1
|
Kang CK, Kim Y, Jo HJ, Lee CM, Kim NJ, Lee CH, Choe PG, Park WB, Oh MD. Anti-SARS-CoV-2 Nucleocapsid Antibody Positivity Three Years after COVID-19. Jpn J Infect Dis 2024; 77:285-288. [PMID: 38684427 DOI: 10.7883/yoken.jjid.2024.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
The accurate identification of individuals without prior infection with severe acute respiratory syndrome coronavirus 2 is pivotal for seroepidemiological studies and vaccine trials. Owing to widespread vaccination against coronavirus disease 2019 (COVID-19), the anti-nucleocapsid antibody continues to serve as a valuable marker for individuals without a history of COVID-19. This study aimed to comprehensively assess anti-nucleocapsid antibody positivity using diverse commercial and in-house immunoassays among individuals who contracted COVID-19 more than three years earlier. We enrolled 44 participants with laboratory-confirmed COVID-19 between January and May 2020 from the Seoul National University Hospital and its community treatment centers. The results showed anti-nucleocapsid antibody positivity ranging from 45.5% to 87.9%, depending on the immunoassay. This study highlights the importance of considering the limited anti-nucleocapsid antibody positivity in individuals, with a history of distant COVID-19, in seroepidemiological or vaccine research.
Collapse
Affiliation(s)
- Chang Kyung Kang
- Department of Internal Medicine, Seoul National University College of Medicine, Republic of Korea
| | - Youngju Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Republic of Korea
- Department of Pharmacology, Seoul National University College of Medicine, Republic of Korea
| | - Hyeon Jae Jo
- Department of Internal Medicine, Seoul National University College of Medicine, Republic of Korea
| | - Chan Mi Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Republic of Korea
| | - Nam Joong Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Republic of Korea
| | - Chang-Han Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Republic of Korea
- Department of Pharmacology, Seoul National University College of Medicine, Republic of Korea
- Wide River Institute of Immunology, Seoul National University, Republic of Korea
- Convergence Research Center for Dementia, Seoul National University Medical Research Center, Republic of Korea
- Cancer Research Institute, Seoul National University Medical Research Center, Republic of Korea
| | - Pyoeng Gyun Choe
- Department of Internal Medicine, Seoul National University College of Medicine, Republic of Korea
| | - Wan Beom Park
- Department of Internal Medicine, Seoul National University College of Medicine, Republic of Korea
| | - Myoung-Don Oh
- Department of Internal Medicine, Seoul National University College of Medicine, Republic of Korea
| |
Collapse
|
2
|
Pathirana SL, Deepachandi B, Gunasekara P, Fernando N, Perera IC, Gangani D, Thambyarajah J, Dasanayake D, de Silva R, Premawansa S, Nitsche A, Handunnetti SM. Development and Evaluation of In-House ELISAs for the Detection of SARS-CoV-2-Specific Antibodies in COVID-19 Patients in Sri Lanka. Int J Microbiol 2024; 2024:1331067. [PMID: 39346702 PMCID: PMC11427722 DOI: 10.1155/2024/1331067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 05/29/2024] [Accepted: 08/27/2024] [Indexed: 10/01/2024] Open
Abstract
COVID-19 serological tests complement the molecular diagnostics and can be used as important tool for serosurveillance and vaccine efficiency evaluation. The aim of this study was to develop and evaluate the diagnostic performance of an in-house ELISA for retrospective serosurveillance of SARS-CoV-2. Total IgG and IgM levels in sera of PCR positive SARS-CoV-2 patients (n = 50) from North Colombo Teaching Hospital were evaluated and compared with sera (n = 50) collected from prepandemic healthy individuals as controls. Patient sample collection was initiated before vaccination programme was widely started within the country. Seropositivity of 94.0% (n = 47/50) was observed for either IgG or IgM anti-SARS-CoV-2 antibodies against receptor binding domain of spike protein or nucleocapsid protein in confirmed cases while none of controls were seropositive. In contrast, the seropositivity of only 48.0% (n = 24/50) was demonstrated with commercial ELISA kits for detection of IgG or IgM. All samples detected seropositive by commercially available kits remained seropositive with either in-house IgM or IgG ELISA. Significant correlations (p ≤ 0.001) were observed between Ab levels and day of sampling from the onset of illness. The overall sensitivity values of the in-house assays were 66.7%, 96.9%, and 100.0% for the first, second, and third week or longer after onset of symptoms for either in-house IgM or IgG ELISAs. Majority of the patients (>80.0%) were seropositive, regardless of age (<60 vs. >60 years), gender (male vs. female), or clinical severity (mild vs. moderate/severe). These data suggest that the developed in-house ELISAs can be applied to assess anti-SARS-CoV-2 antibody levels induced by either natural infections or vaccination.
Collapse
Affiliation(s)
- Sisira L Pathirana
- Institute of Biochemistry Molecular Biology and Biotechnology University of Colombo, Colombo, Sri Lanka
| | - Bhagya Deepachandi
- Institute of Biochemistry Molecular Biology and Biotechnology University of Colombo, Colombo, Sri Lanka
- Department of Life Sciences Faculty of Science, NSBM Green University, Mahenwaththa, Pitipana, Homagama, Sri Lanka
| | - Peshala Gunasekara
- Institute of Biochemistry Molecular Biology and Biotechnology University of Colombo, Colombo, Sri Lanka
| | - Narmada Fernando
- Institute of Biochemistry Molecular Biology and Biotechnology University of Colombo, Colombo, Sri Lanka
| | - Inoka C Perera
- Department of Zoology and Environment Sciences Faculty of Science University of Colombo, Colombo, Sri Lanka
| | - Dakshika Gangani
- Institute of Biochemistry Molecular Biology and Biotechnology University of Colombo, Colombo, Sri Lanka
| | - James Thambyarajah
- Institute of Biochemistry Molecular Biology and Biotechnology University of Colombo, Colombo, Sri Lanka
| | | | - Rajiva de Silva
- Department of Immunology Medical Research Institute, Colombo, Sri Lanka
| | - Sunil Premawansa
- Department of Zoology and Environment Sciences Faculty of Science University of Colombo, Colombo, Sri Lanka
| | - Andreas Nitsche
- Center for Biological Threats and Special Pathogens Robert Koch Institute, Berlin, Germany
| | - Shiroma M Handunnetti
- Institute of Biochemistry Molecular Biology and Biotechnology University of Colombo, Colombo, Sri Lanka
| |
Collapse
|
3
|
De Kesel W, Vanden Broecke B, Borremans B, Fourchault L, Willems E, Ceulemans A, Sabuni C, Massawe A, Makundi RH, Leirs H, Peeters M, Verheyen E, Gryseels S, Mariën J, Ariën KK. Antibodies against medically relevant arthropod-borne viruses in the ubiquitous African rodent Mastomys natalensis. PLoS Negl Trop Dis 2024; 18:e0012233. [PMID: 39231158 PMCID: PMC11404846 DOI: 10.1371/journal.pntd.0012233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/16/2024] [Accepted: 08/20/2024] [Indexed: 09/06/2024] Open
Abstract
Over the past decades, the number of arthropod-borne virus (arbovirus) outbreaks has increased worldwide. Knowledge regarding the sylvatic cycle (i.e., non-human hosts/environment) of arboviruses is limited, particularly in Africa, and the main hosts for virus maintenance are unknown. Previous studies have shown the presence of antibodies against certain arboviruses (i.e., chikungunya-, dengue-, and Zika virus) in African non-human primates and bats. We hypothesize that small mammals, specifically rodents, may function as amplifying hosts in anthropogenic environments. The detection of RNA of most arboviruses is complicated by the viruses' short viremic period within their hosts. An alternative to determine arbovirus hosts is by detecting antibodies, which can persist several months. Therefore, we developed a high-throughput multiplex immunoassay to detect antibodies against 15 medically relevant arboviruses. We used this assay to assess approximately 1,300 blood samples of the multimammate mouse, Mastomys natalensis from Tanzania. In 24% of the samples, we detected antibodies against at least one of the tested arboviruses, with high seroprevalences of antibodies reacting against dengue virus serotype one (7.6%) and two (8.4%), and chikungunya virus (6%). Seroprevalence was higher in females and increased with age, which could be explained by inherent immunity and behavioral differences between sexes, and the increased chance of exposure to an arbovirus with age. We evaluated whether antibodies against multiple arboviruses co-occur more often than randomly and found that this may be true for some members of the Flaviviridae and Togaviridae. In conclusion, the development of an assay against a wide diversity of medically relevant arboviruses enabled the analysis of a large sample collection of one of the most abundant African small mammals. Our findings highlight that Mastomys natalensis is involved in the transmission cycle of multiple arboviruses and provide a solid foundation to better understand the role of this ubiquitous rodent in arbovirus outbreaks.
Collapse
Affiliation(s)
- Wim De Kesel
- Evolutionary Ecology Group, Department of Biology, Faculty of Science, University of Antwerp, Antwerp, Belgium
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Bram Vanden Broecke
- Evolutionary Ecology Group, Department of Biology, Faculty of Science, University of Antwerp, Antwerp, Belgium
- Terrestrial Ecology Unit, Department of Biology, Ghent University, Ghent, Belgium
| | - Benny Borremans
- Evolutionary Ecology Group, Department of Biology, Faculty of Science, University of Antwerp, Antwerp, Belgium
- Wildlife Health Ecology Research Organization, San Diego, California, United States of America
| | - Léa Fourchault
- OD Taxonomy & Phylogeny, Royal Belgian Institute of Natural Sciences, Brussels, Belgium
| | - Elisabeth Willems
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Ann Ceulemans
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
- Virus Ecology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Christopher Sabuni
- Institute of Pest Management, Sokoine University of Agriculture, Morogoro, Tanzania
| | - Apia Massawe
- Institute of Pest Management, Sokoine University of Agriculture, Morogoro, Tanzania
| | - Rhodes H Makundi
- Institute of Pest Management, Sokoine University of Agriculture, Morogoro, Tanzania
| | - Herwig Leirs
- Evolutionary Ecology Group, Department of Biology, Faculty of Science, University of Antwerp, Antwerp, Belgium
| | - Martine Peeters
- TransVIHMI, University of Montpellier, Institut de Recherche pour le Développement (IRD), INSERM, Montpellier, France
| | - Erik Verheyen
- Evolutionary Ecology Group, Department of Biology, Faculty of Science, University of Antwerp, Antwerp, Belgium
- OD Taxonomy & Phylogeny, Royal Belgian Institute of Natural Sciences, Brussels, Belgium
| | - Sophie Gryseels
- Evolutionary Ecology Group, Department of Biology, Faculty of Science, University of Antwerp, Antwerp, Belgium
- OD Taxonomy & Phylogeny, Royal Belgian Institute of Natural Sciences, Brussels, Belgium
| | - Joachim Mariën
- Evolutionary Ecology Group, Department of Biology, Faculty of Science, University of Antwerp, Antwerp, Belgium
- Virus Ecology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Kevin K Ariën
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
- Department of Biomedical sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
4
|
Kuthning D, Raafat D, Holtfreter S, Gramenz J, Wittmann N, Bröker BM, Meyer-Bahlburg A. Variant-specific antibody profiling for tracking SARS-CoV-2 variant infections in children and adolescents. Front Immunol 2024; 15:1434291. [PMID: 39257574 PMCID: PMC11384586 DOI: 10.3389/fimmu.2024.1434291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 08/05/2024] [Indexed: 09/12/2024] Open
Abstract
Monitoring the seroprevalence of SARS-CoV-2 in children and adolescents can provide valuable information for effective SARS-CoV-2 surveillance, and thus guide vaccination strategies. In this study, we quantified antibodies against the spike S1 domains of several SARS-CoV-2 variants (wild-type, Alpha, Delta, and Omicron variants) as well as endemic human coronaviruses (HCoVs) in 1,309 children and adolescents screened between December 2020 and March 2023. Their antibody binding profiles were compared with those of 22 pre-pandemic samples from children and adolescents using an in-house Luminex®-based Corona Array (CA). The primary objectives of this study were to (i) monitor SARS-CoV-2-specific antibodies in children and adolescents, (ii) evaluate whether the S1-specific antibody response can identify the infecting variant of concern (VoC), (iii) estimate the prevalence of silent infections, and (iv) test whether vaccination or infection with SARS-CoV-2 induce HCoV cross-reactive antibodies. Both SARS-CoV-2 infection and vaccination induced a robust antibody response against the S1 domain of WT and VoCs in children and adolescents. Antibodies specific for the S1 domain were able to distinguish between SARS-CoV-2 VoCs in infected children. The serologically identified VoC was typically the predominant VoC at the time of infection. Furthermore, our highly sensitive CA identified more silent SARS-CoV-2 infections than a commercial ELISA (12.1% vs. 6.3%, respectively), and provided insights into the infecting VoC. Seroconversion to endemic HCoVs occurred in early childhood, and vaccination or infection with SARS-CoV-2 did not induce HCoV S1 cross-reactive antibodies. In conclusion, the antibody response to the S1 domain of the spike protein of SARS-CoV-2 is highly specific, providing information about the infecting VoC and revealing clinically silent infections.
Collapse
Affiliation(s)
- Daniela Kuthning
- Pediatric Rheumatology, Department of Pediatric and Adolescent Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Dina Raafat
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Silva Holtfreter
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Jana Gramenz
- Pediatric Rheumatology, Department of Pediatric and Adolescent Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Nico Wittmann
- Pediatric Rheumatology, Department of Pediatric and Adolescent Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Barbara M Bröker
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Almut Meyer-Bahlburg
- Pediatric Rheumatology, Department of Pediatric and Adolescent Medicine, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
5
|
Madinga J, Mbala-Kingebeni P, Nkuba-Ndaye A, Baketana-Kinzonzi L, Matungulu-Biyala E, Mutombo-Lupola P, Seghers CA, Smekens T, Ariën KK, Van Damme W, Kalk A, Peeters M, Ahuka-Mundeke S, Muyembe-Tamfum JJ, Vanlerberghe V. COVID-19 seroprevalence cohort survey among health care workers and their household members in Kinshasa, DR Congo, 2020-2022. JOURNAL OF HEALTH, POPULATION, AND NUTRITION 2024; 43:74. [PMID: 38824595 PMCID: PMC11144309 DOI: 10.1186/s41043-024-00536-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 03/17/2024] [Indexed: 06/03/2024]
Abstract
INTRODUCTION Serological surveys offer the most direct measurement to define the immunity status for numerous infectious diseases, such as COVID-19, and can provide valuable insights into understanding transmission patterns. This study describes seroprevalence changes over time in the context of the Democratic Republic of Congo, where COVID-19 case presentation was apparently largely oligo- or asymptomatic, and vaccination coverage remained extremely low. METHODS A cohort of 635 health care workers (HCW) from 5 health zones of Kinshasa and 670 of their household members was interviewed and sampled in 6 rounds between July 2020 and January 2022. At each round, information on risk exposure and a blood sample were collected. Serology was defined as positive when binding antibodies against SARS-CoV-2 spike and nucleocapsid proteins were simultaneously present. RESULTS The SARS-CoV-2 antibody seroprevalence was high at baseline, 17.3% (95% CI 14.4-20.6) and 7.8% (95% CI 5.5-10.8) for HCW and household members, respectively, and fluctuated over time, between 9% and 62.1%. Seropositivity was heterogeneously distributed over the health zones (p < 0.001), ranging from 12.5% (95% CI 6.6-20.8) in N'djili to 33.7% (95% CI 24.6-43.8) in Bandalungwa at baseline for HCW. Seropositivity was associated with increasing rounds adjusted Odds Ratio (aOR) 1.75 (95% CI 1.66-1.85), with increasing age aOR 1.11 (95% CI 1.02-1.20), being a female aOR 1.35 (95% CI 1.10-1.66) and being a HCW aOR 2.38 (95% CI 1.80-3.14). There was no evidence that HCW brought the COVID-19 infection back home, with an aOR of 0.64 (95% CI 0.46-0.91) of seropositivity risk among household members in subsequent surveys. There was seroreversion and seroconversion over time, and HCW had a lower risk of seroreverting than household members (aOR 0.60 (95% CI 0.42-0.86)). CONCLUSION SARS-CoV-2 IgG antibody levels were high and dynamic over time in this African setting with low clinical case rates. The absence of association with health profession or general risk behaviors and with HCW positivity in subsequent rounds in HH members, shows the importance of the time-dependent, and not work-related, force of infection. Cohort seroprevalence estimates in a 'new disease' epidemic seem insufficient to guide policy makers for defining control strategies.
Collapse
Affiliation(s)
- Joule Madinga
- Department of Epidemiology and Global Health, Institut National de Recherche Biomédicale, Faculty of Medicine, University of Kikwit, Kinshasa, Democratic Republic of Congo
| | - Placide Mbala-Kingebeni
- Department of Epidemiology and Global Health, Institut National de Recherche Biomédicale & Faculty of Medicine, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Antoine Nkuba-Ndaye
- Virology Unit, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of Congo
| | - Leonel Baketana-Kinzonzi
- Virology Unit, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of Congo
| | - Elysé Matungulu-Biyala
- Virology Unit, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of Congo
| | - Patrick Mutombo-Lupola
- Department of Epidemiology and Global Health, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of Congo
| | | | - Tom Smekens
- Public Health Department, Institute of Tropical Medicine, Antwerp, Belgium
| | - Kevin K Ariën
- Virology Unit, Institute of Tropical Medicine & Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Wim Van Damme
- Public Health Department, Institute of Tropical Medicine, Antwerp, Belgium
| | - Andreas Kalk
- Deutsche Gesellschaft für Internationale Zusammenarbeit (GIZ), Bonn, Germany
| | - Martine Peeters
- Unit Trans VIHMI, University of Montpellier, IRD/INSERM, Montpellier, France
| | - Steve Ahuka-Mundeke
- Virology Unit, Institut National de Recherche Biomédicale & Faculty of Medicine, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Jean-Jacques Muyembe-Tamfum
- Virology Unit, Institut National de Recherche Biomédicale & Faculty of Medicine, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Veerle Vanlerberghe
- Emerging Infectious Diseases Unit, Public Health Department, Institute of Tropical Medicine, Antwerp, Belgium.
| |
Collapse
|
6
|
Grayo S, Sagno H, Diassy O, Zogbelemou JB, Kondabo SJ, Houndekon M, Dellagi K, Vigan-Womas I, Rourou S, Hamouda WB, Benabdessalem C, Ahmed MB, Tordo N. Snapshot of Anti-SARS-CoV-2 IgG Antibodies in COVID-19 Recovered Patients in Guinea. J Clin Med 2024; 13:2965. [PMID: 38792506 PMCID: PMC11122401 DOI: 10.3390/jcm13102965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/10/2024] [Accepted: 05/11/2024] [Indexed: 05/26/2024] Open
Abstract
Background: Because the regular vaccine campaign started in Guinea one year after the COVID-19 index case, the profile of naturally acquired immunity following primary SARS-CoV-2 infection needs to be deepened. Methods: Blood samples were collected once from 200 patients (90% of African extraction) who were recovered from COVID-19 for at least ~2.4 months (72 days), and their sera were tested for IgG antibodies to SARS-CoV-2 using an in-house ELISA assay against the Receptor Binding Domain (RBD) of the SARS-CoV-2 spike1 protein (RBD/S1-IH kit). Results: Results revealed that 73% of sera (146/200) were positive for IgG to SARS-CoV-2 with an Optical Density (OD) ranging from 0.13 to 1.19 and a median value of 0.56 (IC95: 0.51-0.61). The median OD value at 3 months (1.040) suddenly decreased thereafter and remained stable around OD 0.5 until 15 months post-infection. The OD median value was slightly higher in males compared to females (0.62 vs. 0.49), but the difference was not statistically significant (p-value: 0.073). In contrast, the OD median value was significantly higher among the 60-100 age group (0.87) compared to other groups, with a noteworthy odds ratio compared to the 0-20 age group (OR: 9.69, p-value: 0.044*). Results from the RBD/S1-IH ELISA kit demonstrated superior concordance with the whole spike1 protein ELISA commercial kit compared to a nucleoprotein ELISA commercial kit. Furthermore, anti-spike1 protein ELISAs (whole spike1 and RBD/S1) revealed higher seropositivity rates. Conclusions: These findings underscore the necessity for additional insights into naturally acquired immunity against COVID-19 and emphasize the relevance of specific ELISA kits for accurate seropositivity rates.
Collapse
Affiliation(s)
- Solène Grayo
- Institut Pasteur de Guinée, Conakry BP 4416, Guinea; (H.S.); (N.T.)
| | - Houlou Sagno
- Institut Pasteur de Guinée, Conakry BP 4416, Guinea; (H.S.); (N.T.)
| | - Oumar Diassy
- Agence Nationale de Sécurité Sanitaire, Conakry BP 797, Guinea;
| | | | | | - Marilyn Houndekon
- Centre Médico-Social de L’ambassade de France, Conakry BP 295, Guinea; (J.-B.Z.); (M.H.)
| | - Koussay Dellagi
- Direction Internationale, Institut Pasteur, 75724 Paris, France;
| | | | - Samia Rourou
- Institut Pasteur de Tunis, Tunis BP 74-1002, Tunisia; (S.R.); : (C.B.); (M.B.A.)
| | - Wafa Ben Hamouda
- Institut Pasteur de Tunis, Tunis BP 74-1002, Tunisia; (S.R.); : (C.B.); (M.B.A.)
| | | | - Melika Ben Ahmed
- Institut Pasteur de Tunis, Tunis BP 74-1002, Tunisia; (S.R.); : (C.B.); (M.B.A.)
| | - Noël Tordo
- Institut Pasteur de Guinée, Conakry BP 4416, Guinea; (H.S.); (N.T.)
| |
Collapse
|
7
|
Olvera-Collantes L, Moares N, Fernandez-Cisnal R, Muñoz-Miranda JP, Gonzalez-Garcia P, Gabucio A, Freyre-Carrillo C, Jordan-Chaves JDD, Trujillo-Soto T, Rodriguez-Martinez MP, Martin-Rubio MI, Escuer E, Rodriguez-Iglesias M, Fernandez-Ponce C, Garcia-Cozar F. Development and Validation of a Highly Sensitive Multiplex Immunoassay for SARS-CoV-2 Humoral Response Monitorization: A Study of the Antibody Response in COVID-19 Patients with Different Clinical Profiles during the First and Second Waves in Cadiz, Spain. Microorganisms 2023; 11:2997. [PMID: 38138141 PMCID: PMC10746014 DOI: 10.3390/microorganisms11122997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/04/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
There is still a long way ahead regarding the COVID-19 pandemic, since emerging waves remain a daunting challenge to the healthcare system. For this reason, the development of new preventive tools and therapeutic strategies to deal with the disease have been necessary, among which serological assays have played a key role in the control of COVID-19 outbreaks and vaccine development. Here, we have developed and evaluated an immunoassay capable of simultaneously detecting multiple IgG antibodies against different SARS-CoV-2 antigens through the use of Bio-PlexTM technology. Additionally, we have analyzed the antibody response in COVID-19 patients with different clinical profiles in Cadiz, Spain. The multiplex immunoassay presented is a high-throughput and robust immune response monitoring tool capable of concurrently detecting anti-S1, anti-NC and anti-RBD IgG antibodies in serum with a very high sensitivity (94.34-97.96%) and specificity (91.84-100%). Therefore, the immunoassay proposed herein may be a useful monitoring tool for individual humoral immunity against SARS-CoV-2, as well as for epidemiological surveillance. In addition, we show the values of antibodies against multiple SARS-CoV-2 antigens and their correlation with the different clinical profiles of unvaccinated COVID-19 patients in Cadiz, Spain, during the first and second waves of the pandemic.
