151
|
Zhang M, Liang Y, Yu D, Du B, Cheng W, Li L, Yu Z, Luo S, Zhang Y, Wang H, Zhang X, Zhang W. A systematic review of Vaccine Breakthrough Infections by SARS-CoV-2 Delta Variant. Int J Biol Sci 2022; 18:889-900. [PMID: 35002532 PMCID: PMC8741840 DOI: 10.7150/ijbs.68973] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/02/2021] [Indexed: 12/11/2022] Open
Abstract
Vaccines are proving to be highly effective in controlling hospitalization and deaths associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, as shown by clinical trials and real-world evidence. However, a deadly second wave of coronavirus disease 2019 (COVID-19), infected by SARS-CoV-2 variants, especially the Delta (B.1.617.2) variant, with an increased number of post-vaccination breakthrough infections were reported in the world recently. Actually, Delta variant not only resulted in a severe surge of vaccine breakthrough infections which was accompanied with high viral load and transmissibility, but also challenged the development of effective vaccines. Therefore, the biological characteristics and epidemiological profile of Delta variant, the current status of Delta variant vaccine breakthrough infections and the mechanism of vaccine breakthrough infections were discussed in this article. In addition, the significant role of the Delta variant spike (S) protein in the mechanism of immune escape of SARS-CoV-2 was highlighted in this article. In particular, we further discussed key points on the future SARS-CoV-2 vaccine research and development, hoping to make a contribution to the early, accurate and rapid control of the COVID-19 epidemic.
Collapse
Affiliation(s)
- Mengxin Zhang
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| | - Ying Liang
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| | - Dongsheng Yu
- Department of Chinese Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Bang Du
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| | - Weyland Cheng
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| | - Lifeng Li
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| | - Zhidan Yu
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| | - Shuying Luo
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| | - Yaodong Zhang
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| | - Huanmin Wang
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| | - Xianwei Zhang
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| | - Wancun Zhang
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| |
Collapse
|
152
|
Joannes-Boyau O, Dahyot-Fizelier C. Lack of vaccination in ventilated patients for SARS-Cov-2 in France. Anaesth Crit Care Pain Med 2022; 41:101021. [PMID: 35124566 PMCID: PMC8800504 DOI: 10.1016/j.accpm.2022.101021] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 11/19/2022]
|
153
|
Angyal A, Longet S, Moore SC, Payne RP, Harding A, Tipton T, Rongkard P, Ali M, Hering LM, Meardon N, Austin J, Brown R, Skelly D, Gillson N, Dobson SL, Cross A, Sandhar G, Kilby JA, Tyerman JK, Nicols AR, Spegarova JS, Mehta H, Hornsby H, Whitham R, Conlon CP, Jeffery K, Goulder P, Frater J, Dold C, Pace M, Ogbe A, Brown H, Ansari MA, Adland E, Brown A, Chand M, Shields A, Matthews PC, Hopkins S, Hall V, James W, Rowland-Jones SL, Klenerman P, Dunachie S, Richter A, Duncan CJA, Barnes E, Carroll M, Turtle L, de Silva TI. T-cell and antibody responses to first BNT162b2 vaccine dose in previously infected and SARS-CoV-2-naive UK health-care workers: a multicentre prospective cohort study. THE LANCET. MICROBE 2022; 3:e21-e31. [PMID: 34778853 PMCID: PMC8577846 DOI: 10.1016/s2666-5247(21)00275-5] [Citation(s) in RCA: 100] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND Previous infection with SARS-CoV-2 affects the immune response to the first dose of the SARS-CoV-2 vaccine. We aimed to compare SARS-CoV-2-specific T-cell and antibody responses in health-care workers with and without previous SARS-CoV-2 infection following a single dose of the BNT162b2 (tozinameran; Pfizer-BioNTech) mRNA vaccine. METHODS We sampled health-care workers enrolled in the PITCH study across four hospital sites in the UK (Oxford, Liverpool, Newcastle, and Sheffield). All health-care workers aged 18 years or older consenting to participate in this prospective cohort study were included, with no exclusion criteria applied. Blood samples were collected where possible before vaccination and 28 (±7) days following one or two doses (given 3-4 weeks apart) of the BNT162b2 vaccine. Previous infection was determined by a documented SARS-CoV-2-positive RT-PCR result or the presence of positive anti-SARS-CoV-2 nucleocapsid antibodies. We measured spike-specific IgG antibodies and quantified T-cell responses by interferon-γ enzyme-linked immunospot assay in all participants where samples were available at the time of analysis, comparing SARS-CoV-2-naive individuals to those with previous infection. FINDINGS Between Dec 9, 2020, and Feb 9, 2021, 119 SARS-CoV-2-naive and 145 previously infected health-care workers received one dose, and 25 SARS-CoV-2-naive health-care workers received two doses, of the BNT162b2 vaccine. In previously infected health-care workers, the median time from previous infection to vaccination was 268 days (IQR 232-285). At 28 days (IQR 27-33) after a single dose, the spike-specific T-cell response measured in fresh peripheral blood mononuclear cells (PBMCs) was higher in previously infected (n=76) than in infection-naive (n=45) health-care workers (median 284 [IQR 150-461] vs 55 [IQR 24-132] spot-forming units [SFUs] per 106 PBMCs; p<0·0001). With cryopreserved PBMCs, the T-cell response in previously infected individuals (n=52) after one vaccine dose was equivalent to that of infection-naive individuals (n=19) after receiving two vaccine doses (median 152 [IQR 119-275] vs 162 [104-258] SFUs/106 PBMCs; p=1·00). Anti-spike IgG antibody responses following a single dose in 142 previously infected health-care workers (median 270 373 [IQR 203 461-535 188] antibody units [AU] per mL) were higher than in 111 infection-naive health-care workers following one dose (35 001 [17 099-55 341] AU/mL; p<0·0001) and higher than in 25 infection-naive individuals given two doses (180 904 [108 221-242 467] AU/mL; p<0·0001). INTERPRETATION A single dose of the BNT162b2 vaccine is likely to provide greater protection against SARS-CoV-2 infection in individuals with previous SARS-CoV-2 infection, than in SARS-CoV-2-naive individuals, including against variants of concern. Future studies should determine the additional benefit of a second dose on the magnitude and durability of immune responses in individuals vaccinated following infection, alongside evaluation of the impact of extending the interval between vaccine doses. FUNDING UK Department of Health and Social Care, and UK Coronavirus Immunology Consortium.
