101
|
Theel ES. Performance Characteristics of High-Throughput Serologic Assays for Severe Acute Respiratory Syndrome Coronavirus 2 with Food and Drug Administration Emergency Use Authorization: A Review. Clin Lab Med 2022; 42:15-29. [PMID: 35153046 PMCID: PMC8563341 DOI: 10.1016/j.cll.2021.10.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
This review provides a broad summary of the performance characteristics of high-throughput severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) serologic assays with Food and Drug Administration Emergency Use Authorization, which are commonly found in central clinical laboratories. In addition, this review discusses the current roles of serologic testing for SARS-CoV-2 and provides a perspective for the future.
Collapse
|
102
|
Sherman AC, Desjardins M, Baden LR. Vaccine-Induced Severe Acute Respiratory Syndrome Coronavirus 2 Antibody Response and the Path to Accelerating Development (Determining a Correlate of Protection). Clin Lab Med 2022; 42:111-128. [PMID: 35153045 PMCID: PMC8563351 DOI: 10.1016/j.cll.2021.10.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
As new public health challenges relating to COVID-19 emerge, such as variant strains, waning vaccine efficacy over time, and decreased vaccine efficacy for special populations (immunocompromised hosts), it is important to determine a correlate of protection (CoP) to allow accurate bridging studies for special populations and against variants of concern. Large-scale phase 3 clinical trials are inefficient to rapidly assess novel vaccine candidates for variant strains or special populations, because these trials are slow and costly. Defining a practical CoP will aid in efficiently conducting future assessments to further describe protection for individuals and on a population level for surveillance.
Collapse
Affiliation(s)
- Amy C. Sherman
- Division of Infectious Diseases, Brigham and Women’s Hospital, 75 Francis Street, Boston, MA 02115, USA,Harvard Medical School, Boston, MA 02115, USA,Corresponding author. Division of Infectious Diseases, Brigham and Women’s Hospital, 75 Francis Street, Boston, MA 02115, USA
| | - Michaël Desjardins
- Division of Infectious Diseases, Brigham and Women’s Hospital, 75 Francis Street, Boston, MA 02115, USA,Harvard Medical School, Boston, MA 02115, USA,Division of Infectious Diseases, Centre Hospitalier de l’Université de Montréal, 1000 Rue Saint-Denis, Bureau F06.1102b, Montreal, Quebec H2X 0C1, Canada
| | - Lindsey R. Baden
- Division of Infectious Diseases, Brigham and Women’s Hospital, 75 Francis Street, Boston, MA 02115, USA,Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
103
|
Freeman J, Olson K, Conklin J, Shalhoub V, Johnson BA, Bopp NE, Fernandez D, Menachery VD, Aguilar PV. Analytical characterization of the SARS-CoV-2 EURM-017 reference material. Clin Biochem 2022; 101:19-25. [PMID: 34933006 PMCID: PMC8684092 DOI: 10.1016/j.clinbiochem.2021.12.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND Current serological methods for SARS-CoV-2 lack adequate standardization to a universal standard reference material. Standardization will allow comparison of results across various lab-developed and commercial assays and publications. SARS-CoV-2 EURM-017 is human sera reference material containing antibodies directed against SARS-CoV-2 proteins, S1/S2 (full-length spike [S]), S1 receptor-binding domain (S1 RBD), S1, S2, and nucleocapsid (N) protein. The goal of this study was to characterize five antigen-specific serum fractions in EURM-017 for standardization of serology assays. METHODS Five antigen-specific serum fractions were affinity purified, quantified, and PRNT50 titers compared. Standardization methods were established for two anti-S1 RBD (IgG and Total Ig) and one N protein assay. For the anti-S1 RBD assays, standardization involved determining assay index values for serial dilutions of S1-RBD anti-sera. Index values for the anti-S1 RBD IgG assay and PRNT50 titers were determined for 44 symptomatic COVID-19 patient sera. The index values were converted to EURM-017 ug/mL. RESULTS Anti-sera protein content was as follows: S1 (17.7 µg/mL), S1 RBD (17.4 µg/mL), S1/S2 (full-length S) (34.1 µg/mL), S2 (29.7 µg/mL), and N protein (72.5 µg/mL). S1 anti-serum had the highest neutralization activity. A standardization method for S1 RBD anti-serum and an anti-S1 RBD IgG assay yielded the linear equation (y = 0.75x-0.10; y = index, x=µg/mL anti-serum). Patient sample index values for the S1-RBD IgG assay correlated well with PRNT50 titers (Pearson r = 0.84). Using the equation above, patient index values were converted to standardized µg/mL. CONCLUSIONS Standardization of different lab-developed and commercial assays to EURM-017 antigen-specific anti-sera will allow comparison of results across studies globally due to traceability to a single standard reference material.
Collapse
Affiliation(s)
- James Freeman
- Siemens Healthcare Diagnostics, 511 Benedict Ave, Tarrytown, NY 10591, USA.
| | - Kalen Olson
- Siemens Healthcare Diagnostics, 511 Benedict Ave, Tarrytown, NY 10591, USA
| | - Justin Conklin
- Siemens Healthcare Diagnostics, 511 Benedict Ave, Tarrytown, NY 10591, USA
| | - Victoria Shalhoub
- Siemens Healthcare Diagnostics, 511 Benedict Ave, Tarrytown, NY 10591, USA
| | - Bryan A Johnson
- Department of Microbiology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA
| | - Nathen E Bopp
- Department of Pathology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA
| | - Diana Fernandez
- Department of Pathology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA
| | - Vineet D Menachery
- Department of Microbiology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA
| | - Patricia V Aguilar
- Department of Pathology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA
| |
Collapse
|
104
|
Romero-Pinedo S, Quesada M, Horndler L, Álvarez-Fernández S, Olmo A, Abia D, Alarcón B, Delgado P. Vaccine Type-, Age- and Past Infection-Dependence of the Humoral Response to SARS-CoV-2 Spike S Protein. Front Immunol 2022; 13:809285. [PMID: 35296086 PMCID: PMC8918633 DOI: 10.3389/fimmu.2022.809285] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 02/07/2022] [Indexed: 12/20/2022] Open
Abstract
The emergence of COVID-19 has led to a worldwide challenge for the rapid development of vaccines. Several types of safe and effective vaccines have been available in a time frame never seen before. Now that several hundred million people have been vaccinated there is an opportunity to compare vaccines in terms of protection and immune response. Here, we have applied a highly sensitive multiplexed flow cytometry method to measure simultaneously IgM, IgG1 and IgA anti-spike protein antibodies generated in response to three vaccines: ChAdOx1 (Oxford-AstraZeneca), mRNA-1273 (Moderna), and BNT162b2 (Pfizer-BioNTech). We have found that mRNA vaccines (mRNA-1273 and BNT162b2) induce a stronger humoral response, both after the first and the second dose, than the adenovirus-based ChAdOx1 vaccine. We also found that, in the elderly, antibody titers negatively correlate with the age of the donor but, also, that antibody titers remain stable for at least 6 months after complete vaccination. Finally, we found that one dose of BNT162b2 is sufficient to induce the highest antibody titers in seropositive pre-vaccination donors. We hope these data will help to guide future decisions on vaccination strategies.
Collapse
Affiliation(s)
| | | | - Lydia Horndler
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid, Madrid, Spain
| | | | | | - David Abia
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid, Madrid, Spain
| | - Balbino Alarcón
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid, Madrid, Spain
| | - Pilar Delgado
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
105
|
Basheer A, Kanungo R, Ratnam VJ, Kandasamy R. Immunoglobulin G Antibodies to SARS-CoV-2 Among Healthcare Workers at a Tertiary Care Center in South India. Cureus 2022; 14:e22520. [PMID: 35345731 PMCID: PMC8956499 DOI: 10.7759/cureus.22520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2022] [Indexed: 11/07/2022] Open
Abstract
Introduction Healthcare workers (HCWs) are at risk of exposure to SARS-CoV-2. Seroprevalence in this group may offer insights into trends to monitor and revise strategies to prevent transmission. Methods A cross-sectional study was conducted in two phases among healthcare workers at a tertiary care center to detect IgG antibodies to SARS-CoV-2. Seropositivity was calculated during both phases, and possible associations were determined using regression analysis. Results A total of 382 and 168 HCWs took part in the two phases, respectively. IgG antibodies were detected in 13 of 382 (3.4%; 95% confidence interval (CI): 2%-5.7%) and 71 of 168 (42.3%) participants in the first and second phases, respectively. Receiving at least one dose of vaccine (p < 0.001) and age (p = 0.028) were factors associated with the presence of antibodies, while gender, job type, exposure to COVID-19 cases, and comorbidities were not associated with seropositivity. Conclusion Serosurveys among HCWs may help identify transmission patterns and redesign infection control practices in the healthcare setting.
Collapse
|
106
|
Zhang S, Xu K, Li C, Zhou L, Kong X, Peng J, Zhu F, Bao C, Jin H, Gao Q, Zhao X, Zhu L. Long-Term Kinetics of SARS-CoV-2 Antibodies and Impact of Inactivated Vaccine on SARS-CoV-2 Antibodies Based on a COVID-19 Patients Cohort. Front Immunol 2022; 13:829665. [PMID: 35154152 PMCID: PMC8828498 DOI: 10.3389/fimmu.2022.829665] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 01/10/2022] [Indexed: 12/14/2022] Open
Abstract
Background Understanding the long-term kinetic characteristics of SARS-CoV-2 antibodies and the impact of inactivated vaccines on SARS-CoV-2 antibodies in convalescent patients can provide information for developing and improving vaccination strategies in such populations. Methods In this cohort, 402 convalescent patients who tested positive for SARS-CoV-2 by RT-PCR from 1 January to 22 June 2020 in Jiangsu, China, were enrolled. The epidemiological data included demographics, symptom onset, and vaccination history. Blood samples were collected and tested for antibody levels of specific IgG, IgM, RBD-IgG, S-IgG, and neutralizing antibodies using a the commercial magnetic chemiluminescence enzyme immunoassay. Results The median follow-up time after symptom onset was 15.6 months (IQR, 14.6 to 15.8). Of the 402 convalescent patients, 44 (13.84%) received an inactivated vaccine against COVID-19. A total of 255 (80.19%) patients were IgG-positive and 65 (20.44%) were IgM-positive. The neutralizing antibody was 83.02%. Compared with non-vaccinated individuals, the IgG antibody levels in vaccinated people were higher (P=0.007). Similarly, antibody levels for RBD-IgG, S-IgG, and neutralizing antibodies were all highly increased in vaccinated individuals (P<0.05). IgG levels were significantly higher after vaccination than before vaccination in the same population. IgG levels in those who received ‘single dose and ≥14d’ were similar to those with two doses (P>0.05). Similar conclusions were drawn for RBD-IgG and the neutralizing antibody. Conclusion 15.6 months after symptom onset, the majority of participants remained positive for serum-specific IgG, RBD-IgG, S-IgG, and neutralizing antibodies. For convalescent patients, a single dose of inactivated vaccine against COVID-19 can further boost antibody titres.
Collapse
Affiliation(s)
- Shihan Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, China.,Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Ke Xu
- Department of Acute Infectious Disease Control and Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Chuchu Li
- Department of Acute Infectious Disease Control and Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Lu Zhou
- Department of Acute Infectious Disease Control and Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Xiaoxiao Kong
- Department of Acute Infectious Disease Control and Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Jiefu Peng
- Department of Acute Infectious Disease Control and Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Fengcai Zhu
- Department of Acute Infectious Disease Control and Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China.,National Health Commission (NHC) Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China.,Key Laboratory of Infectious Diseases, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Changjun Bao
- Department of Acute Infectious Disease Control and Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Hui Jin
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, China.,Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Qiang Gao
- Department of Acute Infectious Disease Control and Prevention, Huai'an Center for Disease Control and Prevention, Huaian, China
| | - Xing Zhao
- Department of Acute Infectious Disease Control and Prevention, Lianyungang Center for Disease Control and Prevention, Lianyungang, China
| | - Liguo Zhu
- Department of Acute Infectious Disease Control and Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China.,National Health Commission (NHC) Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China.,Key Laboratory of Infectious Diseases, School of Public Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
107
|
Comparative Investigation of Methods for Analysis of SARS-CoV-2-Spike-Specific Antisera. Viruses 2022; 14:v14020410. [PMID: 35216003 PMCID: PMC8879086 DOI: 10.3390/v14020410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/24/2022] [Accepted: 02/14/2022] [Indexed: 11/17/2022] Open
Abstract
In light of an increasing number of vaccinated and convalescent individuals, there is a major need for the development of robust methods for the quantification of neutralizing antibodies; although, a defined correlate of protection is still missing. Sera from hospitalized COVID-19 patients suffering or not suffering from acute respiratory distress syndrome (ARDS) were comparatively analyzed by plaque reduction neutralization test (PRNT) and pseudotype-based neutralization assays to quantify their neutralizing capacity. The two neutralization assays showed comparable data. In case of the non-ARDS sera, there was a distinct correlation between the data from the neutralization assays on the one hand, and enzyme-linked immune sorbent assay (ELISA), as well as biophysical analyses, on the other hand. As such, surface plasmon resonance (SPR)-based assays for quantification of binding antibodies or analysis of the stability of the antigen–antibody interaction and inhibition of syncytium formation, determined by cell fusion assays, were performed. In the case of ARDS sera, which are characterized by a significantly higher fraction of RBD-binding IgA antibodies, there is a clear correlation between the neutralization assays and the ELISA data. In contrast to this, a less clear correlation between the biophysical analyses on the one hand and ELISAs and neutralization assays on the other hand was observed, which might be explained by the heterogeneity of the antibodies. To conclude, for less complex immune sera—as in cases of non-ARDS sera—combinations of titer quantification by ELISA with inhibition of syncytium formation, SPR-based analysis of antibody binding, determination of the stability of the antigen–antibody complex, and competition of the RBD-ACE2 binding represent alternatives to the classic PRNT for analysis of the neutralizing potential of SARS-CoV-2-specific sera, without the requirement for a BSL3 facility.
Collapse
|
108
|
Dulovic A, Kessel B, Harries M, Becker M, Ortmann J, Griesbaum J, Jüngling J, Junker D, Hernandez P, Gornyk D, Glöckner S, Melhorn V, Castell S, Heise JK, Kemmling Y, Tonn T, Frank K, Illig T, Klopp N, Warikoo N, Rath A, Suckel C, Marzian AU, Grupe N, Kaiser PD, Traenkle B, Rothbauer U, Kerrinnes T, Krause G, Lange B, Schneiderhan-Marra N, Strengert M. Comparative Magnitude and Persistence of Humoral SARS-CoV-2 Vaccination Responses in the Adult Population in Germany. Front Immunol 2022; 13:828053. [PMID: 35251012 PMCID: PMC8888837 DOI: 10.3389/fimmu.2022.828053] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/17/2022] [Indexed: 12/01/2022] Open
Abstract
Recent increases in SARS-CoV-2 infections have led to questions about duration and quality of vaccine-induced immune protection. While numerous studies have been published on immune responses triggered by vaccination, these often focus on studying the impact of one or two immunisation schemes within subpopulations such as immunocompromised individuals or healthcare workers. To provide information on the duration and quality of vaccine-induced immune responses against SARS-CoV-2, we analyzed antibody titres against various SARS-CoV-2 antigens and ACE2 binding inhibition against SARS-CoV-2 wild-type and variants of concern in samples from a large German population-based seroprevalence study (MuSPAD) who had received all currently available immunisation schemes. We found that homologous mRNA-based or heterologous prime-boost vaccination produced significantly higher antibody responses than vector-based homologous vaccination. Ad26.CoV2S.2 performance was particularly concerning with reduced titres and 91.7% of samples classified as non-responsive for ACE2 binding inhibition, suggesting that recipients require a booster mRNA vaccination. While mRNA vaccination induced a higher ratio of RBD- and S1-targeting antibodies, vector-based vaccines resulted in an increased proportion of S2-targeting antibodies. Given the role of RBD- and S1-specific antibodies in neutralizing SARS-CoV-2, their relative over-representation after mRNA vaccination may explain why these vaccines have increased efficacy compared to vector-based formulations. Previously infected individuals had a robust immune response once vaccinated, regardless of which vaccine they received, which could aid future dose allocation should shortages arise for certain manufacturers. Overall, both titres and ACE2 binding inhibition peaked approximately 28 days post-second vaccination and then decreased.
Collapse
Affiliation(s)
- Alex Dulovic
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Barbora Kessel
- Department of Epidemiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Manuela Harries
- Department of Epidemiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Matthias Becker
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Julia Ortmann
- Department of Epidemiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Johanna Griesbaum
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Jennifer Jüngling
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Daniel Junker
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Pilar Hernandez
- Department of Epidemiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Daniela Gornyk
- Department of Epidemiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Stephan Glöckner
- Department of Epidemiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Vanessa Melhorn
- Department of Epidemiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Stefanie Castell
- Department of Epidemiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Jana-Kristin Heise
- Department of Epidemiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Yvonne Kemmling
- Department of Epidemiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Torsten Tonn
- German Red Cross Blood Donation Service North East, Dresden, Germany
| | - Kerstin Frank
- German Red Cross Blood Donation Service North East, Dresden, Germany
| | - Thomas Illig
- Hannover Unified Biobank, Hannover Medical School, Hannover, Germany
| | - Norman Klopp
- Hannover Unified Biobank, Hannover Medical School, Hannover, Germany
| | - Neha Warikoo
- Department of Epidemiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Angelika Rath
- Department of Epidemiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Christina Suckel
- Department of Epidemiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Anne Ulrike Marzian
- Department of Epidemiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Nicole Grupe
- Department of Epidemiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Philipp D. Kaiser
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Bjoern Traenkle
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Ulrich Rothbauer
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
- Pharmaceutical Biotechnology, Department of Pharmacy and Biochemistry, University of Tübingen, Tübingen, Germany
| | - Tobias Kerrinnes
- Department of RNA-Biology of Bacterial Infections, Helmholtz Institute for RNA-Based Infection Research, Würzburg, Germany
| | - Gérard Krause
- Department of Epidemiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture of the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Braunschweig, Germany
| | - Berit Lange
- Department of Epidemiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Braunschweig, Germany
| | | | - Monika Strengert
- Department of Epidemiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture of the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
| |
Collapse
|
109
|
Uprichard SL, O’Brien A, Evdokimova M, Rowe CL, Joyce C, Hackbart M, Cruz-Pulido YE, Cohen CA, Rock ML, Dye JM, Kuehnert P, Ricks KM, Casper M, Linhart L, Anderson K, Kirk L, Maggiore JA, Herbert AS, Clark NM, Reid GE, Baker SC. Antibody Response to SARS-CoV-2 Infection and Vaccination in COVID-19-naïve and Experienced Individuals. Viruses 2022; 14:370. [PMID: 35215962 PMCID: PMC8878640 DOI: 10.3390/v14020370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/01/2022] [Accepted: 02/04/2022] [Indexed: 11/25/2022] Open
Abstract
Understanding the magnitude of responses to vaccination during the ongoing SARS-CoV-2 pandemic is essential for ultimate mitigation of the disease. Here, we describe a cohort of 102 subjects (70 COVID-19-naïve, 32 COVID-19-experienced) who received two doses of one of the mRNA vaccines (BNT162b2 (Pfizer-BioNTech) and mRNA-1273 (Moderna)). We document that a single exposure to antigen via infection or vaccination induces a variable antibody response which is affected by age, gender, race, and co-morbidities. In response to a second antigen dose, both COVID-19-naïve and experienced subjects exhibited elevated levels of anti-spike and SARS-CoV-2 neutralizing activity; however, COVID-19-experienced individuals achieved higher antibody levels and neutralization activity as a group. The COVID-19-experienced subjects exhibited no significant increase in antibody or neutralization titer in response to the second vaccine dose (i.e., third antigen exposure). Finally, we found that COVID-19-naïve individuals who received the Moderna vaccine exhibited a more robust boost response to the second vaccine dose (p = 0.004) as compared to the response to Pfizer-BioNTech. Ongoing studies with this cohort will continue to contribute to our understanding of the range and durability of responses to SARS-CoV-2 mRNA vaccines.