Collapse
Affiliation(s)
- Lucia Olvera-Collantes
- Department of Biomedicine, Biotechnology and Public Health, School of Medicine, University of Cadiz, 11003 Cadiz, Spain (A.G.); (M.R.-I.)
- Institute of Biomedical Research Cadiz (INIBICA), 11009 Cadiz, Spain
| | - Noelia Moares
- Department of Biomedicine, Biotechnology and Public Health, School of Medicine, University of Cadiz, 11003 Cadiz, Spain (A.G.); (M.R.-I.)
| | - Ricardo Fernandez-Cisnal
- Department of Biomedicine, Biotechnology and Public Health, School of Medicine, University of Cadiz, 11003 Cadiz, Spain (A.G.); (M.R.-I.)
- Institute of Biomedical Research Cadiz (INIBICA), 11009 Cadiz, Spain
| | - Juan P. Muñoz-Miranda
- Department of Biomedicine, Biotechnology and Public Health, School of Medicine, University of Cadiz, 11003 Cadiz, Spain (A.G.); (M.R.-I.)
- Institute of Biomedical Research Cadiz (INIBICA), 11009 Cadiz, Spain
| | | | - Antonio Gabucio
- Department of Biomedicine, Biotechnology and Public Health, School of Medicine, University of Cadiz, 11003 Cadiz, Spain (A.G.); (M.R.-I.)
| | | | | | | | | | | | - Eva Escuer
- Jerez University Hospital, 11407 Jerez de la Frontera, Spain;
| | - Manuel Rodriguez-Iglesias
- Department of Biomedicine, Biotechnology and Public Health, School of Medicine, University of Cadiz, 11003 Cadiz, Spain (A.G.); (M.R.-I.)
- Institute of Biomedical Research Cadiz (INIBICA), 11009 Cadiz, Spain
- Microbiology Service, Puerta del Mar University Hospital, 11009 Cadiz, Spain
| | - Cecilia Fernandez-Ponce
- Department of Biomedicine, Biotechnology and Public Health, School of Medicine, University of Cadiz, 11003 Cadiz, Spain (A.G.); (M.R.-I.)
- Institute of Biomedical Research Cadiz (INIBICA), 11009 Cadiz, Spain
| | - Francisco Garcia-Cozar
- Department of Biomedicine, Biotechnology and Public Health, School of Medicine, University of Cadiz, 11003 Cadiz, Spain (A.G.); (M.R.-I.)
- Institute of Biomedical Research Cadiz (INIBICA), 11009 Cadiz, Spain
| |
Collapse
|
8
|
Stern D, Meyer TC, Treindl F, Mages HW, Krüger M, Skiba M, Krüger JP, Zobel CM, Schreiner M, Grossegesse M, Rinner T, Peine C, Stoliaroff-Pépin A, Harder T, Hofmann N, Michel J, Nitsche A, Stahlberg S, Kneuer A, Sandoni A, Kubisch U, Schlaud M, Mankertz A, Schwarz T, Corman VM, Müller MA, Drosten C, de la Rosa K, Schaade L, Dorner MB, Dorner BG. A bead-based multiplex assay covering all coronaviruses pathogenic for humans for sensitive and specific surveillance of SARS-CoV-2 humoral immunity. Sci Rep 2023; 13:21846. [PMID: 38071261 PMCID: PMC10710470 DOI: 10.1038/s41598-023-48581-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
Serological assays measuring antibodies against SARS-CoV-2 are key to describe the epidemiology, pathobiology or induction of immunity after infection or vaccination. Of those, multiplex assays targeting multiple antigens are especially helpful as closely related coronaviruses or other antigens can be analysed simultaneously from small sample volumes, hereby shedding light on patterns in the immune response that would otherwise remain undetected. We established a bead-based 17-plex assay detecting antibodies targeting antigens from all coronaviruses pathogenic for humans: SARS-CoV-2, SARS-CoV, MERS-CoV, HCoV strains 229E, OC43, HKU1, and NL63. The assay was validated against five commercial serological immunoassays, a commercial surrogate virus neutralisation test, and a virus neutralisation assay, all targeting SARS-CoV-2. It was found to be highly versatile as shown by antibody detection from both serum and dried blot spots and as shown in three case studies. First, we followed seroconversion for all four endemic HCoV strains and SARS-CoV-2 in an outbreak study in day-care centres for children. Second, we were able to link a more severe clinical course to a stronger IgG response with this 17-plex-assay, which was IgG1 and IgG3 dominated. Finally, our assay was able to discriminate recent from previous SARS-CoV-2 infections by calculating the IgG/IgM ratio on the N antigen targeting antibodies. In conclusion, due to the comprehensive method comparison, thorough validation, and the proven versatility, our multiplex assay is a valuable tool for studies on coronavirus serology.
Collapse
Affiliation(s)
- Daniel Stern
- Biological Toxins (ZBS 3), Centre for Biological Threats and Special Pathogens, Robert Koch Institute, 13353, Berlin, Germany.
| | - Tanja C Meyer
- Biological Toxins (ZBS 3), Centre for Biological Threats and Special Pathogens, Robert Koch Institute, 13353, Berlin, Germany
| | - Fridolin Treindl
- Biological Toxins (ZBS 3), Centre for Biological Threats and Special Pathogens, Robert Koch Institute, 13353, Berlin, Germany
| | - Hans Werner Mages
- Biological Toxins (ZBS 3), Centre for Biological Threats and Special Pathogens, Robert Koch Institute, 13353, Berlin, Germany
| | - Maren Krüger
- Biological Toxins (ZBS 3), Centre for Biological Threats and Special Pathogens, Robert Koch Institute, 13353, Berlin, Germany
| | - Martin Skiba
- Biological Toxins (ZBS 3), Centre for Biological Threats and Special Pathogens, Robert Koch Institute, 13353, Berlin, Germany
| | - Jan Philipp Krüger
- Department of Microbiology and Hospital Hygiene, Bundeswehr Hospital Berlin, Berlin, Germany
| | - Christian M Zobel
- Department of Internal Medicine, Bundeswehr Hospital Berlin, Berlin, Germany
| | | | - Marica Grossegesse
- Highly Pathogenic Viruses (ZBS 1), Centre for Biological Threats and Special Pathogens, Robert Koch Institute, 13353, Berlin, Germany
| | - Thomas Rinner
- Highly Pathogenic Viruses (ZBS 1), Centre for Biological Threats and Special Pathogens, Robert Koch Institute, 13353, Berlin, Germany
| | - Caroline Peine
- Immunization Unit (FG 33), Department for Infectious Disease Epidemiology, Robert Koch Institute, 13353, Berlin, Germany
| | - Anna Stoliaroff-Pépin
- Immunization Unit (FG 33), Department for Infectious Disease Epidemiology, Robert Koch Institute, 13353, Berlin, Germany
| | - Thomas Harder
- Immunization Unit (FG 33), Department for Infectious Disease Epidemiology, Robert Koch Institute, 13353, Berlin, Germany
| | - Natalie Hofmann
- Highly Pathogenic Viruses (ZBS 1), Centre for Biological Threats and Special Pathogens, Robert Koch Institute, 13353, Berlin, Germany
| | - Janine Michel
- Highly Pathogenic Viruses (ZBS 1), Centre for Biological Threats and Special Pathogens, Robert Koch Institute, 13353, Berlin, Germany
| | - Andreas Nitsche
- Highly Pathogenic Viruses (ZBS 1), Centre for Biological Threats and Special Pathogens, Robert Koch Institute, 13353, Berlin, Germany
| | - Silke Stahlberg
- Central Epidemiological Laboratory (FG 22), Department of Epidemiology and Health Monitoring, Robert Koch Institute, 12101, Berlin, Germany
| | - Antje Kneuer
- Central Epidemiological Laboratory (FG 22), Department of Epidemiology and Health Monitoring, Robert Koch Institute, 12101, Berlin, Germany
| | - Anna Sandoni
- Central Epidemiological Laboratory (FG 22), Department of Epidemiology and Health Monitoring, Robert Koch Institute, 12101, Berlin, Germany
| | - Ulrike Kubisch
- Central Epidemiological Laboratory (FG 22), Department of Epidemiology and Health Monitoring, Robert Koch Institute, 12101, Berlin, Germany
| | - Martin Schlaud
- Central Epidemiological Laboratory (FG 22), Department of Epidemiology and Health Monitoring, Robert Koch Institute, 12101, Berlin, Germany
| | - Annette Mankertz
- Measles, Mumps, Rubella, and Viruses Affecting Immunocompromised Patients (FG 12), Robert Koch Institute, 13353, Berlin, Germany
| | - Tatjana Schwarz
- Institute of Virology, Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany
| | - Victor M Corman
- Institute of Virology, Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany
- Corporate Member, Freie Universität Berlin, 10117, Berlin, Germany
- Corporate Member, Humboldt-Universität zu Berlin, 14195, Berlin, Germany
| | - Marcel A Müller
- Institute of Virology, Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany
| | - Christian Drosten
- Institute of Virology, Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany
| | - Kathrin de la Rosa
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125, Berlin, Germany
- Berlin Institute of Health (BIH), Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany
| | - Lars Schaade
- Centre for Biological Threats and Special Pathogens, Robert Koch Institute, 13353, Berlin, Germany
| | - Martin B Dorner
- Biological Toxins (ZBS 3), Centre for Biological Threats and Special Pathogens, Robert Koch Institute, 13353, Berlin, Germany
| | - Brigitte G Dorner
- Biological Toxins (ZBS 3), Centre for Biological Threats and Special Pathogens, Robert Koch Institute, 13353, Berlin, Germany.
| |
Collapse
|
9
|
Kaufmann SHE. Vaccine development against tuberculosis before and after Covid-19. Front Immunol 2023; 14:1273938. [PMID: 38035095 PMCID: PMC10684952 DOI: 10.3389/fimmu.2023.1273938] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 10/16/2023] [Indexed: 12/02/2023] Open
Abstract
Coronavirus disease (Covid-19) has not only shaped awareness of the impact of infectious diseases on global health. It has also provided instructive lessons for better prevention strategies against new and current infectious diseases of major importance. Tuberculosis (TB) is a major current health threat caused by Mycobacterium tuberculosis (Mtb) which has claimed more lives than any other pathogen over the last few centuries. Hence, better intervention measures, notably novel vaccines, are urgently needed to accomplish the goal of the World Health Organization to end TB by 2030. This article describes how the research and development of TB vaccines can benefit from recent developments in the Covid-19 vaccine pipeline from research to clinical development and outlines how the field of TB research can pursue its own approaches. It begins with a brief discussion of major vaccine platforms in general terms followed by a short description of the most widely applied Covid-19 vaccines. Next, different vaccination regimes and particular hurdles for TB vaccine research and development are described. This specifically considers the complex immune mechanisms underlying protection and pathology in TB which involve innate as well as acquired immune mechanisms and strongly depend on fine tuning the response. A brief description of the TB vaccine candidates that have entered clinical trials follows. Finally, it discusses how experiences from Covid-19 vaccine research, development, and rollout can and have been applied to the TB vaccine pipeline, emphasizing similarities and dissimilarities.
Collapse
Affiliation(s)
- Stefan H. E. Kaufmann
- Max Planck Institute for Infection Biology, Berlin, Germany
- Systems Immunology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Hagler Institute for Advanced Study, Texas A&M University, College Station, TX, United States
| |
Collapse
|
10
|
Angelot RAKOTOMALALAF, Dauphin RANDRIARIMANANAS, Nathanaella RANDRIAMANANJARAH, Xavier BABINF, Haja RANDRIANTSARA, Tahinamandranto RASAMOELINA, Doll RAKOTODA, Hervé SAMISONL, Ahidjo AYOUBA, Eric NERRIENET. IMPACT OF COVID-19 ON PEOPLE LIVING WITH HIV IN MADAGASCAR: A SARS-COV2 SERO-PREVALENCE SURVEY. Afr J Infect Dis 2023; 18:1-4. [PMID: 38058416 PMCID: PMC10696651 DOI: 10.21010/ajidv18i1.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 08/18/2023] [Accepted: 08/18/2023] [Indexed: 12/08/2023] Open
Abstract
Background In Madagascar, no study has reported the impact of COVID-19 on people living with HIV (PLHIV). The present work aimed to analyze the seroprevalence of SARS-CoV-2 in Malagasy PLHIV before and during the three waves of COVID-19 pandemic.This is a retrospective study. Materials and Methods We conducted a retrospective serological survey in PLHIV followed up for HIV viral load (VL) monitoring at the Centre d'Infectiologie Charles Mérieux Madagascar (CICM) between June 2019 and April 2022. The presence of IgM and/or IgG antibodies against SARS-CoV-2 nucleoprotein was detected using a rapid diagnostic test (COVID-PRESTO®). Results A seroprevalence of 2.5% was found in the 877 patients tested before March 2020, compared to 25.4% (512/2,011) between March 2020 and April 2022. This seroprevalence was 21.7%, 22.3% and 60.1% after the first, second and third waves of COVID-19, respectively. We observed a marginally significant difference (p = 0.043) in SARS-CoV-2 seroprevalence between patients on highly active antiretroviral therapy (HAART) (27.5%) and those who were not (23.7%). No statistically significant difference was observed between PLHIV with undetectable HIV VL (27.4%) and the different detectable VL categories (p>0.05). Conclusions Our data show the presence of antibodies to SARS-CoV-2 among PLHIV as early as December 2019 in Madagascar. At least 25.4% (512/2,011) of Malagasy PLHIV have been in contact with SARS-CoV-2 since March 2020. There is no significant relation between HIV-1 VL and SARS-CoV-2 seroprevalence. Additional studies with more robust assays in the general population are needed for a detailed knowledge of SARS-CoV-2 impact in Madagascar.
Collapse
Affiliation(s)
- RAKOTOMALALA Fetra Angelot
- Centre d’Infectiologie Charles Mérieux, Madagascar
- Recherches Translationnelles sur le VIH et les Maladies Infectieuses (TransVIHMI), Université de Montpellier, IRD, INSERM, France
| | | | | | | | | | | | | | | | - AYOUBA Ahidjo
- Recherches Translationnelles sur le VIH et les Maladies Infectieuses (TransVIHMI), Université de Montpellier, IRD, INSERM, France
| | | |
Collapse
|
11
|
Tapela K, Opurum PC, Nuokpem FY, Tetteh B, Siaw GK, Humbert MV, Tawiah-Eshun S, Barakisu AI, Asiedu K, Arhin SK, Manu AA, Appiedu-Addo SNA, Obbeng L, Quansah D, Languon S, Anyigba C, Dosoo D, Edu NKO, Oduro-Mensah D, Ampofo W, Tagoe E, Quaye O, Donkor IO, Akorli J, Aniweh Y, Christodoulides M, Mutungi J, Bediako Y, Rayner JC, Awandare GA, McCormick CJ, Quashie PK. Development of an Affordable ELISA Targeting the SARS-CoV-2 Nucleocapsid and Its Application to Samples from the Ongoing COVID-19 Epidemic in Ghana. Mol Diagn Ther 2023; 27:583-592. [PMID: 37462793 PMCID: PMC10435612 DOI: 10.1007/s40291-023-00655-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2023] [Indexed: 08/18/2023]
Abstract
INTRODUCTION The true nature of the population spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in populations is often not fully known as most cases, particularly in Africa, are asymptomatic. Finding the true magnitude of SARS-CoV-2 spread is crucial to provide actionable data about the epidemiological progress of the disease for researchers and policymakers. This study developed and optimized an antibody enzyme-linked immunosorbent assay (ELISA) using recombinant nucleocapsid antigen expressed in-house using a simple bacterial expression system. METHODS Nucleocapsid protein from SARS-CoV-2 was expressed and purified from Escherichia coli. Plasma samples used for the assay development were obtained from Ghanaian SARS-CoV-2 seropositive individuals during the pandemic, while seronegative controls were plasma samples collected from blood donors before the coronavirus disease 2019 (COVID-19) pandemic. Another set of seronegative controls was collected during the COVID-19 pandemic. Antibody detection and levels within the samples were validated using commercial kits and Luminex. Analyses were performed using GraphPad Prism, and the sensitivity, specificity and background cut-off were calculated. RESULTS AND DISCUSSION This low-cost ELISA (£0.96/test) assay has a high prediction of 98.9%, and sensitivity and specificity of 97% and 99%, respectively. The assay was subsequently used to screen plasma from SARS-CoV-2 RT-PCR-positive Ghanaians. The assay showed no significant difference in nucleocapsid antibody levels between symptomatic and asymptomatic, with an increase of the levels over time. This is in line with our previous publication. CONCLUSION This study developed a low-cost and transferable assay that enables highly sensitive and specific detection of human anti-SARS-CoV-2 IgG antibodies. This assay can be modified to include additional antigens and used for continuous monitoring of sero-exposure to SARS-CoV-2 in West Africa.
Collapse
Affiliation(s)
- Kesego Tapela
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
- Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana
| | - Precious C Opurum
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
| | - Franklin Y Nuokpem
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
- Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana
| | - Becky Tetteh
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
- Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana
| | - Godfred K Siaw
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
| | - Maria V Humbert
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
| | - Sylvia Tawiah-Eshun
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
| | - Anna Ibrahim Barakisu
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
| | - Kwame Asiedu
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
| | - Samuel Kojo Arhin
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
- Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana
| | - Aaron A Manu
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
- Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana
| | - Sekyibea N A Appiedu-Addo
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
| | - Louisa Obbeng
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
| | - Darius Quansah
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
- Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana
- Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Accra, Ghana
| | - Sylvester Languon
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
| | - Claudia Anyigba
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
- Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana
| | - Daniel Dosoo
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
| | - Nelson K O Edu
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
| | - Daniel Oduro-Mensah
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
- Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana
| | - William Ampofo
- Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Accra, Ghana
| | - Emmanuel Tagoe
- Department of Medical Laboratory Sciences, School of Biomedical and Allied Health, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Osbourne Quaye
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
- Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana
| | - Irene Owusu Donkor
- Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Accra, Ghana
| | - Jewelna Akorli
- Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Accra, Ghana
| | - Yaw Aniweh
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
- The Francis Crick Institute, 1 Midland Rd, London, NW1 1AT, UK
| | - Myron Christodoulides
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
| | - Joe Mutungi
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
| | - Yaw Bediako
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
- The Francis Crick Institute, 1 Midland Rd, London, NW1 1AT, UK
- Yemaachi Biotech Inc., 222 Swaniker St, Accra, Ghana
| | - Julian C Rayner
- Cambridge Institute for Medical Research, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Gordon A Awandare
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
- Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana
| | - Christopher J McCormick
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
| | - Peter Kojo Quashie
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana.
- The Francis Crick Institute, 1 Midland Rd, London, NW1 1AT, UK.
| |
Collapse
|
12
|
Kinganda-Lusamaki E, Baketana LK, Ndomba-Mukanya E, Bouillin J, Thaurignac G, Aziza AA, Luakanda-Ndelemo G, Nuñez NF, Kalonji-Mukendi T, Pukuta ES, Nkuba-Ndaye A, Lofiko EL, Kibungu EM, Lushima RS, Ayouba A, Mbala-Kingebeni P, Muyembe-Tamfum JJ, Delaporte E, Peeters M, Ahuka-Mundeke S. Use of Mpox Multiplex Serology in the Identification of Cases and Outbreak Investigations in the Democratic Republic of the Congo (DRC). Pathogens 2023; 12:916. [PMID: 37513764 PMCID: PMC10385798 DOI: 10.3390/pathogens12070916] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 06/29/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
Human Mpox cases are increasingly reported in Africa, with the highest burden in the Democratic Republic of Congo (DRC). While case reporting on a clinical basis can overestimate infection rates, laboratory confirmation by PCR can underestimate them, especially on suboptimal samples like blood, commonly used in DRC. Here we used a Luminex-based assay to evaluate whether antibody testing can be complementary to confirm cases and to identify human transmission chains during outbreak investigations. We used left-over blood samples from 463 patients, collected during 174 outbreaks between 2013 and 2022, with corresponding Mpox and VZV PCR results. In total, 157 (33.9%) samples were orthopox-PCR positive and classified as Mpox+; 124 (26.8%) had antibodies to at least one of the three Mpox peptides. The proportion of antibody positive samples was significantly higher in Mpox positive samples (36.9%) versus negative (21.6%) (p < 0.001). By combining PCR and serology, 66 additional patients were identified, leading to an Mpox infection rate of 48.2% (223/463) versus 33.9% when only PCR positivity is considered. Mpox infections were as such identified in 14 additional health zones and 23 additional outbreaks (111/174 (63.8%) versus 88/174 (50.6%)). Our findings highlight the urgent need of rapid on-site diagnostics to circumvent Mpox spread.