Collapse
Affiliation(s)
- Adrienn Angyal
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Stephanie Longet
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Shona C Moore
- Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Rebecca P Payne
- Translational and Clinical Research Institute Immunity and Inflammation Theme, Newcastle University, Newcastle, UK
| | - Adam Harding
- Sir William Dunn School of Pathology, Division of Medical Sciences, University of Oxford, Oxford, UK
| | - Tom Tipton
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Patpong Rongkard
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK,Centre For Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK,Mahidol-Oxford Tropical Medicine Research Unit, Bangkok, Thailand
| | - Mohammad Ali
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK,Centre For Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Luisa M Hering
- Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Naomi Meardon
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - James Austin
- Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Rebecca Brown
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Donal Skelly
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK,Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, UK,Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | | | - Sue L Dobson
- Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Andrew Cross
- Department of Infection and Tropical Medicine, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle, UK
| | - Gurjinder Sandhar
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Jonathan A Kilby
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Jessica K Tyerman
- Translational and Clinical Research Institute Immunity and Inflammation Theme, Newcastle University, Newcastle, UK
| | - Alexander R Nicols
- Translational and Clinical Research Institute Immunity and Inflammation Theme, Newcastle University, Newcastle, UK
| | - Jarmila S Spegarova
- Translational and Clinical Research Institute Immunity and Inflammation Theme, Newcastle University, Newcastle, UK
| | - Hema Mehta
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Hailey Hornsby
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Rachel Whitham
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | | | - Katie Jeffery
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK,Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Philip Goulder
- Peter Medawar Building for Pathogen Research, Department of Paediatrics, University of Oxford, Oxford, UK
| | - John Frater
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK,Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Christina Dold
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK,NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Matthew Pace
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Ane Ogbe
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Helen Brown
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - M Azim Ansari
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Emily Adland
- Peter Medawar Building for Pathogen Research, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Anthony Brown
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Meera Chand
- Public Health England, Colindale, London, UK
| | - Adrian Shields
- Institute of Cancer and Genomic Science, College of Medical and Dental Science, University of Birmingham, Birmingham, UK,University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Philippa C Matthews
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK,Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Susan Hopkins
- NIHR Health Protection Research Unit in Healthcare Associated Infection and Antimicrobial Resistance, University of Oxford, Oxford, UK,Public Health England, Colindale, London, UK,Faculty of Medicine, Department of Infectious Disease, Imperial College London, London, UK
| | - Victoria Hall
- NIHR Health Protection Research Unit in Healthcare Associated Infection and Antimicrobial Resistance, University of Oxford, Oxford, UK,Public Health England, Colindale, London, UK
| | - William James
- Sir William Dunn School of Pathology, Division of Medical Sciences, University of Oxford, Oxford, UK
| | - Sarah L Rowland-Jones
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK,NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK,Translational Gastroenterology Unit, University of Oxford, Oxford, UK,Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Susanna Dunachie
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK,Centre For Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK,Mahidol-Oxford Tropical Medicine Research Unit, Bangkok, Thailand,Oxford University Hospitals NHS Foundation Trust, Oxford, UK,Correspondence to: Prof Susanna Dunachie, Peter Medawar Building for Pathogen Research, Oxford OX1 3SY, UK
| | - Alex Richter
- Institute of Cancer and Genomic Science, College of Medical and Dental Science, University of Birmingham, Birmingham, UK,University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Christopher J A Duncan
- Translational and Clinical Research Institute Immunity and Inflammation Theme, Newcastle University, Newcastle, UK,Department of Infection and Tropical Medicine, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle, UK
| | - Eleanor Barnes
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK,NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK,Translational Gastroenterology Unit, University of Oxford, Oxford, UK,Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Miles Carroll
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Lance Turtle
- Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK,Tropical and Infectious Disease Unit, Liverpool University Hospitals NHS Foundation Trust, member of Liverpool Health Partners, Liverpool, UK