Collapse
Affiliation(s)
- Susan L. Uprichard
- Department of Medicine, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (M.C.); (L.L.); (K.A.); (L.K.); (N.M.C.); (G.E.R.)
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (A.O.); (M.E.); (C.L.R.); (M.H.); (Y.E.C.-P.); (S.C.B.)
- Infectious Disease and Immunology Research Institute, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| | - Amornrat O’Brien
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (A.O.); (M.E.); (C.L.R.); (M.H.); (Y.E.C.-P.); (S.C.B.)
| | - Monika Evdokimova
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (A.O.); (M.E.); (C.L.R.); (M.H.); (Y.E.C.-P.); (S.C.B.)
| | - Cynthia L. Rowe
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (A.O.); (M.E.); (C.L.R.); (M.H.); (Y.E.C.-P.); (S.C.B.)
| | - Cara Joyce
- Department of Public Health Sciences, Parkinson School of Health Sciences and Public Health, Loyola University Chicago, Maywood, IL 60153, USA;
| | - Matthew Hackbart
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (A.O.); (M.E.); (C.L.R.); (M.H.); (Y.E.C.-P.); (S.C.B.)
| | - Yazmin E. Cruz-Pulido
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (A.O.); (M.E.); (C.L.R.); (M.H.); (Y.E.C.-P.); (S.C.B.)
| | - Courtney A. Cohen
- Viral Immunology Branch, Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD 21702, USA; (C.A.C.); (M.L.R.); (J.M.D.); (A.S.H.)
- The Geneva Foundation, Tacoma, WA 98042, USA
| | - Michelle L. Rock
- Viral Immunology Branch, Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD 21702, USA; (C.A.C.); (M.L.R.); (J.M.D.); (A.S.H.)
- The Geneva Foundation, Tacoma, WA 98042, USA
| | - John M. Dye
- Viral Immunology Branch, Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD 21702, USA; (C.A.C.); (M.L.R.); (J.M.D.); (A.S.H.)
| | - Paul Kuehnert
- Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD 21702, USA; (P.K.); (K.M.R.)
| | - Keersten M. Ricks
- Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD 21702, USA; (P.K.); (K.M.R.)
| | - Marybeth Casper
- Department of Medicine, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (M.C.); (L.L.); (K.A.); (L.K.); (N.M.C.); (G.E.R.)
| | - Lori Linhart
- Department of Medicine, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (M.C.); (L.L.); (K.A.); (L.K.); (N.M.C.); (G.E.R.)
| | - Katrina Anderson
- Department of Medicine, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (M.C.); (L.L.); (K.A.); (L.K.); (N.M.C.); (G.E.R.)
| | - Laura Kirk
- Department of Medicine, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (M.C.); (L.L.); (K.A.); (L.K.); (N.M.C.); (G.E.R.)
| | - Jack A. Maggiore
- Department of Pathology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA;
| | - Andrew S. Herbert
- Viral Immunology Branch, Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD 21702, USA; (C.A.C.); (M.L.R.); (J.M.D.); (A.S.H.)
| | - Nina M. Clark
- Department of Medicine, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (M.C.); (L.L.); (K.A.); (L.K.); (N.M.C.); (G.E.R.)
- Infectious Disease and Immunology Research Institute, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| | - Gail E. Reid
- Department of Medicine, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (M.C.); (L.L.); (K.A.); (L.K.); (N.M.C.); (G.E.R.)
- Infectious Disease and Immunology Research Institute, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| | - Susan C. Baker
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (A.O.); (M.E.); (C.L.R.); (M.H.); (Y.E.C.-P.); (S.C.B.)
- Infectious Disease and Immunology Research Institute, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| |
Collapse
|
110
|
Magnetic Enrichment of SARS-CoV-2 Antigen-Binding B Cells for Analysis of Transcriptome and Antibody Repertoire. MAGNETOCHEMISTRY 2022. [DOI: 10.3390/magnetochemistry8020023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The ongoing COVID-19 pandemic has had devastating health impacts across the globe. The development of effective diagnostics and therapeutics will depend on the understanding of immune responses to natural infection and vaccination to the causative agent of COVID-19, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). While both B-cell immunity and T-cell immunity are generated in SARS-CoV-2-infected and vaccinated individuals, B-cell-secreted antibodies are known to neutralize SARS-CoV-2 virus and protect from the disease. Although interest in characterizing SARS-CoV-2-reactive B cells is great, the low frequency of antigen-binding B cells in human blood limits in-depth cellular profiling. To overcome this obstacle, we developed a magnetic bead-based approach to enrich SARS-CoV-2-reactive B cells prior to transcriptional and antibody repertoire analysis by single-cell RNA sequencing (scRNA-seq). Here, we describe isolation of SARS-CoV-2 antigen-binding B cells from two seropositive donors and comparison to nonspecific B cells from a seronegative donor. We demonstrate that SARS-CoV-2 antigen-binding B cells can be distinguished on the basis of transcriptional profile and antibody repertoire. Furthermore, SARS-CoV-2 antigen-binding B cells exhibit a gene expression pattern indicative of antigen experience and memory status. Combining scRNA-seq methods with magnetic enrichment enables the rapid characterization of SARS-CoV-2 antigen-binding B cells.
Collapse
|
111
|
Jeewandara C, Fernando S, Pushpakumara PD, Ramu ST, Kamaladasa A, Gunasekara B, Aberathna IS, Kuruppu H, Ranasinghe T, Dayarathne S, Dissanayake O, Gamalath N, Ekanayake D, Jayamali J, Wijesinghe A, Dissanayake M, Somathilake G, Harvie M, Danasekara S, Jayathilaka D, Wijayatilake HDK, Weerasooriya N, Kekulandara C, Schimanski L, Rijal P, Tan TK, Dong T, Townsend A, Ogg GS, Malavige GN. Immune responses following the first dose of the Sputnik V (Gam-COVID-Vac). Sci Rep 2022; 12:1727. [PMID: 35110645 PMCID: PMC8810924 DOI: 10.1038/s41598-022-05788-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 01/12/2022] [Indexed: 01/08/2023] Open
Abstract
As the first dose of Gam-COVID-Vac, is currently used as a single dose vaccine in some countries, we investigated the immunogenicity of this at 4 weeks (327 naïve individuals). 88.7% seroconverted, with significantly lower seroconversion rates in those over 60 years (p = 0.004) and significantly lower than previously seen with AZD1222 (p = 0.018). 82.6% developed ACE2 receptor blocking antibodies, although levels were significantly lower than following natural infection (p = 0.0009) and a single dose of AZD1222 (p < 0.0001). Similar titres of antibodies were observed to the receptor binding domain of WT, B.1.1.7 and B.1.617.2 compared to AZD1222, while the levels for B.1.351 were significantly higher (p = 0.006) for Gam-COVID-Vac. 30% developed ex vivo IFNγ ELISpot responses (significantly lower than AZD1222), and high frequency of CD107a expressing T cells along with memory B cell responses. Although single dose of Gam-COVID-Vac was highly immunogenic, administration of a second dose is likely to be beneficial.
Collapse
Affiliation(s)
- Chandima Jeewandara
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | | | - Pradeep Darshana Pushpakumara
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Shyrar Tanussiya Ramu
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Achala Kamaladasa
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Banuri Gunasekara
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Inoka Sepali Aberathna
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Heshan Kuruppu
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Thushali Ranasinghe
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Shashika Dayarathne
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Osanda Dissanayake
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Nayanathara Gamalath
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Dinithi Ekanayake
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Jewantha Jayamali
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Ayesha Wijesinghe
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Madushika Dissanayake
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Gayasha Somathilake
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Michael Harvie
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Saubhagya Danasekara
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Deshni Jayathilaka
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | | | | | | | - Lisa Schimanski
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Centre for Translational Immunology, Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK
| | - Pramila Rijal
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Centre for Translational Immunology, Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK
| | - Tiong K Tan
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Centre for Translational Immunology, Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK
| | - Tao Dong
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Centre for Translational Immunology, Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK
| | - Alain Townsend
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Centre for Translational Immunology, Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK
| | - Graham S Ogg
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Centre for Translational Immunology, Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK
| | - Gathsaurie Neelika Malavige
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka.
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
112
|
Mousa ZS, Abdulamir AS. Application and Validation of SARS-CoV-2 RBD Neutralizing ELISA Assay. ARCHIVES OF RAZI INSTITUTE 2022; 77:391-402. [PMID: 35891753 PMCID: PMC9288645 DOI: 10.22092/ari.2021.356677.1890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 12/04/2021] [Indexed: 06/15/2023]
Abstract
The establishment of an approach for detecting the anti-severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2)-receptor-binding domain (RBD) neutralizing antibodies (nAbs) by a safe, easy, and rapid technique without requiring the use of live viruses is essential for facing the coronavirus disease 2019 (COVID-19) pandemic. Depending on competitive enzyme-linked immunosorbent assay (ELISA) methodology, the current study assay was designed to simulate the virus-host interaction using purified SARS-COV-2-RBD from the spike protein and the host cell receptor human angiotensin-converting enzyme 2 protein. The performance of this in-house neutralizing ELISA assay was validated using freshly prepared standards with different known concentrations of the assay. In this regard, a cohort of 50 serum samples from convalescent COVID-19 individuals with different disease severity at different time points post-recovery and a cohort of 50 serum samples from healthy individuals were processed by the in-house developed assay for detecting SARS-CoV-2 nAbs, in comparison with a commercial total anti-SARS-CoV-2 IgG antibody assay as a gold standard. The assay obtained a sensitivity of 88% (95% CI: 75.69-95.47) and a specificity of 92% (95% CI: 80.77- 97.78%). A negative strong correlation was demonstrated in the standard curve between the optical density absorbance and log concentration of the nAbs with a statistical measure of r2 (coefficient of determination) = 0.9539. The SARS-COV-2-RBD neutralizing ELISA assay serves as a high throughput qualitative and quantitative tool that can be applied in most laboratory settings without special biosafety requirements to detect anti-RBD nAbs for seroprevalence, pre-clinical, and clinical evaluation of COVID-19 vaccines efficiency and the rapid selection of convalescent plasma donors for the treatment of COVID-19 patients.
Collapse
Affiliation(s)
- Z S Mousa
- Baghdad Veterinary Hospital, Baghdad, Iraq
| | - A S Abdulamir
- Department of Medical Microbiology, College of Medicine, Al-Nahrain University, Baghdad, Iraq
| |
Collapse
|
113
|
A Prospective, Longitudinal Evaluation of SARS-CoV-2 COVID-19 Exposure, Use of Protective Equipment and Social Distancing in a Group of Community Physicians. Healthcare (Basel) 2022; 10:healthcare10020285. [PMID: 35206899 PMCID: PMC8871673 DOI: 10.3390/healthcare10020285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 01/26/2022] [Accepted: 01/28/2022] [Indexed: 11/16/2022] Open
Abstract
Introduction: Healthcare workers experience a significant risk of exposure to and infection from SARS-CoV-2, COVID-19. Nonetheless, little research has focused on physicians’ use of personal protective equipment (PPE), their concerns about becoming infected and their social distancing maneuvers. Methods: All staff physicians at Advocate Lutheran General Hospital were invited to participate. Their COVID-19 IgG antibody level was measured and an online questionnaire was completed. The questionnaire assessed the risk of COVID-19 exposure, PPE usage, concern for contracting COVID-19, the performance of high-risk procedures, work in high-risk settings, and social distancing practices. Testing was performed in September (T0), and December 2020 (T1) at the height of the global pandemic. Results: A total of 481 (26.7%) of 1800 AGLH physicians were enrolled at T0 and 458 (95% of the original group) at T1. A total of 21 (4.3%) and 39 (8.5%) participants had antibodies at T0 and T1. A total of 63 (13.8%) worked in high-risk settings and 111 (24.2%) performed high-risk procedures. Participants working in high-risk settings had increased exposure to COVID-19 infected patients (OR = 4.464 (CI = 2.522–8.459, p < 0.001). Participants were highly adherent to the use of PPE and social distancing practices including mask-wearing in public (86%, 82.1%), avoiding crowds (85.1%, 85.6%), six feet distancing (83.8%, 83.4%), and avoiding public transportation (78%, 83.8%). A total of 251 (55.4%) participants expressed moderate to extreme concern about becoming infected with COVID-19. Conclusions and Relevance: Among a group of community physicians, consistent PPE use and social distancing practices were common. These practices were associated with a low level of initial acquisition of COVID-19 infections and a relatively low longitudinal risk of infection.
Collapse
|
114
|
Broketa M, Bruhns P. Single-Cell Technologies for the Study of Antibody-Secreting Cells. Front Immunol 2022; 12:821729. [PMID: 35173713 PMCID: PMC8841722 DOI: 10.3389/fimmu.2021.821729] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 12/29/2021] [Indexed: 01/05/2023] Open
Abstract
Antibody-secreting cells (ASC), plasmablasts and plasma cells, are terminally differentiated B cells responsible for large-scale production and secretion of antibodies. ASC are derived from activated B cells, which may differentiate extrafollicularly or form germinal center (GC) reactions within secondary lymphoid organs. ASC therefore consist of short-lived, poorly matured plasmablasts that generally secrete lower-affinity antibodies, or long-lived, highly matured plasma cells that generally secrete higher-affinity antibodies. The ASC population is responsible for producing an immediate humoral B cell response, the polyclonal antibody repertoire, as well as in parallel building effective humoral memory and immunity, or potentially driving pathology in the case of autoimmunity. ASC are phenotypically and transcriptionally distinct from other B cells and further distinguishable by morphology, varied lifespans, and anatomical localization. Single cell analyses are required to interrogate the functional and transcriptional diversity of ASC and their secreted antibody repertoire and understand the contribution of individual ASC responses to the polyclonal humoral response. Here we summarize the current and emerging functional and molecular techniques for high-throughput characterization of ASC with single cell resolution, including flow and mass cytometry, spot-based and microfluidic-based assays, focusing on functional approaches of the secreted antibodies: specificity, affinity, and secretion rate.
Collapse
Affiliation(s)
- Matteo Broketa
- Institut Pasteur, Université de Paris, INSERM UMR 1222, Unit of Antibodies in Therapy and Pathology, Paris, France
- Sorbonne Université, Collège doctoral, Paris, France
| | - Pierre Bruhns
- Institut Pasteur, Université de Paris, INSERM UMR 1222, Unit of Antibodies in Therapy and Pathology, Paris, France
| |
Collapse
|
115
|
Winklmeier S, Eisenhut K, Taskin D, Rübsamen H, Gerhards R, Schneider C, Wratil PR, Stern M, Eichhorn P, Keppler OT, Klein M, Mader S, Kümpfel T, Meinl E. Persistence of functional memory B cells recognizing SARS-CoV-2 variants despite loss of specific IgG. iScience 2022; 25:103659. [PMID: 34957380 PMCID: PMC8686444 DOI: 10.1016/j.isci.2021.103659] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 11/17/2021] [Accepted: 12/15/2021] [Indexed: 01/22/2023] Open
Abstract
Although some COVID-19 patients maintain SARS-CoV-2-specific serum immunoglobulin G (IgG) for more than 6 months postinfection, others eventually lose IgG levels. We assessed the persistence of SARS-CoV-2-specific B cells in 17 patients, 5 of whom had lost specific IgGs after 5-8 months. Differentiation of blood-derived B cells in vitro revealed persistent SARS-CoV-2-specific IgG B cells in all patients, whereas IgA B cells were maintained in 11. Antibodies derived from cultured B cells blocked binding of viral receptor-binding domain (RBD) to the cellular receptor ACE-2, had neutralizing activity to authentic virus, and recognized the RBD of the variant of concern Alpha similarly to the wild type, whereas reactivity to Beta and Gamma were decreased. Thus, differentiation of memory B cells could be more sensitive for detecting previous infection than measuring serum antibodies. Understanding the persistence of SARS-CoV-2-specific B cells even in the absence of specific serum IgG will help to promote long-term immunity.
