101
|
Reynolds L, Dewey C, Asfour G, Little M. Vaccine efficacy against SARS-CoV-2 for Pfizer BioNTech, Moderna, and AstraZeneca vaccines: a systematic review. Front Public Health 2023; 11:1229716. [PMID: 37942238 PMCID: PMC10628441 DOI: 10.3389/fpubh.2023.1229716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 09/15/2023] [Indexed: 11/10/2023] Open
Abstract
The purpose of this systematic review was to report on the vaccine efficacy (VE) of three SARS-CoV-2 vaccines approved by Health Canada: Pfizer BioNTech, Moderna, and AstraZeneca. Four databases were searched for primary publications on population-level VE. Ninety-two publications matched the inclusion criteria, and the extracted data were separated by vaccine type: mRNA vaccines (Pfizer and Moderna) and the AstraZeneca vaccine. The median VE for PCR-positive patients and various levels of clinical disease was determined for the first and second doses of both vaccine types against multiple SARS-CoV-2 variants. The median VE for PCR-positive infections against unidentified variants from an mRNA vaccine was 64.5 and 89%, respectively, after one or two doses. The median VE for PCR-positive infections against unidentified variants from the AstraZeneca vaccine was 53.4 and 69.6%, respectively, after one or two doses. The median VE for two doses of mRNA for asymptomatic, symptomatic, and severe infection against unidentified variants was 85.5, 93.2, and 92.2%, respectively. The median VE for two doses of AstraZeneca for asymptomatic, symptomatic, and severe infection against unidentified variants was 69.7, 71, and 90.2%, respectively. Vaccine efficacy numerically increased from the first to the second dose, increased from the first 2 weeks to the second 2 weeks post-vaccination for both doses, but decreased after 4 months from the second dose. Vaccine efficacy did not differ by person's age.
Collapse
Affiliation(s)
- Lia Reynolds
- Department of Population Medicine, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Cate Dewey
- Department of Population Medicine, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Ghaid Asfour
- Department of Population Medicine, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Matthew Little
- Department of Population Medicine, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
- School of Public Health and Social Policy, Faculty of Human and Social Development, University of Victoria, Victoria, BC, Canada
| |
Collapse
|
102
|
Bean DJ, Monroe J, Liang YM, Borberg E, Senussi Y, Swank Z, Chalise S, Walt D, Weinberg J, Sagar M. Heterotypic responses against nsp12/nsp13 from prior SARS-CoV-2 infection associates with lower subsequent endemic coronavirus incidence. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.23.563621. [PMID: 37961343 PMCID: PMC10634759 DOI: 10.1101/2023.10.23.563621] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Immune responses from prior SARS-CoV-2 infection and COVID-19 vaccination do not prevent re-infections and may not protect against future novel coronaviruses (CoVs). We examined the incidence of and immune differences against human endemic CoVs (eCoV) as a proxy for response against future emerging CoVs. Assessment was among those with known SARS-CoV-2 infection, COVID-19 vaccination but no documented SARS-CoV-2 infection, or neither exposure. Retrospective cohort analyses suggest that prior SARS-CoV-2 infection, but not COVID-19 vaccination alone, protects against subsequent symptomatic eCoV infection. CD8+ T cell responses to the non-structural eCoV proteins, nsp12 and nsp13, were significantly higher in individuals with previous SARS-CoV-2 infection as compared to the other groups. The three groups had similar cellular responses against the eCoV spike and nucleocapsid, and those with prior spike exposure had lower eCoV-directed neutralizing antibodies. Incorporation of non-structural viral antigens in a future pan-CoV vaccine may improve protection against future heterologous CoV infections.
Collapse
Affiliation(s)
- David J. Bean
- Department of Virology, Immunology and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA
| | - Janet Monroe
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA
| | - Yan Mei Liang
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA
| | - Ella Borberg
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA
| | - Yasmeen Senussi
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA
| | - Zoe Swank
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA
| | - Sujata Chalise
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA
| | - David Walt
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA
| | - Janice Weinberg
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Manish Sagar
- Department of Virology, Immunology and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA
| |
Collapse
|
103
|
He X, Zeng B, Wang Y, Pang Y, Zhang M, Hu T, Liang Y, Kang M, Tang S. Effectiveness of booster vaccination with inactivated COVID-19 vaccines against SARS-CoV-2 Omicron BA.2 infection in Guangdong, China: a cohort study. Front Immunol 2023; 14:1257360. [PMID: 37915583 PMCID: PMC10616523 DOI: 10.3389/fimmu.2023.1257360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 09/29/2023] [Indexed: 11/03/2023] Open
Abstract
The effectiveness of COVID-19 vaccines wanes over time and the emergence of the SARS-CoV-2 Omicron variant led to the accelerated expansion of efforts for booster vaccination. However, the effect and contribution of booster vaccination with inactivated COVID-19 vaccines remain to be evaluated. We conducted a retrospective close contacts cohort study to analyze the epidemiological characteristics and Omicron infection risk, and to evaluate the effectiveness of booster vaccination with inactivated COVID-19 vaccines against SARS-CoV-2 infection, symptomatic COVID-19, and COVID-19 pneumonia during the outbreaks of Omicron BA.2 infection from 1 February to 31 July 2022 in Guangdong, China. A total of 46,547 close contacts were identified while 6.3% contracted Omicron BA.2 infection, 1.8% were asymptomatic infection, 4.1% developed mild COVID-19, and 0.3% had COVID-19 pneumonia. We found that females and individuals aged 0-17 or ≥ 60 years old were more prone to SARS-CoV-2 infection. The vaccinated individuals showed lower infection risk when compared with the unvaccinated people. The effectiveness of booster vaccination with inactivated COVID-19 vaccines against SARS-CoV-2 infection and symptomatic COVID-19 was 28.6% (95% CI: 11.6%, 35.0%) and 39.6% (95% CI: 30.0, 47.9) among adults aged ≥ 18 years old, respectively when compared with full vaccination. Booster vaccination provided a moderate level of protection against SARS-CoV-2 infection (VE: 49.9%, 95% CI: 22.3%-67.7%) and symptomatic COVID-19 (VE: 62.6%, 95% CI: 36.2%-78.0%) among adults aged ≥ 60 years old. Moreover, the effectiveness of booster vaccination was 52.2% (95% CI: 21.3%, 70.9%) and 83.8% (95% CI: 28.1%, 96.3%) against COVID-19 pneumonia in adults aged ≥ 18 and ≥ 60 years old, respectively. The reduction of absolute risk rate of COVID-19 pneumonia in the booster vaccination group was 0·96% (95% CI: 0.33%, 1.11%), and the number needed to vaccinate to prevent one case of COVID-19 pneumonia was 104 (95% CI: 91, 303) in adults aged ≥ 60 years old. In summary, booster vaccination with inactivated COVID-19 vaccines provides a low level of protection against infection and symptomatic in adults of 18-59 years old, and a moderate level of protection in older adults of more than 60 years old, but a high level of protection against COVID-19 pneumonia in older adults.
Collapse
Affiliation(s)
- Xiaofeng He
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
- Institute of Evidence-Based Medicine, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, China
| | - Biao Zeng
- Institute of Infectious Disease Control and Prevention, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, China
| | - Ye Wang
- Department of Epidemiology, School of Medicine, Jinan University, Guangzhou, China
| | - Yulian Pang
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Meng Zhang
- Institute of Infectious Disease Control and Prevention, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, China
| | - Ting Hu
- Institute of Infectious Disease Control and Prevention, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, China
| | - Yuanhao Liang
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Min Kang
- Institute of Infectious Disease Control and Prevention, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, China
- Department of Epidemiology, School of Medicine, Jinan University, Guangzhou, China
| | - Shixing Tang
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
104
|
Wee LE, Pang D, Chiew C, Tan J, Lee V, Ong B, Lye DC, Tan KB. Long-term Real-world Protection Afforded by Third mRNA Doses Against Symptomatic Severe Acute Respiratory Syndrome Coronavirus 2 Infections, Coronavirus Disease 19-related Emergency Attendances and Hospitalizations Amongst Older Singaporeans During an Omicron XBB Wave. Clin Infect Dis 2023; 77:1111-1119. [PMID: 37280047 DOI: 10.1093/cid/ciad345] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 05/23/2023] [Accepted: 06/02/2023] [Indexed: 06/08/2023] Open
Abstract
BACKGROUND Literature on long-term real-world vaccine effectiveness of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) booster vaccines (up to and beyond 360 days) is scarce. We report estimates of protection against symptomatic infection, emergency department (ED) attendances and hospitalizations up to and beyond 360 days post-receipt of booster messenger RNA (mRNA) vaccines among Singaporeans aged ≥60 years during an Omicron XBB wave. METHODS We conducted a population-based cohort study including all Singaporeans aged ≥60 years with no documented prior SARS-CoV-2 infection who had previously received ≥3 doses of mRNA vaccines (BNT162b2/mRNA-1273), over a 4-month period during transmission of Omicron XBB. We reported the adjusted incidence-rate-ratio (IRR) for symptomatic infections, ED attendances and hospitalizations at different time-intervals from both first and second boosters, using Poisson regression; with the reference group being those who received their first booster 90 to 179 days prior. RESULTS In total, 506 856 boosted adults were included, contributing 55 846 165 person-days of observation. Protection against symptomatic infections among those who received a third vaccine dose (first booster) waned after 180 days with increasing adjusted IRRs; however, protection against ED attendances and hospitalizations held up, with comparable adjusted IRRs with increasing time from third vaccine doses (≥360 days from third dose: adjusted IRR [ED attendances] = 0.73, 95% confidence interval [CI] = .62-.85; adjusted IRR [hospitalization] = 0.58, 95% CI = .49-.70). CONCLUSIONS Our results highlight the benefit of a booster dose in reducing ED attendances and hospitalizations amongst older adults aged ≥60 years with no documented prior SARS-CoV-2 infection, during an Omicron XBB wave; up to and beyond 360 days post-booster. A second booster provided further reduction.
Collapse
Affiliation(s)
- Liang En Wee
- National Centre for Infectious Diseases, Singapore, Singapore
- Duke-NUS Graduate Medical School, National University of Singapore, Singapore, Singapore
- Department of Infectious Diseases, Singapore General Hospital, Singapore, Singapore
| | | | - Calvin Chiew
- National Centre for Infectious Diseases, Singapore, Singapore
- Ministry of Health, Singapore, Singapore
| | - Janice Tan
- Ministry of Health, Singapore, Singapore
| | - Vernon Lee
- Ministry of Health, Singapore, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| | - Benjamin Ong
- Ministry of Health, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - David Chien Lye
- National Centre for Infectious Diseases, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
| | - Kelvin Bryan Tan
- Ministry of Health, Singapore, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| |
Collapse
|
105
|
Liang Y, Sun Q, Liu Q, Pang Y, Tang S. SARS-CoV-2 incidence, seroprevalence, and COVID-19 vaccination coverage in the homeless population: a systematic review and meta-analysis. Front Public Health 2023; 11:1044788. [PMID: 37900041 PMCID: PMC10600393 DOI: 10.3389/fpubh.2023.1044788] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 09/25/2023] [Indexed: 10/31/2023] Open
Abstract
Objectives SARS-CoV-2 infection and COVID-19 vaccination of homeless people are a serious public health concern during COVID-19 pandemic. We aimed to systematically assess SARS-CoV-2 incidence, seroprevalence, and COVID-19 vaccination coverage in homeless people, which are important to inform resource allocation and policy adjustment for the prevention and control of COVID-19. Methods We searched PubMed, Web of Science, and the World Health Organization COVID-19 database for the studies of SARS-CoV-2 incidence, seroprevalence, and COVID-19 vaccination coverage in the homeless population. Subgroup analyses were conducted to pool SARS-CoV-2 incidence and seroprevalence in sheltered homeless, unsheltered homeless, and mixed population, respectively. Potential sources of heterogeneity in the estimates were explored by meta-regression analysis. Results Forty-nine eligible studies with a total of 75,402 homeless individuals and 5,000 shelter staff were included in the meta-analysis. The pooled incidence of SARS-CoV-2 infection was 10% (95% CI: 7 to 12%) in the homeless population and 8% (5 to 12%) for shelter staff. In addition, the overall estimated SARS-CoV-2 specific seroprevalence was 19% (8 to 33%) for homeless populations and 22% (3 to 52%) for shelter staff, respectively. Moreover, for the homeless subjects, the pooled incidence was 10% (4 to 23%) for asymptomatic SARS-CoV-2 infections, 6% (1 to 12%) for symptomatic SARS-CoV-2 infections, 3% (1 to 4%) for hospitalization for COVID-19, and 1% (0 to 2%) for severe COVID-19 cases, respectively while no COVID-19-related death was reported. Furthermore, the data derived from 12 included studies involving 225,448 homeless individuals revealed that the pooled proportion of one dose COVID-19 vaccination was 41% (35 to 47%), which was significantly lower than those in the general population. Conclusion Our study results indicate that the homeless people remain highly susceptible to SARS-CoV-2 infection, but COVID-19 vaccination coverage was lower than the general population, underscoring the need for prioritizing vaccine deployment and implementing enhanced preventive measures targeting this vulnerable group.
Collapse
Affiliation(s)
| | | | | | | | - Shixing Tang
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| |
Collapse
|
106
|
Paternina-Caicedo A, Quevedo DS, Ríos DS, Moyano D, Alvis-Guzmán N, Alviz-Zakzuk NR, Salcedo F, Moyano L, Ramírez-Suarez J, Smith AD, De la Hoz-Restrepo F. Comparative effectiveness and duration of protection of ChAdOx1, CoronaVac, BNT162b2, mRNA-1273, and Ad26.COV2.S COVID-19 vaccines for symptomatic and hospitalized Mu, Delta, and Omicron: A test-negative case-control study. Vaccine 2023; 41:6291-6299. [PMID: 37679278 DOI: 10.1016/j.vaccine.2023.08.072] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 08/23/2023] [Accepted: 08/25/2023] [Indexed: 09/09/2023]
Abstract
OBJECTIVE We carried out a study to estimate the vaccine effectiveness (VE) of homologous vaccination schedules against COVID-19, using data from mandatory information systems from Bogota, Colombia. METHODS A test-negative case-control study in adults from Bogota (Colombia), between March 1st of 2021 and February 25th of 2022. We assess VE among symptomatic COVID-19 cases during the Mul, Delta, and Omicron predominance periods in Bogota, with controls matched by sex, age (±5 years), and date of testing (±7 days), using a case:control ratio of 1:1. We selected homologous vaccination schedules with ChAdOx1, CoronaVac, BNT162b2, mRNA-1273, and Ad26.COV2.S. VE was reported as one minus the odds ratio in adjusted conditional logistic regressions, with their 95% confidence intervals (CI). A p-value < 0.05 was considered statistically significant. RESULTS 52,913 cases were matched to controls, 16,722 for Mu, 14,094 for Delta, and 22,097 for Omicron. VE was high against COVID-19 during Mu weeks with full vaccination using the monovalent BNT162b2 (VE: 69; 95% CI, 65 to 72) vaccine and ChAdOx1 (VE: 64; 95% CI, 31 to 81) and significantly lower with CoronaVac (P < 0.001) and Ad26.COV2.S (P = 0.005). During Delta, VE against COVID-19 was higher with BNT162b2 (VE: 55; 95% CI, 51 to 58). The VE for COVID-19 cases during Omicron was higher with a booster dose of monovalent BNT162b2 (VE: 45; 95% CI, 34 to 54). The VE of primary series and booster for ChAdOx1, Ad26.COV2.S, and CoronaVac did not show protection for Omicron. CONCLUSION Our study provides further evidence on the protective effect of mRNA vaccines for Omicron, and warrant that the duration of protection against symptomatic infection may last for only a few months.
Collapse
Affiliation(s)
| | | | | | | | - Nelson Alvis-Guzmán
- ALZAK Foundation, Cartagena, Colombia; Universidad de Cartagena, Cartagena, Colombia
| | | | | | | | | | | | | |
Collapse
|
107
|
Meeraus W, Stuurman AL, Durukal I, Conde-Sousa E, Lee A, Maria AS, Furtado BE, Ouwens M, Gray CM, Valverde DA, da Silva HG, Taylor S. COVID-19 vaccine booster doses provide increased protection against COVID-19 hospitalization compared with previously vaccinated individuals: Interim findings from the REFORCO-Brazil real-world effectiveness study during Delta and Omicron. Vaccine 2023; 41:6366-6378. [PMID: 37704499 DOI: 10.1016/j.vaccine.2023.08.085] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/15/2023] [Accepted: 08/30/2023] [Indexed: 09/15/2023]
Abstract
BACKGROUND Although COVID-19 booster vaccination is widely recommended, there is limited long-term, population-level, real-world evidence on the magnitude of improved protection against severe COVID-19 conferred by boosting with monovalent COVID-19 vaccines developed against ancestral SARS-CoV-2, especially in low- or middle-income countries. We present interim results from the first large-scale assessment of the relative vaccine effectiveness (rVE) of first and second booster doses against severe COVID-19 in a low-/middle-income country. METHODS REFORCO-Brazil is an ongoing, test-negative case-control study (NCT05697705) utilizing Brazil national severe acute respiratory syndrome (SARS) surveillance and vaccination data. In SARS hospitalizations from August 1, 2021 to July 31, 2022, we matched test-positive (via SARS-CoV-2 antigen/reverse transcription polymerase chain reaction [RT-PCR]) cases and test-negative case-controls (via RT-PCR) based on admission date, preceding vaccinations, and age. We evaluated the rVEs of four monovalent COVID-19 vaccines (AZD1222, Ad26.COV2.S, CoronaVac, and BNT162b2) as second boosters compared with any first boosters received ≥4 months previously, and as first boosters compared with primary-series vaccinations completed ≥4 months previously. RESULTS The overall rVE of second boosters, from 5668 (2238 test-positive) evaluated hospitalizations, was 24.7 % (95 % confidence interval [CI]: 12.6-35.1); the overall rVE of first boosters, from 30,272 (12,063 test-positive) hospitalizations, was 46.8 % (95 % CI: 43.3-50.0). The rVEs of AZD1222 and BNT162b2 were similar: 29.4 % (95 % CI: 8.6-45.5) and 25.5 % (95 % CI: 4.2-42.2), respectively, for second boosters; and 42.5 % (95 % CI: 28.0-54.0) and 50.8 % (95 % CI: 47.5-54.0), respectively, for first boosters. In general, rVEs were higher in elderly (≥80 years) and immunocompromised/high-risk individuals. CONCLUSIONS Our results support the use of AZD1222 and other adenoviral/mRNA vaccine boosters to maintain protection against COVID-19 hospitalization from Omicron subvariants, including in elderly and immunocompromised individuals at increased risk of accelerated waning or severe outcomes.
Collapse
Affiliation(s)
- Wilhelmine Meeraus
- Medical Evidence, Vaccines & Immune Therapies, AstraZeneca, Cambridge, UK.
| | - Anke L Stuurman
- Medical Evidence, Vaccines & Immune Therapies, AstraZeneca, Cambridge, UK; P95 Epidemiology & Pharmacovigilance, Leuven, Belgium
| | - Ilgaz Durukal
- Real World Science, BioPharmaceuticals Medical, AstraZeneca, Cambridge, UK
| | - Eduardo Conde-Sousa
- Medical Evidence, Vaccines & Immune Therapies, AstraZeneca, Cambridge, UK; P95 Epidemiology & Pharmacovigilance, Leuven, Belgium
| | - Andrew Lee
- Medical and Payor Statistics, BioPharmaceuticals Business Unit, AstraZeneca, Cambridge, UK
| | | | | | - Mario Ouwens
- Medical and Payor Statistics, BioPharmaceuticals Business Unit, AstraZeneca, Mölndal, Sweden
| | - Christen M Gray
- Real World Science, BioPharmaceuticals Medical, AstraZeneca, Cambridge, UK
| | | | | | - Sylvia Taylor
- Medical Evidence, Vaccines & Immune Therapies, AstraZeneca, Cambridge, UK
| |
Collapse
|
108
|
Zhang S, Agyeman AA, Hadjichrysanthou C, Standing JF. SARS-CoV-2 viral dynamic modeling to inform model selection and timing and efficacy of antiviral therapy. CPT Pharmacometrics Syst Pharmacol 2023; 12:1450-1460. [PMID: 37534815 PMCID: PMC10583246 DOI: 10.1002/psp4.13022] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 07/15/2023] [Accepted: 07/19/2023] [Indexed: 08/04/2023] Open
Abstract
Mathematical models of viral dynamics have been reported to describe adequately the dynamic changes of severe acute respiratory syndrome-coronavirus 2 viral load within an individual host. In this study, eight published viral dynamic models were assessed, and model selection was performed. Viral load data were collected from a community surveillance study, including 2155 measurements from 162 patients (124 household and 38 non-household contacts). An extended version of the target-cell limited model that includes an eclipse phase and an immune response component that enhances viral clearance described best the data. In general, the parameter estimates showed good precision (relative standard error <10), apart from the death rate of infected cells. The parameter estimates were used to simulate the outcomes of a clinical trial of the antiviral tixagevimab-cilgavimab, a monoclonal antibody combination which blocks infection of the target cells by neutralizing the virus. The simulated outcome of the effectiveness of the antiviral therapy in controlling viral replication was in a good agreement with the clinical trial data. Early treatment with high antiviral efficacy is important for desired therapeutic outcome.