| | - Thushan I de Silva
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK,Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | | |
Collapse
|
154
|
Brüssow H, Zuber S. Can a combination of vaccination and face mask wearing contain the COVID-19 pandemic? Microb Biotechnol 2021; 15:721-737. [PMID: 34962710 PMCID: PMC8913850 DOI: 10.1111/1751-7915.13997] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 12/16/2021] [Indexed: 12/13/2022] Open
Abstract
The COVID‐19 pandemic is going into its third year with Europe again being the focus of major epidemic activity. The present review tries to answer the question whether one can come to grip with the pandemic by a combination of vaccinations and non‐pharmaceutical interventions (NPIs). Several COVID‐19 vaccines are of remarkable efficacy and achieve high protection rates against symptomatic disease, especially severe disease, but mathematical models suggest that the current vaccination coverage in many countries is insufficient to achieve pandemic control. NPIs are needed as complementary measures because recent research has also revealed the limits of vaccination alone. Here, we review the evidence for efficacy of face mask wearing in various settings. Overall pooled analysis showed significant reduction in COVID‐19 incidence with mask wearing, although heterogeneity between studies was substantial. Controlled trials of mask wearing are difficult to conduct, separating mask wearing effects in population studies from the impact of other NPIs is challenging and the efficacy of masks depend on mask material and mask fit. The combination of vaccination and mask wearing is potentially synergistic since vaccination protects so far well from disease development (the omicron variant is currently an unknown) but immunity from infection wanes over few months after vaccination. In comparison, masks interfere with the virus transmission process at a level of a physical barrier independent of coronavirus variant. Vaccination and masks are much less costly to apply than other NPI measures which are associated with high economic and social costs, but paradoxically both measures are the target of a vocal opposition by a sizable minority of the society. In parallel with biomedical research, we need more social science research into this opposition to guide political decisions on how to end the pandemic. The present review tries to answer the question whether one can control the pandemic by a combination of vaccinations and non‐pharmaceutical interventions. The combination of vaccination and mask wearing is potentially synergistic since vaccination protects so far well from disease while masks interfere with the virus transmission process as a physical barrier against any type of coronavirus variant.
Collapse
Affiliation(s)
- Harald Brüssow
- Laboratory of Gene Technology, Department of Biosystems, KU Leuven, Leuven, Belgium
| | - Sophie Zuber
- Institute of Food Safety and Analytical Science, Nestlé Research, Lausanne 26, 1000, Switzerland
| |
Collapse
|
155
|
Elliott P, Haw D, Wang H, Eales O, Walters CE, Ainslie KEC, Atchison C, Fronterre C, Diggle PJ, Page AJ, Trotter AJ, Prosolek SJ, Ashby D, Donnelly CA, Barclay W, Taylor G, Cooke G, Ward H, Darzi A, Riley S. Exponential growth, high prevalence of SARS-CoV-2, and vaccine effectiveness associated with the Delta variant. Science 2021; 374:eabl9551. [PMID: 34726481 PMCID: PMC10763627 DOI: 10.1126/science.abl9551] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 10/29/2021] [Indexed: 01/05/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections were rising during early summer 2021 in many countries as a result of the Delta variant. We assessed reverse transcription polymerase chain reaction swab positivity in the Real-time Assessment of Community Transmission–1 (REACT-1) study in England. During June and July 2021, we observed sustained exponential growth with an average doubling time of 25 days, driven by complete replacement of the Alpha variant by Delta and by high prevalence at younger, less-vaccinated ages. Prevalence among unvaccinated people [1.21% (95% credible interval 1.03%, 1.41%)] was three times that among double-vaccinated people [0.40% (95% credible interval 0.34%, 0.48%)]. However, after adjusting for age and other variables, vaccine effectiveness for double-vaccinated people was estimated at between ~50% and ~60% during this period in England. Increased social mixing in the presence of Delta had the potential to generate sustained growth in infections, even at high levels of vaccination.
Collapse
Affiliation(s)
- Paul Elliott
- School of Public Health, Imperial College London, London, UK
- Imperial College Healthcare NHS Trust, London, UK
- National Institute for Health Research Imperial Biomedical Research Centre, London, UK
- MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- Health Data Research UK London at Imperial College London, London, UK
- UK Dementia Research Institute Centre at Imperial, London, UK
| | - David Haw
- School of Public Health, Imperial College London, London, UK
- MRC Centre for Global Infectious Disease Analysis and Jameel Institute, Imperial College London, London, UK
| | - Haowei Wang
- School of Public Health, Imperial College London, London, UK
- MRC Centre for Global Infectious Disease Analysis and Jameel Institute, Imperial College London, London, UK
| | - Oliver Eales
- School of Public Health, Imperial College London, London, UK
- MRC Centre for Global Infectious Disease Analysis and Jameel Institute, Imperial College London, London, UK
| | - Caroline E. Walters
- School of Public Health, Imperial College London, London, UK
- MRC Centre for Global Infectious Disease Analysis and Jameel Institute, Imperial College London, London, UK
| | - Kylie E. C. Ainslie
- School of Public Health, Imperial College London, London, UK
