51
|
Brown ER, O’Brien MP, Snow B, Isa F, Forleo-Neto E, Chan KC, Hou P, Cohen MS, Herman G, Barnabas RV. A Prospective Study of Key Correlates for Household Transmission of Severe Acute Respiratory Syndrome Coronavirus 2. Open Forum Infect Dis 2023; 10:ofad271. [PMID: 37416758 PMCID: PMC10319621 DOI: 10.1093/ofid/ofad271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 05/22/2023] [Indexed: 07/08/2023] Open
Abstract
Background Randomized controlled trials evaluated monoclonal antibodies for the treatment (Study 2067) and prevention (Study 2069) of coronavirus disease 2019 (COVID-19). Household contacts of the infected index case in Study 2067 were enrolled in Study 2069 and prospectively followed; these cohorts provided a unique opportunity to evaluate correlates of transmission, specifically viral load. Methods This post hoc analysis was designed to identify and evaluate correlates of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) transmission, adjusting for potential confounding factors related to source SARS-CoV-2 viral load and risk of SARS-CoV-2 acquisition in this population. Correlates of transmission were evaluated in potential transmission pairs (any infected household member plus susceptible household contact). Results In total, 943 participants were included. In multivariable regression, 2 potential correlates were determined to have a statistically significant (P < .05) association with transmission risk. A 10-fold increase in viral load was associated with a 40% increase in odds of transmission; sharing a bedroom with the index participant was associated with a 199% increase in odds of transmission. Conclusions In this prospective, post hoc analysis that controlled for confounders, the 2 key correlates for transmission of SARS-CoV-2 within a household are sharing a bedroom and increased viral load, consistent with increased exposure to the infected individual.
Collapse
Affiliation(s)
- Elizabeth R Brown
- Vaccine and Infectious Disease and Public Health Services Divisions, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Meagan P O’Brien
- Global Development, Regeneron Pharmaceuticals, Inc, Tarrytown, New York, USA
| | - Brian Snow
- Global Development, Regeneron Pharmaceuticals, Inc, Tarrytown, New York, USA
| | - Flonza Isa
- Global Development, Regeneron Pharmaceuticals, Inc, Tarrytown, New York, USA
| | - Eduardo Forleo-Neto
- Global Development, Regeneron Pharmaceuticals, Inc, Tarrytown, New York, USA
| | - Kuo-Chen Chan
- Global Development, Regeneron Pharmaceuticals, Inc, Tarrytown, New York, USA
| | - Peijie Hou
- Global Development, Regeneron Pharmaceuticals, Inc, Tarrytown, New York, USA
| | - Myron S Cohen
- Institute for Global Health and Infectious Diseases, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Gary Herman
- Global Development, Regeneron Pharmaceuticals, Inc, Tarrytown, New York, USA
| | - Ruanne V Barnabas
- Division of Infectious Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
52
|
Figueiredo GM, Tengan FM, Campos SR, Luna EJ. Seroprevalence of SARS-CoV-2 in Brazil: A systematic review and meta-analysis. Clinics (Sao Paulo) 2023; 78:100233. [PMID: 37348256 PMCID: PMC10261714 DOI: 10.1016/j.clinsp.2023.100233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 04/19/2023] [Accepted: 06/01/2023] [Indexed: 06/24/2023] Open
Abstract
OBJECTIVES To summarize the data on SARS-CoV-2 seroprevalence surveys conducted in Brazil before the introduction of vaccines METHODS: The authors conducted a systematic review and meta-analysis on the seroprevalence of SARS-CoV-2 infection in Brazil. The present review followed the PRISMA guidelines. The authors searched Medline, Embase, and LILACS databases for serologic surveys conducted in the Brazilian population, in the period from 01/10/2019 to 07/11/2021, without language restrictions. The authors included studies that presented data concerning SARS-CoV-2 antibodies seroprevalence in Brazil and had a sample size ≥50 individuals. Considering the expected heterogeneity between studies, all analyses were performed using the random effects model, and heterogeneity was assessed using the I2 statistic RESULTS: Of 586 publications identified in the initial searches, 54 were included in the review and meta-analysis, which contained the results of 135 surveys, with 336,620 participants. The estimated seroprevalence was 11.0%, ranging from 1.0% to 83.0%, with a substantial heterogeneity (I2 = 99.55%). In subgroup analyses, the authors observed that the prevalence of SARS-CoV-2 antibodies was 13.0% in blood donors, 9.0% in the population-based surveys, 13% in schoolchildren, and 11.0% in healthcare workers. CONCLUSIONS Seroprevalence increases over time. Large differences were observed among the regions of the country. It was higher in the Northern region, decreasing towards the South. The present results may contribute to the analysis of the spread of SARS-CoV-2 infection in the Brazilian population before vaccination, one of the factors that may be influencing the clinical presentation of COVID-19 cases related to the new variants, as well as the effectiveness of the vaccination program.
Collapse
Affiliation(s)
- Gerusa Maria Figueiredo
- Departamento de Medicina Preventiva da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil.
| | - Fátima Mitiko Tengan
- Departamento de Moléstias Infecciosas e Parasitarias da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil.
| | - Sergio Roberto Campos
- Departamento de Medicina Preventiva da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil.
| | - Expedito José Luna
- Departamento de Medicina Preventiva da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
53
|
Jones JM, Manrique IM, Stone MS, Grebe E, Saa P, Germanio CD, Spencer BR, Notari E, Bravo M, Lanteri MC, Green V, Briggs-Hagen M, Coughlin MM, Stramer SL, Opsomer J, Busch MP. Estimates of SARS-CoV-2 Seroprevalence and Incidence of Primary SARS-CoV-2 Infections Among Blood Donors, by COVID-19 Vaccination Status - United States, April 2021-September 2022. MMWR. MORBIDITY AND MORTALITY WEEKLY REPORT 2023; 72:601-605. [PMID: 37262007 DOI: 10.15585/mmwr.mm7222a3] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Changes in testing behaviors and reporting requirements have hampered the ability to estimate the U.S. SARS-CoV-2 incidence (1). Hybrid immunity (immunity derived from both previous infection and vaccination) has been reported to provide better protection than that from infection or vaccination alone (2). To estimate the incidence of infection and the prevalence of infection- or vaccination-induced antibodies (or both), data from a nationwide, longitudinal cohort of blood donors were analyzed. During the second quarter of 2021 (April-June), an estimated 68.4% of persons aged ≥16 years had infection- or vaccination-induced SARS-CoV-2 antibodies, including 47.5% from vaccination alone, 12.0% from infection alone, and 8.9% from both. By the third quarter of 2022 (July-September), 96.4% had SARS-CoV-2 antibodies from previous infection or vaccination, including 22.6% from infection alone and 26.1% from vaccination alone; 47.7% had hybrid immunity. Prevalence of hybrid immunity was lowest among persons aged ≥65 years (36.9%), the group with the highest risk for severe disease if infected, and was highest among those aged 16-29 years (59.6%). Low prevalence of infection-induced and hybrid immunity among older adults reflects the success of public health infection prevention efforts while also highlighting the importance of older adults staying up to date with recommended COVID-19 vaccination, including at least 1 bivalent dose.*,†.
Collapse
|
54
|
Sciaudone M, Cutshaw MK, McClean CM, Lacayo R, Kharabora O, Murray K, Strohminger S, Zivanovich MM, Gurnett R, Markmann AJ, Salgado EM, Bhowmik DR, Castro-Arroyo E, Boyce RM, Aiello AE, Richardson D, Juliano JJ, Bowman NM. Seroepidemiology and risk factors for SARS-CoV-2 infection among household members of food processing and farm workers in North Carolina. IJID REGIONS 2023; 7:164-169. [PMID: 37034427 PMCID: PMC10032047 DOI: 10.1016/j.ijregi.2023.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 03/24/2023]
Abstract
Background Racial and ethnic minorities have borne a disproportionate burden from coronavirus disease 2019 (COVID-19). Certain essential occupations, including food processing and farm work, employ large numbers of Hispanic migrant workers and have been shown to carry an especially high risk of infection. Methods This observational cohort study measured the seroprevalence of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and assessed the risk factors for seropositivity among food processing and farm workers, and members of their households, in North Carolina, USA. Participants completed questionnaires, blood samples were collected, and an enzyme-linked immunosorbent assay was used to assess SARS-CoV-2 seropositivity. Univariate and multi-variate analyses were undertaken to identify risk factors associated with seropositivity, using generalized estimating equations to account for household clustering. Findings Among the 218 participants, 94.5% were Hispanic, and SARS-CoV-2 seropositivity was 50.0%. Most seropositive individuals did not report a history of illness compatible with COVID-19. Attending church, having a prior history of COVID-19, having a seropositive household member, and speaking Spanish as one's primary language were associated with SARS-CoV-2 seropositivity, while preventive behaviours were not. Interpretation These findings underscore the substantial burden of COVID-19 among a population of mostly Hispanic essential workers and their households in rural North Carolina. This study contributes to a large body of evidence showing that Hispanic Americans have suffered a disproportionate burden of COVID-19. This study also highlights the epidemiologic importance of viral transmission within the household.
Collapse
Affiliation(s)
- Michael Sciaudone
- Department of Medicine, Section of Infectious Diseases, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Center for Intelligent Molecular Diagnostics, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | | | | | - Roberto Lacayo
- Infectious Disease Epidemiology and Ecology Laboratory, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Oksana Kharabora
- Infectious Disease Epidemiology and Ecology Laboratory, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Katherine Murray
- Infectious Disease Epidemiology and Ecology Laboratory, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Stephen Strohminger
- Infectious Disease Epidemiology and Ecology Laboratory, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Miriana Moreno Zivanovich
- Infectious Disease Epidemiology and Ecology Laboratory, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Rachel Gurnett
- Infectious Disease Epidemiology and Ecology Laboratory, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Alena J. Markmann
- Department of Medicine, Division of Infectious Diseases, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Emperatriz Morales Salgado
- Infectious Disease Epidemiology and Ecology Laboratory, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - D. Ryan Bhowmik
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Edwin Castro-Arroyo
- Infectious Disease Epidemiology and Ecology Laboratory, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Ross M. Boyce
- Institute for Global Health and Infectious Diseases, University of North Carolina, Chapel Hill, North Carolina, USA
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina, USA
- Carolina Population Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Allison E. Aiello
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York, USA
- Robert N Butler Columbia Aging Center, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - David Richardson
- Department of Environmental and Occupational Health, Program in Public Health, University of California – Irvine, Irvine, California, USA
| | - Jonathan J. Juliano
- Department of Medicine, Division of Infectious Diseases, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina, USA
- Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Natalie M. Bowman
- Department of Medicine, Division of Infectious Diseases, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| |
Collapse
|
55
|
Fox SJ, Javan E, Pasco R, Gibson GC, Betke B, Herrera-Diestra JL, Woody S, Pierce K, Johnson KE, Johnson-León M, Lachmann M, Meyers LA. Disproportionate impacts of COVID-19 in a large US city. PLoS Comput Biol 2023; 19:e1011149. [PMID: 37262052 DOI: 10.1371/journal.pcbi.1011149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 05/02/2023] [Indexed: 06/03/2023] Open
Abstract
COVID-19 has disproportionately impacted individuals depending on where they live and work, and based on their race, ethnicity, and socioeconomic status. Studies have documented catastrophic disparities at critical points throughout the pandemic, but have not yet systematically tracked their severity through time. Using anonymized hospitalization data from March 11, 2020 to June 1, 2021 and fine-grain infection hospitalization rates, we estimate the time-varying burden of COVID-19 by age group and ZIP code in Austin, Texas. During this 15-month period, we estimate an overall 23.7% (95% CrI: 22.5-24.8%) infection rate and 29.4% (95% CrI: 28.0-31.0%) case reporting rate. Individuals over 65 were less likely to be infected than younger age groups (11.2% [95% CrI: 10.3-12.0%] vs 25.1% [95% CrI: 23.7-26.4%]), but more likely to be hospitalized (1,965 per 100,000 vs 376 per 100,000) and have their infections reported (53% [95% CrI: 49-57%] vs 28% [95% CrI: 27-30%]). We used a mixed effect poisson regression model to estimate disparities in infection and reporting rates as a function of social vulnerability. We compared ZIP codes ranking in the 75th percentile of vulnerability to those in the 25th percentile, and found that the more vulnerable communities had 2.5 (95% CrI: 2.0-3.0) times the infection rate and only 70% (95% CrI: 60%-82%) the reporting rate compared to the less vulnerable communities. Inequality persisted but declined significantly over the 15-month study period. Our results suggest that further public health efforts are needed to mitigate local COVID-19 disparities and that the CDC's social vulnerability index may serve as a reliable predictor of risk on a local scale when surveillance data are limited.
Collapse
Affiliation(s)
- Spencer J Fox
- Department of Epidemiology & Biostatistics, University of Georgia, Athens, Georgia, United States of America
- Institute of Bioinformatics, University of Georgia, Athens, Georgia, United States of America
- Center for the Ecology of Infectious Diseases, University of Georgia, Athens, Georgia, United States of America
| | - Emily Javan
- Department of Integrative Biology, The University of Texas at Austin, Austin, Texas, United States of America
| | - Remy Pasco
- Department of Industrial Engineering, The University of Texas at Austin, Austin, Texas, United States of America
| | - Graham C Gibson
- Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Briana Betke
- Department of Integrative Biology, The University of Texas at Austin, Austin, Texas, United States of America
| | - José L Herrera-Diestra
- Department of Integrative Biology, The University of Texas at Austin, Austin, Texas, United States of America
| | - Spencer Woody
- Department of Integrative Biology, The University of Texas at Austin, Austin, Texas, United States of America
| | - Kelly Pierce
- The Texas Advanced Computing Center, The University of Texas at Austin, Austin, Texas, United States of America
| | - Kaitlyn E Johnson
- The Rockefeller Foundation, New York, New York, United States of America
| | - Maureen Johnson-León
- Department of Integrative Biology, The University of Texas at Austin, Austin, Texas, United States of America
| | - Michael Lachmann
- The Santa Fe Institute, Santa Fe, New Mexico, United States of America
| | - Lauren Ancel Meyers
- Department of Integrative Biology, The University of Texas at Austin, Austin, Texas, United States of America
- The Santa Fe Institute, Santa Fe, New Mexico, United States of America
| |
Collapse
|
56
|
Weinberger DM, Bhaskaran K, Korves C, Lucas BP, Columbo JA, Vashi A, Davies L, Justice AC, Rentsch CT. Absolute and relative excess mortality across demographic and clinical subgroups during the COVID-19 pandemic: an individual-level cohort study from a nationwide healthcare system of US Veterans. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.05.12.23289900. [PMID: 37293086 PMCID: PMC10246058 DOI: 10.1101/2023.05.12.23289900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Background Most analyses of excess mortality during the COVID-19 pandemic have employed aggregate data. Individual-level data from the largest integrated healthcare system in the US may enhance understanding of excess mortality. Methods We performed an observational cohort study following patients receiving care from the Department of Veterans Affairs (VA) between 1 March 2018 and 28 February 2022. We estimated excess mortality on an absolute scale (i.e., excess mortality rates, number of excess deaths), and a relative scale by measuring the hazard ratio (HR) for mortality comparing pandemic and pre-pandemic periods, overall, and within demographic and clinical subgroups. Comorbidity burden and frailty were measured using the Charlson Comorbidity Index and Veterans Aging Cohort Study Index, respectively. Results Of 5,905,747 patients, median age was 65.8 years and 91% were men. Overall, the excess mortality rate was 10.0 deaths/1000 person-years (PY), with a total of 103,164 excess deaths and pandemic HR of 1.25 (95% CI 1.25-1.26). Excess mortality rates were highest among the most frail patients (52.0/1000 PY) and those with the highest comorbidity burden (16.3/1000 PY). However, the largest relative mortality increases were observed among the least frail (HR 1.31, 95% CI 1.30-1.32) and those with the lowest comorbidity burden (HR 1.44, 95% CI 1.43-1.46). Conclusions Individual-level data offered crucial clinical and operational insights into US excess mortality patterns during the COVID-19 pandemic. Notable differences emerged among clinical risk groups, emphasising the need for reporting excess mortality in both absolute and relative terms to inform resource allocation in future outbreaks.
