. Structure of the SARS-CoV-2 spike receptor-binding domain bound to the ACE2 receptor. Nature 2020, 581, 215– 220, DOI: 10.1038/s41586-020-2180-5[Crossref], [PubMed], [CAS], Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXoslOqtL8%253D&md5=279c60143e8e5eb505457e0778baa8efStructure of the SARS-CoV-2 spike receptor-binding domain bound to the ACE2 receptorLan, Jun; Ge, Jiwan; Yu, Jinfang; Shan, Sisi; Zhou, Huan; Fan, Shilong; Zhang, Qi; Shi, Xuanling; Wang, Qisheng; Zhang, Linqi; Wang, XinquanNature (London, United Kingdom) (2020), 581 (7807), 215-220CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Abstr.: A new and highly pathogenic coronavirus (severe acute respiratory syndrome coronavirus-2, SARS-CoV-2) caused an outbreak in Wuhan city, Hubei province, China, starting from Dec. 2019 that quickly spread nationwide and to other countries around the world1-3. Here, to better understand the initial step of infection at an at. level, we detd. the crystal structure of the receptor-binding domain (RBD) of the spike protein of SARS-CoV-2 bound to the cell receptor ACE2. The overall ACE2-binding mode of the SARS-CoV-2 RBD is nearly identical to that of the SARS-CoV RBD, which also uses ACE2 as the cell receptor4. Structural anal. identified residues in the SARS-CoV-2 RBD that are essential for ACE2 binding, the majority of which either are highly conserved or share similar side chain properties with those in the SARS-CoV RBD. Such similarity in structure and sequence strongly indicate convergent evolution between the SARS-CoV-2 and SARS-CoV RBDs for improved binding to ACE2, although SARS-CoV-2 does not cluster within SARS and SARS-related coronaviruses1-3,5. The epitopes of two SARS-CoV antibodies that target the RBD are also analyzed for binding to the SARS-CoV-2 RBD, providing insights into the future identification of cross-reactive antibodies.
- 10Li, F.; Li, W.; Farzan, M.; Harrison, S. C. Structure of SARS coronavirus spike receptor-binding domain complexed with receptor. Science 2005, 309, 1864– 1868, DOI: 10.1126/science.1116480[Crossref], [PubMed], [CAS], Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXpvFCisLw%253D&md5=0a7ac2fb7a76c97979d859c0489c682fStructure of SARS Coronavirus Spike Receptor-Binding Domain Complexed with ReceptorLi, Fang; Li, Wenhui; Farzan, Michael; Harrison, Stephen C.Science (Washington, DC, United States) (2005), 309 (5742), 1864-1868CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)The spike protein (S) of SARS coronavirus (SARS-CoV) attaches the virus to its cellular receptor, angiotensin-converting enzyme 2 (ACE2). A defined receptor-binding domain (RBD) on S mediates this interaction. The crystal structure at 2.9 angstrom resoln. of the RBD bound with the peptidase domain of human ACE2 shows that the RBD presents a gently concave surface, which cradles the N-terminal lobe of the peptidase. The at. details at the interface between the two proteins clarify the importance of residue changes that facilitate efficient cross-species infection and human-to-human transmission. The structure of the RBD suggests ways to make truncated disulfide-stabilized RBD variants for use in the design of coronavirus vaccines.
- 11Liu, S.; Xiao, G.; Chen, Y.; He, Y.; Niu, J.; Escalante, C. R.; Xiong, H.; Farmar, J.; Debnath, A. K.; Tien, P.; Jiang, S. Interaction between heptad repeat 1 and 2 regions in spike protein of SARS-associated coronavirus: implications for virus fusogenic mechanism and identification of fusion inhibitors. Lancet 2004, 363, 938– 947, DOI: 10.1016/s0140-6736(04)15788-7[Crossref], [PubMed], [CAS], Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXisVSktbo%253D&md5=578507918c57e2419c24213bea2c978fInteraction between heptad repeat 1 and 2 regions in spike protein of SARS-associated coronavirus: implications for virus fusogenic mechanism and identification of fusion inhibitorsLiu, Shuwen; Xiao, Gengfu; Chen, Yibang; He, Yuxian; Niu, Jinkui; Escalante, Carlos R.; Xiong, Huabao; Farmar, James; Debnath, Asim K.; Tien, Po; Jiang, ShiboLancet (2004), 363 (9413), 938-947CODEN: LANCAO; ISSN:0140-6736. (Elsevier Science Ltd.)Studies on the fusion-inhibitory peptides derived from the heptad repeat 1 and 2 (HR1 and HR2) regions of the HIV-1 envelope glycoprotein gp41 provided crucial information on the viral fusogenic mechanism. We used a similar approach to study the fusogenic mechanism of severe-acute-respiratory-syndrome-assocd. coronavirus (SARS-CoV). We tested the inhibitory activity against infection of two sets of peptides corresponding to sequences of SARS-CoV spike protein HR1 and HR2 regions and investigated the interactions between the HR1 and HR2 peptides by surface plasmon resonance, sedimentation equilibration anal., CD, native polyacrylamide-gel electrophoresis, size exclusion high-performance liq. chromatog., and computer-aided homol. modeling and mol. docking anal. One peptide, CP-1, derived from the HR2 region, inhibited SARS-CoV infection in the micromolar range. CP-1 bound with high affinity to a peptide from the HR1 region, NP-1. CP-1 alone had low α-helicity and self-assocd. to form a trimer in phosphate buffer (pH 7.2). CP-1 and NP-1 mixed in equimolar concns. formed a six-helix bundle, similar to the fusogenic core structure of HIV-1 gp41. After binding to the target cell, the transmembrane spike protein might change conformation by assocn. between the HR1 and HR2 regions to form an oligomeric structure, leading to fusion between the viral and target-cell membranes. At the pre-fusion intermediate state, CP-1 could bind to the HR1 region and interfere with the conformational changes, resulting in inhibition of SARS-CoV fusion with the target cells. CP-1 might be modifiable to increase its anti-SARS-CoV activity and could be further developed as an antiviral agent for treatment or prophylaxis of SARS-CoV infection.
- 12Tai, W.; He, L.; Zhang, X.; Pu, J.; Voronin, D.; Jiang, S.; Zhou, Y.; Du, L. Characterization of the receptor-binding domain (RBD) of 2019 novel coronavirus: implication for development of RBD protein as a viral attachment inhibitor and vaccine. Cell. Mol. Immunol. 2020, 17, 613– 620, DOI: 10.1038/s41423-020-0400-4[Crossref], [PubMed], [CAS], Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXlt1Chsrw%253D&md5=87bc49d070c84e78b01230518aaa465aCharacterization of the receptor-binding domain (RBD) of 2019 novel coronavirus: implication for development of RBD protein as a viral attachment inhibitor and vaccineTai, Wanbo; He, Lei; Zhang, Xiujuan; Pu, Jing; Voronin, Denis; Jiang, Shibo; Zhou, Yusen; Du, LanyingCellular & Molecular Immunology (2020), 17 (6), 613-620CODEN: CMIEAO; ISSN:1672-7681. (Nature Research)The outbreak of Coronavirus Disease 2019 (COVID-19) has posed a serious threat to global public health, calling for the development of safe and effective prophylactics and therapeutics against infection of its causative agent, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), also known as 2019 novel coronavirus (2019-nCoV). The CoV spike (S) protein plays the most important roles in viral attachment, fusion and entry, and serves as a target for development of antibodies, entry inhibitors and vaccines. Here, we identified the receptor-binding domain (RBD) in SARS-CoV-2 S protein and found that the RBD protein bound strongly to human and bat angiotensin-converting enzyme 2 (ACE2) receptors. SARS-CoV-2 RBD exhibited significantly higher binding affinity to ACE2 receptor than SARS-CoV RBD and could block the binding and, hence, attachment of SARS-CoV-2 RBD and SARS-CoV RBD to ACE2-expressing cells, thus inhibiting their infection to host cells. SARS-CoV RBD-specific antibodies could cross-react with SARS-CoV-2 RBD protein, and SARS-CoV RBD-induced antisera could cross-neutralize SARS-CoV-2, suggesting the potential to develop SARS-CoV RBD-based vaccines for prevention of SARS-CoV-2 and SARS-CoV infection.
- 13Dai, L.; Gao, G. F. Viral targets for vaccines against COVID-19. Nat. Rev. Immunol. 2021, 21, 73– 82, DOI: 10.1038/s41577-020-00480-0[Crossref], [PubMed], [CAS], Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXktFeksg%253D%253D&md5=4e84b56201379ab888192ff41d3b30ffViral targets for vaccines against COVID-19Dai, Lianpan; Gao, George F.Nature Reviews Immunology (2021), 21 (2), 73-82CODEN: NRIABX; ISSN:1474-1733. (Nature Research)A review of the progress of which viral elements are used in COVID-19 vaccine candidates, why they might act as good targets for the immune system and the implications for protective immunity. Vaccines are urgently needed to control the coronavirus disease 2019 (COVID-19) pandemic and to help the return to pre-pandemic normalcy. A great many vaccine candidates are being developed, several of which have completed late-stage clin. trials and are reporting pos. results.
- 14Khan, S.; Tombuloglu, H.; Hassanein, S. E.; Rehman, S.; Bozkurt, A.; Cevik, E.; Abdel-Ghany, S.; Nabi, G.; Ali, A.; Sabit, H. Coronavirus diseases 2019: current biological situation and potential therapeutic perspective. Eur. J. Pharmacol. 2020, 886, 1733447, DOI: 10.1016/j.ejphar.2020.173447
- 15Tombuloglu, H.; Sabit, H.; Al-Suhaimi, E.; Al Jindan, R.; Alkharsah, K. R. Development of multiplex real-time RT-PCR assay for the detection of SARS-CoV-2. PLoS One 2021, 16, e0250942 DOI: 10.1371/journal.pone.0250942[Crossref], [PubMed], [CAS], Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhtVegur3I&md5=734b90db068d5762645c84d93e456cd2Development of multiplex real-time RT-PCR assay for the detection of SARS-CoV-2Tombuloglu, Huseyin; Sabit, Hussein; Al-Suhaimi, Ebtesam; Al Jindan, Reem; Alkharsah, Khaled R.PLoS One (2021), 16 (4), e0250942CODEN: POLNCL; ISSN:1932-6203. (Public Library of Science)The outbreak of the new human coronavirus SARS-CoV-2 (also known as 2019-nCoV) continues to increase globally. The real-time reverse transcription polymerase chain reaction (rRT-PCR) is the most used technique in virus detection. However, possible false-neg. and false-pos. results produce misleading consequences, making it necessary to improve existing methods. Here, we developed a multiplex rRT-PCR diagnostic method, which targets two viral genes (RdRP and E) and one human gene (RP) simultaneously. The reaction was tested by using pseudoviral RNA and human target mRNA sequences as a template. Also, the protocol was validated by using 14 clin. SARS-CoV-2 pos. samples. The results are in good agreement with the CDC authorized Cepheid's Xpert Xpress SARS-CoV-2 diagnostic system (100%). Unlike single gene targeting strategies, the current method provides the amplification of two viral regions in the same PCR reaction. Therefore, an accurate SARS-CoV-2 diagnostic assay was provided, which allows testing of 91 samples in 96-well plates in per run. Thanks to this strategy, fast, reliable, and easy-to-use rRT-PCR method is obtained to diagnose SARS-CoV-2.
- 16Ravi, N.; Cortade, D. L.; Ng, E.; Wang, S. X. Diagnostics for SARS-CoV-2 detection: a comprehensive review of the FDA-EUA COVID-19 testing landscape. Biosens. Bioelectron. 2020, 165, 112454, DOI: 10.1016/j.bios.2020.112454[Crossref], [PubMed], [CAS], Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsFShsbvN&md5=1e88bde679e0507c706e21fa9c2d54d7Diagnostics for SARS-CoV-2 detection: A comprehensive review of the FDA-EUA COVID-19 testing landscapeRavi, Neeraja; Cortade, Dana L.; Ng, Elaine; Wang, Shan X.Biosensors & Bioelectronics (2020), 165 (), 112454CODEN: BBIOE4; ISSN:0956-5663. (Elsevier B.V.)A review. The rapidly spreading outbreak of COVID-19 disease is caused by the SARS-CoV-2 virus, first reported in Dec. 2019 in Wuhan, China. As of June 17, 2020, this virus has infected over 8.2 million people but ranges in symptom severity, making it difficult to assess its overall infection rate. There is a need for rapid and accurate diagnostics to better monitor and prevent the spread of COVID-19. In this review, we present and evaluate two main types of diagnostics with FDA-EUA status for COVID-19: nucleic acid testing for detection of SARS-CoV-2 RNA, and serol. assays for detection of SARS-CoV-2 specific IgG and IgM patient antibodies, along with the necessary sample prepn. for accurate diagnoses. In particular, we cover and compare tests such as the CDC 2019-nCoV RT-PCR Diagnostic Panel, Cellex's qSARS-CoV-2 IgG/IgM Rapid Test, and point-of-care tests such as Abbott's ID NOW COVID-19 Test. Antibody testing is esp. important in understanding the prevalence of the virus in the community and to identify those who have gained immunity. We conclude by highlighting the future of COVID-19 diagnostics, which include the need for quant. testing and the development of emerging biosensors as point-of-care tests.
- 17Abbott Diagnostics Scarborough Inc. BinaxNOWTM COVID-19 Ag CARD [package insert, EUA]; Scarborough: MA, USA, 2020.Google ScholarThere is no corresponding record for this reference.
- 18LumiraDx Group Limited. LumiraDxTM SARS-CoV-2 Ag Test [package insert, EUA]; Dumyat Business Park Alloa FK10 2PB: U.K., 2020.Google ScholarThere is no corresponding record for this reference.
- 19Becton, Dickinson and Company. BD VeritorTM System for Rapid Detection of SARS-CoV-2 [package insert, EUA]; Sparks-Glencoe: MD, USA, 2020.Google ScholarThere is no corresponding record for this reference.
- 20Quidel Corporation. Sofia Severe Acute Respiratory Syndrome Antigen FIA [package insert, EUA]: San Diego, CA, USA, 2020.Google ScholarThere is no corresponding record for this reference.
- 21Barlev-Gross, M.; Weiss, S.; Ben-Shmuel, A.; Sittner, A.; Eden, K.; Mazuz, N.; Glinert, I.; Bar-David, E.; Puni, R.; Amit, S.; Kriger, O.; Schuster, O.; Alcalay, R.; Makdasi, E.; Epstein, E.; Noy-Porat, T.; Rosenfeld, R.; Achdout, H.; Mazor, O.; Israely, T.; Levy, H.; Mechaly, A. Spike vs nucleocapsid SARS-CoV-2 antigen detection: application in nasopharyngeal swab specimens. Anal. Bioanal. Chem. 2021, 413, 3501– 3510, DOI: 10.1007/s00216-021-03298-4[Crossref], [PubMed], [CAS], Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXnsVGnsr8%253D&md5=af42bca2a60bfe68687276dfde045b72Spike vs. nucleocapsid SARS-CoV-2 antigen detection: application in nasopharyngeal swab specimensBarlev-Gross, Moria; Weiss, Shay; Ben-Shmuel, Amir; Sittner, Assa; Eden, Keren; Mazuz, Noam; Glinert, Itai; Bar-David, Elad; Puni, Reut; Amit, Sharon; Kriger, Or; Schuster, Ofir; Alcalay, Ron; Makdasi, Efi; Epstein, Eyal; Noy-Porat, Tal; Rosenfeld, Ronit; Achdout, Hagit; Mazor, Ohad; Israely, Tomer; Levy, Haim; Mechaly, AdvaAnalytical and Bioanalytical Chemistry (2021), 413 (13), 3501-3510CODEN: ABCNBP; ISSN:1618-2642. (Springer)Public health experts emphasize the need for quick, point-of-care SARS-CoV-2 detection as an effective strategy for controlling virus spread. To this end, many antigen detection devices were developed and commercialized. These devices are mostly based on detecting SARS-CoV-2's nucleocapsid protein. Recently, alerts issued by both the FDA and the CDC raised concerns regarding the devices' tendency to exhibit false pos. results. We developed a novel alternative spike-based antigen assay, comprising 4 high-affinity, specific monoclonal antibodies, directed against different epitopes on the spike's S1 subunit. The assay's performance was evaluated for COVID-19 detection from nasopharyngeal swabs, compared to an inhouse nucleocapsid-based assay, composed of novel antibodies directed against the nucleocapsid. Detection of COVID-19 was carried out in a cohort of 284 qRT-PCR pos. and neg. nasopharyngeal swab samples. The time resolved fluorescence (TRF) ELISA spike assay displayed very high specificity (99%) accompanied with a somewhat lower sensitivity (66% for Ct < 25), compared to the nucleocapsid ELISA assay which was more sensitive (85% for Ct < 25) while less specific (87% specificity). Despite being outperformed by qRT-PCR, we suggest that there is room for such tests in the clin. setting, as cheap and rapid pre-screening tools. Our results further suggest that when applying antigen detection, one must consider its intended application (sensitivity vs. specificity), taking into consideration that the nucleocapsid might not be the optimal target. In this regard, we propose that a combination of both antigens might contribute to the validity of the results.
- 22Aoki, K.; Nagasawa, T.; Ishii, Y.; Yagi, S.; Okuma, S.; Kashiwagi, K.; Maeda, T.; Miyazaki, T.; Yoshizawa, S.; Tateda, K. Clinical validation of quantitative SARS-CoV-2 antigen assays to estimate SARS-CoV-2 viral loads in nasopharyngeal swabs. J. Infect. Chemother. 2021, 27, 613– 616, DOI: 10.1016/j.jiac.2020.11.021[Crossref], [PubMed], [CAS], Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhslSru7c%253D&md5=98e3f6fbd1cf1041ba11fcc2c6dcc54aClinical validation of quantitative SARS-CoV-2 antigen assays to estimate SARS-CoV-2 viral loads in nasopharyngeal swabsAoki, Kotaro; Nagasawa, Tatsuya; Ishii, Yoshikazu; Yagi, Shintaro; Okuma, Sadatsugu; Kashiwagi, Katsuhito; Maeda, Tadashi; Miyazaki, Taito; Yoshizawa, Sadako; Tateda, KazuhiroJournal of Infection and Chemotherapy (2021), 27 (4), 613-616CODEN: JICHFN; ISSN:1341-321X. (Elsevier B.V.)Expansion of the testing capacity for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an important issue to mitigate the pandemic of coronavirus disease-2019 (COVID-19) caused by this virus. Recently, a sensitive quant. antigen test (SQT), Lumipulse SARS-CoV-2 Ag, was developed. It is a fully automated chemiluminescent enzyme immunoassay system for SARS-CoV-2. In this study, the anal. performance of SQT was examd. using clin. specimens from nasopharyngeal swabs using reverse transcription polymerase chain reaction (RT-PCR) as a control. Receiver operating characteristic anal. of 24 SARS-CoV-2-pos. and 524 -neg. patients showed an area under the curve of 0.957 ± 0.063. Using a cut-off value of 1.34 pg/mL, the sensitivity was 91.7%, the specificity was 98.5%, and the overall rate of agreement was 98.2%. In the distribution of neg. cases, the 99.5 percentile value was 1.03 pg/mL. There was a high correlation between the viral load calcd. using the cycle threshold value of RT-PCR and the concn. of antigen. The tendency for the antigen concn. to decrease with time after disease onset correlated with that of the viral load. Presented results indicate that SQT is highly concordant with RT-PCR and should be useful for the diagnosis of COVID-19 in any clin. setting. Therefore, this fully automated kit will contribute to the expansion of the testing capability for SARS-CoV-2.
- 23Hirotsu, Y.; Maejima, M.; Shibusawa, M.; Nagakubo, Y.; Hosaka, K.; Amemiya, K.; Sueki, H.; Hayakawa, M.; Mochizuki, H.; Tsutsui, T.; Kakizaki, Y.; Miyashita, Y.; Yagi, S.; Kojima, S.; Omata, M. Comparison of automated SARS-CoV-2 antigen test for COVID-19 infection with quantitative RT-PCR using 313 nasopharyngeal swabs, including from seven serially followed patients. Int. J. Infect. Dis. 2020, 99, 397– 402, DOI: 10.1016/j.ijid.2020.08.029[Crossref], [PubMed], [CAS], Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvVWmtb%252FP&md5=2b97870f9db15fdb5136fe7ddda6294eComparison of automated SARS-CoV-2 antigen test for COVID-19 infection with quantitative RT-PCR using 313 nasopharyngeal swabs, including from seven serially followed patientsHirotsu, Yosuke; Maejima, Makoto; Shibusawa, Masahiro; Nagakubo, Yuki; Hosaka, Kazuhiro; Amemiya, Kenji; Sueki, Hitomi; Hayakawa, Miyoko; Mochizuki, Hitoshi; Tsutsui, Toshiharu; Kakizaki, Yumiko; Miyashita, Yoshihiro; Yagi, Shintaro; Kojima, Satoshi; Omata, MasaoInternational Journal of Infectious Diseases (2020), 99 (), 397-402CODEN: IJIDF3; ISSN:1201-9712. (Elsevier Ltd.)In routine clin. practice, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is detd. by reverse-transcription PCR (RT-PCR). In the current pandemic, a more rapid and high-throughput method is in growing demand. Here, the authors validated the performance of a new antigen test (LUMIPULSE) based on chemiluminescence enzyme immunoassay. A total of 313 nasopharyngeal swabs (82 serial samples from 7 infected patients and 231 individual samples from 4 infected patients and 215 uninfected individuals) were analyzed for SARS-CoV-2 with quant. RT-PCR (RT-qPCR) and then subjected to LUMIPULSE. The authors detd. the cutoff value for antigen detection using receiver operating characteristic curve anal. and compared the performance of the antigen test with that of RT-qPCR. The authors also compared the viral loads and antigen levels in serial samples from seven infected patients. Using RT-qPCR as the ref., the antigen test exhibited 55.2% sensitivity and 99.6% specificity, with a 91.4% overall agreement rate (286/313). In specimens with > 100 viral copies and between 10 and 100 copies, the antigen test showed 100% and 85% concordance with RT-qPCR, resp. This concordance declined with lower viral loads. In the serially followed patients, the antigen levels showed a steady decline, along with viral clearance. This gradual decline was in contrast with the abrupt pos.-to-neg. and neg.-to-pos. status changes obsd. with RT-qPCR, particularly in the late phase of infection. In summary, the LUMIPULSE antigen test can rapidly identify SARS-CoV-2-infected individuals with moderate to high viral loads and may be helpful for monitoring viral clearance in hospitalized patients.
- 24Pollock, N. R.; Savage, T. J.; Wardell, H.; Lee, R.; Mathew, A.; Stengelin, M.; Sigal, G. B. Correlation of SARS-CoV-2 nucleocapsid antigen and RNA concentrations in nasopharyngeal samples from children and adults using an ultrasensitive and quantitative antigen assay. J. Clin. Microbiol. 2021, 59, e03077– 20, DOI: 10.1128/JCM.03077-20[Crossref], [PubMed], [CAS], Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXpvFyks70%253D&md5=bb1100be3ef32c227627e5f53c7be92cCorrelation of SARS-CoV-2 nucleocapsid antigen and RNA concentrations in nasopharyngeal samples from children and adults using an ultrasensitive and quantitative antigen assayPollock, Nira R.; Savage, Timothy J.; Wardell, Hanna; Lee, Rose A.; Mathew, Anu; Stengelin, Martin; Sigal, George B.Journal of Clinical Microbiology (2021), 59 (4), e03077CODEN: JCMIDW; ISSN:1098-660X. (American Society for Microbiology)Diagnosis of COVID-19 by PCR offers high sensitivity, but the utility of detecting samples with high cycle threshold (CT) values remains controversial. Currently available rapid diagnostic tests (RDTs) for SARS-CoV-2 nucleocapsid antigens (Ag) have sensitivity well below PCR. The correlation of Ag and RNA quantities in clin. nasopharyngeal (NP) samples is unknown. An ultrasensitive, quant. electrochemiluminescence immunoassay for SARS-CoV-2 nucleocapsid (the MSD S-PLEX SARS-CoV-2 N assay) was used to measure Ag in clin. NP samples from adults and children previously tested by PCR. The S-PLEX Ag assay had a limit of detection (LOD) of 0.16 pg/mL and a cutoff of 0.32 pg/mL. Ag concns. measured in clin. NP samples (collected in 3.0 mL of media) ranged from less than 160 fg/mL to 2.7μg/mL. Log-transformed Ag concns. correlated tightly with CT values. In 35 adult and 101 pediatric PCR-pos. samples, the sensitivities were 91% (95% confidence interval, 77 to 98%) and 79% (70 to 87%), resp. In samples with a CT of ≤35, the sensitivities were 100% (88 to 100%) and 96% (88 to 99%), resp. In 50 adult and 40 pediatric PCR-neg. specimens, the specificities were 100% (93 to 100%) and 98% (87 to 100%), resp. Nucleocapsid concns. in clin. NP samples span 8 orders of magnitude and correlate closely with RNA concns. (CT values). The S-PLEX Ag assay showed 96 to 100% sensitivity in samples from children and adults with CT values of ≤35, and a specificity of 98 to 100%. These results clarify Ag concn. distributions in clin. samples, providing insight into the performance of Ag RDTs and offering a new approach to diagnosis of COVID-19.
- 25Pekosz, A.; Cooper, C. K.; Parvu, V.; Li, M.; Andrews, J. C.; Manabe, Y. C.; Kodsi, S.; Leitch, J.; Gary, D. S.; Roger-Dalbert, C. Antigen-based testing but not real-time polymerase chain reaction correlates with Severe Acute Respiratory Syndrome Coronavirus 2 viral culture. Clin. Infect. Dis. 2021, 73, e2861– e2866, DOI: 10.1093/cid/ciaa1706[Crossref], [PubMed], [CAS], Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXisVKrtLjI&md5=72f2f571f0341b63249a3b702b275717Antigen-based testing but not real-time polymerase chain reaction correlates with severe acute respiratory syndrome Coronavirus 2 viral culturePekosz, Andrew; Parvu, Valentin; Li, Maggie; Andrews, Jeffrey C.; Manabe, Yukari C.; Kodsi, Salma; Gary, Devin S.; Roger-Dalbert, Celine; Leitch, Jeffry; Cooper, Charles K.Clinical Infectious Diseases (2021), 73 (9), e2861-e2866CODEN: CIDIEL; ISSN:1537-6591. (Oxford University Press)Individuals can test pos. for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) by mol. assays following the resoln. of their clin. disease. Recent studies indicate that SARS-CoV-2 antigen-based tests are likely to be pos. early in the disease course, when there is an increased likelihood of high levels of infectious virus. Upper respiratory specimens from 251 participants with coronavirus disease 2019 symptoms (≤7 days from symptom onset) were prospectively collected and tested with a lateral flow antigen test and a real-time polymerase chain reaction (rt-PCR) assay for detection of SARS-CoV-2. Specimens from a subset of the study specimens were utilized to det. the presence of infectious virus in the VeroE6TMPRSS2 cell culture model. The antigen test demonstrated a higher pos. predictive value (90%) than rt-PCR (70%) when compared to culture-pos. results. The pos. percentage agreement for detection of infectious virus for the antigen test was similar to rt-PCR when compared to culture results. The correlation between SARS-CoV-2 antigen and SARS-CoV-2 culture positivity represents a significant advancement in detg. the risk for potential transmissibility beyond that which can be achieved by detection of SARS-CoV-2 genomic RNA. SARS-CoV-2 antigen testing can facilitate low-cost, scalable, and rapid time-to-result, while providing good risk detn. of those who are likely harboring infectious virus, compared to rt-PCR.