Collapse
Affiliation(s)
- Shengyuan Zhang
- Department of Pharmaceutics, School of PharmacyUniversity College LondonLondonUK
| | - Akosua A. Agyeman
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child HealthUniversity College LondonLondonUK
| | - Christoforos Hadjichrysanthou
- Department of MathematicsUniversity of SussexBrightonUK
- Department of Infectious Disease Epidemiology, School of Public HealthImperial College LondonLondonUK
| | - Joseph F. Standing
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child HealthUniversity College LondonLondonUK
| |
Collapse
|
109
|
Ferranna M, Robinson LA, Cadarette D, Eber MR, Bloom DE. The benefits and costs of U.S. employer COVID-19 vaccine mandates. RISK ANALYSIS : AN OFFICIAL PUBLICATION OF THE SOCIETY FOR RISK ANALYSIS 2023; 43:2053-2068. [PMID: 36649917 DOI: 10.1111/risa.14090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 11/07/2022] [Accepted: 12/12/2022] [Indexed: 06/17/2023]
Abstract
In 2021, the Biden Administration issued mandates requiring COVID-19 vaccinations for U.S. federal employees and contractors and for some healthcare and private sector workers. These mandates have been challenged in court; some have been halted or delayed. However, their costs and benefits have not been rigorously appraised. This study helps fill that gap. We estimate the direct costs and health-related benefits that would have accrued if these vaccination requirements had been implemented as intended. Compared with the January 2022 vaccination rates, we find that the mandates could have led to 15 million additional vaccinated individuals, increasing the overall proportion of the fully vaccinated U.S. population from 64% to 68%. The associated net benefits depend on the subsequent evolution of the pandemic-information unavailable ex ante to analysts or policymakers. In scenarios involving the emergence of a novel, more transmissible variant, against which vaccination and previous infection offer moderate protection, the estimated net benefits are potentially large. They reach almost $20,000 per additional vaccinated individual, with more than 20,000 total deaths averted over the 6-month period assessed. In scenarios involving a fading pandemic, existing vaccination-acquired or infection-acquired immunity provides sufficient protection, and the mandates' benefits are unlikely to exceed their costs. Thus, mandates may be most useful when the consequences of inaction are catastrophic. However, we do not compare the effects of mandates with alternative policies for increasing vaccination rates or for promoting other protective measures, which may receive stronger public support and be less likely to be overturned by litigation.
Collapse
Affiliation(s)
- Maddalena Ferranna
- Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California, USA
| | - Lisa A Robinson
- Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, USA
| | | | - Michael R Eber
- Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, USA
- Harvard Graduate School of Arts and Sciences, Harvard University, Cambridge, Massachusetts, USA
| | - David E Bloom
- Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, USA
| |
Collapse
|
110
|
Whitaker HJ, Tsang RSM, Byford R, Aspden C, Button E, Sebastian Pillai P, Jamie G, Kar D, Williams J, Sinnathamby M, Marsden G, Elson WH, Leston M, Anand S, Okusi C, Fan X, Linley E, Rowe C, DArcangelo S, Otter AD, Ellis J, Hobbs FDR, Tzortziou-Brown V, Zambon M, Ramsay M, Brown KE, Amirthalingam G, Andrews NJ, de Lusignan S, Lopez Bernal J. COVID-19 vaccine effectiveness against hospitalisation and death of people in clinical risk groups during the Delta variant period: English primary care network cohort study. J Infect 2023; 87:315-327. [PMID: 37579793 DOI: 10.1016/j.jinf.2023.08.005] [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: 02/10/2023] [Revised: 08/07/2023] [Accepted: 08/09/2023] [Indexed: 08/16/2023]
Abstract
BACKGROUND COVID-19 vaccines have been shown to be highly effective against hospitalisation and death following COVID-19 infection. COVID-19 vaccine effectiveness estimates against severe endpoints among individuals with clinical conditions that place them at increased risk of critical disease are limited. METHODS We used English primary care medical record data from the Oxford-Royal College of General Practitioners Research and Surveillance Centre sentinel network (N > 18 million). Data were linked to the National Immunisation Management Service database, Second Generation Surveillance System for virology test data, Hospital Episode Statistics, and death registry data. We estimated adjusted vaccine effectiveness (aVE) against COVID-19 infection followed by hospitalisation and death among individuals in specific clinical risk groups using a cohort design during the delta-dominant period. We also report mortality statistics and results from our antibody surveillance in this population. FINDINGS aVE against severe endpoints was high, 14-69d following a third dose aVE was 96.4% (95.1%-97.4%) and 97.9% (97.2%-98.4%) for clinically vulnerable people given a Vaxzevria and Comirnaty primary course respectively. Lower aVE was observed in the immunosuppressed group: 88.6% (79.1%-93.8%) and 91.9% (85.9%-95.4%) for Vaxzevria and Comirnaty respectively. Antibody levels were significantly lower among the immunosuppressed group than those not in this risk group across all vaccination types and doses. The standardised case fatality rate within 28 days of a positive test was 3.9/1000 in people not in risk groups, compared to 12.8/1000 in clinical risk groups. Waning aVE with time since 2nd dose was also demonstrated, for example, Comirnaty aVE against hospitalisation reduced from 96.0% (95.1-96.7%) 14-69days post-dose 2-82.9% (81.4-84.2%) 182days+ post-dose 2. INTERPRETATION In all clinical risk groups high levels of vaccine effectiveness against severe endpoints were seen. Reduced vaccine effectiveness was noted among the immunosuppressed group.
Collapse
Affiliation(s)
- Heather J Whitaker
- Statistics, Modelling and Economics Department, UK Health Security Agency, 61 Colindale Avenue, London NW9 5EQ, UK
| | - Ruby S M Tsang
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford OX2 6GG, UK
| | - Rachel Byford
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford OX2 6GG, UK
| | - Carole Aspden
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford OX2 6GG, UK
| | - Elizabeth Button
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford OX2 6GG, UK
| | | | - Gavin Jamie
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford OX2 6GG, UK
| | - Debasish Kar
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford OX2 6GG, UK
| | - John Williams
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford OX2 6GG, UK
| | - Mary Sinnathamby
- Immunisation and Vaccine Preventable Diseases Division, UK Health Security Agency, 61 Colindale Avenue, London NW9 5EQ, UK
| | - Gemma Marsden
- Royal College of General Practitioners Research and Surveillance Centre, Euston Square, London NW1 2FB, UK
| | - William H Elson
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford OX2 6GG, UK
| | - Meredith Leston
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford OX2 6GG, UK
| | - Sneha Anand
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford OX2 6GG, UK
| | - Cecilia Okusi
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford OX2 6GG, UK
| | - Xuejuan Fan
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford OX2 6GG, UK
| | - Ezra Linley
- Vaccine Evaluation Unit, UK Health Security Agency, Manchester M13 9WL, UK
| | - Cathy Rowe
- Diagnostics and Genomics, UK Health Security Agency, Porton Down, Salisbury SP4 0JG, UK
| | - Silvia DArcangelo
- Diagnostics and Genomics, UK Health Security Agency, Porton Down, Salisbury SP4 0JG, UK
| | - Ashley D Otter
- Diagnostics and Genomics, UK Health Security Agency, Porton Down, Salisbury SP4 0JG, UK
| | - Joanna Ellis
- Immunisation and Vaccine Preventable Diseases Division, UK Health Security Agency, 61 Colindale Avenue, London NW9 5EQ, UK; Virus Reference Laboratory, UK Health Security Agency, 61 Colindale Avenue, London NW9 5EQ, UK
| | - F D Richard Hobbs
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford OX2 6GG, UK
| | - Victoria Tzortziou-Brown
- Royal College of General Practitioners Research and Surveillance Centre, Euston Square, London NW1 2FB, UK
| | - Maria Zambon
- Virus Reference Laboratory, UK Health Security Agency, 61 Colindale Avenue, London NW9 5EQ, UK
| | - Mary Ramsay
- Immunisation and Vaccine Preventable Diseases Division, UK Health Security Agency, 61 Colindale Avenue, London NW9 5EQ, UK
| | - Kevin E Brown
- Immunisation and Vaccine Preventable Diseases Division, UK Health Security Agency, 61 Colindale Avenue, London NW9 5EQ, UK
| | - Gayatri Amirthalingam
- Immunisation and Vaccine Preventable Diseases Division, UK Health Security Agency, 61 Colindale Avenue, London NW9 5EQ, UK
| | - Nick J Andrews
- Statistics, Modelling and Economics Department, UK Health Security Agency, 61 Colindale Avenue, London NW9 5EQ, UK; Immunisation and Vaccine Preventable Diseases Division, UK Health Security Agency, 61 Colindale Avenue, London NW9 5EQ, UK
| | - Simon de Lusignan
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford OX2 6GG, UK; Royal College of General Practitioners Research and Surveillance Centre, Euston Square, London NW1 2FB, UK
| | - Jamie Lopez Bernal
- Immunisation and Vaccine Preventable Diseases Division, UK Health Security Agency, 61 Colindale Avenue, London NW9 5EQ, UK.
| |
Collapse
|
111
|
Horga A, Saenz R, Yilmaz G, Simón-Campos A, Pietropaolo K, Stubbings WJ, Collinson N, Ishak L, Zrinscak B, Belanger B, Granier C, Lin K, C Hurt A, Zhou XJ, Wildum S, Hammond J. Oral bemnifosbuvir (AT-527) vs placebo in patients with mild-to-moderate COVID-19 in an outpatient setting (MORNINGSKY). Future Virol 2023; 18:10.2217/fvl-2023-0115. [PMID: 37928891 PMCID: PMC10621114 DOI: 10.2217/fvl-2023-0115] [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: 06/13/2023] [Accepted: 09/29/2023] [Indexed: 11/07/2023]
Abstract
Aim: This phase III study assessed the efficacy/safety/antiviral activity/pharmacokinetics of bemnifosbuvir, a novel, oral nucleotide analog to treat COVID-19. Patients & methods: Outpatient adults/adolescents with mild-to-moderate COVID-19 were randomized 2:1 to bemnifosbuvir/placebo. Time to symptom alleviation/improvement (primary outcome), risk of hospitalization/death, viral load and safety were evaluated. Results: Although the study was discontinued prematurely and did not meet its primary end point, bemnifosbuvir treatment resulted in fewer hospitalizations (71% relative risk reduction), COVID-19-related medically attended hospital visits, and COVID-19-related complications compared with placebo. No reduction in viral load was observed. The proportion of patients with adverse events was similar; no deaths occurred. Conclusion: Bemnifosbuvir showed hospitalization reduction in patients with variable disease progression risk and was well tolerated. Clinical Trial Registration: NCT04889040 (ClinicalTrials.gov).
Collapse
Affiliation(s)
| | | | - Gürdal Yilmaz
- Karadeniz Technical University, Trabzon, 61080, Turkey
| | | | | | | | - Neil Collinson
- Roche Products Limited, Welwyn Garden City, AL7 1TW, Hertfordshire, UK
| | - Laura Ishak
- Atea Pharmaceuticals, Inc, Boston, MA 02110, USA
| | | | | | - Catherine Granier
- Roche Products Limited, Welwyn Garden City, AL7 1TW, Hertfordshire, UK
| | - Kai Lin
- Atea Pharmaceuticals, Inc, Boston, MA 02110, USA
| | - Aeron C Hurt
- F. Hoffmann-La Roche Ltd, Basel, 4070, Switzerland
| | | | | | | |
Collapse
|
112
|
Wang H, Cui M, Li S, Wu F, Jiang S, Chen H, Yuan J, Sun C. Perception and willingness toward various immunization routes for COVID-19 vaccines: a cross-sectional survey in China. Front Public Health 2023; 11:1192709. [PMID: 37818300 PMCID: PMC10560725 DOI: 10.3389/fpubh.2023.1192709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 08/31/2023] [Indexed: 10/12/2023] Open
Abstract
Background To date, most vaccines, including the COVID-19 vaccine, are mainly administered by intramuscular injection, which might lead to vaccine hesitancy in some populations due to needle fear. Alternatively, needle-free immunization technology is extensively developed to improve the efficacy and acceptance of vaccination. However, there is no study to report the perception and willingness toward various immunization routes of the COVID-19 vaccine in the general population. Methods A cross-sectional survey was conducted nationwide using an online questionnaire. Bivariate analyses were undertaken to assess variable associations among the participants who reported a hesitancy to receive the COVID-19 booster vaccination. Multivariable logistic regression with a backward step-wise approach was used to analyze the predicted factors associated with the willingness to receive the COVID-19 booster vaccination. Results A total of 3,244 valid respondents were included in this survey, and 63.2% of participants thought they had a good understanding of intramuscular injection, but only 20.7, 9.2, 9.4, and 6.0% of participants had a self-perceived good understanding of inhalation vaccine, nasal spray vaccine, oral vaccine, and microneedle patch vaccine. Correspondingly, there was high acceptance for intramuscular injection (76.5%), followed by oral inhalation (64.4%) and nasal spray (43.0%). Those participants who were only willing to receive an intramuscular vaccine had less vaccine knowledge (OR = 0.78; 95% CI: 0.65-0.94) than those who were willing to receive a needle-free vaccine (OR = 1.97; 95% CI: 1.52-2.57). Some factors were found to be associated with vaccine hesitancy toward booster COVID-19 vaccination. Conclusion Needle-free vaccination is a promising technology for the next generation of vaccines, but we found that intramuscular injection was still the most acceptable immunization route in this survey. One major reason might be that most people lack knowledge about needle-free vaccination. We should strengthen the publicity of needle-free vaccination technology, and thus improve the acceptance and coverage of vaccination in different populations.
Collapse
Affiliation(s)
- Haohang Wang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Mingting Cui
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Shunran Li
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Fan Wu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Shiqiang Jiang
- Nanshan District Center for Disease Control and Prevention, Shenzhen, China
| | - Hongbiao Chen
- Department of Epidemiology and Infectious Disease Control, Longhua Key Discipline of Public Health for the Prevention and Control of Infectious Diseases, Longhua Centre for Disease Control and Prevention, Shenzhen, China
| | - Jianhui Yuan
- Nanshan District Center for Disease Control and Prevention, Shenzhen, China
| | - Caijun Sun
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| |
Collapse
|
113
|
Thakkar K, Spinardi JR, Yang J, Kyaw MH, Ozbilgili E, Mendoza CF, Oh HML. Impact of vaccination and non-pharmacological interventions on COVID-19: a review of simulation modeling studies in Asia. Front Public Health 2023; 11:1252719. [PMID: 37818298 PMCID: PMC10560858 DOI: 10.3389/fpubh.2023.1252719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/07/2023] [Indexed: 10/12/2023] Open
Abstract
Introduction Epidemiological modeling is widely used to offer insights into the COVID-19 pandemic situation in Asia. We reviewed published computational (mathematical/simulation) models conducted in Asia that assessed impacts of pharmacological and non-pharmacological interventions against COVID-19 and their implications for vaccination strategy. Methods A search of the PubMed database for peer-reviewed, published, and accessible articles in English was performed up to November 2022 to capture studies in Asian populations based on computational modeling of outcomes in the COVID-19 pandemic. Extracted data included model type (mechanistic compartmental/agent-based, statistical, both), intervention type (pharmacological, non-pharmacological), and procedures for parameterizing age. Findings are summarized with descriptive statistics and discussed in terms of the evolving COVID-19 situation. Results The literature search identified 378 results, of which 59 met criteria for data extraction. China, Japan, and South Korea accounted for approximately half of studies, with fewer from South and South-East Asia. Mechanistic models were most common, either compartmental (61.0%), agent-based (1.7%), or combination (18.6%) models. Statistical modeling was applied less frequently (11.9%). Pharmacological interventions were examined in 59.3% of studies, and most considered vaccination, except one study of an antiviral treatment. Non-pharmacological interventions were also considered in 84.7% of studies. Infection, hospitalization, and mortality were outcomes in 91.5%, 30.5%, and 30.5% of studies, respectively. Approximately a third of studies accounted for age, including 10 that also examined mortality. Four of these studies emphasized benefits in terms of mortality from prioritizing older adults for vaccination under conditions of a limited supply; however, one study noted potential benefits to infection rates from early vaccination of younger adults. Few studies (5.1%) considered the impact of vaccination among children. Conclusion Early in the COVID-19 pandemic, non-pharmacological interventions helped to mitigate the health burden of COVID-19; however, modeling indicates that high population coverage of effective vaccines will complement and reduce reliance on such interventions. Thus, increasing and maintaining immunity levels in populations through regular booster shots, particularly among at-risk and vulnerable groups, including older adults, might help to protect public health. Future modeling efforts should consider new vaccines and alternative therapies alongside an evolving virus in populations with varied vaccination histories.
Collapse
Affiliation(s)
- Karan Thakkar
- Vaccine Medical Affairs, Emerging Markets, Pfizer Inc., Singapore, Singapore
| | | | - Jingyan Yang
- Vaccine Global Value and Access, Pfizer Inc., New York, NY, United States
| | - Moe H. Kyaw
- Vaccine Medical Affairs, Emerging Markets, Pfizer Inc., Reston, VA, United States
| | - Egemen Ozbilgili
- Asia Cluster Medical Affairs, Emerging Markets, Pfizer Inc., Singapore, Singapore
| | | | - Helen May Lin Oh
- Department of Infectious Diseases, Changi General Hospital, Singapore, Singapore
| |
Collapse
|
114
|
Sternberg MR, Johnson A, King J, Ali AR, Linde L, Awofeso AO, Baker JS, Bayoumi NS, Broadway S, Busen K, Chang C, Cheng I, Cima M, Collingwood A, Dorabawila V, Drenzek C, Fleischauer A, Gent A, Hartley A, Hicks L, Hoskins M, Jara A, Jones A, Khan SI, Kamal-Ahmed I, Kangas S, Kanishka FNU, Kleppinger A, Kocharian A, León TM, Link-Gelles R, Lyons BC, Masarik J, May A, McCormick D, Meyer S, Milroy L, Morris KJ, Nelson L, Omoike E, Patel K, Pietrowski M, Pike MA, Pilishvili T, Peterson Pompa X, Powell C, Praetorius K, Rosenberg E, Schiller A, Smith-Coronado ML, Stanislawski E, Strand K, Tilakaratne BP, Vest H, Wiedeman C, Zaldivar A, Silk B, Scobie HM. Application of a life table approach to assess duration of BNT162b2 vaccine-derived immunity by age using COVID-19 case surveillance data during the Omicron variant period. PLoS One 2023; 18:e0291678. [PMID: 37729332 PMCID: PMC10511074 DOI: 10.1371/journal.pone.0291678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 09/01/2023] [Indexed: 09/22/2023] Open
Abstract
BACKGROUND SARS-CoV-2 Omicron variants have the potential to impact vaccine effectiveness and duration of vaccine-derived immunity. We analyzed U.S. multi-jurisdictional COVID-19 vaccine breakthrough surveillance data to examine potential waning of protection against SARS-CoV-2 infection for the Pfizer-BioNTech (BNT162b) primary vaccination series by age. METHODS Weekly numbers of SARS-CoV-2 infections during January 16, 2022-May 28, 2022 were analyzed by age group from 22 U.S. jurisdictions that routinely linked COVID-19 case surveillance and immunization data. A life table approach incorporating line-listed and aggregated COVID-19 case datasets with vaccine administration and U.S. Census data was used to estimate hazard rates of SARS-CoV-2 infections, hazard rate ratios (HRR) and percent reductions in hazard rate comparing unvaccinated people to people vaccinated with a Pfizer-BioNTech primary series only, by age group and time since vaccination. RESULTS The percent reduction in hazard rates for persons 2 weeks after vaccination with a Pfizer-BioNTech primary series compared with unvaccinated persons was lowest among children aged 5-11 years at 35.5% (95% CI: 33.3%, 37.6%) compared to the older age groups, which ranged from 68.7%-89.6%. By 19 weeks after vaccination, all age groups showed decreases in the percent reduction in the hazard rates compared with unvaccinated people; with the largest declines observed among those aged 5-11 and 12-17 years and more modest declines observed among those 18 years and older. CONCLUSIONS The decline in vaccine protection against SARS-CoV-2 infection observed in this study is consistent with other studies and demonstrates that national case surveillance data were useful for assessing early signals in age-specific waning of vaccine protection during the initial period of SARS-CoV-2 Omicron variant predominance. The potential for waning immunity during the Omicron period emphasizes the importance of continued monitoring and consideration of optimal timing and provision of booster doses in the future.