- MRC Centre for Global Infectious Disease Analysis and Jameel Institute, Imperial College London, London, UK
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | | | - Claudio Fronterre
- CHICAS, Lancaster Medical School, Lancaster University, and Health Data Research UK, Lancaster, UK
| | - Peter J. Diggle
- CHICAS, Lancaster Medical School, Lancaster University, and Health Data Research UK, Lancaster, UK
| | | | | | | | - The COVID-19 Genomics UK (COG-UK) Consortium11‡
- School of Public Health, Imperial College London, London, UK
- Imperial College Healthcare NHS Trust, London, UK
- National Institute for Health Research Imperial Biomedical Research Centre, London, UK
- MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- Health Data Research UK London at Imperial College London, London, UK
- UK Dementia Research Institute Centre at Imperial, London, UK
- MRC Centre for Global Infectious Disease Analysis and Jameel Institute, Imperial College London, London, UK
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
- CHICAS, Lancaster Medical School, Lancaster University, and Health Data Research UK, Lancaster, UK
- Quadram Institute, Norwich, UK
- www.cogconsortium.uk
- Department of Statistics, University of Oxford, Oxford, UK
- Department of Infectious Disease, Imperial College London, London, UK
- Institute of Global Health Innovation at Imperial College London, London, UK
- Health Security Initiative, Flagship Pioneering UK Ltd., Bristol, UK
| | - Deborah Ashby
- School of Public Health, Imperial College London, London, UK
| | - Christl A. Donnelly
- School of Public Health, Imperial College London, London, UK
- MRC Centre for Global Infectious Disease Analysis and Jameel Institute, Imperial College London, London, UK
- Department of Statistics, University of Oxford, Oxford, UK
| | - Wendy Barclay
- Department of Infectious Disease, Imperial College London, London, UK
| | - Graham Taylor
- Department of Infectious Disease, Imperial College London, London, UK
| | - Graham Cooke
- Imperial College Healthcare NHS Trust, London, UK
- National Institute for Health Research Imperial Biomedical Research Centre, London, UK
- Department of Infectious Disease, Imperial College London, London, UK
| | - Helen Ward
- School of Public Health, Imperial College London, London, UK
- Imperial College Healthcare NHS Trust, London, UK
- National Institute for Health Research Imperial Biomedical Research Centre, London, UK
| | - Ara Darzi
- Imperial College Healthcare NHS Trust, London, UK
- National Institute for Health Research Imperial Biomedical Research Centre, London, UK
- Institute of Global Health Innovation at Imperial College London, London, UK
- Health Security Initiative, Flagship Pioneering UK Ltd., Bristol, UK
| | - Steven Riley
- School of Public Health, Imperial College London, London, UK
- MRC Centre for Global Infectious Disease Analysis and Jameel Institute, Imperial College London, London, UK
| |
Collapse
|
156
|
Cheng CJ, Lu CY, Chang YH, Sun Y, Chu HJ, Lee CY, Liu CH, Lin CH, Lu CJ, Li CY. Effectiveness of the WHO-Authorized COVID-19 Vaccines: A Rapid Review of Global Reports till 30 June 2021. Vaccines (Basel) 2021; 9:1489. [PMID: 34960235 PMCID: PMC8708265 DOI: 10.3390/vaccines9121489] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 12/01/2021] [Accepted: 12/13/2021] [Indexed: 01/06/2023] Open
Abstract
Large clinical trials have proven the efficacy of the COVID-19 vaccine, and the number of studies about the effectiveness rapidly grew in the first half of the year after mass vaccination was administrated globally. This rapid review aims to provide evidence syntheses as a means to complement the current evidence on the vaccine effectiveness (VE) against various outcomes in real-world settings. Databases (PubMed, EMBASE, and MedRxiv) were searched up to 30 June 2021, (PROSPERO ID: 266866). A total of 39 studies were included, covering over 15 million participants from 11 nations. Among the general population being fully vaccinated, the VE against symptomatic SARS-CoV-2 infection was estimated at 89-97%, 92% (95% CI, 78-97%), and 94% (95% CI, 86-97%) for BNT162b2, ChAdOx1, and mRNA-1273, respectively. As for the protective effects against B.1.617.2-related symptomatic infection, the VE was 88% (95% CI, 85.3-90.1%) by BNT162b2 and 67.0% (95% CI, 61.3-71.8%) by ChAdOx1 after full vaccination. This review revealed a consistently high effectiveness of certain vaccines among the general population in real-world settings. However, scarce data on the major variants of SARS-CoV-2 and the shortness of the study time may limit the conclusions to the mRNA vaccines and ChAdOx1.
Collapse
Affiliation(s)
- Chang-Jie Cheng
- Department of Neurology, En Chu Kong Hospital, New Taipei City 237, Taiwan; (C.-J.C.); (H.-J.C.); (C.-Y.L.); (C.-H.L.); (C.-H.L.); (C.-J.L.)
- Department of Neurology, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Chun-Yi Lu
- Division of Pediatric Infectious Diseases, Department of Pediatrics, National Taiwan University Hospital, Taipei 100, Taiwan;
- College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Ya-Hui Chang
- Department of Public Health, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan;
| | - Yu Sun
- Department of Neurology, En Chu Kong Hospital, New Taipei City 237, Taiwan; (C.-J.C.); (H.-J.C.); (C.-Y.L.); (C.-H.L.); (C.-H.L.); (C.-J.L.)
- Department of Neurology, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Hai-Jui Chu
- Department of Neurology, En Chu Kong Hospital, New Taipei City 237, Taiwan; (C.-J.C.); (H.-J.C.); (C.-Y.L.); (C.-H.L.); (C.-H.L.); (C.-J.L.)
- Department of Neurology, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Chun-Yu Lee
- Department of Neurology, En Chu Kong Hospital, New Taipei City 237, Taiwan; (C.-J.C.); (H.-J.C.); (C.-Y.L.); (C.-H.L.); (C.-H.L.); (C.-J.L.)