- 26Scohy, A.; Anantharajah, A.; Bodéus, M.; Kabamba-Mukadi, B.; Verroken, A.; Rodriguez-Villalobos, H. Low performance of rapid antigen detection test as frontline testing for COVID-19 diagnosis. J. Clin. Virol. 2020, 129, 104455, DOI: 10.1016/j.jcv.2020.104455[Crossref], [PubMed], [CAS], Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtVGmsbbI&md5=183778b80c7596564747ec676c198e09Low performance of rapid antigen detection test as frontline testing for COVID-19 diagnosisScohy, Anais; Anantharajah, Ahalieyah; Bodeus, Monique; Kabamba-Mukadi, Benoit; Verroken, Alexia; Rodriguez-Villalobos, HectorJournal of Clinical Virology (2020), 129 (), 104455CODEN: JCVIFB; ISSN:1386-6532. (Elsevier B.V.)Ensuring accurate diagnosis is essential to limit the spread of SARS-CoV-2 and for the clin. management of COVID-19. Although real-time reverse transcription polymerase chain reaction (RT- qPCR) is the current recommended lab. method to diagnose SARS-CoV-2 acute infection, several factors such as requirement of special equipment and skilled staff limit the use of these time-consuming mol. techniques. Recently, several easy to perform rapid antigen detection tests were developed and recommended in some countries as the first line of diagnostic. The aim of this study was to evaluate the performances of the Coris COVID-19 Ag Respi-Strip test, a rapid immunochromatog. test for the detection of SARS-CoV-2 antigen, in comparison to RT-qPCR. 148 Nasopharyngeal swabs were tested. Amongst the 106 pos. RT-qPCR samples, 32 were detected by the rapid antigen test, given an overall sensitivity of 30.2%. All the samples detected pos. with the antigen rapid test were also pos. with RT-qPCR. Higher viral loads are assocd. with better antigen detection rates. Unfortunately, the overall poor sensitivity of the COVID-19 Ag Respi-Strip does not allow using it alone as the frontline testing for COVID-19 diagnosis.
- 27Tang, Y. W.; Schmitz, J. E.; Persing, D. H.; Stratton, C. W. Laboratory diagnosis of COVID-19: current issues and challenges. J. Clin. Microbiol. 2020, 58, 10– 1128, DOI: 10.1128/JCM.00512-20
- 28Che, X.-y.; Qiu, L.-w.; Pan, Y.-x.; Wen, K.; Hao, W.; Zhang, L.-y.; Wang, Y.-d.; Liao, Z.-y.; Hua, X.; Cheng, V. C. C.; Yuen, K.-y. Sensitive and specific monoclonal antibody-based capture enzyme immunoassay for detection of nucleocapsid antigen in sera from patients with Severe Acute Respiratory Syndrome. J. Clin. Microbiol. 2004, 42, 2629– 2635, DOI: 10.1128/jcm.42.6.2629-2635.2004[Crossref], [PubMed], [CAS], Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXlslalsr4%253D&md5=53f47dfefc1c75e16abb77b5a6f46315Sensitive and specific monoclonal antibody-based capture enzyme immunoassay for detection of nucleocapsid antigen in sera from patients with severe acute respiratory syndromeChe, Xiao-yan; Qiu, Li-wen; Pan, Yu-xian; Wen, Kun; Hao, Wei; Zhang, Li-ya; Wang, Ya-di; Liao, Zhi-yong; Hua, Xu; Cheng, Vincent C. C.; Yuen, Kwok-yungJournal of Clinical Microbiology (2004), 42 (6), 2629-2635CODEN: JCMIDW; ISSN:0095-1137. (American Society for Microbiology)A rapid antigen test for the diagnosis of severe acute respiratory syndrome (SARS) is essential for control of this disease at the point of management. The nucleocapsid (N) protein of SARS-assocd. coronavirus (SARS-CoV) is abundantly expressed in infected-cell culture filtrate as demonstrable by Western blotting using convalescent-phase sera from patients with SARS. We used monoclonal antibodies specifically directed against N protein to establish a sensitive antigen capture sandwich ELISA for the detection of SARS-CoV. The assay employed a mixt. of three monoclonal antibodies for capture and rabbit polyclonal antibodies for detection of serum antigen in 32 cases of clin. probable SARS as defined by the World Health Organization during the epidemic in Guangzhou, China. Recombinant N protein was used as a std. to establish a detection sensitivity of approximated 50 pg/mL. The linear range of detection in clin. specimens was from 100 pg/mL to 3.2 ng/mL. Using a panel of sera collected at different points in time, the amt. of circulating N antigen was found to peak 6 to 10 days after the onset of symptoms. The sensitivity of the assay was 84.6% in 13 serol. confirmed SARS patients with blood taken during the first 10 days after the onset of symptoms (11 of 13). The specificity of the assay was 98.5% in 1,272 healthy individuals (1,253 of 1,272). There was no cross-reaction with other human and animal coronaviruses in this assay. In conclusion, a sensitive and quant. antigen capture ELISA was established for the early diagnosis and disease monitoring of SARS-CoV infection.
- 29Diao, B.; Wen, K.; Chen, J.; Liu, Y.; Yuan, Z.; Han, C.; Chen, J.; Pan, Y.; Chen, L.; Dan, Y.; Wang, J.; Chen, Y.; Deng, G.; Zhou, H.; Wu, Y. Diagnosis of acute respiratory syndrome coronavirus 2 infection by detection of nucleocapsid protein. medRxiv 2020, 20032524, DOI: 10.1101/2020.03.07.20032524
- 30Grant, B. D.; Anderson, C. E.; Williford, J. R.; Alonzo, L. F.; Glukhova, V. A.; Boyle, D. S.; Weigl, B. H.; Nichols, K. P. SARS-CoV-2 coronavirus nucleocapsid antigen-detecting half-strip lateral flow assay toward the development of point of care tests using commercially available reagents. Anal. Chem. 2020, 92, 11305– 11309, DOI: 10.1021/acs.analchem.0c01975[ACS Full Text
], [CAS], Google Scholar
30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtlSis7jO&md5=e4cb14b8cb5e90bf93381695f5a4f634SARS-CoV-2 Coronavirus Nucleocapsid Antigen-Detecting Half-Strip Lateral Flow Assay Toward the Development of Point of Care Tests Using Commercially Available ReagentsGrant, Benjamin D.; Anderson, Caitlin E.; Williford, John R.; Alonzo, Luis F.; Glukhova, Veronika A.; Boyle, David S.; Weigl, Bernhard H.; Nichols, Kevin P.Analytical Chemistry (Washington, DC, United States) (2020), 92 (16), 11305-11309CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)The SARS-CoV-2 pandemic has created an unprecedented need for rapid diagnostic testing to enable the efficient treatment and mitigation of COVID-19. The primary diagnostic tool currently employed is reverse transcription polymerase chain reaction (RT-PCR), which can have good sensitivity and excellent specificity. Unfortunately, implementation costs and logistical problems with reagents during the global SARS-CoV-2 pandemic have hindered its universal on demand adoption. Lateral flow assays (LFAs) represent a class of diagnostic that, if sufficiently clin. sensitive, may fill many of the gaps in the current RT-PCR testing regime, esp. in low- and middle-income countries (LMICs). To date, many serol. LFAs have been developed, though none meet the performance requirements necessary for diagnostic use cases, primarily due to the relatively long delay between infection and seroconversion. However, on the basis of previously reported results from SARS-CoV-1, antigen-based SARS-CoV-2 assays may have significantly better clin. sensitivity than serol. assays. To date, only a very small no. of antigen-detecting LFAs have been developed. Development of a half-strip LFA is a useful first step in the development of any LFA format. In this work, we present a half-strip LFA using com. available antibodies for the detection of SARS-CoV-2. We have tested this LFA in buffer and measured an LOD of 0.65 ng/mL (95% CI of 0.53 to 0.77 ng/mL) ng/mL with recombinant antigen using an optical reader with sensitivity equiv. to a visual read. Further development, including evaluating the appropriate sample matrix, will be required for this assay approach to be made useful in a point of care setting, though this half-strip LFA may serve as a useful starting point for others developing similar tests. - 31Ahmadivand, A.; Gerislioglu, B.; Ramezani, Z.; Kaushik, A.; Manickam, P.; Ghoreishi, S. A. Functionalized terahertz plasmonic metasensors: femtomolar-level detection of SARS-CoV-2 spike proteins. Biosens. Bioelectron. 2021, 177, 112971, DOI: 10.1016/j.bios.2021.112971[Crossref], [PubMed], [CAS], Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhtVGlsbc%253D&md5=6c090c57524e355c17a9d9f0e77c37a2Functionalized terahertz plasmonic metasensors: Femtomolar-level detection of SARS-CoV-2 spike proteinsAhmadivand, Arash; Gerislioglu, Burak; Ramezani, Zeinab; Kaushik, Ajeet; Manickam, Pandiaraj; Ghoreishi, S. AmirBiosensors & Bioelectronics (2021), 177 (), 112971CODEN: BBIOE4; ISSN:0956-5663. (Elsevier B.V.)Effective and efficient management of human betacoronavirus severe acute respiratory syndrome (SARS)-CoV-2 virus infection i.e., COVID-19 pandemic, required sensitive and selective sensors with short sample-to-result durations for performing desired diagnostics. In this direction, one appropriate alternative approach to detect SARS-CoV-2 virus protein at low level i.e., femtomolar (fM) is exploring plasmonic metasensor technol. for COVID-19 diagnostics, which offers exquisite opportunities in advanced healthcare programs, and modern clin. diagnostics. The intrinsic merits of plasmonic metasensors stem from their capability to squeeze electromagnetic fields, simultaneously in frequency, time, and space. However, the detection of low-mol. wt. biomols. at low densities is a typical drawback of conventional metasensors that has recently been addressed using toroidal metasurface technol. This research is focused on the fabrication of a miniaturized plasmonic immunosensor based on toroidal electrodynamics concept that can sustain robustly confined plasmonic modes with ultranarrow lineshapes in the terahertz (THz) frequencies. By exciting toroidal dipole mode using our quasi-infinite metasurface and a judiciously optimized protocol based on functionalized gold nanoparticles (AuNPs) conjugated with the specific monoclonal antibody specific to spike protein (S1) of SARS-CoV-2 virus onto the metasurface, the resonance shifts for diverse concns. of the spike protein are monitored. Possessing mol. wt. around ∼76 kDa allowed to detect the presence of SARS-CoV-2 virus protein with significantly low as limit of detection (LoD) was achieved as ∼4.2 fM. We envisage that outcomes of this research will pave the way toward the use of toroidal metasensors as practical technologies for rapid and precise screening of SARS-CoV-2 virus carriers, symptomatic or asymptomatic, and spike proteins in hospitals, clinics, labs., and site of infection.
- 32Seo, G.; Lee, G.; Kim, M. J.; Baek, S.-H.; Choi, M.; Ku, K. B.; Lee, C.-S.; Jun, S.; Park, D.; Kim, H. G.; Kim, S.-J.; Lee, J.-O.; Kim, B. T.; Park, E. C.; Kim, S. I. 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, DOI: 10.1021/acsnano.0c02823[ACS Full Text
], [CAS], Google Scholar
32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXnt1SrtrY%253D&md5=735104d0be58eaefaae38aa6f9877459Rapid Detection of COVID-19 Causative Virus (SARS-CoV-2) in Human Nasopharyngeal Swab Specimens Using Field-Effect Transistor-Based BiosensorSeo, Giwan; Lee, Geonhee; Kim, Mi Jeong; Baek, Seung-Hwa; Choi, Minsuk; Ku, Keun Bon; Lee, Chang-Seop; Jun, Sangmi; Park, Daeui; Kim, Hong Gi; Kim, Seong-Jun; Lee, Jeong-O.; Kim, Bum Tae; Park, Edmond Changkyun; Kim, Seung IlACS Nano (2020), 14 (4), 5135-5142CODEN: ANCAC3; ISSN:1936-0851. (American Chemical Society)Coronavirus disease 2019 (COVID-19) is a newly emerging human infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2, previously called 2019-nCoV). Based on the rapid increase in the rate of human infection, the World Health Organization (WHO) has classified the COVID-19 outbreak as a pandemic. Because no specific drugs or vaccines for COVID-19 are yet available, early diagnosis and management are crucial for contg. the outbreak. Here, we report a field-effect transistor (FET)-based biosensing device for detecting SARS-CoV-2 in clin. samples. The sensor was produced by coating graphene sheets of the FET with a specific antibody against SARS-CoV-2 spike protein. The performance of the sensor was detd. using antigen protein, cultured virus, and nasopharyngeal swab specimens from COVID-19 patients. Our FET device could detect the SARS-CoV-2 spike protein at concns. of 1 fg/mL in phosphate-buffered saline and 100 fg/mL clin. transport medium. In addn., the FET sensor successfully detected SARS-CoV-2 in culture medium (limit of detection [LOD]: 1.6 x 101 pfu/mL) and clin. samples (LOD: 2.42 x 102 copies/mL). Thus, we have successfully fabricated a promising FET biosensor for SARS-CoV-2; our device is a highly sensitive immunol. diagnostic method for COVID-19 that requires no sample pretreatment or labeling. - 33Zhong, J.; Rösch, E. L.; Viereck, T.; Schilling, M.; Ludwig, F. Toward rapid and sensitive detection of SARS-CoV-2 with functionalized magnetic nanoparticles. ACS Sens. 2021, 6, 976– 984, DOI: 10.1021/acssensors.0c02160[ACS Full Text
], [CAS], Google Scholar
33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXitV2ku78%253D&md5=e79da90b2367b4017b15e4e00dae2277Toward Rapid and Sensitive Detection of SARS-CoV-2 with Functionalized Magnetic NanoparticlesZhong, Jing; Roesch, Enja Laureen; Viereck, Thilo; Schilling, Meinhard; Ludwig, FrankACS Sensors (2021), 6 (3), 976-984CODEN: ASCEFJ; ISSN:2379-3694. (American Chemical Society)The outbreak of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) threatens global medical systems and economies and rules our daily living life. Controlling the outbreak of SARS-CoV-2 has become 1 of the most important and urgent strategies throughout the whole world. As of Oct. 2020, there have not yet been any medicines or therapies to be effective against SARS-CoV-2. Thus, rapid and sensitive diagnostics is the most important measures to control the outbreak of SARS-CoV-2. Homogeneous biosensing based on magnetic nanoparticles (MNPs) is 1 of the most promising approaches for rapid and highly sensitive detection of biomols. This paper proposes an approach for rapid and sensitive detection of SARS-CoV-2 with functionalized MNPs via the measurement of their magnetic response in an ac magnetic field. For proof of concept, mimic SARS-CoV-2 consisting of spike proteins and polystyrene beads are used for expts. Exptl. results demonstrate that the proposed approach allows the rapid detection of mimic SARS-CoV-2 with a limit of detection of 0.084 nM (5.9 fmol). The proposed approach has great potential for designing a low-cost and point-of-care device for rapid and sensitive diagnostics of SARS-CoV-2. - 34Ventura, 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, DOI: 10.1021/acssensors.0c01742[ACS Full Text
], [CAS], Google Scholar
34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvFaqsL7K&md5=bef9b17a1a0f63018163bcf46372c57aColorimetric Test for Fast Detection of SARS-CoV-2 in Nasal and Throat SwabsVentura, Bartolomeo Della; Cennamo, Michele; Minopoli, Antonio; Campanile, Raffaele; Censi, Sergio Bolletti; Terracciano, Daniela; Portella, Giuseppe; Velotta, RaffaeleACS Sensors (2020), 5 (10), 3043-3048CODEN: ASCEFJ; ISSN:2379-3694. (American Chemical Society)Mass testing is fundamental to face the pandemic caused by the coronavirus SARS-CoV-2 discovered at the end of 2019. To this aim, it is necessary to establish reliable, fast, and cheap tools to detect viral particles in biol. material so to identify the people capable of spreading the infection. We demonstrate that a colorimetric biosensor based on gold nanoparticle (AuNP) interaction induced by SARS-CoV-2 lends itself as an outstanding tool for detecting viral particles in nasal and throat swabs. The extinction spectrum of a colloidal soln. of multiple viral-target gold nanoparticles-AuNPs functionalized with antibodies targeting 3 surface proteins of SARS-CoV-2 (spike, envelope, and membrane)-is red-shifted in few minutes when mixed with a soln. contg. the viral particle. The optical d. of the mixed soln. measured at 560 nm was compared to the threshold cycle (Ct) of a real-time PCR (gold std. for detecting the presence of viruses) finding that the colorimetric method is able to detect very low viral load with a detection limit approaching that of the real-time PCR. Since the method is sensitive to the infecting viral particle rather than to its RNA, the achievements reported here open a new perspective not only in the context of the current and possible future pandemics, but also in microbiol., as the biosensor proves itself to be a powerful though simple tool for measuring the viral particle concn. - 35Pramanik, A.; Gao, Y.; Patibandla, S.; Mitra, D.; McCandless, M. G.; Fassero, L. A.; Gates, K.; Tandon, R.; Chandra Ray, P. The rapid diagnosis and effective inhibition of coronavirus using spike antibody attached gold nanoparticles. Nanoscale Adv. 2021, 3, 1588– 1596, DOI: 10.1039/d0na01007c[Crossref], [PubMed], [CAS], Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhsVOgu7s%253D&md5=610835de6dba6d0c6c2a97d2daf2339fThe rapid diagnosis and effective inhibition of coronavirus using spike antibody attached gold nanoparticlesPramanik, Avijit; Gao, Ye; Patibandla, Shamily; Mitra, Dipanwita; McCandless, Martin G.; Fassero, Lauren A.; Gates, Kalein; Tandon, Ritesh; Chandra Ray, PareshNanoscale Advances (2021), 3 (6), 1588-1596CODEN: NAADAI; ISSN:2516-0230. (Royal Society of Chemistry)Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the cause of the coronavirus disease that began in 2019 (COVID-19), has been responsible for 1.4 million deaths worldwide as of 13 Nov. 2020. Because at the time of writing no vaccine is yet available, a rapid diagnostic assay is very urgently needed. Herein, we present the development of anti-spike antibody attached gold nanoparticles for the rapid diagnosis of specific COVID-19 viral antigen or virus via a simple colorimetric change observation within a 5 min time period. For rapid and highly sensitive identification, surface enhanced Raman spectroscopy (SERS) was employed using 4-aminothiophenol as a reporter mol., which is attached to the gold nanoparticle via an Au-S bond. In the presence of COVID-19 antigen or virus particles, owing to the antigen-antibody interaction, the gold nanoparticles undergo aggregation, changing color from pink to blue, which allows for the detn. of the presence of antigen or virus very rapidly by the naked eye, even at concns. of 1 ng (ng) per mL for COVID-19 antigen and 1000 virus particles per mL for SARS-CoV-2 spike protein pseudotyped baculovirus. Importantly, the aggregated gold nanoparticles form "hot spots" to provide very strong SERS signal enhancement from anti-spike antibody and 4-aminothiophenol attached gold nanoparticles via light-matter interactions. Finite-difference time-domain (FDTD) simulation data indicate a 4-orders-of-magnitude Raman enhancement in "hot spot" positions when gold nanoparticles form aggregates. Using a portable Raman analyzer, our reported data demonstrate that our antibody and 4-aminothiophenol attached gold nanoparticle-based SERS probe has the capability to detect COVID-19 antigen even at a concn. of 4 pg (pg) per mL and virus at a concn. of 18 virus particles per mL within a 5 min time period. Using HEK293T cells, which express angiotensin-converting enzyme 2 (ACE2), by which SARS-CoV-2 enters human cells, we show that anti-spike antibody attached gold nanoparticles have the capability to inhibit infection by the virus. Our reported data show that antibody attached gold nanoparticles bind to SARS-CoV-2 spike protein, thereby inhibiting the virus from binding to cell receptors, which stops virus infection and spread. It also has the capability to destroy the lipid membrane of the virus.
- 36Yousefi, H.; Mahmud, A.; Chang, D.; Das, J.; Gomis, S.; Chen, J. B.; Wang, H.; Been, T.; Yip, L.; Coomes, E.; Li, Z.; Mubareka, S.; McGeer, A.; Christie, N.; Gray-Owen, S.; Cochrane, A.; Rini, J. M.; Sargent, E. H.; Kelley, S. O. Detection of SARS-CoV-2 viral particles using direct, reagent-free electrochemical sensing. J. Am. Chem. Soc. 2021, 143, 1722– 1727, DOI: 10.1021/jacs.0c10810[ACS Full Text
], [CAS], Google Scholar
36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvVWls74%253D&md5=3315d5fa3a22cd8e29f6a78f02d2665aDetection of SARS-CoV-2 Viral Particles Using Direct, Reagent-Free Electrochemical SensingYousefi, Hanie; Mahmud, Alam; Chang, Dingran; Das, Jagotamoy; Gomis, Surath; Chen, Jenise B.; Wang, Hansen; Been, Terek; Yip, Lily; Coomes, Eric; Li, Zhijie; Mubareka, Samira; McGeer, Allison; Christie, Natasha; Gray-Owen, Scott; Cochrane, Alan; Rini, James M.; Sargent, Edward H.; Kelley, Shana O.Journal of the American Chemical Society (2021), 143 (4), 1722-1727CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)The development of new methods for direct viral detection using streamlined and ideally reagent-free assays is a timely and important, but challenging, problem. The challenge of combating the COVID-19 pandemic has been exacerbated by the lack of rapid and effective methods to identify viral pathogens like SARS-CoV-2 on-demand. Existing gold std. nucleic acid-based approaches require enzymic amplification to achieve clin. relevant levels of sensitivity and are not typically used outside of a lab. setting. We report reagent-free viral sensing that directly reads out the presence of viral particles in 5 min using only a sensor-modified electrode chip. The approach relies on a class of electrode-tethered sensors bearing an analyte-binding antibody displayed on a neg. charged DNA linker that also features a tethered redox probe. When a pos. potential is applied, the sensor is transported to the electrode surface. Using chronoamperometry, the presence of viral particles and proteins can be detected as these species increase the hydrodynamic drag on the sensor. This report is the 1st virus-detecting assay that uses the kinetic response of a probe/virus complex to analyze the complexation state of the antibody. We demonstrate the performance of this sensing approach as a means to detect, within 5 min, the presence of the SARS-CoV-2 virus and its assocd. spike protein in test samples and in unprocessed patient saliva. - 37Lee, J.-H.; Choi, M.; Jung, Y.; Lee, S. K.; Lee, C.-S.; Kim, J.; Kim, J.; Kim, N. H.; Kim, B.-T.; Kim, H. G. A novel rapid detection for SARS-CoV-2 spike 1 antigens using human angiotensin converting enzyme 2 (ACE2). Biosens. Bioelectron. 2021, 171, 112715, DOI: 10.1016/j.bios.2020.112715[Crossref], [PubMed], [CAS], Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitFOisbzJ&md5=3f9071ac5e872a5880248ea47a10f3ebA novel rapid detection for SARS-CoV-2 spike 1 antigens using human angiotensin converting enzyme 2 (ACE2)Lee, Jong-Hwan; Choi, Minsuk; Jung, Yujin; Lee, Sung Kyun; Lee, Chang-Seop; Kim, Jung; Kim, Jongwoo; Kim, Nam Hoon; Kim, Bum-Tae; Kim, Hong GiBiosensors & Bioelectronics (2021), 171 (), 112715CODEN: BBIOE4; ISSN:0956-5663. (Elsevier B.V.)Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19), a newly emerging human infectious disease. Because no specific antiviral drugs or vaccines are available to treat COVID-19, early diagnostics, isolation, and prevention are crucial for contg. the outbreak. Mol. diagnostics using reverse transcription polymerase chain reaction (RT-PCR) are the current gold std. for detection. However, viral RNAs are much less stable during transport and storage than proteins such as antigens and antibodies. Consequently, false-neg. RT-PCR results can occur due to inadequate collection of clin. specimens or poor handling of a specimen during testing. Although antigen immunoassays are stable diagnostics for detection of past infection, infection progress, and transmission dynamics, no matched antibody pair for immunoassay of SARS-CoV-2 antigens has yet been reported. In this study, we designed and developed a novel rapid detection method for SARS-CoV-2 spike 1 (S1) protein using the SARS-CoV-2 receptor ACE2, which can form matched pairs with com. available antibodies. ACE2 and S1-mAb were paired with each other for capture and detection in a lateral flow immunoassay (LFIA) that did not cross-react with SARS-CoV Spike 1 or MERS-CoV Spike 1 protein. The SARS-CoV-2 S1 (<5 ng of recombinant proteins/reaction) was detected by the ACE2-based LFIA. The limit of detection of our ACE2-LFIA was 1.86 x 105 copies/mL in the clin. specimen of COVID-19 Patients without no cross-reactivity for nasal swabs from healthy subjects. This is the first study to detect SARS-CoV-2 S1 antigen using an LFIA with matched pair consisting of ACE2 and antibody. Our findings will be helpful to detect the S1 antigen of SARS-CoV-2 from COVID-19 patients.
- 38Pinals, R. L.; Ledesma, F.; Yang, D.; Navarro, N.; Jeong, S.; Pak, J. E.; Kuo, L.; Chuang, Y.-C.; Cheng, Y.-W.; Sun, H.-Y.; Landry, M. P. Rapid SARS-CoV-2 spike protein detection by carbon nanotube-based near-infrared nanosensors. Nano Lett. 2021, 21, 2272– 2280, DOI: 10.1021/acs.nanolett.1c00118[ACS Full Text
], [CAS], Google Scholar
38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXltFWjs78%253D&md5=d62b5397687debc14aac2e3e87583660Rapid SARS-CoV-2 spike protein detection by carbon nanotube-based near-infrared nanosensorsPinals, Rebecca L.; Ledesma, Francis; Yang, Darwin; Navarro, Nicole; Jeong, Sanghwa; Pak, John E.; Kuo, Lili; Chuang, Yung-Chun; Cheng, Yu-Wei; Sun, Hung-Yu; Landry, Markita P.Nano Letters (2021), 21 (5), 2272-2280CODEN: NALEFD; ISSN:1530-6984. (American Chemical Society)To effectively track and eliminate COVID-19, it is crit. to develop tools for rapid and accessible diagnosis of actively infected individuals. Here, we introduce a single-walled carbon nanotube (SWCNT)-based optical sensing approach toward this end. We construct a nanosensor based on SWCNTs noncovalently functionalized with ACE2, a host protein with high binding affinity for the SARS-CoV-2 spike protein. The presence of the SARS-CoV-2 spike protein elicits a robust, 2-fold nanosensor fluorescence increase within 90 min of spike protein exposure. We characterize the nanosensor stability and sensing mechanism and passivate the nanosensor to preserve sensing response in saliva and viral transport medium. We further demonstrate that these ACE2-SWCNT nanosensors retain sensing capacity in a surface-immobilized format, exhibiting a 73% fluorescence turn-on response within 5 s of exposure to 35 mg/L SARS-CoV-2 virus-like particles. Our data demonstrate that ACE2-SWCNT nanosensors can be developed into an optical tool for rapid SARS-CoV-2 detection. - 39Baker, A. N.; Richards, S.-J.; Guy, C. S.; Congdon, T. R.; Hasan, M.; Zwetsloot, A. J.; Gallo, A.; Lewandowski, J. R.; Stansfeld, P. J.; Straube, A.; Walker, M.; Chessa, S.; Pergolizzi, G.; Dedola, S.; Field, R. A.; Gibson, M. I. The SARS-COV-2 spike protein binds sialic acids and enables rapid detection in a lateral flow point of care diagnostic device. ACS Cent. Sci. 2020, 6, 2046– 2052, DOI: 10.1021/acscentsci.0c00855[ACS Full Text
], [CAS], Google Scholar
39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvVOlsbnP&md5=30a56fcd6d32e48834aa542773a21420The SARS-COV-2 Spike Protein Binds Sialic Acids and Enables Rapid Detection in a Lateral Flow Point of Care Diagnostic DeviceBaker, Alexander N.; Richards, Sarah-Jane; Guy, Collette S.; Congdon, Thomas R.; Hasan, Muhammad; Zwetsloot, Alexander J.; Gallo, Angelo; Lewandowski, Jozef R.; Stansfeld, Phillip J.; Straube, Anne; Walker, Marc; Chessa, Simona; Pergolizzi, Giulia; Dedola, Simone; Field, Robert A.; Gibson, Matthew I.ACS Central Science (2020), 6 (11), 2046-2052CODEN: ACSCII; ISSN:2374-7951. (American Chemical Society)There is an urgent need to understand the behavior of the novel coronavirus (SARS-COV-2), which is the causative agent of COVID-19, and to develop point-of-care diagnostics. Here, a glyconanoparticle platform is used to discover that N-acetyl neuraminic acid has affinity toward the SARS-COV-2 spike glycoprotein, demonstrating its glycan-binding function. Optimization of the particle size and coating enabled detection of the spike glycoprotein in lateral flow and showed selectivity over the SARS-COV-1 spike protein. Using a virus-like particle and a pseudotyped lentivirus model, paper-based lateral flow detection was demonstrated in under 30 min, showing the potential of this system as a low-cost detection platform. The spike-protein from SARS-COV-2 is shown to bind sialic acids, which is exploited to assemble a lateral flow diagnostic tool, using glycans rather than antibodies, as the recognition unit. - 40Tuerk, C.; Gold, L. Systematic evolution of ligands by exponential enrichment: RNA ligands to bacteriophage T4 DNA polymerase. Science 1990, 249, 505– 510, DOI: 10.1126/science.2200121[Crossref], [PubMed], [CAS], Google Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3cXlt1OltLY%253D&md5=a40a28db9df4a5da4c1b1652a34aa715Systematic evolution of ligands by exponential enrichment: RNA ligands to bacteriophage T4 DNA polymeraseTuerk, Craig; Gold, LarryScience (Washington, DC, United States) (1990), 249 (4968), 505-10CODEN: SCIEAS; ISSN:0036-8075.A novel method was developed for rapidly selecting preferred binding sequences from a population of random sequences. The method is designated systematic evolution of ligands by exponential enrichment (SELEX). High-affinity nucleic acid ligands for a protein were isolated by a procedure that depends on alternate cycles of ligand selection from pools of variant sequences and amplification of the bound species. Multiple rounds exponentially enrich the population for the highest affinity species that can be clonally isolated and characterized. In particular one eight-base region of an RNA that interacts with the T4 DNA polymerase was chosen and randomized. Two different sequences were selected by this procedure from the calcd. pool of 65,536 species. One is the wild-type sequence found in the bacteriophage mRNA; one is varied from wild type at four positions. The binding consts. of these two RNA's to T4 DNA polymerase are equiv. These protocols with minimal modification can yield high-affinity ligands for any protein that binds nucleic acids as part of its function; high affinity ligands could conceivably be developed for any target mol.