Collapse
Affiliation(s)
- Stephan Winklmeier
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377 Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, 82152 Martinsried, Germany
| | - Katharina Eisenhut
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377 Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, 82152 Martinsried, Germany
| | - Damla Taskin
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377 Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, 82152 Martinsried, Germany
| | - Heike Rübsamen
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377 Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, 82152 Martinsried, Germany
| | - Ramona Gerhards
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377 Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, 82152 Martinsried, Germany
| | - Celine Schneider
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377 Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, 82152 Martinsried, Germany
| | - Paul R. Wratil
- Max von Pettenkofer Institute & GeneCenter, Virology, LMU Munich, 80336 Munich, Germany
| | - Marcel Stern
- Max von Pettenkofer Institute & GeneCenter, Virology, LMU Munich, 80336 Munich, Germany
| | - Peter Eichhorn
- Institute of Laboratory Medicine, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Oliver T. Keppler
- Max von Pettenkofer Institute & GeneCenter, Virology, LMU Munich, 80336 Munich, Germany
| | - Matthias Klein
- Department of Neurology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Simone Mader
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377 Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, 82152 Martinsried, Germany
| | - Tania Kümpfel
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377 Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, 82152 Martinsried, Germany
| | - Edgar Meinl
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377 Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, 82152 Martinsried, Germany
- Corresponding author
| |
Collapse
|
116
|
Ramos A, Cardoso MJ, Ribeiro L, Guimarães JT. Assessing SARS-CoV-2 Neutralizing Antibodies after BNT162b2 Vaccination and Their Correlation with SARS-CoV-2 IgG Anti-S1, Anti-RBD and Anti-S2 Serological Titers. Diagnostics (Basel) 2022; 12:205. [PMID: 35054372 PMCID: PMC8775066 DOI: 10.3390/diagnostics12010205] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/07/2022] [Accepted: 01/13/2022] [Indexed: 01/27/2023] Open
Abstract
The humoral response through neutralizing antibodies (NAbs) is a key component of the immune response to COVID-19. However, the plaque reduction neutralization test (PRNT), the gold standard for determining NAbs, is technically demanding, time-consuming and requires BSL-3 conditions. Correlating the NAbs and total antibodies levels, assessed by generalized and automated serological tests, is crucial. Through a commercial surrogate virus neutralization test (sVNT), we aimed to evaluate the production of SARS-CoV-2 NAbs in a set of vaccinated healthcare workers and to correlate these NAbs with the SARS-CoV-2 IgG anti-S1, anti-RBD and anti-S2 serological titers. We found that 6 months after vaccination, only 74% maintain NAbs for the Wuhan strain/UK variant (V1) and 47% maintain NAbs for the South African and Brazil variants (V2). Through Spearman's correlation, we found the following correlations between the percentage of inhibition of NAbs and the SARS-CoV-2 IgG II Quant (Abbott Laboratories, Chicago, IL, USA) and BioPlex 2200 SARS-CoV-2 IgG Panel (Bio-Rad, Hercules, CA, USA) immunoassays: rho = 0.87 (V1) and rho = 0.73 (V2) for anti-S1 assessed by Abbott assay; rho = 0.77 (V1) and rho = 0.72 (V2) for anti-S1, rho = 0.88 (V1) and rho = 0.82 (V2) for anti-RBD, and rho = 0.68 (V1) and rho = 0.60 (V2) for anti-S2 assessed by BioPlex assay (p < 0.001 for all). In conclusion, we found a strong correlation between this fast, user-friendly, mobile and bio-safe sVNT and the serological immunoassays.
Collapse
Affiliation(s)
- Angélica Ramos
- Serviço de Patologia Clínica, Centro Hospitalar e Universitário de São João, 4200-319 Porto, Portugal; (M.J.C.); (L.R.); (J.T.G.)
- EPI Unit, Instituto de Saúde Pública, Universidade do Porto, 4200-135 Porto, Portugal
| | - Maria João Cardoso
- Serviço de Patologia Clínica, Centro Hospitalar e Universitário de São João, 4200-319 Porto, Portugal; (M.J.C.); (L.R.); (J.T.G.)
| | - Luís Ribeiro
- Serviço de Patologia Clínica, Centro Hospitalar e Universitário de São João, 4200-319 Porto, Portugal; (M.J.C.); (L.R.); (J.T.G.)
| | - João Tiago Guimarães
- Serviço de Patologia Clínica, Centro Hospitalar e Universitário de São João, 4200-319 Porto, Portugal; (M.J.C.); (L.R.); (J.T.G.)
- EPI Unit, Instituto de Saúde Pública, Universidade do Porto, 4200-135 Porto, Portugal
- Departamento de Biomedicina, Faculdade de Medicina, Universidade do Porto, 4200-135 Porto, Portugal
| |
Collapse
|
117
|
ELISA-Based Analysis Reveals an Anti-SARS-CoV-2 Protein Immune Response Profile Associated with Disease Severity. J Clin Med 2022; 11:jcm11020405. [PMID: 35054099 PMCID: PMC8781066 DOI: 10.3390/jcm11020405] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/04/2022] [Accepted: 01/11/2022] [Indexed: 01/01/2023] Open
Abstract
Since the start of the COVID-19 pandemic, many studies have investigated the humoral response to SARS-CoV-2 during infection. Studies with native viral proteins constitute a first-line approach to assessing the overall immune response, but small peptides are an accurate and valuable tool for the fine characterization of B-cell epitopes, despite the restriction of this approach to the determination of linear epitopes. In this study, we used ELISA and peptides covering a selection of structural and non-structural SARS-CoV-2 proteins to identify key epitopes eliciting a strong immune response that could serve as a biological signature of disease characteristics, such as severity, in particular. We used 213 plasma samples from a cohort of patients well-characterized clinically and biologically and followed for COVID-19 infection. We found that patients developing severe disease had higher titers of antibodies mapping to multiple specific epitopes than patients with mild to moderate disease. These data are potentially important as they could be used for immunological profiling to improve our knowledge of the quantitative and qualitative characteristics of the humoral response in relation to patient outcome.
Collapse
|
118
|
Performance of a flow cytometry-based immunoassay for detection of antibodies binding to SARS-CoV-2 spike protein. Sci Rep 2022; 12:586. [PMID: 35022478 PMCID: PMC8755750 DOI: 10.1038/s41598-021-04565-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 12/24/2021] [Indexed: 01/08/2023] Open
Abstract
The performance of a laboratory-developed IgG/IgA flow cytometry-based immunoassay (FCI) using Jurkat T cells stably expressing full-length native S protein was compared against Elecsys electrochemiluminiscent (ECLIA) Anti-SARS-CoV-2 S (Roche Diagnostics, Pleasanton, CA, USA), and Liaison SARS-CoV-2 TrimericS IgG chemiluminiscent assay (CLIA) (Diasorin S.p.a, Saluggia, IT) for detection of SARS-CoV-2-specific antibodies. A total of 225 serum/plasma specimens from 120 acute or convalescent COVID-19 individuals were included. Overall, IgG/IgA-FCI yielded the highest number of positives (n = 179), followed by IgA-FCI (n = 177), Roche ECLIA (n = 175), IgG-FCI (n = 172) and Diasorin CLIA (n = 154). For sera collected early after the onset of symptoms (within 15 days) IgG/IgA-FCI also returned the highest number of positive results (52/72; 72.2%). Positive percent agreement between FCI and compared immunoassays was highest for Roche ECLIA, ranging from 96.1 (IgG/IgA-FCI) to 97.7% (IgG-FCI), whereas negative percent agreement was higher between FCI and Diasosin CLIA, regardless of antibody isotype. The data suggest that FCI may outperform Roche ECLIA and Diasorin CLIA in terms of clinical sensitivity for serological diagnosis of SARS-CoV-2 infection.
Collapse
|
119
|
Deshpande PS, Abraham IE, Pitamberwale A, Dhote RH. Review of Clinical Performance of Serology Based Commercial Diagnostic Assays for Detection of Severe Acute Respiratory Syndrome Coronavirus 2 Antibodies. Viral Immunol 2022; 35:82-111. [PMID: 35007431 DOI: 10.1089/vim.2020.0313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus, which caused the coronavirus disease 2019 (COVID-19) pandemic as declared by the World Health Organization, has created havoc worldwide. The highly transmissible infection can be contained only by accurate diagnosis, quarantining, and exercising social distancing. Therefore, quick and massive deployment of SARS-CoV-2 testing plays a crucial role in the identification and isolation of infected patients. Reverse transcription-polymerase chain reaction is the gold standard for COVID-19 detection; however, it needs expertise, facilities, and time. Hence, for the ease of population-wide screening, serology-based diagnostic assays were introduced. These can help determine the prevalence of infection, understand the epidemiology of the disease, and assist in suitable public health interventions while being user-friendly and less time consuming. Although serological testing kits in markets soared, their sensitivity and specificity were questioned in reports from different parts of the world. In this article, we have reviewed 40 Food and Drug Administration (FDA) and CE-approved clinically evaluated serological kits (8 enzyme-linked immunosorbent assay [ELISA] kits, 10 chemiluminescent immunoassay [CLIA] kits, and 22 lateral flow immunoassay [LFIA] kits) for their sensitivity and specificity and discussed the apparent reasons behind their performance. We observed appreciable sensitivity in the kits detecting total antibodies compared to the kits targeting single isotype antibodies. Tests that determined antibodies against nucleocapsid protein were found to be more sensitive and those detecting antibodies against spike protein were found to have greater specificity. This study was conducted to help the decision-making while acquiring antibody kits and concurrently to be mindful of their shortcomings.
Collapse
Affiliation(s)
- Poonam S Deshpande
- Biochemistry Division, Department of Chemistry, Fergusson College, Pune, India
| | - Irene E Abraham
- Biochemistry Division, Department of Chemistry, Fergusson College, Pune, India
| | - Anjali Pitamberwale
- Biochemistry Division, Department of Chemistry, Fergusson College, Pune, India
| | - Radhika H Dhote
- Biochemistry Division, Department of Chemistry, Fergusson College, Pune, India
| |
Collapse
|
120
|
Kanokudom S, Assawakosri S, Suntronwong N, Auphimai C, Nilyanimit P, Vichaiwattana P, Thongmee T, Yorsaeng R, Srimuan D, Thatsanatorn T, Klinfueng S, Sudhinaraset N, Wanlapakorn N, Honsawek S, Poovorawan Y. Safety and Immunogenicity of the Third Booster Dose with Inactivated, Viral Vector, and mRNA COVID-19 Vaccines in Fully Immunized Healthy Adults with Inactivated Vaccine. Vaccines (Basel) 2022; 10:86. [PMID: 35062747 PMCID: PMC8779615 DOI: 10.3390/vaccines10010086] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/31/2021] [Accepted: 01/04/2022] [Indexed: 12/16/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has become a severe healthcare problem worldwide since the first outbreak in late December 2019. Currently, the COVID-19 vaccine has been used in many countries, but it is still unable to control the spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, despite patients receiving full vaccination doses. Therefore, we aimed to appraise the booster effect of the different platforms of vaccines, including inactivated vaccine (BBIBP), viral vector vaccine (AZD122), and mRNA vaccine (BNT162b2), in healthy adults who received the full dose of inactivated vaccine (CoronaVac). The booster dose was safe with no serious adverse events. Moreover, the immunogenicity indicated that the booster dose with viral vector and mRNA vaccine achieved a significant proportion of Ig anti-receptor binding domain (RBD), IgG anti-RBD, and IgA anti-S1 booster response. In contrast, inactivated vaccine achieved a lower booster response than others. Consequently, the neutralization activity of vaccinated serum had a high inhibition of over 90% against SARS-CoV-2 wild-type and their variants (B.1.1.7-alpha, B.1.351-beta, and B.1.617.2-delta). In addition, IgG anti-nucleocapsid was observed only among the group that received the BBIBP booster. Our study found a significant increase in levels of IFN-ɣ secreting T-cell response after the additional viral vector or mRNA booster vaccination. This study showed that administration with either viral vector (AZD1222) or mRNA (BNT162b2) boosters in individuals with a history of two doses of inactivated vaccine (CoronaVac) obtained great immunogenicity with acceptable adverse events.
Collapse
Affiliation(s)
- Sitthichai Kanokudom
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (S.K.); (S.A.); (N.S.); (C.A.); (P.N.); (P.V.); (T.T.); (R.Y.); (D.S.); (T.T.); (S.K.); (N.S.); (N.W.)
- Osteoarthritis and Musculoskeleton Research Unit, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok 10330, Thailand
| | - Suvichada Assawakosri
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (S.K.); (S.A.); (N.S.); (C.A.); (P.N.); (P.V.); (T.T.); (R.Y.); (D.S.); (T.T.); (S.K.); (N.S.); (N.W.)
- Osteoarthritis and Musculoskeleton Research Unit, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok 10330, Thailand
| | - Nungruthai Suntronwong
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (S.K.); (S.A.); (N.S.); (C.A.); (P.N.); (P.V.); (T.T.); (R.Y.); (D.S.); (T.T.); (S.K.); (N.S.); (N.W.)
| | - Chompoonut Auphimai
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (S.K.); (S.A.); (N.S.); (C.A.); (P.N.); (P.V.); (T.T.); (R.Y.); (D.S.); (T.T.); (S.K.); (N.S.); (N.W.)
| | - Pornjarim Nilyanimit
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (S.K.); (S.A.); (N.S.); (C.A.); (P.N.); (P.V.); (T.T.); (R.Y.); (D.S.); (T.T.); (S.K.); (N.S.); (N.W.)
| | - Preeyaporn Vichaiwattana
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (S.K.); (S.A.); (N.S.); (C.A.); (P.N.); (P.V.); (T.T.); (R.Y.); (D.S.); (T.T.); (S.K.); (N.S.); (N.W.)
| | - Thanunrat Thongmee
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (S.K.); (S.A.); (N.S.); (C.A.); (P.N.); (P.V.); (T.T.); (R.Y.); (D.S.); (T.T.); (S.K.); (N.S.); (N.W.)
| | - Ritthideach Yorsaeng
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (S.K.); (S.A.); (N.S.); (C.A.); (P.N.); (P.V.); (T.T.); (R.Y.); (D.S.); (T.T.); (S.K.); (N.S.); (N.W.)
| | - Donchida Srimuan
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (S.K.); (S.A.); (N.S.); (C.A.); (P.N.); (P.V.); (T.T.); (R.Y.); (D.S.); (T.T.); (S.K.); (N.S.); (N.W.)
| | - Thaksaporn Thatsanatorn
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (S.K.); (S.A.); (N.S.); (C.A.); (P.N.); (P.V.); (T.T.); (R.Y.); (D.S.); (T.T.); (S.K.); (N.S.); (N.W.)
| | - Sirapa Klinfueng
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (S.K.); (S.A.); (N.S.); (C.A.); (P.N.); (P.V.); (T.T.); (R.Y.); (D.S.); (T.T.); (S.K.); (N.S.); (N.W.)
| | - Natthinee Sudhinaraset
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (S.K.); (S.A.); (N.S.); (C.A.); (P.N.); (P.V.); (T.T.); (R.Y.); (D.S.); (T.T.); (S.K.); (N.S.); (N.W.)
| | - Nasamon Wanlapakorn
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (S.K.); (S.A.); (N.S.); (C.A.); (P.N.); (P.V.); (T.T.); (R.Y.); (D.S.); (T.T.); (S.K.); (N.S.); (N.W.)
| | - Sittisak Honsawek
- Osteoarthritis and Musculoskeleton Research Unit, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok 10330, Thailand
| | - Yong Poovorawan
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (S.K.); (S.A.); (N.S.); (C.A.); (P.N.); (P.V.); (T.T.); (R.Y.); (D.S.); (T.T.); (S.K.); (N.S.); (N.W.)
- The Royal Society of Thailand (FRS(T)), Sanam Sueapa, Dusit, Bangkok 10330, Thailand
| |
Collapse
|
121
|
Bosa L, Di Chiara C, Gaio P, Cosma C, Padoan A, Cozzani S, Perilongo G, Plebani M, Giaquinto C, Donà D, Cananzi M. Protective SARS-CoV-2 Antibody Response in Children With Inflammatory Bowel Disease. Front Pediatr 2022; 10:815857. [PMID: 35223697 PMCID: PMC8866952 DOI: 10.3389/fped.2022.815857] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/11/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND To date, there's no evidence of an increased risk of SARS-CoV-2 infection or more severe COVID-19 in patients with inflammatory bowel disease (IBD). However, whether COVID-19 alters the clinical course of IBD or whether IBD treatment affects the immunological response to SARS-CoV-2 is still under investigation, especially in children. AIM To assess the serological response to SARS-CoV-2 in children with IBD, and to evaluate the impact of COVID-19 on the clinical course of IBD. MATERIAL AND METHODS This prospective study enrolled children (0-18 years) followed-up at the University Hospital of Padova for IBD, who acquired a confirmed SARS-CoV-2 infection between 02.2020 and 02.2021. The anti-SARS-CoV-2 S-RBD IgG titer was evaluated at 3 months after infection and compared to that of a control group of healthy children matched for age, sex, and COVID-19 severity. RESULTS Twelve children with IBD (M = 5; median age 14 years) contracted COVID-19 during the study period. 11/12 patients were under immunomodulatory treatment (4/12 steroids; 6/12 azathioprine; 3/12 anti-TNFs; 2 vedolizumab; 1 ustekinumab). SARS-CoV-2 infection remained asymptomatic in 4/12 children and caused mild COVID-19 in the remaining 8. Mean anti-SARS-CoV-2 IgG S-RBD titer was similar between IBD patients and controls (27.3 ± 43.8 vs. 36.8 ± 35.3 kAU/L, p = ns). No children experienced IBD flares nor required gastroenterological support during the infection period. DISCUSSION Children with IBD can mount a protective humoral response against SARS-CoV-2, which is comparable to that of their healthy peers regardless of ongoing immunomodulatory treatment. This study also supports the favorable course of PIBD during COVID-19 and vice-versa.
Collapse
Affiliation(s)
- Luca Bosa
- Unit of Pediatric Gastroenterology, Digestive Endoscopy, Hepatology and Care of the Child With Liver Transplantation, Department of Women's and Children's Health, University Hospital of Padova, Padova, Italy
| | - Costanza Di Chiara
- Pediatric Infectious Diseases, Department of Women's and Children's Health, University Hospital of Padova, Padova, Italy
| | - Paola Gaio
- Unit of Pediatric Gastroenterology, Digestive Endoscopy, Hepatology and Care of the Child With Liver Transplantation, Department of Women's and Children's Health, University Hospital of Padova, Padova, Italy
| | - Chiara Cosma
- Department of Laboratory Medicine, University Hospital of Padova, Padova, Italy
| | - Andrea Padoan
- Department of Laboratory Medicine, University Hospital of Padova, Padova, Italy.,Department of Medicine-DIMED, Medical School, University of Padova, Padova, Italy
| | - Sandra Cozzani
- Pediatric Infectious Diseases, Department of Women's and Children's Health, University Hospital of Padova, Padova, Italy
| | - Giorgio Perilongo
- Unit of Pediatric Gastroenterology, Digestive Endoscopy, Hepatology and Care of the Child With Liver Transplantation, Department of Women's and Children's Health, University Hospital of Padova, Padova, Italy.,Department of Women's and Children's Health, University Hospital of Padova, Padova, Italy
| | - Mario Plebani
- Department of Laboratory Medicine, University Hospital of Padova, Padova, Italy.,Department of Medicine-DIMED, Medical School, University of Padova, Padova, Italy
| | - Carlo Giaquinto
- Pediatric Infectious Diseases, Department of Women's and Children's Health, University Hospital of Padova, Padova, Italy
| | - Daniele Donà
- Pediatric Infectious Diseases, Department of Women's and Children's Health, University Hospital of Padova, Padova, Italy
| | - Mara Cananzi
- Unit of Pediatric Gastroenterology, Digestive Endoscopy, Hepatology and Care of the Child With Liver Transplantation, Department of Women's and Children's Health, University Hospital of Padova, Padova, Italy
| |
Collapse
|
122
|
Kotsiou OS, Papagiannis D, Fradelos EC, Siachpazidou DI, Perlepe G, Miziou A, Kyritsis A, Vavougios GD, Kalantzis G, Gourgoulianis KI. Defining Antibody Seroprevalence and Duration of Humoral Responses to SARS-CoV-2 Infection and/or Vaccination in a Greek Community. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 19:407. [PMID: 35010667 PMCID: PMC8744770 DOI: 10.3390/ijerph19010407] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 12/24/2021] [Accepted: 12/28/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND In this work, we aimed to evaluate antibody-response longevity to SARS-CoV-2 infection and/or vaccination in one of the Greek communities that was worst hit by the pandemic, Deskati, five months after a previous serosurveillance and nine months after the pandemic wave initiation (October 2020). METHODS The SARS-CoV-2 IgG II Quant method (Architect, Abbott, IL, USA) was used for antibody testing. RESULTS A total of 69 subjects, who previously tested positive or negative for COVID-19 antibodies, participated in the study. We found that 48% of participants turned positive due to vaccination. 27% of participants were both previously infected and vaccinated. However, all previously infected participants retained antibodies to the virus, irrespective of their vaccination status. The antibody titers were significantly higher in previously infected participants that had been vaccinated than those who were unvaccinated and in those that had been previously hospitalized for COVID-19 than those with mild disease. CONCLUSIONS Antibody responses to SARS-CoV-2 infection were maintained nine months after the pandemic. Vaccination alone had generated an immune response in almost half of the population. Higher antibody titers were found in the case of vaccination in previously infected subjects and especially in those with severe disease leading to hospitalization.