- Department of Neurology, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Chang-Hsiu Liu
- Department of Neurology, En Chu Kong Hospital, New Taipei City 237, Taiwan; (C.-J.C.); (H.-J.C.); (C.-Y.L.); (C.-H.L.); (C.-H.L.); (C.-J.L.)
| | - Cheng-Huai Lin
- Department of Neurology, En Chu Kong Hospital, New Taipei City 237, Taiwan; (C.-J.C.); (H.-J.C.); (C.-Y.L.); (C.-H.L.); (C.-H.L.); (C.-J.L.)
| | - Chien-Jung Lu
- Department of Neurology, En Chu Kong Hospital, New Taipei City 237, Taiwan; (C.-J.C.); (H.-J.C.); (C.-Y.L.); (C.-H.L.); (C.-H.L.); (C.-J.L.)
- Department of Neurology, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Chung-Yi Li
- Department of Public Health, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan;
- Department of Public Health, College of Public Health, China Medical University, Taichung 404, Taiwan
- Department of Healthcare Administration, College of Medical and Health Science, Asia University, Taichung 413, Taiwan
| |
Collapse
|
157
|
Al-Emran HM, Hasan MS, Ahasan Setu MA, Rahman MS, Alam ARU, Sarkar SL, Islam MT, Islam MR, Rahman MM, Islam OK, Jahid IK, Hossain MA. Genomic analysis of SARS-CoV-2 variants of concern identified from the ChAdOx1 nCoV-19 immunized patients from Southwest part of Bangladesh. J Infect Public Health 2021; 15:156-163. [PMID: 34952247 PMCID: PMC8688836 DOI: 10.1016/j.jiph.2021.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 11/25/2021] [Accepted: 12/01/2021] [Indexed: 11/21/2022] Open
Abstract
Background Bangladesh introduced ChAdOx1 nCoV-19 since February, 2021 and in six months, only a small population (12.8%) received either one or two dose of vaccination like other low-income countries. The COVID-19 infections were continued to roll all over the places although the information on genomic variations of SARS-CoV-2 between both immunized and unimmunized group was unavailable. The objective of this study was to compare the proportion of immune escaping variants between those groups. Methods A total of 4718 nasopharygeal samples were collected from March 1 until April 15, 2021, of which, 834 (18%) were SARS-CoV-2 positive. The minimum sample size was calculated as 108 who were randomly selected for telephone interview and provided consent. The prevalence of SARS-CoV-2 variants and disease severity among both immunized and unimmunized groups was measured. A total of 63 spike protein sequences and 14 whole-genome sequences were performed from both groups and phylogenetic reconstruction and mutation analysis were compared. Results A total of 40 respondents (37%, N = 108) received single-dose and 2 (2%) received both doses of ChAdOx1 nCoV-19 vaccine, which significantly reduce dry cough, loss of appetite and difficulties in breathing compared to none. There was no significant difference in hospitalization, duration of hospitalization or reduction of other symptoms like running nose, muscle pain, shortness of breathing or generalized weakness between immunized and unimmunized groups. Spike protein sequence assumed 21 (87.5%) B.1.351, one B.1.526 and two 20B variants in immunized group compared to 27 (69%) B.1.351, 5 (13%) B.1.1.7, 4 (10%) 20B, 2 B.1.526 and one B.1.427 variant in unimmunized group. Whole genome sequence analysis of 14 cases identified seven B.1.351 Beta V2, three B.1.1.7 Alpha V1, one B.1.526 Eta and the rest three 20B variants. Conclusion Our study observed that ChAdOx1 could not prevent the new infection or severe COVID-19 disease outcome with single dose while the infections were mostly caused by B.1.351 variants in Bangladesh.
Collapse
Affiliation(s)
- Hassan M Al-Emran
- Department of Biomedical Engineering, Jashore University of Science and Technology, Jashore 7408, Bangladesh; Genome Centre, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - Md Shazid Hasan
- Genome Centre, Jashore University of Science and Technology, Jashore 7408, Bangladesh; Department of Microbiology, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - Md Ali Ahasan Setu
- Genome Centre, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - M Shaminur Rahman
- Department of Microbiology, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - Asm Rubayet Ul Alam
- Genome Centre, Jashore University of Science and Technology, Jashore 7408, Bangladesh; Department of Microbiology, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - Shovon Lal Sarkar
- Genome Centre, Jashore University of Science and Technology, Jashore 7408, Bangladesh; Department of Microbiology, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - Md Tanvir Islam
- Genome Centre, Jashore University of Science and Technology, Jashore 7408, Bangladesh; Department of Microbiology, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - Mir Raihanul Islam
- Poverty, Health, and Nutrition Division, International Food Policy Research Institute, Dhaka, Bangladesh
| | - Mohammad Mahfuzur Rahman
- Department of Environmental Science and Technology, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - Ovinu Kibria Islam
- Genome Centre, Jashore University of Science and Technology, Jashore 7408, Bangladesh; Department of Microbiology, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - Iqbal Kabir Jahid
- Genome Centre, Jashore University of Science and Technology, Jashore 7408, Bangladesh; Department of Microbiology, Jashore University of Science and Technology, Jashore 7408, Bangladesh.
| | - M Anwar Hossain
- Genome Centre, Jashore University of Science and Technology, Jashore 7408, Bangladesh; Department of Microbiology, University of Dhaka, Dhaka 1000, Bangladesh; Vice-Chancellor, Jashore University of Science and Technology, Jashore 7408, Bangladesh.