- 41Ellington, A. D.; Szostak, J. W. In vitro selection of RNA molecules that bind specific ligands. Nature 1990, 346, 818– 822, DOI: 10.1038/346818a0[Crossref], [PubMed], [CAS], Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3MXitVGgsw%253D%253D&md5=522ad1b4f0284ee88173b02035f9e9c5In vitro selection of RNA molecules that bind specific ligandsEllington, Andrew D.; Szostak, Jack W.Nature (London, United Kingdom) (1990), 346 (6287), 818-22CODEN: NATUAS; ISSN:0028-0836.Subpopulations of RNA mols. that bind specifically to a variety of org. dyes have been isolated from a population of random sequence RNA mols. Roughly one in 1010 random sequence RNA mols. folds in such a way as to create a specific binding site for small ligands.
- 42Robertson, D. L.; Joyce, G. F. Selection in vitro of an RNA enzyme that specifically cleaves single-stranded DNA. Nature 1990, 344, 467– 468, DOI: 10.1038/344467a0[Crossref], [PubMed], [CAS], Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3cXitlWltLk%253D&md5=9f3ef5e76f028aa84277861930d98189Selection in vitro of an RNA enzyme that specifically cleaves single-stranded DNARobertson, Debra L.; Joyce, Gerald F.Nature (London, United Kingdom) (1990), 344 (6265), 467-8CODEN: NATUAS; ISSN:0028-0836.The discovery of RNA enzymes has, for the 1st time, provided a single mol. that has both genetic and catalytic properties. Techniques were previously devised for the mutation, selection, and amplification of catalytic RNA, all of which can be performed rapidly in vitro. This report describes how these techniques can be integrated and performed repeatedly within a single reaction vessel. This allows evolution expts. to be carried out in response to artificially imposed selection constraints. The subject of the investigation was the Tetrahymena ribozyme, a self-splicing group I intron derived from the large rRNA precursor of Tetrahymena thermophila that catalyzes sequence-specific phosphoester transfer reactions involving RNA substrates. It consists of 413 nucleotides, and assumes a well-defined secondary and tertiary structure responsible for its catalytic activity. Variant forms of the enzyme were selected that could best react with a DNA substrate. This led to the recovery of a mutant form of the enzyme that cleaves DNA more efficiently than the wild-type enzyme. The selected mol. represents the discovery of the 1st RNA enzyme known to cleave single-stranded DNA specifically.
- 43Nguyen, V.-T.; Seo, H. B.; Kim, B. C.; Kim, S. K.; Song, C.-S.; Gu, M. B. Highly sensitive sandwich-type SPR based detection of whole H5Nx viruses using a pair of aptamers. Biosens. Bioelectron. 2016, 86, 293– 300, DOI: 10.1016/j.bios.2016.06.064[Crossref], [PubMed], [CAS], Google Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhtFSntrbN&md5=802a134f713241370fc1dabb2e5cd2d3Highly sensitive sandwich-type SPR based detection of whole H5Nx viruses using a pair of aptamersNguyen, Van-Thuan; Seo, Ho Bin; Kim, Byoung Chan; Kim, Sang Kyung; Song, Chang-Seon; Gu, Man BockBiosensors & Bioelectronics (2016), 86 (), 293-300CODEN: BBIOE4; ISSN:0956-5663. (Elsevier B.V.)In this research, we report highly sensitive and specific sandwich-type SPR-based biosensor for the detection H5Nx whole viruses. A few of aptamers, for the first time, were successfully screened and characterized for whole avian influenza (AI) viruses, H5Nx, by using Multi-GO-SELEX method. The affinities of the aptamers developed in this study were ranged from 8×104 to 1×104 EID50/mL, and the aptamers IF22, IF23 were found to be specific to H5N1 and H5N8, resp. In addn., some flexible aptamers IF20, IF15, and IF10 were found to bind to the H5N1 and H5N2, H5N1 and H5N8, or H5N1, H5N2, and H5N8, resp. Moreover, aptamers IF10 and IF22 were found to bind H5N1 virus simultaneously and confirmed to bind the different site of the same H5N1 whole virus. Therefore, this pair of aptamers, IF10 and IF22, were successfully applied to develop the sandwich-type SPR-based biosensor assay which is rapid, accurate for the detection of AI whole virus from H5N1-infected feces samples. The min. detectible concn. of H5N1 whole virus was found to be 200 EID50/mL with this sandwich-type detection using the aptamer pair obtained in this study. In addn., the sensitivity of this biosensor was successfully enhanced by using the signal amplification with the secondary aptamer conjugated with gold nanoparticles.
- 44Park, J.-W.; Jin Lee, S.; Choi, E.-J.; Kim, J.; Song, J.-Y.; Bock Gu, M. An ultra-sensitive detection of a whole virus using dual aptamers developed by immobilization-free screening. Biosens. Bioelectron. 2014, 51, 324– 329, DOI: 10.1016/j.bios.2013.07.052[Crossref], [PubMed], [CAS], Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsFGqt77F&md5=58b49b9f4f48fa7fcc3ba564bc1a3cebAn ultra-sensitive detection of a whole virus using dual aptamers developed by immobilization-free screeningPark, Jee-Woong; Jin Lee, Su; Choi, Eun-Jin; Kim, Jaejo; Song, Jae-Young; Bock Gu, ManBiosensors & Bioelectronics (2014), 51 (), 324-329CODEN: BBIOE4; ISSN:0956-5663. (Elsevier B.V.)In this study, we successfully developed a ssDNA aptamer pairs by using an advanced immobilization-free SELEX method with affinity-based selection and counter-screening process at every round. By implementing this method, two different aptamers specifically binding to bovine viral diarrhea virus type 1 (BVDV type 1) with high affinity were successfully screened. This aptamer pair was applied to ultrasensitive detection platform for BVDV type 1 in a sandwich manner. The ultrasensitive detection of BVDV type 1 using one of aptamers conjugated with gold nanoparticles was obtained in aptamer-aptamer sandwich type sensing format, with the limit of detection of 800 copies/mL, which is comparable to a real-time PCR method.
- 45Seo, H. B.; Gu, M. B. Aptamer-based sandwich-type biosensors. J. Biol. Eng. 2017, 11, 11, DOI: 10.1186/s13036-017-0054-7[Crossref], [PubMed], [CAS], Google Scholar45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXkslyrt7o%253D&md5=b10920617d0342cd0717648a30003830Aptamer-based sandwich-type biosensorsSeo, Ho Bin; Gu, Man BockJournal of Biological Engineering (2017), 11 (), 11/1-11/7CODEN: JBEOBZ; ISSN:1754-1611. (BioMed Central Ltd.)Sandwich-type biosensor platforms have drawn lots of attentions due to its superior features, compared to other platforms, in terms of its stable and reproducible responses and easy enhancement in the detection sensitivity. The sandwich-type assays can be developed by utilizing a pair of receptors, which bind to the different sites of the same target. In this mini-review paper, the sandwich-type biosensors using either pairs of aptamers or aptamer-antibody pairs are reviewed in terms of its targets and platforms, the schematic designs, and their anal. performance.
- 46Song, Y.; Song, J.; Wei, X.; Huang, M.; Sun, M.; Zhu, L.; Lin, B.; Shen, H.; Zhu, Z.; Yang, C. Discovery of aptamers targeting the receptor-binding domain of the SARS-CoV-2 spike glycoprotein. Anal. Chem. 2020, 92, 9895– 9900, DOI: 10.1021/acs.analchem.0c01394[ACS Full Text
], [CAS], Google Scholar
46https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtF2hsr7E&md5=03a6de77c4f4834eeee88edb93b82540Discovery of Aptamers Targeting the Receptor-Binding Domain of the SARS-CoV-2 Spike GlycoproteinSong, Yanling; Song, Jia; Wei, Xinyu; Huang, Mengjiao; Sun, Miao; Zhu, Lin; Lin, Bingqian; Shen, Haicong; Zhu, Zhi; Yang, ChaoyongAnalytical Chemistry (Washington, DC, United States) (2020), 92 (14), 9895-9900CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)The World Health Organization has declared the outbreak of a novel coronavirus (SARS-CoV-2 or 2019-nCoV) as a global pandemic. However, the mechanisms behind the coronavirus infection are not yet fully understood, nor are there any targeted treatments or vaccines. In this study, we identified high-binding-affinity aptamers targeting SARS-CoV-2 RBD, using an ACE2 competition-based aptamer selection strategy and a machine learning screening algorithm. The Kd values of the optimized CoV2-RBD-1C and CoV2-RBD-4C aptamers against RBD were 5.8 nM and 19.9 nM, resp. Simulated interaction modeling, along with competitive expts., suggests that two aptamers may have partially identical binding sites at ACE2 on SARS-CoV-2 RBD. These aptamers present an opportunity for generating new probes for recognition of SARS-CoV-2 and could provide assistance in the diagnosis and treatment of SARS-CoV-2 while providing a new tool for in-depth study of the mechanisms behind the coronavirus infection. - 47Stanborough, T.; Given, F. M.; Koch, B.; Sheen, C. R.; Stowers-Hull, A. B.; Waterland, M. R.; Crittenden, D. L. Optical detection of CoV-SARS-2 viral proteins to sub-picomolar concentrations. ACS Omega 2021, 6, 6404– 6413, DOI: 10.1021/acsomega.1c00008[ACS Full Text
], [CAS], Google Scholar
47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXkslynsb0%253D&md5=d0681c670ebd20831680d4698ba49973Optical detection of CoV-SARS-2 viral proteins to sub-picomolar concentrationsStanborough, Tamsyn; Given, Fiona M.; Koch, Barbara; Sheen, Campbell R.; Stowers-Hull, Andre Buzas; Waterland, Mark R.; Crittenden, Deborah L.ACS Omega (2021), 6 (9), 6404-6413CODEN: ACSODF; ISSN:2470-1343. (American Chemical Society)The emergence of a new strain of coronavirus in late 2019, SARS-CoV-2, led to a global pandemic in 2020. This may have been preventable if large scale, rapid diagnosis of active cases had been possible, and this has highlighted the need for more effective and efficient ways of detecting and managing viral infections. In this work, we investigate three different optical techniques for quantifying the binding of recombinant SARS-CoV-2 spike protein to surface-immobilized oligonucleotide aptamers. Biolayer interferometry is a relatively cheap, robust, and rapid method that only requires very small sample vols. However, its detection limit of 250 nM means that it is not sensitive enough to detect antigen proteins at physiol. relevant levels (sub-pM). Surface plasmon resonance is a more sensitive technique but requires larger sample vols., takes longer, requires more expensive instrumentation, and only reduces the detection limit to 5 nM. Surface-enhanced Raman spectroscopy is far more sensitive, enabling detection of spike protein to sub-picomolar concns. Control expts. performed using scrambled aptamers and using bovine serum albumin as an analyte show that this apta-sensing approach is both sensitive and selective, with no appreciable response obsd. for any controls. Overall, these proof-of-principle results demonstrate that SERS-based aptasensors hold great promise for development into rapid, point-of-use antigen detection systems, enabling mass testing without any need for reagents or lab. expertise and equipment. - 48Pramanik, A.; Gao, Y.; Patibandla, S.; Mitra, D.; McCandless, M. G.; Fassero, L. A.; Gates, K.; Tandon, R.; Ray, P. C. Aptamer conjugated gold nanostar-based distance-dependent nanoparticle surface energy transfer spectroscopy for ultrasensitive detection and inactivation of corona virus. J. Phys. Chem. Lett. 2021, 12, 2166– 2171, DOI: 10.1021/acs.jpclett.0c03570[ACS Full Text
], [CAS], Google Scholar
48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXkvFamurw%253D&md5=8da4b26bba915f2721e957132a26eb96Aptamer conjugated gold nanostar-based distance-dependent nanoparticle surface energy transfer spectroscopy for ultrasensitive detection and inactivation of corona virusPramanik, Avijit; Gao, Ye; Patibandla, Shamily; Mitra, Dipanwita; McCandless, Martin G.; Fassero, Lauren A.; Gates, Kalein; Tandon, Ritesh; Ray, Paresh ChandraJournal of Physical Chemistry Letters (2021), 12 (8), 2166-2171CODEN: JPCLCD; ISSN:1948-7185. (American Chemical Society)The ongoing outbreak of the coronavirus infection has killed more than 2 million people. Herein, we demonstrate that Rhodamine 6G (Rh-6G) dye conjugated DNA aptamer-attached gold nanostar (GNS)-based distance-dependent nanoparticle surface energy transfer (NSET) spectroscopy has the capability of rapid diagnosis of specific SARS-CoV-2 spike recombinant antigen or SARS-CoV-2 spike protein pseudotyped baculovirus within 10 min. Because Rh-6G-attached single-stand DNA aptamer wrapped the GNS, 99% dye fluorescence was quenched because of the NSET process. In the presence of spike antigen or virus, the fluorescence signal persists because of the aptamer-spike protein binding. Specifically, the limit of detection for the NSET assay has been detd. to be 130 fg/mL for antigen and 8 particles/mL for virus. Finally, we have demonstrated that DNA aptamer-attached GNSs can stop virus infection by blocking the angiotensin-converting enzyme 2 (ACE2) receptor binding capability and destroying the lipid membrane of the virus. - 49Schmitz, A.; Weber, A.; Bayin, M.; Breuers, S.; Fieberg, V.; Famulok, M.; Mayer, G. A SARS-CoV-2 spike binding DNA aptamer that inhibits pseudovirus infection by an RBD-independent mechanism. Angew. Chem., Int. Ed. 2021, 60, 2– 9, DOI: 10.1101/2020.12.23.424171
- 50Yang, G.; Li, Z.; Mohammed, I.; Zhao, L.; Wei, W.; Xiao, H.; Guo, W.; Zhao, Y.; Qu, F.; Huang, Y. Identification of SARS-CoV-2-against aptamer with high neutralization activity by blocking the RBD domain of spike protein 1. Signal Transduction Targeted Ther. 2021, 6, 227, DOI: 10.1038/s41392-021-00649-6[Crossref], [PubMed], [CAS], Google Scholar50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhtlegtbzE&md5=368c38793a514ca573275b119ca3c012Identification of SARS-CoV-2-against aptamer with high neutralization activity by blocking the RBD domain of spike protein 1Yang, Ge; Li, Ziyue; Mohammed, Irfan; Zhao, Liping; Wei, Wei; Xiao, Haihua; Guo, Weisheng; Zhao, Yongxiang; Qu, Feng; Huang, YuanyuSignal Transduction and Targeted Therapy (2021), 6 (1), 227CODEN: STTTCB; ISSN:2059-3635. (Nature Research)Identification of SARS-CoV-2-against aptamer with high neutralization activity by blocking the RBD domain of spike protein 1.
- 51Li, J.; Zhang, Z.; Gu, J.; Stacey, H. D.; Ang, J. C.; Capretta, A.; Filipe, C. D. M.; Mossman, K. L.; Balion, C.; Salena, B. J.; Yamamura, D.; Soleymani, L.; Miller, M. S.; Brennan, J. D.; Li, Y. Diverse high-affinity DNA aptamers for wild-type and B.1.1.7 SARS-CoV-2 spike proteins from a pre-structured DNA library. Nucleic Acids Res. 2021, 49, 7267– 7279, DOI: 10.1093/nar/gkab574[Crossref], [PubMed], [CAS], Google Scholar51https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXit12ru7jL&md5=05d191a9e9455c4a809aa0dc261efb3fDiverse high-affinity DNA aptamers for wild-type and B.1.1.7 SARS-CoV-2 spike proteins from a pre-structured DNA libraryLi, Jiuxing; Zhang, Zijie; Gu, Jimmy; Stacey, Hannah D.; Ang, Jann C.; Capretta, Alfredo; Filipe, Carlos D. M.; Mossman, Karen L.; Balion, Cynthia; Salena, Bruno J.; Yamamura, Deborah; Soleymani, Leyla; Miller, Matthew S.; Brennan, John D.; Li, YingfuNucleic Acids Research (2021), 49 (13), 7267-7279CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)We performed in vitro selection expts. to identify DNA aptamers for the S1 subunit of the SARS-CoV-2 spike protein (S1 protein). Using a pool of pre-structured random DNA sequences, we obtained over 100 candidate aptamers after 13 cycles of enrichment under progressively more stringent selection pressure. The top 10 sequences all exhibited strong binding to the S1 protein. Two aptamers, named MSA1 (Kd = 1.8 nM) and MSA5 (Kd = 2.7 nM), were assessed for binding to the heat-treated S1 protein, untreated S1 protein spiked into 50% human saliva and the trimeric spike protein of both the wildtype and the B.1.1.7 variant, demonstrating comparable affinities in all cases. MSA1 and MSA5 also recognized the pseudotyped lentivirus of SARS-CoV-2 with resp. Kd values of 22.7 pM and 11.8 pM. Secondary structure prediction and sequence truncation expts. revealed that both MSA1 and MSA5 adopted a hairpin structure, which was the motif pre-designed into the original library. A colorimetric sandwich assay was developed using MSA1 as both the recognition element and detection element, which was capable of detecting the pseudotyped lentivirus in 50% saliva with a limit of detection of 400 fM, confirming the potential of these aptamers as diagnostic tools for COVID-19 detection.
- 52Gupta, A.; Anand, A.; Jain, N.; Goswami, S.; Anantharaj, A.; Patil, S.; Singh, R.; Kumar, A.; Shrivastava, T.; Bhatnagar, S.; Medigeshi, G. R.; Sharma, T. K.; DBT India Consortium for COVID-19 Research A novel G-quadruplex aptamer-based spike trimeric antigen test for the detection of SARS-CoV-2. Mol. Ther.--Nucleic Acids 2021, 26, 321– 332, DOI: 10.1016/j.omtn.2021.06.014[Crossref], [PubMed], [CAS], Google Scholar52https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvFGrt7nI&md5=9dd950049a6d402e6c37542d4e66185dA novel G-quadruplex aptamer-based spike trimeric antigen test for the detection of SARS-CoV-2Gupta, Ankit; Anand, Anjali; Jain, Neha; Goswami, Sandeep; Anantharaj, Anbalagan; Patil, Sharanabasava; Singh, Rahul; Kumar, Amit; Shrivastava, Tripti; Bhatnagar, Shinjini; Medigeshi, Guruprasad R.; Sharma, Tarun KumarMolecular Therapy--Nucleic Acids (2021), 26 (), 321-332CODEN: MTAOC5; ISSN:2162-2531. (Elsevier Inc.)The recent SARS-CoV-2 outbreak has been declared a global health emergency. It will take years to vaccinate the whole population to protect them from this deadly virus, hence the management of SARS-CoV-2 largely depends on the widespread availability of an accurate diagnostic test. Toward addressing the unmet need of a reliable diagnostic test in the current work by utilizing the power of Systematic Evolution of Ligands by EXponential enrichment, a 44-mer G-quadruplex-forming DNA aptamer against spike trimer antigen of SARS-CoV-2 was identified. The lead aptamer candidate (S14) was characterized thoroughly for its binding, selectivity, affinity, structure, and batch-to-batch variability by utilizing various biochem., biophys., and in silico techniques. S14 has demonstrated a low nanomolar KD, confirming its tight binding to a spike antigen of SARS-CoV-2. S14 can detect as low as 2 nM of antigen. The clin. evaluation of S14 aptamer on nasopharyngeal swab specimens (n = 232) has displayed a highly discriminatory response between SARS-CoV-2 infected individuals from the non-infected one with a sensitivity and specificity of ∼91% and 98%, resp. Importantly, S14 aptamer-based test has evinced a comparable performance with that of RT-PCR-based assay. Altogether, this study established the utility of aptamer technol. for the detection of SARS-CoV-2.
- 53Alves Ferreira-Bravo, I.; DeStefano, J. J. Xeno-nucleic Acid (XNA) 2’-fluoro-arabino nucleic acid (FANA) aptamers to the receptor-binding domain of SARS-CoV-2 S protein block ACE2 binding. Viruses 2021, 13, 1983, DOI: 10.3390/v13101983[Crossref], [PubMed], [CAS], Google Scholar53https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXitlOhsL%252FL&md5=4b8c95f265f1068e63decbcbbb1dc5f7Xeno-Nucleic Acid (XNA) 2'-Fluoro-Arabino Nucleic Acid (FANA) Aptamers to the Receptor-Binding Domain of SARS-CoV-2 S Protein Block ACE2 BindingAlves Ferreira-Bravo, Irani; DeStefano, Jeffrey J.Viruses (2021), 13 (10), 1983CODEN: VIRUBR; ISSN:1999-4915. (MDPI AG)The causative agent of COVID-19, SARS-CoV-2, gains access to cells through interactions of the receptor-binding domain (RBD) on the viral S protein with angiotensin-converting enzyme 2 (ACE2) on the surface of human host cells. Systematic evolution of ligands by exponential enrichment (SELEX) was used to generate aptamers (nucleic acids selected for high binding affinity to a target) to the RBD made from 2'-fluoro-arabinonucleic acid (FANA). The best selected ∼79 nucleotide aptamers bound the RBD (Arg319-Phe541) and the larger S1 domain (Val16-Arg685) of the 1272 amino acid S protein with equil. dissocn. consts. (KD,app) of 10-20 nM, and binding half-life for the RBD, S1 domain, and full trimeric S protein of 53 ± 18, 76 ± 5, and 127 ± 7 min, resp. Aptamers inhibited the binding of the RBD to ACE2 in an ELISA assay. Inhibition, on a per wt. basis, was similar to neutralizing antibodies that were specific for RBD. Aptamers demonstrated high specificity, binding with about 10-fold lower affinity to the related S1 domain from the original SARS virus, which also binds to ACE2. Overall, FANA aptamers show affinities comparable to previous DNA aptamers to RBD and S1 protein and directly block receptor interactions while using an alternative Xeno-nucleic acid (XNA) platform.
- 54Behbahani, M.; Mohabatkar, H.; Hosseini, B. In silico design of quadruplex aptamers against the spike protein of SARS-CoV-2. Inf. Med. 2021, 26, 100757, DOI: 10.1016/j.imu.2021.100757[Crossref], [CAS], Google Scholar54https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB2cjhs12iuw%253D%253D&md5=df8b1a0d86351a2967314d233e85f648In silico design of quadruplex aptamers against the spike protein of SARS-CoV-2Behbahani Mandana; Mohabatkar Hassan; Hosseini BarumandInformatics in medicine unlocked (2021), 26 (), 100757 ISSN:2352-9148.Nucleic acid aptamers are short sequences of nucleic acid ligands that bind to a specific target molecule. Aptamers are experimentally nominated using the well-designed SELEX (systematic evolution of ligands by exponential enrichment) method. Here, we designed a new method for diagnosis and blocking SARS-CoV-2 based on G-quadruplex aptamer. This aptamer was developed against the receptor-binding domain (RBD) region of the spike protein. In the current study, ten quadruplex DNA aptamers entitled AP1, AP2, AP3, AP4, AP5, AP6, AP7, AP8, AP9, and AP10 were designed in silico and had high HADDOCK scores. One quadruplex aptamer sequence (AP1) was selected based on the interaction with RBD of SARS-CoV-2. Results showed that AP1 aptamer could be used as an agent in the diagnosis and therapy of SARS-CoV-2, although more works are still needed.
- 55Devi, A.; Chaitanya, N. S. N. Designing of peptide aptamer targeting the receptor-binding domain of spike protein of SARS-CoV-2: an in silico study. Mol. Diversity 2021, 1– 13, DOI: 10.1007/s11030-020-10171-6
- 56Artyushenko, P. V.; Mironov, V. A.; Mironov, V. A.; Morozov, D. I.; Shchugoreva, I. A.; Borbone, N.; Tomilin, F. N.; Kichkailo, A. S. Computational approach to design of aptamers to the receptor binding domain of SARS-CoV-2. Sib. Med. Rev. 2021, 2, 66– 67, DOI: 10.20333/25000136-2021-2-66-67
- 57Kowada, A. Greater public health impact of COVID-19 antigen detection tests. BMC Med. 2021, 19, 82, DOI: 10.1186/s12916-021-01956-z[Crossref], [PubMed], [CAS], Google Scholar57https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXnsVGgsrc%253D&md5=697f8c67d8f02c80f7b7769f03a1aa19Greater public health impact of COVID-19 antigen detection testsKowada, AkikoBMC Medicine (2021), 19 (1), 82CODEN: BMMECZ; ISSN:1741-7015. (BioMed Central Ltd.)Early diagnosis of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection prevents widespread transmission, manages outbreak control, improves poor prognosis, and reduces mortality of the Coronavirus disease 2019 (COVID-19). Ricks et al. report in BMC Medicine greater healthcare cost-saving and health outcomes of antigen detection rapid diagnostic tests compared to nucleic acid tests for symptomatic persons. The main outcomes in the study are the health system costs and health impacts (deaths averted and infectious days isolated). Antigen-detection rapid diagnostic test-led strategy is compared to a strategy based on nucleic acid tests and clin. judgment.