Collapse
Affiliation(s)
- Ourania S. Kotsiou
- Faculty of Nursing, School of Health Sciences, University of Thessaly, Gaiopolis, 41110 Larissa, Greece;
- Department of Respiratory Medicine, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, 41110 Larissa, Greece; (D.I.S.); (G.P.); (A.M.); (A.K.); (G.D.V.); (G.K.); (K.I.G.)
| | - Dimitrios Papagiannis
- Public Health & Vaccines Lab, Department of Nursing, School of Health Sciences, University of Thessaly, Gaiopolis, 41110 Larissa, Greece;
| | - Evangelos C. Fradelos
- Faculty of Nursing, School of Health Sciences, University of Thessaly, Gaiopolis, 41110 Larissa, Greece;
| | - Dimitra I. Siachpazidou
- Department of Respiratory Medicine, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, 41110 Larissa, Greece; (D.I.S.); (G.P.); (A.M.); (A.K.); (G.D.V.); (G.K.); (K.I.G.)
| | - Garifallia Perlepe
- Department of Respiratory Medicine, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, 41110 Larissa, Greece; (D.I.S.); (G.P.); (A.M.); (A.K.); (G.D.V.); (G.K.); (K.I.G.)
| | - Angeliki Miziou
- Department of Respiratory Medicine, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, 41110 Larissa, Greece; (D.I.S.); (G.P.); (A.M.); (A.K.); (G.D.V.); (G.K.); (K.I.G.)
| | - Athanasios Kyritsis
- Department of Respiratory Medicine, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, 41110 Larissa, Greece; (D.I.S.); (G.P.); (A.M.); (A.K.); (G.D.V.); (G.K.); (K.I.G.)
| | - George D. Vavougios
- Department of Respiratory Medicine, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, 41110 Larissa, Greece; (D.I.S.); (G.P.); (A.M.); (A.K.); (G.D.V.); (G.K.); (K.I.G.)
| | - Georgios Kalantzis
- Department of Respiratory Medicine, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, 41110 Larissa, Greece; (D.I.S.); (G.P.); (A.M.); (A.K.); (G.D.V.); (G.K.); (K.I.G.)
| | - Konstantinos I. Gourgoulianis
- Department of Respiratory Medicine, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, 41110 Larissa, Greece; (D.I.S.); (G.P.); (A.M.); (A.K.); (G.D.V.); (G.K.); (K.I.G.)
| |
Collapse
|
123
|
Duarte N, Yanes-Lane M, Arora RK, Bobrovitz N, Liu M, Bego MG, Yan T, Cao C, Gurry C, Hankins CA, Cheng MP, Gingras AC, Mazer BD, Papenburg J, Langlois MA. Adapting Serosurveys for the SARS-CoV-2 Vaccine Era. Open Forum Infect Dis 2021; 9:ofab632. [PMID: 35103246 PMCID: PMC8755308 DOI: 10.1093/ofid/ofab632] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 12/09/2021] [Indexed: 12/04/2022] Open
Abstract
Population-level immune surveillance, which includes monitoring exposure and assessing vaccine-induced immunity, is a crucial component of public health decision-making during a pandemic. Serosurveys estimating the prevalence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antibodies in the population played a key role in characterizing SARS-CoV-2 epidemiology during the early phases of the pandemic. Existing serosurveys provide infrastructure to continue immune surveillance but must be adapted to remain relevant in the SARS-CoV-2 vaccine era. Here, we delineate how SARS-CoV-2 serosurveys should be designed to distinguish infection- and vaccine-induced humoral immune responses to efficiently monitor the evolution of the pandemic. We discuss how serosurvey results can inform vaccine distribution to improve allocation efficiency in countries with scarce vaccine supplies and help assess the need for booster doses in countries with substantial vaccine coverage.
Collapse
Affiliation(s)
- Nathan Duarte
- Department of Systems Design Engineering, University of Waterloo, Waterloo, Ontario, Canada
| | - Mercedes Yanes-Lane
- COVID-19 Immunity Task Force, Secretariat, McGill University, Montreal, Quebec, Canada
| | - Rahul K Arora
- Department of Community Health Sciences, University of Calgary, Calgary, Alberta, Canada
- Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| | - Niklas Bobrovitz
- Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Critical Care Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Michael Liu
- Harvard Medical School, Boston, Massachusetts, USA
| | - Mariana G Bego
- COVID-19 Immunity Task Force, Secretariat, McGill University, Montreal, Quebec, Canada
| | - Tingting Yan
- Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Christian Cao
- Department of Community Health Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Celine Gurry
- Coalition for Epidemic Preparedness Innovations, Oslo, Norway
| | - Catherine A Hankins
- COVID-19 Immunity Task Force, Secretariat, McGill University, Montreal, Quebec, Canada
- Department of Epidemiology, Biostatistics, and Occupational Health, School of Population and Global Health, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Matthew Pellan Cheng
- Divisions of Infectious Diseases and Medical Microbiology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Bruce D Mazer
- COVID-19 Immunity Task Force, Secretariat, McGill University, Montreal, Quebec, Canada
- Division of Allergy and Immunology, Montreal Children’s Hospital, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada
| | - Jesse Papenburg
- Department of Epidemiology, Biostatistics, and Occupational Health, School of Population and Global Health, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Montreal Children’s Hospital, Montreal, Quebec, Canada
- Division of Microbiology, Department of Clinical Laboratory Medicine, McGill University Health Centre, Montreal, Quebec, Canada
| | - Marc-André Langlois
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
- University of Ottawa Center for Infection, Immunity and Inflammation (CI3), Ottawa, Ontario, Canada
| |
Collapse
|
124
|
Nam M, Seo JD, Moon HW, Kim H, Hur M, Yun YM. Evaluation of Humoral Immune Response after SARS-CoV-2 Vaccination Using Two Binding Antibody Assays and a Neutralizing Antibody Assay. Microbiol Spectr 2021; 9:e0120221. [PMID: 34817223 PMCID: PMC8612149 DOI: 10.1128/spectrum.01202-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 10/24/2021] [Indexed: 01/14/2023] Open
Abstract
Multiple vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have been developed and administered to mitigate the coronavirus disease 2019 (COVID-19) pandemic. We assessed the humoral response of BNT162b2 and ChAdOx1 nCoV-19 using Siemens SARS-CoV-2 IgG (sCOVG; cutoff of ≥1.0 U/ml), Abbott SARS-CoV-2 IgG II Quant (CoV-2 IgG II; cutoff of ≥50.0 AU/ml), and GenScript cPASS SARS-CoV-2 neutralization antibody detection kits (cPASS; cutoff of ≥30% inhibition). We collected 710 serum samples (174 samples after BNT162b2 and 536 samples after ChAdOx1 nCoV-19). Venous blood was obtained 3 weeks after first and second vaccinations. In both vaccines, sCOVG, CoV-2 IgG II, and cPASS showed a high seropositive rate (>95.7%) except for cPASS after the first vaccination with ChAdOx1 nCoV-19 (68.8%). Using sCOVG and CoV-2 IgG II, the ratios of antibody value (second/first) increased 10.6- and 11.4-fold in BNT162b2 (first 14.1, second 134.8 U/ml; first 1,416.2, second 14,326.4 AU/ml) and 2.3- and 2.0-fold in ChAdOx1 nCoV-19 (first 4.0, second 9.1 U/ml; first 431.0, second 9,744.0 AU/ml). cPASS-positive results indicated a very high concordance rate with sCOVG and CoV-2 IgG II (>98%), whereas cPASS-negative results showed a relatively low concordance rate (range of 22.2% to 66.7%). To predict cPASS positivity, we suggested additional cutoffs for sCOVG and CoV-2 IgG II at 2.42 U/ml and 284 AU/ml, respectively. In conclusion, BNT162b2 and ChAdOx1 nCoV-19 evoked robust humoral responses. sCOVG and CoV-2 IgG II showed a very strong correlation with cPASS. sCOVG and CoV-2 IgG II may predict the presence of neutralizing antibodies against SARS-CoV-2. IMPORTANCE The Siemens severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) IgG (sCOVG; Siemens Healthcare Diagnostics Inc., NY, USA) and Abbott SARS-CoV-2 IgG II Quant (CoV-2 IgG II; Abbott Laboratories, Sligo, Ireland), which are automated, quantitative SARS-CoV-2-binding antibody assays, have been recently launched. This study aimed to evaluate the humoral immune response of BNT162b2 and ChAdOx1 nCoV-19 vaccines using sCOVG and CoV-2 IgG II and compare the quantitative values with the results of the GenScript surrogate virus neutralization test (cPASS; GenScript, USA Inc., NJ, USA). Our findings demonstrated that both BNT162b2 and ChAdOx1 nCoV-19 elicited a robust humoral response after the first vaccination and further increased after the second vaccination. sCOVG and CoV-2 IgG II showed a strong correlation, and the concordance rates among sCOVG, CoV-2 IgG II, and cPASS were very high in the cPASS-positive results. The additional cutoff sCOVG and CoV-2 IgG II could predict the results of cPASS.
Collapse
Affiliation(s)
- Minjeong Nam
- Department of Laboratory Medicine, Korea University Anam Hospital, Seoul, South Korea
| | - Jong Do Seo
- Department of Laboratory Medicine, Konkuk University School of Medicine, Seoul, South Korea
| | - Hee-Won Moon
- Department of Laboratory Medicine, Konkuk University School of Medicine, Seoul, South Korea
| | - Hanah Kim
- Department of Laboratory Medicine, Konkuk University School of Medicine, Seoul, South Korea
| | - Mina Hur
- Department of Laboratory Medicine, Konkuk University School of Medicine, Seoul, South Korea
| | - Yeo-Min Yun
- Department of Laboratory Medicine, Konkuk University School of Medicine, Seoul, South Korea
| |
Collapse
|
125
|
Correlation of the Commercial Anti-SARS-CoV-2 Receptor Binding Domain Antibody Test with the Chemiluminescent Reduction Neutralizing Test and Possible Detection of Antibodies to Emerging Variants. Microbiol Spectr 2021; 9:e0056021. [PMID: 34851163 PMCID: PMC8635131 DOI: 10.1128/spectrum.00560-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Serological tests are beneficial for recognizing the immune response against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). To identify protective immunity, optimization of the chemiluminescent reduction neutralizing test (CRNT) is critical. Whether commercial antibody tests have comparable accuracy is unknown. Serum samples were obtained from COVID-19 patients (n = 74), SARS-CoV-2 PCR-negative (n = 179), and suspected healthy individuals (n = 229) before SARS-CoV-2 variants had been detected locally. The convalescent phase was defined as the period after day 10 from disease onset or the episode of close contact. The CRNT using pseudotyped viruses displaying the wild-type (WT) spike protein and a commercial anti-receptor-binding domain (RBD) antibody test were assayed. Serology for the B.1.1.7 and B.1.351 variants was also assayed. Both tests concurred for symptomatic COVID-19 patients in the convalescent phase. They clearly differentiated between patients and suspected healthy individuals (sensitivity: 95.8% and 100%, respectively; specificity: 99.1% and 100%, respectively). Anti-RBD antibody test results correlated with neutralizing titers (r = 0.31, 95% confidence interval [CI] 0.22–0.38). Compared with the WT, lower CRNT values were observed for the variants. Of the samples with ≥100 U/mL by the anti-RBD antibody test, 77.8% and 88.9% showed ≥50% neutralization against the B.1.1.7 and the B.1.351 variants, respectively. Exceeding 100 U/mL in the anti-RBD antibody test was associated with neutralization of variants (P < 0.01). The CRNT and commercial anti-RBD antibody test effectively classified convalescent COVID-19 patients. Strong positive results with the anti-RBD antibody test can reflect neutralizing activity against emerging variants. IMPORTANCE This study provides a diagnostic evidence of test validity, which can lead to vaccine efficacy and proof of recovery after COVID-19. It is not easy to know neutralization against SARS-CoV-2 in the clinical laboratory because of technical and biohazard issues. The correlation of the quantitative anti-receptor-binding domain antibody test, which is widely available, with neutralizing test indicates that we can know indirectly the state of acquisition of functional immunity against wild and variant-type viruses in the clinical laboratory.
Collapse
|
126
|
Guerrieri M, Francavilla B, Fiorelli D, Nuccetelli M, Passali FM, Coppeta L, Somma G, Bernardini S, Magrini A, Di Girolamo S. Nasal and Salivary Mucosal Humoral Immune Response Elicited by mRNA BNT162b2 COVID-19 Vaccine Compared to SARS-CoV-2 Natural Infection. Vaccines (Basel) 2021; 9:vaccines9121499. [PMID: 34960244 PMCID: PMC8708818 DOI: 10.3390/vaccines9121499] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/12/2021] [Accepted: 12/13/2021] [Indexed: 01/22/2023] Open
Abstract
SARS-CoV-2 antibody assays are crucial in managing the COVID-19 pandemic. Approved mRNA COVID-19 vaccines are well known to induce a serum antibody responses against the spike protein and its RBD. Mucosal immunity plays a major role in the fight against COVID-19 directly at the site of virus entry; however, vaccine abilities to elicit mucosal immune responses have not been reported. We detected anti-SARS-CoV-2 IgA-S1 and IgG-RBD in three study populations (healthy controls, vaccinated subjects, and subjects recovered from COVID-19 infection) on serum, saliva, and nasal secretions using two commercial immunoassays (ELISA for IgA-S1 and chemiluminescent assay for IgG-RBD). Our results show that the mRNA BNT162b2 vaccine Comirnaty (Pfizer/BioNTech, New York, NY, USA) determines the production of nasal and salivary IgA-S1 and IgG-RBD against SARS-CoV-2. This mucosal humoral immune response is stronger after the injection of the second vaccine dose compared to subjects recovered from COVID-19. Since there is a lack of validated assays on saliva and nasal secretions, this study shows that our pre-analytical and analytical procedures are consistent with the data. Our findings indicate that the mRNA COVID-19 vaccine elicits antigen-specific nasal and salivary immune responses, and that mucosal antibody assays could be used as candidates for non-invasive monitoring of vaccine-induced protection against viral infection.
Collapse
Affiliation(s)
- Mariapia Guerrieri
- Department of Otorhinolaryngology, University of Rome “Tor Vergata”, 00100 Rome, Italy; (M.G.); (F.M.P.); (S.D.G.)
| | - Beatrice Francavilla
- Department of Otorhinolaryngology, University of Rome “Tor Vergata”, 00100 Rome, Italy; (M.G.); (F.M.P.); (S.D.G.)
- Correspondence:
| | - Denise Fiorelli
- Department of Experimental Medicine, University of Rome “Tor Vergata”, 00100 Rome, Italy; (D.F.); (M.N.); (S.B.)
| | - Marzia Nuccetelli
- Department of Experimental Medicine, University of Rome “Tor Vergata”, 00100 Rome, Italy; (D.F.); (M.N.); (S.B.)
| | - Francesco Maria Passali
- Department of Otorhinolaryngology, University of Rome “Tor Vergata”, 00100 Rome, Italy; (M.G.); (F.M.P.); (S.D.G.)
| | - Luca Coppeta
- Department of Occupational Medicine, University of Rome “Tor Vergata”, 00100 Rome, Italy; (L.C.); (G.S.); (A.M.)
| | - Giuseppina Somma
- Department of Occupational Medicine, University of Rome “Tor Vergata”, 00100 Rome, Italy; (L.C.); (G.S.); (A.M.)
| | - Sergio Bernardini
- Department of Experimental Medicine, University of Rome “Tor Vergata”, 00100 Rome, Italy; (D.F.); (M.N.); (S.B.)
- Department of Laboratory Medicine, Tor Vergata University Hospital, 00100 Rome, Italy
| | - Andrea Magrini
- Department of Occupational Medicine, University of Rome “Tor Vergata”, 00100 Rome, Italy; (L.C.); (G.S.); (A.M.)
| | - Stefano Di Girolamo
- Department of Otorhinolaryngology, University of Rome “Tor Vergata”, 00100 Rome, Italy; (M.G.); (F.M.P.); (S.D.G.)
| |
Collapse
|
127
|
Mihaescu G, Chifiriuc MC, Vrancianu CO, Constantin M, Filip R, Popescu MR, Burlibasa L, Nicoara AC, Bolocan A, Iliescu C, Gradisteanu Pircalabioru G. Antiviral Immunity in SARS-CoV-2 Infection: From Protective to Deleterious Responses. Microorganisms 2021; 9:2578. [PMID: 34946179 PMCID: PMC8703918 DOI: 10.3390/microorganisms9122578] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 12/26/2022] Open
Abstract
After two previous episodes, in 2002 and 2012, when two highly pathogenic coronaviruses (SARS, MERS) with a zoonotic origin emerged in humans and caused fatal respiratory illness, we are today experiencing the COVID-19 pandemic produced by SARS-CoV-2. The main question of the year 2021 is if naturally- or artificially-acquired active immunity will be effective against the evolving SARS-CoV-2 variants. This review starts with the presentation of the two compartments of antiviral immunity-humoral and cellular, innate and adaptive-underlining how the involved cellular and molecular actors are intrinsically connected in the development of the immune response in SARS-CoV-2 infection. Then, the SARS-CoV-2 immunopathology, as well as the derived diagnosis and therapeutic approaches, will be discussed.