| |
Collapse
|
158
|
Joshi G, Borah P, Thakur S, Sharma P, Mayank, Poduri R. Exploring the COVID-19 vaccine candidates against SARS-CoV-2 and its variants: where do we stand and where do we go? Hum Vaccin Immunother 2021; 17:4714-4740. [PMID: 34856868 PMCID: PMC8726002 DOI: 10.1080/21645515.2021.1995283] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/20/2021] [Accepted: 10/15/2021] [Indexed: 12/23/2022] Open
Abstract
As of September 2021, 117 COVID-19 vaccines are in clinical development, and 194 are in preclinical development as per the World Health Organization (WHO) published draft landscape. Among the 117 vaccines undergoing clinical trials, the major platforms include protein subunit; RNA; inactivated virus; viral vector, among others. So far, USFDA recognized to approve the Pfizer-BioNTech (Comirnaty) COVID-19 vaccine for its full use in individuals of 16 years of age and older. Though the approved vaccines are being manufactured at a tremendous pace, the wealthiest countries have about 28% of total vaccines despite possessing only 10.8% of the total world population, suggesting an inequity of vaccine distribution. The review comprehensively summarizes the history of vaccines, mainly focusing on vaccines for SARS-CoV-2. The review also connects relevant topics, including measurement of vaccines efficacy against SARS-CoV-2 and its variants, associated challenges, and limitations, as hurdles in global vaccination are also kept forth.
Collapse
Affiliation(s)
- Gaurav Joshi
- School of Pharmacy, Graphic Era Hill University, Dehradun, India
- Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences, Central University of Punjab, Bathinda, India
| | - Pobitra Borah
- School of Pharmacy, Graphic Era Hill University, Dehradun, India
| | - Shweta Thakur
- Laboratory of Genomic Instability and Diseases, Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneshwar, India
| | - Praveen Sharma
- Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences, Central University of Punjab, Bathinda, India
| | - Mayank
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Ramarao Poduri
- Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences, Central University of Punjab, Bathinda, India
| |
Collapse
|
159
|
Jamiruddin R, Haq A, Khondoker MU, Ali T, Ahmed F, Khandker SS, Jawad I, Hossain R, Ahmed S, Rahman SR, Mustafi M, Kaitsuka T, Mie M, Tomizawa K, Kobatake E, Haque M, Adnan N. Antibody response to the first dose of AZD1222 vaccine in COVID-19 convalescent and uninfected individuals in Bangladesh. Expert Rev Vaccines 2021; 20:1651-1660. [PMID: 34503369 PMCID: PMC8442763 DOI: 10.1080/14760584.2021.1977630] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 09/03/2021] [Indexed: 10/31/2022]
Abstract
BACKGROUND Vaccination with the Oxford-AstraZeneca COVID-19 vaccine (AZD1222) initially started in the UK and quickly implemented around the Globe, including Bangladesh. Up to date, more than nine million doses administrated to the Bangladeshi public. METHOD Herein, we studied the antibody response to the first dose of AZD1222 in 86 Bangladeshi individuals using in-house ELISA kits. Study subjects were categorized into two groups, convalescent and uninfected, based on prior infection history and SARS-CoV-2 nucleocapsid-IgG profiles. RESULTS All the convalescent individuals presented elevated spike-1-IgG compared to 90% of uninfected ones after the first dose. Day >28 post-vaccination, the convalescent group showed six times higher antibody titer than the uninfected ones. The most elevated antibody titers for the former and later group were found at Day 14 and Days >28 post-vaccination, respectively. The spike-1-IgA titer showed a similar pattern as spike-1-IgG, although in a low-titer. In contrast, the IgM titer did not show any significant change in either group. CONCLUSION High antibody titer in the convalescent group, signify the importance of the first dose among the uninfected group. This study advocates the integration of antibody tests in vaccination programs in the healthcare system for maximizing benefit.