Collapse
Affiliation(s)
- Daniel M. Weinberger
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, US
- Center for Interdisciplinary Research on AIDS, Yale School of Public Health, New Haven, CT, US
| | - Krishnan Bhaskaran
- Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
| | - Caroline Korves
- Clinical Epidemiology Program, Department of Veterans Affairs Medical Center, White River Junction, VT
| | - Brian P. Lucas
- VA Outcomes Group, Department of Veterans Affairs Medical Center, White River Junction, VT, US
- The Dartmouth Institute for Health Policy & Clinical Practice, Geisel School of Medicine at Dartmouth, Hanover, NH, US
| | - Jesse A. Columbo
- VA Outcomes Group, Department of Veterans Affairs Medical Center, White River Junction, VT, US
- The Dartmouth Institute for Health Policy & Clinical Practice, Geisel School of Medicine at Dartmouth, Hanover, NH, US
- Section of Vascular Surgery, Dartmouth Hitchcock Medical Center, Lebanon, NH, US
| | - Anita Vashi
- Center for Innovation to Implementation, VA Palo Alto Health Care System, Menlo Park, CA, US
- Department of Emergency Medicine, University of California, San Francisco, CA, US
| | - Louise Davies
- VA Outcomes Group, Department of Veterans Affairs Medical Center, White River Junction, VT, US
- The Dartmouth Institute for Health Policy & Clinical Practice, Geisel School of Medicine at Dartmouth, Hanover, NH, US
- Department of Surgery - Otolaryngology Head & Neck Surgery, Geisel School of Medicine at Dartmouth, Hanover, NH, US
| | - Amy C. Justice
- Center for Interdisciplinary Research on AIDS, Yale School of Public Health, New Haven, CT, US
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, US
- VA Connecticut Healthcare System, Department of Veterans Affairs, West Haven, CT, US
| | - Christopher T. Rentsch
- Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, US
- VA Connecticut Healthcare System, Department of Veterans Affairs, West Haven, CT, US
| |
Collapse
|
57
|
Byrum JR, Waltari E, Janson O, Guo SM, Folkesson J, Chhun BB, Vinden J, Ivanov IE, Forst ML, Li H, Larson AG, Blackmon L, Liu Z, Wu W, Ahyong V, Tato CM, McCutcheon KM, Hoh R, Kelly JD, Martin JN, Peluso MJ, Henrich TJ, Deeks SG, Prakash M, Greenhouse B, Mehta SB, Pak JE. MultiSero: An Open-Source Multiplex-ELISA Platform for Measuring Antibody Responses to Infection. Pathogens 2023; 12:671. [PMID: 37242341 PMCID: PMC10221076 DOI: 10.3390/pathogens12050671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/24/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
A multiplexed enzyme-linked immunosorbent assay (ELISA) that simultaneously measures antibody binding to multiple antigens can extend the impact of serosurveillance studies, particularly if the assay approaches the simplicity, robustness, and accuracy of a conventional single-antigen ELISA. Here, we report on the development of multiSero, an open-source multiplex ELISA platform for measuring antibody responses to viral infection. Our assay consists of three parts: (1) an ELISA against an array of proteins in a 96-well format; (2) automated imaging of each well of the ELISA array using an open-source plate reader; and (3) automated measurement of optical densities for each protein within the array using an open-source analysis pipeline. We validated the platform by comparing antibody binding to Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) antigens in 217 human sera samples, showing high sensitivity (0.978), specificity (0.977), positive predictive value (0.978), and negative predictive value (0.977) for classifying seropositivity, a high correlation of multiSero determined antibody titers with commercially available SARS-CoV-2 antibody tests, and antigen-specific changes in antibody titer dynamics upon vaccination. The open-source format and accessibility of our multiSero platform can contribute to the adoption of multiplexed ELISA arrays for serosurveillance studies, for SARS-CoV-2 and other pathogens of significance.
Collapse
Affiliation(s)
- Janie R. Byrum
- Chan Zuckerberg Biohub—San Francisco, San Francisco, CA 94158, USA
| | - Eric Waltari
- Chan Zuckerberg Biohub—San Francisco, San Francisco, CA 94158, USA
| | - Owen Janson
- Division of HIV, Infectious Disease, and Global Medicine, University of California, San Francisco, CA 94143, USA
- EPPIcenter Program, University of California, San Francisco, CA 94143, USA
| | - Syuan-Ming Guo
- Chan Zuckerberg Biohub—San Francisco, San Francisco, CA 94158, USA
| | - Jenny Folkesson
- Chan Zuckerberg Biohub—San Francisco, San Francisco, CA 94158, USA
| | - Bryant B. Chhun
- Chan Zuckerberg Biohub—San Francisco, San Francisco, CA 94158, USA
| | - Joanna Vinden
- Infectious Diseases and Immunity Graduate Program, University of California, Berkeley, CA 94720-3370, USA
| | - Ivan E. Ivanov
- Chan Zuckerberg Biohub—San Francisco, San Francisco, CA 94158, USA
| | - Marcus L. Forst
- Chan Zuckerberg Biohub—San Francisco, San Francisco, CA 94158, USA
- Department of Applied Physics, Stanford University, Stanford, CA 94305, USA
| | - Hongquan Li
- Department of Electrical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Adam G. Larson
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Lena Blackmon
- Chan Zuckerberg Biohub—San Francisco, San Francisco, CA 94158, USA
| | - Ziwen Liu
- Chan Zuckerberg Biohub—San Francisco, San Francisco, CA 94158, USA
| | - Wesley Wu
- Chan Zuckerberg Biohub—San Francisco, San Francisco, CA 94158, USA
| | - Vida Ahyong
- Chan Zuckerberg Biohub—San Francisco, San Francisco, CA 94158, USA
| | - Cristina M. Tato
- Chan Zuckerberg Biohub—San Francisco, San Francisco, CA 94158, USA
| | | | - Rebecca Hoh
- Division of HIV, Infectious Disease, and Global Medicine, University of California, San Francisco, CA 94143, USA
| | - J. Daniel Kelly
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA 94158, USA
| | - Jeffrey N. Martin
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA 94158, USA
| | - Michael J. Peluso
- Division of HIV, Infectious Disease, and Global Medicine, University of California, San Francisco, CA 94143, USA
| | - Timothy J. Henrich
- Division of Experimental Medicine, University of California, San Francisco, CA 94110, USA
| | - Steven G. Deeks
- Division of HIV, Infectious Disease, and Global Medicine, University of California, San Francisco, CA 94143, USA
| | - Manu Prakash
- Chan Zuckerberg Biohub—San Francisco, San Francisco, CA 94158, USA
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Bryan Greenhouse
- Chan Zuckerberg Biohub—San Francisco, San Francisco, CA 94158, USA
- Division of HIV, Infectious Disease, and Global Medicine, University of California, San Francisco, CA 94143, USA
- EPPIcenter Program, University of California, San Francisco, CA 94143, USA
| | - Shalin B. Mehta
- Chan Zuckerberg Biohub—San Francisco, San Francisco, CA 94158, USA
| | - John E. Pak
- Chan Zuckerberg Biohub—San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
58
|
Dean NE, Howard DH, Lopman BA. Serological Studies and the Value of Information. Am J Public Health 2023; 113:517-519. [PMID: 36893371 PMCID: PMC10088957 DOI: 10.2105/ajph.2023.307245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2023] [Indexed: 03/11/2023]
Affiliation(s)
- Natalie E Dean
- Natalie E. Dean, David H. Howard, and Benjamin A. Lopman are with the Rollins School of Public Health, Emory University, Atlanta, GA
| | - David H Howard
- Natalie E. Dean, David H. Howard, and Benjamin A. Lopman are with the Rollins School of Public Health, Emory University, Atlanta, GA
| | - Benjamin A Lopman
- Natalie E. Dean, David H. Howard, and Benjamin A. Lopman are with the Rollins School of Public Health, Emory University, Atlanta, GA
| |
Collapse
|
59
|
Owusu-Boaitey N, Russell TW, Meyerowitz-Katz G, Levin AT, Herrera-Esposito D. Dynamics of SARS-CoV-2 seroassay sensitivity: a systematic review and modelling study. Euro Surveill 2023; 28:2200809. [PMID: 37227301 PMCID: PMC10283460 DOI: 10.2807/1560-7917.es.2023.28.21.2200809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 03/10/2023] [Indexed: 05/26/2023] Open
Abstract
BackgroundSerological surveys have been the gold standard to estimate numbers of SARS-CoV-2 infections, the dynamics of the epidemic, and disease severity. Serological assays have decaying sensitivity with time that can bias their results, but there is a lack of guidelines to account for this phenomenon for SARS-CoV-2.AimOur goal was to assess the sensitivity decay of seroassays for detecting SARS-CoV-2 infections, the dependence of this decay on assay characteristics, and to provide a simple method to correct for this phenomenon.MethodsWe performed a systematic review and meta-analysis of SARS-CoV-2 serology studies. We included studies testing previously diagnosed, unvaccinated individuals, and excluded studies of cohorts highly unrepresentative of the general population (e.g. hospitalised patients).ResultsOf the 488 screened studies, 76 studies reporting on 50 different seroassays were included in the analysis. Sensitivity decay depended strongly on the antigen and the analytic technique used by the assay, with average sensitivities ranging between 26% and 98% at 6 months after infection, depending on assay characteristics. We found that a third of the included assays departed considerably from manufacturer specifications after 6 months.ConclusionsSeroassay sensitivity decay depends on assay characteristics, and for some types of assays, it can make manufacturer specifications highly unreliable. We provide a tool to correct for this phenomenon and to assess the risk of decay for a given assay. Our analysis can guide the design and interpretation of serosurveys for SARS-CoV-2 and other pathogens and quantify systematic biases in the existing serology literature.
Collapse
Affiliation(s)
- Nana Owusu-Boaitey
- Case Western Reserve University School of Medicine, Cleveland, United States
- These authors contributed equally to this work
| | - Timothy W Russell
- Centre for the Mathematical Modelling of Infectious Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | | | - Andrew T Levin
- Dartmouth College, Hanover, United States
- National Bureau for Economic Research, Cambridge, United States
- Centre for Economic Policy Research, London, United Kingdom
| | - Daniel Herrera-Esposito
- These authors contributed equally to this work
- Department of Psychology, University of Pennsylvania, Philadelphia, United States
- Laboratorio de Neurociencias, Universidad de la República, Montevideo, Uruguay
- Centro Interdisciplinario en Ciencia de Datos y Aprendizaje Automático, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
60
|
Gallian P, Hozé N, Brisbarre N, Saba Villarroel PM, Nurtop E, Isnard C, Pastorino B, Richard P, Morel P, Cauchemez S, de Lamballerie X. SARS-CoV-2 IgG seroprevalence surveys in blood donors before the vaccination campaign, France 2020-2021. iScience 2023; 26:106222. [PMID: 36818722 PMCID: PMC9930380 DOI: 10.1016/j.isci.2023.106222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 12/20/2022] [Accepted: 02/11/2023] [Indexed: 02/17/2023] Open
Abstract
We conducted a cross-sectional study for SARS-CoV-2 anti-S1 IgG prevalence in French blood donors (n = 32605), from March-2020 to January-2021. A mathematical model combined seroprevalence with a daily number of hospital admissions to estimate the probability of hospitalization upon infection and determine the number of infections while correcting for antibody decay. There was an overall seroprevalence increase over the study period and we estimate that ∼15% of the French population had been infected by SARS-CoV-2 by January-2021. The infection/hospitalization ratio increased with age, from 0.31% (18-30yo) to 4.5% (61-70yo). Half of the IgG-S1 positive individuals had no detectable antibodies 4 to 5 months after infection. The seroprevalence in group O donors (7.43%) was lower (p = 0.003) than in A, B, and AB donors (8.90%). We conclude, based on seroprevalence data and mathematical modeling, that a large proportion of the French population was unprotected against severe disease prior to the vaccination campaign.
Collapse
Affiliation(s)
- Pierre Gallian
- Établissement Français du Sang, La Plaine Saint Denis 93218, France
- Unité des Virus Émergents (UVE: Aix-Marseille University - IRD 190 - Inserm 1207), 13005 Marseille, France
| | - Nathanaël Hozé
- Mathematical Modelling of Infectious Diseases Unit, Institut Pasteur, Université Paris Cité, UMR2000, CNRS, 75015 Paris, France
| | - Nadège Brisbarre
- Unité des Virus Émergents (UVE: Aix-Marseille University - IRD 190 - Inserm 1207), 13005 Marseille, France
- Établissement Français du Sang Provence Alpes Côte d'Azur et Corse, 13005 Marseille France
| | | | - Elif Nurtop
- Unité des Virus Émergents (UVE: Aix-Marseille University - IRD 190 - Inserm 1207), 13005 Marseille, France
| | - Christine Isnard
- Unité des Virus Émergents (UVE: Aix-Marseille University - IRD 190 - Inserm 1207), 13005 Marseille, France
- Établissement Français du Sang Provence Alpes Côte d'Azur et Corse, 13005 Marseille France
| | - Boris Pastorino
- Unité des Virus Émergents (UVE: Aix-Marseille University - IRD 190 - Inserm 1207), 13005 Marseille, France
| | - Pascale Richard
- Établissement Français du Sang, La Plaine Saint Denis 93218, France
| | - Pascal Morel
- Établissement Français du Sang, La Plaine Saint Denis 93218, France
- UMR RIGHT 1098, Inserm, Établissement Français du Sang, University of Franche-Comté, 25000 Besançon, France
| | - Simon Cauchemez
- Mathematical Modelling of Infectious Diseases Unit, Institut Pasteur, Université Paris Cité, UMR2000, CNRS, 75015 Paris, France
| | - Xavier de Lamballerie
- Unité des Virus Émergents (UVE: Aix-Marseille University - IRD 190 - Inserm 1207), 13005 Marseille, France
| |
Collapse
|
61
|
García-Carreras B, Hitchings MDT, Johansson MA, Biggerstaff M, Slayton RB, Healy JM, Lessler J, Quandelacy T, Salje H, Huang AT, Cummings DAT. Accounting for assay performance when estimating the temporal dynamics in SARS-CoV-2 seroprevalence in the U.S. Nat Commun 2023; 14:2235. [PMID: 37076502 PMCID: PMC10115837 DOI: 10.1038/s41467-023-37944-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 04/06/2023] [Indexed: 04/21/2023] Open
Abstract
Reconstructing the incidence of SARS-CoV-2 infection is central to understanding the state of the pandemic. Seroprevalence studies are often used to assess cumulative infections as they can identify asymptomatic infection. Since July 2020, commercial laboratories have conducted nationwide serosurveys for the U.S. CDC. They employed three assays, with different sensitivities and specificities, potentially introducing biases in seroprevalence estimates. Using models, we show that accounting for assays explains some of the observed state-to-state variation in seroprevalence, and when integrating case and death surveillance data, we show that when using the Abbott assay, estimates of proportions infected can differ substantially from seroprevalence estimates. We also found that states with higher proportions infected (before or after vaccination) had lower vaccination coverages, a pattern corroborated using a separate dataset. Finally, to understand vaccination rates relative to the increase in cases, we estimated the proportions of the population that received a vaccine prior to infection.
Collapse
Affiliation(s)
- Bernardo García-Carreras
- Department of Biology, University of Florida, Gainesville, FL, USA.
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA.
| | - Matt D T Hitchings
- Department of Biostatistics, University of Florida, Gainesville, FL, USA
| | - Michael A Johansson
- COVID-19 Response, US Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Matthew Biggerstaff
- COVID-19 Response, US Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Rachel B Slayton
- COVID-19 Response, US Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Jessica M Healy
- COVID-19 Response, US Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Justin Lessler
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Carolina Population Center, Chapel Hill, NC, USA
| | | | - Henrik Salje
- Department of Genetics, University of Cambridge, Cambridge, UK
| | - Angkana T Huang
- Department of Genetics, University of Cambridge, Cambridge, UK
| | - Derek A T Cummings
- Department of Biology, University of Florida, Gainesville, FL, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA
| |
Collapse
|
62
|
Offergeld R, Preußel K, Zeiler T, Aurich K, Baumann-Baretti BI, Ciesek S, Corman VM, Dienst V, Drosten C, Görg S, Greinacher A, Grossegesse M, Haller S, Heuft HG, Hofmann N, Horn PA, Houareau C, Gülec I, Jiménez Klingberg CL, Juhl D, Lindemann M, Martin S, Neuhauser HK, Nitsche A, Ohme J, Peine S, Sachs UJ, Schaade L, Schäfer R, Scheiblauer H, Schlaud M, Schmidt M, Umhau M, Vollmer T, Wagner FF, Wieler LH, Wilking H, Ziemann M, Zimmermann M, der Heiden MA. Monitoring the SARS-CoV-2 Pandemic: Prevalence of Antibodies in a Large, Repetitive Cross-Sectional Study of Blood Donors in Germany—Results from the SeBluCo Study 2020–2022. Pathogens 2023; 12:pathogens12040551. [PMID: 37111436 PMCID: PMC10144823 DOI: 10.3390/pathogens12040551] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/17/2023] [Accepted: 03/27/2023] [Indexed: 04/05/2023] Open
Abstract
SARS-CoV-2 serosurveillance is important to adapt infection control measures and estimate the degree of underreporting. Blood donor samples can be used as a proxy for the healthy adult population. In a repeated cross-sectional study from April 2020 to April 2021, September 2021, and April/May 2022, 13 blood establishments collected 134,510 anonymised specimens from blood donors in 28 study regions across Germany. These were tested for antibodies against the SARS-CoV-2 spike protein and nucleocapsid, including neutralising capacity. Seroprevalence was adjusted for test performance and sampling and weighted for demographic differences between the sample and the general population. Seroprevalence estimates were compared to notified COVID-19 cases. The overall adjusted SARS-CoV-2 seroprevalence remained below 2% until December 2020 and increased to 18.1% in April 2021, 89.4% in September 2021, and to 100% in April/May 2022. Neutralising capacity was found in 74% of all positive specimens until April 2021 and in 98% in April/May 2022. Our serosurveillance allowed for repeated estimations of underreporting from the early stage of the pandemic onwards. Underreporting ranged between factors 5.1 and 1.1 in the first two waves of the pandemic and remained well below 2 afterwards, indicating an adequate test strategy and notification system in Germany.