Collapse
Affiliation(s)
- Maya R. Sternberg
- COVID-19 Response, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Amelia Johnson
- COVID-19 Response, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Justice King
- COVID-19 Response, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Akilah R. Ali
- COVID-19 Response, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Lauren Linde
- COVID-19 Response, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Abiola O. Awofeso
- Community Health Administration, DC Department of Health, Washington, District of Columbia, United States of America
| | - Jodee S. Baker
- Division of Population Health, Utah Department of Health and Human Services, Salt Lake City, Utah, United States of America
| | - Nagla S. Bayoumi
- Communicable Disease Service, New Jersey Department of Health, Trenton, New Jersey, United States of America
| | - Steven Broadway
- Division of Disease Control and Health Protection, Florida Department of Health, Tallahassee, Florida, United States of America
| | - Katherine Busen
- Division of Communicable Disease, Michigan Department of Health and Human Services, Lansing, Michigan, United States of America
| | - Carolyn Chang
- Communicable Disease Service, New York City Department of Health and Mental Hygiene, Long Island City, New York, United States of America
| | - Iris Cheng
- Bureau of Immunization, New York City Department of Health and Mental Hygiene, Long Island City, New York, United States of America
| | - Mike Cima
- Epidemilogy, Arkansas Department of Health, Little Rock, Arkansas, United States of America
| | - Abi Collingwood
- Division of Population Health, Utah Department of Health and Human Services, Salt Lake City, Utah, United States of America
| | - Vajeera Dorabawila
- Bureau of Surveillance and Data Systems, Division of Epidemiology, Albany, New York State Department of Health, New York, NY, United States of America
| | - Cherie Drenzek
- Acute Epidemiology, Georgia Department of Public Health, Atlanta, Georgia, United States of America
| | - Aaron Fleischauer
- COVID-19 Response, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Ashley Gent
- Division of Disease Control and Health Protection, Florida Department of Health, Tallahassee, Florida, United States of America
| | - Amanda Hartley
- Communicable and Environmental Diseases and Emergency Preparedness, Nashville, Tennessee Department of Health, Nashville, Tennessee, United States of America
| | - Liam Hicks
- Bureau of Infectious Disease and Services, Arizona Department of Health Services, Phoenix, Arizona, United States of America
| | - Mikhail Hoskins
- Communicable Disease, North Carolina Department of Health and Human Services, Raleigh, North Carolina, United States of America
| | - Amanda Jara
- Acute Epidemiology, Georgia Department of Public Health, Atlanta, Georgia, United States of America
| | - Amanda Jones
- COVID-19 Response, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Saadiah I. Khan
- Communicable Disease Service, New Jersey Department of Health, Trenton, New Jersey, United States of America
| | - Ishrat Kamal-Ahmed
- Division of Public Health, Nebraska Department of Health and Human Services, Lincoln, Nebraska, United States of America
| | - Sarah Kangas
- COVID-19 Data and Surveillance Unit, Wisconsin Department of Health Services, Madison, Wisconsin, United States of America
| | - FNU Kanishka
- Division of Public Health, Nebraska Department of Health and Human Services, Lincoln, Nebraska, United States of America
| | - Alison Kleppinger
- Epidemiology and Infectious Disease Section, Connecticut Department of Public Health, Hartford, Connecticut, United States of America
| | - Anna Kocharian
- COVID-19 Data and Surveillance Unit, Wisconsin Department of Health Services, Madison, Wisconsin, United States of America
| | - Tomás M. León
- Center for Infectious Diseases, California Department of Public Health, Sacramento, California, United States of America
| | - Ruth Link-Gelles
- COVID-19 Response, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - B. Casey Lyons
- COVID-19 Response, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - John Masarik
- Community Health Administration, DC Department of Health, Washington, District of Columbia, United States of America
| | - Andrea May
- Bureau of Epidemiology and Public Health Informatics, Kansas Department of Health and Environment, Kansas, Missouri, United States of America
| | - Donald McCormick
- Epidemilogy, Arkansas Department of Health, Little Rock, Arkansas, United States of America
| | - Stephanie Meyer
- Infectious Disease Epidemiology, Prevention and Control Division, Minnesota Department of Health, Saint Paul, Minnesota, United States of America
| | - Lauren Milroy
- Disease Epidemiology and Prevention Division, Indiana Department of Health, Indianapolis, Indiana, United States of America
| | - Keeley J. Morris
- Infectious Disease Epidemiology, Prevention and Control Division, Minnesota Department of Health, Saint Paul, Minnesota, United States of America
| | - Lauren Nelson
- Center for Infectious Diseases, California Department of Public Health, Sacramento, California, United States of America
| | - Enaholo Omoike
- Division of Communicable Disease, Michigan Department of Health and Human Services, Lansing, Michigan, United States of America
| | - Komal Patel
- Acute Epidemiology, Georgia Department of Public Health, Atlanta, Georgia, United States of America
| | - Michael Pietrowski
- Division of Disease Control, Philadelphia Department of Public Health, Philadelphia, Pennsylvania, United States of America
| | - Melissa A. Pike
- Disease Control and Public Health Response Division, Colorado Department of Public Health and Environment, Denver, Colorado, United States of America
| | - Tamara Pilishvili
- COVID-19 Response, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Xandy Peterson Pompa
- Bureau of Infectious Disease and Services, Arizona Department of Health Services, Phoenix, Arizona, United States of America
| | - Charles Powell
- Epidemiology and Infectious Disease Section, Connecticut Department of Public Health, Hartford, Connecticut, United States of America
| | | | - Eli Rosenberg
- Bureau of Surveillance and Data Systems, Division of Epidemiology, Albany, New York State Department of Health, New York, NY, United States of America
| | - Adam Schiller
- COVID-19 Response, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Mayra L. Smith-Coronado
- Disease Control and Public Health Response Division, Colorado Department of Public Health and Environment, Denver, Colorado, United States of America
| | - Emma Stanislawski
- Epidemiology and Response Division, New Mexico Department of Health, Santa Fe, New Mexico, United States of America
| | - Kyle Strand
- Division of Public Health, Nebraska Department of Health and Human Services, Lincoln, Nebraska, United States of America
| | - Buddhi P. Tilakaratne
- Community Health Administration, DC Department of Health, Washington, District of Columbia, United States of America
| | - Hailey Vest
- Disease Epidemiology and Prevention Division, Indiana Department of Health, Indianapolis, Indiana, United States of America
| | - Caleb Wiedeman
- Communicable and Environmental Diseases and Emergency Preparedness, Nashville, Tennessee Department of Health, Nashville, Tennessee, United States of America
| | - Allison Zaldivar
- Bureau of Epidemiology and Public Health Informatics, Kansas Department of Health and Environment, Kansas, Missouri, United States of America
| | - Benjamin Silk
- COVID-19 Response, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Heather M. Scobie
- COVID-19 Response, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| |
Collapse
|
115
|
Yao T, Guo Y, Xu X, Zhang X, Mu S, Huo J, Wei Z, Liu L, Li X, Li H, Xing R, Feng Y, Chen J, Feng L, Wang S. Predictors of immune persistence induced by two-dose BBIBP-CorV vaccine in high-risk occupational population. Vaccine 2023; 41:5910-5917. [PMID: 37604725 DOI: 10.1016/j.vaccine.2023.08.042] [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: 08/09/2022] [Revised: 08/15/2023] [Accepted: 08/15/2023] [Indexed: 08/23/2023]
Abstract
BACKGROUND The immune protection from infection may wane over time as neutralizing antibody levels decline. We aimed to develop a nomogram to predict long-term immune persistence induced by two-dose BBIBP-CorV vaccine and calculate the neutralizing antibody decline probability of individuals. METHODS In the initial study, a total of 809 participants were recruited and randomly allocated (1:1:1) to vaccination group with three two-dose schedules on days 0 and 14, 0 and 21, or 0 and 28. The participants with neutralizing antibody titers of 16 or above on day 28 after the second dose were followed up at month 3, 6 and 10. Multivariable Cox proportional hazards regression model and nomogram model were used to identify predictors associated with maintaining of neutralizing antibody levels during 10 months after the second dose. RESULTS A total of 744 participants followed up at day 28 after the second dose. The participants with age ≥ 50 (aHR = 3.556, 95 %CI: 1.141-4.884, P = 0.028) were associated with a high risk of response loss (titers < 16). The participants who were in 0-28 d group (aHR = 0.403, 95 %CI: 0.177-0.919, P = 0.031), had an influenza vaccination history (aHR = 0.468, 95 %CI: 0.267-0.921, P = 0.033) or were female (aHR = 0.542, 95 %CI: 0.269-0.935, P = 0.035) tended to maintain immune persistence during 10 months after the second dose. The nomogram was constructed and showed moderate discrimination[C-index:0.711 (95 %CI: 0.652-0.770); AUC: 0.731 (95 %CI: 0.663-0.792)] and good calibration. CONCLUSIONS From 28 days to 10 months after receipt of the second dose of the BBIBP-CorV vaccine, neutralizing antibody levels were substantially decreased, especially among men, among persons 50 years of age or older, among persons with the 0-14 d group, and among persons without history of influenza vaccination. TRIAL REGISTRATION Chinese Clinical Trial Registry, ChiCTR2100041705, ChiCTR2100041706.
Collapse
Affiliation(s)
- Tian Yao
- First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, China; Center of Clinical Epidemiology and Evidence Based Medicine, Shanxi Medical University, Taiyuan, China
| | - Yana Guo
- School of Public Health, Shanxi Medical University, Taiyuan, China; Center of Clinical Epidemiology and Evidence Based Medicine, Shanxi Medical University, Taiyuan, China
| | - Xiuyang Xu
- School of Public Health, Shanxi Medical University, Taiyuan, China; Center of Clinical Epidemiology and Evidence Based Medicine, Shanxi Medical University, Taiyuan, China
| | - Xiaohong Zhang
- Shanxi Provincial Center for Disease Control and Prevention, Taiyuan, China; Shanxi Provincial Key Laboratory for Major Infectious Disease Response, Taiyuan, China
| | - Shengcai Mu
- Shanxi Provincial Center for Disease Control and Prevention, Taiyuan, China; Shanxi Provincial Key Laboratory for Major Infectious Disease Response, Taiyuan, China
| | - Junfeng Huo
- Shanxi Provincial Center for Disease Control and Prevention, Taiyuan, China; Shanxi Provincial Key Laboratory for Major Infectious Disease Response, Taiyuan, China
| | - Zhiyun Wei
- Shanxi Provincial Center for Disease Control and Prevention, Taiyuan, China; Shanxi Provincial Key Laboratory for Major Infectious Disease Response, Taiyuan, China
| | - Ling Liu
- Shanxi Provincial Center for Disease Control and Prevention, Taiyuan, China; Shanxi Provincial Key Laboratory for Major Infectious Disease Response, Taiyuan, China
| | - Xiaoqing Li
- Shanxi Provincial Center for Disease Control and Prevention, Taiyuan, China; Shanxi Provincial Key Laboratory for Major Infectious Disease Response, Taiyuan, China
| | - Hong Li
- Shanxi Provincial Center for Disease Control and Prevention, Taiyuan, China; Shanxi Provincial Key Laboratory for Major Infectious Disease Response, Taiyuan, China
| | - Rongqin Xing
- Outpatient Department of Shanxi Aviation Industry Group Co. LTD, Taiyuan, China
| | - Yongliang Feng
- School of Public Health, Shanxi Medical University, Taiyuan, China; Center of Clinical Epidemiology and Evidence Based Medicine, Shanxi Medical University, Taiyuan, China
| | - Jing Chen
- Shanxi Provincial Center for Disease Control and Prevention, Taiyuan, China; Shanxi Provincial Key Laboratory for Major Infectious Disease Response, Taiyuan, China
| | - Lizhong Feng
- Shanxi Provincial Center for Disease Control and Prevention, Taiyuan, China; Shanxi Provincial Key Laboratory for Major Infectious Disease Response, Taiyuan, China.
| | - Suping Wang
- School of Public Health, Shanxi Medical University, Taiyuan, China; Center of Clinical Epidemiology and Evidence Based Medicine, Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
116
|
Rebelo M, Tang C, Coelho AR, Labão-Almeida C, Schneider MM, Tatalick L, Ruivo P, de Miranda MP, Gomes A, Carvalho T, Walker MJ, Ausserwoeger H, Pedro Simas J, Veldhoen M, Knowles TPJ, Silva DA, Shoultz D, Bernardes GJL. De Novo Human Angiotensin-Converting Enzyme 2 Decoy NL-CVX1 Protects Mice From Severe Disease After Severe Acute Respiratory Syndrome Coronavirus 2 Infection. J Infect Dis 2023; 228:723-733. [PMID: 37279654 PMCID: PMC10503951 DOI: 10.1093/infdis/jiad135] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 05/27/2023] [Indexed: 06/08/2023] Open
Abstract
The emergence of novel variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) underscores the need to investigate alternative approaches to prevent infection and treat patients with coronavirus disease 2019. Here, we report the preclinical efficacy of NL-CVX1, a de novo decoy that blocks virus entry into cells by binding with nanomolar affinity and high specificity to the receptor-binding domain of the SARS-CoV-2 spike protein. Using a transgenic mouse model of SARS-CoV-2 infection, we showed that a single prophylactic intranasal dose of NL-CVX1 conferred complete protection from severe disease following SARS-CoV-2 infection. Multiple therapeutic administrations of NL-CVX1 also protected mice from succumbing to infection. Finally, we showed that infected mice treated with NL-CVX1 developed both anti-SARS-CoV-2 antibodies and memory T cells and were protected against reinfection a month after treatment. Overall, these observations suggest NL-CVX1 is a promising therapeutic candidate for preventing and treating severe SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Maria Rebelo
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Cong Tang
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Ana R Coelho
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Carlos Labão-Almeida
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Matthias M Schneider
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | | | - Pedro Ruivo
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Marta Pires de Miranda
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Andreia Gomes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Tânia Carvalho
- Histopathology Unit, Champalimaud Research, Lisboa, Portugal
| | | | - Hannes Ausserwoeger
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - J Pedro Simas
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Católica Biomedical Research and Católica Medical School, Universidade Católica Portuguesa, Lisboa, Portugal
| | - Marc Veldhoen
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Tuomas P J Knowles
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | | | | | - Gonçalo J L Bernardes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
117
|
Zhao Y, Wong SWK. A comparative study of compartmental models for COVID-19 transmission in Ontario, Canada. Sci Rep 2023; 13:15050. [PMID: 37700081 PMCID: PMC10497623 DOI: 10.1038/s41598-023-42043-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 09/04/2023] [Indexed: 09/14/2023] Open
Abstract
The number of confirmed COVID-19 cases reached over 1.3 million in Ontario, Canada by June 4, 2022. The continued spread of the virus underlying COVID-19 has been spurred by the emergence of variants since the initial outbreak in December, 2019. Much attention has thus been devoted to tracking and modelling the transmission of COVID-19. Compartmental models are commonly used to mimic epidemic transmission mechanisms and are easy to understand. Their performance in real-world settings, however, needs to be more thoroughly assessed. In this comparative study, we examine five compartmental models-four existing ones and an extended model that we propose-and analyze their ability to describe COVID-19 transmission in Ontario from January 2022 to June 2022.
Collapse
Affiliation(s)
- Yuxuan Zhao
- Department of Statistics and Actuarial Science, University of Waterloo, Waterloo, N2L 3G1, Canada
| | - Samuel W K Wong
- Department of Statistics and Actuarial Science, University of Waterloo, Waterloo, N2L 3G1, Canada.
| |
Collapse
|
118
|
Norton NJ, Ings DP, Fifield KE, Barnes DA, Barnable KA, Harnum DOA, Holder KA, Russell RS, Grant MD. Characteristics of Vaccine- and Infection-Induced Systemic IgA Anti-SARS-CoV-2 Spike Responses. Vaccines (Basel) 2023; 11:1462. [PMID: 37766138 PMCID: PMC10537135 DOI: 10.3390/vaccines11091462] [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: 07/31/2023] [Revised: 09/03/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Mucosal IgA is widely accepted as providing protection against respiratory infections, but stimulation of mucosal immunity, collection of mucosal samples and measurement of mucosal IgA can be problematic. The relationship between mucosal and circulating IgA responses is unclear, however, whole blood is readily collected and circulating antigen-specific IgA easily measured. We measured circulating IgA against SARS-CoV-2 spike (S) to investigate vaccine- and infection-induced production and correlation with protection. Circulating IgA against ancestral (Wuhan-Hu-1) and Omicron (BA.1) S proteins was measured at different time points in a total of 143 subjects with varied backgrounds of vaccination and infection. Intramuscular vaccination induced circulating anti-SARS-CoV-2 S IgA. Subjects with higher levels of vaccine-induced IgA against SARS-CoV-2 S (p = 0.0333) or receptor binding domain (RBD) (p = 0.0266) were less likely to experience an Omicron breakthrough infection. The same associations did not hold for circulating IgG anti-SARS-CoV-2 S levels. Breakthrough infection following two vaccinations generated stronger IgA anti-SARS-CoV-2 S responses (p = 0.0002) than third vaccinations but did not selectively increase circulating IgA against Omicron over ancestral S, indicating immune imprinting of circulating IgA responses. Circulating IgA against SARS-CoV-2 S following breakthrough infection remained higher than vaccine-induced levels for over 150 days. In conclusion, intramuscular mRNA vaccination induces circulating IgA against SARS-CoV-2 S, and higher levels are associated with protection from breakthrough infection. Vaccination with ancestral S enacts imprinting within circulating IgA responses that become apparent after breakthrough infection with Omicron. Breakthrough infection generates stronger and more durable circulating IgA responses against SARS-CoV-2 S than vaccination alone.
Collapse
Affiliation(s)
- Natasha J. Norton
- Immunology & Infectious Diseases Program, Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6, Canada; (N.J.N.); (D.P.I.); (K.E.F.); (D.A.B.); (K.A.B.); (K.A.H.); (R.S.R.)
| | - Danielle P. Ings
- Immunology & Infectious Diseases Program, Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6, Canada; (N.J.N.); (D.P.I.); (K.E.F.); (D.A.B.); (K.A.B.); (K.A.H.); (R.S.R.)
| | - Kathleen E. Fifield
- Immunology & Infectious Diseases Program, Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6, Canada; (N.J.N.); (D.P.I.); (K.E.F.); (D.A.B.); (K.A.B.); (K.A.H.); (R.S.R.)
| | - David A. Barnes
- Immunology & Infectious Diseases Program, Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6, Canada; (N.J.N.); (D.P.I.); (K.E.F.); (D.A.B.); (K.A.B.); (K.A.H.); (R.S.R.)
| | - Keeley A. Barnable
- Immunology & Infectious Diseases Program, Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6, Canada; (N.J.N.); (D.P.I.); (K.E.F.); (D.A.B.); (K.A.B.); (K.A.H.); (R.S.R.)
| | | | - Kayla A. Holder
- Immunology & Infectious Diseases Program, Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6, Canada; (N.J.N.); (D.P.I.); (K.E.F.); (D.A.B.); (K.A.B.); (K.A.H.); (R.S.R.)
| | - Rodney S. Russell
- Immunology & Infectious Diseases Program, Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6, Canada; (N.J.N.); (D.P.I.); (K.E.F.); (D.A.B.); (K.A.B.); (K.A.H.); (R.S.R.)
| | - Michael D. Grant
- Immunology & Infectious Diseases Program, Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6, Canada; (N.J.N.); (D.P.I.); (K.E.F.); (D.A.B.); (K.A.B.); (K.A.H.); (R.S.R.)
| |
Collapse
|
119
|
Farnsworth CW, O’Neil CA, Dalton C, McDonald D, Vogt L, Hock K, Arter O, Wallace MA, Muenks C, Amor M, Alvarado K, Peacock K, Jolani K, Fraser VJ, Burnham CAD, Babcock HM, Budge PJ, Kwon JH. Association between SARS-CoV-2 Symptoms, Ct Values, and Serological Response in Vaccinated and Unvaccinated Healthcare Personnel. J Appl Lab Med 2023; 8:871-886. [PMID: 37478837 PMCID: PMC10482509 DOI: 10.1093/jalm/jfad042] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 05/15/2023] [Indexed: 07/23/2023]
Abstract
BACKGROUND SARS-CoV-2 vaccines are effective at reducing symptomatic and asymptomatic COVID-19. Limited studies have compared symptoms, threshold cycle (Ct) values from reverse transcription (RT)-PCR testing, and serological testing results between previously vaccinated vs unvaccinated populations with SARS-CoV-2 infection. METHODS Healthcare personnel (HCP) with a positive SARS-CoV-2 RT-PCR test within the previous 14 to 28 days completed surveys including questions about demographics, medical conditions, social factors, and symptoms of COVID-19. Ct values were observed, and serological testing was performed for anti-nucleocapsid (anti-N) and anti-Spike (anti-S) antibodies at enrollment and 40 to 90 days later. Serological results were compared to HCP with no known SARS-CoV-2 infection and negative anti-N testing. RESULTS There were 104 unvaccinated/not fully vaccinated and 77 vaccinated HCP with 2 doses of an mRNA vaccine at time of infection. No differences in type or duration of symptoms were reported (P = 0.45). The median (interquartile range [IQR]) Ct was 21.4 (17.6-24.6) and 21.5 (18.1-24.6) for the unvaccinated and vaccinated HCP, respectively. Higher anti-N IgG was observed in unvaccinated HCP (5.08 S/CO, 3.08-6.92) than vaccinated (3.61 signal to cutoff ratio [S/CO], 2.16-5.05). Anti-S IgG was highest among vaccinated HCP with infection (34 285 aribitrary units [AU]/mL, 17 672-61 775), followed by vaccinated HCP with no prior infection (1452 AU/mL, 791-2943), then unvaccinated HCP with infection (829 AU/mL, 290-1555). Anti-S IgG decreased 1.56% (0.9%-1.79%) per day in unvaccinated and 0.38% (0.03%-0.94%) in vaccinated HCP. CONCLUSIONS Vaccinated HCP infected with SARS-CoV-2 reported comparable symptoms and had similar Ct values relative to unvaccinated. However, vaccinated HCP had increased and prolonged anti-S and decreased anti-N response relative to unvaccinated.