Collapse
Affiliation(s)
- Raeed Jamiruddin
- Department of Pharmacy, Brac University, Dhaka, Bangladesh
- Gonoshasthaya-RNA Molecular Diagnostic & Research Center, Dhanmondi, Dhaka, Bangladesh
| | - Ahsanul Haq
- Gonoshasthaya-RNA Molecular Diagnostic & Research Center, Dhanmondi, Dhaka, Bangladesh
| | - Mohib Ullah Khondoker
- Gonoshasthaya-RNA Molecular Diagnostic & Research Center, Dhanmondi, Dhaka, Bangladesh
- Department of Community Medicine, Gonoshasthaya Samaj Vittik Medical College, Savar, Dhaka, Bangladesh
| | - Tamanna Ali
- Gonoshasthaya-RNA Molecular Diagnostic & Research Center, Dhanmondi, Dhaka, Bangladesh
| | - Firoz Ahmed
- Department of Microbiology, Jahangirnagar University, Savar, Dhaka, Bangladesh
| | - Shahad Saif Khandker
- Gonoshasthaya-RNA Molecular Diagnostic & Research Center, Dhanmondi, Dhaka, Bangladesh
| | - Irfan Jawad
- Department of Microbiology, Jahangirnagar University, Savar, Dhaka, Bangladesh
| | - Rubel Hossain
- Department of Microbiology, Gono Bishwabidyalay, Savar, Dhaka, Bangladesh
| | - Sohel Ahmed
- Department of Biochemistry and Molecular Biology, Jahangirnagar University, Savar, Dhaka, Bangladesh
| | | | - Mamun Mustafi
- Department of Community Medicine, Gonoshasthaya Samaj Vittik Medical College, Savar, Dhaka, Bangladesh
| | - Taku Kaitsuka
- Department of Pharmaceutical Sciences, School of Pharmacy, International University of Health and Welfare, Okawa, Fukuoka, Japan
| | - Masayasu Mie
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan
| | - Kazuhito Tomizawa
- Department of Molecular Physiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Eiry Kobatake
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan
| | - Mainul Haque
- The Unit of Pharmacology, Faculty of Medicine and Defence Health Universiti Pertahanan, Nasional Malaysia (National Defence University of Malaysia), Kem Perdana Sugai Besi, Kuala Lumpur, Malaysia
| | - Nihad Adnan
- Gonoshasthaya-RNA Molecular Diagnostic & Research Center, Dhanmondi, Dhaka, Bangladesh
- Department of Microbiology, Jahangirnagar University, Savar, Dhaka, Bangladesh
| |
Collapse
|
160
|
Marfe G, Perna S, Shukla AK. Effectiveness of COVID-19 vaccines and their challenges (Review). Exp Ther Med 2021; 22:1407. [PMID: 34676000 PMCID: PMC8524740 DOI: 10.3892/etm.2021.10843] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 08/24/2021] [Indexed: 12/13/2022] Open
Abstract
At the end of 2019, a new disease recognized such as severe acute respiratory syndrome (SARS), was reported in Wuhan, China. This disease was caused by an unknown SARS coronavirus 2 (SARS-CoV-2); a virus is characterized by high infectivity among humans. In some cases, this disease can be asymptomatic, while in other cases can induce flu-like symptoms or acute respiratory distress syndrome, pneumonia and death. For this reason, the World Health Organization and Public Health Emergency of International Concern declared a pandemic status in January 2020. Currently, numerous countries have been involved in the development of effective vaccines to protect humans against SARS-CoV-2 infection. The present review will discuss the four vaccines, AZD1222 (AstraZeneca or Vaxzevria), Janssen (Ad26.COV2.S), Moderna/mRNA-1273 and BioNTech/Fosun/Pfizer BNT162b1, that are currently in use worldwide to understand their efficacy, but also evaluate the difficulties and challenges of vaccine development. Although several questions should be addressed regarding these vaccines, the current review will examine the viral elements used in the coronavirus-19 vaccine that can play a crucial role in inducing a strong immune response, as well as the different adverse effects that they can cause to individuals.
Collapse
Affiliation(s)
- Gabriella Marfe
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania ‘Luigi Vanvitelli’, 81100 Caserta, Italy
| | - Stefania Perna
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania ‘Luigi Vanvitelli’, 81100 Caserta, Italy
| | - Arvind Kumar Shukla
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan, Gyeongsangnam-do 50612, Republic of Korea
- Inventra Medclin Biomedical Healthcare and Research Center, Katemanivli, Kalyan, Thane, Maharashtra 421306, India
| |
Collapse
|
161
|
Fendler A, Shepherd ST, Au L, Wilkinson KA, Wu M, Byrne F, Cerrone M, Schmitt AM, Joharatnam-Hogan N, Shum B, Tippu Z, Rzeniewicz K, Boos LA, Harvey R, Carlyle E, Edmonds K, Del Rosario L, Sarker S, Lingard K, Mangwende M, Holt L, Ahmod H, Korteweg J, Foley T, Bazin J, Gordon W, Barber T, Emslie-Henry A, Xie W, Gerard CL, Deng D, Wall EC, Agua-Doce A, Namjou S, Caidan S, Gavrielides M, MacRae JI, Kelly G, Peat K, Kelly D, Murra A, Kelly K, O’Flaherty M, Dowdie L, Ash N, Gronthoud F, Shea RL, Gardner G, Murray D, Kinnaird F, Cui W, Pascual J, Rodney S, Mencel J, Curtis O, Stephenson C, Robinson A, Oza B, Farag S, Leslie I, Rogiers A, Iyengar S, Ethell M, Messiou C, Cunningham D, Chau I, Starling N, Turner N, Welsh L, van As N, Jones RL, Droney J, Banerjee S, Tatham KC, O’Brien M, Harrington K, Bhide S, Okines A, Reid A, Young K, Furness AJ, Pickering L, Swanton C, Gandhi S, Gamblin S, Bauer DLV, Kassiotis G, Kumar S, Yousaf N, Jhanji S, Nicholson E, Howell M, Walker S, Wilkinson RJ, Larkin J, Turajlic S. Adaptive immunity and neutralizing antibodies against SARS-CoV-2 variants of concern following vaccination in patients with cancer: The CAPTURE study. NATURE CANCER 2021; 2:1321-1337. [PMID: 34950880 PMCID: PMC7612125 DOI: 10.1038/s43018-021-00274-w] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 09/17/2021] [Indexed: 12/13/2022]
Abstract
CAPTURE (NCT03226886) is a prospective cohort study of COVID-19 immunity in patients with cancer. Here we evaluated 585 patients following administration of two doses of BNT162b2 or AZD1222 vaccines, administered 12 weeks apart. Seroconversion rates after two doses were 85% and 59% in patients with solid and hematological malignancies, respectively. A lower proportion of patients had detectable neutralizing antibody titers (NAbT) against SARS-CoV-2 variants of concern (VOCs) vs wildtype (WT). Patients with hematological malignancies were more likely to have undetectable NAbT and had lower median NAbT vs solid cancers against both WT and VOCs. In comparison with individuals without cancer, patients with haematological, but not solid, malignancies had reduced NAb responses. Seroconversion showed poor concordance with NAbT against VOCs. Prior SARS-CoV-2 infection boosted NAb response including against VOCs, and anti-CD20 treatment was associated with undetectable NAbT. Vaccine-induced T-cell responses were detected in 80% of patients, and were comparable between vaccines or cancer types. Our results have implications for the management of cancer patients during the ongoing COVID-19 pandemic.