Collapse
Affiliation(s)
- Ruth Offergeld
- Robert Koch Institute, Nordufer 20, 13353 Berlin, Germany
| | - Karina Preußel
- Robert Koch Institute, Nordufer 20, 13353 Berlin, Germany
| | - Thomas Zeiler
- German Red Cross Blood Service West, 58097 Hagen, Germany
| | - Konstanze Aurich
- Institute for Immunology and Transfusion Medicine, University Medicine Greifswald, Sauerbruchstrasse, 17475 Greifswald, Germany
| | | | - Sandra Ciesek
- Institute for Medical Virology, German Centre for Infection Research, External Partner Site Frankfurt, University Hospital, Goethe University Frankfurt am Main, 39120 Frankfurt am Main, Germany
| | - Victor M. Corman
- Institute of Virology, German National Reference Laboratory for Coronavirus, Charité—University Medicine Berlin, 10117 Berlin, Germany
| | | | - Christian Drosten
- Institute of Virology, German National Reference Laboratory for Coronavirus, Charité—University Medicine Berlin, 10117 Berlin, Germany
| | - Siegfried Görg
- Institute of Transfusion Medicine, University Hospital of Schleswig-Holstein, Lübeck/Kiel, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - Andreas Greinacher
- Institute for Immunology and Transfusion Medicine, University Medicine Greifswald, Sauerbruchstrasse, 17475 Greifswald, Germany
| | | | | | - Hans-Gert Heuft
- Institute of Transfusion Medicine and Immunohaematology/Blood Bank, University Hospital Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
| | | | - Peter A. Horn
- Institute for Transfusion Medicine, University Hospital Essen, Hufelandstraße 55, 45147 Essen, Germany
| | | | - Ilay Gülec
- Institute of Transfusion Medicine and Immunohematology, German Red Cross Blood Transfusion Service Baden-Württemberg—Hessen, Sandhofstraße 1, 60528 Frankfurt am Main, Germany
| | | | - David Juhl
- Institute of Transfusion Medicine, University Hospital of Schleswig-Holstein, Lübeck/Kiel, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - Monika Lindemann
- Institute for Transfusion Medicine, University Hospital Essen, Hufelandstraße 55, 45147 Essen, Germany
| | - Silke Martin
- Bavarian Red Cross Blood Service, Herzog-Heinrich-Str. 2, 80336 München, Germany
| | | | | | - Julia Ohme
- German Red Cross Blood Service NSTOB, Eldagsener Straße 38, 31832 Springe, Germany
| | - Sven Peine
- Institute of Transfusion Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Ulrich J. Sachs
- Center for Transfusion Medicine and Haemotherapy, University Hospital Giessen and Marburg, Langhansstr. 7, 35392 Giessen, Germany
| | - Lars Schaade
- Robert Koch Institute, Nordufer 20, 13353 Berlin, Germany
| | - Richard Schäfer
- Institute for Transfusion Medicine and Gene Therapy, Faculty of Medicine, Medical Center—University of Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany
| | | | - Martin Schlaud
- Robert Koch Institute, Nordufer 20, 13353 Berlin, Germany
| | - Michael Schmidt
- Institute of Transfusion Medicine and Immunohematology, German Red Cross Blood Transfusion Service Baden-Württemberg—Hessen, Sandhofstraße 1, 60528 Frankfurt am Main, Germany
| | - Markus Umhau
- Institute for Transfusion Medicine and Gene Therapy, Faculty of Medicine, Medical Center—University of Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany
| | - Tanja Vollmer
- Heart and Diabetes Centre NRW, Institute for Laboratory and Transfusion Medicine, Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany
| | - Franz F. Wagner
- German Red Cross Blood Service NSTOB, Eldagsener Straße 38, 31832 Springe, Germany
| | | | | | - Malte Ziemann
- Institute of Transfusion Medicine, University Hospital of Schleswig-Holstein, Lübeck/Kiel, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | | | | |
Collapse
|
63
|
Thon V, Piler P, Pavlík T, Andrýsková L, Doležel K, Kostka D, Pikhart H, Bobák M, Klánová J. Investigation of SARS-CoV-2 seroprevalence in relation to natural infection and vaccination between October 2020 and September 2021 in the Czech Republic: a prospective national cohort study. BMJ Open 2023; 13:e068258. [PMID: 36898746 PMCID: PMC10008433 DOI: 10.1136/bmjopen-2022-068258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 02/15/2023] [Indexed: 03/12/2023] Open
Abstract
OBJECTIVE Examine changes in SARS-CoV-2 seropositivity before and during the national vaccination campaign in the Czech Republic. DESIGN Prospective national population-based cohort study. SETTING Masaryk University, RECETOX, Brno. PARTICIPANTS 22 130 persons provided blood samples at two time points approximately 5-7 months apart, between October 2020 and March 2021 (phase I, before vaccination), and between April and September 2021 (during vaccination campaign). OUTCOME MEASURES Antigen-specific humoral immune response was analysed by detection of IgG antibodies against the SARS-CoV-2 spike protein by commercial chemiluminescent immunoassays. Participants completed a questionnaire that included personal information, anthropometric data, self-reported results of previous RT-PCR tests (if performed), history of symptoms compatible with COVID-19 and records of COVID-19 vaccination. Seroprevalence was compared between calendar periods, previous RT-PCR results, vaccination and other individual characteristics. RESULTS Before vaccination (phase I), seroprevalence increased from 15% in October 2020 to 56% in March 2021. By the end of phase II, in September 2021, prevalence increased to 91%; the highest seroprevalence was seen among vaccinated persons with and without previous SARS-CoV-2 infection (99.7% and 97.2%, respectively), while the lowest seroprevalence was found among unvaccinated persons with no signs of disease (26%). Vaccination rates were lower in persons who were seropositive in phase I but increased with age and body mass index. Only 9% of unvaccinated subjects who were seropositive in phase I became seronegative by phase II. CONCLUSIONS The rapid increase in seropositivity during the second wave of the COVID-19 epidemic (covered by phase I of this study) was followed by a similarly steep rise in seroprevalence during the national vaccination campaign, reaching seropositivity rates of over 97% among vaccinated persons.
Collapse
Affiliation(s)
- Vojtěch Thon
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Pavel Piler
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Tomáš Pavlík
- Institute of Biostatistics and Analyses, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Lenka Andrýsková
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
| | | | - David Kostka
- Health Insurance Company of the Ministry of the Interior of the Czech Republic, Prague, Czech Republic
| | - Hynek Pikhart
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
- Department of Epidemiology and Public Health, University College London, London WC1E 6BT, UK
| | - Martin Bobák
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
- Department of Epidemiology and Public Health, University College London, London WC1E 6BT, UK
| | - Jana Klánová
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
| |
Collapse
|
64
|
Plumb ID, Fette LM, Tjaden AH, Feldstein L, Saydah S, Ahmed A, Link-Gelles R, Wierzba TF, Berry AA, Friedman-Klabanoff D, Larsen MP, Runyon MS, Ward LM, Santos RP, Ward J, Weintraub WS, Edelstein S, Uschner D. Estimated COVID-19 vaccine effectiveness against seroconversion from SARS-CoV-2 Infection, March-October, 2021. Vaccine 2023; 41:2596-2604. [PMID: 36932031 PMCID: PMC9995303 DOI: 10.1016/j.vaccine.2023.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 03/02/2023] [Accepted: 03/03/2023] [Indexed: 03/11/2023]
Abstract
BACKGROUND Monitoring the effectiveness of COVID-19 vaccines against SARS-CoV-2 infections remains important to inform public health responses. Estimation of vaccine effectiveness (VE) against serological evidence of SARS-CoV-2 infection might provide an alternative measure of the benefit of vaccination against infection. METHODS We estimated mRNA COVID-19 vaccine effectiveness (VE) against development of SARS-CoV-2 anti-nucleocapsid antibodies in March-October 2021, during which the Delta variant became predominant. Participants were enrolled from four participating healthcare systems in the United States, and completed electronic surveys that included vaccination history. Dried blood spot specimens collected on a monthly basis were analyzed for anti-spike antibodies, and, if positive, anti-nucleocapsid antibodies. We used detection of new anti-nucleocapsid antibodies to indicate SARS-CoV-2 infection, and estimated VE by comparing 154 case-participants with new detection of anti-nucleocapsid antibodies to 1,540 seronegative control-participants matched by calendar period. Using conditional logistic regression, we estimated VE ≥ 14 days after the 2nd dose of an mRNA vaccine compared with no receipt of a COVID-19 vaccine dose, adjusting for age group, healthcare worker occupation, urban/suburban/rural residence, healthcare system region, and reported contact with a person testing positive for SARS-CoV-2. RESULTS Among individuals who completed a primary series, estimated VE against seroconversion from SARS-CoV-2 infection was 88.8% (95% confidence interval [CI], 79.6%-93.9%) after any mRNA vaccine, 87.8% (95% CI, 75.9%-93.8%) after BioNTech vaccine and 91.7% (95% CI, 75.7%-97.2%) after Moderna vaccine. VE was estimated to be lower ≥ 3 months after dose 2 compared with < 3 months after dose 2, and among participants who were older or had underlying health conditions, although confidence intervals overlapped between subgroups. CONCLUSIONS VE estimates generated using infection-induced antibodies were consistent with published estimates from clinical trials and observational studies that used virologic tests to confirm infection during the same period. Our findings support recommendations for eligible adults to remain up to date with COVID-19 vaccination.
Collapse
Affiliation(s)
- Ian D Plumb
- Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA 30333, USA.
| | - Lida M Fette
- Biostatistics Center, Milken Institute School of Public Health, The George Washington University, 6110 Executive Blvd., Suite 750, Rockville, MD 20852, USA
| | - Ashley H Tjaden
- Biostatistics Center, Milken Institute School of Public Health, The George Washington University, 6110 Executive Blvd., Suite 750, Rockville, MD 20852, USA
| | - Leora Feldstein
- Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA 30333, USA
| | - Sharon Saydah
- Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA 30333, USA
| | - Amina Ahmed
- Atrium Health Levine Children's Hospital, 1000 Blythe Blvd, Charlotte, NC 28203, USA
| | - Ruth Link-Gelles
- Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA 30333, USA
| | - Thomas F Wierzba
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Andrea A Berry
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, 685 W. Baltimore Street, Room 480, Baltimore, MD 21201, USA
| | - DeAnna Friedman-Klabanoff
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, 685 W. Baltimore Street, Room 480, Baltimore, MD 21201, USA
| | - Moira P Larsen
- Medstar Health Research Institute, 6525 Belcrest Road, Suite 700, Hyattsville, MD 20782, USA
| | - Michael S Runyon
- Department of Emergency Medicine, Atrium Health Carolinas Medical Center, 1000 Blythe Blvd, Charlotte, NC 28203, USA
| | - Lori M Ward
- University of Mississippi Medical Center, 2500 N State St, Jackson, MS 39216, USA
| | - Roberto P Santos
- University of Mississippi Medical Center, 2500 N State St, Jackson, MS 39216, USA
| | - Johnathan Ward
- Vysnova Partners, 8400 Corporate Drive Suite 130, Landover, MD 20785, USA
| | - William S Weintraub
- Medstar Health Research Institute, 6525 Belcrest Road, Suite 700, Hyattsville, MD 20782, USA
| | - Sharon Edelstein
- Biostatistics Center, Milken Institute School of Public Health, The George Washington University, 6110 Executive Blvd., Suite 750, Rockville, MD 20852, USA
| | - Diane Uschner
- Biostatistics Center, Milken Institute School of Public Health, The George Washington University, 6110 Executive Blvd., Suite 750, Rockville, MD 20852, USA
| |
Collapse
|
65
|
Pisanic N, Antar AAR, Kruczynski KL, Gregory Rivera M, Dhakal S, Spicer K, Randad PR, Pekosz A, Klein SL, Betenbaugh MJ, Detrick B, Clarke W, Thomas DL, Manabe YC, Heaney CD. Methodological approaches to optimize multiplex oral fluid SARS-CoV-2 IgG assay performance and correlation with serologic and neutralizing antibody responses. J Immunol Methods 2023; 514:113440. [PMID: 36773929 PMCID: PMC9911157 DOI: 10.1016/j.jim.2023.113440] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 01/25/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023]
Abstract
BACKGROUND Oral fluid (hereafter, saliva) is a non-invasive and attractive alternative to blood for SARS-CoV-2 IgG testing; however, the heterogeneity of saliva as a matrix poses challenges for immunoassay performance. OBJECTIVES To optimize performance of a magnetic microparticle-based multiplex immunoassay (MIA) for SARS-CoV-2 IgG measurement in saliva, with consideration of: i) threshold setting and validation across different MIA bead batches; ii) sample qualification based on salivary total IgG concentration; iii) calibration to U.S. SARS-CoV-2 serological standard binding antibody units (BAU); and iv) correlations with blood-based SARS-CoV-2 serological and neutralizing antibody (nAb) assays. METHODS The salivary SARS-CoV-2 IgG MIA included 2 nucleocapsid (N), 3 receptor-binding domain (RBD), and 2 spike protein (S) antigens. Gingival crevicular fluid (GCF) swab saliva samples were collected before December 2019 (n = 555) and after molecular test-confirmed SARS-CoV-2 infection from 113 individuals (providing up to 5 repeated-measures; n = 398) and used to optimize and validate MIA performance (total n = 953). Combinations of IgG responses to N, RBD and S and total salivary IgG concentration (μg/mL) as a qualifier of nonreactive samples were optimized and validated, calibrated to the U.S. SARS-CoV-2 serological standard, and correlated with blood-based SARS-CoV-2 IgG ELISA and nAb assays. RESULTS The sum of signal to cutoff (S/Co) to all seven MIA SARS-CoV-2 antigens and disqualification of nonreactive saliva samples with ≤15 μg/mL total IgG led to correct classification of 62/62 positives (sensitivity [Se] = 100.0%; 95% confidence interval [CI] = 94.8%, 100.0%) and 108/109 negatives (specificity [Sp] = 99.1%; 95% CI = 97.3%, 100.0%) at 8-million beads coupling scale and 80/81 positives (Se = 98.8%; 95% CI = 93.3%, 100.0%] and 127/127 negatives (Sp = 100%; 95% CI = 97.1%, 100.0%) at 20-million beads coupling scale. Salivary SARS-CoV-2 IgG crossed the MIA cutoff of 0.1 BAU/mL on average 9 days post-COVID-19 symptom onset and peaked around day 30. Among n = 30 matched saliva and plasma samples, salivary SARS-CoV-2 MIA IgG levels correlated with corresponding-antigen plasma ELISA IgG (N: ρ = 0.76, RBD: ρ = 0.83, S: ρ = 0.82; all p < 0.001). Correlations of plasma SARS-CoV-2 nAb assay area under the curve (AUC) with salivary MIA IgG (N: ρ = 0.68, RBD: ρ = 0.78, S: ρ = 0.79; all p < 0.001) and with plasma ELISA IgG (N: ρ = 0.76, RBD: ρ = 0.79, S: ρ = 0.76; p < 0.001) were similar. CONCLUSIONS A salivary SARS-CoV-2 IgG MIA produced consistently high Se (> 98.8%) and Sp (> 99.1%) across two bead coupling scales and correlations with nAb responses that were similar to blood-based SARS-CoV-2 IgG ELISA data. This non-invasive salivary SARS-CoV-2 IgG MIA could increase engagement of vulnerable populations and improve broad understanding of humoral immunity (kinetics and gaps) within the evolving context of booster vaccination, viral variants and waning immunity.
Collapse
Affiliation(s)
- Nora Pisanic
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA.
| | - Annukka A R Antar
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Kate L Kruczynski
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Magdielis Gregory Rivera
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Santosh Dhakal
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Kristoffer Spicer
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Pranay R Randad
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Andrew Pekosz
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA; Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Sabra L Klein
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA; Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Michael J Betenbaugh
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Barbara Detrick
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - William Clarke
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - David L Thomas
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA; Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Yukari C Manabe
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA; Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Christopher D Heaney
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA; Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA; Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
66
|
Van Buren NL, Rajbhandary S, Reynolds V, Gorlin JB, Stramer SL, Notari EP, Conti G, Katz L, Stubbs JR, van Buskirk CM, Kuttner K, Smith DL, Ngamsuntikul SG, Pandey S, Ward DC, Ziman A, Hiskey M, Townsend M, Sachais BS. Demographics of first-time donors returning for donation during the pandemic: COVID-19 convalescent plasma versus standard blood product donors. Transfusion 2023; 63:552-563. [PMID: 36550639 PMCID: PMC9880744 DOI: 10.1111/trf.17229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/30/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Previous studies have demonstrated low first-time donor return rates (DRR) following catastrophic events. Little is known, however, about the influence of demographic factors on the DRR of first-time donors during the COVID-19 pandemic, including the unique motivation of COVID-19 convalescent plasma (CCP) donors as compared to non-CCP donors. STUDY DESIGN AND METHODS Thirteen blood collection organizations submitted deidentified data from first-time CCP and non-CCP donors returning for regular (non-CCP) donations during the pandemic. DRR was calculated as frequencies. Demographic factors associated with returning donors: race/ethnicity, gender, and generation (Gen Z: 19-24, Millennial: 25-40, Gen X: 41-56, and Boomer: ≥57 years old), within the CCP and non-CCP first-time cohorts were compared using chi-square test at p < .05 statistical significance. RESULTS From March 2020 through December 2021, there were a total of 44,274 first-time CCP and 980,201 first-time non-CCP donors. DRR were 14.6% (range 11.9%-43.3%) and 46.6% (range 10.0%-76.9%) for CCP and non-CCP cohorts, respectively. Age over 40 years (Gen X and Boomers), female gender, and White race were each associated with higher return in both donor cohorts (p < .001). For the non-CCP return donor cohort, the Millennial and Boomers were comparable. CONCLUSION The findings demonstrate differences in returning donor trends between the two donor cohorts. The motivation of a first-time CCP donor may be different than that of a non-CCP donor. Further study to improve first-time donor engagement would be worthwhile to expand the donor base with a focus on blood donor diversity emphasizing engagement of underrepresented minorities and younger donors.