- 58Pomplun, S.; Jbara, M.; Quartararo, A. J.; Zhang, G.; Brown, J. S.; Lee, Y.-C.; Ye, X.; Hanna, S.; Pentelute, B. L. De novo discovery of high-affinity peptide binders for the SARS-CoV-2 spike protein. ACS Cent. Sci. 2021, 7, 156– 163, DOI: 10.1021/acscentsci.0c01309[ACS Full Text
], [CAS], Google Scholar
58https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXis1WrsbfM&md5=1cee08cf67569ada5d5ca76bdd2ae059De novo discovery of high-affinity peptide binders for the SARS-CoV-2 spike proteinPomplun, Sebastian; Jbara, Muhammad; Quartararo, Anthony J.; Zhang, Genwei; Brown, Joseph S.; Lee, Yen-Chun; Ye, Xiyun; Hanna, Stephanie; Pentelute, Bradley L.ACS Central Science (2021), 7 (1), 156-163CODEN: ACSCII; ISSN:2374-7951. (American Chemical Society)The β-coronavirus SARS-CoV-2 has caused a global pandemic. Affinity reagents targeting the SARS-CoV-2 spike protein are of interest for the development of therapeutics and diagnostics. We used affinity selection-mass spectrometry for the rapid discovery of synthetic high-affinity peptide binders for the receptor binding domain (RBD) of the SARS-CoV-2 spike protein. From library screening with 800 million synthetic peptides, we identified three sequences with nanomolar affinities (dissocn. consts. Kd = 80-970 nM) for RBD and selectivity over human serum proteins. Nanomolar RBD concns. in a biol. matrix could be detected using the biotinylated lead peptide in ELISA format. These peptides do not compete for ACE2 binding, and their site of interaction on the SARS-CoV-2-spike-RBD might be unrelated to the ACE2 binding site, making them potential orthogonal reagents for sandwich immunoassays. These findings serve as a starting point for the development of SARS-CoV-2 diagnostics or conjugates for virus-directed delivery of therapeutics. Using affinity selection-mass spectrometry, we discovered synthetic peptides with nanomolar affinity for the SARS-CoV-2-spike-RBD and selectivity over human proteins. - 59Kaur, N.; Singh, R.; Dar, Z.; Bijarnia, R. K.; Dhingra, N.; Kaur, T. Genetic comparison among various coronavirus strains for the identification of potential vaccine targets of SARS-CoV2. Infect., Genet. Evol. 2021, 89, 104490, DOI: 10.1016/j.meegid.2020.104490[Crossref], [PubMed], [CAS], Google Scholar59https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXivVWrsr0%253D&md5=dbed715a9b6a000a681c9d83d1e136b5Genetic comparison among various coronavirus strains for the identification of potential vaccine targets of SARS-CoV2Kaur, Navpreet; Singh, Rimaljot; Dar, Zahid; Bijarnia, Rakesh Kumar; Dhingra, Neelima; Kaur, TanzeerInfection, Genetics and Evolution (2021), 89 (), 104490CODEN: IGENCN; ISSN:1567-1348. (Elsevier B.V.)A review. On-going pandemic pneumonia outbreak COVID-19 has raised an urgent public health issue worldwide impacting millions of people with a continuous increase in both morbidity and mortality. The causative agent of this disease is identified and named as SARS-CoV2 because of its genetic relatedness to SARS-CoV species that was responsible for the 2003 coronavirus outbreak. The immense spread of the disease in a very small period demands urgent development of therapeutic and prophylactic interventions for the treatment of SARS-CoV2 infected patients. A plethora of research is being conducted globally on this novel coronavirus strain to gain knowledge about its origin, evolutionary history, and phylogeny. This review is an effort to compare genetic similarities and diversifications among coronavirus strains, which can hint towards the susceptible antigen targets of SARS-CoV2 to come up with the potential therapeutic and prophylactic interventions for the prevention of this public threat.
- 60Loos, C.; Atyeo, C.; Fischinger, S.; Burke, J.; Slein, M. D.; Streeck, H.; Lauffenburger, D.; Ryan, E. T.; Charles, R. C.; Alter, G. Evolution of early SARS-CoV-2 and cross-coronavirus immunity. mSphere 2020, 5, e00622– 20, DOI: 10.1128/mSphere.00622-20[Crossref], [PubMed], [CAS], Google Scholar60https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvFOksbzK&md5=e23c18b6205cff7e4addd50d989c6441Evolution of early SARS-CoV-2 and cross-coronavirus immunityLoos, Carolin; Atyeo, Caroline; Fischinger, Stephanie; Burke, John; Slein, Matthew D.; Streeck, Hendrik; Lauffenburger, Douglas; Ryan, Edward T.; Charles, Richelle C.; Alter, GalitmSphere (2020), 5 (5), e00622-20CODEN: MSPHCI; ISSN:2379-5042. (American Society for Microbiology)The novel coronavirus, SARS-coronavirus (CoV)-2 (SARS-CoV-2), has caused over 17 million infections in just a few months, with disease manifestations ranging from largely asymptomatic infection to critically severe disease. The remarkable spread and unpredictable disease outcomes continue to challenge management of this infection. Among the hypotheses to explain the heterogeneity of symptoms is the possibility that exposure to other coronaviruses (CoVs), or overall higher capability to develop immunity against respiratory pathogens, may influence the evolution of immunity to SARS-CoV-2. Thus, we profiled the immune response across multiple coronavirus receptor binding domains (RBDs), respiratory viruses, and SARS-CoV-2, to det. whether heterologous immunity to other CoV-RBDs or other infections influenced the evolution of the SARS-CoV-2 humoral immune response. Overall changes in subclass, isotype, and Fc-receptor binding were profiled broadly across a cohort of 43 individuals against different coronaviruses-RBDs of SARS-CoV-2 and the more common HKU1 and NL63 viruses. We found rapid functional evolution of responses to SARS-CoV-2 over time, along with broad but relatively more time-invariant responses to the more common CoVs. Moreover, there was little evidence of correlation between SARS-CoV-2 responses and HKU1, NL63, and respiratory infection (influenza and respiratory syncytial virus) responses. These findings suggest that common viral infections including common CoV immunity, targeting the receptor binding domain involved in viral infection, do not appear to influence the rapid functional evolution of SARS-CoV-2 immunity, and thus should not impact diagnostics or shape vaccine-induced immunity.
- 61Li, F. Structure, function, and evolution of coronavirus spike proteins. Annu. Rev. Virol. 2016, 3, 237– 261, DOI: 10.1146/annurev-virology-110615-042301[Crossref], [PubMed], [CAS], Google Scholar61https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsVegsb%252FF&md5=7a64c5330380b5e82b65594512294ffbStructure, Function, and Evolution of Coronavirus Spike ProteinsLi, FangAnnual Review of Virology (2016), 3 (), 237-261CODEN: ARVNBS; ISSN:2327-0578. (Annual Reviews)The coronavirus spike protein is a multifunctional mol. machine that mediates coronavirus entry into host cells. It first binds to a receptor on the host cell surface through its S1 subunit and then fuses viral and host membranes through its S2 subunit. Two domains in S1 from different coronaviruses recognize a variety of host receptors, leading to viral attachment. The spike protein exists in two structurally distinct conformations, prefusion and postfusion. The transition from prefusion to postfusion conformation of the spike protein must be triggered, leading to membrane fusion. This article reviews current knowledge about the structures and functions of coronavirus spike proteins, illustrating how the two S1 domains recognize different receptors and how the spike proteins are regulated to undergo conformational transitions. I further discuss the evolution of these two crit. functions of coronavirus spike proteins, receptor recognition and membrane fusion, in the context of the corresponding functions from other viruses and host cells.
- 62Andersen, K. G.; Rambaut, A.; Lipkin, W. I.; Holmes, E. C.; Garry, R. F. The proximal origin of SARS-CoV-2. Nat. Med. 2020, 26, 450– 452, DOI: 10.1038/s41591-020-0820-9[Crossref], [PubMed], [CAS], Google Scholar62https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXltFCjtbY%253D&md5=3489259c33e29365c0c1cf7fc5613407The proximal origin of SARS-CoV-2Andersen, Kristian G.; Rambaut, Andrew; Lipkin, W. Ian; Holmes, Edward C.; Garry, Robert F.Nature Medicine (New York, NY, United States) (2020), 26 (4), 450-452CODEN: NAMEFI; ISSN:1078-8956. (Nature Research)There is no expanded citation for this reference.
- 63Ou, X.; Liu, Y.; Lei, X.; Li, P.; Mi, D.; Ren, L.; Guo, L.; Guo, R.; Chen, T.; Hu, J.; Xiang, Z.; Mu, Z.; Chen, X.; Chen, J.; Hu, K.; Jin, Q.; Wang, J.; Qian, Z. Characterization of spike glycoprotein of SARS-CoV-2 on virus entry and its immune cross-reactivity with SARS-CoV. Nat. Commun. 2020, 11, 1620, DOI: 10.1038/s41467-020-15562-9[Crossref], [PubMed], [CAS], Google Scholar63https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXlvFyjt78%253D&md5=6b0b1ef5a68f4a35da4aabecb0f99544Characterization of spike glycoprotein of SARS-CoV-2 on virus entry and its immune cross-reactivity with SARS-CoVOu, Xiuyuan; Liu, Yan; Lei, Xiaobo; Li, Pei; Mi, Dan; Ren, Lili; Guo, Li; Guo, Ruixuan; Chen, Ting; Hu, Jiaxin; Xiang, Zichun; Mu, Zhixia; Chen, Xing; Chen, Jieyong; Hu, Keping; Jin, Qi; Wang, Jianwei; Qian, ZhaohuiNature Communications (2020), 11 (1), 1620CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)Since 2002, beta coronaviruses (CoV) have caused three zoonotic outbreaks, SARS-CoV in 2002-2003, MERS-CoV in 2012, and the newly emerged SARS-CoV-2 in late 2019. However, little is currently known about the biol. of SARS-CoV-2. Here, using SARS-CoV-2 S protein pseudovirus system, we confirm that human angiotensin converting enzyme 2 (hACE2) is the receptor for SARS-CoV-2, find that SARS-CoV-2 enters 293/hACE2 cells mainly through endocytosis, that PIKfyve, TPC2, and cathepsin L are crit. for entry, and that SARS-CoV-2 S protein is less stable than SARS-CoV S. Polyclonal anti-SARS S1 antibodies T62 inhibit entry of SARS-CoV S but not SARS-CoV-2 S pseudovirions. Further studies using recovered SARS and COVID-19 patients' sera show limited cross-neutralization, suggesting that recovery from one infection might not protect against the other. Our results present potential targets for development of drugs and vaccines for SARS-CoV-2.
- 64Idili, A.; Parolo, C.; Alvarez-Diduk, R.; Merkoçi, A. Rapid and efficient detection of the SARS-CoV-2 spike protein using an electrochemical aptamer-based sensor. ACS Sens. 2021, 6, 3093– 3101, DOI: 10.1021/acssensors.1c01222[ACS Full Text
], [CAS], Google Scholar
64https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhslegsLvI&md5=eed8021b69f60f966c493363ef2aa479Rapid and Efficient Detection of the SARS-CoV-2 Spike Protein Using an Electrochemical Aptamer-Based SensorIdili, Andrea; Parolo, Claudio; Alvarez-Diduk, Ruslan; Merkoci, ArbenACS Sensors (2021), 6 (8), 3093-3101CODEN: ASCEFJ; ISSN:2379-3694. (American Chemical Society)The availability of sensors able to rapidly detect SARS-CoV-2 directly in biol. fluids in a single step would allow performing massive diagnostic testing to track in real time and contain the spread of COVID-19. Motivated by this, here, we developed an electrochem. aptamer-based (EAB) sensor able to achieve the rapid, reagentless, and quant. measurement of the SARS-CoV-2 spike (S) protein. First, we demonstrated the ability of the selected aptamer to undergo a binding-induced conformational change in the presence of its target using fluorescence spectroscopy. Then, we engineered the aptamer to work as a bioreceptor in the EAB platform and we demonstrated its sensitivity and specificity. Finally, to demonstrate the clin. potential of the sensor, we tested it directly in biol. fluids (serum and artificial saliva), achieving the rapid (minutes) and single-step detection of the S protein in its clin. range. - 65Zakashansky, J. A.; Imamura, A. H.; Salgado, D. F., 2nd; Romero Mercieca, H. C.; Aguas, R. F. L.; Lao, A. M.; Pariser, J.; Arroyo-Currás, N.; Khine, M. Detection of the SARS-CoV-2 spike protein in saliva with Shrinky-Dink electrodes. Anal. Methods 2021, 13, 874– 883, DOI: 10.1039/d1ay00041a[Crossref], [PubMed], [CAS], Google Scholar65https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXjvVCru7c%253D&md5=42942831b25fbc59924f0f80d848b087Detection of the SARS-CoV-2 spike protein in saliva with Shrinky-Dink electrodesZakashansky, Julia A.; Imamura, Amanda H.; Salgado, Darwin F., II; Romero Mercieca, Heather C.; Aguas, Raphael F. L.; Lao, Angelou M.; Pariser, Joseph; Arroyo-Curras, Netzahualcoyotl; Khine, MichelleAnalytical Methods (2021), 13 (7), 874-883CODEN: AMNEGX; ISSN:1759-9679. (Royal Society of Chemistry)A review. Using the children's toy, Shrinky-Dink, we present an aptamer-based electrochem. (E-AB) assay that recognizes the spike protein of SARS-CoV-2 in saliva for viral infection detection. The low-cost electrodes are implementable at population scale and demonstrate detection down to 1 ag mL-1 of the S1 subunit of the spike protein.
- 66Cennamo, N.; Pasquardini, L.; Arcadio, F.; Lunelli, L.; Vanzetti, L.; Carafa, V.; Altucci, L.; Zeni, L. SARS-CoV-2 spike protein detection through a plasmonic D-shaped plastic optical fiber aptasensor. Talanta 2021, 233, 122532, DOI: 10.1016/j.talanta.2021.122532[Crossref], [PubMed], [CAS], Google Scholar66https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhtFKrs77M&md5=918249e712869852a7e75668d1e1f903SARS-CoV-2 spike protein detection through a plasmonic D-shaped plastic optical fiber aptasensorCennamo, Nunzio; Pasquardini, Laura; Arcadio, Francesco; Lunelli, Lorenzo; Vanzetti, Lia; Carafa, Vincenzo; Altucci, Lucia; Zeni, LuigiTalanta (2021), 233 (), 122532CODEN: TLNTA2; ISSN:0039-9140. (Elsevier B.V.)A specific aptameric sequence has been immobilized on short polyethyleneglycol (PEG) interface on gold nano-film deposited on a D-shaped plastic optical fiber (POFs) probe, and the protein binding has been monitored exploiting the very sensitive surface plasmon resonance (SPR) phenomenon. The receptor-binding domain (RBD) of the SARS-CoV-2 spike glycoprotein has been specifically used to develop an aptasensor. Surface anal. techniques coupled to fluorescence microscopy and plasmonic anal. have been utilized to characterize the biointerface. Spanning a wide protein range (25 ÷ 1000 nM), the SARS-Cov-2 spike protein was detected with a Limit of Detection (LoD) of about 37 nM. Different interferents (BSA, AH1N1 hemagglutinin protein and MERS spike protein) have been tested confirming the specificity of our aptasensor. Finally, a preliminary test in dild. human serum encouraged its application in a point-of-care device, since POF-based aptasensor represent a potentially low-cost compact biosensor, characterized by a rapid response, a small size and could be an ideal lab. portable diagnostic tool.
- 67Aithal, S.; Mishriki, S.; Gupta, R.; Sahu, R. P.; Botos, G.; Tanvir, S.; Hanson, R. W.; Puri, I. K. SARS-CoV-2 detection with aptamer-functionalized gold nanoparticles. Talanta 2022, 236, 122841, DOI: 10.1016/j.talanta.2021.122841[Crossref], [PubMed], [CAS], Google Scholar67https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvFyjsbbP&md5=5f4e1f429d578a71f079547f697cb496SARS-CoV-2 detection with aptamer-functionalized gold nanoparticlesAithal, Srivatsa; Mishriki, Sarah; Gupta, Rohit; Sahu, Rakesh P.; Botos, George; Tanvir, Shazia; Hanson, Russell W.; Puri, Ishwar K.Talanta (2022), 236 (), 122841CODEN: TLNTA2; ISSN:0039-9140. (Elsevier B.V.)A rapid detection test for SARS-CoV-2 is urgently required to monitor virus spread and containment. Here, we describe a test that uses nanoprobes, which are gold nanoparticles functionalized with an aptamer specific to the spike membrane protein of SARS-CoV-2. An ELISA confirms aptamer binding with the spike protein on gold surfaces. Protein recognition occurs by adding a coagulant, where nanoprobes with no bound protein agglomerate while those with sufficient bound protein do not. Using plasmon absorbance spectra, the nanoprobes detect 16 nM and higher concns. of spike protein in phosphate-buffered saline. The time-varying light absorbance is examd. at 540 nm to det. the crit. coagulant concn. required to agglomerates the nanoprobes, which depends on the protein concn. This approach detects 3540 genome copies/μl of inactivated SARS-CoV-2.
Cited By
This article is cited by 16 publications.
- Payel Sen, Zijie Zhang, Phoebe Li, Bal Ram Adhikari, Tianyi Guo, Jimmy Gu, Adam R MacIntosh, Colin van der Kuur, Yingfu Li, Leyla Soleymani. Integrating Water Purification with Electrochemical Aptamer Sensing for Detecting SARS-CoV-2 in Wastewater. ACS Sensors 2023, 8 (4) , 1558-1567. https://doi.org/10.1021/acssensors.2c02655
- Aysha Sarfraz Rizvi, Ghulam Murtaza, Xu Xu, Peifeng Gao, Lili Qiu, Zihui Meng. Aptamer-Linked Photonic Crystal Assay for High-Throughput Screening of HIV and SARS-CoV-2. Analytical Chemistry 2023, 95 (2) , 917-923. https://doi.org/10.1021/acs.analchem.2c03467
- Ghulam Murtaza, Aysha Sarfraz Rizvi, Min Xue, Lili Qiu, Zihui Meng. Consensus Receptor-Binding Domain-Targeted Aptamer Selection and Designing of a Photonic Crystal-Decorated Aptasensor for SARS-CoV-2. Analytical Chemistry 2022, 94 (20) , 7391-7399. https://doi.org/10.1021/acs.analchem.2c00937
- Federica Curti, Simone Fortunati, Wolfgang Knoll, Marco Giannetto, Roberto Corradini, Alessandro Bertucci, Maria Careri. A Folding-Based Electrochemical Aptasensor for the Single-Step Detection of the SARS-CoV-2 Spike Protein. ACS Applied Materials & Interfaces 2022, 14 (17) , 19204-19211. https://doi.org/10.1021/acsami.2c02405
- Fatemeh Yaghoobizadeh, Mohammad Roayaei Ardakani, Mohammad Mehdi Ranjbar, Hamid Galehdari, Mohammad Khosravi. Expression, purification, and study on the efficiency of a new potent recombinant scFv antibody against the SARS-CoV-2 spike RBD in E. coli BL21. Protein Expression and Purification 2023, 203 , 106210. https://doi.org/10.1016/j.pep.2022.106210
- Jin Wang, Masaki Ando, Hiroki Nagata, Sota Yoshida, Kenji Sakai, Toshihiko Kiwa. Multifunctional terahertz microscopy for biochemical and chemical imaging and sensing. Biosensors and Bioelectronics 2023, 220 , 114901. https://doi.org/10.1016/j.bios.2022.114901
- Jiuxing Li, Zijie Zhang, Rudi Liu, Ryan Amini, Bruno J. Salena, Yingfu Li. Discovery and translation of functional nucleic acids for clinically diagnosing infectious diseases: Opportunities and challenges. TrAC Trends in Analytical Chemistry 2023, 158 , 116886. https://doi.org/10.1016/j.trac.2022.116886
- Ji Woong Kim, Ah Hyun Cho, Ha Gyeong Shin, Sung Hoon Jang, Su Yeon Cho, Ye Rim Lee, Sukmook Lee. Development and Characterization of Phage Display-Derived Monoclonal Antibodies to the S2 Domain of Spike Proteins of Wild-Type SARS-CoV-2 and Multiple Variants. Viruses 2023, 15 (1) , 174. https://doi.org/10.3390/v15010174
- Fan Zhang, Hongxia Du, Linsen Li, Tengfei Li, Jing Wang, Zilei Chen, Mengmeng Yan, Chao Zhu, Feng Qu. Enzyme-Linked Aptamer Kits for Rapid, Visual, and Sensitive Determination of Lactoferrin in Dairy Products. Foods 2022, 11 (23) , 3763. https://doi.org/10.3390/foods11233763
- Pablo Alberto Franco-Urquijo, Mónica Sierra-Martínez, Mariana Jarquín-Martínez, Mateo Alejandro Martínez-Roque, Victor Miguel García-Velásquez, Gustavo Acosta-Altamirano, Nancy Jannet Ruiz-Pérez, Julia Dolores Toscano-Garibay, Luis Marat Alvarez-Salas. Fluorescence-Linked Aptamer Assay for SARS-CoV-2 Spike-Protein: A Step-by-Step Performance Analysis in Clinical Samples. Diagnostics 2022, 12 (11) , 2829. https://doi.org/10.3390/diagnostics12112829
- Michael Shola David, Damira Kanayeva. Enzyme linked oligonucleotide assay for the sensitive detection of SARS-CoV-2 variants. Frontiers in Cellular and Infection Microbiology 2022, 12 https://doi.org/10.3389/fcimb.2022.1017542
- Ryan Amini, Zijie Zhang, Jiuxing Li, Jimmy Gu, John D. Brennan, Yingfu Li. Aptamers for SARS‐CoV‐2: Isolation, Characterization, and Diagnostic and Therapeutic Developments. Analysis & Sensing 2022, 2 (5) https://doi.org/10.1002/anse.202200012
- Mariia V. Samodelova, Olesya O. Kapitanova, Nadezda F. Meshcheryakova, Sergey. M. Novikov, Nikita R. Yarenkov, Oleg A. Streletskii, Dmitry I. Yakubovsky, Fedor I. Grabovenko, Gleb A. Zhdanov, Aleksey V. Arsenin, Valentyn S. Volkov, Elena G. Zavyalova, Irina A. Veselova, Maria I. Zvereva. Model of the SARS-CoV-2 Virus for Development of a DNA-Modified, Surface-Enhanced Raman Spectroscopy Sensor with a Novel Hybrid Plasmonic Platform in Sandwich Mode. Biosensors 2022, 12 (9) , 768. https://doi.org/10.3390/bios12090768
- Behnaz Hatamluyi, Majid Rezayi, Saeid Amel Jamehdar, Kobra Salimian Rizi, Majid Mojarrad, Zahra Meshkat, Hamzeh Choobin, Saman Soleimanpour, Mohammad Taher Boroushaki. Sensitive and specific clinically diagnosis of SARS-CoV-2 employing a novel biosensor based on boron nitride quantum dots/flower-like gold nanostructures signal amplification. Biosensors and Bioelectronics 2022, 207 , 114209. https://doi.org/10.1016/j.bios.2022.114209
- Jin Wang, Kenji Sakai, Toshihiko Kiwa. Rational Design of Peptides Derived from Odorant-Binding Proteins for SARS-CoV-2-Related Volatile Organic Compounds Recognition. Molecules 2022, 27 (12) , 3917. https://doi.org/10.3390/molecules27123917
- Abilash Gangula, Brandon Kim, Benjamin Casey, Allison Hamill, Hariharan Regunath, Anandhi Upendran. Point-of-Care Testing of COVID-19: Current Status, Clinical Impact, and Future Therapeutic Perspectives. 2022, 1-70. https://doi.org/10.1007/978-981-19-4957-9_1
Abstract
Figure 1
Figure 1. Detection of the SARS-CoV-2 S antigen with the aptamer sandwich assay.
Figure 2
Figure 2. Specificity of the aptamers evaluated by direct ELAA. The relative binding of each aptamer was calculated using constant concentrations of aptamers (200 nM) and S1 proteins from the different HCoV’s (2 μg/mL). The error bars correspond to the standard deviation from duplicate measurements.
Figure 3
Figure 3. Screening of aptamer pairs for the detection of SARS-CoV-2 S1 protein. Each thiolated capture aptamer (AptxT) was analyzed in combination with each of the biotinylated reporter aptamers (AptxB) using a constant concentration of S1. No protein was added for the control samples. The number refers to the number of the aptamer tested, T indicates the thiolated aptamer and B indicates the biotinylated aptamer.
Figure 4
Figure 4. Sensitivity of the sandwich assays for SARS-CoV-2 S1 detection using different combinations of aptamer pairs.
Figure 5
Figure 5. Specificity of the sandwich assays using different aptamer pairs and S1 from different HCoV’s. Each protein was used at 2 μg/mL, and the error bars indicate the standard deviations from duplicate measurements.
Figure 6
Figure 6. Sensitivity of the sandwich assay based on the Apt1T/Apt5B aptamer pair in aptamer binding buffer and in VTM.
References
ARTICLE SECTIONSThis article references 67 other publications.
- 1Weiss, S. R. Forty years with coronaviruses. J. Exp. Med. 2020, 217, e20200537 DOI: 10.1084/jem.20200537[Crossref], [PubMed], [CAS], Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXovFSgtLY%253D&md5=eb058fb12d70e155c40213f30c36ab66Forty years with coronavirusesWeiss, Susan R.Journal of Experimental Medicine (2020), 217 (5), e20200537CODEN: JEMEAV; ISSN:1540-9538. (Rockefeller University Press)A review. I have been researching coronaviruses for more than forty years. This viewpoint summarizes some of the major findings in corona virus research made before the SARS epidemic and how they inform current research on the newly emerged SARS-CoV-2.
- 2He, F.; Deng, Y.; Li, W. Coronavirus disease 2019: What we know?. J. Med. Virol. 2020, 92, 719– 725, DOI: 10.1002/jmv.25766[Crossref], [PubMed], [CAS], Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXnsVWqtLc%253D&md5=4b071ade1e6bc6b8a70b2d389a93bc22Coronavirus disease 2019: What we know?He, Feng; Deng, Yu; Li, WeinaJournal of Medical Virology (2020), 92 (7), 719-725CODEN: JMVIDB; ISSN:0146-6615. (Wiley-Blackwell)A review. In late Dec. 2019, a cluster of unexplained pneumonia cases has been reported in Wuhan, China. A few days later, the causative agent of this mysterious pneumonia was identified as a novel coronavirus. This causative virus has been temporarily named as severe acute respiratory syndrome coronavirus 2 and the relevant infected disease has been named as coronavirus disease 2019 (COVID-19) by the World Health Organization, resp. The COVID-19 epidemic is spreading in China and all over the world now. The purpose of this review is primarily to review the pathogen, clin. features, diagnosis, and treatment of COVID-19, but also to comment briefly on the epidemiol. and pathol. based on the current evidence.