Collapse
Affiliation(s)
- Christopher W Farnsworth
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| | - Caroline A O’Neil
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, United States
| | - Claire Dalton
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| | - David McDonald
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, United States
| | - Lucy Vogt
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, United States
| | - Karl Hock
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| | - Olivia Arter
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, United States
| | - Meghan A Wallace
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| | - Carol Muenks
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| | - Mostafa Amor
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, United States
| | - Kelly Alvarado
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| | - Kate Peacock
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, United States
| | - Kevin Jolani
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, United States
| | - Victoria J Fraser
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, United States
| | - Carey-Ann D Burnham
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| | - Hilary M Babcock
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, United States
| | - Phillip J Budge
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, United States
| | - Jennie H Kwon
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
120
|
Wan EYF, Zhang R, Mathur S, Yan VKC, Lai FTT, Chui CSL, Li X, Wong CKH, Chan EWY, Lau CS, Wong ICK. Post-acute sequelae of COVID-19 in older persons: multi-organ complications and mortality. J Travel Med 2023; 30:taad082. [PMID: 37310901 DOI: 10.1093/jtm/taad082] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/29/2023] [Accepted: 06/10/2023] [Indexed: 06/15/2023]
Abstract
INTRODUCTION Evidence on long-term associations between coronavirus disease 2019 (COVID-19) and risks of multi-organ complications and mortality in older population is limited. This study evaluates these associations. RESEARCH DESIGN AND METHODS The cohorts included patients aged ≥60 year diagnosed with COVID-19 infection (cases), between 16 March 2020 and 31 May 2021 from the UK Biobank; and between 01 April 2020 and 31 May 2022 from the electronic health records in Hong Kong. Each patient was randomly matched with individuals without COVID-19 infection based on year of birth and sex and were followed for up to 18 months until 31 August 2021 for UKB, and up to 28 months until 15 August 2022 for HK cohort. Patients with COVID-19 infection over 6 months after the date of last dose of vaccination and their corresponding controls were excluded from our study. Characteristics between cohorts were further adjusted with Inverse Probability Treatment Weighting. For evaluating long-term association of COVID-19 with multi-organ disease complications and mortality after 21-days of diagnosis, Cox regression was employed. RESULT 10,759 (UKB) and 165,259 (HK) older adults with COVID-19 infection with matched 291,077 (UKB) and 1,100,394 (HK) non-COVID-19-diagnosed older adults were recruited. Older adults with COVID-19 were associated with a significantly higher risk of cardiovascular outcomes [major cardiovascular disease (stroke, heart failure and coronary heart disease): hazard ratio(UKB): 1.4 (95% Confidence interval: 1.1,1.6), HK:1.2 (95% CI: 1.1,1.3)]; myocardial infarction: HR(UKB): 1.8 (95% CI: 1.3,2.4), HK:1.2 (95% CI: 1.0,1.4)]; respiratory outcomes [interstitial lung disease: HR(UKB: 3.4 (95% CI: 2.5,4.5), HK: 4.0 (95% CI: 1.3,12.8); chronic pulmonary disease: HR(UKB): 1.7 (95% CI: 1.3,2.2), HK:1.6 (95% CI: 1.3,2.1)]; neuropsychiatric outcomes [seizure: HR(UKB): 2.6 (95% CI: 1.7,4.1), HK: 1.6 (95% CI: 1.2,2.1)]; and renal outcomes [acute kidney disease: HR(UKB): 1.4 (95% CI: 1.1,1.6), HK:1.6 (95% CI: 1.3,2.1)]; and all-cause mortality [HR(UKB): 4.9 (95% CI: 4.4,5.4), HK:2.5 (95% CI: 2.5,2.6)]. CONCLUSION COVID-19 is associated with long-term risks of multi-organ complications in older adults (aged ≥ 60). Infected patients in this age-group may benefit from appropriate monitoring of signs/symptoms for developing these complications.
Collapse
Affiliation(s)
- Eric Yuk Fai Wan
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Laboratory of Data Discovery for Health (D24H), Hong Kong Science and Technology Park, Hong Kong, China
- Department of Family Medicine and Primary Care, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ran Zhang
- Department of Family Medicine and Primary Care, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Sukriti Mathur
- Department of Family Medicine and Primary Care, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Vincent Ka Chun Yan
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Francisco Tsz Tsun Lai
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Laboratory of Data Discovery for Health (D24H), Hong Kong Science and Technology Park, Hong Kong, China
| | - Celine Sze Ling Chui
- Laboratory of Data Discovery for Health (D24H), Hong Kong Science and Technology Park, Hong Kong, China
- School of Nursing, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Xue Li
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Laboratory of Data Discovery for Health (D24H), Hong Kong Science and Technology Park, Hong Kong, China
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Carlos King Ho Wong
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Laboratory of Data Discovery for Health (D24H), Hong Kong Science and Technology Park, Hong Kong, China
- Department of Family Medicine and Primary Care, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Esther Wai Yin Chan
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Laboratory of Data Discovery for Health (D24H), Hong Kong Science and Technology Park, Hong Kong, China
- Department of Pharmacy, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- The University of Hong Kong Shenzhen Institute of Research and Innovation, Shenzhen 518053, China
| | - Chak Sing Lau
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ian Chi Kei Wong
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Laboratory of Data Discovery for Health (D24H), Hong Kong Science and Technology Park, Hong Kong, China
- Aston Pharmacy School, Aston University, Birmingham B4 7ET, UK
| |
Collapse
|
121
|
Ko L, Malet G, Chang LL, Nguyen H, Mayes R. COVID-19 Infection Rates in Vaccinated and Unvaccinated Inmates: A Retrospective Cohort Study. Cureus 2023; 15:e44684. [PMID: 37680261 PMCID: PMC10482361 DOI: 10.7759/cureus.44684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2023] [Indexed: 09/09/2023] Open
Abstract
Background In 2023, breakthrough COVID-19 infections among vaccinated individuals and reinfections in previously infected people have become common. Additionally, infections are due to Omicron subvariants of the virus that behave differently from those at the onset of the pandemic. Understanding how vaccination and natural immunity influence COVID-19 infection rates is crucial, especially in high-density congregate settings such as prisons, to inform public health strategies. Methods We analyzed COVID-19 surveillance data from January to July 2023 across 33 California state prisons, primarily a male population of 96,201 individuals. We computed the incidence rate of new COVID-19 infections among COVID-bivalent-vaccinated and entirely unvaccinated groups (those not having received either the bivalent or monovalent vaccine). Results Our results indicate that the infection rates in the bivalent-vaccinated and entirely unvaccinated groups are 3.24% (95% confidence interval (CI): 3.06-3.42%) and 2.72% (CI: 2.50-2.94%), respectively, with an absolute risk difference of only 0.52%. When the data were filtered for those aged 50 and above, the infection rates were 4.07% (CI: 3.77-4.37%) and 3.1% (CI: 2.46-3.74%), respectively, revealing a mere 0.97% absolute risk difference. Among those aged 65 and above, the infection rates were 6.45% (CI: 5.74-7.16%) and 4.5% (CI: 2.57-6.43%), respectively, with an absolute risk difference of 1.95%. Conclusion We note low infection rates in both the vaccinated and unvaccinated groups, with a small absolute difference between the two across age groups. A combination of monovalent and bivalent vaccines and natural infections likely contributed to immunity and a lower level of infection rates compared to the height of the pandemic. It is possible that a degree of 'herd immunity' has been achieved. Yet, using p<0.05 as the threshold for statistical significance, the bivalent-vaccinated group had a slightly but statistically significantly higher infection rate than the unvaccinated group in the statewide category and the age ≥50 years category. However, in the older age category (≥65 years), there was no significant difference in infection rates between the two groups. This suggests that while the bivalent vaccine might offer protection against severe outcomes, it may not significantly reduce the risk of infections entirely. Further research is needed to understand the reasons behind these findings and to consider other factors, such as underlying health conditions. This study underscores the importance of developing vaccines that target residual COVID-19 infections, especially in regard to evolving COVID-19 variants.
Collapse
Affiliation(s)
- Luke Ko
- Biomedical Sciences Pathway Program, California High School, San Ramon, USA
| | - Gary Malet
- Internal Medicine, California Correctional Healthcare Services, Stockton, USA
| | - Lisa L Chang
- College of Education, Governors State University, University Park, USA
| | - Huu Nguyen
- Internal Medicine, California Correctional Healthcare Services, Stockton, USA
| | - Robert Mayes
- Internal Medicine, California Correctional Healthcare Services, Stockton, USA
| |
Collapse
|
122
|
Liossi S, Tsiambas E, Maipas S, Papageorgiou E, Lazaris A, Kavantzas N. Mathematical modeling for Delta and Omicron variant of SARS-CoV-2 transmission dynamics in Greece. Infect Dis Model 2023; 8:794-805. [PMID: 37496829 PMCID: PMC10366468 DOI: 10.1016/j.idm.2023.07.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/02/2023] [Accepted: 07/05/2023] [Indexed: 07/28/2023] Open
Abstract
A compartmental, epidemiological, mathematical model was developed in order to analyze the transmission dynamics of Delta and Omicron variant, of SARS-CoV-2, in Greece. The model was parameterized twice during the 4th and 5th wave of the pandemic. The 4th wave refers to the period during which the Delta variant was dominant (approximately July to December of 2021) and the 5th wave to the period during which the Omicron variant was dominant (approximately January to May of 2022), in accordance with the official data from the National Public Health Organization (NPHO). Fitting methods were applied to evaluate important parameters in connection with the transmission of the variants, as well as the social behavior of population during these periods of interest. Mathematical models revealed higher numbers of contagiousness and cases of asymptomatic disease during the Omicron variant period, but a decreased rate of hospitalization compared to the Delta period. Also, parameters related to the behavior of the population in Greece were also assessed. More specifically, the use of protective masks and the abidance of social distancing measures. Simulations revealed that over 5,000 deaths could have been avoided, if mask usage and social distancing were 20% more efficient, during the short period of the Delta and Omicron outbreak. Furthermore, the spread of the variants was assessed using viral load data. The data were recorded from PCR tests at 417 Army Equity Fund Hospital (NIMTS), in Athens and the Ct values from 746 patients with COVID-19 were processed, to explain transmission phenomena and disease severity in patients. The period when the Delta variant prevailed in the country, the average Ct value was calculated as 25.19 (range: 12.32-39.29), whereas during the period when the Omicron variant prevailed, the average Ct value was calculated as 28 (range: 14.41-39.36). In conclusion, our experimental study showed that the higher viral load, which is related to the Delta variant, may interpret the severity of the disease. However, no correlation was confirmed regarding contagiousness phenomena. The results of the model, Ct analysis and official data from NPHO are consistent.
Collapse
Affiliation(s)
- Sofia Liossi
- 1st Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, Athens General Hospital “Laikon”, Athens, Greece
| | - E. Tsiambas
- Department of Cytopathology, 417 Army Equity Fund Hospital (NIMTS), Athens, Greece
| | - S. Maipas
- 1st Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, Athens General Hospital “Laikon”, Athens, Greece
- Master Program “Environment and Health. Management of Environmental Health Effects”, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - E. Papageorgiou
- Department of Biomedical Sciences, School of Health & Welfare Sciences, University of West Attica, Egaleo, Greece
| | - A. Lazaris
- 1st Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, Athens General Hospital “Laikon”, Athens, Greece
- Master Program “Environment and Health. Management of Environmental Health Effects”, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - N. Kavantzas
- 1st Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, Athens General Hospital “Laikon”, Athens, Greece
- Master Program “Environment and Health. Management of Environmental Health Effects”, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
123
|
Tamada Y, Takeuchi K, Kusama T, Maeda M, Murata F, Osaka K, Fukuda H. Effectiveness of COVID-19 vaccines against infection in Japan: A test-negative study from the VENUS study. Vaccine 2023; 41:5447-5453. [PMID: 37487845 DOI: 10.1016/j.vaccine.2023.07.035] [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: 11/10/2022] [Revised: 07/03/2023] [Accepted: 07/16/2023] [Indexed: 07/26/2023]
Abstract
BACKGROUND Although the effectiveness of coronavirus disease 2019 (COVID-19) vaccines is a crucial public health concern, evidence from Western Pacific countries is limited, including Japan. This study aimed to estimate the COVID-19 vaccines effectiveness (VE) against infection during the Delta variant predominance (July-September 2021) in Japan. METHODS We performed a test-negative study using COVID-19 test data of ≥20-year-old residents in four municipalities who were tested in medical institutions between July 1 and September 30, 2021. We extracted COVID-19 test data from healthcare claims data, and the vaccination status at the testing date was ascertained using the Vaccination Record System data. Confirmed positive cases were identified using data from the national system for COVID-19, Health Center Real-time Information-sharing System on COVID-19. Logistic regression analyses were conducted to estimate the odds of testing positive according to vaccination status. VE was calculated as (1 - odds ratio) × 100%. RESULTS This study included 530 positive and 15,650 negative results. Adjusted manufacturer-unspecified VE was 4.1% (95% confidence interval [CI], -36.5-32.6) at 0-13 days after the first dose, 45.2% (95% CI, 13.4-65.3) at ≥14 days after the first dose, 85.2% (95% CI, 69.9-92.7) at 0-13 days after the second dose, and 79.6% (95% CI, 72.6-84.8) at ≥14 days after the second dose. In addition, the VE after the second dose was highest at 14-34 days after the dose (VE, 89.1%; 95% CI, 80.5-93.9). CONCLUSIONS High real-world effectiveness of COVID-19 vaccines, especially two doses, against infection during the Delta variant predominance in Japan was confirmed.
Collapse
Affiliation(s)
- Yudai Tamada
- Department of International and Community Oral Health, Tohoku University Graduate School of Dentistry, Miyagi, Japan; Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Kenji Takeuchi
- Department of International and Community Oral Health, Tohoku University Graduate School of Dentistry, Miyagi, Japan; Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Aichi, Japan; Division of Statistics and Data Science, Liaison Center for Innovative Dentistry, Tohoku University Graduate School of Dentistry, Miyagi, Japan.
| | - Taro Kusama
- Department of International and Community Oral Health, Tohoku University Graduate School of Dentistry, Miyagi, Japan; Division of Statistics and Data Science, Liaison Center for Innovative Dentistry, Tohoku University Graduate School of Dentistry, Miyagi, Japan
| | - Megumi Maeda
- Department of Health Care Administration and Management, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Fumiko Murata
- Department of Health Care Administration and Management, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Ken Osaka
- Department of International and Community Oral Health, Tohoku University Graduate School of Dentistry, Miyagi, Japan
| | - Haruhisa Fukuda
- Department of Health Care Administration and Management, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| |
Collapse
|
124
|
Lind ML, Dorion M, Houde AJ, Lansing M, Lapidus S, Thomas R, Yildirim I, Omer SB, Schulz WL, Andrews JR, Hitchings MDT, Kennedy BS, Richeson RP, Cummings DAT, Ko AI. Evidence of leaky protection following COVID-19 vaccination and SARS-CoV-2 infection in an incarcerated population. Nat Commun 2023; 14:5055. [PMID: 37598213 PMCID: PMC10439918 DOI: 10.1038/s41467-023-40750-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 08/07/2023] [Indexed: 08/21/2023] Open
Abstract
Whether SARS-CoV-2 infection and COVID-19 vaccines confer exposure-dependent ("leaky") protection against infection remains unknown. We examined the effect of prior infection, vaccination, and hybrid immunity on infection risk among residents of Connecticut correctional facilities during periods of predominant Omicron and Delta transmission. Residents with cell, cellblock, and no documented exposure to SARS-CoV-2 infected residents were matched by facility and date. During the Omicron period, prior infection, vaccination, and hybrid immunity reduced the infection risk of residents without a documented exposure (HR: 0.36 [0.25-0.54]; 0.57 [0.42-0.78]; 0.24 [0.15-0.39]; respectively) and with cellblock exposures (0.61 [0.49-0.75]; 0.69 [0.58-0.83]; 0.41 [0.31-0.55]; respectively) but not with cell exposures (0.89 [0.58-1.35]; 0.96 [0.64-1.46]; 0.80 [0.46-1.39]; respectively). Associations were similar during the Delta period and when analyses were restricted to tested residents. Although associations may not have been thoroughly adjusted due to dataset limitations, the findings suggest that prior infection and vaccination may be leaky, highlighting the potential benefits of pairing vaccination with non-pharmaceutical interventions in crowded settings.
Collapse
Affiliation(s)
- Margaret L Lind
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA.
| | - Murilo Dorion
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Amy J Houde
- Connecticut Department of Correction, Wethersfield, CT, USA
| | - Mary Lansing
- Connecticut Department of Correction, Wethersfield, CT, USA
| | - Sarah Lapidus
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Russell Thomas
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Inci Yildirim
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
| | - Saad B Omer
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
- Yale Institute for Global Health, Yale School of Public Health, New Haven, CT, USA
- UT Southwestern, School of Public Health, Dallas, TX, USA
| | - Wade L Schulz
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Jason R Andrews
- Division of Infectious Diseases and Geographic Medicine, Stanford University, Stanford, CA, USA
| | - Matt D T Hitchings
- Department of Biostatistics, College of Public Health & Health Professions, University of Florida, Gainesville, FL, USA
| | | | | | - Derek A T Cummings
- Department of Biology, University of Florida, Gainesville, FL, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA
| | - Albert I Ko
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA.
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, BA, Brazil.
| |
Collapse
|
125
|
Morgan G, Casalino S, Chowdhary S, Frangione E, Fung CYJ, Haller S, Lapadula E, Scott M, Wolday D, Young J, Arnoldo S, Aujla N, Bearss E, Binnie A, Bombard Y, Borgundvaag B, Briollais L, Dagher M, Devine L, Faghfoury H, Friedman SM, Gingras AC, Goneau LW, Khan Z, Mazzulli T, McLeod SL, Nomigolzar R, Noor A, Pugh TJ, Richardson D, Satnam Singh HK, Simpson J, Stern S, Strug L, Taher A, Lerner-Ellis J, Taher J. Characterizing Risk Factors for Hospitalization and Clinical Characteristics in a Cohort of COVID-19 Patients Enrolled in the GENCOV Study. Viruses 2023; 15:1764. [PMID: 37632107 PMCID: PMC10457914 DOI: 10.3390/v15081764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/02/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
The GENCOV study aims to identify patient factors which affect COVID-19 severity and outcomes. Here, we aimed to evaluate patient characteristics, acute symptoms and their persistence, and associations with hospitalization. Participants were recruited at hospital sites across the Greater Toronto Area in Ontario, Canada. Patient-reported demographics, medical history, and COVID-19 symptoms and complications were collected through an intake survey. Regression analyses were performed to identify associations with outcomes including hospitalization and COVID-19 symptoms. In total, 966 responses were obtained from 1106 eligible participants (87% response rate) between November 2020 and May 2022. Increasing continuous age (aOR: 1.05 [95%CI: 1.01-1.08]) and BMI (aOR: 1.17 [95%CI: 1.10-1.24]), non-White/European ethnicity (aOR: 2.72 [95%CI: 1.22-6.05]), hypertension (aOR: 2.78 [95%CI: 1.22-6.34]), and infection by viral variants (aOR: 5.43 [95%CI: 1.45-20.34]) were identified as risk factors for hospitalization. Several symptoms including shortness of breath and fever were found to be more common among inpatients and tended to persist for longer durations following acute illness. Sex, age, ethnicity, BMI, vaccination status, viral strain, and underlying health conditions were associated with developing and having persistent symptoms. By improving our understanding of risk factors for severe COVID-19, our findings may guide COVID-19 patient management strategies by enabling more efficient clinical decision making.