Collapse
Affiliation(s)
- Eddy Kinganda-Lusamaki
- TransVIHMI, University of Montpellier (UM), French Institute of Health and Medical Research (INSERM), French National Research Institute for Sustainable Development (IRD), 34394 Montpellier, France
- Institut National de Recherche Biomédicale (INRB), Kinshasa P.O. Box 1197, Democratic Republic of the Congo
- Service de Microbiologie, Département de Biologie Médicale, Cliniques Universitaires de Kinshasa (CUK), Université de Kinshasa (UNIKIN), Kinshasa P.O. Box 127, Democratic Republic of the Congo
| | - Lionel Kinzonzi Baketana
- Institut National de Recherche Biomédicale (INRB), Kinshasa P.O. Box 1197, Democratic Republic of the Congo
| | - Etienne Ndomba-Mukanya
- Institut National de Recherche Biomédicale (INRB), Kinshasa P.O. Box 1197, Democratic Republic of the Congo
| | - Julie Bouillin
- TransVIHMI, University of Montpellier (UM), French Institute of Health and Medical Research (INSERM), French National Research Institute for Sustainable Development (IRD), 34394 Montpellier, France
| | - Guillaume Thaurignac
- TransVIHMI, University of Montpellier (UM), French Institute of Health and Medical Research (INSERM), French National Research Institute for Sustainable Development (IRD), 34394 Montpellier, France
| | - Adrienne Amuri Aziza
- Institut National de Recherche Biomédicale (INRB), Kinshasa P.O. Box 1197, Democratic Republic of the Congo
| | - Gradi Luakanda-Ndelemo
- Institut National de Recherche Biomédicale (INRB), Kinshasa P.O. Box 1197, Democratic Republic of the Congo
| | - Nicolas Fernandez Nuñez
- TransVIHMI, University of Montpellier (UM), French Institute of Health and Medical Research (INSERM), French National Research Institute for Sustainable Development (IRD), 34394 Montpellier, France
| | - Thierry Kalonji-Mukendi
- Programme National de Lutte Contre le Monkeypox et les Fièvres Hémorragiques Virales, Ministère de la Santé (PNLMPX-FHV), Kinshasa P.O. Box 1197, Democratic Republic of the Congo
| | - Elisabeth Simbu Pukuta
- Institut National de Recherche Biomédicale (INRB), Kinshasa P.O. Box 1197, Democratic Republic of the Congo
| | - Antoine Nkuba-Ndaye
- TransVIHMI, University of Montpellier (UM), French Institute of Health and Medical Research (INSERM), French National Research Institute for Sustainable Development (IRD), 34394 Montpellier, France
- Institut National de Recherche Biomédicale (INRB), Kinshasa P.O. Box 1197, Democratic Republic of the Congo
- Service de Microbiologie, Département de Biologie Médicale, Cliniques Universitaires de Kinshasa (CUK), Université de Kinshasa (UNIKIN), Kinshasa P.O. Box 127, Democratic Republic of the Congo
| | - Emmanuel Lokilo Lofiko
- Institut National de Recherche Biomédicale (INRB), Kinshasa P.O. Box 1197, Democratic Republic of the Congo
| | - Emile Malembi Kibungu
- Programme National de Lutte Contre le Monkeypox et les Fièvres Hémorragiques Virales, Ministère de la Santé (PNLMPX-FHV), Kinshasa P.O. Box 1197, Democratic Republic of the Congo
| | - Robert Shongo Lushima
- Programme National de Lutte Contre le Monkeypox et les Fièvres Hémorragiques Virales, Ministère de la Santé (PNLMPX-FHV), Kinshasa P.O. Box 1197, Democratic Republic of the Congo
| | - Ahidjo Ayouba
- TransVIHMI, University of Montpellier (UM), French Institute of Health and Medical Research (INSERM), French National Research Institute for Sustainable Development (IRD), 34394 Montpellier, France
| | - Placide Mbala-Kingebeni
- Institut National de Recherche Biomédicale (INRB), Kinshasa P.O. Box 1197, Democratic Republic of the Congo
- Service de Microbiologie, Département de Biologie Médicale, Cliniques Universitaires de Kinshasa (CUK), Université de Kinshasa (UNIKIN), Kinshasa P.O. Box 127, Democratic Republic of the Congo
| | - Jean-Jacques Muyembe-Tamfum
- Institut National de Recherche Biomédicale (INRB), Kinshasa P.O. Box 1197, Democratic Republic of the Congo
- Service de Microbiologie, Département de Biologie Médicale, Cliniques Universitaires de Kinshasa (CUK), Université de Kinshasa (UNIKIN), Kinshasa P.O. Box 127, Democratic Republic of the Congo
| | - Eric Delaporte
- TransVIHMI, University of Montpellier (UM), French Institute of Health and Medical Research (INSERM), French National Research Institute for Sustainable Development (IRD), 34394 Montpellier, France
| | - Martine Peeters
- TransVIHMI, University of Montpellier (UM), French Institute of Health and Medical Research (INSERM), French National Research Institute for Sustainable Development (IRD), 34394 Montpellier, France
| | - Steve Ahuka-Mundeke
- Institut National de Recherche Biomédicale (INRB), Kinshasa P.O. Box 1197, Democratic Republic of the Congo
- Service de Microbiologie, Département de Biologie Médicale, Cliniques Universitaires de Kinshasa (CUK), Université de Kinshasa (UNIKIN), Kinshasa P.O. Box 127, Democratic Republic of the Congo
| |
Collapse
|
13
|
Houngbégnon P, Nouatin O, Yadouléton A, Hounkpatin B, Fievet N, Atindégla E, Dechavanne S, Guichet E, Ayouba A, Pelloquin R, Maman D, Thaurignac G, Peeters M, Aviansou A, Sourakafou S, Delaporte E, Massougbodji A, Cottrell G. Interest of seroprevalence surveys for the epidemiological surveillance of the SARS-CoV-2 pandemic in African populations: Insights from the ARIACOV project in Benin. Trop Med Int Health 2023. [PMID: 37243412 DOI: 10.1111/tmi.13895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
BACKGROUND Many SARS-CoV-2 seroprevalence surveys since the end of 2020 have disqualified the first misconception that Africa had been spared by the pandemic. Through the analysis of three SARS-CoV-2 seroprevalence surveys carried out in Benin as part of the ARIACOV project, we argue that the integration of epidemiological serosurveillance of the SARS-CoV-2 infection in the national surveillance packages would be of great use to refine the understanding of the COVID-19 pandemic in Africa. METHODS We carried out three repeated cross-sectional surveys in Benin: two in Cotonou, the economic capital in March and May 2021, and one in Natitingou, a semi-rural city in the north of the country in August 2021. Total and weighted-by-age-group seroprevalences were estimated and the risk factors for SARS-CoV-2 infection were assessed by multivariate logistic regression. RESULTS In Cotonou, a slight increase in overall age-standardised SARS-CoV-2 seroprevalence from 29.77% (95% CI: 23.12%-37.41%) at the first survey to 34.86% (95% CI: 31.57%-38.30%) at the second survey was observed. In Natitingou, the globally adjusted seroprevalence was 33.34% (95% CI: 27.75%-39.44%). A trend of high risk for SARS-CoV 2 seropositivity was observed in adults over 40 versus the young (less than 18 years old) during the first survey in Cotonou but no longer in the second survey. CONCLUSIONS Our results show that, however, rapid organisation of preventive measures aimed at breaking the chains of transmission, they were ultimately unable to prevent a wide spread of the virus in the population. Routine serological surveillance on strategic sentinel sites and/or populations could constitute a cost-effective compromise to better anticipate the onset of new waves and define public health strategies.
Collapse
Affiliation(s)
| | - Odilon Nouatin
- Institut de Recherche Clinique du Bénin, Abomey-Calavi, Benin
| | - Anges Yadouléton
- Laboratoire des Fièvres Hémorragiques Virales et des Arbovirus du Bénin, Ministère de la Santé, Cotonou, Benin
- Ecole Normale Supérieure de Natitingou, Université Nationale des Sciences Technologies, Ingénierie et Mathématiques, Abomey-Calavi, Benin
| | | | - Nadine Fievet
- Université de Paris Cité, Institut de Recherche pour le Développement, MERIT, Paris, France
| | - Eloïc Atindégla
- Institut de Recherche Clinique du Bénin, Abomey-Calavi, Benin
| | - Sébastien Dechavanne
- Université de Paris Cité, Institut de Recherche pour le Développement, MERIT, Paris, France
| | - Emilande Guichet
- Recherches Translationnelles sur le VIH et les Maladies Infectieuses, Institut de Recherche pour le Développement, University of Montpellier, Montpellier, France
- Institut National de la Santé et de la Recherche Médicale, Montpellier, France
| | - Ahidjo Ayouba
- Recherches Translationnelles sur le VIH et les Maladies Infectieuses, Institut de Recherche pour le Développement, University of Montpellier, Montpellier, France
- Institut National de la Santé et de la Recherche Médicale, Montpellier, France
| | - Raphaël Pelloquin
- Recherches Translationnelles sur le VIH et les Maladies Infectieuses, Institut de Recherche pour le Développement, University of Montpellier, Montpellier, France
- Institut National de la Santé et de la Recherche Médicale, Montpellier, France
| | | | - Guillaume Thaurignac
- Recherches Translationnelles sur le VIH et les Maladies Infectieuses, Institut de Recherche pour le Développement, University of Montpellier, Montpellier, France
- Institut National de la Santé et de la Recherche Médicale, Montpellier, France
| | - Martine Peeters
- Recherches Translationnelles sur le VIH et les Maladies Infectieuses, Institut de Recherche pour le Développement, University of Montpellier, Montpellier, France
- Institut National de la Santé et de la Recherche Médicale, Montpellier, France
| | | | | | - Eric Delaporte
- Recherches Translationnelles sur le VIH et les Maladies Infectieuses, Institut de Recherche pour le Développement, University of Montpellier, Montpellier, France
- Institut National de la Santé et de la Recherche Médicale, Montpellier, France
| | | | - Gilles Cottrell
- Université de Paris Cité, Institut de Recherche pour le Développement, MERIT, Paris, France
| |
Collapse
|
14
|
Diallo MSK, Amougou-Atsama M, Ayouba A, Kpamou C, Mimbe Taze ED, Thaurignac G, Diallo H, Lamare NB, Bouillin J, Soumah AK, Noah SA, Guichet E, Keita AK, Varloteaux M, Peeters M, Bissek ACZK, Toure A, Delaporte E, Kouanfack C. Large Diffusion of Severe Acute Respiratory Syndrome Coronavirus 2 After the Successive Epidemiological Waves, Including Omicron, in Guinea and Cameroon: Implications for Vaccine Strategies. Open Forum Infect Dis 2023; 10:ofad216. [PMID: 37152188 PMCID: PMC10157752 DOI: 10.1093/ofid/ofad216] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 04/18/2023] [Indexed: 05/09/2023] Open
Abstract
Background We aimed to estimate the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) seroprevalence among the general population in Conakry, Guinea and Yaounde, Cameroon after the coronavirus disease 2019 Omicron wave. Methods We conducted population-based, age-stratified seroprevalence surveys in Conakry and Yaounde (May and June 2022). We collected demographic and epidemiologic information and dried blood spot samples that were tested for SARS-CoV-2 immunoglobulin G (IgG) antibodies using recombinant nucleocapsid and spike proteins with Luminex technology. Results Samples were obtained from 1386 and 1425 participants in Guinea and Cameroon, respectively. The overall age-standardized SARS-CoV-2 IgG seroprevalence against spike and nucleocapsid proteins was 71.57% (95% confidence interval [CI], 67.48%-75.33%) in Guinea and 74.71% (95% CI, 71.99%-77.25%) in Cameroon. Seroprevalence increased significantly with age categories. Female participants were more likely than male participants to be seropositive. The seroprevalence in unvaccinated participants was 69.6% (95% CI, 65.5%-73.41%) in Guinea and 74.8% (95% CI, 72.04%-77.38%) in Cameroon. In multivariate analysis, only age, sex, and education were independently associated with seropositivity. Conclusions These findings show a high community transmission after the different epidemiological waves including Omicron, especially among people aged >40 years. In addition, our results suggest that the spread of SARS-CoV-2 has been underestimated as a significant proportion of the population has already contracted the virus and that vaccine strategies should focus on vulnerable populations.
Collapse
Affiliation(s)
- Mamadou Saliou Kalifa Diallo
- Correspondence: Mamadou Saliou Kalifa Diallo, MD, TransVIHMI, University of Montpellier, Inserm, Institut de Recherche pour le Developpement, 911 Avenue Agropolis, 34934 Montpellier, France (); Eric Delaporte, MD, TransVIHMI, University of Montpellier, Inserm, Institut de Recherche pour le Developpement, Montpellier, France ()
| | - Marie Amougou-Atsama
- Centre de Recherche sur les Maladies Émergentes et Re-Emergentes, Yaounde, Cameroon
| | - Ahidjo Ayouba
- TransVIHMI, University of Montpellier, Inserm, Institut de Recherche pour le Developpement, Montpellier, France
| | - Cece Kpamou
- Centre de Recherche et de Formation en Infectiologie de Guinée, Université Gamal Abdel Nasser de Conakry, Conakry, Guinea
| | - Eric Donald Mimbe Taze
- French National Agency for Research on AIDS and Infectious Diseases, Cameroon Site, Central Hospital of Yaounde, Yaounde, Cameroon
| | - Guillaume Thaurignac
- TransVIHMI, University of Montpellier, Inserm, Institut de Recherche pour le Developpement, Montpellier, France
| | - Haby Diallo
- Centre de Recherche et de Formation en Infectiologie de Guinée, Université Gamal Abdel Nasser de Conakry, Conakry, Guinea
| | - Nadine Boutgam Lamare
- Faculty of Medicine and Pharmaceutical Sciences, University of Dschang, Dschang, Cameroon
| | - Julie Bouillin
- TransVIHMI, University of Montpellier, Inserm, Institut de Recherche pour le Developpement, Montpellier, France
| | - Abdoul Karim Soumah
- Centre de Recherche et de Formation en Infectiologie de Guinée, Université Gamal Abdel Nasser de Conakry, Conakry, Guinea
| | - Sébastien Awono Noah
- French National Agency for Research on AIDS and Infectious Diseases, Cameroon Site, Central Hospital of Yaounde, Yaounde, Cameroon
| | - Emilande Guichet
- TransVIHMI, University of Montpellier, Inserm, Institut de Recherche pour le Developpement, Montpellier, France
| | - Alpha Kabinet Keita
- Centre de Recherche et de Formation en Infectiologie de Guinée, Université Gamal Abdel Nasser de Conakry, Conakry, Guinea
- TransVIHMI, University of Montpellier, Inserm, Institut de Recherche pour le Developpement, Montpellier, France
| | - Marie Varloteaux
- French National Agency for Research on AIDS and Infectious Diseases, Cameroon Site, Central Hospital of Yaounde, Yaounde, Cameroon
| | - Martine Peeters
- TransVIHMI, University of Montpellier, Inserm, Institut de Recherche pour le Developpement, Montpellier, France
| | - Anne-Cécile Zoung-Kanyi Bissek
- Centre de Recherche sur les Maladies Émergentes et Re-Emergentes, Yaounde, Cameroon
- Division of Operational Research in Health, Ministry of Public Health of Cameroon, Yaounde, Cameroon
| | | | - Eric Delaporte
- Correspondence: Mamadou Saliou Kalifa Diallo, MD, TransVIHMI, University of Montpellier, Inserm, Institut de Recherche pour le Developpement, 911 Avenue Agropolis, 34934 Montpellier, France (); Eric Delaporte, MD, TransVIHMI, University of Montpellier, Inserm, Institut de Recherche pour le Developpement, Montpellier, France ()
| | | |
Collapse
|
15
|
Kumar D, Roy SS, Rastogi R, Arora K, Undale A, Gupta R, Arora NM, Kundu PK. VLP-ELISA for the Detection of IgG Antibodies against Spike, Envelope, and Membrane Antigens of SARS-CoV-2 in Indian Population. Vaccines (Basel) 2023; 11:vaccines11040743. [PMID: 37112655 PMCID: PMC10145915 DOI: 10.3390/vaccines11040743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/20/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
Background: Serological methods to conduct epidemiological survey are often directed only against the spike protein. To overcome this limitation, we have designed PRAK-03202, a virus-like particle (VLP), by inserting three antigens (Spike, envelope and membrane) of SARS-CoV-2 into a highly characterized S. cerevisiae-based D-Crypt™ platform. Methods: Dot blot analysis was performed to confirm the presence of S, E, and M proteins in PRAK-03202. The number of particles in PRAK-03202 was measured using nanoparticle tracking analysis (NTA). The sensitivity of VLP-ELISA was evaluated in 100 COVID positive. PRAK-03202 was produced at a 5 L scale using fed-batch fermentation. Results: Dot blot confirmed the presence of S, E, and M proteins in PRAK-03202. The number of particles in PRAK-03202 was 1.21 × 109 mL−1. In samples collected >14 days after symptom onset, the sensitivity, specificity, and accuracy of VLP-ELISA were 96%. We did not observe any significant differences in sensitivity, specificity, and accuracy when post-COVID-19 samples were used as negative controls compared to pre-COVID-samples. At a scale of 5 L, the total yield of PRAK-03202 was 100–120 mg/L. Conclusion: In conclusion, we have successfully developed an in-house VLP-ELISA to detect IgG antibodies against three antigens of SARS-CoV-2 as a simple and affordable alternative test.
Collapse
Affiliation(s)
- Dilip Kumar
- Research and Developmental Laboratory, Premas Biotech Private Limited, Sector 4, IMT Manesar, Gurgaon 122050, India (R.G.)
| | - Sourav Singha Roy
- Research and Developmental Laboratory, Premas Biotech Private Limited, Sector 4, IMT Manesar, Gurgaon 122050, India (R.G.)
| | - Ruchir Rastogi
- Research and Developmental Laboratory, Premas Biotech Private Limited, Sector 4, IMT Manesar, Gurgaon 122050, India (R.G.)
| | - Kajal Arora
- Research and Developmental Laboratory, Premas Biotech Private Limited, Sector 4, IMT Manesar, Gurgaon 122050, India (R.G.)
| | - Avinash Undale
- Research and Developmental Laboratory, Premas Biotech Private Limited, Sector 4, IMT Manesar, Gurgaon 122050, India (R.G.)
| | - Reeshu Gupta
- Research and Developmental Laboratory, Premas Biotech Private Limited, Sector 4, IMT Manesar, Gurgaon 122050, India (R.G.)
- Centre of Research for Development, Parul University, Vadodara 391760, India
| | - Nupur Mehrotra Arora
- Research and Developmental Laboratory, Premas Biotech Private Limited, Sector 4, IMT Manesar, Gurgaon 122050, India (R.G.)
| | - Prabuddha K. Kundu
- Research and Developmental Laboratory, Premas Biotech Private Limited, Sector 4, IMT Manesar, Gurgaon 122050, India (R.G.)
- Correspondence: or
| |
Collapse
|
16
|
Rafat N, Brewer L, Das N, Trivedi DJ, Kaszala BK, Sarkar A. Inexpensive High-Throughput Multiplexed Biomarker Detection Using Enzymatic Metallization with Cellphone-Based Computer Vision. ACS Sens 2023; 8:534-542. [PMID: 36753573 PMCID: PMC9972466 DOI: 10.1021/acssensors.2c01429] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Multiplexed biomarker detection can play a critical role in reliable and comprehensive disease diagnosis and prediction of outcome. Enzyme-linked immunosorbent assay (ELISA) is the gold standard method for immunobinding-based biomarker detection. However, this is currently expensive, limited to centralized laboratories, and usually limited to the detection of a single biomarker at a time. We present a low-cost, smartphone-based portable biosensing platform for high-throughput, multiplexed, sensitive, and quantitative detection of biomarkers from single, low-volume drops (<1 μL) of clinical samples. Biomarker binding to spotted capture antigens is converted, via enzymatic metallization, to the localized surface deposition of amplified, dry-stable, silver metal spots whose darkness is proportional to biomarker concentration. A custom smartphone application is developed, which uses real-time computer vision to enable easy optical detection of the deposited metal spots and sensitive and reproducible quantification of the biomarkers. We demonstrate the use of this platform for high-throughput, multiplexed detection of multiple viral antigen-specific antibodies from convalescent COVID-19 patient serum as well as vaccine-elicited antibody responses from uninfected vaccine-recipient serum and show that distinct multiplexed antibody fingerprints are observed among them.
Collapse
Affiliation(s)
- Neda Rafat
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Lee Brewer
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Nabojeet Das
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Dhruti J Trivedi
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Balazs K Kaszala
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Aniruddh Sarkar
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| |
Collapse
|
17
|
Klüpfel J, Paßreiter S, Rumpf M, Christa C, Holthoff HP, Ungerer M, Lohse M, Knolle P, Protzer U, Elsner M, Seidel M. Automated detection of neutralizing SARS-CoV-2 antibodies in minutes using a competitive chemiluminescence immunoassay. Anal Bioanal Chem 2023; 415:391-404. [PMID: 36346456 PMCID: PMC9643999 DOI: 10.1007/s00216-022-04416-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/13/2022] [Accepted: 10/31/2022] [Indexed: 11/09/2022]
Abstract
The SARS-CoV-2 pandemic has shown the importance of rapid and comprehensive diagnostic tools. While there are numerous rapid antigen tests available, rapid serological assays for the detection of neutralizing antibodies are and will be needed to determine not only the amount of antibodies formed after infection or vaccination but also their neutralizing potential, preventing the cell entry of SARS-CoV-2. Current active-virus neutralization assays require biosafety level 3 facilities, while virus-free surrogate assays are more versatile in applications, but still take typically several hours until results are available. To overcome these disadvantages, we developed a competitive chemiluminescence immunoassay that enables the detection of neutralizing SARS-CoV-2 antibodies within 7 min. The neutralizing antibodies bind to the viral receptor binding domain (RBD) and inhibit the binding to the human angiotensin-converting enzyme 2 (ACE2) receptor. This competitive binding inhibition test was characterized with a set of 80 samples, which could all be classified correctly. The assay results favorably compare to those obtained with a more time-intensive ELISA-based neutralization test and a commercial surrogate neutralization assay. Our test could further be used to detect individuals with a high total IgG antibody titer, but only a low neutralizing titer, as well as for monitoring neutralizing antibodies after vaccinations. This effective performance in SARS-CoV-2 seromonitoring delineates the potential for the test to be adapted to other diseases in the future.