Collapse
Affiliation(s)
- Grigore Mihaescu
- Faculty of Biology, University of Bucharest, 030018 Bucharest, Romania; (G.M.); (C.O.V.); (L.B.)
| | - Mariana Carmen Chifiriuc
- Faculty of Biology, University of Bucharest, 030018 Bucharest, Romania; (G.M.); (C.O.V.); (L.B.)
- Life, Environmental and Earth Sciences Division, Research Institute of the University of Bucharest, 050096 Bucharest, Romania;
- The Romanian Academy, 25 Calea Victoriei, Sector 1, 010071 Bucharest, Romania
| | | | | | - Roxana Filip
- Faculty of Medicine and Biological Sciences, Stefan cel Mare University of Suceava, 720229 Suceava, Romania;
- Regional County Emergency Hospital, 720284 Suceava, Romania
| | - Mihaela Roxana Popescu
- Department of Cardiology, Elias Emergency University Hospital “Carol Davila”, University of Medicine and Pharmacy “Carol Davila”, 020021 Bucharest, Romania;
| | - Liliana Burlibasa
- Faculty of Biology, University of Bucharest, 030018 Bucharest, Romania; (G.M.); (C.O.V.); (L.B.)
| | - Anca Cecilia Nicoara
- Faculty of Pharmacy, University of Medicine and Pharmacy “Carol Davila”, 020021 Bucharest, Romania;
| | - Alexandra Bolocan
- General Surgery, University of Medicine and Pharmacy “Carol Davila”, 020021 Bucharest, Romania;
| | - Ciprian Iliescu
- National Institute for Research and Development in Microtechnologies—IMT, 077190 Bucharest, Romania;
- Faculty of Applied Chemistry and Materials Science, University “Politehnica” of Bucharest, 011061 Bucharest, Romania
- Academy of Romanian Scientists, 010071 Bucharest, Romania
| | | |
Collapse
|
128
|
Magicova M, Fialova M, Zahradka I, Rajnochova-Bloudickova S, Hackajlo D, Raska P, Striz I, Viklicky O. Humoral response to SARS-CoV-2 is well preserved and symptom dependent in kidney transplant recipients. Am J Transplant 2021; 21:3926-3935. [PMID: 34212497 PMCID: PMC9906442 DOI: 10.1111/ajt.16746] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/24/2021] [Accepted: 06/24/2021] [Indexed: 01/25/2023]
Abstract
Data on the immune response to SARS-CoV-2 in kidney transplant recipients are scarce. Thus, we conducted a single-center observational study to assess the anti-SARS-CoV-2 IgG seroprevalence in outpatient kidney transplant recipients (KTR; n = 1037) and healthcare workers (HCW; n = 512) during the second wave of the COVID-19 pandemic in fall 2020 and evaluated the clinical variables affecting antibody levels. Antibodies against S1 and S2 subunit of SARS-CoV-2 were evaluated using immunochemiluminescent assay (cut off 9.5 AU/ml, sensitivity of 91.2% and specificity of 90.2%). Anti-SARS-CoV-2 IgG seroprevalence was lower in KTR than in HCW (7% vs. 11.9%, p = .001). Kidney transplant recipients with SARS-CoV-2 infection were younger (p = .001) and received CNI-based immunosuppression more frequently (p = .029) than seronegative KTR. Anti-SARS-CoV-2 IgG positive symptomatic KTR had a higher BMI (p = .04) than asymptomatic KTR. Interestingly, anti-SARS-CoV-2 IgG levels were higher in KTR than in HCW (median 31 AU/ml, IQR 17-84 vs. median 15 AU/ml, IQR 11-39, p < .001). The presence of moderate to severe symptoms in KTR was found to be the only independent factor affecting IgG levels (Beta coefficient = 41.99, 95% CI 9.92-74.06, p = .011) in the multivariable model. In conclusion, KTR exhibit a well-preserved symptom-dependent humoral response to SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Maria Magicova
- Department of Nephrology, Transplant Center, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Martina Fialova
- Department of Immunology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Ivan Zahradka
- Department of Nephrology, Transplant Center, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Silvie Rajnochova-Bloudickova
- Department of Nephrology, Transplant Center, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - David Hackajlo
- Department of Informatics, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Petr Raska
- Department of Informatics, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Ilja Striz
- Department of Immunology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Ondrej Viklicky
- Department of Nephrology, Transplant Center, Institute for Clinical and Experimental Medicine, Prague, Czech Republic,Correspondence Ondrej Viklicky, Department of Nephrology, Transplant Center, Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
| |
Collapse
|
129
|
Sherman AC, Smith T, Zhu Y, Taibl K, Howard-Anderson J, Landay T, Pisanic N, Kleinhenz J, Simon TW, Espinoza D, Edupuganti N, Hammond S, Rouphael N, Shen H, Fairley JK, Edupuganti S, Cardona-Ospina JA, Rodriguez-Morales AJ, Premkumar L, Wrammert J, Tarleton R, Fridkin S, Heaney CD, Scherer EM, Collins MH. Application of SARS-CoV-2 Serology to Address Public Health Priorities. Front Public Health 2021; 9:744535. [PMID: 34888282 PMCID: PMC8650110 DOI: 10.3389/fpubh.2021.744535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 10/14/2021] [Indexed: 11/17/2022] Open
Abstract
Background: Antibodies against SARS-CoV-2 can be detected by various testing platforms, but a detailed understanding of assay performance is critical. Methods: We developed and validated a simple enzyme-linked immunosorbent assay (ELISA) to detect IgG binding to the receptor-binding domain (RBD) of SARS-CoV-2, which was then applied for surveillance. ELISA results were compared to a set of complimentary serologic assays using a large panel of clinical research samples. Results: The RBD ELISA exhibited robust performance in ROC curve analysis (AUC> 0.99; Se = 89%, Sp = 99.3%). Antibodies were detected in 23/353 (6.5%) healthcare workers, 6/9 RT-PCR-confirmed mild COVID-19 cases, and 0/30 non-COVID-19 cases from an ambulatory site. RBD ELISA showed a positive correlation with neutralizing activity (p = <0.0001, R2 = 0.26). Conclusions: We applied a validated SARS-CoV-2-specific IgG ELISA in multiple contexts and performed orthogonal testing on samples. This study demonstrates the utility of a simple serologic assay for detecting prior SARS-CoV-2 infection, particularly as a tool for efficiently testing large numbers of samples as in population surveillance. Our work also highlights that precise understanding of SARS-CoV-2 infection and immunity at the individual level, particularly with wide availability of vaccination, may be improved by orthogonal testing and/or more complex assays such as multiplex bead assays.
Collapse
Affiliation(s)
- Amy C. Sherman
- Division of Infectious Diseases, The Hope Clinic of the Emory Vaccine Center, Emory University, Atlanta, GA, United States
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, United States
| | - Teresa Smith
- Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Yerun Zhu
- Division of Infectious Diseases, The Hope Clinic of the Emory Vaccine Center, Emory University, Atlanta, GA, United States
| | - Kaitlin Taibl
- Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | | | - Taylor Landay
- Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Nora Pisanic
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Jennifer Kleinhenz
- Division of Infectious Diseases, The Hope Clinic of the Emory Vaccine Center, Emory University, Atlanta, GA, United States
- Division of Infectious, Diseases, Department of Pediatrics, Emory University, Atlanta, GA, United States
| | - Trevor W. Simon
- Division of Infectious Diseases, The Hope Clinic of the Emory Vaccine Center, Emory University, Atlanta, GA, United States
| | - Daniel Espinoza
- Division of Infectious Diseases, The Hope Clinic of the Emory Vaccine Center, Emory University, Atlanta, GA, United States
| | - Neena Edupuganti
- Division of Infectious Diseases, The Hope Clinic of the Emory Vaccine Center, Emory University, Atlanta, GA, United States
| | - Skyler Hammond
- Department of Anthropology, Emory University, Atlanta, GA, United States
| | - Nadine Rouphael
- Division of Infectious Diseases, The Hope Clinic of the Emory Vaccine Center, Emory University, Atlanta, GA, United States
| | - Huifeng Shen
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, United States
| | - Jessica K. Fairley
- Division of Infectious Diseases, Emory University, Atlanta, GA, United States
| | - Srilatha Edupuganti
- Division of Infectious Diseases, The Hope Clinic of the Emory Vaccine Center, Emory University, Atlanta, GA, United States
| | - Jaime A. Cardona-Ospina
- Grupo de Investigación Biomedicina, Faculty of Medicine, Fundación Universitaria Autónoma de las Américas, Pereira, Colombia
- Emerging Infectious Diseases and Tropical Medicine Research Group, Sci-Help, Pereira, Colombia
| | - Alfonso J. Rodriguez-Morales
- Grupo de Investigación Biomedicina, Faculty of Medicine, Fundación Universitaria Autónoma de las Américas, Pereira, Colombia
- Master of Clinical Epidemiology and Biostatistics, Universidad Científica del Sur, Lima, Peru
| | - Lakshmanane Premkumar
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Jens Wrammert
- Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA, United States
| | - Rick Tarleton
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, United States
| | - Scott Fridkin
- Division of Infectious Diseases, Emory University, Atlanta, GA, United States
- Georgia Emerging Infections Program, Atlanta, GA, United States
| | | | - Erin M. Scherer
- Division of Infectious Diseases, The Hope Clinic of the Emory Vaccine Center, Emory University, Atlanta, GA, United States
| | - Matthew H. Collins
- Division of Infectious Diseases, The Hope Clinic of the Emory Vaccine Center, Emory University, Atlanta, GA, United States
| |
Collapse
|
130
|
Castillo-Olivares J, Wells DA, Ferrari M, Chan ACY, Smith P, Nadesalingam A, Paloniemi M, Carnell GW, Ohlendorf L, Cantoni D, Mayora-Neto M, Palmer P, Tonks P, Temperton NJ, Peterhoff D, Neckermann P, Wagner R, Doffinger R, Kempster S, Otter AD, Semper A, Brooks T, Albecka A, James LC, Page M, Schwaeble W, Baxendale H, Heeney JL. Analysis of Serological Biomarkers of SARS-CoV-2 Infection in Convalescent Samples From Severe, Moderate and Mild COVID-19 Cases. Front Immunol 2021; 12:748291. [PMID: 34867975 PMCID: PMC8640495 DOI: 10.3389/fimmu.2021.748291] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 10/22/2021] [Indexed: 12/11/2022] Open
Abstract
Precision monitoring of antibody responses during the COVID-19 pandemic is increasingly important during large scale vaccine rollout and rise in prevalence of Severe Acute Respiratory Syndrome-related Coronavirus-2 (SARS-CoV-2) variants of concern (VOC). Equally important is defining Correlates of Protection (CoP) for SARS-CoV-2 infection and COVID-19 disease. Data from epidemiological studies and vaccine trials identified virus neutralising antibodies (Nab) and SARS-CoV-2 antigen-specific (notably RBD and S) binding antibodies as candidate CoP. In this study, we used the World Health Organisation (WHO) international standard to benchmark neutralising antibody responses and a large panel of binding antibody assays to compare convalescent sera obtained from: a) COVID-19 patients; b) SARS-CoV-2 seropositive healthcare workers (HCW) and c) seronegative HCW. The ultimate aim of this study is to identify biomarkers of humoral immunity that could be used to differentiate severe from mild or asymptomatic SARS-CoV-2 infections. Some of these biomarkers could be used to define CoP in further serological studies using samples from vaccination breakthrough and/or re-infection cases. Whenever suitable, the antibody levels of the samples studied were expressed in International Units (IU) for virus neutralisation assays or in Binding Antibody Units (BAU) for ELISA tests. In this work we used commercial and non-commercial antibody binding assays; a lateral flow test for detection of SARS-CoV-2-specific IgG/IgM; a high throughput multiplexed particle flow cytometry assay for SARS-CoV-2 Spike (S), Nucleocapsid (N) and Receptor Binding Domain (RBD) proteins); a multiplex antigen semi-automated immuno-blotting assay measuring IgM, IgA and IgG; a pseudotyped microneutralisation test (pMN) and an electroporation-dependent neutralisation assay (EDNA). Our results indicate that overall, severe COVID-19 patients showed statistically significantly higher levels of SARS-CoV-2-specific neutralising antibodies (average 1029 IU/ml) than those observed in seropositive HCW with mild or asymptomatic infections (379 IU/ml) and that clinical severity scoring, based on WHO guidelines was tightly correlated with neutralisation and RBD/S antibodies. In addition, there was a positive correlation between severity, N-antibody assays and intracellular virus neutralisation.
Collapse
Affiliation(s)
- Javier Castillo-Olivares
- Lab of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - David A. Wells
- Lab of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
- DIOSynVax, University of Cambridge, Cambridge, United Kingdom
| | - Matteo Ferrari
- Lab of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
- DIOSynVax, University of Cambridge, Cambridge, United Kingdom
| | - Andrew C. Y. Chan
- Lab of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Peter Smith
- Lab of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Angalee Nadesalingam
- Lab of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Minna Paloniemi
- Lab of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - George W. Carnell
- Lab of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Luis Ohlendorf
- Lab of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Diego Cantoni
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent, Chatham, United Kingdom
| | - Martin Mayora-Neto
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent, Chatham, United Kingdom
| | - Phil Palmer
- Lab of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Paul Tonks
- Lab of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Nigel J. Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent, Chatham, United Kingdom
| | - David Peterhoff
- Institute of Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
| | - Patrick Neckermann
- Institute of Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
| | - Ralf Wagner
- Institute of Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Rainer Doffinger
- Department of Clinical Biochemistry and Immunology, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Sarah Kempster
- Division of Virology, National Institute for Biological Standards and Control, Potters Bar, United Kingdom
| | | | - Amanda Semper
- UK Health Security Agency, Porton Down, United Kingdom
| | - Tim Brooks
- UK Health Security Agency, Porton Down, United Kingdom
| | - Anna Albecka
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Leo C. James
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Mark Page
- Division of Virology, National Institute for Biological Standards and Control, Potters Bar, United Kingdom
| | - Wilhelm Schwaeble
- Complement Laboratory, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Helen Baxendale
- Royal Papworth Hospital NHS Foundation Trust, Cambridge, United Kingdom
| | - Jonathan L. Heeney
- Lab of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
131
|
Tian X, Jiang W, Zhang H, Lu X, Li L, Liu W, Li J. Persistence of the SARS-CoV-2 Antibody Response in Asymptomatic Patients in Correctional Facilities. Front Microbiol 2021; 12:789374. [PMID: 34858383 PMCID: PMC8631518 DOI: 10.3389/fmicb.2021.789374] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 10/21/2021] [Indexed: 12/23/2022] Open
Abstract
SARS-CoV-2 has caused a global health disaster with millions of death worldwide, and the substantial proportion of asymptomatic carriers poses a huge threat to public health. The long-term antibody responses and neutralization activity during natural asymptomatic SARS-CoV-2 infection are unknown. In this study, we used enzyme-linked immunosorbent assays (ELISA) and neutralization assay with purified SARS-CoV-2S and N proteins to study the antibody responses of 156 individuals with natural asymptomatic infection. We found robust antibody responses to SARS-CoV-2 in 156 patients from 6 to 12 months. Although the antibody responses gradually decreased, S-IgG was more stable than N-IgG. S-IgG was still detected in 79% of naturally infected individuals after 12 months of infection. Moderate to potent neutralization activities were also observed in 98.74% of patients 6 months after infection. However, this proportion decreased at 8-month (46.15%) and 10-month (39.11%) after infection, respectively. Only 23.72% of patients displayed potent neutralization activity at 12 months. This study strongly supports the long-term presence of antibodies against SARS-CoV-2 in individuals with natural asymptomatic infection, although the magnitude of the antibody responses started to cripple 6 months after infection.
Collapse
Affiliation(s)
- Xiaodong Tian
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Wenguo Jiang
- Jining Center for Disease Control and Prevention, Shandong, China
| | - He Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - XiXi Lu
- Jining Center for Disease Control and Prevention, Shandong, China
| | - Libo Li
- Jining Center for Disease Control and Prevention, Shandong, China
| | - Wenjun Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- School of Life Sciences, University of Science and Technology of China, Hefei, China
- Savaid Medical School, University of the Chinese Academy of Sciences, Beijing, China
- Institute of Microbiology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Beijing, China
| | - Jing Li
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- Savaid Medical School, University of the Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
132
|
Hojjat Jodaylami M, Djaïleb A, Ricard P, Lavallée É, Cellier-Goetghebeur S, Parker MF, Coutu J, Stuible M, Gervais C, Durocher Y, Desautels F, Cayer MP, de Grandmont MJ, Rochette S, Brouard D, Trottier S, Boudreau D, Pelletier JN, Masson JF. Cross-reactivity of antibodies from non-hospitalized COVID-19 positive individuals against the native, B.1.351, B.1.617.2, and P.1 SARS-CoV-2 spike proteins. Sci Rep 2021; 11:21601. [PMID: 34750399 PMCID: PMC8575961 DOI: 10.1038/s41598-021-00844-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 10/19/2021] [Indexed: 12/20/2022] Open
Abstract
SARS-CoV-2 variants of concern (VOCs) have emerged worldwide, with implications on the spread of the pandemic. Characterizing the cross-reactivity of antibodies against these VOCs is necessary to understand the humoral response of non-hospitalized individuals previously infected with SARS-CoV-2, a population that remains understudied. Thirty-two SARS-CoV-2-positive (PCR-confirmed) and non-hospitalized Canadian adults were enrolled 14-21 days post-diagnosis in 2020, before the emergence of the B.1.351 (also known as Beta), B.1.617.2 (Delta) and P.1 (Gamma) VOCs. Sera were collected 4 and 16 weeks post-diagnosis. Antibody levels and pseudo-neutralization of the ectodomain of SARS-CoV-2 spike protein/human ACE-2 receptor interaction were analyzed with native, B.1.351, B.1.617.2 and P.1 variant spike proteins. Despite a lower response observed for the variant spike proteins, we report evidence of a sustained humoral response against native, B.1.351, B.1.617.2 and P.1 variant spike proteins among non-hospitalized Canadian adults. Furthermore, this response inhibited the interaction between the spike proteins from the different VOCs and ACE-2 receptor for ≥ 16 weeks post-diagnosis, except for individuals aged 18-49 years who showed no inhibition of the interaction between B.1.617.1 or B.1.617.2 spike and ACE-2. Interestingly, the affinity (KD) measured between the spike proteins (native, B.1.351, B.1.617.2 and P.1) and antibodies elicited in sera of infected and vaccinated (BNT162b2 and ChAdOx1 nCoV-19) individuals was invariant. Relative to sera from vaccine-naïve (and previously infected) individuals, sera from vaccinated individuals had higher antibody levels (as measured with label-free SPR) and more efficiently inhibited the spike-ACE-2 interactions, even among individuals aged 18-49 years, showing the effectiveness of vaccination.