Collapse
Affiliation(s)
- Nancy L. Van Buren
- Division of New York Blood CenterInnovative Blood ResourcesSaint PaulMinnesotaUSA
- Division of New York Blood CenterCommunity Blood Center of Greater Kansas CityKansas CityMissouriUSA
| | | | - Vanessa Reynolds
- Division of New York Blood CenterInnovative Blood ResourcesSaint PaulMinnesotaUSA
| | - Jed B. Gorlin
- Division of New York Blood CenterInnovative Blood ResourcesSaint PaulMinnesotaUSA
- Division of New York Blood CenterCommunity Blood Center of Greater Kansas CityKansas CityMissouriUSA
| | | | | | - Galen Conti
- Scientific AffairsAmerican Red CrossRockvilleMarylandUSA
| | | | - James R. Stubbs
- Division of Transfusion Medicine, Department of Laboratory Medicine and PathologyMayo ClinicRochesterMinnesotaUSA
| | - Camille M. van Buskirk
- Division of Transfusion Medicine, Department of Laboratory Medicine and PathologyMayo ClinicRochesterMinnesotaUSA
| | - Kip Kuttner
- Miller‐Keystone Blood CenterBethlehemPennsylvaniaUSA
| | | | | | | | - Dawn C. Ward
- Division of Transfusion Medicine, Department of Pathology and Laboratory MedicineDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
| | - Alyssa Ziman
- Division of Transfusion Medicine, Department of Pathology and Laboratory MedicineDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
| | | | | | | |
Collapse
|
67
|
Irizar P, Pan D, Kapadia D, Bécares L, Sze S, Taylor H, Amele S, Kibuchi E, Divall P, Gray LJ, Nellums LB, Katikireddi SV, Pareek M. Ethnic inequalities in COVID-19 infection, hospitalisation, intensive care admission, and death: a global systematic review and meta-analysis of over 200 million study participants. EClinicalMedicine 2023; 57:101877. [PMID: 36969795 PMCID: PMC9986034 DOI: 10.1016/j.eclinm.2023.101877] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 02/02/2023] [Accepted: 02/02/2023] [Indexed: 03/08/2023] Open
Abstract
Background COVID-19 has exacerbated existing ethnic inequalities in health. Little is known about whether inequalities in severe disease and deaths, observed globally among minoritised ethnic groups, relates to greater infection risk, poorer prognosis, or both. We analysed global data on COVID-19 clinical outcomes examining inequalities between people from minoritised ethnic groups compared to the ethnic majority group. Methods Databases (MEDLINE, EMBASE, EMCARE, CINAHL, Cochrane Library) were searched from 1st December 2019 to 3rd October 2022, for studies reporting original clinical data for COVID-19 outcomes disaggregated by ethnicity: infection, hospitalisation, intensive care unit (ICU) admission, and mortality. We assessed inequalities in incidence and prognosis using random-effects meta-analyses, with Grading of Recommendations Assessment, Development, and Evaluation (GRADE) use to assess certainty of findings. Meta-regressions explored the impact of region and time-frame (vaccine roll-out) on heterogeneity. PROSPERO: CRD42021284981. Findings 77 studies comprising over 200,000,000 participants were included. Compared with White majority populations, we observed an increased risk of testing positive for infection for people from Black (adjusted Risk Ratio [aRR]:1.78, 95% CI:1.59-1.99, I2 = 99.1), South Asian (aRR:3.00, 95% CI:1.59-5.66, I2 = 99.1), Mixed (aRR:1.64, 95% CI:1.02-1.67, I2 = 93.2) and Other ethnic groups (aRR:1.36, 95% CI:1.01-1.82, I2 = 85.6). Black, Hispanic, and South Asian people were more likely to be seropositive. Among population-based studies, Black and Hispanic ethnic groups and Indigenous peoples had an increased risk of hospitalisation; Black, Hispanic, South Asian, East Asian and Mixed ethnic groups and Indigenous peoples had an increased risk of ICU admission. Mortality risk was increased for Hispanic, Mixed, and Indigenous groups. Smaller differences were seen for prognosis following infection. Following hospitalisation, South Asian, East Asian, Black and Mixed ethnic groups had an increased risk of ICU admission, and mortality risk was greater in Mixed ethnic groups. Certainty of evidence ranged from very low to moderate. Interpretation Our study suggests that systematic ethnic inequalities in COVID-19 health outcomes exist, with large differences in exposure risk and some differences in prognosis following hospitalisation. Response and recovery interventions must focus on tackling drivers of ethnic inequalities which increase exposure risk and vulnerabilities to severe disease, including structural racism and racial discrimination. Funding ESRC:ES/W000849/1.
Collapse
Affiliation(s)
- Patricia Irizar
- School of Social Sciences, University of Manchester, United Kingdom
| | - Daniel Pan
- Department of Respiratory Sciences, University of Leicester, United Kingdom
- Department of Infection and HIV Medicine, University Hospitals Leicester NHS Trust, United Kingdom
- Li Ka Shing Centre for Health Information and Discovery, Oxford Big Data Institute, University of Oxford, United Kingdom
- NIHR Leicester Biomedical Research Centre, United Kingdom
| | - Dharmi Kapadia
- School of Social Sciences, University of Manchester, United Kingdom
| | - Laia Bécares
- Department of Global Health and Social Medicine, King's College London, United Kingdom
| | - Shirley Sze
- Department of Cardiovascular Sciences, University of Leicester, United Kingdom
| | - Harry Taylor
- School of Social Sciences, University of Manchester, United Kingdom
| | - Sarah Amele
- MRC/CSO Social & Public Health Sciences Unit, University of Glasgow, United Kingdom
| | - Eliud Kibuchi
- MRC/CSO Social & Public Health Sciences Unit, University of Glasgow, United Kingdom
| | - Pip Divall
- University Hospitals of Leicester, Education Centre Library, Glenfield Hospital and Leicester Royal Infirmary, United Kingdom
| | - Laura J Gray
- Department of Health Sciences, University of Leicester, United Kingdom
| | - Laura B Nellums
- Division of Epidemiology and Public Health, School of Medicine, University of Nottingham, United Kingdom
| | | | - Manish Pareek
- Department of Respiratory Sciences, University of Leicester, United Kingdom
- Department of Infection and HIV Medicine, University Hospitals Leicester NHS Trust, United Kingdom
- NIHR Leicester Biomedical Research Centre, United Kingdom
| |
Collapse
|
68
|
Germain M, Lewin A, Bazin R, Dieudé M, Perreault J, Boivin A, Grégoire Y, Renaud C. Cohort profile: A Québec-based plasma donor biobank to study COVID-19 immunity (PlasCoV). BMJ Open 2023; 13:e068803. [PMID: 36822809 PMCID: PMC9950588 DOI: 10.1136/bmjopen-2022-068803] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/25/2023] Open
Abstract
PURPOSE The long-term humoral immunity to COVID-19 is not well understood owing to the continuous emergence of new variants of concern, the evolving vaccine-induced and infection-induced immunity, and the limited duration of follow-up in previous studies. As the sole blood service in Québec (Canada), Héma-Québec established a COVID-19-focused biobank ('PlasCoV') in April 2021. PARTICIPANTS As of January 2022, the biobank included 86 483 plasma samples from 15 502 regular donors (age range=18-84 years, females=49.7%), for an average of 5.6 donations per donor. Nearly two-thirds (65.6%) of biobank donors made at least two donations, with many donors having provided samples prevaccination and postvaccination (3061 (19.7%)) or preinfection and postinfection (131 (0.8%)), thus allowing for longitudinal studies on vaccine-induced and infection-induced immunity. FINDINGS TO DATE A study that used PlasCoV samples revealed that previously infected individuals who received a single dose of the BNT162b2 COVID-19 vaccine exhibited the strongest immune response. By contrast, SARS-CoV-2-naïve individuals required two vaccine doses to produce a maximal immune response. Furthermore, the results of a four-phase seroprevalence study indicated that the antinucleocapsid (N) response wanes rapidly, so that up to one-third of previously infected donors were seronegative for anti-N. FUTURE PLANS Donations from individuals who consented to participate before 1 October 2022 will be collected up until 31 March 2023. This plasma biobank will facilitate the conduct of longitudinal studies on COVID-19 immunity, thus helping to provide valuable insights into the anti-SARS-CoV-2 immune response and its persistence, and the effects of vaccination and variants on the specificity of the anti-SARS-CoV-2 immune response.
Collapse
Affiliation(s)
- Marc Germain
- Medical Affairs and Innovation, Héma-Québec, Québec, Montreal and Québec, Canada
| | - Antoine Lewin
- Medical Affairs and Innovation, Héma-Québec, Québec, Montreal and Québec, Canada
| | - Renée Bazin
- Medical Affairs and Innovation, Héma-Québec, Québec, Montreal and Québec, Canada
| | - Mélanie Dieudé
- Medical Affairs and Innovation, Héma-Québec, Québec, Montreal and Québec, Canada
| | - Josée Perreault
- Medical Affairs and Innovation, Héma-Québec, Québec, Montreal and Québec, Canada
| | - Amélie Boivin
- Medical Affairs and Innovation, Héma-Québec, Québec, Montreal and Québec, Canada
| | - Yves Grégoire
- Medical Affairs and Innovation, Héma-Québec, Québec, Montreal and Québec, Canada
| | - Christian Renaud
- Medical Affairs and Innovation, Héma-Québec, Québec, Montreal and Québec, Canada
| |
Collapse
|
69
|
O’Brien SF, Caffrey N, Yi QL, Bolotin S, Janjua NZ, Binka M, Thanh CQ, Stein DR, Lang A, Colquhoun A, Pambrun C, Reedman CN, Drews SJ. Cross-Canada Variability in Blood Donor SARS-CoV-2 Seroprevalence by Social Determinants of Health. Microbiol Spectr 2023; 11:e0335622. [PMID: 36625634 PMCID: PMC9927354 DOI: 10.1128/spectrum.03356-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 12/07/2022] [Indexed: 01/11/2023] Open
Abstract
We compared the seroprevalence of SARS-CoV-2 anti-nucleocapsid antibodies in blood donors across Canadian regions in 2021. The seroprevalence was the highest in Alberta and the Prairies, and it was so low in Atlantic Canada that few correlates were observed. Being male and of young age were predictive of seropositivity. Racialization was associated with higher seroprevalence in British Columbia and Ontario but not in Alberta and the Prairies. Living in a materially deprived neighborhood predicted higher seroprevalence, but it was more linear across quintiles in Alberta and the Prairies, whereas in British Columbia and Ontario, the most affluent 60% were similarly low and the most deprived 40% similarly elevated. Living in a more socially deprived neighborhood (more single individuals and one parent families) was associated with lower seroprevalence in British Columbia and Ontario but not in Alberta and the Prairies. These data show striking variability in SARS-CoV-2 seroprevalence across regions by social determinants of health. IMPORTANCE Canadian blood donors are a healthy adult population that shows clear disparities associated with racialization and material deprivation. This underscores the pervasiveness of the socioeconomic gradient on SARS-CoV-2 infections in Canada. We identify regional differences in the relationship between SARS-CoV-2 seroprevalence and social determinants of health. Cross-Canada studies, such as ours, are rare because health information is under provincial jurisdiction and is not available in sufficient detail in national data sets, whereas other national seroprevalence studies have insufficient sample sizes for regional comparisons. Ours is the largest seroprevalence study in Canada. An important strength of our study is the interpretation input from a public health team that represented multiple Canadian provinces. Our blood donor seroprevalence study has informed Canadian public health policy at national and provincial levels since the start of the SARS-CoV-2 pandemic.
Collapse
Affiliation(s)
- Sheila F. O’Brien
- Epidemiology and Surveillance, Canadian Blood Services, Ottawa, Ontario, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
| | - Niamh Caffrey
- Epidemiology and Surveillance, Canadian Blood Services, Ottawa, Ontario, Canada
| | - Qi-Long Yi
- Epidemiology and Surveillance, Canadian Blood Services, Ottawa, Ontario, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
| | - Shelly Bolotin
- Center for Vaccine Preventable Disease, University of Toronto, Toronto, Ontario, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Public Health Ontario, Toronto, Ontario, Canada
| | - Naveed Z. Janjua
- BC Centre for Disease Control, British Columbia, Vancouver, Canada
- School of Population and Public Health, University of British Columbia, British Columbia, Vancouver, Canada
| | - Mawuena Binka
- BC Centre for Disease Control, British Columbia, Vancouver, Canada
| | - Caroline Quach Thanh
- Department of Microbiology, Infectious Diseases & Immunology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
- Infection Prevention & Control, Clinical Department of Laboratory Medicine, CHU Sainte-Justine, Montreal, Quebec, Canada
| | - Derek R. Stein
- Cadham Provincial Laboratory, Winnipeg, Manitoba, Canada
- Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Amanda Lang
- Roy Romanow Provincial laboratory, Saskatchewan Health Authority, Regina, Saskatchewan, Canada
| | - Amy Colquhoun
- Population Health Assessment, Alberta Health, Edmonton, Alberta, Canada
- School of Public Health, University of Alberta, Edmonton, Alberta, Canada
| | - Chantale Pambrun
- Medical Affairs & Innovation, Canadian Blood Services, Ottawa, Ontario, Canada
- Department of Pathology & Laboratory Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Cassandra N. Reedman
- Epidemiology and Surveillance, Canadian Blood Services, Ottawa, Ontario, Canada
- Public Health Agency of Canada, Ottawa, Ontario, Canada
| | - Steven J. Drews
- Medical Microbiology Department, Canadian Blood Services, Edmonton, Alberta, Canada
- Department of Laboratory Medicine & Pathology, Division of Diagnostic and Applied Microbiology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
70
|
Klaassen F, Chitwood MH, Cohen T, Pitzer VE, Russi M, Swartwood NA, Salomon JA, Menzies NA. Population Immunity to Pre-Omicron and Omicron Severe Acute Respiratory Syndrome Coronavirus 2 Variants in US States and Counties Through 1 December 2021. Clin Infect Dis 2023; 76:e350-e359. [PMID: 35717642 PMCID: PMC9214178 DOI: 10.1093/cid/ciac438] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/20/2022] [Accepted: 05/28/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Both severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and coronavirus disease 2019 (COVID-19) vaccination contribute to population-level immunity against SARS-CoV-2. This study estimated the immunological exposure and effective protection against future SARS-CoV-2 infection in each US state and county over 2020-2021 and how this changed with the introduction of the Omicron variant. METHODS We used a Bayesian model to synthesize estimates of daily SARS-CoV-2 infections, vaccination data and estimates of the relative rates of vaccination conditional on infection status to estimate the fraction of the population with (1) immunological exposure to SARS-CoV-2 (ever infected with SARS-CoV-2 and/or received ≥1 doses of a COVID-19 vaccine), (2) effective protection against infection, and (3) effective protection against severe disease, for each US state and county from 1 January 2020 to 1 December 2021. RESULTS The estimated percentage of the US population with a history of SARS-CoV-2 infection or vaccination as of 1 December 2021 was 88.2% (95% credible interval [CrI], 83.6%-93.5%). Accounting for waning and immune escape, effective protection against the Omicron variant on 1 December 2021 was 21.8% (95% CrI, 20.7%-23.4%) nationally and ranged between 14.4% (13.2%-15.8%; West Virginia) and 26.4% (25.3%-27.8%; Colorado). Effective protection against severe disease from Omicron was 61.2% (95% CrI, 59.1%-64.0%) nationally and ranged between 53.0% (47.3%-60.0%; Vermont) and 65.8% (64.9%-66.7%; Colorado). CONCLUSIONS While more than four-fifths of the US population had prior immunological exposure to SARS-CoV-2 via vaccination or infection on 1 December 2021, only a fifth of the population was estimated to have effective protection against infection with the immune-evading Omicron variant.
Collapse
Affiliation(s)
- Fayette Klaassen
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Melanie H Chitwood
- Department of Epidemiology of Microbial Diseases and Public Health Modeling Unit, Yale School of Public Health, New Haven, Connecticut, USA
| | - Ted Cohen
- Department of Epidemiology of Microbial Diseases and Public Health Modeling Unit, Yale School of Public Health, New Haven, Connecticut, USA
| | - Virginia E Pitzer
- Department of Epidemiology of Microbial Diseases and Public Health Modeling Unit, Yale School of Public Health, New Haven, Connecticut, USA
| | - Marcus Russi
- Department of Epidemiology of Microbial Diseases and Public Health Modeling Unit, Yale School of Public Health, New Haven, Connecticut, USA
| | - Nicole A Swartwood
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Joshua A Salomon
- Department of Health Policy, Stanford University School of Medicine, Stanford, California, USA
| | - Nicolas A Menzies
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| |
Collapse
|
71
|
Wiegand RE, Deng Y, Deng X, Lee A, Meyer WA, Letovsky S, Charles MD, Gundlapalli AV, MacNeil A, Hall AJ, Thornburg NJ, Jones J, Iachan R, Clarke KE. Estimated SARS-CoV-2 antibody seroprevalence trends and relationship to reported case prevalence from a repeated, cross-sectional study in the 50 states and the District of Columbia, United States-October 25, 2020-February 26, 2022. LANCET REGIONAL HEALTH. AMERICAS 2023; 18:100403. [PMID: 36479424 PMCID: PMC9716971 DOI: 10.1016/j.lana.2022.100403] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 11/05/2022] [Accepted: 11/14/2022] [Indexed: 12/03/2022]
Abstract
Background Sero-surveillance of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can reveal trends and differences in subgroups and capture undetected or unreported infections that are not included in case-based surveillance systems. Methods Cross-sectional, convenience samples of remnant sera from clinical laboratories from 51 U.S. jurisdictions were assayed for infection-induced SARS-CoV-2 antibodies biweekly from October 25, 2020, to July 11, 2021, and monthly from September 6, 2021, to February 26, 2022. Test results were analyzed for trends in infection-induced, nucleocapsid-protein seroprevalence using mixed effects models that adjusted for demographic variables and assay type. Findings Analyses of 1,469,792 serum specimens revealed U.S. infection-induced SARS-CoV-2 seroprevalence increased from 8.0% (95% confidence interval (CI): 7.9%-8.1%) in November 2020 to 58.2% (CI: 57.4%-58.9%) in February 2022. The U.S. ratio of the change in estimated seroprevalence to the change in reported case prevalence was 2.8 (CI: 2.8-2.9) during winter 2020-2021, 2.3 (CI: 2.0-2.5) during summer 2021, and 3.1 (CI: 3.0-3.3) during winter 2021-2022. Change in seroprevalence to change in case prevalence ratios ranged from 2.6 (CI: 2.3-2.8) to 3.5 (CI: 3.3-3.7) by region in winter 2021-2022. Interpretation Ratios of the change in seroprevalence to the change in case prevalence suggest a high proportion of infections were not detected by case-based surveillance during periods of increased transmission. The largest increases in the seroprevalence to case prevalence ratios coincided with the spread of the B.1.1.529 (Omicron) variant and with increased accessibility of home testing. Ratios varied by region and season with the highest ratios in the midwestern and southern United States during winter 2021-2022. Our results demonstrate that reported case counts did not fully capture differing underlying infection rates and demonstrate the value of sero-surveillance in understanding the full burden of infection. Levels of infection-induced antibody seroprevalence, particularly spikes during periods of increased transmission, are important to contextualize vaccine effectiveness data as the susceptibility to infection of the U.S. population changes. Funding This work was supported by the Centers for Disease Control and Prevention, Atlanta, Georgia.