Collapse
Affiliation(s)
- Julia Klüpfel
- Institute of Water Chemistry, Chair of Analytical Chemistry and Water Chemistry, Technical University of Munich, Lichtenbergstr. 4, 85748 Garching, Germany
| | - Sandra Paßreiter
- Institute of Water Chemistry, Chair of Analytical Chemistry and Water Chemistry, Technical University of Munich, Lichtenbergstr. 4, 85748 Garching, Germany
| | - Melina Rumpf
- Institute of Water Chemistry, Chair of Analytical Chemistry and Water Chemistry, Technical University of Munich, Lichtenbergstr. 4, 85748 Garching, Germany
| | - Catharina Christa
- Institute of Virology, Technical University of Munich/Helmholtz Zentrum München, Trogerstr. 30, 81675 Munich, Germany
| | | | - Martin Ungerer
- ISAR Bioscience GmbH, Semmelweisstr. 5, 82152 Planegg, Germany
| | - Martin Lohse
- ISAR Bioscience GmbH, Semmelweisstr. 5, 82152 Planegg, Germany
| | - Percy Knolle
- Institute of Molecular Immunology/Experimental Oncology, Technical University of Munich, Ismaningerstr. 22, 81675 Munich, Germany
| | - Ulrike Protzer
- Institute of Virology, Technical University of Munich/Helmholtz Zentrum München, Trogerstr. 30, 81675 Munich, Germany ,German Center for Infection Research (DZIF), 81675 Munich, Germany
| | - Martin Elsner
- Institute of Water Chemistry, Chair of Analytical Chemistry and Water Chemistry, Technical University of Munich, Lichtenbergstr. 4, 85748 Garching, Germany
| | - Michael Seidel
- Institute of Water Chemistry, Chair of Analytical Chemistry and Water Chemistry, Technical University of Munich, Lichtenbergstr. 4, 85748 Garching, Germany
| |
Collapse
|
18
|
Mensah BA, Ndong IC, Quashie PK, Guichet E, Abuaku B, Effah-Baafi Y, Tapela K, Asiedu K, Appiedu-Addo SNA, Obbeng LB, Amponsah JA, Kusi KA, Ofori M, Ayouba A, Courtin D, Tahar R, Delaporte E, Awandare G, Ndam NT. Population-based sero-epidemiological investigation of the dynamics of SARS-CoV-2 infections in the Greater Accra Region of Ghana. Sci Rep 2022; 12:21582. [PMID: 36517505 PMCID: PMC9748398 DOI: 10.1038/s41598-022-25598-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 12/01/2022] [Indexed: 12/15/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic devastated countries worldwide, and resulted in a global shutdown. Not all infections are symptomatic and hence the extent of SARS-CoV-2 infection in the community is unknown. The paper presents the dynamics of the SARS-CoV-2 epidemic in the Greater Accra Metropolis, describing the evolution of seroprevalence through time and by age group. Three repeated independent population-based surveys at 6-week intervals were conducted in from November 2020 to July 2021. The global and by age-groups weighted seroprevalences were estimated and the risk factors for SARS-CoV-2 antibody seropositivity were assessed using logistic regression. The overall age-standardized SARS-CoV-2 antibody seroprevalence for both spike and nucleocapsid increased from 13.8% (95% CI 11.9, 16.1) in November 2020 to 39.6% (95% CI 34.8, 44.6) in July 2021. After controlling for gender, marital status, education level, and occupation, the older age group over 40 years had a higher odds of seropositivity than the younger age group (OR 3.0 [95% CI 1.1-8.5]) in the final survey. Pupils or students had 3.3-fold increased odds of seropositivity (OR 3.2 [95% CI 1.1-8.5]) compared to the unemployed. This study reinforces that, SARS-CoV-2 infections have been significantly higher than reported.
Collapse
Affiliation(s)
| | - Ignatius Cheng Ndong
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
- Department of Biochemistry, Catholic University of Cameroon, Bamenda, Cameroon
| | - Peter Kojo Quashie
- West African Centre for Cell Biology of Infectious Pathogens, University of Ghana, Legon, Accra, Ghana
| | - Emilande Guichet
- TransVIHMI, University of Montpellier, IRD and Institut National de La Santé Et de La Recherche Médicale (INSERM), Montpellier, France
| | - Benjamin Abuaku
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Yaw Effah-Baafi
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Kesego Tapela
- West African Centre for Cell Biology of Infectious Pathogens, University of Ghana, Legon, Accra, Ghana
| | - Kwame Asiedu
- West African Centre for Cell Biology of Infectious Pathogens, University of Ghana, Legon, Accra, Ghana
| | | | - Louisa Baaba Obbeng
- West African Centre for Cell Biology of Infectious Pathogens, University of Ghana, Legon, Accra, Ghana
| | - Jones Amo Amponsah
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Kwadwo Asamoah Kusi
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Michael Ofori
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Ahidjo Ayouba
- TransVIHMI, University of Montpellier, IRD and Institut National de La Santé Et de La Recherche Médicale (INSERM), Montpellier, France
| | - David Courtin
- MERIT, IRD, Université de Paris, 75006, Paris, France
| | - Rachida Tahar
- MERIT, IRD, Université de Paris, 75006, Paris, France
| | - Eric Delaporte
- TransVIHMI, University of Montpellier, IRD and Institut National de La Santé Et de La Recherche Médicale (INSERM), Montpellier, France
| | - Gordon Awandare
- West African Centre for Cell Biology of Infectious Pathogens, University of Ghana, Legon, Accra, Ghana
| | | |
Collapse
|
19
|
Ali A, Zhang GF, Hu C, Yuan B, Jahan S, Kitsios GD, Morris A, Gao SJ, Panat R. Ultrarapid and ultrasensitive detection of SARS-CoV-2 antibodies in COVID-19 patients via a 3D-printed nanomaterial-based biosensing platform. J Med Virol 2022; 94:5808-5826. [PMID: 35981973 PMCID: PMC9538259 DOI: 10.1002/jmv.28075] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/07/2022] [Accepted: 08/17/2022] [Indexed: 01/06/2023]
Abstract
Rapid detection of antibodies during infection and after vaccination is critical for the control of infectious outbreaks, understanding immune response, and evaluating vaccine efficacy. In this manuscript, we evaluate a simple ultrarapid test for SARS-CoV-2 antibodies in COVID-19 patients, which gives quantitative results (i.e., antibody concentration) in 10-12 s using a previously reported nanomaterial-based three-dimensional (3D)-printed biosensing platform. This platform consists of a micropillar array electrode fabricated via 3D printing of aerosolized gold nanoparticles and coated with nanoflakes of graphene and specific SARS-CoV-2 antigens, including spike S1, S1 receptor-binding domain (RBD) and nucleocapsid (N). The sensor works on the principle of electrochemical transduction, where the change of sensor impedance is realized by the interactions between the viral proteins attached to the sensor electrode surface and the antibodies. The three sensors were used to test samples from 17 COVID-19 patients and 3 patients without COVID-19. Unlike other serological tests, the 3D sensors quantitatively detected antibodies at a concentration as low as picomole within 10-12 s in human plasma samples. We found that the studied COVID-19 patients had higher concentrations of antibodies to spike proteins (RBD and S1) than to the N protein. These results demonstrate the enormous potential of the rapid antibody test platform for understanding patients' immunity, disease epidemiology and vaccine efficacy, and facilitating the control and prevention of infectious epidemics.
Collapse
Affiliation(s)
- Azahar Ali
- Department of Mechanical Engineering, Carnegie Mellon
University, Pittsburgh, PA, 15213 USA
- Current address: Department of Animal and Poultry Sciences,
Virginia Tech, Blacksburg, VA, 24061 USA
| | - George Fei Zhang
- Cancer Virology Program, UPMC Hillman Cancer Center and
Department of Microbiology and Molecular Genetics, University of Pittsburgh School
of Medicine, Pittsburgh, PA, 15213 USA
| | - Chunshan Hu
- Department of Mechanical Engineering, Carnegie Mellon
University, Pittsburgh, PA, 15213 USA
| | - Bin Yuan
- Department of Mechanical Engineering, Carnegie Mellon
University, Pittsburgh, PA, 15213 USA
| | - Sanjida Jahan
- Department of Mechanical Engineering, Carnegie Mellon
University, Pittsburgh, PA, 15213 USA
| | - Georgios D. Kitsios
- Division of Pulmonary, Allergy and Critical Care Medicine,
Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA,
15213 USA
| | - Alison Morris
- Division of Pulmonary, Allergy and Critical Care Medicine,
Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA,
15213 USA
| | - Shou-Jiang Gao
- Cancer Virology Program, UPMC Hillman Cancer Center and
Department of Microbiology and Molecular Genetics, University of Pittsburgh School
of Medicine, Pittsburgh, PA, 15213 USA
| | - Rahul Panat
- Department of Mechanical Engineering, Carnegie Mellon
University, Pittsburgh, PA, 15213 USA
| |
Collapse
|
20
|
Rafat N, Zhang H, Rudge J, Kim YN, Peddireddy SP, Das N, Sarkar A. Enhanced Enzymatically Amplified Metallization on Nanostructured Surfaces for Multiplexed Point-of-Care Electrical Detection of COVID-19 Biomarkers. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2203309. [PMID: 36036173 PMCID: PMC9538889 DOI: 10.1002/smll.202203309] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/27/2022] [Indexed: 06/15/2023]
Abstract
Inexpensive yet sensitive and specific biomarker detection is a critical bottleneck in diagnostics, monitoring, and surveillance of infectious diseases such as COVID-19. Multiplexed detection of several biomarkers can achieve wider diagnostic applicability, accuracy, and ease-of-use, while reducing cost. Current biomarker detection methods often use enzyme-linked immunosorbent assays (ELISA) with optical detection which offers high sensitivity and specificity. However, this is complex, expensive, and limited to detecting only a single analyte at a time. Here, it is found that biomarker-bound enzyme-labeled probes act synergistically with nanostructured catalytic surfaces and can be used to selectively reduce a soluble silver substrate to generate highly dense and conductive, localized surface silver metallization on microelectrode arrays. This enables a sensitive and quantitative, simple, direct electronic readout of biomarker binding without the use of any intermediate optics. Furthermore, the localized and dry-phase stable nature of the metallization enables multiplexed electronic measurement of several biomarkers from a single drop (<10 µL) of sample on a microchip.This method is applied for the multiplexed point-of-care (POC) quantitative detection of multiple COVID-19 antigen-specific antibodies. Combining a simple microchip and an inexpensive, cellphone-interfaced, portable reader, the detection and discrimination of biomarkers of prior infection versus vaccination is demonstrated.
Collapse
Affiliation(s)
- Neda Rafat
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, 30332, USA
| | - Hanhao Zhang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, 30332, USA
| | - Josiah Rudge
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, 30332, USA
| | - Yoo Na Kim
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, 30332, USA
| | - Sai Preetham Peddireddy
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, 30332, USA
| | - Nabojeet Das
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, 30332, USA
| | - Aniruddh Sarkar
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, 30332, USA
| |
Collapse
|
21
|
Fox T, Geppert J, Dinnes J, Scandrett K, Bigio J, Sulis G, Hettiarachchi D, Mathangasinghe Y, Weeratunga P, Wickramasinghe D, Bergman H, Buckley BS, Probyn K, Sguassero Y, Davenport C, Cunningham J, Dittrich S, Emperador D, Hooft L, Leeflang MM, McInnes MD, Spijker R, Struyf T, Van den Bruel A, Verbakel JY, Takwoingi Y, Taylor-Phillips S, Deeks JJ. Antibody tests for identification of current and past infection with SARS-CoV-2. Cochrane Database Syst Rev 2022; 11:CD013652. [PMID: 36394900 PMCID: PMC9671206 DOI: 10.1002/14651858.cd013652.pub2] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
BACKGROUND The diagnostic challenges associated with the COVID-19 pandemic resulted in rapid development of diagnostic test methods for detecting SARS-CoV-2 infection. Serology tests to detect the presence of antibodies to SARS-CoV-2 enable detection of past infection and may detect cases of SARS-CoV-2 infection that were missed by earlier diagnostic tests. Understanding the diagnostic accuracy of serology tests for SARS-CoV-2 infection may enable development of effective diagnostic and management pathways, inform public health management decisions and understanding of SARS-CoV-2 epidemiology. OBJECTIVES To assess the accuracy of antibody tests, firstly, to determine if a person presenting in the community, or in primary or secondary care has current SARS-CoV-2 infection according to time after onset of infection and, secondly, to determine if a person has previously been infected with SARS-CoV-2. Sources of heterogeneity investigated included: timing of test, test method, SARS-CoV-2 antigen used, test brand, and reference standard for non-SARS-CoV-2 cases. SEARCH METHODS The COVID-19 Open Access Project living evidence database from the University of Bern (which includes daily updates from PubMed and Embase and preprints from medRxiv and bioRxiv) was searched on 30 September 2020. We included additional publications from the Evidence for Policy and Practice Information and Co-ordinating Centre (EPPI-Centre) 'COVID-19: Living map of the evidence' and the Norwegian Institute of Public Health 'NIPH systematic and living map on COVID-19 evidence'. We did not apply language restrictions. SELECTION CRITERIA We included test accuracy studies of any design that evaluated commercially produced serology tests, targeting IgG, IgM, IgA alone, or in combination. Studies must have provided data for sensitivity, that could be allocated to a predefined time period after onset of symptoms, or after a positive RT-PCR test. Small studies with fewer than 25 SARS-CoV-2 infection cases were excluded. We included any reference standard to define the presence or absence of SARS-CoV-2 (including reverse transcription polymerase chain reaction tests (RT-PCR), clinical diagnostic criteria, and pre-pandemic samples). DATA COLLECTION AND ANALYSIS We use standard screening procedures with three reviewers. Quality assessment (using the QUADAS-2 tool) and numeric study results were extracted independently by two people. Other study characteristics were extracted by one reviewer and checked by a second. We present sensitivity and specificity with 95% confidence intervals (CIs) for each test and, for meta-analysis, we fitted univariate random-effects logistic regression models for sensitivity by eligible time period and for specificity by reference standard group. Heterogeneity was investigated by including indicator variables in the random-effects logistic regression models. We tabulated results by test manufacturer and summarised results for tests that were evaluated in 200 or more samples and that met a modification of UK Medicines and Healthcare products Regulatory Agency (MHRA) target performance criteria. MAIN RESULTS We included 178 separate studies (described in 177 study reports, with 45 as pre-prints) providing 527 test evaluations. The studies included 64,688 samples including 25,724 from people with confirmed SARS-CoV-2; most compared the accuracy of two or more assays (102/178, 57%). Participants with confirmed SARS-CoV-2 infection were most commonly hospital inpatients (78/178, 44%), and pre-pandemic samples were used by 45% (81/178) to estimate specificity. Over two-thirds of studies recruited participants based on known SARS-CoV-2 infection status (123/178, 69%). All studies were conducted prior to the introduction of SARS-CoV-2 vaccines and present data for naturally acquired antibody responses. Seventy-nine percent (141/178) of studies reported sensitivity by week after symptom onset and 66% (117/178) for convalescent phase infection. Studies evaluated enzyme-linked immunosorbent assays (ELISA) (165/527; 31%), chemiluminescent assays (CLIA) (167/527; 32%) or lateral flow assays (LFA) (188/527; 36%). Risk of bias was high because of participant selection (172, 97%); application and interpretation of the index test (35, 20%); weaknesses in the reference standard (38, 21%); and issues related to participant flow and timing (148, 82%). We judged that there were high concerns about the applicability of the evidence related to participants in 170 (96%) studies, and about the applicability of the reference standard in 162 (91%) studies. Average sensitivities for current SARS-CoV-2 infection increased by week after onset for all target antibodies. Average sensitivity for the combination of either IgG or IgM was 41.1% in week one (95% CI 38.1 to 44.2; 103 evaluations; 3881 samples, 1593 cases), 74.9% in week two (95% CI 72.4 to 77.3; 96 evaluations, 3948 samples, 2904 cases) and 88.0% by week three after onset of symptoms (95% CI 86.3 to 89.5; 103 evaluations, 2929 samples, 2571 cases). Average sensitivity during the convalescent phase of infection (up to a maximum of 100 days since onset of symptoms, where reported) was 89.8% for IgG (95% CI 88.5 to 90.9; 253 evaluations, 16,846 samples, 14,183 cases), 92.9% for IgG or IgM combined (95% CI 91.0 to 94.4; 108 evaluations, 3571 samples, 3206 cases) and 94.3% for total antibodies (95% CI 92.8 to 95.5; 58 evaluations, 7063 samples, 6652 cases). Average sensitivities for IgM alone followed a similar pattern but were of a lower test accuracy in every time slot. Average specificities were consistently high and precise, particularly for pre-pandemic samples which provide the least biased estimates of specificity (ranging from 98.6% for IgM to 99.8% for total antibodies). Subgroup analyses suggested small differences in sensitivity and specificity by test technology however heterogeneity in study results, timing of sample collection, and smaller sample numbers in some groups made comparisons difficult. For IgG, CLIAs were the most sensitive (convalescent-phase infection) and specific (pre-pandemic samples) compared to both ELISAs and LFAs (P < 0.001 for differences across test methods). The antigen(s) used (whether from the Spike-protein or nucleocapsid) appeared to have some effect on average sensitivity in the first weeks after onset but there was no clear evidence of an effect during convalescent-phase infection. Investigations of test performance by brand showed considerable variation in sensitivity between tests, and in results between studies evaluating the same test. For tests that were evaluated in 200 or more samples, the lower bound of the 95% CI for sensitivity was 90% or more for only a small number of tests (IgG, n = 5; IgG or IgM, n = 1; total antibodies, n = 4). More test brands met the MHRA minimum criteria for specificity of 98% or above (IgG, n = 16; IgG or IgM, n = 5; total antibodies, n = 7). Seven assays met the specified criteria for both sensitivity and specificity. In a low-prevalence (2%) setting, where antibody testing is used to diagnose COVID-19 in people with symptoms but who have had a negative PCR test, we would anticipate that 1 (1 to 2) case would be missed and 8 (5 to 15) would be falsely positive in 1000 people undergoing IgG or IgM testing in week three after onset of SARS-CoV-2 infection. In a seroprevalence survey, where prevalence of prior infection is 50%, we would anticipate that 51 (46 to 58) cases would be missed and 6 (5 to 7) would be falsely positive in 1000 people having IgG tests during the convalescent phase (21 to 100 days post-symptom onset or post-positive PCR) of SARS-CoV-2 infection. AUTHORS' CONCLUSIONS Some antibody tests could be a useful diagnostic tool for those in whom molecular- or antigen-based tests have failed to detect the SARS-CoV-2 virus, including in those with ongoing symptoms of acute infection (from week three onwards) or those presenting with post-acute sequelae of COVID-19. However, antibody tests have an increasing likelihood of detecting an immune response to infection as time since onset of infection progresses and have demonstrated adequate performance for detection of prior infection for sero-epidemiological purposes. The applicability of results for detection of vaccination-induced antibodies is uncertain.
Collapse
Affiliation(s)
- Tilly Fox
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Julia Geppert
- Division of Health Sciences, Warwick Medical School, University of Warwick, Coventry, UK
| | - Jacqueline Dinnes
- Test Evaluation Research Group, Institute of Applied Health Research, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
| | - Katie Scandrett
- Test Evaluation Research Group, Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Jacob Bigio
- Research Institute of the McGill University Health Centre, Montreal, Canada
- McGill International TB Centre, Montreal, Canada
| | - Giorgia Sulis
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Canada
| | - Dineshani Hettiarachchi
- Department of Anatomy Genetics and Biomedical Informatics, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - Yasith Mathangasinghe
- Department of Anatomy Genetics and Biomedical Informatics, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
- Australian Regenerative Medicine Institute, Monash University, Clayton, Australia
| | - Praveen Weeratunga
- Department of Clinical Medicine, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | | | | | - Brian S Buckley
- Cochrane Response, Cochrane, London, UK
- Department of Surgery, University of the Philippines, Manila, Philippines
| | | | | | - Clare Davenport
- Test Evaluation Research Group, Institute of Applied Health Research, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
| | - Jane Cunningham
- Global Malaria Programme, World Health Organization, Geneva, Switzerland
| | | | | | - Lotty Hooft
- Cochrane Netherlands, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht , Netherlands
| | - Mariska Mg Leeflang
- Epidemiology and Data Science, Amsterdam UMC location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Public Health, Amsterdam, Netherlands
| | | | - René Spijker
- Medical Library, Amsterdam UMC, University of Amsterdam, Amsterdam Public Health, Amsterdam, Netherlands
- Cochrane Netherlands, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Thomas Struyf
- Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
| | - Ann Van den Bruel
- Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
| | - Jan Y Verbakel
- Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
| | - Yemisi Takwoingi
- Test Evaluation Research Group, Institute of Applied Health Research, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
| | - Sian Taylor-Phillips
- Division of Health Sciences, Warwick Medical School, University of Warwick, Coventry, UK
- Test Evaluation Research Group, Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Jonathan J Deeks
- Test Evaluation Research Group, Institute of Applied Health Research, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
| |
Collapse
|
22
|
Tapela K, Oyawoye FO, Olwal CO, Opurum PC, Amponsah JA, Segbedzi KAL, Tetteh B, Kumi-Ansah F, Mutungi JK, Obodai E, Amoako E, Agyemang S, Ndam NT, Ampofo WK, Rayner JC, Awandare GA, Paemka L, Bediako Y, Quashie PK. Probing SARS-CoV-2-positive plasma to identify potential factors correlating with mild COVID-19 in Ghana, West Africa. BMC Med 2022; 20:370. [PMID: 36184636 PMCID: PMC9527094 DOI: 10.1186/s12916-022-02571-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 09/20/2022] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND West Africa has recorded a relatively higher proportion of asymptomatic coronavirus disease 2019 (COVID-19) cases than the rest of the world, and West Africa-specific host factors could play a role in this discrepancy. Here, we assessed the association between COVID-19 severity among Ghanaians with their immune profiles and ABO blood groups. METHODS Plasma samples were obtained from Ghanaians PCR-confirmed severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-positive individuals. The participants were categorized into symptomatic and asymptomatic cases. Cytokine profiling and antibody quantification were performed using Luminex™ multiplex assay whereas antigen-driven agglutination assay was used to assess the ABO blood groups. Immune profile levels between symptomatic and asymptomatic groups were compared using the two-tailed Mann-Whitney U test. Multiple comparisons of cytokine levels among and between days were tested using Kruskal-Wallis with Dunn's post hoc test. Correlations within ABO blood grouping (O's and non-O's) and between cytokines were determined using Spearman correlations. Logistic regression analysis was performed to assess the association of various cytokines with asymptomatic phenotype. RESULTS There was a trend linking blood group O to reduced disease severity, but this association was not statistically significant. Generally, symptomatic patients displayed significantly (p < 0.05) higher cytokine levels compared to asymptomatic cases with exception of Eotaxin, which was positively associated with asymptomatic cases. There were also significant (p < 0.05) associations between other immune markers (IL-6, IL-8 and IL-1Ra) and disease severity. Cytokines' clustering patterns differ between symptomatic and asymptomatic cases. We observed a steady decrease in the concentration of most cytokines over time, while anti-SARS-CoV-2 antibody levels were stable for at least a month, regardless of the COVID-19 status. CONCLUSIONS The findings suggest that genetic background and pre-existing immune response patterns may in part shape the nature of the symptomatic response against COVID-19 in a West African population. This study offers clear directions to be explored further in larger studies.