- 3Zhu, Z.; Lian, X.; Su, X.; Wu, W.; Marraro, G. A.; Zeng, Y. From SARS and MERS to COVID-19: a brief summary and comparison of severe acute respiratory infections caused by three highly pathogenic human coronaviruses. Respir. Res. 2020, 21, 224, DOI: 10.1186/s12931-020-01479-w[Crossref], [PubMed], [CAS], Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhslWqtrfE&md5=7c9a3bd77971d16156df4aadb70a2a72From SARS and MERS to COVID-19: a brief summary and comparison of severe acute respiratory infections caused by three highly pathogenic human coronavirusesZhu, Zhixing; Lian, Xihua; Su, Xiaoshan; Wu, Weijing; Marraro, Giuseppe A.; Zeng, YimingRespiratory Research (2020), 21 (1), 224CODEN: RREEBZ; ISSN:1465-993X. (BioMed Central Ltd.)A review. Within 2 decades, there have emerged 3 highly pathogenic and deadly human coronaviruses, namely SARS-CoV, MERS-CoV, and SARS-CoV-2. The economic burden and health threats caused by these coronaviruses are extremely dreadful and getting more serious as the increasing no. of global infections and attributed deaths of SARS-CoV-2 and MERS-CoV. Unfortunately, specific medical countermeasures for these hCoVs remain absent. Moreover, the fast spread of misinformation about the ongoing SARS-CoV-2 pandemic uniquely places the virus alongside an annoying infodemic and causes unnecessary worldwide panic. SARS-CoV-2 shares many similarities with SARS-CoV and MERS-CoV, certainly, obvious differences exist as well. Lessons learned from SARS-CoV and MERS-CoV, timely updated information of SARS-CoV-2 and MERS-CoV, and summarized specific knowledge of these hCoVs are extremely invaluable for effectively and efficiently contain the outbreak of SARS-CoV-2 and MERS-CoV. By gaining a deeper understanding of hCoVs and the illnesses caused by them, we can bridge knowledge gaps, provide cultural weapons for fighting and controlling the spread of MERS-CoV and SARS-CoV-2, and prep. effective and robust defense lines against hCoVs that may emerge or reemerge in the future. To this end, the state-of-the-art knowledge and comparing the biol. features of these lethal hCoVs and the clin. characteristics of illnesses caused by them are systematically summarized in the review.
- 4Gorbalenya, A. E.; Baker, S. C.; Baric, R. S.; de Groot, R. J.; Drosten, C.; Gulyaeva, A. A.; Haagmans, B. L.; Lauber, C.; Leontovich, A. M.; Neuman, B. W.; Penzar, D.; Perlman, S.; Poon, L. L. M.; Samborskiy, D. V.; Sidorov, I. A.; Sola, I.; Ziebuhr, J. The species Severe Acute Respiratory Syndrome-related coronavirus: classifying 2019-nCoV and naming it SARS-CoV-2. Nat. Microbiol. 2020, 5, 536– 544, DOI: 10.1038/s41564-020-0695-z
- 5Wrapp, 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, DOI: 10.1126/science.abb2507[Crossref], [PubMed], [CAS], Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXkvFemt70%253D&md5=27d08cbb9a43d1da051a8a92a9f68aa5Cryo-EM structure of the 2019-nCoV spike in the prefusion conformationWrapp, Daniel; Wang, Nianshuang; Corbett, Kizzmekia S.; Goldsmith, Jory A.; Hsieh, Ching-Lin; Abiona, Olubukola; Graham, Barney S.; McLellan, Jason S.Science (Washington, DC, United States) (2020), 367 (6483), 1260-1263CODEN: SCIEAS; ISSN:1095-9203. (American Association for the Advancement of Science)The outbreak of a novel coronavirus (2019-nCoV) represents a pandemic threat that has been declared a public health emergency of international concern. The CoV spike (S) glycoprotein is a key target for vaccines, therapeutic antibodies, and diagnostics. To facilitate medical countermeasure development, we detd. a 3.5-angstrom-resoln. cryo-electron microscopy structure of the 2019-nCoV S trimer in the prefusion conformation. The predominant state of the trimer has one of the three receptor-binding domains (RBDs) rotated up in a receptor-accessible conformation. We also provide biophys. and structural evidence that the 2019-nCoV S protein binds angiotensin-converting enzyme 2 (ACE2) with higher affinity than does severe acute respiratory syndrome (SARS)-CoV S. Addnl., we tested several published SARS-CoV RBD-specific monoclonal antibodies and found that they do not have appreciable binding to 2019-nCoV S, suggesting that antibody cross-reactivity may be limited between the two RBDs. The structure of 2019-nCoV S should enable the rapid development and evaluation of medical countermeasures to address the ongoing public health crisis.
- 6Wang, N.; Shang, J.; Jiang, S.; Du, L. Subunit vaccines against emerging pathogenic human coronaviruses. Front. Microbiol. 2020, 11, 298, DOI: 10.3389/fmicb.2020.00298[Crossref], [PubMed], [CAS], Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB38zislyluw%253D%253D&md5=0bc5d7ff9839b3cc6678b0d3bb57a56fSubunit Vaccines Against Emerging Pathogenic Human CoronavirusesWang Ning; Jiang Shibo; Du Lanying; Shang Jian; Jiang ShiboFrontiers in microbiology (2020), 11 (), 298 ISSN:1664-302X.Seven coronaviruses (CoVs) have been isolated from humans so far. Among them, three emerging pathogenic CoVs, including severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome coronavirus (MERS-CoV), and a newly identified CoV (2019-nCoV), once caused or continue to cause severe infections in humans, posing significant threats to global public health. SARS-CoV infection in humans (with about 10% case fatality rate) was first reported from China in 2002, while MERS-CoV infection in humans (with about 34.4% case fatality rate) was first reported from Saudi Arabia in June 2012. 2019-nCoV was first reported from China in December 2019, and is currently infecting more than 70000 people (with about 2.7% case fatality rate). Both SARS-CoV and MERS-CoV are zoonotic viruses, using bats as their natural reservoirs, and then transmitting through intermediate hosts, leading to human infections. Nevertheless, the intermediate host for 2019-nCoV is still under investigation and the vaccines against this new CoV have not been available. Although a variety of vaccines have been developed against infections of SARS-CoV and MERS-CoV, none of them has been approved for use in humans. In this review, we have described the structure and function of key proteins of emerging human CoVs, overviewed the current vaccine types to be developed against SARS-CoV and MERS-CoV, and summarized recent advances in subunit vaccines against these two pathogenic human CoVs. These subunit vaccines are introduced on the basis of full-length spike (S) protein, receptor-binding domain (RBD), non-RBD S protein fragments, and non-S structural proteins, and the potential factors affecting these subunit vaccines are also illustrated. Overall, this review will be helpful for rapid design and development of vaccines against the new 2019-nCoV and any future CoVs with pandemic potential. This review was written for the topic of Antivirals for Emerging Viruses: Vaccines and Therapeutics in the Virology section of Frontiers in Microbiology.
- 7Yao, H.; Song, Y.; Chen, Y.; Wu, N.; Xu, J.; Sun, C.; Zhang, J.; Weng, T.; Zhang, Z.; Wu, Z.; Cheng, L.; Shi, D.; Lu, X.; Lei, J.; Crispin, M.; Shi, Y.; Li, L.; Li, S. Molecular architecture of the SARS-CoV-2 virus. Cell 2020, 183, 730– 738, e13 DOI: 10.1016/j.cell.2020.09.018[Crossref], [PubMed], [CAS], Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvFKjs7nM&md5=7bd110334cb42931034a4eb5f9d2ff00Molecular Architecture of the SARS-CoV-2 VirusYao, Hangping; Song, Yutong; Chen, Yong; Wu, Nanping; Xu, Jialu; Sun, Chujie; Zhang, Jiaxing; Weng, Tianhao; Zhang, Zheyuan; Wu, Zhigang; Cheng, Linfang; Shi, Danrong; Lu, Xiangyun; Lei, Jianlin; Crispin, Max; Shi, Yigong; Li, Lanjuan; Li, SaiCell (Cambridge, MA, United States) (2020), 183 (3), 730-738.e13CODEN: CELLB5; ISSN:0092-8674. (Cell Press)SARS-CoV-2 is an enveloped virus responsible for the COVID-19 pandemic. Despite recent advances in the structural elucidation of SARS-CoV-2 proteins, the detailed architecture of the intact virus remains to be unveiled. Here we report the mol. assembly of the authentic SARS-CoV-2 virus using cryoelectron tomog. (cryo-ET) and subtomogram averaging (STA). Native structures of the S proteins in pre- and postfusion conformations were detd. to av. resolns. of 8.7-11 Å. Compns. of the N-linked glycans from the native spikes were analyzed by mass spectrometry, which revealed overall processing states of the native glycans highly similar to that of the recombinant glycoprotein glycans. The native conformation of the ribonucleoproteins (RNPs) and their higher-order assemblies were revealed. Overall, these characterizations revealed the architecture of the SARS-CoV-2 virus in exceptional detail and shed light on how the virus packs its ∼30-kb-long single-segmented RNA in the ∼80-nm-diam. lumen.
- 8Zhou, Y.; Jiang, S.; Du, L. Prospects for a MERS-CoV spike vaccine. Expert Rev. Vaccines 2018, 17, 677– 686, DOI: 10.1080/14760584.2018.1506702[Crossref], [PubMed], [CAS], Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhsV2isb3J&md5=f249563636d50985a7a70127ac0db27aProspects for a MERS-CoV spike vaccineZhou, Yusen; Jiang, Shibo; Du, LanyingExpert Review of Vaccines (2018), 17 (8), 677-686CODEN: ERVXAX; ISSN:1476-0584. (Taylor & Francis Ltd.): Six years have passed since Middle East respiratory syndrome (MERS) coronavirus (MERS-CoV), a newly emerging infectious virus, was first reported in 2012. Although MERS-CoV has had a consistently high mortality rate in humans, no vaccines have been approved to prevent MERS-CoV infection in humans. MERS-CoV spike (S) protein is a key target for development of MERS vaccines.: In this review, we illustrate the structure and function of S protein as a vaccine target, describe available animal models for evaluating MERS vaccines, and summarize recent progress on MERS-CoV S-based vaccines, focusing on their ability to elicit antibody and/or cellular immune responses, neutralizing antibodies, and protection against MERS-CoV infection in different models. Prospects for future MERS-CoV S-based vaccines are discussed.: The majority of MERS vaccines under development are based on MERS-CoV S protein, including full-length S, S1, and receptor-binding domain (RBD). While it is essential to evaluate the safety of full-length S and S1-based MERS vaccines, further improvement of the efficacy of RBD-based vaccines using novel strategies would be necessary. Overall, this review provides informative guidance for designing and developing safe and effective MERS vaccines based on viral S protein.
- 9Lan, J.; Ge, J.; Yu, J.; Shan, S.; Zhou, H.; Fan, S.; Zhang, Q.; Shi, X.; Wang, Q.; Zhang, L.; Wang, X. Structure of the SARS-CoV-2 spike receptor-binding domain bound to the ACE2 receptor. Nature 2020, 581, 215– 220, DOI: 10.1038/s41586-020-2180-5[Crossref], [PubMed], [CAS], Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXoslOqtL8%253D&md5=279c60143e8e5eb505457e0778baa8efStructure of the SARS-CoV-2 spike receptor-binding domain bound to the ACE2 receptorLan, Jun; Ge, Jiwan; Yu, Jinfang; Shan, Sisi; Zhou, Huan; Fan, Shilong; Zhang, Qi; Shi, Xuanling; Wang, Qisheng; Zhang, Linqi; Wang, XinquanNature (London, United Kingdom) (2020), 581 (7807), 215-220CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Abstr.: A new and highly pathogenic coronavirus (severe acute respiratory syndrome coronavirus-2, SARS-CoV-2) caused an outbreak in Wuhan city, Hubei province, China, starting from Dec. 2019 that quickly spread nationwide and to other countries around the world1-3. Here, to better understand the initial step of infection at an at. level, we detd. the crystal structure of the receptor-binding domain (RBD) of the spike protein of SARS-CoV-2 bound to the cell receptor ACE2. The overall ACE2-binding mode of the SARS-CoV-2 RBD is nearly identical to that of the SARS-CoV RBD, which also uses ACE2 as the cell receptor4. Structural anal. identified residues in the SARS-CoV-2 RBD that are essential for ACE2 binding, the majority of which either are highly conserved or share similar side chain properties with those in the SARS-CoV RBD. Such similarity in structure and sequence strongly indicate convergent evolution between the SARS-CoV-2 and SARS-CoV RBDs for improved binding to ACE2, although SARS-CoV-2 does not cluster within SARS and SARS-related coronaviruses1-3,5. The epitopes of two SARS-CoV antibodies that target the RBD are also analyzed for binding to the SARS-CoV-2 RBD, providing insights into the future identification of cross-reactive antibodies.
- 10Li, F.; Li, W.; Farzan, M.; Harrison, S. C. Structure of SARS coronavirus spike receptor-binding domain complexed with receptor. Science 2005, 309, 1864– 1868, DOI: 10.1126/science.1116480[Crossref], [PubMed], [CAS], Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXpvFCisLw%253D&md5=0a7ac2fb7a76c97979d859c0489c682fStructure of SARS Coronavirus Spike Receptor-Binding Domain Complexed with ReceptorLi, Fang; Li, Wenhui; Farzan, Michael; Harrison, Stephen C.Science (Washington, DC, United States) (2005), 309 (5742), 1864-1868CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)The spike protein (S) of SARS coronavirus (SARS-CoV) attaches the virus to its cellular receptor, angiotensin-converting enzyme 2 (ACE2). A defined receptor-binding domain (RBD) on S mediates this interaction. The crystal structure at 2.9 angstrom resoln. of the RBD bound with the peptidase domain of human ACE2 shows that the RBD presents a gently concave surface, which cradles the N-terminal lobe of the peptidase. The at. details at the interface between the two proteins clarify the importance of residue changes that facilitate efficient cross-species infection and human-to-human transmission. The structure of the RBD suggests ways to make truncated disulfide-stabilized RBD variants for use in the design of coronavirus vaccines.
- 11Liu, S.; Xiao, G.; Chen, Y.; He, Y.; Niu, J.; Escalante, C. R.; Xiong, H.; Farmar, J.; Debnath, A. K.; Tien, P.; Jiang, S. Interaction between heptad repeat 1 and 2 regions in spike protein of SARS-associated coronavirus: implications for virus fusogenic mechanism and identification of fusion inhibitors. Lancet 2004, 363, 938– 947, DOI: 10.1016/s0140-6736(04)15788-7[Crossref], [PubMed], [CAS], Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXisVSktbo%253D&md5=578507918c57e2419c24213bea2c978fInteraction between heptad repeat 1 and 2 regions in spike protein of SARS-associated coronavirus: implications for virus fusogenic mechanism and identification of fusion inhibitorsLiu, Shuwen; Xiao, Gengfu; Chen, Yibang; He, Yuxian; Niu, Jinkui; Escalante, Carlos R.; Xiong, Huabao; Farmar, James; Debnath, Asim K.; Tien, Po; Jiang, ShiboLancet (2004), 363 (9413), 938-947CODEN: LANCAO; ISSN:0140-6736. (Elsevier Science Ltd.)Studies on the fusion-inhibitory peptides derived from the heptad repeat 1 and 2 (HR1 and HR2) regions of the HIV-1 envelope glycoprotein gp41 provided crucial information on the viral fusogenic mechanism. We used a similar approach to study the fusogenic mechanism of severe-acute-respiratory-syndrome-assocd. coronavirus (SARS-CoV). We tested the inhibitory activity against infection of two sets of peptides corresponding to sequences of SARS-CoV spike protein HR1 and HR2 regions and investigated the interactions between the HR1 and HR2 peptides by surface plasmon resonance, sedimentation equilibration anal., CD, native polyacrylamide-gel electrophoresis, size exclusion high-performance liq. chromatog., and computer-aided homol. modeling and mol. docking anal. One peptide, CP-1, derived from the HR2 region, inhibited SARS-CoV infection in the micromolar range. CP-1 bound with high affinity to a peptide from the HR1 region, NP-1. CP-1 alone had low α-helicity and self-assocd. to form a trimer in phosphate buffer (pH 7.2). CP-1 and NP-1 mixed in equimolar concns. formed a six-helix bundle, similar to the fusogenic core structure of HIV-1 gp41. After binding to the target cell, the transmembrane spike protein might change conformation by assocn. between the HR1 and HR2 regions to form an oligomeric structure, leading to fusion between the viral and target-cell membranes. At the pre-fusion intermediate state, CP-1 could bind to the HR1 region and interfere with the conformational changes, resulting in inhibition of SARS-CoV fusion with the target cells. CP-1 might be modifiable to increase its anti-SARS-CoV activity and could be further developed as an antiviral agent for treatment or prophylaxis of SARS-CoV infection.
- 12Tai, W.; He, L.; Zhang, X.; Pu, J.; Voronin, D.; Jiang, S.; Zhou, Y.; Du, L. Characterization of the receptor-binding domain (RBD) of 2019 novel coronavirus: implication for development of RBD protein as a viral attachment inhibitor and vaccine. Cell. Mol. Immunol. 2020, 17, 613– 620, DOI: 10.1038/s41423-020-0400-4[Crossref], [PubMed], [CAS], Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXlt1Chsrw%253D&md5=87bc49d070c84e78b01230518aaa465aCharacterization of the receptor-binding domain (RBD) of 2019 novel coronavirus: implication for development of RBD protein as a viral attachment inhibitor and vaccineTai, Wanbo; He, Lei; Zhang, Xiujuan; Pu, Jing; Voronin, Denis; Jiang, Shibo; Zhou, Yusen; Du, LanyingCellular & Molecular Immunology (2020), 17 (6), 613-620CODEN: CMIEAO; ISSN:1672-7681. (Nature Research)The outbreak of Coronavirus Disease 2019 (COVID-19) has posed a serious threat to global public health, calling for the development of safe and effective prophylactics and therapeutics against infection of its causative agent, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), also known as 2019 novel coronavirus (2019-nCoV). The CoV spike (S) protein plays the most important roles in viral attachment, fusion and entry, and serves as a target for development of antibodies, entry inhibitors and vaccines. Here, we identified the receptor-binding domain (RBD) in SARS-CoV-2 S protein and found that the RBD protein bound strongly to human and bat angiotensin-converting enzyme 2 (ACE2) receptors. SARS-CoV-2 RBD exhibited significantly higher binding affinity to ACE2 receptor than SARS-CoV RBD and could block the binding and, hence, attachment of SARS-CoV-2 RBD and SARS-CoV RBD to ACE2-expressing cells, thus inhibiting their infection to host cells. SARS-CoV RBD-specific antibodies could cross-react with SARS-CoV-2 RBD protein, and SARS-CoV RBD-induced antisera could cross-neutralize SARS-CoV-2, suggesting the potential to develop SARS-CoV RBD-based vaccines for prevention of SARS-CoV-2 and SARS-CoV infection.
- 13Dai, L.; Gao, G. F. Viral targets for vaccines against COVID-19. Nat. Rev. Immunol. 2021, 21, 73– 82, DOI: 10.1038/s41577-020-00480-0[Crossref], [PubMed], [CAS], Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXktFeksg%253D%253D&md5=4e84b56201379ab888192ff41d3b30ffViral targets for vaccines against COVID-19Dai, Lianpan; Gao, George F.Nature Reviews Immunology (2021), 21 (2), 73-82CODEN: NRIABX; ISSN:1474-1733. (Nature Research)A review of the progress of which viral elements are used in COVID-19 vaccine candidates, why they might act as good targets for the immune system and the implications for protective immunity. Vaccines are urgently needed to control the coronavirus disease 2019 (COVID-19) pandemic and to help the return to pre-pandemic normalcy. A great many vaccine candidates are being developed, several of which have completed late-stage clin. trials and are reporting pos. results.
- 14Khan, S.; Tombuloglu, H.; Hassanein, S. E.; Rehman, S.; Bozkurt, A.; Cevik, E.; Abdel-Ghany, S.; Nabi, G.; Ali, A.; Sabit, H. Coronavirus diseases 2019: current biological situation and potential therapeutic perspective. Eur. J. Pharmacol. 2020, 886, 1733447, DOI: 10.1016/j.ejphar.2020.173447
- 15Tombuloglu, H.; Sabit, H.; Al-Suhaimi, E.; Al Jindan, R.; Alkharsah, K. R. Development of multiplex real-time RT-PCR assay for the detection of SARS-CoV-2. PLoS One 2021, 16, e0250942 DOI: 10.1371/journal.pone.0250942[Crossref], [PubMed], [CAS], Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhtVegur3I&md5=734b90db068d5762645c84d93e456cd2Development of multiplex real-time RT-PCR assay for the detection of SARS-CoV-2Tombuloglu, Huseyin; Sabit, Hussein; Al-Suhaimi, Ebtesam; Al Jindan, Reem; Alkharsah, Khaled R.PLoS One (2021), 16 (4), e0250942CODEN: POLNCL; ISSN:1932-6203. (Public Library of Science)The outbreak of the new human coronavirus SARS-CoV-2 (also known as 2019-nCoV) continues to increase globally. The real-time reverse transcription polymerase chain reaction (rRT-PCR) is the most used technique in virus detection. However, possible false-neg. and false-pos. results produce misleading consequences, making it necessary to improve existing methods. Here, we developed a multiplex rRT-PCR diagnostic method, which targets two viral genes (RdRP and E) and one human gene (RP) simultaneously. The reaction was tested by using pseudoviral RNA and human target mRNA sequences as a template. Also, the protocol was validated by using 14 clin. SARS-CoV-2 pos. samples. The results are in good agreement with the CDC authorized Cepheid's Xpert Xpress SARS-CoV-2 diagnostic system (100%). Unlike single gene targeting strategies, the current method provides the amplification of two viral regions in the same PCR reaction. Therefore, an accurate SARS-CoV-2 diagnostic assay was provided, which allows testing of 91 samples in 96-well plates in per run. Thanks to this strategy, fast, reliable, and easy-to-use rRT-PCR method is obtained to diagnose SARS-CoV-2.
- 16Ravi, N.; Cortade, D. L.; Ng, E.; Wang, S. X. Diagnostics for SARS-CoV-2 detection: a comprehensive review of the FDA-EUA COVID-19 testing landscape. Biosens. Bioelectron. 2020, 165, 112454, DOI: 10.1016/j.bios.2020.112454[Crossref], [PubMed], [CAS], Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsFShsbvN&md5=1e88bde679e0507c706e21fa9c2d54d7Diagnostics for SARS-CoV-2 detection: A comprehensive review of the FDA-EUA COVID-19 testing landscapeRavi, Neeraja; Cortade, Dana L.; Ng, Elaine; Wang, Shan X.Biosensors & Bioelectronics (2020), 165 (), 112454CODEN: BBIOE4; ISSN:0956-5663. (Elsevier B.V.)A review. The rapidly spreading outbreak of COVID-19 disease is caused by the SARS-CoV-2 virus, first reported in Dec. 2019 in Wuhan, China. As of June 17, 2020, this virus has infected over 8.2 million people but ranges in symptom severity, making it difficult to assess its overall infection rate. There is a need for rapid and accurate diagnostics to better monitor and prevent the spread of COVID-19. In this review, we present and evaluate two main types of diagnostics with FDA-EUA status for COVID-19: nucleic acid testing for detection of SARS-CoV-2 RNA, and serol. assays for detection of SARS-CoV-2 specific IgG and IgM patient antibodies, along with the necessary sample prepn. for accurate diagnoses. In particular, we cover and compare tests such as the CDC 2019-nCoV RT-PCR Diagnostic Panel, Cellex's qSARS-CoV-2 IgG/IgM Rapid Test, and point-of-care tests such as Abbott's ID NOW COVID-19 Test. Antibody testing is esp. important in understanding the prevalence of the virus in the community and to identify those who have gained immunity. We conclude by highlighting the future of COVID-19 diagnostics, which include the need for quant. testing and the development of emerging biosensors as point-of-care tests.
- 17Abbott Diagnostics Scarborough Inc. BinaxNOWTM COVID-19 Ag CARD [package insert, EUA]; Scarborough: MA, USA, 2020.Google ScholarThere is no corresponding record for this reference.
- 18LumiraDx Group Limited. LumiraDxTM SARS-CoV-2 Ag Test [package insert, EUA]; Dumyat Business Park Alloa FK10 2PB: U.K., 2020.Google ScholarThere is no corresponding record for this reference.
- 19Becton, Dickinson and Company. BD VeritorTM System for Rapid Detection of SARS-CoV-2 [package insert, EUA]; Sparks-Glencoe: MD, USA, 2020.Google ScholarThere is no corresponding record for this reference.
- 20Quidel Corporation. Sofia Severe Acute Respiratory Syndrome Antigen FIA [package insert, EUA]: San Diego, CA, USA, 2020.Google ScholarThere is no corresponding record for this reference.
- 21Barlev-Gross, M.; Weiss, S.; Ben-Shmuel, A.; Sittner, A.; Eden, K.; Mazuz, N.; Glinert, I.; Bar-David, E.; Puni, R.; Amit, S.; Kriger, O.; Schuster, O.; Alcalay, R.; Makdasi, E.; Epstein, E.; Noy-Porat, T.; Rosenfeld, R.; Achdout, H.; Mazor, O.; Israely, T.; Levy, H.; Mechaly, A. Spike vs nucleocapsid SARS-CoV-2 antigen detection: application in nasopharyngeal swab specimens. Anal. Bioanal. Chem. 2021, 413, 3501– 3510, DOI: 10.1007/s00216-021-03298-4[Crossref], [PubMed], [CAS], Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXnsVGnsr8%253D&md5=af42bca2a60bfe68687276dfde045b72Spike vs. nucleocapsid SARS-CoV-2 antigen detection: application in nasopharyngeal swab specimensBarlev-Gross, Moria; Weiss, Shay; Ben-Shmuel, Amir; Sittner, Assa; Eden, Keren; Mazuz, Noam; Glinert, Itai; Bar-David, Elad; Puni, Reut; Amit, Sharon; Kriger, Or; Schuster, Ofir; Alcalay, Ron; Makdasi, Efi; Epstein, Eyal; Noy-Porat, Tal; Rosenfeld, Ronit; Achdout, Hagit; Mazor, Ohad; Israely, Tomer; Levy, Haim; Mechaly, AdvaAnalytical and Bioanalytical Chemistry (2021), 413 (13), 3501-3510CODEN: ABCNBP; ISSN:1618-2642. (Springer)Public health experts emphasize the need for quick, point-of-care SARS-CoV-2 detection as an effective strategy for controlling virus spread. To this end, many antigen detection devices were developed and commercialized. These devices are mostly based on detecting SARS-CoV-2's nucleocapsid protein. Recently, alerts issued by both the FDA and the CDC raised concerns regarding the devices' tendency to exhibit false pos. results. We developed a novel alternative spike-based antigen assay, comprising 4 high-affinity, specific monoclonal antibodies, directed against different epitopes on the spike's S1 subunit. The assay's performance was evaluated for COVID-19 detection from nasopharyngeal swabs, compared to an inhouse nucleocapsid-based assay, composed of novel antibodies directed against the nucleocapsid. Detection of COVID-19 was carried out in a cohort of 284 qRT-PCR pos. and neg. nasopharyngeal swab samples. The time resolved fluorescence (TRF) ELISA spike assay displayed very high specificity (99%) accompanied with a somewhat lower sensitivity (66% for Ct < 25), compared to the nucleocapsid ELISA assay which was more sensitive (85% for Ct < 25) while less specific (87% specificity). Despite being outperformed by qRT-PCR, we suggest that there is room for such tests in the clin. setting, as cheap and rapid pre-screening tools. Our results further suggest that when applying antigen detection, one must consider its intended application (sensitivity vs. specificity), taking into consideration that the nucleocapsid might not be the optimal target. In this regard, we propose that a combination of both antigens might contribute to the validity of the results.