Collapse
Affiliation(s)
- Gregory Morgan
- Lunenfeld-Tanenbaum Research Institute, Toronto, ON M5G 1X5, Canada
- Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Selina Casalino
- Lunenfeld-Tanenbaum Research Institute, Toronto, ON M5G 1X5, Canada
- Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Sunakshi Chowdhary
- Lunenfeld-Tanenbaum Research Institute, Toronto, ON M5G 1X5, Canada
- Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Erika Frangione
- Lunenfeld-Tanenbaum Research Institute, Toronto, ON M5G 1X5, Canada
- Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Chun Yiu Jordan Fung
- Lunenfeld-Tanenbaum Research Institute, Toronto, ON M5G 1X5, Canada
- Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Simona Haller
- Lunenfeld-Tanenbaum Research Institute, Toronto, ON M5G 1X5, Canada
- Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Elisa Lapadula
- Lunenfeld-Tanenbaum Research Institute, Toronto, ON M5G 1X5, Canada
- Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Mackenzie Scott
- Lunenfeld-Tanenbaum Research Institute, Toronto, ON M5G 1X5, Canada
- Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Dawit Wolday
- Lunenfeld-Tanenbaum Research Institute, Toronto, ON M5G 1X5, Canada
- Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Juliet Young
- Lunenfeld-Tanenbaum Research Institute, Toronto, ON M5G 1X5, Canada
- Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Saranya Arnoldo
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- William Osler Health System, Brampton, ON L6R 3J7, Canada
| | - Navneet Aujla
- Lunenfeld-Tanenbaum Research Institute, Toronto, ON M5G 1X5, Canada
- Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Erin Bearss
- Mount Sinai Academic Family Health Team, Mount Sinai Hospital, Toronto, ON M5T 3L9, Canada
- Department of Family and Community Medicine, University of Toronto, Toronto, ON M5G 1V7, Canada
| | - Alexandra Binnie
- Department of Critical Care, William Osler Health System, Etobicoke, ON M9V 1R8, Canada
| | - Yvonne Bombard
- Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, ON M5B 1A6, Canada
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON M5T 3M6, Canada
| | - Bjug Borgundvaag
- Schwartz/Reisman Emergency Medicine Institute, Sinai Health System, Toronto, ON M5G 2A2, Canada
| | | | - Marc Dagher
- Department of Family and Community Medicine, University of Toronto, Toronto, ON M5G 1V7, Canada
- Women’s College Hospital, Toronto, ON M5S 1B2, Canada
| | - Luke Devine
- Division of General Internal Medicine, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
- Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Hanna Faghfoury
- Fred A Litwin Family Centre in Genetic Medicine, University Health Network & Mount Sinai Hospital, Toronto, ON M5T 3H7, Canada
| | - Steven M. Friedman
- Department of Family and Community Medicine, University of Toronto, Toronto, ON M5G 1V7, Canada
- Emergency Medicine, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute, Toronto, ON M5G 1X5, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Lee W. Goneau
- Dynacare Medical Laboratories, Brampton, ON L6T 5V1, Canada
| | - Zeeshan Khan
- Mackenzie Health, Richmond Hill, ON L4C 4Z3, Canada
| | - Tony Mazzulli
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Microbiology, Mount Sinai Hospital, Sinai Health, Toronto, ON M5G 1X5, Canada
| | - Shelley L. McLeod
- Department of Family and Community Medicine, University of Toronto, Toronto, ON M5G 1V7, Canada
- Schwartz/Reisman Emergency Medicine Institute, Sinai Health System, Toronto, ON M5G 2A2, Canada
| | | | - Abdul Noor
- Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Trevor J. Pugh
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C4, Canada
- Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada
| | | | - Harpreet Kaur Satnam Singh
- Lunenfeld-Tanenbaum Research Institute, Toronto, ON M5G 1X5, Canada
- Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Jared Simpson
- Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada
- Department of Computer Science, University of Toronto, Toronto, ON M5S 2E4, Canada
| | - Seth Stern
- Mackenzie Health, Richmond Hill, ON L4C 4Z3, Canada
| | - Lisa Strug
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Statistical Sciences, University of Toronto, Toronto, ON M5G 1Z5, Canada
| | - Ahmed Taher
- Mackenzie Health, Richmond Hill, ON L4C 4Z3, Canada
- Division of Emergency Medicine, University of Toronto, Toronto, ON M5S 3H2, Canada
| | - Jordan Lerner-Ellis
- Lunenfeld-Tanenbaum Research Institute, Toronto, ON M5G 1X5, Canada
- Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jennifer Taher
- Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
126
|
Dai YC, Lin YC, Ching LL, Tsai JJ, Ishikawa K, Tsai WY, Chen JJ, Nerurkar VR, Wang WK. Determining the Time of Booster Dose Based on the Half-Life and Neutralization Titers against SARS-CoV-2 Variants of Concern in Fully Vaccinated Individuals. Microbiol Spectr 2023; 11:e0408122. [PMID: 37428104 PMCID: PMC10434144 DOI: 10.1128/spectrum.04081-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 06/20/2023] [Indexed: 07/11/2023] Open
Abstract
Although mRNA-based COVID-19 vaccines reduce the risk of severe disease, hospitalization and death, vaccine effectiveness (VE) against infection and disease from variants of concern (VOC) wanes over time. Neutralizing antibodies (NAb) are surrogates of protection and are enhanced by a booster dose, but their kinetics and durability remain understudied. Current recommendation of a booster dose does not consider the existing NAb in each individual. Here, we investigated 50% neutralization (NT50) titers against VOC among COVID-19-naive participants receiving the Moderna (n = 26) or Pfizer (n = 25) vaccine for up to 7 months following the second dose, and determined their half-lives. We found that the time it took for NT50 titers to decline to 24, equivalent to 50% inhibitory dilution of 10 international units/mL, was longer in the Moderna (325/324/235/274 days for the D614G/alpha/beta/delta variants) group than in the Pfizer (253/252/174/226 days) group, which may account for the slower decline in VE of the Moderna vaccine observed in real-world settings and supports our hypothesis that measuring the NT50 titers against VOC, together with information on NAb half-lives, can be used to dictate the time of booster vaccination. Our study provides a framework to determine the optimal time of a booster dose against VOC at the individual level. In response to future VOC with high morbidity and mortality, a quick evaluation of NAb half-lives using longitudinal serum samples from clinical trials or research programs of different primary-series vaccinations and/or one or two boosters could provide references for determining the time of booster in different individuals. IMPORTANCE Despite improved understanding of the biology of SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2), the evolutionary trajectory of the virus is uncertain, and the concern of future antigenically distinct variants remains. Current recommendations for a COVID-19 vaccine booster dose are primarily based on neutralization capacity, effectiveness against circulating variants of concern (VOC), and other host factors. We hypothesized that measuring neutralizing antibody titers against SARS-CoV-2 VOC together with half-life information can be used to dictate the time of booster vaccination. Through detailed analysis of neutralizing antibodies against VOC among COVID-19-naive vaccinees receiving either of two mRNA vaccines, we found that the time it took for 50% neutralization titers to decline to a reference level of protection was longer in the Moderna than in the Pfizer group, which supports our hypothesis. In response to future VOC with potentially high morbidity and mortality, our proof-of-concept study provides a framework to determine the optimal time of a booster dose at the individual level.
Collapse
Affiliation(s)
- Yu-Ching Dai
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Yen-Chia Lin
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Lauren L. Ching
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
- Pacific Center for Emerging Infectious Diseases, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Jih-Jin Tsai
- Tropical Medicine Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Division of Infectious Diseases, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Kyle Ishikawa
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Wen-Yang Tsai
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
- Pacific Center for Emerging Infectious Diseases, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - John J. Chen
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Vivek R. Nerurkar
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
- Pacific Center for Emerging Infectious Diseases, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Wei-Kung Wang
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
- Pacific Center for Emerging Infectious Diseases, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| |
Collapse
|
127
|
Wood AJ, Kao RR. Empirical distributions of time intervals between COVID-19 cases and more severe outcomes in Scotland. PLoS One 2023; 18:e0287397. [PMID: 37585389 PMCID: PMC10431635 DOI: 10.1371/journal.pone.0287397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 06/05/2023] [Indexed: 08/18/2023] Open
Abstract
A critical factor in infectious disease control is the risk of an outbreak overwhelming local healthcare capacity. The overall demand on healthcare services will depend on disease severity, but the precise timing and size of peak demand also depends on the time interval (or clinical time delay) between initial infection, and development of severe disease. A broader distribution of intervals may draw that demand out over a longer period, but have a lower peak demand. These interval distributions are therefore important in modelling trajectories of e.g. hospital admissions, given a trajectory of incidence. Conversely, as testing rates decline, an incidence trajectory may need to be inferred through the delayed, but relatively unbiased signal of hospital admissions. Healthcare demand has been extensively modelled during the COVID-19 pandemic, where localised waves of infection have imposed severe stresses on healthcare services. While the initial acute threat posed by this disease has since subsided with immunity buildup from vaccination and prior infection, prevalence remains high and waning immunity may lead to substantial pressures for years to come. In this work, then, we present a set of interval distributions, for COVID-19 cases and subsequent severe outcomes; hospital admission, ICU admission, and death. These may be used to model more realistic scenarios of hospital admissions and occupancy, given a trajectory of infections or cases. We present a method for obtaining empirical distributions using COVID-19 outcomes data from Scotland between September 2020 and January 2022 (N = 31724 hospital admissions, N = 3514 ICU admissions, N = 8306 mortalities). We present separate distributions for individual age, sex, and deprivation of residing community. While the risk of severe disease following COVID-19 infection is substantially higher for the elderly and those residing in areas of high deprivation, the length of stay shows no strong dependence, suggesting that severe outcomes are equally severe across risk groups. As Scotland and other countries move into a phase where testing is no longer abundant, these intervals may be of use for retrospective modelling of patterns of infection, given data on severe outcomes.
Collapse
Affiliation(s)
- Anthony J. Wood
- The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Rowland R. Kao
- The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
- Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
128
|
Meeraus W, de Munter L, Gray CM, Dwivedi A, Wyndham-Thomas C, Ouwens M, Hartig-Merkel W, Drikite L, Rebry G, Carmona A, Stuurman AL, Chi Nguyen TY, Mena G, Mira-Iglesias A, Icardi G, Otero-Romero S, Baumgartner S, Martin C, Taylor S, Bollaerts K. Protection against COVID-19 hospitalisation conferred by primary-series vaccination with AZD1222 in non-boosted individuals: first vaccine effectiveness results of the European COVIDRIVE study and meta-regression analysis. THE LANCET REGIONAL HEALTH. EUROPE 2023; 31:100675. [PMID: 37547274 PMCID: PMC10398604 DOI: 10.1016/j.lanepe.2023.100675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/30/2023] [Accepted: 06/14/2023] [Indexed: 08/08/2023]
Abstract
Background Vaccine effectiveness (VE) studies with long-term follow-up are needed to understand durability of protection against severe COVID-19 outcomes conferred by primary-series vaccination in individuals not receiving boosters. COVIDRIVE is a European public-private partnership evaluating brand-specific vaccine effectiveness (VE). We report a prespecified interim analysis of primary-series AZD1222 (ChAdOx1 nCoV-19) VE. Methods Seven Study Contributors in Europe collected data on individuals aged ≥18 years who were hospitalised with severe acute respiratory infection (June 1st, 2021-September 5th, 2022) and eligible for COVID-19 vaccination prior to hospitalisation. In this test-negative case-control study, individuals were defined as test-positive cases or test-negative controls (SARS-CoV-2 RT-PCR) and were either fully vaccinated (two AZD1222 doses, 4-12 weeks apart, completed ≥14 days prior to symptom onset; no booster doses) or unvaccinated (no COVID-19 vaccine prior to hospitalisation). The primary objective was to estimate AZD1222 VE against COVID-19 hospitalisation. A literature review and meta-regression were conducted to contextualise findings on durability of protection. Findings 761 individuals were included during the 15-month analysis period. Overall AZD1222 VE estimate was 72.8% (95% CI, 53.4-84.1). VE was 93.8% (48.6-99.3) in participants who received second AZD1222 doses ≤8 weeks prior to hospitalisation, with spline-based VE estimates demonstrating protection (VE ≥ 50%) 30 weeks post-second dose. Meta-regression analysis (data from seven publications) showed consistent results, with ≥80% protection against COVID-19 hospitalisation through ∼43 weeks post-second dose, with some degree of waning. Interpretation Primary-series AZD1222 vaccination confers protection against COVID-19 hospitalisation with enduring levels of VE through ≥6 months. Funding AstraZeneca.
Collapse
Affiliation(s)
- Wilhelmine Meeraus
- Medical Evidence, Vaccines & Immune Therapies, AstraZeneca, Cambridge, UK
| | | | - Christen M. Gray
- Real World Science, BioPharmaceuticals Medical, AstraZeneca, Cambridge, UK
| | | | | | - Mario Ouwens
- Medical and Payor Statistics, BioPharmaceutical Business Unit, AstraZeneca, Mölndal, Sweden
| | | | - Laura Drikite
- P95 Pharmacovigilance and Epidemiology, Leuven, Belgium
| | - Griet Rebry
- P95 Pharmacovigilance and Epidemiology, Leuven, Belgium
| | - Antonio Carmona
- Vaccine Research Department, Fundación para el Fomento de la Investigación Sanitaria y Biomédica (FISABIO) de la Comunitat Valenciana, Salud Pública, Valencia, Spain
- Biomedical Research Consortium of Epidemiology and Public Health (CIBER-ESP), Instituto de Salud Carlos III, Madrid, Spain
| | - Anke L. Stuurman
- Medical Evidence, Vaccines & Immune Therapies, AstraZeneca, Cambridge, UK
- P95 Pharmacovigilance and Epidemiology, Leuven, Belgium
| | - Thi Yen Chi Nguyen
- Medical Evidence, Vaccines & Immune Therapies, AstraZeneca, Cambridge, UK
- P95 Pharmacovigilance and Epidemiology, Leuven, Belgium
| | - Guillermo Mena
- Preventive Medicine Department - Germans Trias i Pujol University Hospital, Badalona, Spain
- Autonomous University of Barcelona, Bellaterra, Spain
- Germans Trias I Pujol Research Institute (IGTP), Badalona, Spain
| | - Ainara Mira-Iglesias
- Vaccine Research Department, Fundación para el Fomento de la Investigación Sanitaria y Biomédica (FISABIO) de la Comunitat Valenciana, Salud Pública, Valencia, Spain
- Biomedical Research Consortium of Epidemiology and Public Health (CIBER-ESP), Instituto de Salud Carlos III, Madrid, Spain
| | - Giancarlo Icardi
- Interuniversity Research Centre on Influenza and Other Transmissible Infections (CIRI-IT), Genoa, Italy
- Department of Health Sciences, University of Genoa, Italy
| | - Susana Otero-Romero
- Servicio de Medicina Preventiva y Epidemiología, Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Campus Hospitalari, Barcelona, Spain
| | - Sebastian Baumgartner
- Fourth Medical Department with Infectious Diseases and Tropical Medicine, Klinik Favoriten/Kaiser-Franz-Josef Hospital, Vienna, Austria
| | - Charlotte Martin
- Infectious Diseases Department, Centre Hospitalier Universitaire Saint-Pierre, Brussels, Belgium
| | - Sylvia Taylor
- Medical Evidence, Vaccines & Immune Therapies, AstraZeneca, Cambridge, UK
| | | |
Collapse
|
129
|
Marziano V, Guzzetta G, Menegale F, Sacco C, Petrone D, Mateo Urdiales A, Del Manso M, Bella A, Fabiani M, Vescio MF, Riccardo F, Poletti P, Manica M, Zardini A, d'Andrea V, Trentini F, Stefanelli P, Rezza G, Palamara AT, Brusaferro S, Ajelli M, Pezzotti P, Merler S. Estimating SARS-CoV-2 infections and associated changes in COVID-19 severity and fatality. Influenza Other Respir Viruses 2023; 17:e13181. [PMID: 37599801 PMCID: PMC10432583 DOI: 10.1111/irv.13181] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 06/21/2023] [Accepted: 07/19/2023] [Indexed: 08/22/2023] Open
Abstract
Background The difficulty in identifying SARS-CoV-2 infections has not only been the major obstacle to control the COVID-19 pandemic but also to quantify changes in the proportion of infections resulting in hospitalization, intensive care unit (ICU) admission, or death. Methods We developed a model of SARS-CoV-2 transmission and vaccination informed by official estimates of the time-varying reproduction number to estimate infections that occurred in Italy between February 2020 and 2022. Model outcomes were compared with the Italian National surveillance data to estimate changes in the SARS-CoV-2 infection ascertainment ratio (IAR), infection hospitalization ratio (IHR), infection ICU ratio (IIR), and infection fatality ratio (IFR) in five different sub-periods associated with the dominance of the ancestral lineages and Alpha, Delta, and Omicron BA.1 variants. Results We estimate that, over the first 2 years of pandemic, the IAR ranged between 15% and 40% (range of 95%CI: 11%-61%), with a peak value in the second half of 2020. The IHR, IIR, and IFR consistently decreased throughout the pandemic with 22-44-fold reductions between the initial phase and the Omicron period. At the end of the study period, we estimate an IHR of 0.24% (95%CI: 0.17-0.36), IIR of 0.015% (95%CI: 0.011-0.023), and IFR of 0.05% (95%CI: 0.04-0.08). Conclusions Since 2021, changes in the dominant SARS-CoV-2 variant, vaccination rollout, and the shift of infection to younger ages have reduced SARS-CoV-2 infection ascertainment. The same factors, combined with the improvement of patient management and care, contributed to a massive reduction in the severity and fatality of COVID-19.
Collapse
Affiliation(s)
| | - Giorgio Guzzetta
- Center for Health EmergenciesBruno Kessler FoundationTrentoItaly
| | - Francesco Menegale
- Center for Health EmergenciesBruno Kessler FoundationTrentoItaly
- Department of MathematicsUniversity of TrentoTrentoItaly
| | - Chiara Sacco
- Department of Infectious DiseasesIstituto Superiore di SanitàRomeItaly
| | - Daniele Petrone
- Department of Infectious DiseasesIstituto Superiore di SanitàRomeItaly
| | | | - Martina Del Manso
- Department of Infectious DiseasesIstituto Superiore di SanitàRomeItaly
| | - Antonino Bella
- Department of Infectious DiseasesIstituto Superiore di SanitàRomeItaly
| | - Massimo Fabiani
- Department of Infectious DiseasesIstituto Superiore di SanitàRomeItaly
| | | | - Flavia Riccardo
- Department of Infectious DiseasesIstituto Superiore di SanitàRomeItaly
| | - Piero Poletti
- Center for Health EmergenciesBruno Kessler FoundationTrentoItaly
| | - Mattia Manica
- Center for Health EmergenciesBruno Kessler FoundationTrentoItaly
| | - Agnese Zardini
- Center for Health EmergenciesBruno Kessler FoundationTrentoItaly
| | - Valeria d'Andrea
- Center for Health EmergenciesBruno Kessler FoundationTrentoItaly
| | - Filippo Trentini
- Center for Health EmergenciesBruno Kessler FoundationTrentoItaly
- Dondena Centre for Research on Social Dynamics and Public PolicyBocconi UniversityMilanItaly
- COVID Crisis LabBocconi UniversityMilanItaly
| | - Paola Stefanelli
- Department of Infectious DiseasesIstituto Superiore di SanitàRomeItaly
| | - Giovanni Rezza
- Health Prevention directorateMinistry of HealthRomeItaly
| | | | - Silvio Brusaferro
- Department of Infectious DiseasesIstituto Superiore di SanitàRomeItaly
| | - Marco Ajelli
- Laboratory for Computational Epidemiology and Public Health, Department of Epidemiology and BiostatisticsIndiana University School of Public HealthBloomingtonIndianaUSA
| | - Patrizio Pezzotti
- Department of Infectious DiseasesIstituto Superiore di SanitàRomeItaly
| | - Stefano Merler
- Center for Health EmergenciesBruno Kessler FoundationTrentoItaly
| |
Collapse
|
130
|
España PP, Bilbao-González A, Larrea N, Castillo-Sintes I, García-Gutiérrez S, Portuondo J, Villanueva A, Uranga A, Legarreta MJ, Gascon M, Quintana JM. Impact of prior SARS-COV-2 infection and vaccination on COVID-19 hospital admission and mortality amongst nursing home residents. Aging Clin Exp Res 2023; 35:1771-1778. [PMID: 37249860 PMCID: PMC10228436 DOI: 10.1007/s40520-023-02446-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/17/2023] [Indexed: 05/31/2023]
Abstract
BACKGROUND Nursing home residents (NHRs) have experienced disproportionately high risk of severe outcomes due to COVID-19 infection. AIM We investigated the impact of COVID-19 vaccinations and previous SARS-CoV-2 episodes in preventing hospitalization and mortality in NHRs. METHODS Retrospective study of a cohort of all NHRs in our area who were alive at the start of the vaccination campaign. The first three doses of SARS-CoV-2 vaccine and prior COVID-19 infections were registered. The main outcomes were hospital admission and mortality during each follow up. Random effects time-varying Cox models adjusted for age, sex, and comorbidities were fitted to estimate hazard ratios (HRs) according to vaccination status. RESULTS COVID-19 hospitalization and death rates for unvaccinated NHRs were respectively 2.39 and 1.42 per 10,000 person-days, falling after administration of the second dose (0.37 and 0.34) and rising with the third dose (1.08 and 0.8). Rates were much lower amongst people who had previously had COVID-19. Adjusted HRs indicated a significant decrease in hospital admission amongst those with a two- and three-dose status; those who had had a previous COVID-19 infection had even lower hospital admission rates. Death rates decreased as NHRs received two and three doses, and the probability of death was much lower among those who had previously had the infection. CONCLUSIONS The effectiveness of current vaccines against severe COVID-19 disease in NHRs remains high and SARS-CoV-2 episodes prior to vaccination entail a major reduction in hospitalization and mortality rates. The protection conferred by vaccines appears to decline in the following months. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT04463706.
Collapse
Affiliation(s)
- Pedro P España
- Respiratory Service, Osakidetza Basque Health Service, Galdakao-Usansolo University Hospital, Galdakao, Spain.
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain.