Collapse
Affiliation(s)
- Ryan E. Wiegand
- COVID-19 Response, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | | | | | | | | | - Myrna D. Charles
- COVID-19 Response, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Adi V. Gundlapalli
- COVID-19 Response, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Adam MacNeil
- COVID-19 Response, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Aron J. Hall
- COVID-19 Response, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | - Jefferson Jones
- COVID-19 Response, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | - Kristie E.N. Clarke
- COVID-19 Response, Centers for Disease Control and Prevention, Atlanta, GA, USA
| |
Collapse
|
72
|
Lau JJ, Cheng SMS, Leung K, Lee CK, Hachim A, Tsang LCH, Yam KWH, Chaothai S, Kwan KKH, Chai ZYH, Lo THK, Mori M, Wu C, Valkenburg SA, Amarasinghe GK, Lau EHY, Hui DSC, Leung GM, Peiris M, Wu JT. Real-world COVID-19 vaccine effectiveness against the Omicron BA.2 variant in a SARS-CoV-2 infection-naive population. Nat Med 2023; 29:348-357. [PMID: 36652990 PMCID: PMC9941049 DOI: 10.1038/s41591-023-02219-5] [Citation(s) in RCA: 96] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 01/13/2023] [Indexed: 01/19/2023]
Abstract
The SARS-CoV-2 Omicron variant has demonstrated enhanced transmissibility and escape of vaccine-derived immunity. Although first-generation vaccines remain effective against severe disease and death, robust evidence on vaccine effectiveness (VE) against all Omicron infections, irrespective of symptoms, remains sparse. We used a community-wide serosurvey with 5,310 subjects to estimate how vaccination histories modulated risk of infection in infection-naive Hong Kong during a large wave of Omicron BA.2 epidemic in January-July 2022. We estimated that Omicron infected 45% (41-48%) of the local population. Three and four doses of BNT162b2 or CoronaVac were effective against Omicron infection 7 days after vaccination (VE of 48% (95% credible interval 34-64%) and 69% (46-98%) for three and four doses of BNT162b2, respectively; VE of 30% (1-66%) and 56% (6-97%) for three and four doses of CoronaVac, respectively). At 100 days after immunization, VE waned to 26% (7-41%) and 35% (10-71%) for three and four doses of BNT162b2, and to 6% (0-29%) and 11% (0-54%) for three and four doses of CoronaVac. The rapid waning of VE against infection conferred by first-generation vaccines and an increasingly complex viral evolutionary landscape highlight the necessity for rapidly deploying updated vaccines followed by vigilant monitoring of VE.
Collapse
Affiliation(s)
- Jonathan J Lau
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Laboratory of Data Discovery for Health (D24H), Hong Kong SAR, China
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Samuel M S Cheng
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Kathy Leung
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Laboratory of Data Discovery for Health (D24H), Hong Kong SAR, China
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- The University of Hong Kong - Shenzhen Hospital, Shenzhen, China
| | - Cheuk Kwong Lee
- Hong Kong Red Cross Blood Transfusion Service, Hong Kong SAR, People's Republic of China
| | - Asmaa Hachim
- HKU-Pasteur Research Pole, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Leo C H Tsang
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Kenny W H Yam
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Sara Chaothai
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Kelvin K H Kwan
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Zacary Y H Chai
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Tiffany H K Lo
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Laboratory of Data Discovery for Health (D24H), Hong Kong SAR, China
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Masashi Mori
- Research Institute for Bioresources and Biotechnology, Ishikawa Prefectural University, Nonoichi, Japan
| | - Chao Wu
- Department of Pathology and Immunology, Washington University School of Medicine at St. Louis, St. Louis, MO, USA
| | - Sophie A Valkenburg
- HKU-Pasteur Research Pole, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Gaya K Amarasinghe
- Department of Pathology and Immunology, Washington University School of Medicine at St. Louis, St. Louis, MO, USA
| | - Eric H Y Lau
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Laboratory of Data Discovery for Health (D24H), Hong Kong SAR, China
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - David S C Hui
- Department of Medicine and Therapeutics and Stanley Ho Centre for Emerging Infectious Diseases, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Gabriel M Leung
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Laboratory of Data Discovery for Health (D24H), Hong Kong SAR, China
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Malik Peiris
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Centre for Immunology and Infection, Hong Kong SAR, China
| | - Joseph T Wu
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
- Laboratory of Data Discovery for Health (D24H), Hong Kong SAR, China.
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
- The University of Hong Kong - Shenzhen Hospital, Shenzhen, China.
| |
Collapse
|
73
|
Gallian P, Slimani A, Malard L, Morel P, de Lamballerie X. Impact of vaccination on SARS-CoV-2 seroprevalence rate in French blood donors: An assessment as of July 2021. Transfus Clin Biol 2023; 30:25-26. [PMID: 35934226 PMCID: PMC9352401 DOI: 10.1016/j.tracli.2022.08.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 08/02/2022] [Indexed: 02/07/2023]
Affiliation(s)
- Pierre Gallian
- Établissement français du Sang, La Plaine Saint Denis, France; Unité des Virus Émergents (UVE: Aix-Marseille Univ - IRD 190 - Inserm 1207), Marseille, France.
| | - Ahmed Slimani
- Établissement Français du Sang Ile de France, Ivry sur Seine, France
| | - Lucile Malard
- Établissement français du Sang, La Plaine Saint Denis, France
| | - Pascal Morel
- Établissement français du Sang, La Plaine Saint Denis, France
| | - Xavier de Lamballerie
- Unité des Virus Émergents (UVE: Aix-Marseille Univ – IRD 190 – Inserm 1207), Marseille, France
| |
Collapse
|
74
|
Krogsgaard LW, Espenhain L, Tribler S, Sværke Jørgensen C, Hansen CH, Møller FT, Glode Helmuth I, Sönksen UW, Vangsted AM, Ullum H, Ethelberg S. Seroprevalence of SARS-CoV-2 Antibodies in Denmark: Results of Two Nationwide Population-Based Surveys, February and May 2021. Infect Drug Resist 2023; 16:301-312. [PMID: 36683911 PMCID: PMC9851711 DOI: 10.2147/idr.s383491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 12/14/2022] [Indexed: 01/15/2023] Open
Abstract
Background Seroprevalence studies can be used to measure the progression of national COVID-19 epidemics. The Danish National Seroprevalence Survey of SARS-CoV-2 infections (DSS) was conducted as five separate surveys between May 2020 and May 2021. Here, we present results from the two last surveys conducted in February and May 2021. Methods Persons aged 12 or older were randomly selected from the Danish Population Register and those having received COVID-19 vaccination subsequently excluded. Invitations to have blood drawn in local test centers were sent by mail. Samples were analyzed for whole Immunoglobulin by ELISA. Seroprevalence was estimated by sex, age and geography. Comparisons to vaccination uptake and RT-PCR test results were made. Results In February 2021, we found detectable antibodies in 7.2% (95% CI: 6.3-7.9%) of the invited participants (participation rate 25%) and in May 2021 in 8.6% (95% CI: 7.6-9.5%) of the invited (participation rate: 14%). Seroprevalence did not differ by sex, but by age group, generally being higher among the <50 than 50+ year-olds. In May 2021, levels of seroprevalence varied from an estimated 13% (95% CI: 12-15%) in the capital to 5.2% (95% CI: 3.4-7.4%) in rural areas. Combining seroprevalence results with vaccine coverage, estimates of protection against infection in May 2021 varied from 95% among 65+ year-olds down to 10-20% among 12-40 year-olds. In March-May 2021, an estimated 80% of all community SARS-CoV-2 infections were diagnosed by RT-PCR and captured by surveillance. Conclusion Seroprevalence estimates doubled during the 2020-21 winter wave of SARS-CoV-2 infections and then stabilized as vaccinations were rolled out. The epidemic affected large cities and younger people the most. Denmark saw comparatively low infections rates, but high test coverage; an estimated four out of five infections were detected by RT-PCR in March-May 2021.
Collapse
Affiliation(s)
- Lene Wulff Krogsgaard
- Department of Infectious Disease Epidemiology and Prevention, Statens Serum Institut, Copenhagen, Denmark
| | - Laura Espenhain
- Department of Infectious Disease Epidemiology and Prevention, Statens Serum Institut, Copenhagen, Denmark
| | - Siri Tribler
- Department of Infectious Disease Epidemiology and Prevention, Statens Serum Institut, Copenhagen, Denmark
| | | | - Christian Holm Hansen
- Department of Infectious Disease Epidemiology and Prevention, Statens Serum Institut, Copenhagen, Denmark
| | - Frederik Trier Møller
- Department of Infectious Disease Epidemiology and Prevention, Statens Serum Institut, Copenhagen, Denmark
| | - Ida Glode Helmuth
- Department of Infectious Disease Epidemiology and Prevention, Statens Serum Institut, Copenhagen, Denmark
| | - Ute Wolff Sönksen
- Department of Bacteria, Parasites and Fungi, Statens Serum Institut, Copenhagen, Denmark
- TestCentre Denmark, Statens Serum Institut, Copenhagen, Denmark
| | | | - Henrik Ullum
- Division of Infectious Disease Preparedness, Statens Serum Institut, Copenhagen, Denmark
| | - Steen Ethelberg
- Department of Infectious Disease Epidemiology and Prevention, Statens Serum Institut, Copenhagen, Denmark
- Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
75
|
Gatto NM, Freund D, Ogata P, Diaz L, Ibarrola A, Desai M, Aspelund T, Gluckstein D. Correlates of Coronavirus Disease 2019 Inpatient Mortality at a Southern California Community Hospital With a Predominantly Hispanic/Latino Adult Population. Open Forum Infect Dis 2023; 10:ofad011. [PMID: 36726553 PMCID: PMC9887269 DOI: 10.1093/ofid/ofad011] [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: 12/15/2022] [Accepted: 12/06/2023] [Indexed: 01/11/2023] Open
Abstract
Background Studies of inpatient coronavirus disease 2019 (COVID-19) mortality risk factors have mainly used data from academic medical centers or large multihospital databases and have not examined populations with large proportions of Hispanic/Latino patients. In a retrospective cohort study of 4881 consecutive adult COVID-19 hospitalizations at a single community hospital in Los Angeles County with a majority Hispanic/Latino population, we evaluated factors associated with mortality. Methods Data on demographic characteristics, comorbidities, laboratory and clinical results, and COVID-19 therapeutics were abstracted from the electronic medical record. Cox proportional hazards regression modeled statistically significant, independently associated predictors of hospital mortality. Results Age ≥65 years (hazard ratio [HR] = 2.66; 95% confidence interval [CI] = 1.90-3.72), male sex (HR = 1.31; 95% CI = 1.07-1.60), renal disease (HR = 1.52; 95% CI = 1.18-1.95), cardiovascular disease (HR = 1.45; 95% CI = 1.18-1.78), neurological disease (HR = 1.84; 95% CI = 1.41-2.39), D-dimer ≥500 ng/mL (HR = 2.07; 95% CI = 1.43-3.0), and pulse oxygen level <88% (HR = 1.39; 95% CI = 1.13-1.71) were independently associated with increased mortality. Patient household with (1) multiple COVID-19 cases and (2) Asian, Black, or Hispanic compared with White non-Hispanic race/ethnicity were associated with reduced mortality. In hypoxic COVID-19 inpatients, remdesivir, tocilizumab, and convalescent plasma were associated with reduced mortality, and corticosteroid use was associated with increased mortality. Conclusions We corroborate several previously identified mortality risk factors and find evidence that the combination of factors associated with mortality differ between populations.
Collapse
Affiliation(s)
- Nicole M Gatto
- Correspondence: Nicole M. Gatto, MPH, PhD, Adjunct Research Assistant Professor Department of Population and Public Health Sciences Keck School of Medicine University of Southern California 1845 N Soto St, Los Angeles, CA 90032, USA ()
| | - Debbie Freund
- School of Community and Global Health, Claremont Graduate University, Claremont, California, USA,Department of Economic Sciences, Claremont Graduate University, Claremont, California, USA,Department of Health Policy and Management, Fielding School of Public Health, University of California Los Angeles, Los Angeles, California, USA
| | - Pamela Ogata
- School of Community and Global Health, Claremont Graduate University, Claremont, California, USA
| | - Lisa Diaz
- Pomona Valley Hospital and Medical Center, Pomona, California, USA
| | - Ace Ibarrola
- Pomona Valley Hospital and Medical Center, Pomona, California, USA
| | - Mamta Desai
- Pomona Valley Hospital and Medical Center, Pomona, California, USA
| | - Thor Aspelund
- Center for Public Health Sciences, University of Iceland, Reykjavik, Iceland
| | | |
Collapse
|
76
|
Glasser JW, Feng Z, Vo M, Jones JN, Clarke KEN. Analysis of serological surveys of antibodies to SARS-CoV-2 in the United States to estimate parameters needed for transmission modeling and to evaluate and improve the accuracy of predictions. J Theor Biol 2023; 556:111296. [PMID: 36208669 PMCID: PMC9532270 DOI: 10.1016/j.jtbi.2022.111296] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/02/2022] [Accepted: 09/28/2022] [Indexed: 11/23/2022]
Abstract
Seroprevalence studies can estimate proportions of the population that have been infected or vaccinated, including infections that were not reported because of the lack of symptoms or testing. Based on information from studies in the United States from mid-summer 2020 through the end of 2021, we describe proportions of the population with antibodies to SARS-CoV-2 as functions of age and time. Slices through these surfaces at arbitrary times provide initial and target conditions for simulation modeling. They also provide the information needed to calculate age-specific forces of infection, attack rates, and - together with contact rates - age-specific probabilities of infection on contact between susceptible and infectious people. We modified the familiar Susceptible-Exposed-Infectious-Removed (SEIR) model to include features of the biology of COVID-19 that might affect transmission of SARS-CoV-2 and stratified by age and location. We consulted the primary literature or subject matter experts for contact rates and other parameter values. Using time-varying Oxford COVID-19 Government Response Tracker assessments of US state and DC efforts to mitigate the pandemic and compliance with non-pharmaceutical interventions (NPIs) from a YouGov survey fielded in the US during 2020, we estimate that the efficacy of social-distancing when possible and mask-wearing otherwise at reducing susceptibility or infectiousness was 31% during the fall of 2020. Initialized from seroprevalence among people having commercial laboratory tests for purposes other than SARS-CoV-2 infection assessments on 7 September 2020, our age- and location-stratified SEIR population model reproduces seroprevalence among members of the same population on 25 December 2020 quite well. Introducing vaccination mid-December 2020, first of healthcare and other essential workers, followed by older adults, people who were otherwise immunocompromised, and then progressively younger people, our metapopulation model reproduces seroprevalence among blood donors on 4 April 2021 less well, but we believe that the discrepancy is due to vaccinations being under-reported or blood donors being disproportionately vaccinated, if not both. As experimenting with reliable transmission models is the best way to assess the indirect effects of mitigation measures, we determined the impact of vaccination, conditional on NPIs. Results indicate that, during this period, vaccination substantially reduced infections, hospitalizations and deaths. This manuscript was submitted as part of a theme issue on "Modelling COVID-19 and Preparedness for Future Pandemics."
Collapse
Affiliation(s)
- John W Glasser
- National Center for Immunization and Respiratory Diseases, CDC, USA.
| | - Zhilan Feng
- Department of Mathematics, Purdue University, USA; Division of Mathematical Sciences, NSF, USA
| | - MyVan Vo
- Department of Mathematics, Purdue University, USA
| | | | - Kristie E N Clarke
- Center For Surveillance, Epidemiology, and Laboratory Services, CDC, USA
| |
Collapse
|
77
|
Strengers P, O'Brien SF, Politis C, Mayr W, Seifried E, Spencer BR. White paper on pandemic preparedness in the blood supply. Vox Sang 2023; 118:8-15. [PMID: 36427057 DOI: 10.1111/vox.13378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/30/2022] [Accepted: 10/28/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND AND OBJECTIVES In March 2020, the WHO declared the SARS-CoV-2 corona virus a pandemic which caused a great disruption to global society and had a pronounced effect on the worldwide supply of blood. MATERIALS AND METHODS In 2022 an on-line meeting was organised with experts from Austria, Canada, Germany, Greece, Netherlands and United States to explore the opportunities for increasing preparedness within blood systems for a potential future pandemic with similar, or more devastating, consequences. The main themes included the value of preparedness, current risks to the blood supply, supply chain vulnerabilities, and the role of innovation in increasing resiliency and safety. RESULTS Seven key recommendations were formulated and including required actions at different levels. CONCLUSION Although SARS-CoV-2 might be seen as a unique event, global health risks are expected to increase and will affect blood transfusion medicine if no preparedness plans are developed.