Collapse
Affiliation(s)
- Kesego Tapela
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana.,Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana
| | - Fatima O Oyawoye
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana.,Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana
| | - Charles Ochieng' Olwal
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana.,Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana
| | - Precious C Opurum
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
| | - Jones Amo Amponsah
- Department of Immunology, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Accra, Ghana
| | - Kekeli Aku Lumor Segbedzi
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana.,Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana
| | - Becky Tetteh
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
| | | | - Joe K Mutungi
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
| | - Evangeline Obodai
- Virology Department, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Accra, Ghana
| | - Emmanuella Amoako
- Department of Pediatrics, Cape Coast Teaching Hospital, Cape Coast, Ghana.,Yemaachi Biotech Inc., 222 Swaniker St, Accra, Ghana
| | - Seth Agyemang
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana.,Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana
| | - Nicaise Tuikue Ndam
- UMR261 MERIT and Head of IRD Branch in Benin-Nigeria-Togo-Ghana, Accra, Ghana
| | - William Kwabena Ampofo
- Virology Department, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Accra, Ghana
| | - Julian C Rayner
- Cambridge Institute for Medical Research, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Gordon A Awandare
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana.,Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana
| | - Lily Paemka
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana.,Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana.,Yemaachi Biotech Inc., 222 Swaniker St, Accra, Ghana
| | - Yaw Bediako
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana.,Yemaachi Biotech Inc., 222 Swaniker St, Accra, Ghana.,The Francis Crick Institute, 1 Midland Rd, London, NW1 1AT, UK
| | - Peter Kojo Quashie
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana. .,The Francis Crick Institute, 1 Midland Rd, London, NW1 1AT, UK.
| |
Collapse
|
23
|
Ellipilli S, Wang H, Lee WJ, Shu D, Guo P. Proof-of-concept for speedy development of rapid and simple at-home method for potential diagnosis of early COVID-19 mutant infections using nanogold and aptamer. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2022; 45:102590. [PMID: 35905841 PMCID: PMC9315840 DOI: 10.1016/j.nano.2022.102590] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 07/14/2022] [Accepted: 07/16/2022] [Indexed: 06/15/2023]
Abstract
The positive single-stranded nature of COVID-19 mRNA led to the low proof-reading efficacy for its genome authentication. Thus mutant covid-19 strains have been rapidly evolving. Besides Alpha, Beta, Gamma, Delta, and Omicron variants, currently, subvariants of omicron are circulating, including BA.4, BA.5, and BA.2.12.1. Therefore, the speedy development of a rapid, simple, and easier diagnosis method to deal with new mutant covid viral infection is critically important. Many diagnosis methods have been developed for COVID-19 detection such as RT-PCR and antibodies detection. However, the former is time-consuming, laborious, and expensive, and the latter relies on the production of antibodies making it not suitable for the early diagnosis of viral infection. Many lateral-flow methods are available but might not be suitable for detecting the mutants, Here we proved the concept for the speedy development of a simple, rapid, and cost-effective early at-home diagnosis method for mutant Covid-19 infection by combining a new aptamer. The idea is to use the current lateral flow Covid-19 diagnosis system available in the market or to use one existing antibody for the Lateral Flow Nitrocellulose filter. To prove the concept, the DNA aptamer specific to spike proteins (S-proteins) was conjugated to gold nanoparticles and served as a detection probe. An antibody that is specific to spike proteins overexpressed on COVID viral particles was used as a second probe immobilized to the nitrocellulose membrane. The aptamer conjugated nanoparticles were incubated with spike proteins for half an hour and tested for their ability to bind to antibodies anchored on the nitrocellulose membrane. The gold nanoparticles were visualized on the nitrocellulose membrane due to interaction between the antigen (S-protein) with both the aptamer and the antibody. Thus, the detection of viral antigen can be obtained within 2 h, with a cost of less than $5 for the diagnosis reagent. In the future, as long as the mutant of the newly emerged viral surface protein is reported, a peptide or protein corresponding to the mutation can be produced by peptide synthesis or gene cloning within several days. An RNA or DNA aptamer can be generated quickly via SELEX. A gold-labeled aptamer specific to spike proteins (S-proteins) will serve as a detection probe. Any available lateral-flow diagnosis kits with an immobilized antibody that has been available on the market, or simply an antibody that binds COVID-19 virus might be used as a second probe immobilized on the nitrocellulose. The diagnosis method can be carried out by patients at home if a clinical trial verifies the feasibility and specificity of this method.
Collapse
Affiliation(s)
- Satheesh Ellipilli
- Center for RNA Nanobiotechnology and Nanomedicine, The Ohio State University, Columbus, OH 43210, USA; College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; College of Medicine, The Ohio State University, Columbus, OH 43210, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Hongzhi Wang
- Center for RNA Nanobiotechnology and Nanomedicine, The Ohio State University, Columbus, OH 43210, USA; College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; College of Medicine, The Ohio State University, Columbus, OH 43210, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Wen-Jui Lee
- Center for RNA Nanobiotechnology and Nanomedicine, The Ohio State University, Columbus, OH 43210, USA; College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; College of Medicine, The Ohio State University, Columbus, OH 43210, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Dan Shu
- Center for RNA Nanobiotechnology and Nanomedicine, The Ohio State University, Columbus, OH 43210, USA; College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; College of Medicine, The Ohio State University, Columbus, OH 43210, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA.
| | - Peixuan Guo
- Center for RNA Nanobiotechnology and Nanomedicine, The Ohio State University, Columbus, OH 43210, USA; College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; College of Medicine, The Ohio State University, Columbus, OH 43210, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
24
|
Colombo VC, Sluydts V, Mariën J, Vanden Broecke B, Van Houtte N, Leirs W, Jacobs L, Iserbyt A, Hubert M, Heyndrickx L, Goris H, Delputte P, De Roeck N, Elst J, Ariën KK, Leirs H, Gryseels S. SARS-CoV-2 surveillance in Norway rats (Rattus norvegicus) from Antwerp sewer system, Belgium. Transbound Emerg Dis 2022; 69:3016-3021. [PMID: 34224205 PMCID: PMC8447303 DOI: 10.1111/tbed.14219] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 06/30/2021] [Accepted: 07/02/2021] [Indexed: 12/27/2022]
Abstract
SARS-CoV-2 human-to-animal transmission can lead to the establishment of novel reservoirs and the evolution of new variants with the potential to start new outbreaks in humans. We tested Norway rats inhabiting the sewer system of Antwerp, Belgium, for the presence of SARS-CoV-2 following a local COVID-19 epidemic peak. In addition, we discuss the use and interpretation of SARS-CoV-2 serological tests on non-human samples. Between November and December 2020, Norway rat oral swabs, faeces and tissues from the sewer system of Antwerp were collected to be tested by RT-qPCR for the presence of SARS-CoV-2. Serum samples were screened for the presence of anti-SARS-CoV-2 IgG antibodies using a Luminex microsphere immunoassay (MIA). Samples considered positive were then checked for neutralizing antibodies using a conventional viral neutralization test (cVNT). The serum of 35 rats was tested by MIA showing three potentially positive sera that were later negative by cVNT. All tissue samples of 39 rats analysed tested negative for SARS-CoV-2 RNA. This is the first study that evaluates SARS-CoV-2 infection in urban rats. We can conclude that the sample of rats analysed had never been infected with SARS-CoV-2. However, monitoring activities should continue due to the emergence of new variants prone to infect Muridae rodents.
Collapse
Affiliation(s)
- Valeria Carolina Colombo
- Evolutionary Ecology GroupDepartment of BiologyUniversity of AntwerpAntwerpBelgium
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)Buenos AiresArgentina
| | - Vincent Sluydts
- Evolutionary Ecology GroupDepartment of BiologyUniversity of AntwerpAntwerpBelgium
| | - Joachim Mariën
- Evolutionary Ecology GroupDepartment of BiologyUniversity of AntwerpAntwerpBelgium
- Virology UnitDepartment of Biomedical SciencesInstitute of Tropical MedicineAntwerpBelgium
| | - Bram Vanden Broecke
- Evolutionary Ecology GroupDepartment of BiologyUniversity of AntwerpAntwerpBelgium
| | - Natalie Van Houtte
- Evolutionary Ecology GroupDepartment of BiologyUniversity of AntwerpAntwerpBelgium
| | - Wannes Leirs
- Evolutionary Ecology GroupDepartment of BiologyUniversity of AntwerpAntwerpBelgium
| | - Lotte Jacobs
- Laboratory for MicrobiologyParasitology and Hygiene (LMPH)University of AntwerpAntwerpBelgium
| | - Arne Iserbyt
- Evolutionary Ecology GroupDepartment of BiologyUniversity of AntwerpAntwerpBelgium
| | - Marine Hubert
- Evolutionary Ecology GroupDepartment of BiologyUniversity of AntwerpAntwerpBelgium
| | - Leo Heyndrickx
- Virology UnitDepartment of Biomedical SciencesInstitute of Tropical MedicineAntwerpBelgium
| | - Hanne Goris
- Evolutionary Ecology GroupDepartment of BiologyUniversity of AntwerpAntwerpBelgium
| | - Peter Delputte
- Laboratory for MicrobiologyParasitology and Hygiene (LMPH)University of AntwerpAntwerpBelgium
| | - Naomi De Roeck
- Laboratory for MicrobiologyParasitology and Hygiene (LMPH)University of AntwerpAntwerpBelgium
| | - Joris Elst
- Evolutionary Ecology GroupDepartment of BiologyUniversity of AntwerpAntwerpBelgium
| | - Kevin K. Ariën
- Virology UnitDepartment of Biomedical SciencesInstitute of Tropical MedicineAntwerpBelgium
| | - Herwig Leirs
- Evolutionary Ecology GroupDepartment of BiologyUniversity of AntwerpAntwerpBelgium
| | - Sophie Gryseels
- Evolutionary Ecology GroupDepartment of BiologyUniversity of AntwerpAntwerpBelgium
- OD Taxonomy and PhylogenyRoyal Belgian Institute of Natural SciencesBrusselsBelgium
| |
Collapse
|
25
|
Gao K, Wang R, Chen J, Cheng L, Frishcosy J, Huzumi Y, Qiu Y, Schluckbier T, Wei X, Wei GW. Methodology-Centered Review of Molecular Modeling, Simulation, and Prediction of SARS-CoV-2. Chem Rev 2022; 122:11287-11368. [PMID: 35594413 PMCID: PMC9159519 DOI: 10.1021/acs.chemrev.1c00965] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Despite tremendous efforts in the past two years, our understanding of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), virus-host interactions, immune response, virulence, transmission, and evolution is still very limited. This limitation calls for further in-depth investigation. Computational studies have become an indispensable component in combating coronavirus disease 2019 (COVID-19) due to their low cost, their efficiency, and the fact that they are free from safety and ethical constraints. Additionally, the mechanism that governs the global evolution and transmission of SARS-CoV-2 cannot be revealed from individual experiments and was discovered by integrating genotyping of massive viral sequences, biophysical modeling of protein-protein interactions, deep mutational data, deep learning, and advanced mathematics. There exists a tsunami of literature on the molecular modeling, simulations, and predictions of SARS-CoV-2 and related developments of drugs, vaccines, antibodies, and diagnostics. To provide readers with a quick update about this literature, we present a comprehensive and systematic methodology-centered review. Aspects such as molecular biophysics, bioinformatics, cheminformatics, machine learning, and mathematics are discussed. This review will be beneficial to researchers who are looking for ways to contribute to SARS-CoV-2 studies and those who are interested in the status of the field.
Collapse
Affiliation(s)
- Kaifu Gao
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Rui Wang
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Jiahui Chen
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Limei Cheng
- Clinical
Pharmacology and Pharmacometrics, Bristol
Myers Squibb, Princeton, New Jersey 08536, United States
| | - Jaclyn Frishcosy
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Yuta Huzumi
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Yuchi Qiu
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Tom Schluckbier
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Xiaoqi Wei
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Guo-Wei Wei
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
- Department
of Electrical and Computer Engineering, Michigan State University, East Lansing, Michigan 48824, United States
- Department
of Biochemistry and Molecular Biology, Michigan
State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
26
|
Soumah AA, Diallo MSK, Guichet E, Maman D, Thaurignac G, Keita AK, Bouillin J, Diallo H, Pelloquin R, Ayouba A, Kpamou C, Peeters M, Delaporte E, Etard JF, Toure A. High and Rapid Increase in Seroprevalence for SARS-CoV-2 in Conakry, Guinea: Results From 3 Successive Cross-Sectional Surveys (ANRS COV16-ARIACOV). Open Forum Infect Dis 2022; 9:ofac152. [PMID: 35493112 PMCID: PMC8992319 DOI: 10.1093/ofid/ofac152] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 03/21/2022] [Indexed: 11/14/2022] Open
Abstract
We conducted 3 successive seroprevalence surveys, 3 months apart, using multistage cluster sampling to measure the extent and dynamics of the severe acute respiratory syndrome coronavirus 2 epidemic in Conakry, the capital city of Guinea. Seroprevalence increased from 17.3% (95% CI, 12.4%-23.8%) in December 2020 during the first survey (S1) to 28.9% (95% CI, 25.6%-32.4%) in March/April 2021 (S2), then to 42.4% (95% CI, 39.5%-45.3%) in June 2021 (S3). This significant overall trend of increasing seroprevalence (P < .0001) was also significant in every age class, illustrating a sustained transmission within the whole community. These data may contribute to defining cost-effective response strategies.
Collapse
Affiliation(s)
- Abou Aissata Soumah
- Centre de Recherche et de Formation en Infectiologie de Guinée, Université Gamal Abdel Nasser de Conakry, Conakry, Guinea
| | - Mamadou Saliou Kalifa Diallo
- Centre de Recherche et de Formation en Infectiologie de Guinée, Université Gamal Abdel Nasser de Conakry, Conakry, Guinea
- Recherches Translationnelles sur le VIH et les Maladies Infectieuses (TransVIHMI), University of Montpellier, Institut de Recherche pour le Développement (IRD), Institut National de la Santé et de la Recherche Médicale (INSERM), Montpellier, France
| | - Emilande Guichet
- Recherches Translationnelles sur le VIH et les Maladies Infectieuses (TransVIHMI), University of Montpellier, Institut de Recherche pour le Développement (IRD), Institut National de la Santé et de la Recherche Médicale (INSERM), Montpellier, France
| | | | - Guillaume Thaurignac
- Recherches Translationnelles sur le VIH et les Maladies Infectieuses (TransVIHMI), University of Montpellier, Institut de Recherche pour le Développement (IRD), Institut National de la Santé et de la Recherche Médicale (INSERM), Montpellier, France
| | - Alpha Kabinet Keita
- Centre de Recherche et de Formation en Infectiologie de Guinée, Université Gamal Abdel Nasser de Conakry, Conakry, Guinea
- Recherches Translationnelles sur le VIH et les Maladies Infectieuses (TransVIHMI), University of Montpellier, Institut de Recherche pour le Développement (IRD), Institut National de la Santé et de la Recherche Médicale (INSERM), Montpellier, France
| | - Julie Bouillin
- Recherches Translationnelles sur le VIH et les Maladies Infectieuses (TransVIHMI), University of Montpellier, Institut de Recherche pour le Développement (IRD), Institut National de la Santé et de la Recherche Médicale (INSERM), Montpellier, France
| | - Haby Diallo
- Centre de Recherche et de Formation en Infectiologie de Guinée, Université Gamal Abdel Nasser de Conakry, Conakry, Guinea
| | - Raphael Pelloquin
- Recherches Translationnelles sur le VIH et les Maladies Infectieuses (TransVIHMI), University of Montpellier, Institut de Recherche pour le Développement (IRD), Institut National de la Santé et de la Recherche Médicale (INSERM), Montpellier, France
| | - Ahidjo Ayouba
- Recherches Translationnelles sur le VIH et les Maladies Infectieuses (TransVIHMI), University of Montpellier, Institut de Recherche pour le Développement (IRD), Institut National de la Santé et de la Recherche Médicale (INSERM), Montpellier, France
| | - Cece Kpamou
- Centre de Recherche et de Formation en Infectiologie de Guinée, Université Gamal Abdel Nasser de Conakry, Conakry, Guinea
| | - Martine Peeters
- Recherches Translationnelles sur le VIH et les Maladies Infectieuses (TransVIHMI), University of Montpellier, Institut de Recherche pour le Développement (IRD), Institut National de la Santé et de la Recherche Médicale (INSERM), Montpellier, France
| | - Eric Delaporte
- Recherches Translationnelles sur le VIH et les Maladies Infectieuses (TransVIHMI), University of Montpellier, Institut de Recherche pour le Développement (IRD), Institut National de la Santé et de la Recherche Médicale (INSERM), Montpellier, France
| | - Jean-Francois Etard
- Recherches Translationnelles sur le VIH et les Maladies Infectieuses (TransVIHMI), University of Montpellier, Institut de Recherche pour le Développement (IRD), Institut National de la Santé et de la Recherche Médicale (INSERM), Montpellier, France
| | - Abdoulaye Toure
- Centre de Recherche et de Formation en Infectiologie de Guinée, Université Gamal Abdel Nasser de Conakry, Conakry, Guinea
| |
Collapse
|
27
|
Ndongo FA, Guichet E, Mimbé ED, Ndié J, Pelloquin R, Varloteaux M, Esemu L, Mpoudi-Etame M, Lamare N, Edoul G, Wouambo RK, Djomsi DM, Tongo M, Tabala FN, Dongmo RK, Diallo MSK, Bouillin J, Thaurignac G, Ayouba A, Peeters M, Delaporte E, Bissek ACZK, Mpoudi-Ngolé E. Rapid Increase of Community SARS-CoV-2 Seroprevalence during Second Wave of COVID-19, Yaoundé, Cameroon. Emerg Infect Dis 2022; 28:1233-1236. [PMID: 35470795 PMCID: PMC9155890 DOI: 10.3201/eid2806.212580] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
We conducted 2 independent population-based SARS-CoV-2 serosurveys in Yaoundé, Cameroon, during January 27–February 6 and April 24–May 19, 2021. Overall age-standardized SARS-CoV-2 IgG seroprevalence increased from 18.6% in the first survey to 51.3% in the second (p<0.001). This finding illustrates high community transmission during the second wave of COVID-19.
Collapse
|
28
|
Jugwanth S, Gededzha MP, Mampeule N, Zwane N, David A, Burgers WA, Blackburn JM, Grove JS, George JA, Sanne I, Scott L, Stevens W, Mayne ES. Performance of the Abbott SARS-CoV-2 IgG serological assay in South African 2 patients. PLoS One 2022; 17:e0262442. [PMID: 35120133 PMCID: PMC8815965 DOI: 10.1371/journal.pone.0262442] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 12/23/2021] [Indexed: 12/21/2022] Open
Abstract
In late December 2019, pneumonia cases of unknown origin were reported in Wuhan, China. This virus was named SARS-CoV2 and the clinical syndrome was named coronavirus disease 19 (COVID-19). South Africa, despite strict and early lockdown has the highest infection rate in Africa. A key component of South Africa's response to SARSCoV2 was the rapid scale-up of diagnostic testing. The Abbott SARS-CoV2 assay detects IgG antibodies against the Nucleocapsid (N) protein of the SARS-CoV2 virus. This study undertook to validate and evaluate performance criteria of the Abbott assay and to establish whether this assay would show clinical utility in our population. Positive patients (n = 391) and negative controls (n = 139) were included. The Architect-i and Alinity-i systems were analyzers that were used to perform the SARS-CoV-2 IgG assay. In-house ELISA was incorporated into the study as a confirmatory serology test. A total of number of 530 participants was tested, 87% were symptomatic with infection and 13% were asymptomatic. When compared to RT-qPCR, the sensitivity of Architect and Alinity SARS-CoV2 assays was 69.5% and 64.8%, respectively. Specificity for Architect and Alinity assays was 95% and 90.3%, respectively. The Abbott assay was also compared to in house ELISA assay, with sensitivity for the Architect and Alinity assays of 94.7% and 92.5%, respectively. Specificity for Abbott Alinity assays was 91.7% higher than Abbott Architect 88.1%. Based on the current findings testing of IgG after 14 days is recommended in South Africa and supports other studies performed around the world.
Collapse
Affiliation(s)
- Sarika Jugwanth
- Department of Immunology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- National Health Laboratory Services, Johannesburg, South Africa
| | - Maemu P. Gededzha
- Department of Immunology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- National Health Laboratory Services, Johannesburg, South Africa
| | - Nakampe Mampeule
- Department of Immunology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- National Health Laboratory Services, Johannesburg, South Africa
| | - Nontobeko Zwane
- Department of Immunology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Anura David
- Department of Molecular Medicine and Haematology, School of Pathology, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| | - Wendy A. Burgers
- Division of Medical Virology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, Cape Town, South Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Jonathan M. Blackburn
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Divisions of Chemical and System Biology, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Jurette S. Grove
- National Health Laboratory Services, Johannesburg, South Africa
- Department of Chemical Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Jaya A. George
- National Health Laboratory Services, Johannesburg, South Africa
- Department of Chemical Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Ian Sanne
- Clinical HIV Research Unit, Department of Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Lesley Scott
- National Health Laboratory Services, Johannesburg, South Africa
- Department of Molecular Medicine and Haematology, School of Pathology, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| | - Wendy Stevens
- National Health Laboratory Services, Johannesburg, South Africa
- Department of Molecular Medicine and Haematology, School of Pathology, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| | - Elizabeth S. Mayne
- Department of Immunology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- National Health Laboratory Services, Johannesburg, South Africa
| |
Collapse
|
29
|
A SYSTEMATIC REVIEW AND META-ANALYSIS OF THE ACCURACY OF SARS-COV-2 IGM AND IGG TESTS IN INDIVIDUALS WITH COVID-19. J Clin Virol 2022; 148:105121. [PMID: 35245882 PMCID: PMC8863416 DOI: 10.1016/j.jcv.2022.105121] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 01/22/2022] [Accepted: 02/21/2022] [Indexed: 01/21/2023]
Abstract
Introduction Objective Methods Results Conclusion
Collapse
|
30
|
Simultaneous detection of antibody responses to multiple SARS-CoV-2 antigens by a Western blot serological assay. Appl Microbiol Biotechnol 2022; 106:8183-8194. [PMID: 36404356 PMCID: PMC9676789 DOI: 10.1007/s00253-022-12288-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/08/2022] [Accepted: 11/11/2022] [Indexed: 11/22/2022]
Abstract
The nucleic acid test is still the standard assessment for the diagnosis of coronavirus disease 2019 (COVID-19), which is caused by human infection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In addition to supporting the confirmation of disease cases, serological assays are used for the analysis of antibody status and epidemiological surveys. In this study, a single Western blot strip (WBS) coated with multiple Escherichia coli (E. coli)-expressed SARS-CoV-2 antigens was developed for comprehensive studies of antibody profiles in COVID-19 patient sera. The levels of specific antibodies directed to SARS-CoV-2 spike (S), S2, and nucleocapsid (N) proteins were gradually increased with the same tendency as the disease progressed after hospitalization. The signal readouts of S, S2, and N revealed by the multi-antigen-coated WBS (mWBS)-based serological assay (mWBS assay) also demonstrated a positive correlation with the SARS-CoV-2 neutralizing potency of the sera measured by the plaque reduction neutralization test (PRNT) assays. Surprisingly, the detection signals against the unstructured receptor-binding domain (RBD) purified from E. coli inclusion bodies were not observed, although the COVID-19 patient sera exhibited strong neutralizing potency in the PRNT assays, suggesting that the RBD-specific antibodies in patient sera mostly recognize the conformational epitopes. Furthermore, the mWBS assay identified a unique and major antigenic epitope at the residues 1148, 1149, 1152, 1155, and 1156 located within the 1127-1167 fragment of the S2 subunit, which was specifically recognized by the COVID-19 patient serum. The mWBS assay can be finished within 14-16 min by using the automatic platform of Western blotting by thin-film direct coating with suction (TDCS WB). Collectively, the mWBS assay can be applied for the analysis of antibody responses, prediction of the protective antibody status, and identification of the specific epitope. KEY POINTS: • A Western blot strip (WBS) coated with multiple SARS-CoV-2 antigens was developed for the serological assay. • The multi-antigen-coated WBS (mWBS) can be utilized for the simultaneous detection of antibody responses to multiple SARS-CoV-2 antigens. • The mWBS-based serological assay (mWBS assay) identified a unique epitope recognized by the COVID-19 patient serum.