- Respiratory Unit, Galdakao-Usansolo University Hospital, Galdakao, Spain.
| | - Amaia Bilbao-González
- Research and Innovation Unit, Osakidetza Basque Health Service, Basurto University Hospital, Bilbao, Spain
- Kronikgune Institute for Health Services Research, Barakaldo, Spain
- Health Service Research Network On Chronic Diseases (REDISSEC), Bilbao, Spain
- Network for Research On Chronicity, Primary Care, and Health Promotion (RICAPPS), Galdakao, Spain
| | - Nere Larrea
- Research Unit, Osakidetza Basque Health Service, Galdakao-Usansolo University Hospital, Galdakao, Spain
- Kronikgune Institute for Health Services Research, Barakaldo, Spain
- Health Service Research Network On Chronic Diseases (REDISSEC), Bilbao, Spain
- Network for Research On Chronicity, Primary Care, and Health Promotion (RICAPPS), Galdakao, Spain
| | - Idoia Castillo-Sintes
- Research and Innovation Unit, Osakidetza Basque Health Service, Basurto University Hospital, Bilbao, Spain
- Kronikgune Institute for Health Services Research, Barakaldo, Spain
- Network for Research On Chronicity, Primary Care, and Health Promotion (RICAPPS), Galdakao, Spain
| | - Susana García-Gutiérrez
- Research Unit, Osakidetza Basque Health Service, Galdakao-Usansolo University Hospital, Galdakao, Spain
- Kronikgune Institute for Health Services Research, Barakaldo, Spain
- Health Service Research Network On Chronic Diseases (REDISSEC), Bilbao, Spain
- Network for Research On Chronicity, Primary Care, and Health Promotion (RICAPPS), Galdakao, Spain
| | - Janire Portuondo
- Osakidetza Basque Health Service, Sub-Directorate for Primary Care Coordination, Vitoria-Gasteiz, Spain
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Network for Research On Chronicity, Primary Care, and Health Promotion (RICAPPS), Galdakao, Spain
| | - Ane Villanueva
- Research Unit, Osakidetza Basque Health Service, Galdakao-Usansolo University Hospital, Galdakao, Spain
- Kronikgune Institute for Health Services Research, Barakaldo, Spain
- Health Service Research Network On Chronic Diseases (REDISSEC), Bilbao, Spain
- Network for Research On Chronicity, Primary Care, and Health Promotion (RICAPPS), Galdakao, Spain
| | - Ane Uranga
- Respiratory Service, Osakidetza Basque Health Service, Galdakao-Usansolo University Hospital, Galdakao, Spain
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Maria J Legarreta
- Research Unit, Osakidetza Basque Health Service, Galdakao-Usansolo University Hospital, Galdakao, Spain
- Kronikgune Institute for Health Services Research, Barakaldo, Spain
- Health Service Research Network On Chronic Diseases (REDISSEC), Bilbao, Spain
- Network for Research On Chronicity, Primary Care, and Health Promotion (RICAPPS), Galdakao, Spain
| | - Maria Gascon
- Research Unit, Osakidetza Basque Health Service, Galdakao-Usansolo University Hospital, Galdakao, Spain
- Kronikgune Institute for Health Services Research, Barakaldo, Spain
- Health Service Research Network On Chronic Diseases (REDISSEC), Bilbao, Spain
- Network for Research On Chronicity, Primary Care, and Health Promotion (RICAPPS), Galdakao, Spain
| | - Jose M Quintana
- Research Unit, Osakidetza Basque Health Service, Galdakao-Usansolo University Hospital, Galdakao, Spain
- Kronikgune Institute for Health Services Research, Barakaldo, Spain
- Health Service Research Network On Chronic Diseases (REDISSEC), Bilbao, Spain
- Network for Research On Chronicity, Primary Care, and Health Promotion (RICAPPS), Galdakao, Spain
| |
Collapse
|
131
|
Hara M, Furue T, Fukuoka M, Iwanaga K, Matsuishi E, Miike T, Sakamoto Y, Mukai N, Kinugasa Y, Shigyo M, Sonoda N, Tanaka M, Arase Y, Tanaka Y, Nakashima H, Irie S, Hirota Y. Comparison of self-reported symptoms in COVID-19 patients who had or had not previously received COVID-19 mRNA vaccination. Hum Vaccin Immunother 2023; 19:2226575. [PMID: 37357433 PMCID: PMC10294725 DOI: 10.1080/21645515.2023.2226575] [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: 03/13/2023] [Accepted: 06/13/2023] [Indexed: 06/27/2023] Open
Abstract
Although mRNA coronavirus disease 2019 (COVID-19) vaccines have been reported for high effectiveness against symptoms, it remains unclear whether post-vaccination infections are less symptomatic than infections in vaccine-naive individuals. We included patients with COVID-19 diagnosed by polymerase chain reaction tests during Japan's alpha and delta variant epidemics. COVID-19 symptoms at approximately 4 weeks were compared based on COVID-19 vaccination status. In total, 398 cases (372 symptomatic and 26 asymptomatic; 286 unvaccinated, 66 vaccinated with one dose, and 46 with two doses) were analyzed. The most common symptoms were fever (78.4%), fatigue (78.4%), cough (74.4%), loss of taste or smell (62.8%), and headache (59.8%). Post-vaccination infections were significantly less likely to be symptomatic. Possible confounder-adjusted odds ratios of two vaccine doses against fatigue, dry eyes and mouth, insomnia, fever, shortness of breath, unusual muscle pains, and loss of taste or smell were 0.18 (95% confidence interval [CI]: 0.09-0.38), 0.22 (95% CI: 0.08-0.59), 0.33 (95% CI: 0.14-0.80), 0.31 (95% CI: 0.15-0.63), 0.36 (95% CI: 0.16-0.76), 0.40 (95% CI: 0.19-0.82), and 0.44 (95% CI: 0.22-0.87), respectively. Post-vaccination infections after two mRNA COVID-19 vaccine doses show milder and fewer symptoms than infections in unvaccinated patients, highlighting the effectiveness of vaccination.
Collapse
Affiliation(s)
- Megumi Hara
- Department of Preventive Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | | | | | | | | | - Toru Miike
- Department of Emergency Medicine, Saga University Hospital, Saga, Japan
| | - Yuichiro Sakamoto
- Department of Emergency Medicine, Saga University Hospital, Saga, Japan
| | - Naoko Mukai
- Fukuoka City Jonan Ward Public Health Center, Fukuoka, Japan
| | - Yuki Kinugasa
- Fukuoka City Higashi Ward Public Health Center, Fukuoka, Japan
| | - Mutsumi Shigyo
- Fukuoka City Hakata Ward Public Health Center, Fukuoka, Japan
| | - Noriko Sonoda
- Fukuoka City Sawara Ward Public Health Center, Fukuoka, Japan
| | | | | | - Yosuke Tanaka
- Kanenokuma Hospital, SOUSEIKAI Medical Group (Medical Co. LTA), Fukuoka, Japan
| | | | - Shin Irie
- SOUSEIKAI Medical Group (Medical Co. LTA), Fukuoka, Japan
| | - Yoshio Hirota
- Clinical Epidemiology Research Center, SOUSEIKAI Medical Group (Medical Co. LTA), Fukuoka, Japan
| |
Collapse
|
132
|
Chavda VP, Apostolopoulos V. Mosaic receptor binding domain nanoparticles: towards fourth-generation vaccination. Nanomedicine (Lond) 2023; 18:1223-1226. [PMID: 37593937 DOI: 10.2217/nnm-2022-0316] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/19/2023] Open
Affiliation(s)
- Vivek P Chavda
- Department of Pharmaceutics & Pharmaceutical Technology, LM College of Pharmacy, Ahmedabad, 380008, Gujarat, India
| | - Vasso Apostolopoulos
- Institute for Health & Sport, Victoria University, Melbourne, VIC, 3030, Australia
| |
Collapse
|
133
|
Gromowski GD, Cincotta CM, Mayer S, King J, Swafford I, McCracken MK, Coleman D, Enoch J, Storme C, Darden J, Peel S, Epperson D, McKee K, Currier JR, Okulicz J, Paquin-Proulx D, Cowden J, Peachman K. Humoral immune responses associated with control of SARS-CoV-2 breakthrough infections in a vaccinated US military population. EBioMedicine 2023; 94:104683. [PMID: 37413891 PMCID: PMC10345251 DOI: 10.1016/j.ebiom.2023.104683] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 06/09/2023] [Accepted: 06/13/2023] [Indexed: 07/08/2023] Open
Abstract
BACKGROUND COVID-19 vaccines have been critical for protection against severe disease following infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) but gaps remain in our understanding of the immune responses that contribute to controlling subclinical and mild infections. METHODS Vaccinated, active-duty US military service members were enrolled in a non-interventional, minimal-risk, observational study starting in May, 2021. Clinical data, serum, and saliva samples were collected from study participants and were used to characterise the humoral immune responses to vaccination and to assess its impact on clinical and subclinical infections, as well as virologic outcomes of breakthrough infections (BTI) including viral load and infection duration. FINDINGS The majority of VIRAMP participants had received the Pfizer COVID-19 vaccine and by January, 2022, N = 149 had a BTI. The median BTI duration (PCR+ days) was 4 days and the interquartile range was 1-8 days. Participants that were nucleocapsid seropositive prior to their BTI had significantly higher levels of binding and functional antibodies to the spike protein, shorter median duration of infections, and lower median peak viral loads compared to seronegative participants. Furthermore, levels of neutralising antibody, ACE2 blocking activity, and spike-specific IgA measured prior to BTI also correlated with the duration of infection. INTERPRETATION We extended previous findings and demonstrate that a subset of vaccine-induced humoral immune responses, along with nucleocapsid serostatus are associated with control of SARS-CoV-2 breakthrough infections in the upper airways. FUNDING This work was funded by the DoD Joint Program Executive Office for Chemical, Biological, Radiological and Nuclear Defense (JPEO-CBRND) in collaboration with the Defense Health Agency (DHA) COVID-19 funding initiative for the VIRAMP study.
Collapse
Affiliation(s)
- Gregory D Gromowski
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA.
| | - Camila Macedo Cincotta
- Diagnostics and Countermeasures Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Sandra Mayer
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA; Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Jocelyn King
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA; Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Isabella Swafford
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA; U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Michael K McCracken
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Dante Coleman
- Diagnostics and Countermeasures Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Jennifer Enoch
- Diagnostics and Countermeasures Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Casey Storme
- Diagnostics and Countermeasures Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Janice Darden
- Diagnostics and Countermeasures Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Sheila Peel
- Diagnostics and Countermeasures Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Diane Epperson
- Booz Allen Hamilton, McLean, VA, USA; Enabling Biotechnologies, Joint Program Executive Office for Chemical, Biological, Radiological and Nuclear Defense, Frederick, MD, USA
| | | | - Jeffrey R Currier
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Jason Okulicz
- Department of Infectious Disease, Brooke Army Medical Center, San Antonio, TX, USA
| | - Dominic Paquin-Proulx
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA; U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Jessica Cowden
- Enabling Biotechnologies, Joint Program Executive Office for Chemical, Biological, Radiological and Nuclear Defense, Frederick, MD, USA; Department of Retrovirology, U.S. Army Medical Directorate-Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand.
| | - Kristina Peachman
- Diagnostics and Countermeasures Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| |
Collapse
|
134
|
Shang S, Fu B, Geng Y, Zhang J, Zhang D, Xiao F, Sheng Z, Zhai J, Li W, Chen X, Zheng C, Li Q. Azvudine therapy of common COVID-19 in hemodialysis patients. J Med Virol 2023; 95:e29007. [PMID: 37522276 DOI: 10.1002/jmv.29007] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 07/13/2023] [Accepted: 07/15/2023] [Indexed: 08/01/2023]
Abstract
There is no antiviral study on hemodialysis patients infected with coronavirus disease 2019 (COVID-19), especially on the application of 2'-deoxy-2'-β -fluoro-4'-azidocytidine (Azvudine, FNC) antiviral therapy. We conducted a multicenter observational study involving 1008 hemodialysis patients. After matching for age, sex, and other factors, 182 patients in the basic treatment group and 182 in the FNC group were included. The negative nucleic acid conversion rate of the FNC group was significantly higher than that of the basic treatment group, and viral loads, interleukin-6, and C-reactive protein were significantly lower than those of the basic treatment group (p < 0.05). There were no significant differences in liver function, renal function, or the number of adverse events between the two groups (p > 0.05). In conclusion, our study has provided novel evidence suggesting that the FNC scheme may be safe and effective compared to the basic treatment of hemodialysis patients with common COVID-19.
Collapse
Affiliation(s)
- Shunlai Shang
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China
- Department of Nephrology, The First Medical Center, Chinese PLA General Hospital, Medical School of Chinese PLA, Chinese PLA Institute of Nephrology, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| | - Bo Fu
- Department of Nephrology, The First Medical Center, Chinese PLA General Hospital, Medical School of Chinese PLA, Chinese PLA Institute of Nephrology, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| | - Yanqiu Geng
- Department of Nephrology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jian Zhang
- Department of Nephrology, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Dawei Zhang
- Senior Department of Infectious Diseases of the Fifth Medical Center, PLA General Hospital, Beijing, China
| | - Fenglin Xiao
- Department of Nephrology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhaojun Sheng
- College of Pulmonary and Critical Care Medicine, the First Medical Center Chinese PLA General Hospital, Beijing, China
| | - Jingbo Zhai
- Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Medical College, Inner Mongolia Minzu University, Tongliao, China
| | - Wenge Li
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China
| | - Xiangmei Chen
- Department of Nephrology, The First Medical Center, Chinese PLA General Hospital, Medical School of Chinese PLA, Chinese PLA Institute of Nephrology, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| | - Chunfu Zheng
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou City, Guangdong Province, China
| | - Qinggang Li
- Department of Nephrology, The First Medical Center, Chinese PLA General Hospital, Medical School of Chinese PLA, Chinese PLA Institute of Nephrology, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| |
Collapse
|
135
|
Ruiz JG, Andrew MK. COVID-19 vaccination and hybrid immunity in older adults. THE LANCET. HEALTHY LONGEVITY 2023; 4:e364-e365. [PMID: 37459880 DOI: 10.1016/s2666-7568(23)00112-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 08/07/2023] Open
Affiliation(s)
- Jorge G Ruiz
- Division of Geriatric Medicine, Department of Medicine, Memorial Healthcare System, Hollywood, FL, USA.
| | - Melissa K Andrew
- Division of Geriatric Medicine, Department of Medicine, Department of Community Health and Epidemiology, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
136
|
Pierre CN, Adams LE, Anasti K, Goodman D, Stanfield-Oakley S, Powers JM, Li D, Rountree W, Wang Y, Edwards RJ, Munir Alam S, Ferrari G, Tomaras GD, Haynes BF, Baric RS, Saunders KO. Non-neutralizing SARS-CoV-2 N-terminal domain antibodies protect mice against severe disease using Fc-mediated effector functions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.25.550460. [PMID: 37546738 PMCID: PMC10402036 DOI: 10.1101/2023.07.25.550460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Antibodies perform both neutralizing and non-neutralizing effector functions that protect against certain pathogen-induced diseases. A human antibody directed at the SARS-CoV-2 Spike N-terminal domain (NTD), DH1052, was recently shown to be non-neutralizing yet it protected mice and cynomolgus macaques from severe disease. The mechanisms of this non-neutralizing antibody-mediated protection are unknown. Here we show that Fc effector functions mediate non-neutralizing antibody (non-nAb) protection against SARS-CoV-2 MA10 viral challenge in mice. Though non-nAb infusion did not suppress infectious viral titers in the lung as potently as NTD neutralizing antibody (nAb) infusion, disease markers including gross lung discoloration were similar in nAb and non-nAb groups. Fc functional knockout substitutions abolished non-nAb protection and increased viral titers in the nAb group. Finally, Fc enhancement increased non-nAb protection relative to WT, supporting a positive association between Fc functionality and degree of protection in SARS-CoV-2 infection. This study demonstrates that non-nAbs can utilize Fc-mediated mechanisms to lower viral load and prevent lung damage due to coronavirus infection.
Collapse
Affiliation(s)
- Camille N Pierre
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC USA
- Duke University School of Medicine, Durham, NC USA
| | - Lily E Adams
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Kara Anasti
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC USA
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Derrick Goodman
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC USA
| | | | - John M Powers
- Department of Immunology, Duke University, Durham, NC USA
| | - Dapeng Li
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC USA
| | - Wes Rountree
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC USA
- Department of Medicine, Duke University School of Medicine, Durham, NC USA
| | - Yunfei Wang
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC USA
- Department of Medicine, Duke University School of Medicine, Durham, NC USA
| | - Robert J Edwards
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC USA
- Department of Medicine, Duke University School of Medicine, Durham, NC USA
| | - S Munir Alam
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC USA
- Department of Medicine, Duke University School of Medicine, Durham, NC USA
| | - Guido Ferrari
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC USA
- Department of Surgery, Duke University School of Medicine, Durham, NC USA
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC USA
| | - Georgia D Tomaras
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC USA
- Department of Surgery, Duke University School of Medicine, Durham, NC USA
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC USA
- Department of Immunology, Duke University, Durham, NC USA
| | - Barton F Haynes
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC USA
- Duke University School of Medicine, Durham, NC USA
- Department of Immunology, Duke University, Durham, NC USA
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Kevin O Saunders
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC USA
- Department of Surgery, Duke University School of Medicine, Durham, NC USA
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC USA
- Department of Immunology, Duke University, Durham, NC USA
| |
Collapse
|
137
|
Hogan AB, Doohan P, Wu SL, Mesa DO, Toor J, Watson OJ, Winskill P, Charles G, Barnsley G, Riley EM, Khoury DS, Ferguson NM, Ghani AC. Estimating long-term vaccine effectiveness against SARS-CoV-2 variants: a model-based approach. Nat Commun 2023; 14:4325. [PMID: 37468463 DOI: 10.1038/s41467-023-39736-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 06/27/2023] [Indexed: 07/21/2023] Open
Abstract
With the ongoing evolution of the SARS-CoV-2 virus updated vaccines may be needed. We fitted a model linking immunity levels and protection to vaccine effectiveness data from England for three vaccines (Oxford/AstraZeneca AZD1222, Pfizer-BioNTech BNT162b2, Moderna mRNA-1273) and two variants (Delta, Omicron). Our model reproduces the observed sustained protection against hospitalisation and death from the Omicron variant over the first six months following dose 3 with the ancestral vaccines but projects a gradual waning to moderate protection after 1 year. Switching the fourth dose to a variant-matched vaccine against Omicron BA.1/2 is projected to prevent nearly twice as many hospitalisations and deaths over a 1-year period compared to administering the ancestral vaccine. This result is sensitive to the degree to which immunogenicity data can be used to predict vaccine effectiveness and uncertainty regarding the impact that infection-induced immunity (not captured here) may play in modifying future vaccine effectiveness.
Collapse
Affiliation(s)
- Alexandra B Hogan
- School of Population Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
- MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, UK
| | - Patrick Doohan
- MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, UK
| | - Sean L Wu
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, USA
| | - Daniela Olivera Mesa
- MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, UK
| | - Jaspreet Toor
- MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, UK
| | - Oliver J Watson
- MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, UK
- London School of Hygiene and Tropical Medicine, London, UK
| | - Peter Winskill
- MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, UK
| | - Giovanni Charles
- MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, UK
| | - Gregory Barnsley
- MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, UK
- London School of Hygiene and Tropical Medicine, London, UK
| | - Eleanor M Riley
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - David S Khoury
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| | - Neil M Ferguson
- MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, UK
| | - Azra C Ghani
- MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, UK.
| |
Collapse
|
138
|
Ates I, Batirel A, Aydin M, Karadag FY, Erden A, Kucuksahin O, Armagan B, Guven SC, Karakas O, Gokdemir S, Altunal LN, Buber AA, Gemcioglu E, Zengin O, Inan O, Sahiner ES, Korukluoglu G, Sezer Z, Ozdarendeli A, Omma A, Kara A. Long-Term Results of Immunogenicity of Booster Vaccination against SARS-CoV-2 (Hybrid COV-RAPEL TR Study) in Turkiye: A Double-Blind, Randomized, Controlled, Multicenter Phase 2 Clinical Study. Vaccines (Basel) 2023; 11:1234. [PMID: 37515050 PMCID: PMC10416156 DOI: 10.3390/vaccines11071234] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/21/2023] [Accepted: 06/23/2023] [Indexed: 07/30/2023] Open
Abstract
The immunogenicity of vaccines decreases over time, causing a need for booster doses. This study aimed to present the long-term (Day 84) immunogenicity results of the double-blind, randomized, controlled, phase II Hybrid COV-RAPEL TR Study (NCT04979949), in which the TURKOVAC or CoronaVac vaccines were used as a booster after the second dose of primary vaccination with CoronaVac. A total of 190 participants from the Hybrid COV-RAPEL TR Study, who had both Day 28 and Day 84 immunogenicity results, were included. The immunogenicity on Day 84, regarding the neutralizing antibody positivity (Wuhan and Delta variants) and anti-spike immunoglobulin (Ig) G (IgG) antibody positivity, was compared between TURKOVAC and CoronaVac vaccine arms according to sex and age groups. Overall, antibody positivity showed a slight decrease on Day 84 vs. Day 28, but was not different between TURKOVAC and CoronaVac arms either for sexes or for age groups. However, TURKOVAC produced better antibody response against the Delta variant than CoronaVac, while CoronaVac was superior over TURKOVAC regarding neutralizing antibody positivity in the 50-60 years age group, regardless of the variant. A single booster dose, after the completion of the primary vaccination, increases antibody positivity on Day 28 which persists until Day 84 with a slight decrease. However, an additional booster dose may be required thereafter, since the decrease in antibody titer may be faster over time.