Collapse
Affiliation(s)
- Maryam Hojjat Jodaylami
- Department of Chemistry, Québec Centre for Advanced Materials (QCAM), Regroupement Québécois sur les Matériaux de Pointe (RQMP), and Centre Interdisciplinaire de Recherche sur le Cerveau et l'apprentissage (CIRCA), Université de Montréal, CP 6128 Succ. Centre-Ville, Montréal, QC, H3C 3J7, Canada
| | - Abdelhadi Djaïleb
- Department of Chemistry, Department of Biochemistry and PROTEO, The Québec Network for Research On Protein Function, Engineering and Applications, Université de Montréal, CP 6128 Succ. Centre-Ville, Montréal, QC, H3C 3J7, Canada
| | - Pierre Ricard
- Department of Chemistry, Québec Centre for Advanced Materials (QCAM), Regroupement Québécois sur les Matériaux de Pointe (RQMP), and Centre Interdisciplinaire de Recherche sur le Cerveau et l'apprentissage (CIRCA), Université de Montréal, CP 6128 Succ. Centre-Ville, Montréal, QC, H3C 3J7, Canada
| | - Étienne Lavallée
- Department of Chemistry, Department of Biochemistry and PROTEO, The Québec Network for Research On Protein Function, Engineering and Applications, Université de Montréal, CP 6128 Succ. Centre-Ville, Montréal, QC, H3C 3J7, Canada
| | - Stella Cellier-Goetghebeur
- Department of Chemistry, Department of Biochemistry and PROTEO, The Québec Network for Research On Protein Function, Engineering and Applications, Université de Montréal, CP 6128 Succ. Centre-Ville, Montréal, QC, H3C 3J7, Canada
| | - Megan-Faye Parker
- Department of Chemistry, Department of Biochemistry and PROTEO, The Québec Network for Research On Protein Function, Engineering and Applications, Université de Montréal, CP 6128 Succ. Centre-Ville, Montréal, QC, H3C 3J7, Canada
| | - Julien Coutu
- Department of Chemistry, Québec Centre for Advanced Materials (QCAM), Regroupement Québécois sur les Matériaux de Pointe (RQMP), and Centre Interdisciplinaire de Recherche sur le Cerveau et l'apprentissage (CIRCA), Université de Montréal, CP 6128 Succ. Centre-Ville, Montréal, QC, H3C 3J7, Canada
| | - Matthew Stuible
- Mammalian Cell Expression, Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, QC, Canada
| | - Christian Gervais
- Mammalian Cell Expression, Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, QC, Canada
| | - Yves Durocher
- Mammalian Cell Expression, Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, QC, Canada
| | - Florence Desautels
- Héma-Québec, Affaires médicales et innovation, 1070, avenue des Sciences-de-la-Vie, Québec, QC, G1V 5C3, Canada
| | - Marie-Pierre Cayer
- Héma-Québec, Affaires médicales et innovation, 1070, avenue des Sciences-de-la-Vie, Québec, QC, G1V 5C3, Canada
| | - Marie Joëlle de Grandmont
- Héma-Québec, Affaires médicales et innovation, 1070, avenue des Sciences-de-la-Vie, Québec, QC, G1V 5C3, Canada
| | - Samuel Rochette
- Héma-Québec, Affaires médicales et innovation, 1070, avenue des Sciences-de-la-Vie, Québec, QC, G1V 5C3, Canada
| | - Danny Brouard
- Héma-Québec, Affaires médicales et innovation, 1070, avenue des Sciences-de-la-Vie, Québec, QC, G1V 5C3, Canada
| | - Sylvie Trottier
- Centre de recherche du Centre hospitalier universitaire de Québec and Département de microbiologie-infectiologie et d'immunologie, Université Laval, 2705, boulevard Laurier, Québec, QC, G1V 4G2, Canada
| | - Denis Boudreau
- Department of Chemistry and Centre for Optics, Photonics and Lasers (COPL), Université Laval, 1045, av. de la Médecine, Québec, QC, G1V 0A6, Canada
| | - Joelle N Pelletier
- Department of Chemistry, Department of Biochemistry and PROTEO, The Québec Network for Research On Protein Function, Engineering and Applications, Université de Montréal, CP 6128 Succ. Centre-Ville, Montréal, QC, H3C 3J7, Canada.
| | - Jean-Francois Masson
- Department of Chemistry, Québec Centre for Advanced Materials (QCAM), Regroupement Québécois sur les Matériaux de Pointe (RQMP), and Centre Interdisciplinaire de Recherche sur le Cerveau et l'apprentissage (CIRCA), Université de Montréal, CP 6128 Succ. Centre-Ville, Montréal, QC, H3C 3J7, Canada.
| |
Collapse
|
133
|
Nerenz RD, Hubbard JA, Cervinski MA. Review of SARS-CoV-2 Antigen and Antibody Testing in Diagnosis and Community Surveillance. ADVANCES IN MOLECULAR PATHOLOGY 2021. [PMCID: PMC8220942 DOI: 10.1016/j.yamp.2021.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
134
|
Kinetics of SARS-CoV-2 Specific and Neutralizing Antibodies over Seven Months after Symptom Onset in COVID-19 Patients. Microbiol Spectr 2021; 9:e0059021. [PMID: 34550000 PMCID: PMC8557935 DOI: 10.1128/spectrum.00590-21] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
To assess the persistence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antibodies produced by natural infection and describe the serological characteristics over 7 months after symptom onset among coronavirus disease 2019 (COVID-19) patients by age and severity group, we followed up COVID-19 convalescent patients confirmed from 1 January to 20 March 2020 in Jiangsu, China and collected serum samples for testing IgM/IgG and neutralizing antibodies against SARS-CoV-2 between 26 August and 28 October 2020. In total, 284 recovered participants with COVID-19 were enrolled in our study. Patients had a mean age of 46.72 years (standard deviation [SD], 17.09), and 138 (48.59%) were male. The median follow-up time after symptom onset was 225.5 (interquartile range [IQR], 219 to 232) days. During the follow-up period (162 to 282 days after symptom onset), the seropositive rate of IgM fluctuated around 25.70% (95% confidence interval [CI], 20.72% to 31.20%) and that of IgG fluctuated around 79.93% (95% CI, 74.79% to 84.43%). Of the 284 patients, 64 participants were tested when discharged from hospital. Compared with that at the acute phase, the IgM/IgG antibody levels and IgM seropositivity have decreased; however, the seropositivity of IgG was not significantly lower at this follow-up (78.13% versus 82.81%). Fifty percent inhibitory dilution (ID50) titers of neutralizing antibody for samples when discharged from hospital (geometric mean titer [GMT], 82; 95% CI, 56 to 121) were significantly higher than those at 6 to 7 months after discharge (GMT, 47; 95% CI, 35 to 63) (P < 0.001). After 7 months from symptom onset, the convalescent COVID-19 patients continued to have high IgG seropositive; however, many plasma samples decreased neutralizing activity. IMPORTANCE The long-term characteristics of anti-SARS-CoV-2 antibodies among COVID-19 patients remain largely unclear. Tracking the longevity of these antibodies can provide a forward-looking reference for monitoring COVID-19. We conducted a comprehensive assessment combining the kinetics of specific and neutralizing antibodies over 7 months with age and disease severity and revealed influencing factors of the protection period of convalescent patients. By observing the long-term antibody levels against SARS-CoV-2 and comparing antibody levels at two time points after symptom onset, we found that the convalescent COVID-19 patients continued to have a high IgG seropositive rate; however, their plasma samples decreased neutralizing activity. These findings provide evidence supporting that the neutralizing activity of SARS-CoV-2-infected persons should be monitored and the administration of vaccine may be needed.
Collapse
|
135
|
Wack M, Péré H, Demory-Guinet N, Kassis-Chikhani N, Janot L, Vedie B, Izquierdo L, Bélec L, Veyer D. No SARS-CoV-2 reinfection among staff health-care workers: Prospective hospital-wide screening during the first and second waves in Paris. J Clin Virol 2021; 145:104999. [PMID: 34695725 PMCID: PMC8525071 DOI: 10.1016/j.jcv.2021.104999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 09/28/2021] [Accepted: 10/16/2021] [Indexed: 11/27/2022]
Abstract
Objectives Risk of reinfection with SARS-CoV-2 among health-care workers (HCWs) is unknown. We assessed the incidence rate of SARS-CoV-2 reinfection in the real-life setting of a longitudinal observational cohort of HCWs from the Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, France, during the first and second waves of COVID-19 epidemic. Methods From March to December 2020, HCWs were subjected to molecular and serology testing of SARS-CoV-2. Reinfection was defined as a positive test result during the first wave, either by serology or PCR, followed by a positive PCR during the second wave. Evolution of COVID-19 status of HWCs was assessed by a Sankey diagram. Results A total of 7765 tests (4579 PCR and 3186 serology) were carried out and 4168 HCWs had at least one test result during the follow-up period with a positivity rate of 15.9%. No case of reinfection during the second wave could be observed among 102 positive HCWs of the first wave, nor among 175 HCWs found positive by PCR during the second wave who were negative during the first wave. Conclusions SARS-CoV-2 reinfection was not observed among HCWs, suggesting a protective immunity against reinfection that lasts at least 8 months post infection.
Collapse
Affiliation(s)
- Maxime Wack
- Département d'Informatique Médicale, Biostatistiques et Santé Publique, hôpital européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, 75015, France; Faculté de Médecine, Université de Paris, Paris, 75005, France
| | - Hélène Péré
- Laboratoire de Virologie, Service de Microbiologie, hôpital européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, 75015, France; Unité de Génomique Fonctionnelle des Tumeurs Solides, Centre de Recherche des Cordeliers, INSERM, Université Paris, Paris, 75005, France
| | - Nathalie Demory-Guinet
- Service de Médecine du Travail, hôpital européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, 75015, France
| | - Najiby Kassis-Chikhani
- Unité d'Hygiène Hospitalière, Service de Microbiologie, hôpital européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, 75015, France
| | - Laurence Janot
- Service de Médecine du Travail, hôpital européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, 75015, France
| | - Benoit Vedie
- Laboratoire de Biochimie, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, 75015, France
| | - Laure Izquierdo
- Laboratoire de Virologie, Service de Microbiologie, hôpital européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, 75015, France
| | - Laurent Bélec
- Faculté de Médecine, Université de Paris, Paris, 75005, France; Laboratoire de Virologie, Service de Microbiologie, hôpital européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, 75015, France; INSERM U970, PARCC, hôpital européen Georges Pompidou, Faculté de Médecine, Université de Paris, Paris, 75015, France
| | - David Veyer
- Laboratoire de Virologie, Service de Microbiologie, hôpital européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, 75015, France; Unité de Génomique Fonctionnelle des Tumeurs Solides, Centre de Recherche des Cordeliers, INSERM, Université Paris, Paris, 75005, France.
| |
Collapse
|
136
|
Jagtap S, K R, Valloly P, Sharma R, Maurya S, Gaigore A, Ardhya C, Biligi DS, Desiraju BK, Natchu UCM, Saini DK, Roy R. Evaluation of spike protein antigens for SARS-CoV-2 serology. J Virol Methods 2021; 296:114222. [PMID: 34197839 PMCID: PMC8239204 DOI: 10.1016/j.jviromet.2021.114222] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 06/20/2021] [Accepted: 06/26/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Spike protein domains are being used in various serology-based assays to detect prior exposure to SARS-CoV-2 virus. However, there has been limited comparison of antibody titers against various spike protein antigens among COVID-19 infected patients. METHODS We compared four spike proteins (RBD, S1, S2 and a stabilized spike trimer (ST)) representing commonly used antigens for their reactivity to human IgG antibodies using indirect ELISA in serum from COVID-19 patients and pre-2020 samples. ST ELISA was also compared against the EUROIMMUN IgG ELISA test. Further, we estimated time appropriate IgG and IgA seropositivity rates in COVID-19 patients using a panel of sera samples collected longitudinally from the day of onset of symptoms (DOS). RESULTS Among the four spike antigens tested, the ST demonstrated the highest sensitivity (86.2 %; 95 % CI: 77.8-91.7 %), while all four antigens showed high specificity to COVID-19 sera (94.7-96.8 %). 13.8 % (13/94) of the samples did not show seroconversion in any of the four antigen-based assays. In a double-blinded head-to-head comparison, ST based IgG ELISA displayed a better sensitivity (87.5 %, 95 % CI: 76.4-93.8 %) than the EUROIMMUN IgG ELISA (67.9 %, 95 % CI: 54.8-78.6 %). Further, in ST-based assays, we found 48 % and 50 % seroconversion in the first six days (from DOS) for IgG and IgA antibodies, respectively, which increased to 84 % (IgG) and 85 % (IgA) for samples collected ≥22 days from DOS. CONCLUSIONS Comparison of spike antigens demonstrates that spike trimer protein is a superior option as an ELISA antigen for COVID-19 serology.
Collapse
Affiliation(s)
- Suraj Jagtap
- Department of Chemical Engineering, Indian Institute of Science, Bangalore, 560012, India
| | - Ratnasri K
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, 560012, India
| | - Priyanka Valloly
- Department of Chemical Engineering, Indian Institute of Science, Bangalore, 560012, India
| | - Rakhi Sharma
- Department of Chemical Engineering, Indian Institute of Science, Bangalore, 560012, India
| | - Satyaghosh Maurya
- Department of Chemical Engineering, Indian Institute of Science, Bangalore, 560012, India
| | - Anushree Gaigore
- Department of Chemical Engineering, Indian Institute of Science, Bangalore, 560012, India
| | - Chitra Ardhya
- Department of Pathology, Bangalore Medical College and Research Institute, Bangalore, 560002, India
| | - Dayananda S Biligi
- Department of Pathology, Bangalore Medical College and Research Institute, Bangalore, 560002, India
| | - Bapu Koundinya Desiraju
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad, 121001, India
| | | | - Deepak Kumar Saini
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, 560012, India; Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, 560012, India
| | - Rahul Roy
- Department of Chemical Engineering, Indian Institute of Science, Bangalore, 560012, India; Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, 560012, India.
| |
Collapse
|
137
|
Giannella M, Pierrotti LC, Helanterä I, Manuel O. SARS-CoV-2 vaccination in solid-organ transplant recipients: What the clinician needs to know. Transpl Int 2021; 34:1776-1788. [PMID: 34450686 PMCID: PMC8646251 DOI: 10.1111/tri.14029] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/30/2021] [Accepted: 08/20/2021] [Indexed: 12/21/2022]
Abstract
In response to the COVID-19 pandemic, SARS-CoV-2 vaccines have been developed at an unparalleled speed, with 14 SARS-CoV-2 vaccines currently authorized. Solid-organ transplant (SOT) recipients are at risk for developing a higher rate of COVID-19-related complications and therefore they are at priority for immunization against SARS-CoV-2. Preliminary data suggest that although SARS-CoV-2 vaccines are safe in SOT recipients (with similar rate of adverse events than in the general population), the antibody responses are decreased in this population. Risk factors for poor vaccine immunogenicity include older age, shorter time from transplantation, use of mycophenolate and belatacept, and worse allograft function. SOT recipients should continue to be advised to maintain hand hygiene, use of facemasks, and social distancing after SARS-CoV-2 vaccine. Vaccination of household contacts should be also prioritized. Although highly encouraged for research purposes, systematic assessment in clinical practice of humoral and cellular immune responses after SARS-CoV-2 vaccination is controversial, since correlation between immunological findings and clinical protection from severe COVID-19, and cutoffs for protection are currently unknown in SOT recipients. Alternative immunization schemes, including a booster dose, higher doses, and modulation of immunosuppression during vaccination, need to be assessed in the context of well-designed clinical trials.
Collapse
Affiliation(s)
- Maddalena Giannella
- Infectious Diseases UnitDepartment of Medical and Surgical SciencesIRCCS Azienda Ospedaliero‐Universitaria di Bologna, Policlinico di Sant’OrsolaAlma Mater Studiorum University of BolognaBolognaItaly
| | - Lígia C. Pierrotti
- Department of Infectious DiseasesUniversity of São Paulo School of Medicine Hospital das ClínicasSão PauloBrazil
| | - Ilkka Helanterä
- Transplantation and Liver SurgeryHelsinki University Hospital and University of HelsinkiHelsinkiFinland
| | - Oriol Manuel
- Infectious Diseases Service and Transplantation CenterLausanne University HospitalLausanneSwitzerland
| |
Collapse
|
138
|
Smith I, Mc Callum GJ, Sabljic AV, Marfia JI, Bombicino SS, Trabucchi A, Iacono RF, Birenbaum JM, Vazquez SC, Minoia JM, Cascone O, López MG, Taboga O, Targovnik AM, Wolman FJ, Fingermann M, Alonso LG, Valdez SN, Miranda MV. Rapid and cost-effective process based on insect larvae for scale-up production of SARS-COV-2 spike protein for serological COVID-19 testing. Biotechnol Bioeng 2021; 118:4129-4137. [PMID: 34264519 PMCID: PMC8426952 DOI: 10.1002/bit.27889] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/07/2021] [Accepted: 07/12/2021] [Indexed: 11/09/2022]
Abstract
Serology testing for COVID-19 is important in evaluating active immune response against SARS-CoV-2, studying the antibody kinetics, and monitoring reinfections with genetic variants and new virus strains, in particular, the duration of antibodies in virus-exposed individuals and vaccine-mediated immunity. In this study, recombinant S protein of SARS-CoV-2 was expressed in Rachiplusia nu, an important agronomic plague. One gram of insect larvae produces an amount of S protein sufficient for 150 determinations in the ELISA method herein developed. We established a rapid production process for SARS-CoV-2 S protein that showed immunoreactivity for anti-SARS-CoV-2 antibodies and was used as a single antigen for developing the ELISA method with high sensitivity (96.2%) and specificity (98.8%). Our findings provide an efficient and cost-effective platform for large-scale S protein production, and the scale-up is linear, thus avoiding the use of complex equipment like bioreactors.