Collapse
|
31
|
Exinger J, Hartard C, Lafferrière F, Fenninger C, Charbonnière LJ, Jeulin H. Evaluation of a Lateral Flow Immunoassay COVIDTECH ® SARS-CoV-2 IgM/IgG Antibody Rapid Test. Jpn J Infect Dis 2021; 75:334-340. [PMID: 34980703 DOI: 10.7883/yoken.jjid.2021.273] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The new epidemic coronavirus SARS-CoV-2 is responsible for severe respiratory illness (i.e. COVID-19). RT-PCR on respiratory samples is the gold standard in COVID-19 diagnosis, while serological tests may contribute to detect post-infection and post-vaccination immunity, and permit seroprevalence studies. The lateral flow immunoassay (LFIA) COVIDTECH® SARS-CoV-2 IgM/IgG Antibody Rapid Test that detects anti-SARS-CoV-2 IgM and IgG using a S-protein recombinant antigen has been independently evaluated in two laboratories. The specificity evaluated on 65 pre-pandemic samples reached 100% for IgM/IgG. Analyzing samples from patients with RT-PCR-confirmed infection, IgM/IgG antibodies were detected in 18/26 (69%) and 58/58 (100%) samples before day 13th and from the 14th day post-symptom onset respectively. Before the 14th post-symptom onset, the COVIDTECH test was less sensitive than another LFIA method (BioSynex BSS IgM/IgG) and a chemiluminescent Immunoassay (LIAISON® SARS-CoV-2 TrimericS IgG assay). Overall, this LFIA method is suitable for SARS-CoV-2 serological diagnosis, when the patient is > 14th day after onset of symptoms.
Collapse
Affiliation(s)
- Julien Exinger
- Laboratory of Medical Biology, Haguenau Hospital Center, France
| | - Cédric Hartard
- LCPME, Lorraine University, CNRS; Virology Laboratory, CHRU Nancy Brabois, France
| | | | | | - Loic J Charbonnière
- Synthesis for analysis team, IPHC, UMR 7178 CNRS/Strasbourg University, ECPM, France
| | - Hélène Jeulin
- LCPME, Lorraine University, CNRS; Virology Laboratory, CHRU Nancy Brabois, France
| |
Collapse
|
32
|
Keuning MW, Grobben M, de Groen AEC, Berman-de Jong EP, Bijlsma MW, Cohen S, Felderhof M, de Groof F, Molanus D, Oeij N, Rijpert M, van Eijk HWM, Koen G, van der Straten K, Oomen M, Visser R, Linty F, Steenhuis M, Vidarsson G, Rispens T, Plötz FB, van Gils MJ, Pajkrt D. Saliva SARS-CoV-2 Antibody Prevalence in Children. Microbiol Spectr 2021; 9:e0073121. [PMID: 34523985 PMCID: PMC8557814 DOI: 10.1128/spectrum.00731-21] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/12/2021] [Indexed: 12/23/2022] Open
Abstract
COVID-19 patients produce circulating and mucosal antibodies. In adults, specific saliva antibodies have been detected. Nonetheless, seroprevalence is routinely investigated, while little attention has been paid to mucosal antibodies. We therefore assessed SARS-CoV-2-specific antibody prevalence in serum and saliva in children in the Netherlands. We assessed SARS-CoV-2 antibody prevalence in serum and saliva of 517 children attending medical services in the Netherlands (irrespective of COVID-19 exposure) from April to October 2020. The prevalence of SARS-CoV-2 spike (S), receptor binding domain (RBD), and nucleocapsid (N)-specific IgG and IgA were evaluated with an exploratory Luminex assay in serum and saliva and with the Wantai SARS-CoV-2 RBD total antibody enzyme-linked immunosorbent assay in serum. Using the Wantai assay, the RBD-specific antibody prevalence in serum was 3.3% (95% confidence interval [CI]. 1.9 to 5.3%). With the Luminex assay, we detected heterogeneity between antibodies for S, RBD, and N antigens, as IgG and IgA prevalence ranged between 3.6 and 4.6% in serum and between 0 and 4.4% in saliva. The Luminex assay also revealed differences between serum and saliva, with SARS-CoV-2-specific IgG present in saliva but not in serum for 1.5 to 2.7% of all children. Using multiple antigen assays, the IgG prevalence for at least two out of three antigens (S, RBD, or N) in serum or saliva can be calculated as 3.8% (95% CI, 2.3 to 5.6%). Our study displays the heterogeneity of the SARS-CoV-2 antibody response in children and emphasizes the additional value of saliva antibody detection and the combined use of different antigens. IMPORTANCE Comprehending humoral immunity to SARS-CoV-2, including in children, is crucial for future public health and vaccine strategies. Others have suggested that mucosal antibody measurement could be an important and more convenient tool to evaluate humoral immunity compared to circulating antibodies. Nonetheless, seroprevalence is routinely investigated, while little attention has been paid to mucosal antibodies. We show the heterogeneity of SARS-CoV-2 antibodies, in terms of both antigen specificity and differences between circulating and mucosal antibodies, emphasizing the additional value of saliva antibody detection next to detection of antibodies in serum.
Collapse
Affiliation(s)
- Maya W. Keuning
- Department of Pediatric Infectious Diseases, Rheumatology, & Immunology, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Marloes Grobben
- Department of Medical Microbiology and Infection Prevention, Amsterdam Institute of Infection and Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Anne-Elise C. de Groen
- Department of Pediatric Infectious Diseases, Rheumatology, & Immunology, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Eveline P. Berman-de Jong
- Department of Pediatrics, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Merijn W. Bijlsma
- Department of Pediatrics, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Sophie Cohen
- Department of Pediatrics, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Mariet Felderhof
- Department of Pediatrics, Flevoziekenhuis, Almere, The Netherlands
| | - Femke de Groof
- Department of Pediatrics, Noordwest Ziekenhuisgroep, Alkmaar, The Netherlands
| | - Daniel Molanus
- Department of Pediatrics, Amstellandziekenhuis, Amstelveen, The Netherlands
| | - Nadia Oeij
- Department of Pediatrics, Amstellandziekenhuis, Amstelveen, The Netherlands
| | - Maarten Rijpert
- Department of Pediatrics, Zaans Medisch Centrum, Zaandam, The Netherlands
| | - Hetty W. M. van Eijk
- Department of Medical Microbiology and Infection Prevention, Amsterdam Institute of Infection and Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Gerrit Koen
- Department of Medical Microbiology and Infection Prevention, Amsterdam Institute of Infection and Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Karlijn van der Straten
- Department of Medical Microbiology and Infection Prevention, Amsterdam Institute of Infection and Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Melissa Oomen
- Department of Medical Microbiology and Infection Prevention, Amsterdam Institute of Infection and Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Remco Visser
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, The Netherlands
- Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Federica Linty
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, The Netherlands
- Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Maurice Steenhuis
- Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Immunopathology, Sanquin Research, Amsterdam, The Netherlands
| | - Gestur Vidarsson
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, The Netherlands
- Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Theo Rispens
- Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Immunopathology, Sanquin Research, Amsterdam, The Netherlands
| | - Frans B. Plötz
- Department of Pediatrics, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Pediatrics, Tergooi Hospital, Blaricum, The Netherlands
| | - Marit J. van Gils
- Department of Medical Microbiology and Infection Prevention, Amsterdam Institute of Infection and Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Dasja Pajkrt
- Department of Pediatric Infectious Diseases, Rheumatology, & Immunology, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
33
|
Larese Filon F, Purpuri A, Camata D, Bovenzi M, Rui F, Ronchese F, De Michieli P, Marcello A, Poggianella M, Confalonieri M, Salton F, Confalonieri P, Ruscio M, Belgrano A, Segat L, D'Agaro P, Negro C. Low sensitivity of rapid tests detecting anti-CoV-2 IgG and IgM in health care workers' serum for COVID-19 screening. LA MEDICINA DEL LAVORO 2021; 112:331-339. [PMID: 34726663 PMCID: PMC8600574 DOI: 10.23749/mdl.v112i5.11798] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/12/2021] [Indexed: 11/17/2022]
Abstract
Background: the sensitivity and specificity of a rapid antibody test were investigated for the screening of healthcare workers. Methods: the serum of 389 health care workers exposed to COVID-19 patients or with symptoms, were analysed. All workers underwent monthly the screening for SARS-CoV-2 with detection of viral RNA in nasopharyngeal swabs by RT-PCR. IgG antibody detection in serum was performed by Chemiluminescence Immunoassay (CLIA) and by the Rapid test (KHB diagnostic kit for SARS CoV-2 IgM/IgG antibody after a median of 7.6 weeks (25°-75° percentiles 6.6-11.5). Results: the rapid test resulted positive in 31/132 (23.5%), 16/135 (11.8%) and 0/122 cases in COVID-19 positive individuals, in those with only SARS-CoV-2 IgG antibodies and in those negative for both tests, respectively. Sensitivity was 17.6% (CI95% 13.2-22.7) and 23.5% (CI95% 16.5-31.6), and specificity was 100% (CI95% 97-100) and 100% (CI95% 97-100) considering Rapid test vs CLIA IgG or Rapid test vs SARS-CoV-2 positive RNA detection, respectively. Conclusion: the KHB Rapid test is not suitable for the screening of workers with previous COVID-19 infection.
Collapse
Affiliation(s)
| | - Antonio Purpuri
- a:1:{s:5:"en_US";s:59:"UCO Medicina del Lavoro, Università degli Studi di Trieste";}.
| | - Davide Camata
- Unit of Occupational Medicine, University of Trieste.
| | | | - Francesca Rui
- Unit of Occupational Medicine, University of Trieste.
| | | | | | - Alessandro Marcello
- Laboratory of Molecular Virology, International Centre for Genetic Engineering and Biotechnology (ICGEB), Padriciano 99, 34149 Trieste, Italy.
| | - Monica Poggianella
- Laboratory of Molecular Virology, International Centre for Genetic Engineering and Biotechnology (ICGEB), Padriciano 99, 34149 Trieste, Italy.
| | | | | | | | - Maurizio Ruscio
- Division of Laboratory Medicine, University Hospital Giuliano Isontina (ASU GI).
| | - Anna Belgrano
- Division of Laboratory Medicine, University Hospital Giuliano Isontina (ASU GI).
| | - Ludovica Segat
- Laboratory of Virology, Public Health Division, University of Trieste.
| | | | - Corrado Negro
- Unit of Occupational Medicine, University of Trieste.
| |
Collapse
|
34
|
Haneef K, Saleem R, Iqbal Khan MS, Adeyinka OS, Banday S, Asghar MU, Rahman ZU, Fatima Z. Severe acute respiratory syndrome coronavirus 2 targeted antibodies cocktail and B cell receptor interplay: interventions to trigger vaccine development. EXPLORATION OF IMMUNOLOGY 2021. [DOI: 10.37349/ei.2021.00011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/11/2021] [Indexed: 01/25/2024]
Abstract
Coronavirus disease-2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus (SARS-CoV)-2 spread globally and creates an alarming situation. Following the SARS-CoV-2 paradigm, therapeutic efficacy is achieved via repurposing several antiviral, antibacterial, and antimalarial drugs. Innate and adaptive immune cells work close to combat infection through the intricate production of antibodies (Abs) and inflammatory cytokines. As an essential component of the immune system, Abs play an important role in eliminating viruses and maintaining homeostasis. B lymphocytes (B cells) are effector cells, stringent to produce neutralizing Abs to combat infection. After recognizing SARS-CoV-2 antigens by a surface receptor called B cell receptors (BCRs) on the plasma membrane, the BCRs transmembrane signal transduction and immune activation results in Ab production and development of immune memory. Thus, it ensures that plasma B cells can quickly start an intricate immune response to generate efficient protective Abs to clear the pathogen. Nevertheless, considering therapeutic challenges in the context of the new coronavirus pandemic, this review addresses the molecular mechanism of the immune activation and function of novel SARS-CoV-2 specific B cells in the production of SARS-CoV-2 specific Abs. Additionally, these studies highlighted the Ab-mediated pathogenesis, the intriguing role of nano-scale signaling subunits, non-structural proteins during COVID-19 infection, and structural insights of SARS-CoV-2 specific Abs.
Collapse
Affiliation(s)
- Kabeer Haneef
- School of Life Science, Institute for Immunology, Tsinghua University, Beijing 100084, China
| | - Rabia Saleem
- Department of Pediatric Haematology, Oncology and BMT, The Children’s Hospital and Institute of Child Health, Lahore 53700, Pakistan
| | - Muhammad Saleem Iqbal Khan
- Department of Biochemistry, Cancer Institute of the second affiliated hospital, School of Medicine, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | | | - Sadeeq Banday
- School of Agriculture Sciences and Food Technology, Shandong University of Technology, Zibo 255000, Shandong, China
| | - Muhammad Umer Asghar
- 6National Institute for Biotechnology and Genetic engineering (NIBGE), Faisalabad 38000, Punjab Pakistan 7Pakistan Institute of Engineering and Applied Sciences (PIEAS), Nilore, Islamabad 45650, Pakistan
| | - Zia Ur Rahman
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore 53700, Pakistan
| | - Zainab Fatima
- Department of Psychology, University of Gujrat, Punjab 50991, Pakistan
| |
Collapse
|
35
|
Wang J, Li D, Zhou Q, Wiltse A, Zand MS. Antibody Mediated Immunity to SARS-CoV-2 and Human Coronaviruses: Multiplex Beads Assay and Volumetric Absorptive Microsampling to Generate Immune Repertoire Cartography. Front Immunol 2021; 12:696370. [PMID: 34386006 PMCID: PMC8353270 DOI: 10.3389/fimmu.2021.696370] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 06/21/2021] [Indexed: 12/20/2022] Open
Abstract
The COVID-19 pandemic is caused by SARS-CoV-2, a novel zoonotic coronavirus. Emerging evidence indicates that preexisting humoral immunity against other seasonal human coronaviruses (HCoVs) plays a critical role in the specific antibody response to SARS-CoV-2. However, current work to assess the effects of preexisting and cross-reactive anti-HCoVs antibodies has been limited. To address this issue, we have adapted our previously reported multiplex assay to simultaneously and quantitatively measure anti-HCoV antibodies. The full mPlex-CoV panel covers the spike (S) and nucleocapsid (N) proteins of three highly pathogenic HCoVs (SARS-CoV-1, SARS-CoV-2, MERS) and four human seasonal strains (OC43, HKU1, NL63, 229E). Combining this assay with volumetric absorptive microsampling (VAMS), we measured the anti-HCoV IgG, IgA, and IgM antibodies in fingerstick blood samples. The results demonstrate that the mPlex-CoV assay has high specificity and sensitivity. It can detect strain-specific anti-HCoV antibodies down to 0.1 ng/ml with 4 log assay range and with low intra- and inter-assay coefficients of variation (%CV). We also estimate multiple strain HCoVs IgG, IgA and IgM concentration in VAMS samples in three categories of subjects: pre-COVID-19 (n=21), post-COVID-19 convalescents (n=19), and COVID-19 vaccine recipients (n=14). Using metric multidimensional scaling (MDS) analysis, HCoVs IgG concentrations in fingerstick blood samples were well separated between the pre-COVID-19, post-COVID-19 convalescents, and COVID-19 vaccine recipients. In addition, we demonstrate how multi-dimensional scaling analysis can be used to visualize IgG mediated antibody immunity against multiple human coronaviruses. We conclude that the combination of VAMS and the mPlex-Cov assay is well suited to performing remote study sample collection under pandemic conditions to monitor HCoVs antibody responses in population studies.
Collapse
Affiliation(s)
- Jiong Wang
- Department of Medicine, Division of Nephrology, University of Rochester Medical Center, Rochester, NY, United States
| | - Dongmei Li
- Clinical and Translational Science Institute, University of Rochester Medical Center, Rochester, NY, United States
| | - Qian Zhou
- Department of Medicine, Division of Nephrology, University of Rochester Medical Center, Rochester, NY, United States
| | - Alexander Wiltse
- Department of Medicine, Division of Nephrology, University of Rochester Medical Center, Rochester, NY, United States
| | - Martin S. Zand
- Department of Medicine, Division of Nephrology, University of Rochester Medical Center, Rochester, NY, United States
- Clinical and Translational Science Institute, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
36
|
Hober S, Hellström C, Olofsson J, Andersson E, Bergström S, Jernbom Falk A, Bayati S, Mravinacova S, Sjöberg R, Yousef J, Skoglund L, Kanje S, Berling A, Svensson AS, Jensen G, Enstedt H, Afshari D, Xu LL, Zwahlen M, von Feilitzen K, Hanke L, Murrell B, McInerney G, Karlsson Hedestam GB, Lendel C, Roth RG, Skoog I, Svenungsson E, Olsson T, Fogdell-Hahn A, Lindroth Y, Lundgren M, Maleki KT, Lagerqvist N, Klingström J, Da Silva Rodrigues R, Muschiol S, Bogdanovic G, Arroyo Mühr LS, Eklund C, Lagheden C, Dillner J, Sivertsson Å, Havervall S, Thålin C, Tegel H, Pin E, Månberg A, Hedhammar M, Nilsson P. Systematic evaluation of SARS-CoV-2 antigens enables a highly specific and sensitive multiplex serological COVID-19 assay. Clin Transl Immunology 2021; 10:e1312. [PMID: 34295471 PMCID: PMC8288725 DOI: 10.1002/cti2.1312] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 06/20/2021] [Accepted: 06/22/2021] [Indexed: 12/14/2022] Open
Abstract
Objective The COVID-19 pandemic poses an immense need for accurate, sensitive and high-throughput clinical tests, and serological assays are needed for both overarching epidemiological studies and evaluating vaccines. Here, we present the development and validation of a high-throughput multiplex bead-based serological assay. Methods More than 100 representations of SARS-CoV-2 proteins were included for initial evaluation, including antigens produced in bacterial and mammalian hosts as well as synthetic peptides. The five best-performing antigens, three representing the spike glycoprotein and two representing the nucleocapsid protein, were further evaluated for detection of IgG antibodies in samples from 331 COVID-19 patients and convalescents, and in 2090 negative controls sampled before 2020. Results Three antigens were finally selected, represented by a soluble trimeric form and the S1-domain of the spike glycoprotein as well as by the C-terminal domain of the nucleocapsid. The sensitivity for these three antigens individually was found to be 99.7%, 99.1% and 99.7%, and the specificity was found to be 98.1%, 98.7% and 95.7%. The best assay performance was although achieved when utilising two antigens in combination, enabling a sensitivity of up to 99.7% combined with a specificity of 100%. Requiring any two of the three antigens resulted in a sensitivity of 99.7% and a specificity of 99.4%. Conclusion These observations demonstrate that a serological test based on a combination of several SARS-CoV-2 antigens enables a highly specific and sensitive multiplex serological COVID-19 assay.
Collapse
|
37
|
Nkuba AN, Makiala SM, Guichet E, Tshiminyi PM, Bazitama YM, Yambayamba MK, Kazenza BM, Kabeya TM, Matungulu EB, Baketana LK, Mitongo NM, Thaurignac G, Leendertz FH, Vanlerberghe V, Pelloquin R, Etard JF, Maman D, Mbala PK, Ayouba A, Peeters M, Muyembe JJT, Delaporte E, Ahuka SM. High prevalence of anti-SARS-CoV-2 antibodies after the first wave of COVID-19 in Kinshasa, Democratic Republic of the Congo: results of a cross-sectional household-based survey. Clin Infect Dis 2021; 74:882-890. [PMID: 34089598 PMCID: PMC8244674 DOI: 10.1093/cid/ciab515] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Indexed: 02/06/2023] Open
Abstract
Background In October 2020, after the first wave of coronavirus disease 2019 (COVID-19), only 8290 confirmed cases were reported in Kinshasa, Democratic Republic of the Congo, but the real prevalence remains unknown. To guide public health policies, we aimed to describe the prevalence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) immunoglobulin G (IgG) antibodies in the general population in Kinshasa. Methods We conducted a cross-sectional, household-based serosurvey between 22 October 2020 and 8 November 2020. Participants were interviewed at home and tested for antibodies against SARS-CoV-2 spike and nucleocapsid proteins in a Luminex-based assay. A positive serology was defined as a sample that reacted with both SARS-CoV-2 proteins (100% sensitivity, 99.7% specificity). The overall weighted, age-standardized prevalence was estimated and the infection-to-case ratio was calculated to determine the proportion of undiagnosed SARS-CoV-2 infections. Results A total of 1233 participants from 292 households were included (mean age, 32.4 years; 764 [61.2%] women). The overall weighted, age-standardized SARS-CoV-2 seroprevalence was 16.6% (95% CI: 14.0–19.5%). The estimated infection-to-case ratio was 292:1. Prevalence was higher among participants ≥40 years than among those <18 years (21.2% vs 14.9%, respectively; P < .05). It was also higher in participants who reported hospitalization than among those who did not (29.8% vs 16.0%, respectively; P < .05). However, differences were not significant in the multivariate model (P = .1). Conclusions The prevalence of SARS-CoV-2 is much higher than the number of COVID-19 cases reported. These results justify the organization of a sequential series of serosurveys by public health authorities to adapt response measures to the dynamics of the pandemic.