Collapse
Affiliation(s)
| | | | - Constantina Politis
- Department of Medicine, Athens University, Athens, Greece.,National Public Health Organization, Athens, Greece
| | - Wolfgang Mayr
- Medical University of Vienna, Vienna, Austria.,Austrian Red Cross Blood Transfusion Service, Vienna, Austria.,European Blood Alliance, Amsterdam, Netherlands
| | - Erhard Seifried
- European Blood Alliance, Amsterdam, Netherlands.,DRK Blutspendedienst, Frankfurt, Germany
| | | |
Collapse
|
78
|
Galea LA, Lee BH, de leon RG, Rajah MN, Einstein G. Beyond sex and gender differences: The case for women's health research. PRINCIPLES OF GENDER-SPECIFIC MEDICINE 2023:699-711. [DOI: 10.1016/b978-0-323-88534-8.00045-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
79
|
Alexopoulos H, Trougakos IP, Dimopoulos MA, Terpos E. Clinical usefulness of testing for severe acute respiratory syndrome coronavirus 2 antibodies. Eur J Intern Med 2023; 107:7-16. [PMID: 36379820 PMCID: PMC9647045 DOI: 10.1016/j.ejim.2022.11.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 10/11/2022] [Accepted: 11/08/2022] [Indexed: 11/11/2022]
Abstract
In the COVID-19 pandemic era, antibody testing against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has proven an invaluable tool and herein we highlight some of the most useful clinical and/or epidemiological applications of humoral immune responses recording. Anti-spike circulating IgGs and SARS-CoV-2 neutralizing antibodies can serve as predictors of disease progression or disease prevention, whereas anti-nucleocapsid antibodies can help distinguishing infection from vaccination. Also, in the era of immunotherapies we address the validity of anti-SARS-CoV-2 antibody monitoring post-infection and/or vaccination following therapies with the popular anti-CD20 monoclonals, as well as in the context of various cancers or autoimmune conditions such as rheumatoid arthritis and multiple sclerosis. Additional crucial applications include population immunosurveillance, either at the general population or at specific communities such as health workers. Finally, we discuss how testing of antibodies in cerebrospinal fluid can inform us on the neurological complications that often accompany COVID-19.
Collapse
Affiliation(s)
- Harry Alexopoulos
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens, 15784, Greece
| | - Ioannis P Trougakos
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens, 15784, Greece
| | - Meletios-Athanasios Dimopoulos
- Department of Clinical Therapeutics, School of Medicine, Alexandra General Hospital, National and Kapodistrian University of Athens, Athens, 11528, Greece
| | - Evangelos Terpos
- Department of Clinical Therapeutics, School of Medicine, Alexandra General Hospital, National and Kapodistrian University of Athens, Athens, 11528, Greece.
| |
Collapse
|
80
|
Tonnetti L, Dodd RY, Burke DD, Saá P, Spencer BR, Xu M, Haynes JM, Stramer SL. A Longitudinal Study of Severe Acute Respiratory Syndrome Coronavirus 2 Antibody Response in a Subset of United States Blood Donors. Open Forum Infect Dis 2022; 10:ofac697. [PMID: 36751647 PMCID: PMC9898875 DOI: 10.1093/ofid/ofac697] [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: 09/23/2022] [Accepted: 12/26/2022] [Indexed: 12/29/2022] Open
Abstract
Background Blood donors were tested for antibodies to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2); resulting antibody levels were monitored over time. Methods Donors reactive to anti-SARS-CoV-2 spike protein (S1-total antibodies) participated in a follow-up study of 18 months. Testing for nucleocapsid antibodies distinguished between vaccination and infection. Vaccination and symptom information were collected for anti-S1-reactive donors by completing a survey. Results The majority of 249 followed donors were over 60 years old (54%), White (90%), and female (58%); 83% had not been vaccinated at enrollment, but by study completion, only 29% remained nonvaccinated. Of the 210 (84%) anti-N-reactive donors, 138 (66%) reported vaccination, whereas 37 (95%) of donors vaccinated and anti-N negative at enrollment remained uninfected. Vaccinated (2 doses) and infected donors showed a steady increase in anti-S1 that increased markedly for vaccinated donors after a booster and infected donors after vaccination (slightly higher for those with hybrid immunity), whereas anti-N levels declined. Most surveyed nonvaccinated donors (65%) reported symptoms, whereas 85% of vaccinated donors were asymptomatic. A coronavirus disease 2019 (COVID-19) diagnosis was reported by 48 (31%) nonvaccinated and 3 (8%) vaccinated donors. Of asymptomatic donors, 38% never tested diagnostically for COVID-19, and 35% tested negative, suggesting an absence of knowledge of the infection. Conclusions Healthy blood donors were vaccinated at high rates and remained mostly asymptomatic and noninfected, whereas approximately two thirds of infected donors reported symptoms. Anti-S1 levels increased while anti-N decreased over 18 months but remained comparable between vaccinated and hybrid immune individuals with dramatic anti-S1 increases after vaccination or boosting.
Collapse
Affiliation(s)
- Laura Tonnetti
- Correspondence: L. Tonnetti, PhD, American Red Cross, Scientific Affairs, 15601 Crabbs Branch Way, Rockville, MD 20855 ()
| | - Roger Y Dodd
- Scientific Affairs, American Red Cross, Rockville, Maryland, USA
| | - Donna D Burke
- Scientific Affairs, American Red Cross, St. Paul, Minnesota, USA
| | - Paula Saá
- Scientific Affairs, American Red Cross, Rockville, Maryland, USA
| | - Bryan R Spencer
- Scientific Affairs, American Red Cross, Dedham, Massachusetts, USA
| | - Meng Xu
- Scientific Affairs, American Red Cross, Rockville, Maryland, USA
| | - James M Haynes
- Scientific Affairs, American Red Cross, Rockville, Maryland, USA
| | | |
Collapse
|
81
|
Pisanic N, Antar AAR, Kruczynski K, Rivera MG, Dhakal S, Spicer K, Randad PR, Pekosz A, Klein SL, Betenbaugh MJ, Detrick B, Clarke W, Thomas DL, Manabe YC, Heaney CD. Methodological approaches to optimize multiplex oral fluid SARS-CoV-2 IgG assay performance and correlation with serologic and neutralizing antibody responses. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.12.22.22283858. [PMID: 36597525 PMCID: PMC9810233 DOI: 10.1101/2022.12.22.22283858] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Background Oral fluid (hereafter, saliva) is a non-invasive and attractive alternative to blood for SARS-CoV-2 IgG testing; however, the heterogeneity of saliva as a matrix poses challenges for immunoassay performance. Objectives To optimize performance of a magnetic microparticle-based multiplex immunoassay (MIA) for SARS-CoV-2 IgG measurement in saliva, with consideration of: i) threshold setting and validation across different MIA bead batches; ii) sample qualification based on salivary total IgG concentration; iii) calibration to U.S. SARS-CoV-2 serological standard binding antibody units (BAU); and iv) correlations with blood-based SARS-CoV-2 serological and neutralizing antibody (nAb) assays. Methods The salivary SARS-CoV-2 IgG MIA included 2 nucleocapsid (N), 3 receptor-binding domain (RBD), and 2 spike protein (S) antigens. Gingival crevicular fluid (GCF) swab saliva samples were collected before December, 2019 (n=555) and after molecular test-confirmed SARS-CoV-2 infection from 113 individuals (providing up to 5 repeated-measures; n=398) and used to optimize and validate MIA performance (total n=953). Combinations of IgG responses to N, RBD and S and total salivary IgG concentration (μg/mL) as a qualifier of nonreactive samples were optimized and validated, calibrated to the U.S. SARS-CoV-2 serological standard, and correlated with blood-based SARS-CoV-2 IgG ELISA and nAb assays. Results The sum of signal to cutoff (S/Co) to all seven MIA SARS-CoV-2 antigens and disqualification of nonreactive saliva samples with ≤15 μg/mL total IgG led to correct classification of 62/62 positives (sensitivity [Se]=100.0%; 95% confidence interval [CI]=94.8%, 100.0%) and 108/109 negatives (specificity [Sp]=99.1%; 95% CI=97.3%, 100.0%) at 8-million beads coupling scale and 80/81 positives (Se=98.8%; 95% CI=93.3%, 100.0%] and 127/127 negatives (Sp=100%; 95% CI=97.1%, 100.0%) at 20-million beads coupling scale. Salivary SARS-CoV-2 IgG crossed the MIA cutoff of 0.1 BAU/mL on average 9 days post-COVID-19 symptom onset and peaked around day 30. Among n=30 matched saliva and plasma samples, salivary SARS-CoV-2 MIA IgG levels correlated with corresponding-antigen plasma ELISA IgG (N: ρ=0.67, RBD: ρ=0.76, S: ρ=0.82; all p <0.0001). Correlations of plasma SARS-CoV-2 nAb assay area under the curve (AUC) with salivary MIA IgG (N: ρ=0.68, RBD: ρ=0.78, S: ρ=0.79; all p <0.0001) and with plasma ELISA IgG (N: ρ=0.76, RBD: ρ=0.79, S: ρ=0.76; p <0.0001) were similar. Conclusions A salivary SARS-CoV-2 IgG MIA produced consistently high Se (>98.8%) and Sp (>99.1%) across two bead coupling scales and correlations with nAb responses that were similar to blood-based SARS-CoV-2 IgG ELISA data. This non-invasive salivary SARS-CoV-2 IgG MIA could increase engagement of vulnerable populations and improve broad understanding of humoral immunity (kinetics and gaps) within the evolving context of booster vaccination, viral variants and waning immunity.
Collapse
Affiliation(s)
- Nora Pisanic
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Annukka A. R. Antar
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Kate Kruczynski
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Magdielis Gregory Rivera
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Santosh Dhakal
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Kristoffer Spicer
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Pranay R. Randad
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Andrew Pekosz
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Sabra L. Klein
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Michael J. Betenbaugh
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Barbara Detrick
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - William Clarke
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - David L. Thomas
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Yukari C. Manabe
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Christopher D. Heaney
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
- Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
82
|
Akinbami LJ, Kruszon-Moran D, Wang CY, Storandt RJ, Clark J, Riddles MK, Mohadjer LK. SARS-CoV-2 Serology and Self-Reported Infection Among Adults - National Health and Nutrition Examination Survey, United States, August 2021-May 2022. MMWR. MORBIDITY AND MORTALITY WEEKLY REPORT 2022; 71:1522-1525. [PMID: 36454698 PMCID: PMC9721142 DOI: 10.15585/mmwr.mm7148a4] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
CDC COVID-19 surveillance systems monitor SARS-CoV-2 antibody prevalence to collect information about asymptomatic, undiagnosed, and unreported disease using national convenience samples of blood donor data from commercial laboratories (1,2). However, nonrandom sampling of data from these systems could affect prevalence estimates (1-3). The National Health and Nutrition Examination Survey (NHANES) collects SARS-CoV-2 serology data among a sample of the general U.S. civilian population (4). In addition, NHANES collects self-reported COVID-19 vaccination and disease history, and its statistical sampling design is not based on health care access or blood donation. Therefore, NHANES data can be used to better quantify asymptomatic SARS-CoV-2 infection prevalence and seropositivity attained through infection without vaccination. Preliminary NHANES 2021-2022 results indicated that 41.6% of adults aged ≥18 years had serology indicative of past infection and that 43.7% of these adults, including 57.1% of non-Hispanic Black or African American (Black) adults, reported never having had COVID-19, possibly representing asymptomatic infection. In addition, 25.5% of adults whose serology indicated past infection reported never having received COVID-19 vaccination. Prevalences of seropositivity in the absence of vaccination were higher among younger adults and Black adults, reflecting the lower observed vaccination rates among these groups (5). These findings raise health equity concerns given the disparities observed in SARS-CoV-2 infection and COVID-19 vaccination. Results from NHANES 2021-2022 can guide ongoing efforts to achieve vaccine equity in COVID-19 primary vaccination series and booster dose coverage.
Collapse
|
83
|
Deng L, Li P, Zhang X, Jiang Q, Turner D, Zhou C, Gao Y, Qian F, Zhang C, Lu H, Zou H, Vermund SH, Qian HZ. Risk of SARS-CoV-2 reinfection: a systematic review and meta-analysis. Sci Rep 2022; 12:20763. [PMID: 36456577 PMCID: PMC9714387 DOI: 10.1038/s41598-022-24220-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 11/11/2022] [Indexed: 12/02/2022] Open
Abstract
This meta-analysis aims to synthesize global evidence on the risk of reinfection among people previously infected with SARS-CoV-2. We systematically searched PubMed, Scopus, Embase and Web of Science as of April 5, 2021. We conducted: (1) meta-analysis of cohort studies containing data sufficient for calculating the incidence rate of SARS-CoV-2 reinfection; (2) systematic review of case reports with confirmed SARS-CoV-2 reinfection cases. The reinfection incidence was pooled by zero-inflated beta distribution. The hazard ratio (HR) between reinfection incidence among previously infected individuals and new infection incidence among infection-naïve individuals was calculated using random-effects models. Of 906 records retrieved and reviewed, 11 studies and 11 case reports were included in the meta-analysis and the systematic review, respectively. The pooled SARS-CoV-2 reinfection incidence rate was 0.70 (standard deviation [SD] 0.33) per 10,000 person-days. The incidence of reinfection was lower than the incidence of new infection (HR = 0.12, 95% confidence interval 0.09-0.17). Our meta-analysis of studies conducted prior to the emergency of the more transmissible Omicron variant showed that people with a prior SARS-CoV-2 infection could be re-infected, and they have a lower risk of infection than those without prior infection. Continuing reviews are needed as the reinfection risk may change due to the rapid evolution of SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Luojia Deng
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Peiqi Li
- School of Acupuncture-Moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xuezhixing Zhang
- Yale School of Public Health, Yale University, 300 George Street, New Haven, CT, USA
| | - Qianxue Jiang
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | | | - Chao Zhou
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yanxiao Gao
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Frank Qian
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Ci Zhang
- Xiangya Nursing School, Central South University, Changsha, China
| | - Hui Lu
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Huachun Zou
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Sten H Vermund
- Yale School of Public Health, Yale University, 300 George Street, New Haven, CT, USA
| | - Han-Zhu Qian
- Yale School of Public Health, Yale University, 300 George Street, New Haven, CT, USA.
- GSK plc, Rockville, MD, USA.
| |
Collapse
|
84
|
Dallas TA, Foster G, Richards RL, Elderd BD. Epidemic time series similarity is related to geographic distance and age structure. Infect Dis Model 2022; 7:690-697. [PMID: 36313152 PMCID: PMC9579807 DOI: 10.1016/j.idm.2022.09.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 11/07/2022] Open
Abstract
Objective More similar locations may have similar infectious disease dynamics. There is clear overlap in putative causes for epidemic similarity, such as geographic distance, age structure, and population size. We compare the effects of these potential drivers on epidemic similarity compared to a baseline assumption that differences in the basic reproductive number (R 0) will translate to differences in epidemic trajectories. Methods Using COVID-19 case counts from United States counties, we explore the importance of geographic distance, population size differences, and age structure dissimilarity on resulting epidemic similarity. Results We find clear effects of geographic space, age structure, population size, and R 0 on epidemic similarity, but notably the effect of age structure was stronger than the baseline assumption that differences in R 0 would be most related to epidemic similarity. Conclusions Together, this highlights the role of spatial and demographic processes on SARS-CoV2 epidemics in the United States.
Collapse
Affiliation(s)
- Tad A. Dallas
- Department of Biological Sciences, University of South Carolina, Columbia, SC, 29208, USA
| | - Grant Foster
- Department of Biological Sciences, University of South Carolina, Columbia, SC, 29208, USA
| | - Robert L. Richards
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, 70802, USA
| | - Bret D. Elderd
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, 70802, USA
| |
Collapse
|
85
|
O'Brien SF, Drews SJ, Lewin A, Russell A, Davison K, Goldman M. How do we decide how representative our donors are for public health surveillance? Transfusion 2022; 62:2431-2437. [PMID: 36193865 DOI: 10.1111/trf.17140] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 09/13/2022] [Accepted: 09/13/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Surveillance of blood donors is fundamental to safety of the blood supply. Such data can also be useful for public health policy but tend to be underutilized. When the COVID-19 pandemic arrived, blood centers around the world measured blood donor SARS-CoV-2 seroprevalence to inform public health policy. There is now a movement toward blood centers becoming more involved in public health research and surveillance post-pandemic. However, blood donors are a healthy population and not representative of all segments of the general population. In this article, we explain how blood centers can evaluate their donor base to understand which part of the general population they are representative of. STUDY DESIGN AND METHODS Methodologic approaches for evaluating samples relative to the target population were reviewed. Blood donor data that are available to most blood centers were identified and application to assess representativeness of blood donors was evaluated. RESULTS Key aspects of blood donor data to compare with general population data include donor selection criteria, health indicators, geography, and demographics. In some cases, statistical adjustment can improve representativeness. DISCUSSION Comparing key blood donor data with corresponding general population data can define the subset of the general population for which a particular blood center's donors may be representative of. We suggest that donors are an ideal convenience population for surveillance of infectious agents which are frequently asymptomatic and main routes of transmission are not deferrable, for studying the natural history of disease in an initially well population, and for vaccination serology surveillance.