- 22Aoki, K.; Nagasawa, T.; Ishii, Y.; Yagi, S.; Okuma, S.; Kashiwagi, K.; Maeda, T.; Miyazaki, T.; Yoshizawa, S.; Tateda, K. Clinical validation of quantitative SARS-CoV-2 antigen assays to estimate SARS-CoV-2 viral loads in nasopharyngeal swabs. J. Infect. Chemother. 2021, 27, 613– 616, DOI: 10.1016/j.jiac.2020.11.021[Crossref], [PubMed], [CAS], Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhslSru7c%253D&md5=98e3f6fbd1cf1041ba11fcc2c6dcc54aClinical validation of quantitative SARS-CoV-2 antigen assays to estimate SARS-CoV-2 viral loads in nasopharyngeal swabsAoki, Kotaro; Nagasawa, Tatsuya; Ishii, Yoshikazu; Yagi, Shintaro; Okuma, Sadatsugu; Kashiwagi, Katsuhito; Maeda, Tadashi; Miyazaki, Taito; Yoshizawa, Sadako; Tateda, KazuhiroJournal of Infection and Chemotherapy (2021), 27 (4), 613-616CODEN: JICHFN; ISSN:1341-321X. (Elsevier B.V.)Expansion of the testing capacity for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an important issue to mitigate the pandemic of coronavirus disease-2019 (COVID-19) caused by this virus. Recently, a sensitive quant. antigen test (SQT), Lumipulse SARS-CoV-2 Ag, was developed. It is a fully automated chemiluminescent enzyme immunoassay system for SARS-CoV-2. In this study, the anal. performance of SQT was examd. using clin. specimens from nasopharyngeal swabs using reverse transcription polymerase chain reaction (RT-PCR) as a control. Receiver operating characteristic anal. of 24 SARS-CoV-2-pos. and 524 -neg. patients showed an area under the curve of 0.957 ± 0.063. Using a cut-off value of 1.34 pg/mL, the sensitivity was 91.7%, the specificity was 98.5%, and the overall rate of agreement was 98.2%. In the distribution of neg. cases, the 99.5 percentile value was 1.03 pg/mL. There was a high correlation between the viral load calcd. using the cycle threshold value of RT-PCR and the concn. of antigen. The tendency for the antigen concn. to decrease with time after disease onset correlated with that of the viral load. Presented results indicate that SQT is highly concordant with RT-PCR and should be useful for the diagnosis of COVID-19 in any clin. setting. Therefore, this fully automated kit will contribute to the expansion of the testing capability for SARS-CoV-2.
- 23Hirotsu, Y.; Maejima, M.; Shibusawa, M.; Nagakubo, Y.; Hosaka, K.; Amemiya, K.; Sueki, H.; Hayakawa, M.; Mochizuki, H.; Tsutsui, T.; Kakizaki, Y.; Miyashita, Y.; Yagi, S.; Kojima, S.; Omata, M. Comparison of automated SARS-CoV-2 antigen test for COVID-19 infection with quantitative RT-PCR using 313 nasopharyngeal swabs, including from seven serially followed patients. Int. J. Infect. Dis. 2020, 99, 397– 402, DOI: 10.1016/j.ijid.2020.08.029[Crossref], [PubMed], [CAS], Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvVWmtb%252FP&md5=2b97870f9db15fdb5136fe7ddda6294eComparison of automated SARS-CoV-2 antigen test for COVID-19 infection with quantitative RT-PCR using 313 nasopharyngeal swabs, including from seven serially followed patientsHirotsu, Yosuke; Maejima, Makoto; Shibusawa, Masahiro; Nagakubo, Yuki; Hosaka, Kazuhiro; Amemiya, Kenji; Sueki, Hitomi; Hayakawa, Miyoko; Mochizuki, Hitoshi; Tsutsui, Toshiharu; Kakizaki, Yumiko; Miyashita, Yoshihiro; Yagi, Shintaro; Kojima, Satoshi; Omata, MasaoInternational Journal of Infectious Diseases (2020), 99 (), 397-402CODEN: IJIDF3; ISSN:1201-9712. (Elsevier Ltd.)In routine clin. practice, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is detd. by reverse-transcription PCR (RT-PCR). In the current pandemic, a more rapid and high-throughput method is in growing demand. Here, the authors validated the performance of a new antigen test (LUMIPULSE) based on chemiluminescence enzyme immunoassay. A total of 313 nasopharyngeal swabs (82 serial samples from 7 infected patients and 231 individual samples from 4 infected patients and 215 uninfected individuals) were analyzed for SARS-CoV-2 with quant. RT-PCR (RT-qPCR) and then subjected to LUMIPULSE. The authors detd. the cutoff value for antigen detection using receiver operating characteristic curve anal. and compared the performance of the antigen test with that of RT-qPCR. The authors also compared the viral loads and antigen levels in serial samples from seven infected patients. Using RT-qPCR as the ref., the antigen test exhibited 55.2% sensitivity and 99.6% specificity, with a 91.4% overall agreement rate (286/313). In specimens with > 100 viral copies and between 10 and 100 copies, the antigen test showed 100% and 85% concordance with RT-qPCR, resp. This concordance declined with lower viral loads. In the serially followed patients, the antigen levels showed a steady decline, along with viral clearance. This gradual decline was in contrast with the abrupt pos.-to-neg. and neg.-to-pos. status changes obsd. with RT-qPCR, particularly in the late phase of infection. In summary, the LUMIPULSE antigen test can rapidly identify SARS-CoV-2-infected individuals with moderate to high viral loads and may be helpful for monitoring viral clearance in hospitalized patients.
- 24Pollock, N. R.; Savage, T. J.; Wardell, H.; Lee, R.; Mathew, A.; Stengelin, M.; Sigal, G. B. Correlation of SARS-CoV-2 nucleocapsid antigen and RNA concentrations in nasopharyngeal samples from children and adults using an ultrasensitive and quantitative antigen assay. J. Clin. Microbiol. 2021, 59, e03077– 20, DOI: 10.1128/JCM.03077-20[Crossref], [PubMed], [CAS], Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXpvFyks70%253D&md5=bb1100be3ef32c227627e5f53c7be92cCorrelation of SARS-CoV-2 nucleocapsid antigen and RNA concentrations in nasopharyngeal samples from children and adults using an ultrasensitive and quantitative antigen assayPollock, Nira R.; Savage, Timothy J.; Wardell, Hanna; Lee, Rose A.; Mathew, Anu; Stengelin, Martin; Sigal, George B.Journal of Clinical Microbiology (2021), 59 (4), e03077CODEN: JCMIDW; ISSN:1098-660X. (American Society for Microbiology)Diagnosis of COVID-19 by PCR offers high sensitivity, but the utility of detecting samples with high cycle threshold (CT) values remains controversial. Currently available rapid diagnostic tests (RDTs) for SARS-CoV-2 nucleocapsid antigens (Ag) have sensitivity well below PCR. The correlation of Ag and RNA quantities in clin. nasopharyngeal (NP) samples is unknown. An ultrasensitive, quant. electrochemiluminescence immunoassay for SARS-CoV-2 nucleocapsid (the MSD S-PLEX SARS-CoV-2 N assay) was used to measure Ag in clin. NP samples from adults and children previously tested by PCR. The S-PLEX Ag assay had a limit of detection (LOD) of 0.16 pg/mL and a cutoff of 0.32 pg/mL. Ag concns. measured in clin. NP samples (collected in 3.0 mL of media) ranged from less than 160 fg/mL to 2.7μg/mL. Log-transformed Ag concns. correlated tightly with CT values. In 35 adult and 101 pediatric PCR-pos. samples, the sensitivities were 91% (95% confidence interval, 77 to 98%) and 79% (70 to 87%), resp. In samples with a CT of ≤35, the sensitivities were 100% (88 to 100%) and 96% (88 to 99%), resp. In 50 adult and 40 pediatric PCR-neg. specimens, the specificities were 100% (93 to 100%) and 98% (87 to 100%), resp. Nucleocapsid concns. in clin. NP samples span 8 orders of magnitude and correlate closely with RNA concns. (CT values). The S-PLEX Ag assay showed 96 to 100% sensitivity in samples from children and adults with CT values of ≤35, and a specificity of 98 to 100%. These results clarify Ag concn. distributions in clin. samples, providing insight into the performance of Ag RDTs and offering a new approach to diagnosis of COVID-19.
- 25Pekosz, A.; Cooper, C. K.; Parvu, V.; Li, M.; Andrews, J. C.; Manabe, Y. C.; Kodsi, S.; Leitch, J.; Gary, D. S.; Roger-Dalbert, C. Antigen-based testing but not real-time polymerase chain reaction correlates with Severe Acute Respiratory Syndrome Coronavirus 2 viral culture. Clin. Infect. Dis. 2021, 73, e2861– e2866, DOI: 10.1093/cid/ciaa1706[Crossref], [PubMed], [CAS], Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXisVKrtLjI&md5=72f2f571f0341b63249a3b702b275717Antigen-based testing but not real-time polymerase chain reaction correlates with severe acute respiratory syndrome Coronavirus 2 viral culturePekosz, Andrew; Parvu, Valentin; Li, Maggie; Andrews, Jeffrey C.; Manabe, Yukari C.; Kodsi, Salma; Gary, Devin S.; Roger-Dalbert, Celine; Leitch, Jeffry; Cooper, Charles K.Clinical Infectious Diseases (2021), 73 (9), e2861-e2866CODEN: CIDIEL; ISSN:1537-6591. (Oxford University Press)Individuals can test pos. for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) by mol. assays following the resoln. of their clin. disease. Recent studies indicate that SARS-CoV-2 antigen-based tests are likely to be pos. early in the disease course, when there is an increased likelihood of high levels of infectious virus. Upper respiratory specimens from 251 participants with coronavirus disease 2019 symptoms (≤7 days from symptom onset) were prospectively collected and tested with a lateral flow antigen test and a real-time polymerase chain reaction (rt-PCR) assay for detection of SARS-CoV-2. Specimens from a subset of the study specimens were utilized to det. the presence of infectious virus in the VeroE6TMPRSS2 cell culture model. The antigen test demonstrated a higher pos. predictive value (90%) than rt-PCR (70%) when compared to culture-pos. results. The pos. percentage agreement for detection of infectious virus for the antigen test was similar to rt-PCR when compared to culture results. The correlation between SARS-CoV-2 antigen and SARS-CoV-2 culture positivity represents a significant advancement in detg. the risk for potential transmissibility beyond that which can be achieved by detection of SARS-CoV-2 genomic RNA. SARS-CoV-2 antigen testing can facilitate low-cost, scalable, and rapid time-to-result, while providing good risk detn. of those who are likely harboring infectious virus, compared to rt-PCR.
- 26Scohy, A.; Anantharajah, A.; Bodéus, M.; Kabamba-Mukadi, B.; Verroken, A.; Rodriguez-Villalobos, H. Low performance of rapid antigen detection test as frontline testing for COVID-19 diagnosis. J. Clin. Virol. 2020, 129, 104455, DOI: 10.1016/j.jcv.2020.104455[Crossref], [PubMed], [CAS], Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtVGmsbbI&md5=183778b80c7596564747ec676c198e09Low performance of rapid antigen detection test as frontline testing for COVID-19 diagnosisScohy, Anais; Anantharajah, Ahalieyah; Bodeus, Monique; Kabamba-Mukadi, Benoit; Verroken, Alexia; Rodriguez-Villalobos, HectorJournal of Clinical Virology (2020), 129 (), 104455CODEN: JCVIFB; ISSN:1386-6532. (Elsevier B.V.)Ensuring accurate diagnosis is essential to limit the spread of SARS-CoV-2 and for the clin. management of COVID-19. Although real-time reverse transcription polymerase chain reaction (RT- qPCR) is the current recommended lab. method to diagnose SARS-CoV-2 acute infection, several factors such as requirement of special equipment and skilled staff limit the use of these time-consuming mol. techniques. Recently, several easy to perform rapid antigen detection tests were developed and recommended in some countries as the first line of diagnostic. The aim of this study was to evaluate the performances of the Coris COVID-19 Ag Respi-Strip test, a rapid immunochromatog. test for the detection of SARS-CoV-2 antigen, in comparison to RT-qPCR. 148 Nasopharyngeal swabs were tested. Amongst the 106 pos. RT-qPCR samples, 32 were detected by the rapid antigen test, given an overall sensitivity of 30.2%. All the samples detected pos. with the antigen rapid test were also pos. with RT-qPCR. Higher viral loads are assocd. with better antigen detection rates. Unfortunately, the overall poor sensitivity of the COVID-19 Ag Respi-Strip does not allow using it alone as the frontline testing for COVID-19 diagnosis.
- 27Tang, Y. W.; Schmitz, J. E.; Persing, D. H.; Stratton, C. W. Laboratory diagnosis of COVID-19: current issues and challenges. J. Clin. Microbiol. 2020, 58, 10– 1128, DOI: 10.1128/JCM.00512-20
- 28Che, X.-y.; Qiu, L.-w.; Pan, Y.-x.; Wen, K.; Hao, W.; Zhang, L.-y.; Wang, Y.-d.; Liao, Z.-y.; Hua, X.; Cheng, V. C. C.; Yuen, K.-y. Sensitive and specific monoclonal antibody-based capture enzyme immunoassay for detection of nucleocapsid antigen in sera from patients with Severe Acute Respiratory Syndrome. J. Clin. Microbiol. 2004, 42, 2629– 2635, DOI: 10.1128/jcm.42.6.2629-2635.2004[Crossref], [PubMed], [CAS], Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXlslalsr4%253D&md5=53f47dfefc1c75e16abb77b5a6f46315Sensitive and specific monoclonal antibody-based capture enzyme immunoassay for detection of nucleocapsid antigen in sera from patients with severe acute respiratory syndromeChe, Xiao-yan; Qiu, Li-wen; Pan, Yu-xian; Wen, Kun; Hao, Wei; Zhang, Li-ya; Wang, Ya-di; Liao, Zhi-yong; Hua, Xu; Cheng, Vincent C. C.; Yuen, Kwok-yungJournal of Clinical Microbiology (2004), 42 (6), 2629-2635CODEN: JCMIDW; ISSN:0095-1137. (American Society for Microbiology)A rapid antigen test for the diagnosis of severe acute respiratory syndrome (SARS) is essential for control of this disease at the point of management. The nucleocapsid (N) protein of SARS-assocd. coronavirus (SARS-CoV) is abundantly expressed in infected-cell culture filtrate as demonstrable by Western blotting using convalescent-phase sera from patients with SARS. We used monoclonal antibodies specifically directed against N protein to establish a sensitive antigen capture sandwich ELISA for the detection of SARS-CoV. The assay employed a mixt. of three monoclonal antibodies for capture and rabbit polyclonal antibodies for detection of serum antigen in 32 cases of clin. probable SARS as defined by the World Health Organization during the epidemic in Guangzhou, China. Recombinant N protein was used as a std. to establish a detection sensitivity of approximated 50 pg/mL. The linear range of detection in clin. specimens was from 100 pg/mL to 3.2 ng/mL. Using a panel of sera collected at different points in time, the amt. of circulating N antigen was found to peak 6 to 10 days after the onset of symptoms. The sensitivity of the assay was 84.6% in 13 serol. confirmed SARS patients with blood taken during the first 10 days after the onset of symptoms (11 of 13). The specificity of the assay was 98.5% in 1,272 healthy individuals (1,253 of 1,272). There was no cross-reaction with other human and animal coronaviruses in this assay. In conclusion, a sensitive and quant. antigen capture ELISA was established for the early diagnosis and disease monitoring of SARS-CoV infection.
- 29Diao, B.; Wen, K.; Chen, J.; Liu, Y.; Yuan, Z.; Han, C.; Chen, J.; Pan, Y.; Chen, L.; Dan, Y.; Wang, J.; Chen, Y.; Deng, G.; Zhou, H.; Wu, Y. Diagnosis of acute respiratory syndrome coronavirus 2 infection by detection of nucleocapsid protein. medRxiv 2020, 20032524, DOI: 10.1101/2020.03.07.20032524
- 30Grant, B. D.; Anderson, C. E.; Williford, J. R.; Alonzo, L. F.; Glukhova, V. A.; Boyle, D. S.; Weigl, B. H.; Nichols, K. P. SARS-CoV-2 coronavirus nucleocapsid antigen-detecting half-strip lateral flow assay toward the development of point of care tests using commercially available reagents. Anal. Chem. 2020, 92, 11305– 11309, DOI: 10.1021/acs.analchem.0c01975[ACS Full Text
], [CAS], Google Scholar
30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtlSis7jO&md5=e4cb14b8cb5e90bf93381695f5a4f634SARS-CoV-2 Coronavirus Nucleocapsid Antigen-Detecting Half-Strip Lateral Flow Assay Toward the Development of Point of Care Tests Using Commercially Available ReagentsGrant, Benjamin D.; Anderson, Caitlin E.; Williford, John R.; Alonzo, Luis F.; Glukhova, Veronika A.; Boyle, David S.; Weigl, Bernhard H.; Nichols, Kevin P.Analytical Chemistry (Washington, DC, United States) (2020), 92 (16), 11305-11309CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)The SARS-CoV-2 pandemic has created an unprecedented need for rapid diagnostic testing to enable the efficient treatment and mitigation of COVID-19. The primary diagnostic tool currently employed is reverse transcription polymerase chain reaction (RT-PCR), which can have good sensitivity and excellent specificity. Unfortunately, implementation costs and logistical problems with reagents during the global SARS-CoV-2 pandemic have hindered its universal on demand adoption. Lateral flow assays (LFAs) represent a class of diagnostic that, if sufficiently clin. sensitive, may fill many of the gaps in the current RT-PCR testing regime, esp. in low- and middle-income countries (LMICs). To date, many serol. LFAs have been developed, though none meet the performance requirements necessary for diagnostic use cases, primarily due to the relatively long delay between infection and seroconversion. However, on the basis of previously reported results from SARS-CoV-1, antigen-based SARS-CoV-2 assays may have significantly better clin. sensitivity than serol. assays. To date, only a very small no. of antigen-detecting LFAs have been developed. Development of a half-strip LFA is a useful first step in the development of any LFA format. In this work, we present a half-strip LFA using com. available antibodies for the detection of SARS-CoV-2. We have tested this LFA in buffer and measured an LOD of 0.65 ng/mL (95% CI of 0.53 to 0.77 ng/mL) ng/mL with recombinant antigen using an optical reader with sensitivity equiv. to a visual read. Further development, including evaluating the appropriate sample matrix, will be required for this assay approach to be made useful in a point of care setting, though this half-strip LFA may serve as a useful starting point for others developing similar tests. - 31Ahmadivand, A.; Gerislioglu, B.; Ramezani, Z.; Kaushik, A.; Manickam, P.; Ghoreishi, S. A. Functionalized terahertz plasmonic metasensors: femtomolar-level detection of SARS-CoV-2 spike proteins. Biosens. Bioelectron. 2021, 177, 112971, DOI: 10.1016/j.bios.2021.112971[Crossref], [PubMed], [CAS], Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhtVGlsbc%253D&md5=6c090c57524e355c17a9d9f0e77c37a2Functionalized terahertz plasmonic metasensors: Femtomolar-level detection of SARS-CoV-2 spike proteinsAhmadivand, Arash; Gerislioglu, Burak; Ramezani, Zeinab; Kaushik, Ajeet; Manickam, Pandiaraj; Ghoreishi, S. AmirBiosensors & Bioelectronics (2021), 177 (), 112971CODEN: BBIOE4; ISSN:0956-5663. (Elsevier B.V.)Effective and efficient management of human betacoronavirus severe acute respiratory syndrome (SARS)-CoV-2 virus infection i.e., COVID-19 pandemic, required sensitive and selective sensors with short sample-to-result durations for performing desired diagnostics. In this direction, one appropriate alternative approach to detect SARS-CoV-2 virus protein at low level i.e., femtomolar (fM) is exploring plasmonic metasensor technol. for COVID-19 diagnostics, which offers exquisite opportunities in advanced healthcare programs, and modern clin. diagnostics. The intrinsic merits of plasmonic metasensors stem from their capability to squeeze electromagnetic fields, simultaneously in frequency, time, and space. However, the detection of low-mol. wt. biomols. at low densities is a typical drawback of conventional metasensors that has recently been addressed using toroidal metasurface technol. This research is focused on the fabrication of a miniaturized plasmonic immunosensor based on toroidal electrodynamics concept that can sustain robustly confined plasmonic modes with ultranarrow lineshapes in the terahertz (THz) frequencies. By exciting toroidal dipole mode using our quasi-infinite metasurface and a judiciously optimized protocol based on functionalized gold nanoparticles (AuNPs) conjugated with the specific monoclonal antibody specific to spike protein (S1) of SARS-CoV-2 virus onto the metasurface, the resonance shifts for diverse concns. of the spike protein are monitored. Possessing mol. wt. around ∼76 kDa allowed to detect the presence of SARS-CoV-2 virus protein with significantly low as limit of detection (LoD) was achieved as ∼4.2 fM. We envisage that outcomes of this research will pave the way toward the use of toroidal metasensors as practical technologies for rapid and precise screening of SARS-CoV-2 virus carriers, symptomatic or asymptomatic, and spike proteins in hospitals, clinics, labs., and site of infection.
- 32Seo, G.; Lee, G.; Kim, M. J.; Baek, S.-H.; Choi, M.; Ku, K. B.; Lee, C.-S.; Jun, S.; Park, D.; Kim, H. G.; Kim, S.-J.; Lee, J.-O.; Kim, B. T.; Park, E. C.; Kim, S. I. 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, DOI: 10.1021/acsnano.0c02823[ACS Full Text
], [CAS], Google Scholar
32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXnt1SrtrY%253D&md5=735104d0be58eaefaae38aa6f9877459Rapid Detection of COVID-19 Causative Virus (SARS-CoV-2) in Human Nasopharyngeal Swab Specimens Using Field-Effect Transistor-Based BiosensorSeo, Giwan; Lee, Geonhee; Kim, Mi Jeong; Baek, Seung-Hwa; Choi, Minsuk; Ku, Keun Bon; Lee, Chang-Seop; Jun, Sangmi; Park, Daeui; Kim, Hong Gi; Kim, Seong-Jun; Lee, Jeong-O.; Kim, Bum Tae; Park, Edmond Changkyun; Kim, Seung IlACS Nano (2020), 14 (4), 5135-5142CODEN: ANCAC3; ISSN:1936-0851. (American Chemical Society)Coronavirus disease 2019 (COVID-19) is a newly emerging human infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2, previously called 2019-nCoV). Based on the rapid increase in the rate of human infection, the World Health Organization (WHO) has classified the COVID-19 outbreak as a pandemic. Because no specific drugs or vaccines for COVID-19 are yet available, early diagnosis and management are crucial for contg. the outbreak. Here, we report a field-effect transistor (FET)-based biosensing device for detecting SARS-CoV-2 in clin. samples. The sensor was produced by coating graphene sheets of the FET with a specific antibody against SARS-CoV-2 spike protein. The performance of the sensor was detd. using antigen protein, cultured virus, and nasopharyngeal swab specimens from COVID-19 patients. Our FET device could detect the SARS-CoV-2 spike protein at concns. of 1 fg/mL in phosphate-buffered saline and 100 fg/mL clin. transport medium. In addn., the FET sensor successfully detected SARS-CoV-2 in culture medium (limit of detection [LOD]: 1.6 x 101 pfu/mL) and clin. samples (LOD: 2.42 x 102 copies/mL). Thus, we have successfully fabricated a promising FET biosensor for SARS-CoV-2; our device is a highly sensitive immunol. diagnostic method for COVID-19 that requires no sample pretreatment or labeling. - 33Zhong, J.; Rösch, E. L.; Viereck, T.; Schilling, M.; Ludwig, F. Toward rapid and sensitive detection of SARS-CoV-2 with functionalized magnetic nanoparticles. ACS Sens. 2021, 6, 976– 984, DOI: 10.1021/acssensors.0c02160[ACS Full Text
], [CAS], Google Scholar
33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXitV2ku78%253D&md5=e79da90b2367b4017b15e4e00dae2277Toward Rapid and Sensitive Detection of SARS-CoV-2 with Functionalized Magnetic NanoparticlesZhong, Jing; Roesch, Enja Laureen; Viereck, Thilo; Schilling, Meinhard; Ludwig, FrankACS Sensors (2021), 6 (3), 976-984CODEN: ASCEFJ; ISSN:2379-3694. (American Chemical Society)The outbreak of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) threatens global medical systems and economies and rules our daily living life. Controlling the outbreak of SARS-CoV-2 has become 1 of the most important and urgent strategies throughout the whole world. As of Oct. 2020, there have not yet been any medicines or therapies to be effective against SARS-CoV-2. Thus, rapid and sensitive diagnostics is the most important measures to control the outbreak of SARS-CoV-2. Homogeneous biosensing based on magnetic nanoparticles (MNPs) is 1 of the most promising approaches for rapid and highly sensitive detection of biomols. This paper proposes an approach for rapid and sensitive detection of SARS-CoV-2 with functionalized MNPs via the measurement of their magnetic response in an ac magnetic field. For proof of concept, mimic SARS-CoV-2 consisting of spike proteins and polystyrene beads are used for expts. Exptl. results demonstrate that the proposed approach allows the rapid detection of mimic SARS-CoV-2 with a limit of detection of 0.084 nM (5.9 fmol). The proposed approach has great potential for designing a low-cost and point-of-care device for rapid and sensitive diagnostics of SARS-CoV-2. - 34Ventura, 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, DOI: 10.1021/acssensors.0c01742[ACS Full Text
], [CAS], Google Scholar
34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvFaqsL7K&md5=bef9b17a1a0f63018163bcf46372c57aColorimetric Test for Fast Detection of SARS-CoV-2 in Nasal and Throat SwabsVentura, Bartolomeo Della; Cennamo, Michele; Minopoli, Antonio; Campanile, Raffaele; Censi, Sergio Bolletti; Terracciano, Daniela; Portella, Giuseppe; Velotta, RaffaeleACS Sensors (2020), 5 (10), 3043-3048CODEN: ASCEFJ; ISSN:2379-3694. (American Chemical Society)Mass testing is fundamental to face the pandemic caused by the coronavirus SARS-CoV-2 discovered at the end of 2019. To this aim, it is necessary to establish reliable, fast, and cheap tools to detect viral particles in biol. material so to identify the people capable of spreading the infection. We demonstrate that a colorimetric biosensor based on gold nanoparticle (AuNP) interaction induced by SARS-CoV-2 lends itself as an outstanding tool for detecting viral particles in nasal and throat swabs. The extinction spectrum of a colloidal soln. of multiple viral-target gold nanoparticles-AuNPs functionalized with antibodies targeting 3 surface proteins of SARS-CoV-2 (spike, envelope, and membrane)-is red-shifted in few minutes when mixed with a soln. contg. the viral particle. The optical d. of the mixed soln. measured at 560 nm was compared to the threshold cycle (Ct) of a real-time PCR (gold std. for detecting the presence of viruses) finding that the colorimetric method is able to detect very low viral load with a detection limit approaching that of the real-time PCR. Since the method is sensitive to the infecting viral particle rather than to its RNA, the achievements reported here open a new perspective not only in the context of the current and possible future pandemics, but also in microbiol., as the biosensor proves itself to be a powerful though simple tool for measuring the viral particle concn. - 35Pramanik, A.; Gao, Y.; Patibandla, S.; Mitra, D.; McCandless, M. G.; Fassero, L. A.; Gates, K.; Tandon, R.; Chandra Ray, P. The rapid diagnosis and effective inhibition of coronavirus using spike antibody attached gold nanoparticles. Nanoscale Adv. 2021, 3, 1588– 1596, DOI: 10.1039/d0na01007c[Crossref], [PubMed], [CAS], Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhsVOgu7s%253D&md5=610835de6dba6d0c6c2a97d2daf2339fThe rapid diagnosis and effective inhibition of coronavirus using spike antibody attached gold nanoparticlesPramanik, Avijit; Gao, Ye; Patibandla, Shamily; Mitra, Dipanwita; McCandless, Martin G.; Fassero, Lauren A.; Gates, Kalein; Tandon, Ritesh; Chandra Ray, PareshNanoscale Advances (2021), 3 (6), 1588-1596CODEN: NAADAI; ISSN:2516-0230. (Royal Society of Chemistry)Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the cause of the coronavirus disease that began in 2019 (COVID-19), has been responsible for 1.4 million deaths worldwide as of 13 Nov. 2020. Because at the time of writing no vaccine is yet available, a rapid diagnostic assay is very urgently needed. Herein, we present the development of anti-spike antibody attached gold nanoparticles for the rapid diagnosis of specific COVID-19 viral antigen or virus via a simple colorimetric change observation within a 5 min time period. For rapid and highly sensitive identification, surface enhanced Raman spectroscopy (SERS) was employed using 4-aminothiophenol as a reporter mol., which is attached to the gold nanoparticle via an Au-S bond. In the presence of COVID-19 antigen or virus particles, owing to the antigen-antibody interaction, the gold nanoparticles undergo aggregation, changing color from pink to blue, which allows for the detn. of the presence of antigen or virus very rapidly by the naked eye, even at concns. of 1 ng (ng) per mL for COVID-19 antigen and 1000 virus particles per mL for SARS-CoV-2 spike protein pseudotyped baculovirus. Importantly, the aggregated gold nanoparticles form "hot spots" to provide very strong SERS signal enhancement from anti-spike antibody and 4-aminothiophenol attached gold nanoparticles via light-matter interactions. Finite-difference time-domain (FDTD) simulation data indicate a 4-orders-of-magnitude Raman enhancement in "hot spot" positions when gold nanoparticles form aggregates. Using a portable Raman analyzer, our reported data demonstrate that our antibody and 4-aminothiophenol attached gold nanoparticle-based SERS probe has the capability to detect COVID-19 antigen even at a concn. of 4 pg (pg) per mL and virus at a concn. of 18 virus particles per mL within a 5 min time period. Using HEK293T cells, which express angiotensin-converting enzyme 2 (ACE2), by which SARS-CoV-2 enters human cells, we show that anti-spike antibody attached gold nanoparticles have the capability to inhibit infection by the virus. Our reported data show that antibody attached gold nanoparticles bind to SARS-CoV-2 spike protein, thereby inhibiting the virus from binding to cell receptors, which stops virus infection and spread. It also has the capability to destroy the lipid membrane of the virus.