Collapse
Affiliation(s)
- Ihsan Ates
- Department of Internal Medicine, University of Health Sciences, Ankara City Hospital, 06800 Ankara, Türkiye
| | - Ayse Batirel
- Department of Infectious Diseases and Clinical Microbiology, University of Health Sciences, International Medical School, Kartal Dr. Lutfi Kirdar City Hospital, 34865 Istanbul, Türkiye
| | - Mehtap Aydin
- Department of Infectious Diseases and Clinical Microbiology, University of Health Sciences, Umraniye Training and Research Hospital, 34760 Istanbul, Türkiye
| | - Fatma Yilmaz Karadag
- Department of Infectious Diseases, University of Health Sciences, Sancaktepe Sehit Prof. Dr. Ilhan Varank Training and Research Hospital, 34785 Istanbul, Türkiye
| | - Abdulsamet Erden
- Clinic of Rheumatology, Ankara City Hospital, 06800 Ankara, Türkiye (B.A.)
| | - Orhan Kucuksahin
- Clinic of Rheumatology, Ankara City Hospital, 06800 Ankara, Türkiye (B.A.)
| | - Berkan Armagan
- Clinic of Rheumatology, Ankara City Hospital, 06800 Ankara, Türkiye (B.A.)
| | - Serdar Can Guven
- Clinic of Rheumatology, Ankara City Hospital, 06800 Ankara, Türkiye (B.A.)
| | - Ozlem Karakas
- Clinic of Rheumatology, Ankara City Hospital, 06800 Ankara, Türkiye (B.A.)
| | - Selim Gokdemir
- Department of Clinical Pharmacology, University of Health Sciences, Kartal Dr. Lutfi Kirdar City Hospital, 34865 Istanbul, Türkiye
| | - Lutfiye Nilsun Altunal
- Department of Infectious Diseases and Clinical Microbiology, University of Health Sciences, Umraniye Training and Research Hospital, 34760 Istanbul, Türkiye
| | - Aslihan Ayse Buber
- Department of Infectious Diseases, University of Health Sciences, Sancaktepe Sehit Prof. Dr. Ilhan Varank Training and Research Hospital, 34785 Istanbul, Türkiye
| | - Emin Gemcioglu
- Department of Internal Medicine, University of Health Sciences, Ankara City Hospital, 06800 Ankara, Türkiye
| | - Oguzhan Zengin
- Department of Internal Medicine, University of Health Sciences, Ankara City Hospital, 06800 Ankara, Türkiye
| | - Osman Inan
- Department of Internal Medicine, University of Health Sciences, Ankara City Hospital, 06800 Ankara, Türkiye
| | - Enes Seyda Sahiner
- Department of Internal Medicine, University of Health Sciences, Ankara City Hospital, 06800 Ankara, Türkiye
| | - Gulay Korukluoglu
- Virology Laboratory, General Directorate of Public Health, 06560 Ankara, Türkiye
| | - Zafer Sezer
- Department of Pharmacology, Erciyes University, 38030 Kayseri, Türkiye
| | - Aykut Ozdarendeli
- Vaccine Research, Development and Application Center, Erciyes University, 38280 Kayseri, Türkiye
- Department of Microbiology, Medical Faculty, Erciyes University, 38030 Kayseri, Türkiye
| | - Ahmet Omma
- Clinic of Rheumatology, University of Health Sciences, Ankara City Hospital, 06800 Ankara, Türkiye
| | - Ates Kara
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Hacettepe University, 06230 Ankara, Türkiye
- Türkiye Vaccine Institute, 06270 Ankara, Türkiye
| |
Collapse
|
139
|
Bosworth ML, Schofield R, Ayoubkhani D, Charlton L, Nafilyan V, Khunti K, Zaccardi F, Gillies C, Akbari A, Knight M, Wood R, Hardelid P, Zuccolo L, Harrison C. Vaccine effectiveness for prevention of covid-19 related hospital admission during pregnancy in England during the alpha and delta variant dominant periods of the SARS-CoV-2 pandemic: population based cohort study. BMJ MEDICINE 2023; 2:e000403. [PMID: 37564827 PMCID: PMC10410807 DOI: 10.1136/bmjmed-2022-000403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 05/18/2023] [Indexed: 08/12/2023]
Abstract
Objective To estimate vaccine effectiveness for preventing covid-19 related hospital admission in individuals first infected with the SARS-CoV-2 virus during pregnancy compared with those of reproductive age who were not pregnant when first infected with the virus. Design Population based cohort study. Setting Office for National Statistics Public Health Data Asset linked dataset, providing national linked census and administrative data in England, 8 December 2020 to 31 August 2021. Participants 815 477 females aged 18-45 years (mean age 30.4 years) who had documented evidence of a first SARS-CoV-2 infection in the NHS Test and Trace or Hospital Episode Statistics data. Main outcome measures Hospital admission where covid-19 was recorded as the primary diagnosis. Cox proportional hazards models, adjusted for calendar time of infection, sociodemographic factors, and pre-existing health conditions related to uptake of the covid-19 vaccine and risk of severe covid-19 outcomes, were used to estimate vaccine effectiveness as the complement of the hazard ratio for hospital admission for covid-19. Results Compared with pregnant individuals who were not vaccinated, the adjusted rate of hospital admission for covid-19 was 77% (95% confidence interval 70% to 82%) lower for pregnant individuals who had received one dose and 83% (76% to 89%) lower for those who had received two doses of vaccine. These estimates were similar to those found in the non-pregnant group: 79% (77% to 81%) for one dose and 83% (82% to 85%) for two doses of vaccine. Among those who were vaccinated >90 days before infection, having two doses of vaccine was associated with a greater reduction in risk than one dose. Conclusions Covid-19 vaccination was associated with reduced rates of hospital admission in pregnant individuals infected with the SARS-CoV-2 virus, and the reduction in risk was similar to that in non-pregnant individuals. Waning of vaccine effectiveness occurred more quickly after one than after two doses of vaccine.
Collapse
Affiliation(s)
| | | | - Daniel Ayoubkhani
- Office for National Statistics, Newport, UK
- Real World Evidence Unit, Diabetes Research Centre, University of Leicester, Leicester, UK
| | | | - Vahé Nafilyan
- Office for National Statistics, Newport, UK
- London School of Hygiene and Tropical Medicine, London, UK
| | - Kamlesh Khunti
- Real World Evidence Unit, Diabetes Research Centre, University of Leicester, Leicester, UK
| | - Francesco Zaccardi
- Real World Evidence Unit, Diabetes Research Centre, University of Leicester, Leicester, UK
| | - Clare Gillies
- Real World Evidence Unit, Diabetes Research Centre, University of Leicester, Leicester, UK
| | - Ashley Akbari
- Population Data Science, Swansea University Medical School, Swansea University, Swansea, UK
| | - Marian Knight
- National Perinatal Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Rachael Wood
- Public Health Scotland, Edinburgh, UK
- The University of Edinburgh Usher Institute of Population Health Sciences and Informatics, Edinburgh, UK
| | - Pia Hardelid
- NIHR Great Ormond Street Hospital Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Luisa Zuccolo
- Health Data Science Centre, Fondazione Human Technopole, Milan, Italy
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | | |
Collapse
|
140
|
Graham S, Tessier E, Stowe J, Bernal JL, Parker EPK, Nitsch D, Miller E, Andrews N, Walker JL, McDonald HI. Bias assessment of a test-negative design study of COVID-19 vaccine effectiveness used in national policymaking. Nat Commun 2023; 14:3984. [PMID: 37414791 PMCID: PMC10325974 DOI: 10.1038/s41467-023-39674-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 06/21/2023] [Indexed: 07/08/2023] Open
Abstract
National test-negative-case-control (TNCC) studies are used to monitor COVID-19 vaccine effectiveness in the UK. A questionnaire was sent to participants from the first published TNCC COVID-19 vaccine effectiveness study conducted by the UK Health Security Agency, to assess for potential biases and changes in behaviour related to vaccination. The original study included symptomatic adults aged ≥70 years testing for COVID-19 between 08/12/2020 and 21/02/2021. A questionnaire was sent to cases and controls tested from 1-21 February 2021. In this study, 8648 individuals responded to the questionnaire (36.5% response). Using information from the questionnaire to produce a combined estimate that accounted for all potential biases decreased the original vaccine effectiveness estimate after two doses of BNT162b2 from 88% (95% CI: 79-94%) to 85% (95% CI: 68-94%). Self-reported behaviour demonstrated minimal evidence of riskier behaviour after vaccination. These findings offer reassurance to policy makers and clinicians making decisions based on COVID-19 vaccine effectiveness TNCC studies.
Collapse
Affiliation(s)
- Sophie Graham
- London School of Hygiene and Tropical Medicine, London, UK.
- UK Health Security Agency, London, UK.
- National Institute for Health and Care Research (NIHR) Health Protection Research Unit in Vaccines and Immunisation, London, UK.
| | | | | | | | | | - Dorothea Nitsch
- London School of Hygiene and Tropical Medicine, London, UK
- UK Renal Registry, Bristol, UK
- Renal Unit, Royal Free London NHS Foundation Trust, Hertfordshire, UK
| | - Elizabeth Miller
- London School of Hygiene and Tropical Medicine, London, UK
- National Institute for Health and Care Research (NIHR) Health Protection Research Unit in Vaccines and Immunisation, London, UK
| | - Nick Andrews
- UK Health Security Agency, London, UK
- National Institute for Health and Care Research (NIHR) Health Protection Research Unit in Vaccines and Immunisation, London, UK
| | - Jemma L Walker
- London School of Hygiene and Tropical Medicine, London, UK
- UK Health Security Agency, London, UK
- National Institute for Health and Care Research (NIHR) Health Protection Research Unit in Vaccines and Immunisation, London, UK
| | - Helen I McDonald
- London School of Hygiene and Tropical Medicine, London, UK
- National Institute for Health and Care Research (NIHR) Health Protection Research Unit in Vaccines and Immunisation, London, UK
| |
Collapse
|
141
|
Kolla M, Rout NK, Gupta S, Mohapatra SK, Alone VD, Singh P. Outcome of COVID-19 infection and the impact of COVID-19 vaccination in chronic kidney disease patients: A single-center study. Ann Afr Med 2023; 22:347-351. [PMID: 37417024 PMCID: PMC10445720 DOI: 10.4103/aam.aam_81_22] [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: 04/15/2021] [Revised: 01/07/2022] [Accepted: 02/01/2023] [Indexed: 07/08/2023] Open
Abstract
Introduction Chronic kidney disease (CKD) patients have increased vulnerability to infections including Covid-19. There is limited availability of antiviral in CKD patients. All guidelines have prioritized vaccinations for CKD patients. The variability in immunogenic response is multifactorial in CKD group. We have tried to analyze the outcome of Covid-19 infection and the impact of COVID-19 vaccination [COVAXIN / COVISHIELD] in our cohort. Materials Methods and Statistical Analysis In a retrospective observational study, 73 cases of Covid-19 positive CKD patients were selected, who were managed as per MOFHW guidelines. The data of first laboratory values and radiological findings were evaluated. Their treatment outcome and stay during hospitalization were studied. All data were later analyzed using STATA 16.1 software. Results In this study, 73 cases of CKD with Covid-19 were included. There were 38 patients who were vaccinated with at least one dose of the Covid-19 vaccine, while there were 35 patients who were unvaccinated. Out of 38 patients, 20 were vaccinated with 2 doses of Covid-19 while 18 received only one dose. The unvaccinated group was having more hypoxia and raised inflammatory markers, and had more lung involvement [i.e. higher CT severity value] [p value for CTSS-0.0765]. There was a higher mortality rate observed in the unvaccinated group [i.e-65.71%] than the vaccinated group [39.47%] [p-value 0.0249]. Dialysis was needed in 57.50% of the study population either due to failure of conservative management for renal failure or due to maintenance dialysis. The mean duration of hospitalization was 11.47 days with a mortality rate of 52% which is much higher than the reported average data in CKD patients. Conclusion Vaccination seems to be very helpful in combating the adverse effect of Covid-19 in CKD patients. It also reduces mortality significantly in Covid-19 infected CKD patients.
Collapse
Affiliation(s)
- Madhuri Kolla
- Department of Pulmonary Medicine, Kalinga Institute of Medical Sciences, KIIT University, Bhubaneswar, Odisha, India
| | - Nikunj Kishore Rout
- Department Nephrology, Kalinga Institute of Medical Sciences, KIIT University, Bhubaneswar, Odisha, India
| | - Saurabh Gupta
- Department of Critical Care Medicine, Kalinga Institute of Medical Sciences, KIIT University, Bhubaneswar, Odisha, India
| | - Sangram Keshari Mohapatra
- Department of Pulmonary Medicine, Kalinga Institute of Medical Sciences, KIIT University, Bhubaneswar, Odisha, India
| | - Vivek D. Alone
- Department of Pulmonary Medicine, Kalinga Institute of Medical Sciences, KIIT University, Bhubaneswar, Odisha, India
| | - Pratima Singh
- Department of Pulmonary Medicine, Kalinga Institute of Medical Sciences, KIIT University, Bhubaneswar, Odisha, India
| |
Collapse
|
142
|
Morgans HA, Bradley T, Flebbe-Rehwaldt L, Selvarangan R, Bagherian A, Barnes AP, Bass J, Cooper AM, Fischer R, Kleiboeker S, Lee BR, LeMaster C, Markus K, Morrison S, Myers A, Myers D, Payne E, Schuster JE, Standley S, Wieser A, Warady B. Humoral and cellular response to the COVID-19 vaccine in immunocompromised children. Pediatr Res 2023; 94:200-205. [PMID: 36376507 PMCID: PMC9662120 DOI: 10.1038/s41390-022-02374-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 10/11/2022] [Accepted: 10/19/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND A suboptimal response to the 2-dose COVID-19 vaccine series in the immunocompromised population prompted recommendations for a 3rd primary dose. We aimed to determine the humoral and cellular immune response to the 3rd COVID-19 vaccine in immunocompromised children. METHODS Prospective cohort study of immunocompromised participants, 5-21 years old, who received 2 prior doses of an mRNA COVID-19 vaccine. Humoral and CD4/CD8 T-cell responses were measured to SARS-CoV-2 spike antigens prior to receiving the 3rd vaccine dose and 3-4 weeks after the 3rd dose was given. RESULTS Of the 37 participants, approximately half were solid organ transplant recipients. The majority (86.5%) had a detectable humoral response after the 2nd and 3rd vaccine doses, with a significant increase in antibody levels after the 3rd dose. Positive T-cell responses increased from being present in 86.5% to 100% of the cohort after the 3rd dose. CONCLUSIONS Most immunocompromised children mount a humoral and cellular immune response to the 2-dose COVID-19 vaccine series, which is significantly augmented after receiving the 3rd vaccine dose. This supports the utility of the 3rd vaccine dose and the rationale for ongoing emphasis for vaccination against COVID-19 in this population. IMPACT Most immunocompromised children mount a humoral and cellular immune response to the 2-dose COVID-19 vaccine series, which is significantly augmented after receiving the 3rd vaccine dose. This is the first prospective cohort study to analyze both the humoral and T-cell immune response to the 3rd COVID-19 primary vaccine dose in children who are immunocompromised. The results of this study support the utility of the 3rd vaccine dose and the rationale for ongoing emphasis for vaccination against COVID-19 in the immunosuppressed pediatric population.
Collapse
Affiliation(s)
- Heather A Morgans
- Children's Mercy Kansas City, Kansas City, MO, 64108, USA.
- University of Missouri-Kansas City, Kansas City, MO, 64110, USA.
| | - Todd Bradley
- Children's Mercy Kansas City, Kansas City, MO, 64108, USA
| | | | | | | | - Aliessa P Barnes
- Children's Mercy Kansas City, Kansas City, MO, 64108, USA
- University of Missouri-Kansas City, Kansas City, MO, 64110, USA
| | - Julie Bass
- Children's Mercy Kansas City, Kansas City, MO, 64108, USA
- University of Missouri-Kansas City, Kansas City, MO, 64110, USA
| | - Ashley M Cooper
- Children's Mercy Kansas City, Kansas City, MO, 64108, USA
- University of Missouri-Kansas City, Kansas City, MO, 64110, USA
| | - Ryan Fischer
- Children's Mercy Kansas City, Kansas City, MO, 64108, USA
- University of Missouri-Kansas City, Kansas City, MO, 64110, USA
| | | | - Brian R Lee
- Children's Mercy Kansas City, Kansas City, MO, 64108, USA
| | - Cas LeMaster
- Children's Mercy Kansas City, Kansas City, MO, 64108, USA
| | - Kelsey Markus
- Children's Mercy Kansas City, Kansas City, MO, 64108, USA
| | | | - Angela Myers
- Children's Mercy Kansas City, Kansas City, MO, 64108, USA
- University of Missouri-Kansas City, Kansas City, MO, 64110, USA
| | - Doug Myers
- Children's Mercy Kansas City, Kansas City, MO, 64108, USA
- University of Missouri-Kansas City, Kansas City, MO, 64110, USA
| | - Erin Payne
- Children's Mercy Kansas City, Kansas City, MO, 64108, USA
| | - Jennifer E Schuster
- Children's Mercy Kansas City, Kansas City, MO, 64108, USA
- University of Missouri-Kansas City, Kansas City, MO, 64110, USA
| | - Sarah Standley
- Children's Mercy Kansas City, Kansas City, MO, 64108, USA
| | - Andrea Wieser
- Children's Mercy Kansas City, Kansas City, MO, 64108, USA
| | - Bradley Warady
- Children's Mercy Kansas City, Kansas City, MO, 64108, USA
- University of Missouri-Kansas City, Kansas City, MO, 64110, USA
| |
Collapse
|
143
|
Feng S, Lin E, Cowling BJ. Number needed to vaccinate for COVID-19 booster doses: a valuable metric to inform vaccination strategies. LANCET REGIONAL HEALTH. AMERICAS 2023; 23:100548. [PMID: 37397875 PMCID: PMC10304837 DOI: 10.1016/j.lana.2023.100548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 06/16/2023] [Accepted: 06/19/2023] [Indexed: 07/04/2023]
Affiliation(s)
- Shuo Feng
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - E Lin
- Department of Surgical Sciences, Faculty of Medicine, Uppsala University, Sweden
| | - Benjamin J Cowling
- 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 Special Administrative Region, China
- Laboratory of Data Discovery for Health Limited, Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region, China
| |
Collapse
|
144
|
Alami A, Krewski D, Farhat N, Mattison D, Wilson K, Gravel CA, Farrell PJ, Crispo JAG, Haddad N, Perez-Lloret S, Villeneuve PJ. Risk of myocarditis and pericarditis in mRNA COVID-19-vaccinated and unvaccinated populations: a systematic review and meta-analysis. BMJ Open 2023; 13:e065687. [PMID: 37339840 DOI: 10.1136/bmjopen-2022-065687] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/22/2023] Open
Abstract
OBJECTIVE To summarise the available evidence on the risk of myocarditis and/or pericarditis following mRNA COVID-19 vaccination, compared with the risk among unvaccinated individuals in the absence of COVID-19 infection. DESIGN Systematic review and meta-analysis. DATA SOURCES Electronic databases (Medline, Embase, Web of Science and WHO Global Literature on Coronavirus Disease), preprint repositories (medRxiv and bioRxiv), reference lists and grey literature were searched from 1 December 2020 until 31 October 2022. STUDY SELECTION Epidemiological studies of individuals of any age who received at least one dose of an mRNA COVID-19 vaccine, reported a risk of myo/pericarditis and compared the risk of myo/pericarditis to individuals who did not receive any dose of an mRNA COVID-19 vaccine. DATA EXTRACTION AND SYNTHESIS Two reviewers independently conducted screening and data extraction. The rate of myo/pericarditis among vaccinated and unvaccinated groups was recorded, and the rate ratios were calculated. Additionally, the total number of individuals, case ascertainment criteria, percentage of males and history of SARS-CoV-2 infection were extracted for each study. Meta-analysis was done using a random-effects model. RESULTS Seven studies met the inclusion criteria, of which six were included in the quantitative synthesis. Our meta-analysis indicates that within 30-day follow-up period, vaccinated individuals were twice as likely to develop myo/pericarditis in the absence of SARS-CoV-2 infection compared to unvaccinated individuals, with a rate ratio of 2.05 (95% CI 1.49-2.82). CONCLUSION Although the absolute number of observed myo/pericarditis cases remains quite low, a higher risk was detected in those who received mRNA COVID-19 vaccinations compared with unvaccinated individuals in the absence of SARS-CoV-2 infection. Given the effectiveness of mRNA COVID-19 vaccines in preventing severe illnesses, hospitalisations and deaths, future research should focus on accurately determining the rates of myo/pericarditis linked to mRNA COVID-19 vaccines, understanding the biological mechanisms behind these rare cardiac events and identifying those most at risk.
Collapse
Affiliation(s)
- Abdallah Alami
- School of Mathematics and Statistics, Faculty of Science, Carleton University, Ottawa, Ontario, Canada
| | - Daniel Krewski
- School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Risk Sciences International, Ottawa, Ontario, Canada
| | - Nawal Farhat
- School of Mathematics and Statistics, Faculty of Science, Carleton University, Ottawa, Ontario, Canada
| | - Donald Mattison
- School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Risk Sciences International, Ottawa, Ontario, Canada
- Arnold School of Public Health, University of South Carolina, Columbia, South Carolina, USA
| | - Kumanan Wilson
- Bruyère Research Institute, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Christopher A Gravel
- School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Québec, Canada
- Department of Mathematics and Statistics, University of Ottawa, Ottawa, Ontario, Canada
| | - Patrick J Farrell
- School of Mathematics and Statistics, Faculty of Science, Carleton University, Ottawa, Ontario, Canada
| | - James A G Crispo
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
- Division of Human Sciences, NOSM University, Sudbury, Ontario, Canada
| | - Nisrine Haddad
- School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Santiago Perez-Lloret
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Observatorio de Salud Pública, Pontificia Universidad Católica Argentina, Buenos Aires, Argentina
- Department of Physiology, Faculty of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Paul J Villeneuve
- Department of Neurosciences, Faculty of Science, Carleton University, Ottawa, Ontario, Canada
| |
Collapse
|
145
|
Song R, Yang C, Li Q, Wang J, Chen J, Sun K, Lv H, Yang Y, Liang J, Ye Q, Gao Y, Li J, Li Y, Yan J, Liu Y, Wang T, Liu C, Zhu P, Wang F, Yin W, Xiang H. Durability of immune response after SARS-CoV-2 vaccination in patients with chronic liver disease. Front Immunol 2023; 14:1200198. [PMID: 37398662 PMCID: PMC10308026 DOI: 10.3389/fimmu.2023.1200198] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/31/2023] [Indexed: 07/04/2023] Open
Abstract
Aim The present study aimed to evaluate the durability of immune response after basic and booster immunization with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines in patients with chronic liver disease (CLD). Methods Patients with CLD and complete basic or booster immunization with SARS-CoV-2 vaccines were included in this study. Based on the vaccination situation, they were divided into the basic immunity group (Basic) and the booster immunity group (Booster), which were then subdivided into four groups according to the time interval from completion of basic immunization or booster immunization to serological specimen collection. The positive rates and antibody titers of novel coronavirus neutralizing antibody (nCoV NTAb) and novel coronavirus spike receptor-binding domain antibody (nCoV S-RBD) were analyzed. Results A total of 313 patients with CLD were enrolled in this study, including 201 in Basic and 112 in Booster. The positive rates of nCoV NTAb and nCoV S-RBD within 30 days of completing basic immunization were 80.4% and 84.8%, respectively, but decreased rapidly with the extension of vaccination time, and only 29% and 48.4% of patients with CLD remained positive for nCoV NTAb and nCoV S-RBD, respectively, after 120 days of completing basic immunization. Within 30 days of booster immunization, the positive rates of nCoV NTAb and nCoV S-RBD in patients with CLD rapidly increased from 29.0% and 48.4% at the end of basic immunization to 95.2% and 90.5%, and maintained a high level (defined as the positive rate >50%) until 120 days when the positive rates of nCoV NTAb and nCoV S-RBD were still high at 79.5% and 87.2%, respectively. After basic immunization, the time for nCoV NTAb and nCoV S-RBD to turn negative was 120 and 169 days, respectively, and the negative time of nCoV NTAb and nCoV S-RBD was significantly prolonged to 266 days and 329 days, respectively. Conclusion It is safe and effective for patients with CLD to complete basic and booster immunization with SARS-CoV-2 vaccines. After booster immunization, the immune response of patients with CLD was further improved and the durability of the SARS-CoV-2 antibody was significantly prolonged.