Collapse
Affiliation(s)
- Antoine N Nkuba
- TransVIHMI, Institut de Recherche pour le Développement, Institut National de la Santé et de la Recherche Médicale (INSERM), Montpellier University, Montpellier, France.,Département de Virologie, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo.,Département de Biologie Médicale, Université de Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Sheila M Makiala
- Département de Virologie, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo.,Département de Biologie Médicale, Université de Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Emilande Guichet
- TransVIHMI, Institut de Recherche pour le Développement, Institut National de la Santé et de la Recherche Médicale (INSERM), Montpellier University, Montpellier, France
| | - Paul M Tshiminyi
- Département de Virologie, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo
| | - Yannick M Bazitama
- Département de Virologie, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo.,Département de Biologie Médicale, Université de Kinshasa, Kinshasa, Democratic Republic of the Congo.,Center for Zoonosis Control, Graduate School of Infectious Diseases, Hokkaido University, Sapporo, Japan
| | - Marc K Yambayamba
- Département d'Epidémiologie et Statistiques, Ecole de Santé Publique, Université de Kinshasa
| | - Benito M Kazenza
- Département de Nutrition, Ecole de Santé Publique, Université de Kinshasa
| | - Trésor M Kabeya
- Département de Virologie, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo
| | - Elysee B Matungulu
- Département de Virologie, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo
| | - Lionel K Baketana
- Département de Virologie, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo
| | - Naomi M Mitongo
- Département de Virologie, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo
| | - Guillaume Thaurignac
- TransVIHMI, Institut de Recherche pour le Développement, Institut National de la Santé et de la Recherche Médicale (INSERM), Montpellier University, Montpellier, France
| | - Fabian H Leendertz
- Epidemiology of Highly Pathogenic Microorganisms Project Group, Robert Koch Institute, Berlin, Germany
| | - Veerle Vanlerberghe
- Tropical Infectious Diseases Unit, Department of Public Health, Antwerp, Belgium
| | - Raphaël Pelloquin
- TransVIHMI, Institut de Recherche pour le Développement, Institut National de la Santé et de la Recherche Médicale (INSERM), Montpellier University, Montpellier, France
| | - Jean-François Etard
- TransVIHMI, Institut de Recherche pour le Développement, Institut National de la Santé et de la Recherche Médicale (INSERM), Montpellier University, Montpellier, France.,Epigreen, Paris, France
| | | | - Placide K Mbala
- Département de Virologie, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo.,Département de Biologie Médicale, Université de Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Ahidjo Ayouba
- TransVIHMI, Institut de Recherche pour le Développement, Institut National de la Santé et de la Recherche Médicale (INSERM), Montpellier University, Montpellier, France
| | - Martine Peeters
- TransVIHMI, Institut de Recherche pour le Développement, Institut National de la Santé et de la Recherche Médicale (INSERM), Montpellier University, Montpellier, France
| | - Jean-Jacques T Muyembe
- Département de Virologie, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo.,Département de Biologie Médicale, Université de Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Eric Delaporte
- TransVIHMI, Institut de Recherche pour le Développement, Institut National de la Santé et de la Recherche Médicale (INSERM), Montpellier University, Montpellier, France
| | - Steve M Ahuka
- Département de Virologie, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo.,Département de Biologie Médicale, Université de Kinshasa, Kinshasa, Democratic Republic of the Congo
| |
Collapse
|
38
|
de Alwis R, Gan ES, Chen S, Leong YS, Tan HC, Zhang SL, Yau C, Low JGH, Kalimuddin S, Matsuda D, Allen EC, Hartman P, Park KJJ, Alayyoubi M, Bhaskaran H, Dukanovic A, Bao Y, Clemente B, Vega J, Roberts S, Gonzalez JA, Sablad M, Yelin R, Taylor W, Tachikawa K, Parker S, Karmali P, Davis J, Sullivan BM, Sullivan SM, Hughes SG, Chivukula P, Ooi EE. A single dose of self-transcribing and replicating RNA-based SARS-CoV-2 vaccine produces protective adaptive immunity in mice. Mol Ther 2021; 29:1970-1983. [PMID: 33823303 PMCID: PMC8019652 DOI: 10.1016/j.ymthe.2021.04.001] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/26/2021] [Accepted: 03/30/2021] [Indexed: 12/18/2022] Open
Abstract
A self-transcribing and replicating RNA (STARR)-based vaccine (LUNAR-COV19) has been developed to prevent SARS-CoV-2 infection. The vaccine encodes an alphavirus-based replicon and the SARS-CoV-2 full-length spike glycoprotein. Translation of the replicon produces a replicase complex that amplifies and prolongs SARS-CoV-2 spike glycoprotein expression. A single prime vaccination in mice led to robust antibody responses, with neutralizing antibody titers increasing up to day 60. Activation of cell-mediated immunity produced a strong viral antigen-specific CD8+ T lymphocyte response. Assaying for intracellular cytokine staining for interferon (IFN)γ and interleukin-4 (IL-4)-positive CD4+ T helper (Th) lymphocytes as well as anti-spike glycoprotein immunoglobulin G (IgG)2a/IgG1 ratios supported a strong Th1-dominant immune response. Finally, single LUNAR-COV19 vaccination at both 2 μg and 10 μg doses completely protected human ACE2 transgenic mice from both mortality and even measurable infection following wild-type SARS-CoV-2 challenge. Our findings collectively suggest the potential of LUNAR-COV19 as a single-dose vaccine.
Collapse
MESH Headings
- Alphavirus/genetics
- Alphavirus/immunology
- Angiotensin-Converting Enzyme 2/genetics
- Angiotensin-Converting Enzyme 2/immunology
- Animals
- Antibodies, Neutralizing/biosynthesis
- Antibodies, Viral/biosynthesis
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/virology
- COVID-19/immunology
- COVID-19/pathology
- COVID-19/prevention & control
- COVID-19/virology
- COVID-19 Vaccines/administration & dosage
- COVID-19 Vaccines/biosynthesis
- COVID-19 Vaccines/genetics
- COVID-19 Vaccines/immunology
- Female
- Gene Expression
- Humans
- Immunity, Cellular/drug effects
- Immunity, Humoral/drug effects
- Interferon-gamma/genetics
- Interferon-gamma/immunology
- Interleukin-4/genetics
- Interleukin-4/immunology
- Mice
- Mice, Transgenic
- Replicon/immunology
- SARS-CoV-2/drug effects
- SARS-CoV-2/immunology
- SARS-CoV-2/pathogenicity
- Spike Glycoprotein, Coronavirus/chemistry
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/immunology
- Th1 Cells/drug effects
- Th1 Cells/immunology
- Th1 Cells/virology
- Transgenes
- Treatment Outcome
- Vaccination/methods
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/biosynthesis
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- mRNA Vaccines
Collapse
Affiliation(s)
- Ruklanthi de Alwis
- Viral Research and Experimental Medicine Center, SingHealth Duke-NUS Academic Medical Center, Singapore, Singapore; Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Esther S Gan
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Shiwei Chen
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Yan Shan Leong
- Viral Research and Experimental Medicine Center, SingHealth Duke-NUS Academic Medical Center, Singapore, Singapore
| | - Hwee Cheng Tan
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Summer L Zhang
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Clement Yau
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Jenny G H Low
- Viral Research and Experimental Medicine Center, SingHealth Duke-NUS Academic Medical Center, Singapore, Singapore; Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore; Department of Infectious Disease, Singapore General Hospital, Singapore, Singapore
| | - Shirin Kalimuddin
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore; Department of Infectious Disease, Singapore General Hospital, Singapore, Singapore
| | - Daiki Matsuda
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, San Diego, CA 92121, USA
| | - Elizabeth C Allen
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, San Diego, CA 92121, USA
| | - Paula Hartman
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, San Diego, CA 92121, USA
| | | | - Maher Alayyoubi
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, San Diego, CA 92121, USA
| | - Hari Bhaskaran
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, San Diego, CA 92121, USA
| | - Adrian Dukanovic
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, San Diego, CA 92121, USA
| | - Yanjie Bao
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, San Diego, CA 92121, USA
| | - Brenda Clemente
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, San Diego, CA 92121, USA
| | - Jerel Vega
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, San Diego, CA 92121, USA
| | - Scott Roberts
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, San Diego, CA 92121, USA
| | - Jose A Gonzalez
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, San Diego, CA 92121, USA
| | - Marciano Sablad
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, San Diego, CA 92121, USA
| | - Rodrigo Yelin
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, San Diego, CA 92121, USA
| | - Wendy Taylor
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, San Diego, CA 92121, USA
| | - Kiyoshi Tachikawa
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, San Diego, CA 92121, USA
| | - Suezanne Parker
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, San Diego, CA 92121, USA
| | - Priya Karmali
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, San Diego, CA 92121, USA
| | - Jared Davis
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, San Diego, CA 92121, USA
| | - Brian M Sullivan
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, San Diego, CA 92121, USA
| | - Sean M Sullivan
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, San Diego, CA 92121, USA.
| | - Steve G Hughes
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, San Diego, CA 92121, USA
| | - Pad Chivukula
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, San Diego, CA 92121, USA
| | - Eng Eong Ooi
- Viral Research and Experimental Medicine Center, SingHealth Duke-NUS Academic Medical Center, Singapore, Singapore; Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
39
|
Heffron AS, McIlwain SJ, Amjadi MF, Baker DA, Khullar S, Armbrust T, Halfmann PJ, Kawaoka Y, Sethi AK, Palmenberg AC, Shelef MA, O’Connor DH, Ong IM. The landscape of antibody binding in SARS-CoV-2 infection. PLoS Biol 2021; 19:e3001265. [PMID: 34143766 PMCID: PMC8245122 DOI: 10.1371/journal.pbio.3001265] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 06/30/2021] [Accepted: 05/06/2021] [Indexed: 02/08/2023] Open
Abstract
The search for potential antibody-based diagnostics, vaccines, and therapeutics for pandemic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has focused almost exclusively on the spike (S) and nucleocapsid (N) proteins. Coronavirus membrane (M), ORF3a, and ORF8 proteins are humoral immunogens in other coronaviruses (CoVs) but remain largely uninvestigated for SARS-CoV-2. Here, we use ultradense peptide microarray mapping to show that SARS-CoV-2 infection induces robust antibody responses to epitopes throughout the SARS-CoV-2 proteome, particularly in M, in which 1 epitope achieved excellent diagnostic accuracy. We map 79 B cell epitopes throughout the SARS-CoV-2 proteome and demonstrate that antibodies that develop in response to SARS-CoV-2 infection bind homologous peptide sequences in the 6 other known human CoVs. We also confirm reactivity against 4 of our top-ranking epitopes by enzyme-linked immunosorbent assay (ELISA). Illness severity correlated with increased reactivity to 9 SARS-CoV-2 epitopes in S, M, N, and ORF3a in our population. Our results demonstrate previously unknown, highly reactive B cell epitopes throughout the full proteome of SARS-CoV-2 and other CoV proteins.
Collapse
Affiliation(s)
- Anna S. Heffron
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Sean J. McIlwain
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- University of Wisconsin Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Maya F. Amjadi
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - David A. Baker
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Saniya Khullar
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Tammy Armbrust
- Department of Pathobiological Sciences, School of Veterinary Medicine, Influenza Research Institute, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Peter J. Halfmann
- Department of Pathobiological Sciences, School of Veterinary Medicine, Influenza Research Institute, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Yoshihiro Kawaoka
- Department of Pathobiological Sciences, School of Veterinary Medicine, Influenza Research Institute, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Ajay K. Sethi
- Department of Population Health Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Ann C. Palmenberg
- Department of Biochemistry, Institute for Molecular Virology, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Miriam A. Shelef
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, United States of America
| | - David H. O’Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Irene M. Ong
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- University of Wisconsin Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Center for Human Genomics and Precision Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
40
|
Klüpfel J, Koros RC, Dehne K, Ungerer M, Würstle S, Mautner J, Feuerherd M, Protzer U, Hayden O, Elsner M, Seidel M. Automated, flow-based chemiluminescence microarray immunoassay for the rapid multiplex detection of IgG antibodies to SARS-CoV-2 in human serum and plasma (CoVRapid CL-MIA). Anal Bioanal Chem 2021; 413:5619-5632. [PMID: 33983466 PMCID: PMC8116441 DOI: 10.1007/s00216-021-03315-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/10/2021] [Accepted: 03/26/2021] [Indexed: 12/16/2022]
Abstract
In the face of the COVID-19 pandemic, the need for rapid serological tests that allow multiplexing emerged, as antibody seropositivity can instruct about individual immunity after an infection with SARS-CoV-2 or after vaccination. As many commercial antibody tests are either time-consuming or tend to produce false negative or false positive results when only one antigen is considered, we developed an automated, flow-based chemiluminescence microarray immunoassay (CL-MIA) that allows for the detection of IgG antibodies to SARS-CoV-2 receptor-binding domain (RBD), spike protein (S1 fragment), and nucleocapsid protein (N) in human serum and plasma in less than 8 min. The CoVRapid CL-MIA was tested with a set of 65 SARS-CoV-2 serology positive or negative samples, resulting in 100% diagnostic specificity and 100% diagnostic sensitivity, thus even outcompeting commercial tests run on the same sample set. Additionally, the prospect of future quantitative assessments (i.e., quantifying the level of antibodies) was demonstrated. Due to the fully automated process, the test can easily be operated in hospitals, medical practices, or vaccination centers, offering a valuable tool for COVID-19 serosurveillance. Graphical abstract.
Collapse
Affiliation(s)
- Julia Klüpfel
- Institute of Hydrochemistry, Chair of Analytical Chemistry and Water Chemistry, Technical University of Munich, Elisabeth-Winterhalter-Weg 6, 81377, Munich, Germany
| | - Rosa Carolina Koros
- Institute of Hydrochemistry, Chair of Analytical Chemistry and Water Chemistry, Technical University of Munich, Elisabeth-Winterhalter-Weg 6, 81377, Munich, Germany
| | - Kerstin Dehne
- ISAR Bioscience GmbH, Semmelweisstr. 5, 82152, Planegg, Germany
| | - Martin Ungerer
- ISAR Bioscience GmbH, Semmelweisstr. 5, 82152, Planegg, Germany
| | - Silvia Würstle
- Institute of Hydrochemistry, Chair of Analytical Chemistry and Water Chemistry, Technical University of Munich, Elisabeth-Winterhalter-Weg 6, 81377, Munich, Germany
| | - Josef Mautner
- Helmholtz Zentrum München, German Research Center for Environmental Health, Haematologikum, Research Unit Gene Vectors and Technical University of Munich, Children's Hospital, Marchioninistraße 25, 81377, Munich, Germany.,Institute of Virology, Technical University of Munich / Helmholtz Zentrum München, Trogerstr. 30, 81675, Munich, Germany
| | - Martin Feuerherd
- Institute of Virology, Technical University of Munich / Helmholtz Zentrum München, Trogerstr. 30, 81675, Munich, Germany
| | - Ulrike Protzer
- Institute of Virology, Technical University of Munich / Helmholtz Zentrum München, Trogerstr. 30, 81675, Munich, Germany.,German Center for Infection Research (DZIF), Munich partner site, 81675, Munich, Germany
| | - Oliver Hayden
- Heinz-Nixdorf-Chair for Biomedical Electronics, Technical University of Munich, TranslaTUM, Einsteinstr. 25, 81675, Munich, Germany
| | - Martin Elsner
- Institute of Hydrochemistry, Chair of Analytical Chemistry and Water Chemistry, Technical University of Munich, Elisabeth-Winterhalter-Weg 6, 81377, Munich, Germany
| | - Michael Seidel
- Institute of Hydrochemistry, Chair of Analytical Chemistry and Water Chemistry, Technical University of Munich, Elisabeth-Winterhalter-Weg 6, 81377, Munich, Germany.
| |
Collapse
|
41
|
Veyrenche N, Bolloré K, Pisoni A, Bedin A, Mondain A, Ducos J, Segondy M, Montes B, Pastor P, Morquin D, Makinson A, Le Moing V, Van de Perre P, Foulongne V, Tuaillon E. Diagnosis value of SARS-CoV-2 antigen/antibody combined testing using rapid diagnostic tests at hospital admission. J Med Virol 2021; 93:3069-3076. [PMID: 33554363 PMCID: PMC8013599 DOI: 10.1002/jmv.26855] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/16/2021] [Accepted: 02/01/2021] [Indexed: 12/23/2022]
Abstract
The implementation of rapid diagnostic tests (RDTs) may enhance the efficiency of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) testing, as RDTs are widely accessible and easy to use. The aim of this study was to evaluate the performance of a diagnosis strategy based on a combination of antigen and immunoglobulin M (IgM) or immunoglobulin G (IgG) serological RDTs. Plasma and nasopharyngeal samples were collected between 14 March and 11 April 2020 at hospital admission from 45 patients with reverse transcription polymerase chain reaction (RT-PCR) confirmed COVID-19 and 20 negative controls. SARS-CoV-2 antigen (Ag) was assessed in nasopharyngeal swabs using the Coris Respi-Strip. For IgM/IgG detection, SureScreen Diagnostics and Szybio Biotech RDTs were used in addition to laboratory assays (Abbott Alinity i SARS-CoV-2 IgG and Theradiag COVID-19 IgM enzyme-linked immunosorbent assay). Using the Ag RDT, 13 out of 45 (29.0%) specimens tested positive, the sensitivity was 87.0% for cycle threshold (Ct ) values ≤25% and 0% for Ct values greater than 25. IgG detection was associated with high Ct values and the amount of time after the onset of symptoms. The profile of isolated IgM on RDTs was more frequently observed during the first and second week after the onset of symptoms. The combination of Ag and IgM/IgG RDTs enabled the detection of up to 84.0% of COVID-19 confirmed cases at hospital admission. Antigen and antibody-based RDTs showed suboptimal performances when used alone. However when used in combination, they are able to identify most COVID-19 patients admitted in an emergency department.
Collapse
Affiliation(s)
- Nicolas Veyrenche
- Pathogenesis and Control of Chronic Infections, INSERM, Etablissement Français du Sang, CHU MontpellierUniversité de MontpellierMontpellierFrance
| | - Karine Bolloré
- Pathogenesis and Control of Chronic Infections, INSERM, Etablissement Français du Sang, CHU MontpellierUniversité de MontpellierMontpellierFrance
| | - Amandine Pisoni
- Pathogenesis and Control of Chronic Infections, INSERM, Etablissement Français du Sang, CHU MontpellierUniversité de MontpellierMontpellierFrance
| | - Anne‐Sophie Bedin
- Pathogenesis and Control of Chronic Infections, INSERM, Etablissement Français du Sang, CHU MontpellierUniversité de MontpellierMontpellierFrance
| | | | | | - Michel Segondy
- Pathogenesis and Control of Chronic Infections, INSERM, Etablissement Français du Sang, CHU MontpellierUniversité de MontpellierMontpellierFrance
| | | | | | - David Morquin
- Recherches Translationnelles sur le VIH et Maladies Infectieuses/INSERM U1175Institut de Recherche pour le Développement et Université de MontpellierMontpellierFrance
- Département de Maladies Infectieuses et TropicalesCentre Hospitalier Universitaire de MontpellierMontpellierFrance
| | - Alain Makinson
- Recherches Translationnelles sur le VIH et Maladies Infectieuses/INSERM U1175Institut de Recherche pour le Développement et Université de MontpellierMontpellierFrance
- Département de Maladies Infectieuses et TropicalesCentre Hospitalier Universitaire de MontpellierMontpellierFrance
| | - Vincent Le Moing
- Recherches Translationnelles sur le VIH et Maladies Infectieuses/INSERM U1175Institut de Recherche pour le Développement et Université de MontpellierMontpellierFrance
- Département de Maladies Infectieuses et TropicalesCentre Hospitalier Universitaire de MontpellierMontpellierFrance
| | - Philippe Van de Perre
- Pathogenesis and Control of Chronic Infections, INSERM, Etablissement Français du Sang, CHU MontpellierUniversité de MontpellierMontpellierFrance
| | - Vincent Foulongne
- Pathogenesis and Control of Chronic Infections, INSERM, Etablissement Français du Sang, CHU MontpellierUniversité de MontpellierMontpellierFrance
| | - Edouard Tuaillon
- Pathogenesis and Control of Chronic Infections, INSERM, Etablissement Français du Sang, CHU MontpellierUniversité de MontpellierMontpellierFrance
| |
Collapse
|
42
|
Yuen RR, Steiner D, Pihl RMF, Chavez E, Olson A, Smith EL, Baird LA, Korkmaz F, Urick P, Sagar M, Berrigan JL, Gummuluru S, Corley RB, Quillen K, Belkina AC, Mostoslavsky G, Rifkin IR, Kataria Y, Cappione AJ, Gao W, Lin NH, Bhadelia N, Snyder-Cappione JE. Novel ELISA Protocol Links Pre-Existing SARS-CoV-2 Reactive Antibodies With Endemic Coronavirus Immunity and Age and Reveals Improved Serologic Identification of Acute COVID-19 via Multi-Parameter Detection. Front Immunol 2021; 12:614676. [PMID: 33897682 PMCID: PMC8062931 DOI: 10.3389/fimmu.2021.614676] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 03/03/2021] [Indexed: 12/13/2022] Open
Abstract
The COVID-19 pandemic has drastically impacted work, economy, and way of life. Sensitive measurement of SARS-CoV-2 specific antibodies would provide new insight into pre-existing immunity, virus transmission dynamics, and the nuances of SARS-CoV-2 pathogenesis. To date, existing SARS-CoV-2 serology tests have limited utility due to insufficient reliable detection of antibody levels lower than what is typically present after several days of symptoms. To measure lower quantities of SARS-CoV-2 IgM, IgG, and IgA with higher resolution than existing assays, we developed a new ELISA protocol with a distinct plate washing procedure and timed plate development via use of a standard curve. Very low optical densities from samples added to buffer coated wells at as low as a 1:5 dilution are reported using this 'BU ELISA' method. Use of this method revealed circulating SARS-CoV-2 receptor binding domain (RBD) and nucleocapsid protein (N) reactive antibodies (IgG, IgM, and/or IgA) in 44 and 100 percent of pre-pandemic subjects, respectively, and the magnitude of these antibodies tracked with antibody levels of analogous viral proteins from endemic coronavirus (eCoV) strains. The disease status (HIV, SLE) of unexposed subjects was not linked with SARS-CoV-2 reactive antibody levels; however, quantities were significantly lower in subjects over 70 years of age compared with younger counterparts. Also, we measured SARS-CoV-2 RBD- and N- specific IgM, IgG, and IgA antibodies from 29 SARS-CoV-2 infected individuals at varying disease states, including 10 acute COVID-19 hospitalized subjects with negative serology results by the EUA approved Abbott IgG chemiluminescent microparticle immunoassay. Measurements of SARS-CoV-2 RBD- and N- specific IgM, IgG, IgA levels measured by the BU ELISA revealed higher signal from 9 of the 10 Abbott test negative COVID-19 subjects than all pre-pandemic samples for at least one antibody specificity/isotype, implicating improved serologic identification of SARS-CoV-2 infection via multi-parameter, high sensitive antibody detection. We propose that this improved ELISA protocol, which is straightforward to perform, low cost, and uses readily available commercial reagents, is a useful tool to elucidate new information about SARS-CoV-2 infection and immunity and has promising implications for improved detection of all analytes measurable by this platform.