Collapse
Affiliation(s)
- Ignacio Smith
- Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Cátedra de BiotecnologiaUniversidad de Buenos AiresBuenos AiresArgentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Universidad de Buenos AiresInstituto de Nanobiotecnología (NANOBIOTEC)Buenos AiresArgentina
| | - Gregorio Juan Mc Callum
- Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Cátedra de BiotecnologiaUniversidad de Buenos AiresBuenos AiresArgentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Universidad de Buenos AiresInstituto de Nanobiotecnología (NANOBIOTEC)Buenos AiresArgentina
| | - Adriana Victoria Sabljic
- Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Cátedra de InmunologíaUniversidad de Buenos AiresBuenos AiresArgentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Universidad de Buenos AiresInstituto de Estudios de la Inmunidad Humoral “Prof. Ricardo A. Margni” (IDEHU)Buenos AiresArgentina
| | - Juan Ignacio Marfia
- Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Cátedra de InmunologíaUniversidad de Buenos AiresBuenos AiresArgentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Universidad de Buenos AiresInstituto de Estudios de la Inmunidad Humoral “Prof. Ricardo A. Margni” (IDEHU)Buenos AiresArgentina
| | - Silvina Sonia Bombicino
- Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Cátedra de InmunologíaUniversidad de Buenos AiresBuenos AiresArgentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Universidad de Buenos AiresInstituto de Estudios de la Inmunidad Humoral “Prof. Ricardo A. Margni” (IDEHU)Buenos AiresArgentina
| | - Aldana Trabucchi
- Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Cátedra de InmunologíaUniversidad de Buenos AiresBuenos AiresArgentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Universidad de Buenos AiresInstituto de Estudios de la Inmunidad Humoral “Prof. Ricardo A. Margni” (IDEHU)Buenos AiresArgentina
| | - Ruben Francisco Iacono
- Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Cátedra de InmunologíaUniversidad de Buenos AiresBuenos AiresArgentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Universidad de Buenos AiresInstituto de Estudios de la Inmunidad Humoral “Prof. Ricardo A. Margni” (IDEHU)Buenos AiresArgentina
| | - Joaquín Manuel Birenbaum
- Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Cátedra de BiotecnologiaUniversidad de Buenos AiresBuenos AiresArgentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Universidad de Buenos AiresInstituto de Nanobiotecnología (NANOBIOTEC)Buenos AiresArgentina
| | - Susana Claudia Vazquez
- Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Cátedra de BiotecnologiaUniversidad de Buenos AiresBuenos AiresArgentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Universidad de Buenos AiresInstituto de Nanobiotecnología (NANOBIOTEC)Buenos AiresArgentina
| | - Juan Mauricio Minoia
- Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Cátedra de BiotecnologiaUniversidad de Buenos AiresBuenos AiresArgentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Universidad de Buenos AiresInstituto de Nanobiotecnología (NANOBIOTEC)Buenos AiresArgentina
| | - Osvaldo Cascone
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Universidad de Buenos AiresInstituto de Nanobiotecnología (NANOBIOTEC)Buenos AiresArgentina
- Instituto Nacional de Producción de Biológicos (INPB)ANLIS “Dr. Carlos G. Malbrán”Buenos AiresArgentina
| | - María Gabriela López
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)Buenos AiresArgentina
- Instituto de BiotecnologíaInstituto Nacional de Tecnología Agropecuaria (INTA)Buenos AiresArgentina
| | - Oscar Taboga
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)Buenos AiresArgentina
- Instituto de BiotecnologíaInstituto Nacional de Tecnología Agropecuaria (INTA)Buenos AiresArgentina
| | - Alexandra Marisa Targovnik
- Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Cátedra de BiotecnologiaUniversidad de Buenos AiresBuenos AiresArgentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Universidad de Buenos AiresInstituto de Nanobiotecnología (NANOBIOTEC)Buenos AiresArgentina
| | - Federico Javier Wolman
- Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Cátedra de BiotecnologiaUniversidad de Buenos AiresBuenos AiresArgentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Universidad de Buenos AiresInstituto de Nanobiotecnología (NANOBIOTEC)Buenos AiresArgentina
| | - Matías Fingermann
- Instituto Nacional de Producción de Biológicos (INPB)ANLIS “Dr. Carlos G. Malbrán”Buenos AiresArgentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)Buenos AiresArgentina
| | - Leonardo Gabriel Alonso
- Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Cátedra de BiotecnologiaUniversidad de Buenos AiresBuenos AiresArgentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Universidad de Buenos AiresInstituto de Nanobiotecnología (NANOBIOTEC)Buenos AiresArgentina
| | - Silvina Noemí Valdez
- Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Cátedra de InmunologíaUniversidad de Buenos AiresBuenos AiresArgentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Universidad de Buenos AiresInstituto de Estudios de la Inmunidad Humoral “Prof. Ricardo A. Margni” (IDEHU)Buenos AiresArgentina
| | - María Victoria Miranda
- Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Cátedra de BiotecnologiaUniversidad de Buenos AiresBuenos AiresArgentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Universidad de Buenos AiresInstituto de Nanobiotecnología (NANOBIOTEC)Buenos AiresArgentina
| |
Collapse
|
139
|
Neutralizing Antibodies against SARS-CoV-2, Anti-Ad5 Antibodies, and Reactogenicity in Response to Ad5-nCoV (CanSino Biologics) Vaccine in Individuals with and without Prior SARS-CoV-2. Vaccines (Basel) 2021; 9:vaccines9091047. [PMID: 34579284 PMCID: PMC8472849 DOI: 10.3390/vaccines9091047] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/12/2021] [Accepted: 09/17/2021] [Indexed: 12/23/2022] Open
Abstract
This is the first study outside of clinical trials (phase I–III) evaluating the ability of the Ad5-nCoV vaccine to generate neutralizing antibodies and the factors associated with optimal or suboptimal response. In a longitudinal assay, 346 people (117 with prior COVID-19 and 229 without prior COVID-19) vaccinated with Ad5-nCoV were recruited. The percentage of neutralizing antibodies against SARS-CoV-2 (Surrogate Virus Neutralization Test) and antibodies against Ad5 (ADV-Ad5 IgG ELISA) were quantified pre and post-vaccination effects. The Ad5-nCoV vaccine induces higher neutralizing antibodies percentage in individuals with prior COVID-19 than those without prior COVID-19 (median [IQR]: 98% [97–98.1] vs. 72% [54–90], respectively; p < 0.0001). Furthermore, a natural infection (before vaccination) induces more neutralizing antibodies percentage than immunized individuals without prior COVID-19 (p < 0.01). No patient had vaccine-severe adverse effects. The age, antidepressant, and immunosuppressive treatments, reactogenicity, and history of COVID-19 are associated with impaired antibody production. The anti-Ad5 antibodies increased after 21 days of post-vaccination in all groups (p < 0.01). We recommend the application of a booster dose of Ad5-nCoV, especially for those individuals without previous COVID-19 infection. Finally, the induction of anti-Ad5 antibodies after vaccination should be considered if a booster with the same vaccine is planned.
Collapse
|
140
|
Mutantu PN, Ngwe Tun MM, Nabeshima T, Yu F, Mukadi PK, Tanaka T, Tashiro M, Fujita A, Kanie N, Oshiro R, Takazono T, Imamura Y, Hirayama T, Moi ML, Inoue S, Izumikawa K, Yasuda J, Morita K. Development and Evaluation of Quantitative Immunoglobulin G Enzyme-Linked Immunosorbent Assay for the Diagnosis of Coronavirus Disease 2019 Using Truncated Recombinant Nucleocapsid Protein as Assay Antigen. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:9630. [PMID: 34574555 PMCID: PMC8469721 DOI: 10.3390/ijerph18189630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 09/04/2021] [Accepted: 09/09/2021] [Indexed: 11/24/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of coronavirus disease 2019 (COVID-19). Real-time RT-PCR is the most commonly used method for COVID-19 diagnosis. However, serological assays are urgently needed as complementary tools to RT-PCR. Hachim et al. 2020 and Burbelo et al. 2020 demonstrated that anti-nucleocapsid(N) SARS-CoV-2 antibodies are higher and appear earlier than the spike antibodies. Additionally, cross-reactive antibodies against N protein are more prevalent than those against spike protein. We developed a less cross-reactive immunoglobulin G (IgG) indirect ELISA by using a truncated recombinant SARS-CoV-2 N protein as assay antigen. A highly conserved region of coronaviruses N protein was deleted and the protein was prepared using an E. coli protein expression system. A total of 177 samples collected from COVID-19 suspected cases and 155 negative control sera collected during the pre-COVID-19 period were applied to evaluate the assay's performance, with the plaque reduction neutralization test and the commercial SARS-CoV-2 spike protein IgG ELISA as gold standards. The SARS-CoV-2 N truncated protein-based ELISA showed similar sensitivity (91.1% vs. 91.9%) and specificity (93.8% vs. 93.8%) between the PRNT and spike IgG ELISA, as well as also higher specificity compared to the full-length N protein (93.8% vs. 89.9%). Our ELISA can be used for the diagnosis and surveillance of COVID-19.
Collapse
Affiliation(s)
- Pierre Nsele Mutantu
- Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; (P.N.M.); (P.K.M.)
- Department of Virology, Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; (M.M.N.T.); (T.N.); (M.L.M.); (K.M.)
- Program for Nurturing Global Leaders in Tropical and Emerging Communicable Diseases, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Mya Myat Ngwe Tun
- Department of Virology, Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; (M.M.N.T.); (T.N.); (M.L.M.); (K.M.)
| | - Takeshi Nabeshima
- Department of Virology, Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; (M.M.N.T.); (T.N.); (M.L.M.); (K.M.)
| | - Fuxun Yu
- Guizhou Provincial People’s Hospital, Guiyang 550002, China;
| | - Patrick Kakoni Mukadi
- Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; (P.N.M.); (P.K.M.)
- Program for Nurturing Global Leaders in Tropical and Emerging Communicable Diseases, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
- Department of Clinical Medicine, Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Takeshi Tanaka
- Infection Control and Education Center, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan; (T.T.); (M.T.); (A.F.); (K.I.)
| | - Masato Tashiro
- Infection Control and Education Center, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan; (T.T.); (M.T.); (A.F.); (K.I.)
| | - Ayumi Fujita
- Infection Control and Education Center, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan; (T.T.); (M.T.); (A.F.); (K.I.)
| | - Nobuhiro Kanie
- Department of Infectious Diseases, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan; (N.K.); (R.O.)
| | - Ryosaku Oshiro
- Department of Infectious Diseases, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan; (N.K.); (R.O.)
| | - Takahiro Takazono
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan; (T.T.); (Y.I.); (T.H.)
| | - Yoshifumi Imamura
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan; (T.T.); (Y.I.); (T.H.)
- Medical Education Development Center, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| | - Tatsuro Hirayama
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan; (T.T.); (Y.I.); (T.H.)
| | - Meng Ling Moi
- Department of Virology, Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; (M.M.N.T.); (T.N.); (M.L.M.); (K.M.)
| | - Shingo Inoue
- Department of Virology, Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; (M.M.N.T.); (T.N.); (M.L.M.); (K.M.)
| | - Koichi Izumikawa
- Infection Control and Education Center, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan; (T.T.); (M.T.); (A.F.); (K.I.)
| | - Jiro Yasuda
- Department of Emerging Infectious Diseases, Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan;
| | - Kouichi Morita
- Department of Virology, Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; (M.M.N.T.); (T.N.); (M.L.M.); (K.M.)
| |
Collapse
|
141
|
COVID-19 in Liver Transplant Recipients: A Systematic Review. J Clin Med 2021; 10:jcm10174015. [PMID: 34501463 PMCID: PMC8432463 DOI: 10.3390/jcm10174015] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/31/2021] [Accepted: 09/01/2021] [Indexed: 12/16/2022] Open
Abstract
Liver transplant (LT) recipients are considered a vulnerable population amidst the COVID-19 pandemic. To date, available data have been heterogeneous and scarce. Therefore, we conducted a systematic literature review identifying English-language articles published in PubMed between November 2019 and 30 May 2021. We aimed to explore three areas: (1) outcome and clinical course; (2) immunological response after COVID-19 in LT recipients; and (3) vaccination response. After systematic selection, 35, 4, and 5 articles, respectively, were considered suitable for each area of analysis. Despite the heterogeneity of the reports included in this study, we found that gastrointestinal symptoms were common in LT recipients. The outcome of the LT population was not per se worse compared to the general population, although careful management of immunosuppressive therapy is required. While a complete therapy discontinuation is not encouraged, caution needs to be taken with use of mycophenolate mofetil (MMF), favoring tacrolimus (TAC) use. Although data conflicted about acquired immunity after SARS-CoV-2 infection, vaccine immunogenicity appeared to be low, suggesting that the level of surveillance should be kept high in this population.
Collapse
|
142
|
Carr EJ, Kronbichler A, Graham-Brown M, Abra G, Argyropoulos C, Harper L, Lerma EV, Suri RS, Topf J, Willicombe M, Hiremath S. Review of Early Immune Response to SARS-CoV-2 Vaccination Among Patients With CKD. Kidney Int Rep 2021; 6:2292-2304. [PMID: 34250319 PMCID: PMC8257418 DOI: 10.1016/j.ekir.2021.06.027] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/07/2021] [Accepted: 06/21/2021] [Indexed: 02/07/2023] Open
Abstract
The effects of the coronavirus disease-2019 (COVID-19) pandemic, particularly among those with chronic kidney disease (CKD), who commonly have defects in humoral and cellular immunity, and the efficacy of vaccinations against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) are uncertain. To inform public health and clinical practice, we synthesized published studies and preprints evaluating surrogate measures of immunity after SARS-CoV-2 vaccination in patients with CKD, including those receiving dialysis or with a kidney transplant. We found 35 studies (28 published, 7 preprints), with sample sizes ranging from 23 to 1140 participants and follow-up ranging from 1 week to 1 month after vaccination. Seventeen of these studies enrolled a control group. In the 22 studies of patients receiving dialysis, the development of antibodies was observed in 18% to 53% after 1 dose and in 70% to 96% after 2 doses of mRNA vaccine. In the 14 studies of transplant recipients, 3% to 59% mounted detectable humoral or cellular responses after 2 doses of mRNA vaccine. After vaccination, there were a few reported cases of relapse or de novo glomerulonephritis, and acute transplant rejection, suggesting a need for ongoing surveillance. Studies are needed to better evaluate the effectiveness of SARS-CoV-2 vaccination in these populations. Rigorous surveillance is necessary for detection of long-term adverse effects in patients with autoimmune disease and transplant recipients. For transplant recipients and those with suboptimal immune responses, alternate vaccination platforms and strategies should be considered. As additional data arise, the NephJC COVID-19 page will continue to be updated (http://www.nephjc.com/news/covid-vaccine).
Collapse
Affiliation(s)
| | | | | | - Graham Abra
- Satellite Healthcare, San Jose, California, USA
- Division of Nephrology, Department of Medicine, Stanford University, Palo Alto, California, USA
| | - Christos Argyropoulos
- Division of Nephrology, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Lorraine Harper
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Edgar V. Lerma
- Section of Nephrology, University of Illinois at Chicago/Advocate Christ Medical Center, Oak Lawn, Illinois, USA
| | - Rita S. Suri
- Research Institute, University Health Center, Department of Medicine, McGill University, Montreal, Québec, Canada
| | - Joel Topf
- Department of Medicine, Oakland University William Beaumont School of Medicine, Detroit, Michigan, USA
| | | | - Swapnil Hiremath
- Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
143
|
Riester E, Findeisen P, Hegel JK, Kabesch M, Ambrosch A, Rank CM, Pessl F, Laengin T, Niederhauser C. Performance evaluation of the Roche Elecsys Anti-SARS-CoV-2 S immunoassay. J Virol Methods 2021; 297:114271. [PMID: 34461153 PMCID: PMC8393518 DOI: 10.1016/j.jviromet.2021.114271] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/24/2021] [Accepted: 08/25/2021] [Indexed: 12/23/2022]
Abstract
The Elecsys® Anti-SARS-CoV-2 S immunoassay (Roche Diagnostics International Ltd, Rotkreuz, Switzerland) has been developed for the detection of antibodies to the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike (S) protein. We evaluated the assay performance using samples from seven sites in Germany, Austria, and Switzerland. For specificity and sensitivity analyses, 7880 presumed negative pre-pandemic samples and 827 SARS-CoV-2 PCR-confirmed single or sequential samples from 272 different patients were tested, respectively. The overall specificity and sensitivity (≥14 days post-PCR) for the Elecsys Anti-SARS-CoV-2 S immunoassay were 99.95% (95% confidence interval [CI]: 99.87–99.99; 7876/7880) and 97.92% (95% CI: 95.21–99.32; 235/240), respectively. The Elecsys Anti-SARS-CoV-2 S immunoassay had significantly higher specificity compared with the LIAISON® SARS-CoV-2 S1/S2 IgG (99.95% [2032/2033] vs 98.82% [2009/2033]), ADVIA Centaur® SARS-CoV-2 Total (100% [928/928] vs 86.96% [807/928]), ARCHITECT SARS-CoV-2 IgG (99.97% [2931/2932] vs 99.69% [2923/2932]), iFlash-SARS-CoV-2 IgM (100.00% [928/928] vs 99.57% [924/928]), and EUROIMMUN Anti-SARS-CoV-2 IgG (100.00% [903/903] vs 97.45% [880/903]) and IgA (100.00% [895/895] vs 95.75% [857/895]) assays. The Elecsys Anti-SARS-CoV-2 S immunoassay had significantly higher sensitivity (≥14 days post-PCR) compared with the ARCHITECT SARS-CoV-2 IgG (98.70% [76/77] vs 87.01% [67/77]), iFlash-SARS-CoV-2 IgG (100.00% [76/76] vs 93.42% [71/76]) and IgM (100.00% [76/76] vs 35.53% [27/76]), and EUROIMMUN Anti-SARS-CoV-2 IgG (98.26% [113/115] vs 93.91% [108/115]) assays. Therefore, the Elecsys Anti-SARS-CoV-2 S assay demonstrated a reliable performance across various sample populations for the detection of anti-S antibodies.