- 36Yousefi, H.; Mahmud, A.; Chang, D.; Das, J.; Gomis, S.; Chen, J. B.; Wang, H.; Been, T.; Yip, L.; Coomes, E.; Li, Z.; Mubareka, S.; McGeer, A.; Christie, N.; Gray-Owen, S.; Cochrane, A.; Rini, J. M.; Sargent, E. H.; Kelley, S. O. Detection of SARS-CoV-2 viral particles using direct, reagent-free electrochemical sensing. J. Am. Chem. Soc. 2021, 143, 1722– 1727, DOI: 10.1021/jacs.0c10810[ACS Full Text
], [CAS], Google Scholar
36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvVWls74%253D&md5=3315d5fa3a22cd8e29f6a78f02d2665aDetection of SARS-CoV-2 Viral Particles Using Direct, Reagent-Free Electrochemical SensingYousefi, Hanie; Mahmud, Alam; Chang, Dingran; Das, Jagotamoy; Gomis, Surath; Chen, Jenise B.; Wang, Hansen; Been, Terek; Yip, Lily; Coomes, Eric; Li, Zhijie; Mubareka, Samira; McGeer, Allison; Christie, Natasha; Gray-Owen, Scott; Cochrane, Alan; Rini, James M.; Sargent, Edward H.; Kelley, Shana O.Journal of the American Chemical Society (2021), 143 (4), 1722-1727CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)The development of new methods for direct viral detection using streamlined and ideally reagent-free assays is a timely and important, but challenging, problem. The challenge of combating the COVID-19 pandemic has been exacerbated by the lack of rapid and effective methods to identify viral pathogens like SARS-CoV-2 on-demand. Existing gold std. nucleic acid-based approaches require enzymic amplification to achieve clin. relevant levels of sensitivity and are not typically used outside of a lab. setting. We report reagent-free viral sensing that directly reads out the presence of viral particles in 5 min using only a sensor-modified electrode chip. The approach relies on a class of electrode-tethered sensors bearing an analyte-binding antibody displayed on a neg. charged DNA linker that also features a tethered redox probe. When a pos. potential is applied, the sensor is transported to the electrode surface. Using chronoamperometry, the presence of viral particles and proteins can be detected as these species increase the hydrodynamic drag on the sensor. This report is the 1st virus-detecting assay that uses the kinetic response of a probe/virus complex to analyze the complexation state of the antibody. We demonstrate the performance of this sensing approach as a means to detect, within 5 min, the presence of the SARS-CoV-2 virus and its assocd. spike protein in test samples and in unprocessed patient saliva. - 37Lee, J.-H.; Choi, M.; Jung, Y.; Lee, S. K.; Lee, C.-S.; Kim, J.; Kim, J.; Kim, N. H.; Kim, B.-T.; Kim, H. G. A novel rapid detection for SARS-CoV-2 spike 1 antigens using human angiotensin converting enzyme 2 (ACE2). Biosens. Bioelectron. 2021, 171, 112715, DOI: 10.1016/j.bios.2020.112715[Crossref], [PubMed], [CAS], Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitFOisbzJ&md5=3f9071ac5e872a5880248ea47a10f3ebA novel rapid detection for SARS-CoV-2 spike 1 antigens using human angiotensin converting enzyme 2 (ACE2)Lee, Jong-Hwan; Choi, Minsuk; Jung, Yujin; Lee, Sung Kyun; Lee, Chang-Seop; Kim, Jung; Kim, Jongwoo; Kim, Nam Hoon; Kim, Bum-Tae; Kim, Hong GiBiosensors & Bioelectronics (2021), 171 (), 112715CODEN: BBIOE4; ISSN:0956-5663. (Elsevier B.V.)Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19), a newly emerging human infectious disease. Because no specific antiviral drugs or vaccines are available to treat COVID-19, early diagnostics, isolation, and prevention are crucial for contg. the outbreak. Mol. diagnostics using reverse transcription polymerase chain reaction (RT-PCR) are the current gold std. for detection. However, viral RNAs are much less stable during transport and storage than proteins such as antigens and antibodies. Consequently, false-neg. RT-PCR results can occur due to inadequate collection of clin. specimens or poor handling of a specimen during testing. Although antigen immunoassays are stable diagnostics for detection of past infection, infection progress, and transmission dynamics, no matched antibody pair for immunoassay of SARS-CoV-2 antigens has yet been reported. In this study, we designed and developed a novel rapid detection method for SARS-CoV-2 spike 1 (S1) protein using the SARS-CoV-2 receptor ACE2, which can form matched pairs with com. available antibodies. ACE2 and S1-mAb were paired with each other for capture and detection in a lateral flow immunoassay (LFIA) that did not cross-react with SARS-CoV Spike 1 or MERS-CoV Spike 1 protein. The SARS-CoV-2 S1 (<5 ng of recombinant proteins/reaction) was detected by the ACE2-based LFIA. The limit of detection of our ACE2-LFIA was 1.86 x 105 copies/mL in the clin. specimen of COVID-19 Patients without no cross-reactivity for nasal swabs from healthy subjects. This is the first study to detect SARS-CoV-2 S1 antigen using an LFIA with matched pair consisting of ACE2 and antibody. Our findings will be helpful to detect the S1 antigen of SARS-CoV-2 from COVID-19 patients.
- 38Pinals, R. L.; Ledesma, F.; Yang, D.; Navarro, N.; Jeong, S.; Pak, J. E.; Kuo, L.; Chuang, Y.-C.; Cheng, Y.-W.; Sun, H.-Y.; Landry, M. P. Rapid SARS-CoV-2 spike protein detection by carbon nanotube-based near-infrared nanosensors. Nano Lett. 2021, 21, 2272– 2280, DOI: 10.1021/acs.nanolett.1c00118[ACS Full Text
], [CAS], Google Scholar
38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXltFWjs78%253D&md5=d62b5397687debc14aac2e3e87583660Rapid SARS-CoV-2 spike protein detection by carbon nanotube-based near-infrared nanosensorsPinals, Rebecca L.; Ledesma, Francis; Yang, Darwin; Navarro, Nicole; Jeong, Sanghwa; Pak, John E.; Kuo, Lili; Chuang, Yung-Chun; Cheng, Yu-Wei; Sun, Hung-Yu; Landry, Markita P.Nano Letters (2021), 21 (5), 2272-2280CODEN: NALEFD; ISSN:1530-6984. (American Chemical Society)To effectively track and eliminate COVID-19, it is crit. to develop tools for rapid and accessible diagnosis of actively infected individuals. Here, we introduce a single-walled carbon nanotube (SWCNT)-based optical sensing approach toward this end. We construct a nanosensor based on SWCNTs noncovalently functionalized with ACE2, a host protein with high binding affinity for the SARS-CoV-2 spike protein. The presence of the SARS-CoV-2 spike protein elicits a robust, 2-fold nanosensor fluorescence increase within 90 min of spike protein exposure. We characterize the nanosensor stability and sensing mechanism and passivate the nanosensor to preserve sensing response in saliva and viral transport medium. We further demonstrate that these ACE2-SWCNT nanosensors retain sensing capacity in a surface-immobilized format, exhibiting a 73% fluorescence turn-on response within 5 s of exposure to 35 mg/L SARS-CoV-2 virus-like particles. Our data demonstrate that ACE2-SWCNT nanosensors can be developed into an optical tool for rapid SARS-CoV-2 detection. - 39Baker, A. N.; Richards, S.-J.; Guy, C. S.; Congdon, T. R.; Hasan, M.; Zwetsloot, A. J.; Gallo, A.; Lewandowski, J. R.; Stansfeld, P. J.; Straube, A.; Walker, M.; Chessa, S.; Pergolizzi, G.; Dedola, S.; Field, R. A.; Gibson, M. I. The SARS-COV-2 spike protein binds sialic acids and enables rapid detection in a lateral flow point of care diagnostic device. ACS Cent. Sci. 2020, 6, 2046– 2052, DOI: 10.1021/acscentsci.0c00855[ACS Full Text
], [CAS], Google Scholar
39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvVOlsbnP&md5=30a56fcd6d32e48834aa542773a21420The SARS-COV-2 Spike Protein Binds Sialic Acids and Enables Rapid Detection in a Lateral Flow Point of Care Diagnostic DeviceBaker, Alexander N.; Richards, Sarah-Jane; Guy, Collette S.; Congdon, Thomas R.; Hasan, Muhammad; Zwetsloot, Alexander J.; Gallo, Angelo; Lewandowski, Jozef R.; Stansfeld, Phillip J.; Straube, Anne; Walker, Marc; Chessa, Simona; Pergolizzi, Giulia; Dedola, Simone; Field, Robert A.; Gibson, Matthew I.ACS Central Science (2020), 6 (11), 2046-2052CODEN: ACSCII; ISSN:2374-7951. (American Chemical Society)There is an urgent need to understand the behavior of the novel coronavirus (SARS-COV-2), which is the causative agent of COVID-19, and to develop point-of-care diagnostics. Here, a glyconanoparticle platform is used to discover that N-acetyl neuraminic acid has affinity toward the SARS-COV-2 spike glycoprotein, demonstrating its glycan-binding function. Optimization of the particle size and coating enabled detection of the spike glycoprotein in lateral flow and showed selectivity over the SARS-COV-1 spike protein. Using a virus-like particle and a pseudotyped lentivirus model, paper-based lateral flow detection was demonstrated in under 30 min, showing the potential of this system as a low-cost detection platform. The spike-protein from SARS-COV-2 is shown to bind sialic acids, which is exploited to assemble a lateral flow diagnostic tool, using glycans rather than antibodies, as the recognition unit. - 40Tuerk, C.; Gold, L. Systematic evolution of ligands by exponential enrichment: RNA ligands to bacteriophage T4 DNA polymerase. Science 1990, 249, 505– 510, DOI: 10.1126/science.2200121[Crossref], [PubMed], [CAS], Google Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3cXlt1OltLY%253D&md5=a40a28db9df4a5da4c1b1652a34aa715Systematic evolution of ligands by exponential enrichment: RNA ligands to bacteriophage T4 DNA polymeraseTuerk, Craig; Gold, LarryScience (Washington, DC, United States) (1990), 249 (4968), 505-10CODEN: SCIEAS; ISSN:0036-8075.A novel method was developed for rapidly selecting preferred binding sequences from a population of random sequences. The method is designated systematic evolution of ligands by exponential enrichment (SELEX). High-affinity nucleic acid ligands for a protein were isolated by a procedure that depends on alternate cycles of ligand selection from pools of variant sequences and amplification of the bound species. Multiple rounds exponentially enrich the population for the highest affinity species that can be clonally isolated and characterized. In particular one eight-base region of an RNA that interacts with the T4 DNA polymerase was chosen and randomized. Two different sequences were selected by this procedure from the calcd. pool of 65,536 species. One is the wild-type sequence found in the bacteriophage mRNA; one is varied from wild type at four positions. The binding consts. of these two RNA's to T4 DNA polymerase are equiv. These protocols with minimal modification can yield high-affinity ligands for any protein that binds nucleic acids as part of its function; high affinity ligands could conceivably be developed for any target mol.
- 41Ellington, A. D.; Szostak, J. W. In vitro selection of RNA molecules that bind specific ligands. Nature 1990, 346, 818– 822, DOI: 10.1038/346818a0[Crossref], [PubMed], [CAS], Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3MXitVGgsw%253D%253D&md5=522ad1b4f0284ee88173b02035f9e9c5In vitro selection of RNA molecules that bind specific ligandsEllington, Andrew D.; Szostak, Jack W.Nature (London, United Kingdom) (1990), 346 (6287), 818-22CODEN: NATUAS; ISSN:0028-0836.Subpopulations of RNA mols. that bind specifically to a variety of org. dyes have been isolated from a population of random sequence RNA mols. Roughly one in 1010 random sequence RNA mols. folds in such a way as to create a specific binding site for small ligands.
- 42Robertson, D. L.; Joyce, G. F. Selection in vitro of an RNA enzyme that specifically cleaves single-stranded DNA. Nature 1990, 344, 467– 468, DOI: 10.1038/344467a0[Crossref], [PubMed], [CAS], Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3cXitlWltLk%253D&md5=9f3ef5e76f028aa84277861930d98189Selection in vitro of an RNA enzyme that specifically cleaves single-stranded DNARobertson, Debra L.; Joyce, Gerald F.Nature (London, United Kingdom) (1990), 344 (6265), 467-8CODEN: NATUAS; ISSN:0028-0836.The discovery of RNA enzymes has, for the 1st time, provided a single mol. that has both genetic and catalytic properties. Techniques were previously devised for the mutation, selection, and amplification of catalytic RNA, all of which can be performed rapidly in vitro. This report describes how these techniques can be integrated and performed repeatedly within a single reaction vessel. This allows evolution expts. to be carried out in response to artificially imposed selection constraints. The subject of the investigation was the Tetrahymena ribozyme, a self-splicing group I intron derived from the large rRNA precursor of Tetrahymena thermophila that catalyzes sequence-specific phosphoester transfer reactions involving RNA substrates. It consists of 413 nucleotides, and assumes a well-defined secondary and tertiary structure responsible for its catalytic activity. Variant forms of the enzyme were selected that could best react with a DNA substrate. This led to the recovery of a mutant form of the enzyme that cleaves DNA more efficiently than the wild-type enzyme. The selected mol. represents the discovery of the 1st RNA enzyme known to cleave single-stranded DNA specifically.
- 43Nguyen, V.-T.; Seo, H. B.; Kim, B. C.; Kim, S. K.; Song, C.-S.; Gu, M. B. Highly sensitive sandwich-type SPR based detection of whole H5Nx viruses using a pair of aptamers. Biosens. Bioelectron. 2016, 86, 293– 300, DOI: 10.1016/j.bios.2016.06.064[Crossref], [PubMed], [CAS], Google Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhtFSntrbN&md5=802a134f713241370fc1dabb2e5cd2d3Highly sensitive sandwich-type SPR based detection of whole H5Nx viruses using a pair of aptamersNguyen, Van-Thuan; Seo, Ho Bin; Kim, Byoung Chan; Kim, Sang Kyung; Song, Chang-Seon; Gu, Man BockBiosensors & Bioelectronics (2016), 86 (), 293-300CODEN: BBIOE4; ISSN:0956-5663. (Elsevier B.V.)In this research, we report highly sensitive and specific sandwich-type SPR-based biosensor for the detection H5Nx whole viruses. A few of aptamers, for the first time, were successfully screened and characterized for whole avian influenza (AI) viruses, H5Nx, by using Multi-GO-SELEX method. The affinities of the aptamers developed in this study were ranged from 8×104 to 1×104 EID50/mL, and the aptamers IF22, IF23 were found to be specific to H5N1 and H5N8, resp. In addn., some flexible aptamers IF20, IF15, and IF10 were found to bind to the H5N1 and H5N2, H5N1 and H5N8, or H5N1, H5N2, and H5N8, resp. Moreover, aptamers IF10 and IF22 were found to bind H5N1 virus simultaneously and confirmed to bind the different site of the same H5N1 whole virus. Therefore, this pair of aptamers, IF10 and IF22, were successfully applied to develop the sandwich-type SPR-based biosensor assay which is rapid, accurate for the detection of AI whole virus from H5N1-infected feces samples. The min. detectible concn. of H5N1 whole virus was found to be 200 EID50/mL with this sandwich-type detection using the aptamer pair obtained in this study. In addn., the sensitivity of this biosensor was successfully enhanced by using the signal amplification with the secondary aptamer conjugated with gold nanoparticles.
- 44Park, J.-W.; Jin Lee, S.; Choi, E.-J.; Kim, J.; Song, J.-Y.; Bock Gu, M. An ultra-sensitive detection of a whole virus using dual aptamers developed by immobilization-free screening. Biosens. Bioelectron. 2014, 51, 324– 329, DOI: 10.1016/j.bios.2013.07.052[Crossref], [PubMed], [CAS], Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsFGqt77F&md5=58b49b9f4f48fa7fcc3ba564bc1a3cebAn ultra-sensitive detection of a whole virus using dual aptamers developed by immobilization-free screeningPark, Jee-Woong; Jin Lee, Su; Choi, Eun-Jin; Kim, Jaejo; Song, Jae-Young; Bock Gu, ManBiosensors & Bioelectronics (2014), 51 (), 324-329CODEN: BBIOE4; ISSN:0956-5663. (Elsevier B.V.)In this study, we successfully developed a ssDNA aptamer pairs by using an advanced immobilization-free SELEX method with affinity-based selection and counter-screening process at every round. By implementing this method, two different aptamers specifically binding to bovine viral diarrhea virus type 1 (BVDV type 1) with high affinity were successfully screened. This aptamer pair was applied to ultrasensitive detection platform for BVDV type 1 in a sandwich manner. The ultrasensitive detection of BVDV type 1 using one of aptamers conjugated with gold nanoparticles was obtained in aptamer-aptamer sandwich type sensing format, with the limit of detection of 800 copies/mL, which is comparable to a real-time PCR method.
- 45Seo, H. B.; Gu, M. B. Aptamer-based sandwich-type biosensors. J. Biol. Eng. 2017, 11, 11, DOI: 10.1186/s13036-017-0054-7[Crossref], [PubMed], [CAS], Google Scholar45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXkslyrt7o%253D&md5=b10920617d0342cd0717648a30003830Aptamer-based sandwich-type biosensorsSeo, Ho Bin; Gu, Man BockJournal of Biological Engineering (2017), 11 (), 11/1-11/7CODEN: JBEOBZ; ISSN:1754-1611. (BioMed Central Ltd.)Sandwich-type biosensor platforms have drawn lots of attentions due to its superior features, compared to other platforms, in terms of its stable and reproducible responses and easy enhancement in the detection sensitivity. The sandwich-type assays can be developed by utilizing a pair of receptors, which bind to the different sites of the same target. In this mini-review paper, the sandwich-type biosensors using either pairs of aptamers or aptamer-antibody pairs are reviewed in terms of its targets and platforms, the schematic designs, and their anal. performance.
- 46Song, Y.; Song, J.; Wei, X.; Huang, M.; Sun, M.; Zhu, L.; Lin, B.; Shen, H.; Zhu, Z.; Yang, C. Discovery of aptamers targeting the receptor-binding domain of the SARS-CoV-2 spike glycoprotein. Anal. Chem. 2020, 92, 9895– 9900, DOI: 10.1021/acs.analchem.0c01394[ACS Full Text
], [CAS], Google Scholar
46https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtF2hsr7E&md5=03a6de77c4f4834eeee88edb93b82540Discovery of Aptamers Targeting the Receptor-Binding Domain of the SARS-CoV-2 Spike GlycoproteinSong, Yanling; Song, Jia; Wei, Xinyu; Huang, Mengjiao; Sun, Miao; Zhu, Lin; Lin, Bingqian; Shen, Haicong; Zhu, Zhi; Yang, ChaoyongAnalytical Chemistry (Washington, DC, United States) (2020), 92 (14), 9895-9900CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)The World Health Organization has declared the outbreak of a novel coronavirus (SARS-CoV-2 or 2019-nCoV) as a global pandemic. However, the mechanisms behind the coronavirus infection are not yet fully understood, nor are there any targeted treatments or vaccines. In this study, we identified high-binding-affinity aptamers targeting SARS-CoV-2 RBD, using an ACE2 competition-based aptamer selection strategy and a machine learning screening algorithm. The Kd values of the optimized CoV2-RBD-1C and CoV2-RBD-4C aptamers against RBD were 5.8 nM and 19.9 nM, resp. Simulated interaction modeling, along with competitive expts., suggests that two aptamers may have partially identical binding sites at ACE2 on SARS-CoV-2 RBD. These aptamers present an opportunity for generating new probes for recognition of SARS-CoV-2 and could provide assistance in the diagnosis and treatment of SARS-CoV-2 while providing a new tool for in-depth study of the mechanisms behind the coronavirus infection. - 47Stanborough, T.; Given, F. M.; Koch, B.; Sheen, C. R.; Stowers-Hull, A. B.; Waterland, M. R.; Crittenden, D. L. Optical detection of CoV-SARS-2 viral proteins to sub-picomolar concentrations. ACS Omega 2021, 6, 6404– 6413, DOI: 10.1021/acsomega.1c00008[ACS Full Text
], [CAS], Google Scholar
47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXkslynsb0%253D&md5=d0681c670ebd20831680d4698ba49973Optical detection of CoV-SARS-2 viral proteins to sub-picomolar concentrationsStanborough, Tamsyn; Given, Fiona M.; Koch, Barbara; Sheen, Campbell R.; Stowers-Hull, Andre Buzas; Waterland, Mark R.; Crittenden, Deborah L.ACS Omega (2021), 6 (9), 6404-6413CODEN: ACSODF; ISSN:2470-1343. (American Chemical Society)The emergence of a new strain of coronavirus in late 2019, SARS-CoV-2, led to a global pandemic in 2020. This may have been preventable if large scale, rapid diagnosis of active cases had been possible, and this has highlighted the need for more effective and efficient ways of detecting and managing viral infections. In this work, we investigate three different optical techniques for quantifying the binding of recombinant SARS-CoV-2 spike protein to surface-immobilized oligonucleotide aptamers. Biolayer interferometry is a relatively cheap, robust, and rapid method that only requires very small sample vols. However, its detection limit of 250 nM means that it is not sensitive enough to detect antigen proteins at physiol. relevant levels (sub-pM). Surface plasmon resonance is a more sensitive technique but requires larger sample vols., takes longer, requires more expensive instrumentation, and only reduces the detection limit to 5 nM. Surface-enhanced Raman spectroscopy is far more sensitive, enabling detection of spike protein to sub-picomolar concns. Control expts. performed using scrambled aptamers and using bovine serum albumin as an analyte show that this apta-sensing approach is both sensitive and selective, with no appreciable response obsd. for any controls. Overall, these proof-of-principle results demonstrate that SERS-based aptasensors hold great promise for development into rapid, point-of-use antigen detection systems, enabling mass testing without any need for reagents or lab. expertise and equipment. - 48Pramanik, A.; Gao, Y.; Patibandla, S.; Mitra, D.; McCandless, M. G.; Fassero, L. A.; Gates, K.; Tandon, R.; Ray, P. C. Aptamer conjugated gold nanostar-based distance-dependent nanoparticle surface energy transfer spectroscopy for ultrasensitive detection and inactivation of corona virus. J. Phys. Chem. Lett. 2021, 12, 2166– 2171, DOI: 10.1021/acs.jpclett.0c03570[ACS Full Text
], [CAS], Google Scholar
48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXkvFamurw%253D&md5=8da4b26bba915f2721e957132a26eb96Aptamer conjugated gold nanostar-based distance-dependent nanoparticle surface energy transfer spectroscopy for ultrasensitive detection and inactivation of corona virusPramanik, Avijit; Gao, Ye; Patibandla, Shamily; Mitra, Dipanwita; McCandless, Martin G.; Fassero, Lauren A.; Gates, Kalein; Tandon, Ritesh; Ray, Paresh ChandraJournal of Physical Chemistry Letters (2021), 12 (8), 2166-2171CODEN: JPCLCD; ISSN:1948-7185. (American Chemical Society)The ongoing outbreak of the coronavirus infection has killed more than 2 million people. Herein, we demonstrate that Rhodamine 6G (Rh-6G) dye conjugated DNA aptamer-attached gold nanostar (GNS)-based distance-dependent nanoparticle surface energy transfer (NSET) spectroscopy has the capability of rapid diagnosis of specific SARS-CoV-2 spike recombinant antigen or SARS-CoV-2 spike protein pseudotyped baculovirus within 10 min. Because Rh-6G-attached single-stand DNA aptamer wrapped the GNS, 99% dye fluorescence was quenched because of the NSET process. In the presence of spike antigen or virus, the fluorescence signal persists because of the aptamer-spike protein binding. Specifically, the limit of detection for the NSET assay has been detd. to be 130 fg/mL for antigen and 8 particles/mL for virus. Finally, we have demonstrated that DNA aptamer-attached GNSs can stop virus infection by blocking the angiotensin-converting enzyme 2 (ACE2) receptor binding capability and destroying the lipid membrane of the virus. - 49Schmitz, A.; Weber, A.; Bayin, M.; Breuers, S.; Fieberg, V.; Famulok, M.; Mayer, G. A SARS-CoV-2 spike binding DNA aptamer that inhibits pseudovirus infection by an RBD-independent mechanism. Angew. Chem., Int. Ed. 2021, 60, 2– 9, DOI: 10.1101/2020.12.23.424171
- 50Yang, G.; Li, Z.; Mohammed, I.; Zhao, L.; Wei, W.; Xiao, H.; Guo, W.; Zhao, Y.; Qu, F.; Huang, Y. Identification of SARS-CoV-2-against aptamer with high neutralization activity by blocking the RBD domain of spike protein 1. Signal Transduction Targeted Ther. 2021, 6, 227, DOI: 10.1038/s41392-021-00649-6[Crossref], [PubMed], [CAS], Google Scholar50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhtlegtbzE&md5=368c38793a514ca573275b119ca3c012Identification of SARS-CoV-2-against aptamer with high neutralization activity by blocking the RBD domain of spike protein 1Yang, Ge; Li, Ziyue; Mohammed, Irfan; Zhao, Liping; Wei, Wei; Xiao, Haihua; Guo, Weisheng; Zhao, Yongxiang; Qu, Feng; Huang, YuanyuSignal Transduction and Targeted Therapy (2021), 6 (1), 227CODEN: STTTCB; ISSN:2059-3635. (Nature Research)Identification of SARS-CoV-2-against aptamer with high neutralization activity by blocking the RBD domain of spike protein 1.