Collapse
Affiliation(s)
- Rachel R Yuen
- Department of Microbiology, Boston University School of Medicine, Boston, MA, United States
| | - Dylan Steiner
- Department of Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Riley M F Pihl
- PiBS Program, Boston University School of Medicine, Boston, MA, United States
| | - Elizabeth Chavez
- Department of Microbiology, Boston University School of Medicine, Boston, MA, United States
| | - Alex Olson
- Section of Infectious Diseases, Department of Medicine, Boston Medical Center, Boston, MA, United States
| | - Erika L Smith
- Department of Microbiology, Boston University School of Medicine, Boston, MA, United States
| | - Lillia A Baird
- Department of Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Filiz Korkmaz
- Department of Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Patricia Urick
- Department of Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Manish Sagar
- Department of Microbiology, Boston University School of Medicine, Boston, MA, United States.,Department of Medicine, Boston University School of Medicine, Boston, MA, United States.,Section of Infectious Diseases, Department of Medicine, Boston Medical Center, Boston, MA, United States
| | - Jacob L Berrigan
- Department of Microbiology, Boston University School of Medicine, Boston, MA, United States
| | - Suryaram Gummuluru
- Department of Microbiology, Boston University School of Medicine, Boston, MA, United States
| | - Ronald B Corley
- Department of Microbiology, Boston University School of Medicine, Boston, MA, United States.,National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, United States
| | - Karen Quillen
- Department of Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Anna C Belkina
- Flow Cytometry Core Facility, Boston University School of Medicine, Boston, MA, United States.,Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Gustavo Mostoslavsky
- Center for Regenerative Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Ian R Rifkin
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston, MA, United States.,Renal Section, Department of Medicine, VA Boston Healthcare System, Boston, MA, United States
| | - Yachana Kataria
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, United States
| | | | - Wenda Gao
- Antagen Pharmaceuticals, Boston, MA, United States
| | - Nina H Lin
- Section of Infectious Diseases, Department of Medicine, Boston Medical Center, Boston, MA, United States
| | - Nahid Bhadelia
- Department of Medicine, Boston University School of Medicine, Boston, MA, United States.,National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, United States
| | | |
Collapse
|
43
|
Pérez-López B, Mir M. Commercialized diagnostic technologies to combat SARS-CoV2: Advantages and disadvantages. Talanta 2021; 225:121898. [PMID: 33592692 PMCID: PMC7713573 DOI: 10.1016/j.talanta.2020.121898] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 11/09/2020] [Accepted: 11/12/2020] [Indexed: 12/13/2022]
Abstract
The current situation of the Covid-19 pandemic is indicated by a huge number of infections, high lethality, and rapid spread. These circumstances have stopped the activity of almost the entire world, affecting severely the global economy. A rapid diagnosis of the Covid-19 and a generalized testing protocol is essential to fight against the pandemic and to maintain health control in the population. Principal biosensing and diagnostic technologies used to monitor the spread of the SARS-CoV-2 are based on specific genomic analysis and rapid immune tests, both with different technology platforms that include advantages and disadvantages. Most of the in vitro diagnosis companies are competing to be the first on validating under different regulations their technology for placing their platforms for Covid-19 detection as fast as possible in this big international market. A comprehensive analysis of the commercialized technologies for the genomic based sensing and the antibody/antigen detection methods devoted to Covid-19 diagnosis is described in this review, which have been detailed and listed under different countries regulations. The effectiveness of the described technologies throughout the different stages of the disease and a critical comparison of the emerging technologies in the market to counterattack this pandemic have been discussed.
Collapse
Affiliation(s)
- Briza Pérez-López
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC) Barcelona Institute of Science and Technology (BIST), 12 Baldiri Reixac 15-21, Barcelona, 08028, Spain
| | - Mònica Mir
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Monforte de Lemos 3-5, Pabellón 11, 28029, Madrid, Spain; Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC) Barcelona Institute of Science and Technology (BIST), 12 Baldiri Reixac 15-21, Barcelona, 08028, Spain; Department of Electronics and Biomedical Engineering, Universitat de Barcelona, Martí I Franquès 1, 08028, Barcelona, Spain.
| |
Collapse
|
44
|
Fotis C, Meimetis N, Tsolakos N, Politou M, Akinosoglou K, Pliaka V, Minia A, Terpos E, Trougakos IP, Mentis A, Marangos M, Panayiotakopoulos G, Dimopoulos MA, Gogos C, Spyridonidis A, Alexopoulos LG. Accurate SARS-CoV-2 seroprevalence surveys require robust multi-antigen assays. Sci Rep 2021; 11:6614. [PMID: 33758278 PMCID: PMC7988055 DOI: 10.1038/s41598-021-86035-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 03/03/2021] [Indexed: 12/18/2022] Open
Abstract
There is a plethora of severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) serological tests based either on nucleocapsid phosphoprotein (N), S1-subunit of spike glycoprotein (S1) or receptor binding domain (RBD). Although these single-antigen based tests demonstrate high clinical performance, there is growing evidence regarding their limitations in epidemiological serosurveys. To address this, we developed a Luminex-based multiplex immunoassay that detects total antibodies (IgG/IgM/IgA) against the N, S1 and RBD antigens and used it to compare antibody responses in 1225 blood donors across Greece. Seroprevalence based on single-antigen readouts was strongly influenced by both the antigen type and cut-off value and ranged widely [0.8% (95% CI 0.4-1.5%)-7.5% (95% CI 6.0-8.9%)]. A multi-antigen approach requiring partial agreement between RBD and N or S1 readouts (RBD&N|S1 rule) was less affected by cut-off selection, resulting in robust seroprevalence estimation [0.6% (95% CI 0.3-1.1%)-1.2% (95% CI 0.7-2.0%)] and accurate identification of seroconverted individuals.
Collapse
Affiliation(s)
- Christos Fotis
- Biomedical Systems Laboratory, National Technical University of Athens, Athens, Greece
| | - Nikolaos Meimetis
- Biomedical Systems Laboratory, National Technical University of Athens, Athens, Greece
| | | | - Marianna Politou
- Department of Clinical Therapeutics, Alexandra General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Karolina Akinosoglou
- Division of Infectious Diseases, Department of Internal Medicine, University Hospital of Patras, Patras, Greece
| | - Vaia Pliaka
- ProtATonce Ltd, Demokritos Science Park, Athens, Greece
| | | | - Evangelos Terpos
- Department of Clinical Therapeutics, Alexandra General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis P Trougakos
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Andreas Mentis
- Medicinal Microbiology Laboratory, Hellenic Pasteur Institute, Athens, Greece
| | - Markos Marangos
- Division of Infectious Diseases, Department of Internal Medicine, University Hospital of Patras, Patras, Greece
| | - George Panayiotakopoulos
- Pharmacology Laboratory, University of Patras, Patras, Greece
- National Public Health Organization, Athens, Greece
| | - Meletios A Dimopoulos
- Department of Clinical Therapeutics, Alexandra General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Charalampos Gogos
- Division of Infectious Diseases, Department of Internal Medicine, University Hospital of Patras, Patras, Greece
| | - Alexandros Spyridonidis
- Department of Internal Medicine, BMT Unit and CBMDP Donor Center, University of Patras, Patras, Greece.
| | - Leonidas G Alexopoulos
- Biomedical Systems Laboratory, National Technical University of Athens, Athens, Greece.
- ProtATonce Ltd, Demokritos Science Park, Athens, Greece.
| |
Collapse
|
45
|
Tian L, Elsheikh EB, Patrone PN, Kearsley AJ, Gaigalas AK, Inwood S, Lin-Gibson S, Esposito D, Wang L. Towards Quantitative and Standardized Serological and Neutralization Assays for COVID-19. Int J Mol Sci 2021; 22:2723. [PMID: 33800363 PMCID: PMC7962843 DOI: 10.3390/ijms22052723] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 03/04/2021] [Accepted: 03/05/2021] [Indexed: 11/16/2022] Open
Abstract
Quantitative and robust serology assays are critical measurements underpinning global COVID-19 response to diagnostic, surveillance, and vaccine development. Here, we report a proof-of-concept approach for the development of quantitative, multiplexed flow cytometry-based serological and neutralization assays. The serology assays test the IgG and IgM against both the full-length spike antigens and the receptor binding domain (RBD) of the spike antigen. Benchmarking against an RBD-specific SARS-CoV IgG reference standard, the anti-SARS-CoV-2 RBD antibody titer was quantified in the range of 37.6 µg/mL to 31.0 ng/mL. The quantitative assays are highly specific with no correlative cross-reactivity with the spike proteins of MERS, SARS1, OC43 and HKU1 viruses. We further demonstrated good correlation between anti-RBD antibody titers and neutralizing antibody titers. The suite of serology and neutralization assays help to improve measurement confidence and are complementary and foundational for clinical and epidemiologic studies.
Collapse
Affiliation(s)
- Linhua Tian
- Biosystems and Biomaterials Division, National Institute of Standards and Technology (NIST), Gaithersburg, MD 20899, USA; (L.T.); (E.B.E.); (A.K.G.); (S.I.); (S.L.-G.)
| | - Elzafir B. Elsheikh
- Biosystems and Biomaterials Division, National Institute of Standards and Technology (NIST), Gaithersburg, MD 20899, USA; (L.T.); (E.B.E.); (A.K.G.); (S.I.); (S.L.-G.)
| | - Paul N. Patrone
- Applied and Computational Mathematics Division, NIST, Gaithersburg, MD 20899, USA; (P.N.P.); (A.J.K.)
| | - Anthony J. Kearsley
- Applied and Computational Mathematics Division, NIST, Gaithersburg, MD 20899, USA; (P.N.P.); (A.J.K.)
| | - Adolfas K. Gaigalas
- Biosystems and Biomaterials Division, National Institute of Standards and Technology (NIST), Gaithersburg, MD 20899, USA; (L.T.); (E.B.E.); (A.K.G.); (S.I.); (S.L.-G.)
| | - Sarah Inwood
- Biosystems and Biomaterials Division, National Institute of Standards and Technology (NIST), Gaithersburg, MD 20899, USA; (L.T.); (E.B.E.); (A.K.G.); (S.I.); (S.L.-G.)
| | - Sheng Lin-Gibson
- Biosystems and Biomaterials Division, National Institute of Standards and Technology (NIST), Gaithersburg, MD 20899, USA; (L.T.); (E.B.E.); (A.K.G.); (S.I.); (S.L.-G.)
| | - Dominic Esposito
- Frederick National Laboratory for Cancer Research (FNLCR), Frederick, MD 21702, USA;
| | - Lili Wang
- Biosystems and Biomaterials Division, National Institute of Standards and Technology (NIST), Gaithersburg, MD 20899, USA; (L.T.); (E.B.E.); (A.K.G.); (S.I.); (S.L.-G.)
| |
Collapse
|
46
|
Serological antibody testing in the COVID-19 pandemic: their molecular basis and applications. Biochem Soc Trans 2021; 48:2851-2863. [PMID: 33170924 DOI: 10.1042/bst20200744] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/26/2020] [Accepted: 10/30/2020] [Indexed: 12/23/2022]
Abstract
The ongoing COVID-19 pandemic has placed an overwhelming burden on the healthcare system, and caused major disruption to the world economy. COVID-19 is caused by SARS-CoV-2, a novel coronavirus that leads to a variety of symptoms in humans, including cough, fever and respiratory failure. SARS-CoV-2 infection can trigger extensive immune responses, including the production of antibodies. The detection of antibody response by serological testing provides a supplementary diagnostic tool to molecular tests. We hereby present a succinct yet comprehensive review on the antibody response to SARS-CoV-2 infection, as well as molecular mechanisms behind the strengths and limitations of serological antibody tests. The presence of antibodies can be detected in patient sera within days post symptom onset. Serological tests demonstrate superior sensitivity to molecular tests in some periods of time during disease development. Compared with the molecular tests, serological tests can be used for point-of-care testing, providing faster results at a lower cost. Commercially available serological tests show variable sensitivity and specificity, and the molecular basis of these variabilities are analysed. We discuss assays of different complexities that are used to specifically quantitate neutralising antibodies against SARS-CoV-2, which has important implications for vaccine development and herd immunity. Furthermore, we discuss examples of successful applications of serological tests to contact tracing and community-level sero-surveying, which provide invaluable information for pandemic management and assessment.
Collapse
|
47
|
Jain S, Nehra M, Kumar R, Dilbaghi N, Hu T, Kumar S, Kaushik A, Li CZ. Internet of medical things (IoMT)-integrated biosensors for point-of-care testing of infectious diseases. Biosens Bioelectron 2021; 179:113074. [PMID: 33596516 PMCID: PMC7866895 DOI: 10.1016/j.bios.2021.113074] [Citation(s) in RCA: 128] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 02/01/2021] [Accepted: 02/02/2021] [Indexed: 02/06/2023]
Abstract
On global scale, the current situation of pandemic is symptomatic of increased incidences of contagious diseases caused by pathogens. The faster spread of these diseases, in a moderately short timeframe, is threatening the overall population wellbeing and conceivably the economy. The inadequacy of conventional diagnostic tools in terms of time consuming and complex laboratory-based diagnosis process is a major challenge to medical care. In present era, the development of point-of-care testing (POCT) is in demand for fast detection of infectious diseases along with “on-site” results that are helpful in timely and early action for better treatment. In addition, POCT devices also play a crucial role in preventing the transmission of infectious diseases by offering real-time testing and lab quality microbial diagnosis within minutes. Timely diagnosis and further treatment optimization facilitate the containment of outbreaks of infectious diseases. Presently, efforts are being made to support such POCT by the technological development in the field of internet of medical things (IoMT). The IoMT offers wireless-based operation and connectivity of POCT devices with health expert and medical centre. In this review, the recently developed POC diagnostics integrated or future possibilities of integration with IoMT are discussed with focus on emerging and re-emerging infectious diseases like malaria, dengue fever, influenza A (H1N1), human papilloma virus (HPV), Ebola virus disease (EVD), Zika virus (ZIKV), and coronavirus (COVID-19). The IoMT-assisted POCT systems are capable enough to fill the gap between bioinformatics generation, big rapid analytics, and clinical validation. An optimized IoMT-assisted POCT will be useful in understanding the diseases progression, treatment decision, and evaluation of efficacy of prescribed therapy.
Collapse
Affiliation(s)
- Shikha Jain
- Department of Bio and Nano Technology, Guru Jambheshwar University of Science and Technology, Hisar, Haryana, 125001, India
| | - Monika Nehra
- Department of Bio and Nano Technology, Guru Jambheshwar University of Science and Technology, Hisar, Haryana, 125001, India; Department of Mechanical Engineering, UIET, Panjab University, Chandigarh, 160014, India
| | - Rajesh Kumar
- Department of Mechanical Engineering, UIET, Panjab University, Chandigarh, 160014, India
| | - Neeraj Dilbaghi
- Department of Bio and Nano Technology, Guru Jambheshwar University of Science and Technology, Hisar, Haryana, 125001, India
| | - TonyY Hu
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Sandeep Kumar
- Department of Bio and Nano Technology, Guru Jambheshwar University of Science and Technology, Hisar, Haryana, 125001, India.
| | - Ajeet Kaushik
- NanoBioTech Laboratory, Health Systems Engineering, Department of Natural Sciences, Florida Polytechnic University, Lakeland, FL, 33805-8531, United States.
| | - Chen-Zhong Li
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, 70112, USA; Department of Biomedical Engineering, Florida International University, Miami, FL, 33174, USA.
| |
Collapse
|
48
|
Mariën J, Ceulemans A, Michiels J, Heyndrickx L, Kerkhof K, Foque N, Widdowson MA, Mortgat L, Duysburgh E, Desombere I, Jansens H, Van Esbroeck M, Ariën KK. Evaluating SARS-CoV-2 spike and nucleocapsid proteins as targets for antibody detection in severe and mild COVID-19 cases using a Luminex bead-based assay. J Virol Methods 2021; 288:114025. [PMID: 33227340 PMCID: PMC7678438 DOI: 10.1016/j.jviromet.2020.114025] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 11/16/2020] [Accepted: 11/18/2020] [Indexed: 01/09/2023]
Abstract
Large-scale serosurveillance of severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) will only be possible if serological tests are sufficiently reliable, rapid and affordable. Many assays are either labour-intensive and require specialised facilities (e.g. virus neutralization assays), or are expensive with suboptimal specificity (e.g. commercial ELISAs and RDTs). Bead-based assays offer a cost-effective alternative and allow for multiplexing to test for antibodies against multiple antigens and against other pathogens. Here, we compare the performance of spike (S) and nucleocapsid (NP) antigens for the detection of SARS-CoV-2 specific IgG, IgM and IgA antibodies in a panel of sera that includes recent (up to six weeks after symptom onset, severe n = 44; and mild cases n = 52) and old infections (five months after symptom onset, mild n = 104), using a Luminex-bead based assay and comparison to a virus neutralization test. While we show that neutralizing antibody levels are significantly lower in mild than in severe cases, we demonstrate that a combination of the recombinant nucleocapsid protein (NP) and receptor-binding domain (RBD) results in highly specific (99 %) IgG antibody detection five months after infection in 96 % of cases. Although most severe Covid-19 cases developed a clear IgM and IgA response, titers fell below the detection threshold in more than 20 % of mild cases in our bead-based assay. In conclusion, our data supports the use of RBD and NP for the development of SARS-CoV-2 serological IgG bead-based assays.
Collapse
Affiliation(s)
- Joachim Mariën
- Outbreak Research Team, Institute of Tropical Medicine, Antwerp, Belgium.
| | - Ann Ceulemans
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Johan Michiels
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Leo Heyndrickx
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Karen Kerkhof
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Nikki Foque
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | | | - Laure Mortgat
- Epidemiology and Public Health, Sciensano, Brussels, Belgium
| | - Els Duysburgh
- Epidemiology and Public Health, Sciensano, Brussels, Belgium
| | | | | | - Marjan Van Esbroeck
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Kevin K Ariën
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium; University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
49
|
Shen H, Forgacs D, Chapla D, Moremen KW, Wells L, Hamer SA, Tompkins SM, Ross TM, Rouphael N, Edupuganti S, Collins MH, Tarleton RL. A flexible, pan-species, multi-antigen platform for the detection and monitoring of SARS-CoV-2-specific antibody responses. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.01.20.21249279. [PMID: 33532799 PMCID: PMC7852250 DOI: 10.1101/2021.01.20.21249279] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The SARS-CoV-2 pandemic and the vaccination effort that is ongoing has created an unmet need for accessible, affordable, flexible and precise platforms for monitoring the induction, specificity and maintenance of virus-specific immune responses. Herein we validate a multiplex (Luminex-based) assay capable of detecting SARS-CoV-2-specific antibodies irrespective of host species, antibody isotype, and specimen type (e.g. plasma, serum, saliva or blood spots). The well-established precision of Luminex-based assays provides the ability to follow changes in antibody levels over time to many antigens, including multiple permutations of the most common SARS-CoV-2 antigens. This platform can easily measure antibodies known to correlate with neutralization activity as well as multiple non-SARS-CoV-2 antigens such as vaccines (e.g. Tetanus toxoid) or those from frequently encountered agents (influenza), which serve as stable reference points for quantifying the changing SARS-specific responses. All of the antigens utilized in our study can be made in-house, many in E. coli using readily available plasmids. Commercially sourced antigens may also be incorporated and newly available antigen variants can be rapidly produced and integrated, making the platform adaptable to the evolving viral strains in this pandemic.
Collapse
Affiliation(s)
- Huifeng Shen
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens GA, USA
| | - David Forgacs
- Center for Vaccines and Immunology, University of Georgia, Athens GA, USA
| | - Digantkumar Chapla
- Complex Carbohydrate Research Center, University of Georgia, Athens GA, USA
| | - Kelley W. Moremen
- Complex Carbohydrate Research Center, University of Georgia, Athens GA, USA
- Department of Biochemistry, University of Georgia, Athens GA, USA
| | - Lance Wells
- Complex Carbohydrate Research Center, University of Georgia, Athens GA, USA
- Department of Biochemistry, University of Georgia, Athens GA, USA
| | - Sarah A. Hamer
- College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station TX, USA
| | - Stephen M. Tompkins
- Center for Vaccines and Immunology, University of Georgia, Athens GA, USA
- Department of Infectious Diseases, University of Georgia, Athens GA, USA
| | - Ted M. Ross
- Center for Vaccines and Immunology, University of Georgia, Athens GA, USA
- Department of Infectious Diseases, University of Georgia, Athens GA, USA
| | - Nadine Rouphael
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Decatur, Georgia, USA
| | - Srilatha Edupuganti
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Decatur, Georgia, USA
| | - Matthew H. Collins
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Decatur, Georgia, USA
| | - Rick L. Tarleton
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens GA, USA
- Department of Cellular Biology, University of Georgia, Athens GA, USA
| |
Collapse
|
50
|
Heffron AS, McIlwain SJ, Amjadi MF, Baker DA, Khullar S, Sethi AK, Palmenberg AC, Shelef MA, O'Connor DH, Ong IM. The landscape of antibody binding in SARS-CoV-2 infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2020.10.10.334292. [PMID: 33052349 PMCID: PMC7553183 DOI: 10.1101/2020.10.10.334292] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The search for potential antibody-based diagnostics, vaccines, and therapeutics for pandemic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has focused almost exclusively on the spike (S) and nucleocapsid (N) proteins. Coronavirus membrane (M), ORF3a, and ORF8 proteins are humoral immunogens in other coronaviruses (CoVs) but remain largely uninvestigated for SARS-CoV-2. Here we use ultradense peptide microarray mapping to show that SARS-CoV-2 infection induces robust antibody responses to epitopes throughout the SARS-CoV-2 proteome, particularly in M, in which one epitope achieved excellent diagnostic accuracy. We map 79 B cell epitopes throughout the SARS-CoV-2 proteome and demonstrate that antibodies that develop in response to SARS-CoV-2 infection bind homologous peptide sequences in the six other known human CoVs. We also confirm reactivity against four of our top-ranking epitopes by enzyme-linked immunosorbent assay (ELISA). Illness severity correlated with increased reactivity to nine SARS-CoV-2 epitopes in S, M, N, and ORF3a in our population. Our results demonstrate previously unknown, highly reactive B cell epitopes throughout the full proteome of SARS-CoV-2 and other CoV proteins.
Collapse
|