Collapse
Affiliation(s)
- Sheila F O'Brien
- Canadian Blood Services, Ottawa, Ontario, Canada.,School of Epidemiology & Public Health, University of Ottawa, Ottawa, Ontario, Canada
| | - Steven J Drews
- Canadian Blood Services, Edmonton, Alberta, Canada.,Department of Laboratory Medicine & Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Antoine Lewin
- Héma-Québec, Montreal, Quebec, Canada.,Faculty of Medicine & Health Sciences, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Alton Russell
- School of Population and Global Health, McGill University, Montreal, Quebec, Canada
| | | | - Mindy Goldman
- Canadian Blood Services, Ottawa, Ontario, Canada.,Department of Pathology & Laboratory Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | | |
Collapse
|
86
|
Sjörs Dahlman A, Anund A. Seroprevalence of SARS-CoV-2 antibodies among public transport workers in Sweden. JOURNAL OF TRANSPORT & HEALTH 2022; 27:101508. [PMID: 36188635 PMCID: PMC9515328 DOI: 10.1016/j.jth.2022.101508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 06/16/2023]
Abstract
INTRODUCTION Public transportation is an essential societal function in crisis situations like the coronavirus disease 2019 (COVID-19) pandemic. Bus drivers and other public transport workers are essential workers that need to keep working despite the risk of contagion. The SARS-CoV-2 virus may pose an occupational health risk to public transport workers and especially to bus drivers as they interact with passengers in a confined area. By analyzing antibodies towards SARS-CoV-2 proteins in blood samples it is possible to measure if an individual has been infected by COVID-19. Here, we report the prevalence of antibodies among bus drivers and other public transport employees in Stockholm, Sweden and relate it to socio-demographic factors. METHODS Seroprevalence of IgG antibodies towards SARS-CoV-2 proteins was investigated in a sample of 262 non-vaccinated public transport workers (182 men and 40 women) recruited between April 26 and May 7, 2021. Most of the participants were bus drivers (n = 222). The relationship between socio-demographic factors and seroprevalence was investigated with logistic regression. RESULTS The seroprevalence was 50% in the total sample of public transport workers. Among bus drivers, 51% were seropositive compared to 44% seropositive among the other public transport workers. The difference was not significant. The seroprevalence was higher than the national seroprevalence in Sweden during the same period (18.3% in non-vaccinated people aged 20-64 years). The logistic regression model using Wald forward selection showed that men had a higher risk of being seropositive (OR 2.7, 95% CI 1.3 - 5.8) and there was a higher risk with increasing number of people in the household (OR 1.3, 95% CI 1.1 - 1.6). CONCLUSIONS These findings could imply an occupational risk for COVID-19 infection among public transport workers. Infection control measures are warranted during virus epidemics to assure bus drives' safety and reduce transmission in public transport.
Collapse
Affiliation(s)
- Anna Sjörs Dahlman
- The Swedish National Road and Transport Research Institute (VTI), Linköping, Sweden
- Department of Electrical Engineering and SAFER Vehicle and Traffic Safety Centre at Chalmers University of Technology, Gothenburg, Sweden
| | - Anna Anund
- The Swedish National Road and Transport Research Institute (VTI), Linköping, Sweden
- Rehabilitation Medicine, Linköping University, Linköping, Sweden and Stockholm University, Stockholm Stress Centre, Stockholm, Sweden
| |
Collapse
|
87
|
Ankomah PO, Siedner MJ, Bhattacharyya RP. Pre-Existing Population Immunity and severe acute respiratory syndrome coronavirus 2 Variant Establishment and Dominance Dynamics in the United States: An Ecological Study. Open Forum Infect Dis 2022; 9:ofac621. [PMID: 36544861 PMCID: PMC9764332 DOI: 10.1093/ofid/ofac621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/15/2022] [Indexed: 12/24/2022] Open
Abstract
We conducted an ecological analysis of the dynamics of Delta and Omicron establishment and dominance in US states. Omicron became the dominant circulating variant later in states with higher population-level immunity. By contrast, population immunity did not impact the maximum rate of takeover by Delta or Omicron from prior variants.
Collapse
Affiliation(s)
- Pierre O Ankomah
- Massachusetts General Hospital, Boston, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
| | - Mark J Siedner
- Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Roby P Bhattacharyya
- Massachusetts General Hospital, Boston, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
| |
Collapse
|
88
|
Freedman ND, Brown L, Newman LM, Jones JM, Benoit TJ, Averhoff F, Bu X, Bayrak K, Lu A, Coffey B, Jackson L, Chanock SJ, Kerlavage AR. COVID-19 SeroHub, an online repository of SARS-CoV-2 seroprevalence studies in the United States. Sci Data 2022; 9:727. [DOI: 10.1038/s41597-022-01830-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 11/09/2022] [Indexed: 11/28/2022] Open
Abstract
AbstractSeroprevalence studies provide useful information about the proportion of the population either vaccinated against SARS-CoV-2, previously infected with the virus, or both. Numerous studies have been conducted in the United States, but differ substantially by dates of enrollment, target population, geographic location, age distribution, and assays used. This can make it challenging to identify and synthesize available seroprevalence data by geographic region or to compare infection-induced versus combined infection- and vaccination-induced seroprevalence. To facilitate public access and understanding, the National Institutes of Health and the Centers for Disease Control and Prevention developed the COVID-19 Seroprevalence Studies Hub (COVID-19 SeroHub, https://covid19serohub.nih.gov/), a data repository in which seroprevalence studies are systematically identified, extracted using a standard format, and summarized through an interactive interface. Within COVID-19 SeroHub, users can explore and download data from 178 studies as of September 1, 2022. Tools allow users to filter results and visualize trends over time, geography, population, age, and antigen target. Because COVID-19 remains an ongoing pandemic, we will continue to identify and include future studies.
Collapse
|
89
|
Cumulative seroprevalence among healthcare workers after the first wave of the COVID-19 pandemic in El Salvador, Central America. Clin Microbiol Infect 2022; 28:1508-1510. [PMID: 35777604 PMCID: PMC9239702 DOI: 10.1016/j.cmi.2022.06.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 06/14/2022] [Accepted: 06/16/2022] [Indexed: 11/24/2022]
|
90
|
Yamayoshi S, Iwatsuki-Horimoto K, Okuda M, Ujie M, Yasuhara A, Murakami J, Duong C, Hamabata T, Ito M, Chiba S, Kobayashi R, Takahashi S, Mitamura K, Hagihara M, Shibata A, Uwamino Y, Hasegawa N, Ebina T, Izumi A, Kato H, Nakajima H, Sugaya N, Seki Y, Iqbal A, Kamimaki I, Yamazaki M, Kawaoka Y, Furuse Y. Age-Stratified Seroprevalence of SARS-CoV-2 Antibodies before and during the Vaccination Era, Japan, February 2020–March 2022. Emerg Infect Dis 2022; 28:2198-2205. [PMID: 36198306 PMCID: PMC9622230 DOI: 10.3201/eid2811.221127] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Japan has reported a relatively small number of COVID-19 cases. Because not all infected persons receive diagnostic tests for COVID-19, the reported number must be lower than the actual number of infections. We assessed SARS-CoV-2 seroprevalence by analyzing >60,000 samples collected in Japan (Tokyo Metropolitan Area and Hokkaido Prefecture) during February 2020–March 2022. The results showed that ≈3.8% of the population had become seropositive by January 2021. The seroprevalence increased with the administration of vaccinations; however, among the elderly, seroprevalence was not as high as the vaccination rate. Among children, who were not eligible for vaccination, infection was spread during the epidemic waves caused by the SARS-CoV-2 Delta and Omicron variants. Nevertheless, seroprevalence for unvaccinated children <5 years of age was as low as 10% as of March 2022. Our study underscores the low incidence of SARS-CoV-2 infection in Japan and the effects of vaccination on immunity at the population level.
Collapse
|
91
|
Binder RA, Fujimori GF, Forconi CS, Reed GW, Silva LS, Lakshmi PS, Higgins A, Cincotta L, Dutta P, Salive MC, Mangolds V, Anya O, Calvo Calle JM, Nixon T, Tang Q, Wessolossky M, Wang Y, Ritacco DA, Bly CS, Fischinger S, Atyeo C, Oluoch PO, Odwar B, Bailey JA, Maldonado-Contreras A, Haran JP, Schmidt AG, Cavacini L, Alter G, Moormann AM. SARS-CoV-2 Serosurveys: How Antigen, Isotype and Threshold Choices Affect the Outcome. J Infect Dis 2022; 227:371-380. [PMID: 36314635 PMCID: PMC9891417 DOI: 10.1093/infdis/jiac431] [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: 07/13/2022] [Revised: 10/21/2022] [Accepted: 10/27/2022] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND Evaluating the performance of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) serological assays and clearly articulating the utility of selected antigens, isotypes, and thresholds is crucial to understanding the prevalence of infection within selected communities. METHODS This cross-sectional study, implemented in 2020, screened PCRconfirmed coronavirus disease 2019 patients (n 86), banked prepandemic and negative samples (n 96), healthcare workers and family members (n 552), and university employees (n 327) for antiSARS-CoV-2 receptor-binding domain, trimeric spike protein, and nucleocapsid protein immunoglobulin (Ig)G and IgA antibodies with a laboratory-developed enzyme-linked immunosorbent assay and tested how antigen, isotype and threshold choices affected the seroprevalence outcomes. The following threshold methods were evaluated: (i) mean 3 standard deviations of the negative controls; (ii) 100 specificity for each antigen-isotype combination; and (iii) the maximal Youden index. RESULTS We found vastly different seroprevalence estimates depending on selected antigens and isotypes and the applied threshold method, ranging from 0.0 to 85.4. Subsequently, we maximized specificity and reported a seroprevalence, based on more than one antigen, ranging from 9.3 to 25.9. CONCLUSIONS This study revealed the importance of evaluating serosurvey tools for antigen-, isotype-, and threshold-specific sensitivity and specificity, to interpret qualitative serosurvey outcomes reliably and consistently across studies.
Collapse
Affiliation(s)
- Raquel A Binder
- Correspondence: Raquel A. Binder, University of Massachusetts Chan Medical School, Worcester, MA 01605 ()
| | | | | | - George W Reed
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Leandro S Silva
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Priya Saikumar Lakshmi
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Amanda Higgins
- Department of Emergency Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Lindsey Cincotta
- Department of Emergency Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Protiva Dutta
- Department of Emergency Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Marie-Claire Salive
- Department of Emergency Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Virginia Mangolds
- Department of Emergency Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Otuwe Anya
- Department of Emergency Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - J Mauricio Calvo Calle
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Thomas Nixon
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Qiushi Tang
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Mireya Wessolossky
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Yang Wang
- MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts, USA
| | - Dominic A Ritacco
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Courtney S Bly
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | | | - Caroline Atyeo
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
| | - Peter O Oluoch
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Boaz Odwar
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Jeffrey A Bailey
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | - Ana Maldonado-Contreras
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - John P Haran
- Department of Emergency Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA,Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Aaron G Schmidt
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA,Department of Microbiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Lisa Cavacini
- MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
| | | |
Collapse
|
92
|
SARS-CoV-2 Seroprevalence among Canadian Blood Donors: The Advance of Omicron. Viruses 2022; 14:v14112336. [PMID: 36366432 PMCID: PMC9695729 DOI: 10.3390/v14112336] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/13/2022] [Accepted: 10/17/2022] [Indexed: 02/01/2023] Open
Abstract
With the emergence of the SARS-CoV-2 Omicron variant in late 2021, Canadian public health case/contact testing was scaled back due to high infection rates with milder symptoms in a highly vaccinated population. We monitored the seroprevalence of SARS-CoV-2 nucleocapsid (anti-N) and spike protein (anti-S) antibodies in blood donors across Canada from September 2021 to June 2022 in 202,123 randomly selected samples. Multivariable logistic regression of anti-N positivity with month, age, sex, racialization, region, material and social deprivation (based on postal code) identified as independent predictors. Piece-wise logistic regression analysed the association between anti-S concentration and month, and anti-N/anti-S positivity. Infection-related seroprevalence (anti-N positive) was 4.38% (95% CI: 3.96, 4.81) in September reaching 50.70% (50.15, 52.16) in June; nearly 100% were anti-S positive throughout. Anti-N positivity was associated with younger age, male sex, the Alberta and Prairies regions, greater material deprivation and less social deprivation (p < 0.001). Anti-S concentration was high initially (3306 U/mL, IQR 4280 U/mL), increased to (13,659 U/mL, IQR 28,224 U/mL) by June (p < 0.001), following the pattern of deployment of the third and fourth vaccine doses and was higher in those that were anti-N positive (p < 0.001). Despite already high vaccination-related seroprevalence, infection-related seroprevalence increased dramatically with the emergence of the Omicron SARS-CoV-2 variant.
Collapse
|
93
|
Jiang Q, Zhang YF, Hu P. Multisystem Inflammatory Syndrome in Children During 3 SARS-CoV-2 Pandemic Waves in Israel. JAMA 2022; 328:1561-1562. [PMID: 36255432 DOI: 10.1001/jama.2022.15217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- Qi Jiang
- Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yan Fang Zhang
- Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Peng Hu
- Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
94
|
Malijan GMB, Edwards T, Agrupis KA, Suzuki S, Villanueva AMG, Sayo AR, De Guzman F, Dimapilis AQ, Solante RM, Telan EO, Umipig DV, Ota K, Nishimura F, Yanagihara K, Salazar MJ, Lopez EB, Ariyoshi K, Smith C. SARS-CoV-2 seroprevalence and infection rate in Manila, Philippines prior to national vaccination program implementation: a repeated cross-sectional analysis. Trop Med Health 2022; 50:75. [PMID: 36221142 PMCID: PMC9552163 DOI: 10.1186/s41182-022-00468-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 10/02/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND SARS-CoV-2 seroepidemiological studies are used to guide public health decision making and to prepare for emerging infectious diseases. Disease occurrence estimates are limited in the Philippines, the country with the highest reported number of coronavirus disease-related deaths in the Western Pacific region. We aimed to estimate SARS-CoV-2 seroprevalence and infection rate among outpatient clinic attendees in Metro Manila prior to the implementation of the national coronavirus disease vaccination program. METHODS We conducted repeated cross-sectional surveys at the animal bite clinic in San Lazaro Hospital, Manila, the Philippines across four periods, 3 months apart, between May 2020 and March 2021. Multivariable logistic regression was used to assess associations between different characteristics and infection status including seropositivity. RESULTS In total 615 participants were enrolled, ranging from 115 to 174 per period. Seroprevalence quadrupled between the first (11.3%) and second (46.8%) periods and plateaued thereafter (third-46.0%, fourth-44.6%). Among seropositive participants, total antibody concentration was comparable throughout the first to third periods but declined between the third and fourth periods. Infection prevalence was comparable across enrollment periods (range 2.9-9.5%). Post-secondary education [aOR 0.42 (95% CI 0.26, 0.67)] was protective, and frontline work [aOR 1.81 (95% CI 1.18, 2.80)] was associated with increased odds of seropositivity. Frontline work status [aOR 2.27 (95% CI 1.10, 4.75)] and large household size [aOR 2.45 (95% CI 1.18, 5.49)] were associated with increased odds of infection. CONCLUSIONS The quadrupling of seroprevalence over 3 months between the first and second enrollment periods coincided with the high burden of infection in Metro Manila in early 2020. Our findings suggest a limit to the rise and potential decline of population-level SARS-CoV-2 infection-induced immunity without introduction of vaccines. These results may add to our understanding of how immunity develops against emerging infectious diseases including coronaviruses.