- 51Li, J.; Zhang, Z.; Gu, J.; Stacey, H. D.; Ang, J. C.; Capretta, A.; Filipe, C. D. M.; Mossman, K. L.; Balion, C.; Salena, B. J.; Yamamura, D.; Soleymani, L.; Miller, M. S.; Brennan, J. D.; Li, Y. Diverse high-affinity DNA aptamers for wild-type and B.1.1.7 SARS-CoV-2 spike proteins from a pre-structured DNA library. Nucleic Acids Res. 2021, 49, 7267– 7279, DOI: 10.1093/nar/gkab574[Crossref], [PubMed], [CAS], Google Scholar51https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXit12ru7jL&md5=05d191a9e9455c4a809aa0dc261efb3fDiverse high-affinity DNA aptamers for wild-type and B.1.1.7 SARS-CoV-2 spike proteins from a pre-structured DNA libraryLi, Jiuxing; Zhang, Zijie; Gu, Jimmy; Stacey, Hannah D.; Ang, Jann C.; Capretta, Alfredo; Filipe, Carlos D. M.; Mossman, Karen L.; Balion, Cynthia; Salena, Bruno J.; Yamamura, Deborah; Soleymani, Leyla; Miller, Matthew S.; Brennan, John D.; Li, YingfuNucleic Acids Research (2021), 49 (13), 7267-7279CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)We performed in vitro selection expts. to identify DNA aptamers for the S1 subunit of the SARS-CoV-2 spike protein (S1 protein). Using a pool of pre-structured random DNA sequences, we obtained over 100 candidate aptamers after 13 cycles of enrichment under progressively more stringent selection pressure. The top 10 sequences all exhibited strong binding to the S1 protein. Two aptamers, named MSA1 (Kd = 1.8 nM) and MSA5 (Kd = 2.7 nM), were assessed for binding to the heat-treated S1 protein, untreated S1 protein spiked into 50% human saliva and the trimeric spike protein of both the wildtype and the B.1.1.7 variant, demonstrating comparable affinities in all cases. MSA1 and MSA5 also recognized the pseudotyped lentivirus of SARS-CoV-2 with resp. Kd values of 22.7 pM and 11.8 pM. Secondary structure prediction and sequence truncation expts. revealed that both MSA1 and MSA5 adopted a hairpin structure, which was the motif pre-designed into the original library. A colorimetric sandwich assay was developed using MSA1 as both the recognition element and detection element, which was capable of detecting the pseudotyped lentivirus in 50% saliva with a limit of detection of 400 fM, confirming the potential of these aptamers as diagnostic tools for COVID-19 detection.
- 52Gupta, A.; Anand, A.; Jain, N.; Goswami, S.; Anantharaj, A.; Patil, S.; Singh, R.; Kumar, A.; Shrivastava, T.; Bhatnagar, S.; Medigeshi, G. R.; Sharma, T. K.; DBT India Consortium for COVID-19 Research A novel G-quadruplex aptamer-based spike trimeric antigen test for the detection of SARS-CoV-2. Mol. Ther.--Nucleic Acids 2021, 26, 321– 332, DOI: 10.1016/j.omtn.2021.06.014[Crossref], [PubMed], [CAS], Google Scholar52https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvFGrt7nI&md5=9dd950049a6d402e6c37542d4e66185dA novel G-quadruplex aptamer-based spike trimeric antigen test for the detection of SARS-CoV-2Gupta, Ankit; Anand, Anjali; Jain, Neha; Goswami, Sandeep; Anantharaj, Anbalagan; Patil, Sharanabasava; Singh, Rahul; Kumar, Amit; Shrivastava, Tripti; Bhatnagar, Shinjini; Medigeshi, Guruprasad R.; Sharma, Tarun KumarMolecular Therapy--Nucleic Acids (2021), 26 (), 321-332CODEN: MTAOC5; ISSN:2162-2531. (Elsevier Inc.)The recent SARS-CoV-2 outbreak has been declared a global health emergency. It will take years to vaccinate the whole population to protect them from this deadly virus, hence the management of SARS-CoV-2 largely depends on the widespread availability of an accurate diagnostic test. Toward addressing the unmet need of a reliable diagnostic test in the current work by utilizing the power of Systematic Evolution of Ligands by EXponential enrichment, a 44-mer G-quadruplex-forming DNA aptamer against spike trimer antigen of SARS-CoV-2 was identified. The lead aptamer candidate (S14) was characterized thoroughly for its binding, selectivity, affinity, structure, and batch-to-batch variability by utilizing various biochem., biophys., and in silico techniques. S14 has demonstrated a low nanomolar KD, confirming its tight binding to a spike antigen of SARS-CoV-2. S14 can detect as low as 2 nM of antigen. The clin. evaluation of S14 aptamer on nasopharyngeal swab specimens (n = 232) has displayed a highly discriminatory response between SARS-CoV-2 infected individuals from the non-infected one with a sensitivity and specificity of ∼91% and 98%, resp. Importantly, S14 aptamer-based test has evinced a comparable performance with that of RT-PCR-based assay. Altogether, this study established the utility of aptamer technol. for the detection of SARS-CoV-2.
- 53Alves Ferreira-Bravo, I.; DeStefano, J. J. Xeno-nucleic Acid (XNA) 2’-fluoro-arabino nucleic acid (FANA) aptamers to the receptor-binding domain of SARS-CoV-2 S protein block ACE2 binding. Viruses 2021, 13, 1983, DOI: 10.3390/v13101983[Crossref], [PubMed], [CAS], Google Scholar53https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXitlOhsL%252FL&md5=4b8c95f265f1068e63decbcbbb1dc5f7Xeno-Nucleic Acid (XNA) 2'-Fluoro-Arabino Nucleic Acid (FANA) Aptamers to the Receptor-Binding Domain of SARS-CoV-2 S Protein Block ACE2 BindingAlves Ferreira-Bravo, Irani; DeStefano, Jeffrey J.Viruses (2021), 13 (10), 1983CODEN: VIRUBR; ISSN:1999-4915. (MDPI AG)The causative agent of COVID-19, SARS-CoV-2, gains access to cells through interactions of the receptor-binding domain (RBD) on the viral S protein with angiotensin-converting enzyme 2 (ACE2) on the surface of human host cells. Systematic evolution of ligands by exponential enrichment (SELEX) was used to generate aptamers (nucleic acids selected for high binding affinity to a target) to the RBD made from 2'-fluoro-arabinonucleic acid (FANA). The best selected ∼79 nucleotide aptamers bound the RBD (Arg319-Phe541) and the larger S1 domain (Val16-Arg685) of the 1272 amino acid S protein with equil. dissocn. consts. (KD,app) of 10-20 nM, and binding half-life for the RBD, S1 domain, and full trimeric S protein of 53 ± 18, 76 ± 5, and 127 ± 7 min, resp. Aptamers inhibited the binding of the RBD to ACE2 in an ELISA assay. Inhibition, on a per wt. basis, was similar to neutralizing antibodies that were specific for RBD. Aptamers demonstrated high specificity, binding with about 10-fold lower affinity to the related S1 domain from the original SARS virus, which also binds to ACE2. Overall, FANA aptamers show affinities comparable to previous DNA aptamers to RBD and S1 protein and directly block receptor interactions while using an alternative Xeno-nucleic acid (XNA) platform.
- 54Behbahani, M.; Mohabatkar, H.; Hosseini, B. In silico design of quadruplex aptamers against the spike protein of SARS-CoV-2. Inf. Med. 2021, 26, 100757, DOI: 10.1016/j.imu.2021.100757[Crossref], [CAS], Google Scholar54https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB2cjhs12iuw%253D%253D&md5=df8b1a0d86351a2967314d233e85f648In silico design of quadruplex aptamers against the spike protein of SARS-CoV-2Behbahani Mandana; Mohabatkar Hassan; Hosseini BarumandInformatics in medicine unlocked (2021), 26 (), 100757 ISSN:2352-9148.Nucleic acid aptamers are short sequences of nucleic acid ligands that bind to a specific target molecule. Aptamers are experimentally nominated using the well-designed SELEX (systematic evolution of ligands by exponential enrichment) method. Here, we designed a new method for diagnosis and blocking SARS-CoV-2 based on G-quadruplex aptamer. This aptamer was developed against the receptor-binding domain (RBD) region of the spike protein. In the current study, ten quadruplex DNA aptamers entitled AP1, AP2, AP3, AP4, AP5, AP6, AP7, AP8, AP9, and AP10 were designed in silico and had high HADDOCK scores. One quadruplex aptamer sequence (AP1) was selected based on the interaction with RBD of SARS-CoV-2. Results showed that AP1 aptamer could be used as an agent in the diagnosis and therapy of SARS-CoV-2, although more works are still needed.
- 55Devi, A.; Chaitanya, N. S. N. Designing of peptide aptamer targeting the receptor-binding domain of spike protein of SARS-CoV-2: an in silico study. Mol. Diversity 2021, 1– 13, DOI: 10.1007/s11030-020-10171-6
- 56Artyushenko, P. V.; Mironov, V. A.; Mironov, V. A.; Morozov, D. I.; Shchugoreva, I. A.; Borbone, N.; Tomilin, F. N.; Kichkailo, A. S. Computational approach to design of aptamers to the receptor binding domain of SARS-CoV-2. Sib. Med. Rev. 2021, 2, 66– 67, DOI: 10.20333/25000136-2021-2-66-67
- 57Kowada, A. Greater public health impact of COVID-19 antigen detection tests. BMC Med. 2021, 19, 82, DOI: 10.1186/s12916-021-01956-z[Crossref], [PubMed], [CAS], Google Scholar57https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXnsVGgsrc%253D&md5=697f8c67d8f02c80f7b7769f03a1aa19Greater public health impact of COVID-19 antigen detection testsKowada, AkikoBMC Medicine (2021), 19 (1), 82CODEN: BMMECZ; ISSN:1741-7015. (BioMed Central Ltd.)Early diagnosis of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection prevents widespread transmission, manages outbreak control, improves poor prognosis, and reduces mortality of the Coronavirus disease 2019 (COVID-19). Ricks et al. report in BMC Medicine greater healthcare cost-saving and health outcomes of antigen detection rapid diagnostic tests compared to nucleic acid tests for symptomatic persons. The main outcomes in the study are the health system costs and health impacts (deaths averted and infectious days isolated). Antigen-detection rapid diagnostic test-led strategy is compared to a strategy based on nucleic acid tests and clin. judgment.
- 58Pomplun, S.; Jbara, M.; Quartararo, A. J.; Zhang, G.; Brown, J. S.; Lee, Y.-C.; Ye, X.; Hanna, S.; Pentelute, B. L. De novo discovery of high-affinity peptide binders for the SARS-CoV-2 spike protein. ACS Cent. Sci. 2021, 7, 156– 163, DOI: 10.1021/acscentsci.0c01309[ACS Full Text
], [CAS], Google Scholar
58https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXis1WrsbfM&md5=1cee08cf67569ada5d5ca76bdd2ae059De novo discovery of high-affinity peptide binders for the SARS-CoV-2 spike proteinPomplun, Sebastian; Jbara, Muhammad; Quartararo, Anthony J.; Zhang, Genwei; Brown, Joseph S.; Lee, Yen-Chun; Ye, Xiyun; Hanna, Stephanie; Pentelute, Bradley L.ACS Central Science (2021), 7 (1), 156-163CODEN: ACSCII; ISSN:2374-7951. (American Chemical Society)The β-coronavirus SARS-CoV-2 has caused a global pandemic. Affinity reagents targeting the SARS-CoV-2 spike protein are of interest for the development of therapeutics and diagnostics. We used affinity selection-mass spectrometry for the rapid discovery of synthetic high-affinity peptide binders for the receptor binding domain (RBD) of the SARS-CoV-2 spike protein. From library screening with 800 million synthetic peptides, we identified three sequences with nanomolar affinities (dissocn. consts. Kd = 80-970 nM) for RBD and selectivity over human serum proteins. Nanomolar RBD concns. in a biol. matrix could be detected using the biotinylated lead peptide in ELISA format. These peptides do not compete for ACE2 binding, and their site of interaction on the SARS-CoV-2-spike-RBD might be unrelated to the ACE2 binding site, making them potential orthogonal reagents for sandwich immunoassays. These findings serve as a starting point for the development of SARS-CoV-2 diagnostics or conjugates for virus-directed delivery of therapeutics. Using affinity selection-mass spectrometry, we discovered synthetic peptides with nanomolar affinity for the SARS-CoV-2-spike-RBD and selectivity over human proteins. - 59Kaur, N.; Singh, R.; Dar, Z.; Bijarnia, R. K.; Dhingra, N.; Kaur, T. Genetic comparison among various coronavirus strains for the identification of potential vaccine targets of SARS-CoV2. Infect., Genet. Evol. 2021, 89, 104490, DOI: 10.1016/j.meegid.2020.104490[Crossref], [PubMed], [CAS], Google Scholar59https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXivVWrsr0%253D&md5=dbed715a9b6a000a681c9d83d1e136b5Genetic comparison among various coronavirus strains for the identification of potential vaccine targets of SARS-CoV2Kaur, Navpreet; Singh, Rimaljot; Dar, Zahid; Bijarnia, Rakesh Kumar; Dhingra, Neelima; Kaur, TanzeerInfection, Genetics and Evolution (2021), 89 (), 104490CODEN: IGENCN; ISSN:1567-1348. (Elsevier B.V.)A review. On-going pandemic pneumonia outbreak COVID-19 has raised an urgent public health issue worldwide impacting millions of people with a continuous increase in both morbidity and mortality. The causative agent of this disease is identified and named as SARS-CoV2 because of its genetic relatedness to SARS-CoV species that was responsible for the 2003 coronavirus outbreak. The immense spread of the disease in a very small period demands urgent development of therapeutic and prophylactic interventions for the treatment of SARS-CoV2 infected patients. A plethora of research is being conducted globally on this novel coronavirus strain to gain knowledge about its origin, evolutionary history, and phylogeny. This review is an effort to compare genetic similarities and diversifications among coronavirus strains, which can hint towards the susceptible antigen targets of SARS-CoV2 to come up with the potential therapeutic and prophylactic interventions for the prevention of this public threat.
- 60Loos, C.; Atyeo, C.; Fischinger, S.; Burke, J.; Slein, M. D.; Streeck, H.; Lauffenburger, D.; Ryan, E. T.; Charles, R. C.; Alter, G. Evolution of early SARS-CoV-2 and cross-coronavirus immunity. mSphere 2020, 5, e00622– 20, DOI: 10.1128/mSphere.00622-20[Crossref], [PubMed], [CAS], Google Scholar60https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvFOksbzK&md5=e23c18b6205cff7e4addd50d989c6441Evolution of early SARS-CoV-2 and cross-coronavirus immunityLoos, Carolin; Atyeo, Caroline; Fischinger, Stephanie; Burke, John; Slein, Matthew D.; Streeck, Hendrik; Lauffenburger, Douglas; Ryan, Edward T.; Charles, Richelle C.; Alter, GalitmSphere (2020), 5 (5), e00622-20CODEN: MSPHCI; ISSN:2379-5042. (American Society for Microbiology)The novel coronavirus, SARS-coronavirus (CoV)-2 (SARS-CoV-2), has caused over 17 million infections in just a few months, with disease manifestations ranging from largely asymptomatic infection to critically severe disease. The remarkable spread and unpredictable disease outcomes continue to challenge management of this infection. Among the hypotheses to explain the heterogeneity of symptoms is the possibility that exposure to other coronaviruses (CoVs), or overall higher capability to develop immunity against respiratory pathogens, may influence the evolution of immunity to SARS-CoV-2. Thus, we profiled the immune response across multiple coronavirus receptor binding domains (RBDs), respiratory viruses, and SARS-CoV-2, to det. whether heterologous immunity to other CoV-RBDs or other infections influenced the evolution of the SARS-CoV-2 humoral immune response. Overall changes in subclass, isotype, and Fc-receptor binding were profiled broadly across a cohort of 43 individuals against different coronaviruses-RBDs of SARS-CoV-2 and the more common HKU1 and NL63 viruses. We found rapid functional evolution of responses to SARS-CoV-2 over time, along with broad but relatively more time-invariant responses to the more common CoVs. Moreover, there was little evidence of correlation between SARS-CoV-2 responses and HKU1, NL63, and respiratory infection (influenza and respiratory syncytial virus) responses. These findings suggest that common viral infections including common CoV immunity, targeting the receptor binding domain involved in viral infection, do not appear to influence the rapid functional evolution of SARS-CoV-2 immunity, and thus should not impact diagnostics or shape vaccine-induced immunity.
- 61Li, F. Structure, function, and evolution of coronavirus spike proteins. Annu. Rev. Virol. 2016, 3, 237– 261, DOI: 10.1146/annurev-virology-110615-042301[Crossref], [PubMed], [CAS], Google Scholar61https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsVegsb%252FF&md5=7a64c5330380b5e82b65594512294ffbStructure, Function, and Evolution of Coronavirus Spike ProteinsLi, FangAnnual Review of Virology (2016), 3 (), 237-261CODEN: ARVNBS; ISSN:2327-0578. (Annual Reviews)The coronavirus spike protein is a multifunctional mol. machine that mediates coronavirus entry into host cells. It first binds to a receptor on the host cell surface through its S1 subunit and then fuses viral and host membranes through its S2 subunit. Two domains in S1 from different coronaviruses recognize a variety of host receptors, leading to viral attachment. The spike protein exists in two structurally distinct conformations, prefusion and postfusion. The transition from prefusion to postfusion conformation of the spike protein must be triggered, leading to membrane fusion. This article reviews current knowledge about the structures and functions of coronavirus spike proteins, illustrating how the two S1 domains recognize different receptors and how the spike proteins are regulated to undergo conformational transitions. I further discuss the evolution of these two crit. functions of coronavirus spike proteins, receptor recognition and membrane fusion, in the context of the corresponding functions from other viruses and host cells.
- 62Andersen, K. G.; Rambaut, A.; Lipkin, W. I.; Holmes, E. C.; Garry, R. F. The proximal origin of SARS-CoV-2. Nat. Med. 2020, 26, 450– 452, DOI: 10.1038/s41591-020-0820-9[Crossref], [PubMed], [CAS], Google Scholar62https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXltFCjtbY%253D&md5=3489259c33e29365c0c1cf7fc5613407The proximal origin of SARS-CoV-2Andersen, Kristian G.; Rambaut, Andrew; Lipkin, W. Ian; Holmes, Edward C.; Garry, Robert F.Nature Medicine (New York, NY, United States) (2020), 26 (4), 450-452CODEN: NAMEFI; ISSN:1078-8956. (Nature Research)There is no expanded citation for this reference.
- 63Ou, X.; Liu, Y.; Lei, X.; Li, P.; Mi, D.; Ren, L.; Guo, L.; Guo, R.; Chen, T.; Hu, J.; Xiang, Z.; Mu, Z.; Chen, X.; Chen, J.; Hu, K.; Jin, Q.; Wang, J.; Qian, Z. Characterization of spike glycoprotein of SARS-CoV-2 on virus entry and its immune cross-reactivity with SARS-CoV. Nat. Commun. 2020, 11, 1620, DOI: 10.1038/s41467-020-15562-9[Crossref], [PubMed], [CAS], Google Scholar63https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXlvFyjt78%253D&md5=6b0b1ef5a68f4a35da4aabecb0f99544Characterization of spike glycoprotein of SARS-CoV-2 on virus entry and its immune cross-reactivity with SARS-CoVOu, Xiuyuan; Liu, Yan; Lei, Xiaobo; Li, Pei; Mi, Dan; Ren, Lili; Guo, Li; Guo, Ruixuan; Chen, Ting; Hu, Jiaxin; Xiang, Zichun; Mu, Zhixia; Chen, Xing; Chen, Jieyong; Hu, Keping; Jin, Qi; Wang, Jianwei; Qian, ZhaohuiNature Communications (2020), 11 (1), 1620CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)Since 2002, beta coronaviruses (CoV) have caused three zoonotic outbreaks, SARS-CoV in 2002-2003, MERS-CoV in 2012, and the newly emerged SARS-CoV-2 in late 2019. However, little is currently known about the biol. of SARS-CoV-2. Here, using SARS-CoV-2 S protein pseudovirus system, we confirm that human angiotensin converting enzyme 2 (hACE2) is the receptor for SARS-CoV-2, find that SARS-CoV-2 enters 293/hACE2 cells mainly through endocytosis, that PIKfyve, TPC2, and cathepsin L are crit. for entry, and that SARS-CoV-2 S protein is less stable than SARS-CoV S. Polyclonal anti-SARS S1 antibodies T62 inhibit entry of SARS-CoV S but not SARS-CoV-2 S pseudovirions. Further studies using recovered SARS and COVID-19 patients' sera show limited cross-neutralization, suggesting that recovery from one infection might not protect against the other. Our results present potential targets for development of drugs and vaccines for SARS-CoV-2.
- 64Idili, A.; Parolo, C.; Alvarez-Diduk, R.; Merkoçi, A. Rapid and efficient detection of the SARS-CoV-2 spike protein using an electrochemical aptamer-based sensor. ACS Sens. 2021, 6, 3093– 3101, DOI: 10.1021/acssensors.1c01222[ACS Full Text
], [CAS], Google Scholar
64https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhslegsLvI&md5=eed8021b69f60f966c493363ef2aa479Rapid and Efficient Detection of the SARS-CoV-2 Spike Protein Using an Electrochemical Aptamer-Based SensorIdili, Andrea; Parolo, Claudio; Alvarez-Diduk, Ruslan; Merkoci, ArbenACS Sensors (2021), 6 (8), 3093-3101CODEN: ASCEFJ; ISSN:2379-3694. (American Chemical Society)The availability of sensors able to rapidly detect SARS-CoV-2 directly in biol. fluids in a single step would allow performing massive diagnostic testing to track in real time and contain the spread of COVID-19. Motivated by this, here, we developed an electrochem. aptamer-based (EAB) sensor able to achieve the rapid, reagentless, and quant. measurement of the SARS-CoV-2 spike (S) protein. First, we demonstrated the ability of the selected aptamer to undergo a binding-induced conformational change in the presence of its target using fluorescence spectroscopy. Then, we engineered the aptamer to work as a bioreceptor in the EAB platform and we demonstrated its sensitivity and specificity. Finally, to demonstrate the clin. potential of the sensor, we tested it directly in biol. fluids (serum and artificial saliva), achieving the rapid (minutes) and single-step detection of the S protein in its clin. range. - 65Zakashansky, J. A.; Imamura, A. H.; Salgado, D. F., 2nd; Romero Mercieca, H. C.; Aguas, R. F. L.; Lao, A. M.; Pariser, J.; Arroyo-Currás, N.; Khine, M. Detection of the SARS-CoV-2 spike protein in saliva with Shrinky-Dink electrodes. Anal. Methods 2021, 13, 874– 883, DOI: 10.1039/d1ay00041a[Crossref], [PubMed], [CAS], Google Scholar65https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXjvVCru7c%253D&md5=42942831b25fbc59924f0f80d848b087Detection of the SARS-CoV-2 spike protein in saliva with Shrinky-Dink electrodesZakashansky, Julia A.; Imamura, Amanda H.; Salgado, Darwin F., II; Romero Mercieca, Heather C.; Aguas, Raphael F. L.; Lao, Angelou M.; Pariser, Joseph; Arroyo-Curras, Netzahualcoyotl; Khine, MichelleAnalytical Methods (2021), 13 (7), 874-883CODEN: AMNEGX; ISSN:1759-9679. (Royal Society of Chemistry)A review. Using the children's toy, Shrinky-Dink, we present an aptamer-based electrochem. (E-AB) assay that recognizes the spike protein of SARS-CoV-2 in saliva for viral infection detection. The low-cost electrodes are implementable at population scale and demonstrate detection down to 1 ag mL-1 of the S1 subunit of the spike protein.
- 66Cennamo, N.; Pasquardini, L.; Arcadio, F.; Lunelli, L.; Vanzetti, L.; Carafa, V.; Altucci, L.; Zeni, L. SARS-CoV-2 spike protein detection through a plasmonic D-shaped plastic optical fiber aptasensor. Talanta 2021, 233, 122532, DOI: 10.1016/j.talanta.2021.122532[Crossref], [PubMed], [CAS], Google Scholar66https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhtFKrs77M&md5=918249e712869852a7e75668d1e1f903SARS-CoV-2 spike protein detection through a plasmonic D-shaped plastic optical fiber aptasensorCennamo, Nunzio; Pasquardini, Laura; Arcadio, Francesco; Lunelli, Lorenzo; Vanzetti, Lia; Carafa, Vincenzo; Altucci, Lucia; Zeni, LuigiTalanta (2021), 233 (), 122532CODEN: TLNTA2; ISSN:0039-9140. (Elsevier B.V.)A specific aptameric sequence has been immobilized on short polyethyleneglycol (PEG) interface on gold nano-film deposited on a D-shaped plastic optical fiber (POFs) probe, and the protein binding has been monitored exploiting the very sensitive surface plasmon resonance (SPR) phenomenon. The receptor-binding domain (RBD) of the SARS-CoV-2 spike glycoprotein has been specifically used to develop an aptasensor. Surface anal. techniques coupled to fluorescence microscopy and plasmonic anal. have been utilized to characterize the biointerface. Spanning a wide protein range (25 ÷ 1000 nM), the SARS-Cov-2 spike protein was detected with a Limit of Detection (LoD) of about 37 nM. Different interferents (BSA, AH1N1 hemagglutinin protein and MERS spike protein) have been tested confirming the specificity of our aptasensor. Finally, a preliminary test in dild. human serum encouraged its application in a point-of-care device, since POF-based aptasensor represent a potentially low-cost compact biosensor, characterized by a rapid response, a small size and could be an ideal lab. portable diagnostic tool.
- 67Aithal, S.; Mishriki, S.; Gupta, R.; Sahu, R. P.; Botos, G.; Tanvir, S.; Hanson, R. W.; Puri, I. K. SARS-CoV-2 detection with aptamer-functionalized gold nanoparticles. Talanta 2022, 236, 122841, DOI: 10.1016/j.talanta.2021.122841[Crossref], [PubMed], [CAS], Google Scholar67https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvFyjsbbP&md5=5f4e1f429d578a71f079547f697cb496SARS-CoV-2 detection with aptamer-functionalized gold nanoparticlesAithal, Srivatsa; Mishriki, Sarah; Gupta, Rohit; Sahu, Rakesh P.; Botos, George; Tanvir, Shazia; Hanson, Russell W.; Puri, Ishwar K.Talanta (2022), 236 (), 122841CODEN: TLNTA2; ISSN:0039-9140. (Elsevier B.V.)A rapid detection test for SARS-CoV-2 is urgently required to monitor virus spread and containment. Here, we describe a test that uses nanoprobes, which are gold nanoparticles functionalized with an aptamer specific to the spike membrane protein of SARS-CoV-2. An ELISA confirms aptamer binding with the spike protein on gold surfaces. Protein recognition occurs by adding a coagulant, where nanoprobes with no bound protein agglomerate while those with sufficient bound protein do not. Using plasmon absorbance spectra, the nanoprobes detect 16 nM and higher concns. of spike protein in phosphate-buffered saline. The time-varying light absorbance is examd. at 540 nm to det. the crit. coagulant concn. required to agglomerates the nanoprobes, which depends on the protein concn. This approach detects 3540 genome copies/μl of inactivated SARS-CoV-2.
Supporting Information
Supporting Information
ARTICLE SECTIONSThe Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsomega.1c05521.
Recombinant proteins and aptamers used in this work; biosensors and assays developed for the detection of the SARS-CoV-2 S antigen; binding studies by direct ELAA to monitor S proteins interactions with aptamers; checkerboard titration of concentrations of capture and detection aptamers to optimize aptamer sandwich assay; sensitivity of the sandwich aptamer assay at different concentrations of reporter aptamer; and SARS-CoV-2 RNA detection in NP swab samples by droplet digital PCR analysis (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.