Collapse
Affiliation(s)
| | | | - J Kolja Hegel
- Labor Berlin, Charité Vivantes Services GmbH, Berlin, Germany
| | - Michael Kabesch
- University Children's Hospital Regensburg (KUNO) at the Hospital St. Hedwig of the Order of St. John and the University Hospital, University of Regensburg, Germany
| | - Andreas Ambrosch
- Institute for Laboratory Medicine, Microbiology and Hygiene, Barmherzige Brüder Hospital, Regensburg, Germany
| | | | | | | | - Christoph Niederhauser
- Interregionale Blood Transfusion Swiss Red Cross, Bern, Switzerland; Institute for Infectious Diseases (IFIK), University of Bern, Bern, Switzerland.
| |
Collapse
|
144
|
Sjöwall J, Azharuddin M, Frodlund M, Zhang Y, Sandner L, Dahle C, Hinkula J, Sjöwall C. SARS-CoV-2 Antibody Isotypes in Systemic Lupus Erythematosus Patients Prior to Vaccination: Associations With Disease Activity, Antinuclear Antibodies, and Immunomodulatory Drugs During the First Year of the Pandemic. Front Immunol 2021; 12:724047. [PMID: 34512651 PMCID: PMC8430325 DOI: 10.3389/fimmu.2021.724047] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 08/04/2021] [Indexed: 12/23/2022] Open
Abstract
Objectives Impact of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic on individuals with arthritis has been highlighted whereas data on other rheumatic diseases, e.g., systemic lupus erythematosus (SLE), are scarce. Similarly to SLE, severe SARS-CoV-2 infection includes risks for thromboembolism, an unbalanced type I interferon response, and complement activation. Herein, SARS-CoV-2 antibodies in longitudinal samples collected prior to vaccination were analyzed and compared with SLE progression and antinuclear antibody (ANA) levels. Methods One hundred patients (83 women) with established SLE and a regular visit to the rheumatologist (March 2020 to January 2021) were included. All subjects donated blood and had done likewise prior to the pandemic. SARS-CoV-2 antibody isotypes (IgG, IgA, IgM) to the cell receptor-binding S1-spike outer envelope protein were detected by ELISA, and their neutralizing capacity was investigated. IgG-ANA were measured by multiplex technology. Results During the pandemic, 4% had PCR-confirmed infection but 36% showed SARS-CoV-2 antibodies of ≥1 isotype; IgA was the most common (30%), followed by IgM (9%) and IgG (8%). The antibodies had low neutralizing capacity and were detected also in prepandemic samples. Plasma albumin (p = 0.04) and anti-dsDNA (p = 0.003) levels were lower in patients with SARS-CoV-2 antibodies. Blood group, BMI, smoking habits, complement proteins, daily glucocorticoid dose, use of hydroxychloroquine, or self-reported coronavirus disease 2019 (COVID-19) symptoms (except fever, >38.5°C) did not associate with SARS-CoV-2 antibodies. Conclusion Our data from early 2021 indicate that a large proportion of Swedish SLE patients had serological signs of exposure to SARS-CoV-2 but apparently with a minor impact on the SLE course. Use of steroids and hydroxychloroquine showed no distinct effects, and self-reported COVID-19-related symptoms correlated poorly with all antibody isotypes.
Collapse
Affiliation(s)
- Johanna Sjöwall
- Department of Biomedical and Clinical Sciences, Division of Inflammation and Infection/Infectious Diseases, Linköping University, Linköping, Sweden
| | - Mohammad Azharuddin
- Department of Biomedical and Clinical Sciences, Division of Molecular Medicine and Virology, Linköping University, Linköping, Sweden
| | - Martina Frodlund
- Department of Biomedical and Clinical Sciences, Division of Inflammation and Infection/Rheumatology, Linköping University, Linköping, Sweden
| | - Yuming Zhang
- Department of Biomedical and Clinical Sciences, Division of Molecular Medicine and Virology, Linköping University, Linköping, Sweden
| | - Laura Sandner
- Department of Biomedical and Clinical Sciences, Division of Molecular Medicine and Virology, Linköping University, Linköping, Sweden
| | - Charlotte Dahle
- Department of Biomedical and Clinical Sciences, Division of Inflammation and Infection/Clinical Immunology & Transfusion Medicine, Linköping University, Linköping, Sweden
| | - Jorma Hinkula
- Department of Biomedical and Clinical Sciences, Division of Molecular Medicine and Virology, Linköping University, Linköping, Sweden
| | - Christopher Sjöwall
- Department of Biomedical and Clinical Sciences, Division of Inflammation and Infection/Rheumatology, Linköping University, Linköping, Sweden
| |
Collapse
|
145
|
Meng Z, Guo S, Zhou Y, Li M, Wang M, Ying B. Applications of laboratory findings in the prevention, diagnosis, treatment, and monitoring of COVID-19. Signal Transduct Target Ther 2021; 6:316. [PMID: 34433805 PMCID: PMC8386162 DOI: 10.1038/s41392-021-00731-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 07/21/2021] [Accepted: 07/30/2021] [Indexed: 02/07/2023] Open
Abstract
The worldwide pandemic of coronavirus disease 2019 (COVID-19) presents us with a serious public health crisis. To combat the virus and slow its spread, wider testing is essential. There is a need for more sensitive, specific, and convenient detection methods of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Advanced detection can greatly improve the ability and accuracy of the clinical diagnosis of COVID-19, which is conducive to the early suitable treatment and supports precise prophylaxis. In this article, we combine and present the latest laboratory diagnostic technologies and methods for SARS-CoV-2 to identify the technical characteristics, considerations, biosafety requirements, common problems with testing and interpretation of results, and coping strategies of commonly used testing methods. We highlight the gaps in current diagnostic capacity and propose potential solutions to provide cutting-edge technical support to achieve a more precise diagnosis, treatment, and prevention of COVID-19 and to overcome the difficulties with the normalization of epidemic prevention and control.
Collapse
Affiliation(s)
- Zirui Meng
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Shuo Guo
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yanbing Zhou
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Mengjiao Li
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Minjin Wang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Binwu Ying
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
146
|
Dolscheid-Pommerich R, Bartok E, Renn M, Kümmerer BM, Schulte B, Schmithausen RM, Stoffel-Wagner B, Streeck H, Saschenbrecker S, Steinhagen K, Hartmann G. Correlation between a quantitative anti-SARS-CoV-2 IgG ELISA and neutralization activity. J Med Virol 2021; 94:388-392. [PMID: 34415572 PMCID: PMC8426838 DOI: 10.1002/jmv.27287] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 08/17/2021] [Indexed: 12/11/2022]
Abstract
In the current COVID-19 pandemic, a better understanding of the relationship between merely binding and functionally neutralizing antibodies is necessary to characterize protective antiviral immunity following infection or vaccination. This study analyzes the level of correlation between the novel quantitative EUROIMMUN Anti-SARS-CoV-2 QuantiVac ELISA (IgG) and a microneutralization assay. A panel of 123 plasma samples from a COVID-19 outbreak study population, preselected by semiquantitative anti-SARS-CoV-2 IgG testing, was used to assess the relationship between the novel quantitative ELISA (IgG) and a microneutralization assay. Binding IgG targeting the S1 antigen was detected in 106 (86.2%) samples using the QuantiVac ELISA, while 89 (72.4%) samples showed neutralizing antibody activity. Spearman's correlation analysis demonstrated a strong positive relationship between anti-S1 IgG levels and neutralizing antibody titers (rs = 0.819, p < 0.0001). High and low anti-S1 IgG levels were associated with a positive predictive value of 72.0% for high-titer neutralizing antibodies and a negative predictive value of 90.8% for low-titer neutralizing antibodies, respectively. These results substantiate the implementation of the QuantiVac ELISA to assess protective immunity following infection or vaccination.
Collapse
Affiliation(s)
| | - Eva Bartok
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Marcel Renn
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany.,Mildred Scheel School of Oncology, Bonn, Germany.,University Hospital Bonn, Medical Faculty, Bonn, Germany
| | | | - Bianca Schulte
- Institute of Virology, University Hospital Bonn, Bonn, Germany
| | | | - Birgit Stoffel-Wagner
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Hendrik Streeck
- Institute of Virology, University Hospital Bonn, Bonn, Germany
| | - Sandra Saschenbrecker
- Institute for Experimental Immunology, Affiliated to EUROIMMUN Medizinische Labordiagnostika AG, Lübeck, Germany
| | - Katja Steinhagen
- Institute for Experimental Immunology, Affiliated to EUROIMMUN Medizinische Labordiagnostika AG, Lübeck, Germany
| | - Gunther Hartmann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
147
|
Berguido FJ, Burbelo PD, Bortolami A, Bonfante F, Wernike K, Hoffmann D, Balkema-Buschmann A, Beer M, Dundon WG, Lamien CE, Cattoli G. Serological Detection of SARS-CoV-2 Antibodies in Naturally-Infected Mink and Other Experimentally-Infected Animals. Viruses 2021; 13:1649. [PMID: 34452513 PMCID: PMC8402807 DOI: 10.3390/v13081649] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/16/2021] [Accepted: 08/17/2021] [Indexed: 02/07/2023] Open
Abstract
The recent emergence of SARS-CoV-2 in humans from a yet unidentified animal reservoir and the capacity of the virus to naturally infect pets, farmed animals and potentially wild animals has highlighted the need for serological surveillance tools. In this study, the luciferase immunoprecipitation systems (LIPS), employing the spike (S) and nucleocapsid proteins (N) of SARS-CoV-2, was used to examine the suitability of the assay for antibody detection in different animal species. Sera from SARS-CoV-2 naturally-infected mink (n = 77), SARS-CoV-2 experimentally-infected ferrets, fruit bats and hamsters and a rabbit vaccinated with a purified spike protein were examined for antibodies using the SARS-CoV-2 N and/or S proteins. From comparison with the known neutralization status of the serum samples, statistical analyses including calculation of the Spearman rank-order-correlation coefficient and Cohen's kappa agreement were used to interpret the antibody results and diagnostic performance. The LIPS immunoassay robustly detected the presence of viral antibodies in naturally infected SARS-CoV-2 mink, experimentally infected ferrets, fruit bats and hamsters as well as in an immunized rabbit. For the SARS-CoV-2-LIPS-S assay, there was a good level of discrimination between the positive and negative samples for each of the five species tested with 100% agreement with the virus neutralization results. In contrast, the SARS-CoV-2-LIPS-N assay did not consistently differentiate between SARS-CoV-2 positive and negative sera. This study demonstrates the suitability of the SARS-CoV-2-LIPS-S assay for the sero-surveillance of SARS-CoV-2 infection in a range of animal species.
Collapse
Affiliation(s)
- Francisco J. Berguido
- Joint FAO/IAEA Centre for Nuclear Applications in Food and Agriculture, Animal Production and Health Laboratory, Department of Nuclear Sciences and Applications, International Atomic Energy Agency Vienna International Centre, P.O. Box 100, 1400 Vienna, Austria; (W.G.D.); (C.E.L.); (G.C.)
| | - Peter D. Burbelo
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Alessio Bortolami
- Laboratory of Experimental Animal Models, Division of Comparative Biomedical Sciences, Istituto Zooprofilattico Sperimentale delle Venezie, 35020 Legnaro, Italy; (A.B.); (F.B.)
| | - Francesco Bonfante
- Laboratory of Experimental Animal Models, Division of Comparative Biomedical Sciences, Istituto Zooprofilattico Sperimentale delle Venezie, 35020 Legnaro, Italy; (A.B.); (F.B.)
| | - Kerstin Wernike
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Südufer 10, 17493 Greifswald, Insel Riems, Germany; (K.W.); (D.H.); (M.B.)
| | - Donata Hoffmann
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Südufer 10, 17493 Greifswald, Insel Riems, Germany; (K.W.); (D.H.); (M.B.)
| | - Anne Balkema-Buschmann
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Südufer 10, 17493 Greifswald, Insel Riems, Germany;
| | - Martin Beer
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Südufer 10, 17493 Greifswald, Insel Riems, Germany; (K.W.); (D.H.); (M.B.)
| | - William G. Dundon
- Joint FAO/IAEA Centre for Nuclear Applications in Food and Agriculture, Animal Production and Health Laboratory, Department of Nuclear Sciences and Applications, International Atomic Energy Agency Vienna International Centre, P.O. Box 100, 1400 Vienna, Austria; (W.G.D.); (C.E.L.); (G.C.)
| | - Charles E. Lamien
- Joint FAO/IAEA Centre for Nuclear Applications in Food and Agriculture, Animal Production and Health Laboratory, Department of Nuclear Sciences and Applications, International Atomic Energy Agency Vienna International Centre, P.O. Box 100, 1400 Vienna, Austria; (W.G.D.); (C.E.L.); (G.C.)
| | - Giovanni Cattoli
- Joint FAO/IAEA Centre for Nuclear Applications in Food and Agriculture, Animal Production and Health Laboratory, Department of Nuclear Sciences and Applications, International Atomic Energy Agency Vienna International Centre, P.O. Box 100, 1400 Vienna, Austria; (W.G.D.); (C.E.L.); (G.C.)
| |
Collapse
|
148
|
Altawalah H. Antibody Responses to Natural SARS-CoV-2 Infection or after COVID-19 Vaccination. Vaccines (Basel) 2021; 9:910. [PMID: 34452035 PMCID: PMC8402626 DOI: 10.3390/vaccines9080910] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/03/2021] [Accepted: 08/11/2021] [Indexed: 12/14/2022] Open
Abstract
The novel coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is the causative agent of the ongoing pandemic of coronavirus disease 2019 (COVID-19). The clinical severity of COVID-19 ranges from asymptomatic to critical disease and, eventually, death in smaller subsets of patients. The first case of COVID-19 was declared at the end of 2019 and it has since spread worldwide and remained a challenge in 2021, with the emergence of variants of concern. In fact, new concerns were the still unclear situation of SARS-CoV-2 immunity during the ongoing pandemic and progress with vaccination. If maintained at sufficiently high levels, the immune response could effectively block reinfection, which might confer long-lived protection. Understanding the protective capacity and the duration of humoral immunity during SARS-CoV-2 infection or after vaccination is critical for managing the pandemic and would also provide more evidence about the efficacy of SARS-CoV-2 vaccines. However, the exact features of antibody responses that govern SARS-CoV-2 infection or after vaccination remain unclear. This review summarizes the main knowledge that we have about the humoral immune response during COVID-19 disease or after vaccination. Such knowledge should help to optimize vaccination strategies and public health decisions.
Collapse
Affiliation(s)
- Haya Altawalah
- Department of Microbiology, Faculty of Medicine, Kuwait University, Safat 24923, Kuwait; or
- Virology Unit, Yacoub Behbehani Center, Sabah Hospital, Ministry of Health, Safat 24923, Kuwait
| |
Collapse
|
149
|
Singh J, Pandit P, McArthur AG, Banerjee A, Mossman K. Evolutionary trajectory of SARS-CoV-2 and emerging variants. Virol J 2021; 18:166. [PMID: 34389034 PMCID: PMC8361246 DOI: 10.1186/s12985-021-01633-w] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 08/03/2021] [Indexed: 12/17/2022] Open
Abstract
The emergence of a novel coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and more recently, the independent evolution of multiple SARS-CoV-2 variants has generated renewed interest in virus evolution and cross-species transmission. While all known human coronaviruses (HCoVs) are speculated to have originated in animals, very little is known about their evolutionary history and factors that enable some CoVs to co-exist with humans as low pathogenic and endemic infections (HCoV-229E, HCoV-NL63, HCoV-OC43, HCoV-HKU1), while others, such as SARS-CoV, MERS-CoV and SARS-CoV-2 have evolved to cause severe disease. In this review, we highlight the origins of all known HCoVs and map positively selected for mutations within HCoV proteins to discuss the evolutionary trajectory of SARS-CoV-2. Furthermore, we discuss emerging mutations within SARS-CoV-2 and variants of concern (VOC), along with highlighting the demonstrated or speculated impact of these mutations on virus transmission, pathogenicity, and neutralization by natural or vaccine-mediated immunity.
Collapse
Affiliation(s)
- Jalen Singh
- School of Interdisciplinary Science, McMaster University, Hamilton, ON, Canada
| | - Pranav Pandit
- EpiCenter for Disease Dynamics, One Health Institute, School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| | - Andrew G McArthur
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Arinjay Banerjee
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK, Canada.
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada.
- Department of Biology, University of Waterloo, Waterloo, ON, Canada.
| | - Karen Mossman
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada.
- Department of Medicine, McMaster University, Hamilton, ON, Canada.
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
150
|
Cognetti JS, Steiner DJ, Abedin M, Bryan MR, Shanahan C, Tokranova N, Young E, Klose AM, Zavriyev A, Judy N, Piorek B, Meinhart C, Jakubowicz R, Warren H, Cady NC, Miller BL. Disposable photonics for cost-effective clinical bioassays: application to COVID-19 antibody testing. LAB ON A CHIP 2021; 21:2913-2921. [PMID: 34160511 DOI: 10.1039/d1lc00369k] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Decades of research have shown that biosensors using photonic circuits fabricated using CMOS processes can be highly sensitive, selective, and quantitative. Unfortunately, the cost of these sensors combined with the complexity of sample handling systems has limited the use of such sensors in clinical diagnostics. We present a new "disposable photonics" sensor platform in which rice-sized (1 × 4 mm) silicon nitride ring resonator sensor chips are paired with plastic micropillar fluidic cards for sample handling and optical detection. We demonstrate the utility of the platform in the context of detecting human antibodies to SARS-CoV-2, both in convalescent COVID-19 patients and for subjects undergoing vaccination. Given its ability to provide quantitative data on human samples in a simple, low-cost single-use format, we anticipate that this platform will find broad utility in clinical diagnostics for a broad range of assays.
Collapse
Affiliation(s)
- John S Cognetti
- Department of Biomedical Engineering, University of Rochester, Rochester, New York, USA.
| | - Daniel J Steiner
- Department of Biochemistry and Biophysics, University of Rochester, Rochester, New York, USA
| | - Minhaz Abedin
- College of Nanoscale Science and Engineering, SUNY Polytechnic, Albany, New York, USA
| | - Michael R Bryan
- Department of Biochemistry and Biophysics, University of Rochester, Rochester, New York, USA
| | - Conor Shanahan
- Department of Biomedical Engineering, University of Rochester, Rochester, New York, USA.
| | - Natalya Tokranova
- College of Nanoscale Science and Engineering, SUNY Polytechnic, Albany, New York, USA
| | - Ethan Young
- Ortho-Clinical Diagnostics, Rochester, New York, USA
| | - Alanna M Klose
- Department of Dermatology, University of Rochester, Rochester, New York, USA
| | | | - Nicholas Judy
- Department of Mechanical Engineering, University of California at Santa Barbara, Santa Barbara, California, USA
| | - Brian Piorek
- Department of Mechanical Engineering, University of California at Santa Barbara, Santa Barbara, California, USA
| | - Carl Meinhart
- Department of Mechanical Engineering, University of California at Santa Barbara, Santa Barbara, California, USA
| | | | - Harold Warren
- Ortho-Clinical Diagnostics, Rochester, New York, USA
| | - Nathaniel C Cady
- College of Nanoscale Science and Engineering, SUNY Polytechnic, Albany, New York, USA
| | - Benjamin L Miller
- Department of Biomedical Engineering, University of Rochester, Rochester, New York, USA. and Department of Biochemistry and Biophysics, University of Rochester, Rochester, New York, USA and Institute of Optics, University of Rochester, Rochester, New York, USA and Department of Dermatology, University of Rochester, Rochester, New York, USA
| |
Collapse
|