Collapse
Affiliation(s)
- Greco Mark B Malijan
- San Lazaro Hospital-Nagasaki University Collaborative Research Office, San Lazaro Hospital, Quiricada St., Sta. Cruz, 1003, Manila, Philippines.
| | - Tansy Edwards
- School of Tropical Medicine and Global Health, Nagasaki University, Sakamoto, Nagasaki, 852-8102, Japan
- MRC International Statistics and Epidemiology Group, London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
| | - Kristal An Agrupis
- San Lazaro Hospital-Nagasaki University Collaborative Research Office, San Lazaro Hospital, Quiricada St., Sta. Cruz, 1003, Manila, Philippines
| | - Shuichi Suzuki
- San Lazaro Hospital-Nagasaki University Collaborative Research Office, San Lazaro Hospital, Quiricada St., Sta. Cruz, 1003, Manila, Philippines
- School of Tropical Medicine and Global Health, Nagasaki University, Sakamoto, Nagasaki, 852-8102, Japan
| | - Annavi Marie G Villanueva
- San Lazaro Hospital-Nagasaki University Collaborative Research Office, San Lazaro Hospital, Quiricada St., Sta. Cruz, 1003, Manila, Philippines
- San Lazaro Hospital, Quiricada St., Sta. Cruz, 1003, Manila, Philippines
| | - Ana Ria Sayo
- San Lazaro Hospital, Quiricada St., Sta. Cruz, 1003, Manila, Philippines
| | | | - Alexis Q Dimapilis
- San Lazaro Hospital, Quiricada St., Sta. Cruz, 1003, Manila, Philippines
| | - Rontgene M Solante
- San Lazaro Hospital, Quiricada St., Sta. Cruz, 1003, Manila, Philippines
| | - Elizabeth O Telan
- San Lazaro Hospital, Quiricada St., Sta. Cruz, 1003, Manila, Philippines
| | - Dorcas V Umipig
- San Lazaro Hospital, Quiricada St., Sta. Cruz, 1003, Manila, Philippines
| | - Kenji Ota
- Department of Laboratory Medicine, Nagasaki University Hospital, Sakamoto, Nagasaki, 852-8102, Japan
| | - Fumitaka Nishimura
- Department of Laboratory Medicine, Nagasaki University Hospital, Sakamoto, Nagasaki, 852-8102, Japan
| | - Katsunori Yanagihara
- Department of Laboratory Medicine, Nagasaki University Hospital, Sakamoto, Nagasaki, 852-8102, Japan
| | - Mary Jane Salazar
- San Lazaro Hospital-Nagasaki University Collaborative Research Office, San Lazaro Hospital, Quiricada St., Sta. Cruz, 1003, Manila, Philippines
| | - Edmundo B Lopez
- San Lazaro Hospital, Quiricada St., Sta. Cruz, 1003, Manila, Philippines
| | - Koya Ariyoshi
- School of Tropical Medicine and Global Health, Nagasaki University, Sakamoto, Nagasaki, 852-8102, Japan
- Institute of Tropical Medicine, Nagasaki University, Sakamoto, Nagasaki, Nagasaki, 852-8523, Japan
| | - Chris Smith
- School of Tropical Medicine and Global Health, Nagasaki University, Sakamoto, Nagasaki, 852-8102, Japan
- Department of Clinical Research, London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
| |
Collapse
|
95
|
Bassal R, Keinan-Boker L, Cohen D, Mendelson E, Lustig Y, Indenbaum V. Estimated Infection and Vaccine Induced SARS-CoV-2 Seroprevalence in Israel among Adults, January 2020-July 2021. Vaccines (Basel) 2022; 10:vaccines10101663. [PMID: 36298527 PMCID: PMC9609359 DOI: 10.3390/vaccines10101663] [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: 09/19/2022] [Revised: 10/02/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) emerged in Israel in February 2020 and spread from then. In December 2020, the FDA approved an emergency use authorization of the Pfizer-BioNTech vaccine, and on 20 December, an immunization campaign began among adults in Israel. We characterized seropositivity for IgG anti-spike antibodies against SARS-CoV-2 between January 2020 and July 2021, before and after the introduction of the vaccine in Israel among adults. We tested 9520 serum samples, collected between January 2020 and July 2021. Between January and August 2020, seropositivity rates were lower than 5.0%; this rate increased from September 2020 (6.3%) to April 2021 (84.9%) and reached 79.1% in July 2021. Between January and December 2020, low socio-economic rank was an independent, significant correlate for seropositivity. Between January and July 2021, the 40.00–64.99-year-old age group, Jews and others, and residents of the Northern district were significantly more likely to be seropositive. Our findings indicate a slow, non-significant increase in the seropositivity rate to SARS-CoV-2 between January and December 2020. Following the introduction of the Pfizer-BioNTech vaccine in Israel, a significant increase in seropositivity was observed from January until April 2021, with stable rates thereafter, up to July 2021.
Collapse
Affiliation(s)
- Ravit Bassal
- Israel Center for Disease Control, Ministry of Health, Gertner Institute, Chaim Sheba Medical Center, Tel-Hashomer 52621, Israel
- Department of Epidemiology and Preventive Medicine, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
- Correspondence: ; Tel.: +972-3-7371522
| | - Lital Keinan-Boker
- Israel Center for Disease Control, Ministry of Health, Gertner Institute, Chaim Sheba Medical Center, Tel-Hashomer 52621, Israel
- School of Public Health, University of Haifa, Haifa 3498838, Israel
| | - Dani Cohen
- Department of Epidemiology and Preventive Medicine, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Ella Mendelson
- Department of Epidemiology and Preventive Medicine, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
- Central Virology Laboratory, Public Health Services, Ministry of Health, Chaim Sheba Medical Center, Tel Hashomer 52621, Israel
| | - Yaniv Lustig
- Department of Epidemiology and Preventive Medicine, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
- Central Virology Laboratory, Public Health Services, Ministry of Health, Chaim Sheba Medical Center, Tel Hashomer 52621, Israel
| | - Victoria Indenbaum
- Central Virology Laboratory, Public Health Services, Ministry of Health, Chaim Sheba Medical Center, Tel Hashomer 52621, Israel
| |
Collapse
|
96
|
Garcia Quesada M, Hetrich MK, Zeger S, Sharma J, Na YB, Veguilla V, Karron RA, Dawood FS, Knoll MD. Predictors of Severe Acute Respiratory Syndrome Coronavirus 2 Seropositivity Before Coronavirus Disease 2019 Vaccination Among Children 0-4 Years and Their Household Members in the SEARCh Study. Open Forum Infect Dis 2022; 9:ofac507. [PMID: 36324323 PMCID: PMC9619557 DOI: 10.1093/ofid/ofac507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/28/2022] [Indexed: 11/24/2022] Open
Abstract
Background Estimates of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) seroprevalence in young children and risk factors for seropositivity are scarce. Using data from a prospective cohort study of households during the pre-coronavirus disease 2019 (COVID-19) vaccine period, we estimated SARS-CoV-2 seroprevalence by age and evaluated risk factors for SARS-CoV-2 seropositivity. Methods The SARS-CoV-2 Epidemiology and Response in Children (SEARCh) study enrolled 175 Maryland households (690 participants) with ≥1 child aged 0-4 years during November 2020-March 2021; individuals vaccinated against COVID-19 were ineligible. At enrollment, participants completed questionnaires about sociodemographic and health status and work, school, and daycare attendance. Participants were tested for SARS-CoV-2 antibodies in sera. Logistic regression models with generalized estimating equations (GEE) to account for correlation within households assessed predictors of individual- and household-level SARS-CoV-2 seropositivity. Results Of 681 (98.7%) participants with enrollment serology results, 55 (8.1%; 95% confidence interval [CI], 6.3%-10.4%) participants from 21 (12.0%) households were seropositive for SARS-CoV-2. Among seropositive participants, fewer children than adults reported being tested for SARS-CoV-2 infection before enrollment (odds ratio [OR] = 0.23; 95% CI, .06-.73). Seropositivity was similar by age (GEE OR vs 0-4 years: 1.19 for 5-17 years, 1.36 for adults; P = .16) and was significantly higher among adults working outside the home (GEE adjusted OR = 2.2; 95% CI, 1.1-4.4) but not among children attending daycare or school. Conclusions Before study enrollment, children and adults in this cohort had similar rates of SARS-CoV-2 infection as measured by serology. An adult household member working outside the home increased a household's odds of SARS-CoV-2 infection, whereas a child attending daycare or school in person did not.
Collapse
Affiliation(s)
- Maria Garcia Quesada
- Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Marissa K Hetrich
- Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Scott Zeger
- Department of Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jayati Sharma
- Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Yu Bin Na
- Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Vic Veguilla
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Ruth A Karron
- Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Fatimah S Dawood
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Maria D Knoll
- Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
97
|
Drews SJ, O’Brien SF. Lessons Learned from the COVID-19 Pandemic and How Blood Operators Can Prepare for the Next Pandemic. Viruses 2022; 14:2126. [PMID: 36298680 PMCID: PMC9608827 DOI: 10.3390/v14102126] [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: 08/04/2022] [Revised: 09/08/2022] [Accepted: 09/22/2022] [Indexed: 11/17/2022] Open
Abstract
Humans interact with virus-infected animal hosts, travel globally, and maintain social networks that allow for novel viruses to emerge and develop pandemic potential. There are key lessons-learned from the coronavirus diseases 2019 (COVID-19) pandemic that blood operators can apply to the next pandemic. Warning signals to the COVID-19 pandemic included outbreaks of Severe acute respiratory syndrome-related coronavirus-1 (SARS-CoV-1) and Middle East respiratory syndrome-related coronavirus (MERS-CoV) in the prior two decades. It will be critical to quickly determine whether there is a risk of blood-borne transmission of a new pandemic virus. Prior to the next pandemic blood operators should be prepared for changes in activities, policies, and procedures at all levels of the organization. Blood operators can utilize "Plan-Do-Study-Act" cycles spanning from: vigilance for emerging viruses, surveillance activities and studies, operational continuity, donor engagement and trust, and laboratory testing if required. Occupational health and donor safety issues will be key areas of focus even if the next pandemic virus is not transfusion transmitted. Blood operators may also be requested to engage in new activities such as the development of therapeutics or supporting public health surveillance activities. Activities such as scenario development, tabletop exercises, and drills will allow blood operators to prepare for the unknowns of the next pandemic.
Collapse
Affiliation(s)
- Steven J. Drews
- Canadian Blood Services, Microbiology, Donation and Policy Studies, Canadian Blood Services, Edmonton, AB T6G 2R8, Canada
- Division of Applied and Diagnostic Microbiology, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Sheila F. O’Brien
- Epidemiology and Surveillance, Donation Policy and Studies, Canadian Blood Services, Ottawa, ON K1G 4J5, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| |
Collapse
|
98
|
Follmann D, Janes HE, Buhule OD, Zhou H, Girard B, Marks K, Kotloff K, Desjardins M, Corey L, Neuzil KM, Miller JM, El Sahly HM, Baden LR. Antinucleocapsid Antibodies After SARS-CoV-2 Infection in the Blinded Phase of the Randomized, Placebo-Controlled mRNA-1273 COVID-19 Vaccine Efficacy Clinical Trial. Ann Intern Med 2022; 175:1258-1265. [PMID: 35785530 PMCID: PMC9258784 DOI: 10.7326/m22-1300] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Immunoassays for determining past SARS-CoV-2 infection have not been systematically evaluated in vaccinated persons in comparison with unvaccinated persons. OBJECTIVE To evaluate antinucleocapsid antibody (anti-N Ab) seropositivity in mRNA-1273 (Moderna) vaccinees with breakthrough SARS-CoV-2 infection. DESIGN Nested substudy of a phase 3 randomized, double-blind, placebo-controlled vaccine efficacy trial. (ClinicalTrials.gov: NCT04470427). SETTING 99 sites in the United States, July 2020 through March 2021. PARTICIPANTS Participants were aged 18 years or older, had no known history of SARS-CoV-2 infection, and were at risk for SARS-CoV-2 infection or severe COVID-19. Substudy participants were diagnosed with SARS-CoV-2 infection during the trial's blinded phase. INTERVENTION 2 mRNA-1273 or placebo injections 28 days apart. MEASUREMENTS Nasopharyngeal swabs from days 1 and 29 (vaccination days) and from symptom-prompted illness visits were tested for SARS-CoV-2 via polymerase chain reaction (PCR). Serum samples from days 1, 29, and 57 and the participant decision visit (PDV, when participants were informed of treatment assignment; median day 149) were tested for anti-N Abs by the Elecsys immunoassay. RESULTS Among 812 participants with PCR-confirmed COVID-19 illness during the blinded phase of the trial (through March 2021), seroconversion to anti-N Abs (median of 53 days after diagnosis) occurred in 21 of 52 mRNA-1273 vaccinees (40% [95% CI, 27% to 54%]) versus 605 of 648 placebo recipients (93% [CI, 92% to 95%]). Each 1-log increase in SARS-CoV-2 viral copies at diagnosis was associated with 90% higher odds of anti-N Ab seroconversion (odds ratio, 1.90 [CI, 1.59 to 2.28]). LIMITATION The scope was restricted to mRNA-1273 vaccinees and the Elecsys assay, the sample size was small, data on Delta and Omicron infections were lacking, and the analysis did not address a prespecified objective of the trial. CONCLUSION Vaccination status should be considered when interpreting seroprevalence and seropositivity data based solely on anti-N Ab testing. PRIMARY FUNDING SOURCE National Institute of Allergy and Infectious Diseases of the National Institutes of Health.
Collapse
Affiliation(s)
- Dean Follmann
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland (D.F., O.D.B.)
| | - Holly E Janes
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington (H.E.J.)
| | - Olive D Buhule
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland (D.F., O.D.B.)
| | - Honghong Zhou
- Moderna, Cambridge, Massachusetts (H.Z., B.G., J.M.M.)
| | | | | | - Karen Kotloff
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland (K.K., K.M.N.)
| | - Michaël Desjardins
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, and Division of Infectious Diseases, Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada (M.D.)
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, and Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington (L.C.)
| | - Kathleen M Neuzil
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland (K.K., K.M.N.)
| | | | - Hana M El Sahly
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas (H.M.E.)
| | - Lindsey R Baden
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts (L.R.B.)
| |
Collapse
|
99
|
SARS-CoV-2 seroprevalence in blood donors before and after the first wave in Catalonia (Spain). BLOOD TRANSFUSION = TRASFUSIONE DEL SANGUE 2022; 20:353-361. [PMID: 35175189 PMCID: PMC9480965 DOI: 10.2450/2022.0232-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 12/29/2021] [Indexed: 01/24/2023]
Abstract
BACKGROUND Due to the COVID-19 pandemic, a national lockdown was applied in Spain from March to May 2020. It is uncertain when SARS-CoV-2 started to circulate in Catalonia, and only a few cases were diagnosed in this period. We assessed the SARS-CoV-2 seroprevalence in blood donors before and after the first wave and compared it with public health service (PHS) data. MATERIALS AND METHODS Retrospective archive or prospective fresh blood samples were obtained from blood donors aged 18 to 70 and anonymized after demographic data had been recorded (gender, age, place of residence, blood collection date). Two CE-marked enzyme-linked immunosorbent assays were used to test for anti-SARS-CoV-2. A SARS-CoV-2 IgM test was additionally performed in positive samples. Individuals aged 18 to 70 from among the general population diagnosed as having SARS-CoV-2 by the PHS were included for comparison with blood donor results. RESULTS A total of 10,170 blood donations were included in the first period, between 24 February and 9 March 2020, and 6,829 in the second period, between 16 May and 17 June 2020. The observed SARS-CoV-2 seroprevalence among blood donors rose from 0.27% (95% CI: 0.18-0.39) before the first wave to 5.55% (95% CI: 5.03-6.12) after it, and was even higher (6.90% [95% CI: 5.64-8.41]) among blood donors aged 18 to 29. The seroprevalence among blood donors was higher in more populated areas (Barcelona: 7.69%). A comparison of blood donor data with officially diagnosed cases showed a global 87.44% underestimation of SARS-CoV-2 in June 2020. DISCUSSION We analyzed the explosive 3-month increase in blood donor SARS-CoV-2 seroprevalence (from 0.27% to 5.55%) and show that more than 87% of cases went undiagnosed, despite the unprecedented deployment of testing measures. SARS-CoV-2 IgM results suggest that the virus was circulating among blood donors in February 2020. Blood donors are definitively proven to be a valuable resource for emerging disease surveillance studies.
Collapse
|
100
|
Saá P, Fink RV, Bakkour S, Jin J, Simmons G, Muench MO, Dawar H, Di Germanio C, Hui AJ, Wright DJ, Krysztof DE, Kleinman SH, Cheung A, Nester T, Kessler DA, Townsend RL, Spencer BR, Kamel H, Vannoy JM, Dave H, Busch MP, Stramer SL, Stone M, Jackman RP, Norris PJ. Frequent detection but lack of infectivity of SARS-CoV-2 RNA in presymptomatic, infected blood donor plasma. J Clin Invest 2022; 132:e159876. [PMID: 35834347 PMCID: PMC9435642 DOI: 10.1172/jci159876] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 07/12/2022] [Indexed: 11/24/2022] Open
Abstract
Respiratory viruses such as influenza do not typically cause viremia; however, SARS-CoV-2 has been detected in the blood of COVID-19 patients with mild and severe symptoms. Detection of SARS-CoV-2 in blood raises questions about its role in pathogenesis as well as transfusion safety concerns. Blood donor reports of symptoms or a diagnosis of COVID-19 after donation (post-donation information, PDI) preceded or coincided with increased general population COVID-19 mortality. Plasma samples from 2,250 blood donors who reported possible COVID-19-related PDI were tested for the presence of SARS-CoV-2 RNA. Detection of RNAemia peaked at 9%-15% of PDI donors in late 2020 to early 2021 and fell to approximately 4% after implementation of widespread vaccination in the population. RNAemic donors were 1.2- to 1.4-fold more likely to report cough or shortness of breath and 1.8-fold more likely to report change in taste or smell compared with infected donors without detectable RNAemia. No infectious virus was detected in plasma from RNAemic donors; inoculation of permissive cell lines produced less than 0.7-7 plaque-forming units (PFU)/mL and in susceptible mice less than 100 PFU/mL in RNA-positive plasma based on limits of detection in these models. These findings suggest that blood transfusions are highly unlikely to transmit SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Paula Saá
- Scientific Affairs, American Red Cross, Gaithersburg, Maryland, USA
| | | | - Sonia Bakkour
- Vitalant Research Institute, San Francisco, California, USA
- Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| | - Jing Jin
- Vitalant Research Institute, San Francisco, California, USA
- Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| | - Graham Simmons
- Vitalant Research Institute, San Francisco, California, USA
- Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| | - Marcus O. Muench
- Vitalant Research Institute, San Francisco, California, USA
- Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| | - Hina Dawar
- Vitalant Research Institute, San Francisco, California, USA
| | - Clara Di Germanio
- Vitalant Research Institute, San Francisco, California, USA
- Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| | - Alvin J. Hui
- Vitalant Research Institute, San Francisco, California, USA
| | | | | | | | | | | | | | | | - Bryan R. Spencer
- Scientific Affairs, American Red Cross, Gaithersburg, Maryland, USA
| | | | | | - Honey Dave
- Vitalant Research Institute, San Francisco, California, USA
| | - Michael P. Busch
- Vitalant Research Institute, San Francisco, California, USA
- Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| | - Susan L. Stramer
- Scientific Affairs, American Red Cross, Gaithersburg, Maryland, USA
| | - Mars Stone
- Vitalant Research Institute, San Francisco, California, USA
- Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| | - Rachael P. Jackman
- Vitalant Research Institute, San Francisco, California, USA
- Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| | - Philip J. Norris
- Vitalant Research Institute, San Francisco, California, USA
- Department of Laboratory Medicine, UCSF, San Francisco, California, USA
- Department of Medicine, UCSF, San Francisco, California, USA
| | | |
Collapse
|