Collapse
Affiliation(s)
- Ruixin Song
- The Third Central Clinical College of Tianjin Medical University, Department of Gastroenterology and Hepatology, Tianjin Third Central Hospital, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Chao Yang
- Department of Gastroenterology and Hepatology, Tianjin Third Central Hospital, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Institute of Hepatobiliary Disease, Tianjin, China
| | - Qianqian Li
- Department of Gastroenterology and Hepatology, Tianjin Third Central Hospital, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Institute of Hepatobiliary Disease, Tianjin, China
| | - Jiayin Wang
- The Third Central Clinical College of Tianjin Medical University, Department of Gastroenterology and Hepatology, Tianjin Third Central Hospital, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Jing Chen
- The Third Central Clinical College of Tianjin Medical University, Department of Gastroenterology and Hepatology, Tianjin Third Central Hospital, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Kai Sun
- Emergency Department, Tianjin Hongqiao Hospital, Tianjin, China
| | - Hongmin Lv
- Department of Gastroenterology and Hepatology, Tianjin Third Central Hospital, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Institute of Hepatobiliary Disease, Tianjin, China
| | - Yankai Yang
- Department of Gastroenterology and Hepatology, Tianjin Third Central Hospital, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Institute of Hepatobiliary Disease, Tianjin, China
| | - Jing Liang
- Department of Gastroenterology and Hepatology, Tianjin Third Central Hospital, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Institute of Hepatobiliary Disease, Tianjin, China
| | - Qing Ye
- Department of Gastroenterology and Hepatology, Tianjin Third Central Hospital, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Institute of Hepatobiliary Disease, Tianjin, China
| | - YanYing Gao
- Department of Gastroenterology and Hepatology, Tianjin Third Central Hospital, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Institute of Hepatobiliary Disease, Tianjin, China
| | - Jun Li
- Department of Gastroenterology and Hepatology, Tianjin Third Central Hospital, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Institute of Hepatobiliary Disease, Tianjin, China
| | - Ying Li
- Department of Gastroenterology and Hepatology, Tianjin Third Central Hospital, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Institute of Hepatobiliary Disease, Tianjin, China
| | - Junqing Yan
- Department of Gastroenterology and Hepatology, Tianjin Third Central Hospital, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Institute of Hepatobiliary Disease, Tianjin, China
| | - Ying Liu
- Department of Gastroenterology and Hepatology, Tianjin Third Central Hospital, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Institute of Hepatobiliary Disease, Tianjin, China
| | - Tao Wang
- Department of Gastroenterology and Hepatology, Tianjin Third Central Hospital, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Institute of Hepatobiliary Disease, Tianjin, China
| | - Changen Liu
- Department of Gastroenterology and Hepatology, Tianjin Third Central Hospital, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Institute of Hepatobiliary Disease, Tianjin, China
| | - Ping Zhu
- Department of Gastroenterology and Hepatology, Tianjin Third Central Hospital, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Institute of Hepatobiliary Disease, Tianjin, China
| | - Fei Wang
- Department of Gastroenterology and Hepatology, Tianjin Third Central Hospital, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Institute of Hepatobiliary Disease, Tianjin, China
| | - Weili Yin
- Department of Gastroenterology and Hepatology, Tianjin Third Central Hospital, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Institute of Hepatobiliary Disease, Tianjin, China
| | - Huiling Xiang
- Department of Gastroenterology and Hepatology, Tianjin Third Central Hospital, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Institute of Hepatobiliary Disease, Tianjin, China
| |
Collapse
|
146
|
Nowill AE, Caruso M, de Campos-Lima PO. T-cell immunity to SARS-CoV-2: what if the known best is not the optimal course for the long run? Adapting to evolving targets. Front Immunol 2023; 14:1133225. [PMID: 37388738 PMCID: PMC10303130 DOI: 10.3389/fimmu.2023.1133225] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 05/11/2023] [Indexed: 07/01/2023] Open
Abstract
Humanity did surprisingly well so far, considering how unprepared it was to respond to the coronavirus disease 2019 (COVID-19) threat. By blending old and ingenious new technology in the context of the accumulated knowledge on other human coronaviruses, several vaccine candidates were produced and tested in clinical trials in record time. Today, five vaccines account for the bulk of the more than 13 billion doses administered worldwide. The ability to elicit biding and neutralizing antibodies most often against the spike protein is a major component of the protection conferred by immunization but alone it is not enough to limit virus transmission. Thus, the surge in numbers of infected individuals by newer variants of concern (VOCs) was not accompanied by a proportional increase in severe disease and death rate. This is likely due to antiviral T-cell responses, whose evasion is more difficult to achieve. The present review helps navigating the very large literature on T cell immunity induced by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and vaccination. We examine the successes and shortcomings of the vaccinal protection in the light of the emergence of VOCs with breakthrough potential. SARS-CoV-2 and human beings will likely coexist for a long while: it will be necessary to update existing vaccines to improve T-cell responses and attain better protection against COVID-19.
Collapse
Affiliation(s)
- Alexandre E. Nowill
- Integrated Center for Pediatric OncoHaematological Research, State University of Campinas, Campinas, SP, Brazil
| | - Manuel Caruso
- CHU de Québec-Université Laval Research Center (Oncology Division), Université Laval Cancer Research Center, Québec, QC, Canada
| | - Pedro O. de Campos-Lima
- Boldrini Children’s Center, Campinas, SP, Brazil
- Molecular and Morphofunctional Biology Graduate Program, Institute of Biology, State University of Campinas, Campinas, SP, Brazil
| |
Collapse
|
147
|
de Gier B, van Asten L, Boere TM, van Roon A, van Roekel C, Pijpers J, van Werkhoven CHH, van den Ende C, Hahné SJM, de Melker HE, Knol MJ, van den Hof S. Effect of COVID-19 vaccination on mortality by COVID-19 and on mortality by other causes, the Netherlands, January 2021-January 2022. Vaccine 2023:S0264-410X(23)00660-6. [PMID: 37328352 PMCID: PMC10247887 DOI: 10.1016/j.vaccine.2023.06.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/25/2023] [Accepted: 06/01/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND We aimed to estimate vaccine effectiveness (VE) against COVID-19 mortality, and to explore whether an increased risk of non-COVID-19 mortality exists in the weeks following a COVID-19 vaccine dose. METHODS National registries of causes of death, COVID-19 vaccination, specialized health care and long-term care reimbursements were linked by a unique person identifier using data from 1 January 2021 to 31 January 2022. We used Cox regression with calendar time as underlying time scale to, firstly, estimate VE against COVID-19 mortality after primary and first booster vaccination, per month since vaccination and, secondly, estimate risk of non-COVID-19 mortality in the 5 or 8 weeks following a first, second or first booster dose, adjusting for birth year, sex, medical risk group and country of origin. RESULTS VE against COVID-19 mortality was > 90 % for all age groups two months after completion of the primary series. VE gradually decreased thereafter, to around 80 % at 7-8 months post-primary series for most groups, and around 60 % for elderly receiving a high level of long-term care and for people aged 90+ years. Following a first booster dose, the VE increased to > 85 % in all groups. The risk of non-COVID-19 mortality was lower or similar in the 5 or 8 weeks following a first dose compared to no vaccination, as well as following a second dose compared to one dose and a booster compared to two doses, for all age and long-term care groups. CONCLUSION At the population level, COVID-19 vaccination greatly reduced the risk of COVID-19 mortality and no increased risk of death from other causes was observed.
Collapse
Affiliation(s)
- Brechje de Gier
- Center for Epidemiology and Surveillance of Infectious Diseases, National Institute for Public Health and the Environment, PO Box 1, 3720 BA Bilthoven, The Netherlands.
| | - Liselotte van Asten
- Center for Epidemiology and Surveillance of Infectious Diseases, National Institute for Public Health and the Environment, PO Box 1, 3720 BA Bilthoven, The Netherlands
| | - Tjarda M Boere
- Center for Epidemiology and Surveillance of Infectious Diseases, National Institute for Public Health and the Environment, PO Box 1, 3720 BA Bilthoven, The Netherlands
| | - Annika van Roon
- Center for Epidemiology and Surveillance of Infectious Diseases, National Institute for Public Health and the Environment, PO Box 1, 3720 BA Bilthoven, The Netherlands
| | - Caren van Roekel
- Center for Epidemiology and Surveillance of Infectious Diseases, National Institute for Public Health and the Environment, PO Box 1, 3720 BA Bilthoven, The Netherlands
| | - Joyce Pijpers
- Center for Epidemiology and Surveillance of Infectious Diseases, National Institute for Public Health and the Environment, PO Box 1, 3720 BA Bilthoven, The Netherlands
| | - C H Henri van Werkhoven
- Center for Epidemiology and Surveillance of Infectious Diseases, National Institute for Public Health and the Environment, PO Box 1, 3720 BA Bilthoven, The Netherlands; Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, P.O. Box 85500, 3508 GA Utrecht, The Netherlands
| | - Caroline van den Ende
- Center for Epidemiology and Surveillance of Infectious Diseases, National Institute for Public Health and the Environment, PO Box 1, 3720 BA Bilthoven, The Netherlands
| | - Susan J M Hahné
- Center for Epidemiology and Surveillance of Infectious Diseases, National Institute for Public Health and the Environment, PO Box 1, 3720 BA Bilthoven, The Netherlands
| | - Hester E de Melker
- Center for Epidemiology and Surveillance of Infectious Diseases, National Institute for Public Health and the Environment, PO Box 1, 3720 BA Bilthoven, The Netherlands
| | - Mirjam J Knol
- Center for Epidemiology and Surveillance of Infectious Diseases, National Institute for Public Health and the Environment, PO Box 1, 3720 BA Bilthoven, The Netherlands
| | - Susan van den Hof
- Center for Epidemiology and Surveillance of Infectious Diseases, National Institute for Public Health and the Environment, PO Box 1, 3720 BA Bilthoven, The Netherlands
| |
Collapse
|
148
|
Liu Y, Sánchez-Ovando S, Carolan L, Dowson L, Khvorov A, Hadiprodjo J, Tseng YY, Delahunty C, Khatami A, Macnish M, Dougherty S, Hagenauer M, Riley KE, Jadhav A, Harvey J, Kaiser M, Mathew S, Hodgson D, Leung V, Subbarao K, Cheng AC, Macartney K, Koirala A, Marshall H, Clark J, Blyth CC, Wark P, Kucharski AJ, Sullivan SG, Fox A. Comparative B cell and antibody responses induced by adenoviral vectored and mRNA vaccines against COVID-19. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.02.23290871. [PMID: 37333329 PMCID: PMC10275006 DOI: 10.1101/2023.06.02.23290871] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Both vector and mRNA vaccines were an important part of the response to the COVID-19 pandemic and may be required in future outbreaks and pandemics. However, adenoviral vectored (AdV) vaccines may be less immunogenic than mRNA vaccines against SARS-CoV-2. We assessed anti-spike and anti-vector immunity among infection-naïve Health Care Workers (HCW) following two doses of AdV (AZD1222) versus mRNA (BNT162b2) vaccine. 183 AdV and 274 mRNA vaccinees enrolled between April and October 2021. Median ages were 42 and 39 years, respectively. Blood was collected at least once, 10-48 days after vaccine dose 2. Surrogate virus neutralization test (sVNT) and spike binding antibody titres were a median of 4.2 and 2.2 times lower, respectively, for AdV compared to mRNA vaccinees (p<0.001). Median percentages of memory B cells that recognized fluorescent-tagged spike and RBD were 2.9 and 8.3 times lower, respectively for AdV compared to mRNA vaccinees. Titres of IgG reactive with human Adenovirus type 5 hexon protein rose a median of 2.2-fold after AdV vaccination but were not correlated with anti-spike antibody titres. Together the results show that mRNA induced substantially more sVNT antibody than AdV vaccine due to greater B cell expansion and targeting of the RBD. Pre-existing AdV vector cross-reactive antibodies were boosted following AdV vaccination but had no detectable effect on immunogenicity.
Collapse
Affiliation(s)
- Yi Liu
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Stephany Sánchez-Ovando
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Louise Carolan
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Leslie Dowson
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Arseniy Khvorov
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Jessica Hadiprodjo
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Yeu Yang Tseng
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Catherine Delahunty
- Immune Health Program, Hunter Medical Research Institute and School of Medicine and Public Health, University of Newcastle, Newcastle, Australia
| | - Ameneh Khatami
- The Children’s Hospital at Westmead; Sydney Children’s Hospital Network; National Centre for Immunisation Research and Surveillance, Sydney, Australia
- The University of Sydney; and National Centre for Immunisation Research and Surveillance, Sydney, Australia
| | - Marion Macnish
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Australia
| | - Sonia Dougherty
- Queensland Children’s Hospital, Children’s Health Queensland Hospital and Health Service; and University of Queensland, Brisbane, Australia
| | | | - Kathryn E. Riley
- Adelaide Medical School and Robinson Research Institute, The University of Adelaide, Adelaide, Australia
- Division of Paediatric Medicine, Women’s and Children’s Health Network, Adelaide, Australia
| | - Ajay Jadhav
- The Children’s Hospital at Westmead; Sydney Children’s Hospital Network; National Centre for Immunisation Research and Surveillance, Sydney, Australia
| | - Joanne Harvey
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Australia
| | - Marti Kaiser
- Alfred Health and Monash University, Melbourne, Australia
| | - Suja Mathew
- Adelaide Medical School and Robinson Research Institute, The University of Adelaide, Adelaide, Australia
- Division of Paediatric Medicine, Women’s and Children’s Health Network, Adelaide, Australia
| | - David Hodgson
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - Vivian Leung
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Kanta Subbarao
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Allen C. Cheng
- Alfred Health and Monash University, Melbourne, Australia
| | - Kristine Macartney
- The Children’s Hospital at Westmead; Sydney Children’s Hospital Network; National Centre for Immunisation Research and Surveillance, Sydney, Australia
- The University of Sydney; and National Centre for Immunisation Research and Surveillance, Sydney, Australia
| | - Archana Koirala
- The Children’s Hospital at Westmead; Sydney Children’s Hospital Network; National Centre for Immunisation Research and Surveillance, Sydney, Australia
- The University of Sydney; and National Centre for Immunisation Research and Surveillance, Sydney, Australia
| | - Helen Marshall
- Adelaide Medical School and Robinson Research Institute, The University of Adelaide, Adelaide, Australia
- Division of Paediatric Medicine, Women’s and Children’s Health Network, Adelaide, Australia
| | - Julia Clark
- Queensland Children’s Hospital, Children’s Health Queensland Hospital and Health Service; and University of Queensland, Brisbane, Australia
| | - Christopher C. Blyth
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Australia
- School of Medicine, University of Western Australia; Perth Children’s Hospital; and Department of Microbiology, PathWest Laboratory Medicine, QEII medical centre, Perth, Australia
| | - Peter Wark
- Immune Health Program, Hunter Medical Research Institute and School of Medicine and Public Health, University of Newcastle, Newcastle, Australia
| | - Adam J. Kucharski
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - Sheena G. Sullivan
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Department of Epidemiology, University of California, Los Angeles, USA
| | - Annette Fox
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| |
Collapse
|
149
|
Rossi M, Pessolano G, Gambaro G. What has vaccination against COVID-19 in CKD patients taught us? J Nephrol 2023; 36:1257-1266. [PMID: 37140817 PMCID: PMC10157569 DOI: 10.1007/s40620-023-01640-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2023] [Indexed: 05/05/2023]
Abstract
Effective vaccination strategies are of crucial importance to protecting patients who are vulnerable to infections, such as patients with chronic kidney disease. This is because the decreased efficiency of the immune system in chronic kidney disease impairs vaccine-induced immunisation. COVID-19 has prompted investigation of the immune response to SARS-CoV-2 vaccines in chronic kidney disease and in kidney transplant recipients in an effort to improve efficacy. The seroconversion rate after two vaccine doses is reduced, especially in kidney transplant recipients. Furthermore, although the seroconversion rate in chronic kidney disease patients is as high as in healthy subjects, anti-spike antibody titres are lower than in healthy vaccinated individuals, and these titres decrease rapidly. Although the vaccine-induced anti-spike antibody titre correlates with neutralising antibody levels and with protection against COVID-19, the protective prognostic significance of their titre is decreased due to the emergence of SARS-CoV-2 variants other than the Wuhan index virus against which the original vaccines were produced. Cellular immunity is also relevant, and because of cross-reactivity to the spike protein, epitopes of different viral variants confer protection against newly emerging variants of SARS-CoV-2. A multi-dose vaccination strategy is the most effective way to obtain a sufficient serological response. In kidney transplant recipients, a 5-week discontinuation period from antimetabolite drugs in concomitance with vaccine administration may also increase the vaccine's efficacy. The newly acquired knowledge obtained from COVID-19 vaccination is of general interest for the success of other vaccinations in chronic kidney disease patients.
Collapse
Affiliation(s)
- Mattia Rossi
- Division of Nephrology, Department of Medicine, University of Verona, Piazzale A. Stefani 1, 37126, Verona, Italy.
| | - Giuseppina Pessolano
- Division of Nephrology, Department of Medicine, University of Verona, Piazzale A. Stefani 1, 37126, Verona, Italy
| | - Giovanni Gambaro
- Division of Nephrology, Department of Medicine, University of Verona, Piazzale A. Stefani 1, 37126, Verona, Italy
| |
Collapse
|
150
|
Alnemari RF, Roublah FA, Bargawi AA. The Effect of COVID-19 Vaccines on Hospital Admission and Severity of Symptoms Among COVID-19 Patients in Saudi Arabia, 2021. Cureus 2023; 15:e41067. [PMID: 37519611 PMCID: PMC10375302 DOI: 10.7759/cureus.41067] [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] [Accepted: 06/28/2023] [Indexed: 08/01/2023] Open
Abstract
Introduction Following the World Health Organization (WHO) declaration of coronavirus disease 2019 (COVID-19) as a pandemic, Saudi Arabia took unpreceded precautions to prevent and control the spread of the severe acute respiratory syndrome coronavirus 2 (SARS‑CoV‑2) infection. It is one of the first countries in the world to grant the authorization to use the Pfizer-BioNTech vaccine. This study aimed to assess the effect of one dose of COVID-19 vaccines (Pfizer-BioNTech, Manhattan, New York City, and Oxford-AstraZeneca, Cambridge, United Kingdom) among the Saudi population regarding symptom severity, hospital admission rate, and death. Methods An observational retrospective cohort study was conducted using data from COVID-19 surveillance records at King Abdulaziz Medical City (KAMC), Saudi Arabia, from January to May 2021. All confirmed COVID-19 patients who had positive tests by reverse transcription polymerase chain reaction (RT-PCR) assay of a nasopharyngeal swab were included in the study. Patients diagnosed outside KAMC and cases below 18 years old were excluded from the study. The research was approved by King Abdullah International Medical Research Center (NRJ21J/303/12). Multivariable logistic regression was conducted to estimate the odds of hospitalization among vaccinated and unvaccinated patients. Results A total of 1058 cases were included in the analysis. Two hundred sixty-five (265; 25%) patients were vaccinated with one dose of either Pfizer-BioNTech or Oxford Astra-Zeneca, and 793 (75%) were unvaccinated. The median age was 34 (IQR 25-51), primarily Saudi (94.6%) and male (59.5%). The odds of being vaccinated (CI: 1.284-2.882, P 0.002) were 1.924 times greater for males than females. Young patients below 40 had 1.997 times higher odds (CI: 1.238-3.222, P 0.004) of being vaccinated than patients above 60. The hospital admission rate was low among both groups (12.9%); however, it was significantly lower among the vaccinated group (2.3%) as compared to the unvaccinated (16.5%). The results showed significant differences in symptom severity among the groups. For vaccinated, only one patient (0.4%) died, one patient was admitted to the ICU, and one patient (0.4%) was admitted to the hospital isolation ward. On the contrary, among the unvaccinated group, 19 patients (2.4%) died, 17 patients (2.1%) were admitted to the ICU, and 114 patients (14.4%) were admitted to the hospital isolation ward. Conclusion This study demonstrates that one dose of COVID-19 vaccines, either Pfizer-BioNTech or Oxford-AstraZeneca, reduced the probability of death by 2% and hospital admission by 15% before the spread of the Delta variant (B.1.617). For generalizable results, nationwide studies using national surveillance data are recommended to assess multiple doses efficacy on different variants of the SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Reem F Alnemari
- Preventive Medicine, Ministry of National Guard Health Affairs, King Abdullah International Medical Research Center, King Saud bin Abdul-Aziz University for Health Sciences, Jeddah, SAU
| | - Fawziah A Roublah
- Preventive Medicine, Ministry of National Guard Health Affairs, King Abdullah International Medical Research Center, King Saud bin Abdul-Aziz University for Health Sciences, Jeddah, SAU
| | - Amina A Bargawi
- Preventive Medicine, Ministry of National Guard Health Affairs, King Abdullah International Medical Research Center, King Saud bin Abdul-Aziz University for Health Sciences, Jeddah, SAU
| |
Collapse
|