Collapse
Affiliation(s)
- Annika Fendler
- Cancer Dynamics Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Scott T.C. Shepherd
- Cancer Dynamics Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
- Skin and Renal Units, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Lewis Au
- Cancer Dynamics Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
- Skin and Renal Units, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Katalin A. Wilkinson
- Tuberculosis Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
- Wellcome Center for Infectious Disease Research in Africa, University of Cape Town, Observatory, Cape Town, Republic of South Africa
| | - Mary Wu
- Wellcome Center for Infectious Disease Research in Africa, University of Cape Town, Observatory, Cape Town, Republic of South Africa
| | - Fiona Byrne
- Cancer Dynamics Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Maddalena Cerrone
- Tuberculosis Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
- Department of Infectious Disease, Imperial College London, London, UK
| | - Andreas M. Schmitt
- Skin and Renal Units, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | | | - Benjamin Shum
- Cancer Dynamics Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
- Skin and Renal Units, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Zayd Tippu
- Skin and Renal Units, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Karolina Rzeniewicz
- Cancer Dynamics Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Laura Amanda Boos
- Skin and Renal Units, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Ruth Harvey
- Worldwide Influenza Centre, The Francis Crick Institute, London, NW1 1AT, UK
| | - Eleanor Carlyle
- Skin and Renal Units, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Kim Edmonds
- Skin and Renal Units, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Lyra Del Rosario
- Skin and Renal Units, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Sarah Sarker
- Skin and Renal Units, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Karla Lingard
- Skin and Renal Units, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Mary Mangwende
- Skin and Renal Units, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Lucy Holt
- Skin and Renal Units, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Hamid Ahmod
- Skin and Renal Units, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Justine Korteweg
- Skin and Renal Units, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Tara Foley
- Skin and Renal Units, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Jessica Bazin
- Haemato-oncology Unit, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - William Gordon
- Cancer Dynamics Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Taja Barber
- Cancer Dynamics Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Andrea Emslie-Henry
- Cancer Dynamics Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Wenyi Xie
- Cancer Dynamics Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Camille L. Gerard
- Cancer Dynamics Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Daqi Deng
- Cancer Dynamics Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Emma C. Wall
- University College London Hospitals NHS Foundation Trust Biomedical Research Centre, London, NW1 1AT, UK
- Structural Biology of Disease Processes Laboratory, The Francis Crick Institute, London, NW1 1AT, UK; Experimental Histopathology Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Ana Agua-Doce
- Flow Cytometry Scientific Technology Platform, The Francis Crick Institute, London, NW1 1AT, UK
| | - Sina Namjou
- Safety, Health & Sustainability, The Francis Crick Institute, London, NW1 1AT, UK
| | - Simon Caidan
- Safety, Health & Sustainability, The Francis Crick Institute, London, NW1 1AT, UK
| | - Mike Gavrielides
- Scientific Computing Scientific Technology Platform, The Francis Crick Institute, London, NW1 1AT, UK
| | - James I MacRae
- Metabolomics Scientific Technology Platform, The Francis Crick Institute, London, NW1 1AT, UK
| | - Gavin Kelly
- Department of Bioinformatics and Biostatistics, The Francis Crick Institute, London, UK
| | - Kema Peat
- Skin and Renal Units, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Denise Kelly
- Skin and Renal Units, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Aida Murra
- Skin and Renal Units, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Kayleigh Kelly
- Skin and Renal Units, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Molly O’Flaherty
- Skin and Renal Units, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Lauren Dowdie
- Skin and Renal Units, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Natalie Ash
- Skin and Renal Units, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Firza Gronthoud
- Department of Pathology, The Royal Marsden NHS Foundation Trust, London, NW1 1AT, UK
| | - Robyn L. Shea
- Department of Pathology, The Royal Marsden NHS Foundation Trust, London, NW1 1AT, UK
- Translational Cancer Biochemistry Laboratory, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Gail Gardner
- Department of Pathology, The Royal Marsden NHS Foundation Trust, London, NW1 1AT, UK
| | - Darren Murray
- Department of Pathology, The Royal Marsden NHS Foundation Trust, London, NW1 1AT, UK
| | - Fiona Kinnaird
- Clinical Trials Unit, The Royal Marsden NHS Foundation Trust, London, SM2 5PT, UK
| | - Wanyuan Cui
- Lung Unit, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Javier Pascual
- Breast Unit, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Simon Rodney
- Skin and Renal Units, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Justin Mencel
- Gastrointestinal Unit, The Royal Marsden NHS Foundation Trust, London and Surrey SM2 5PT
| | - Olivia Curtis
- Lung Unit, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Clemency Stephenson
- Haemato-oncology Unit, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Anna Robinson
- Haemato-oncology Unit, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Bhavna Oza
- Skin and Renal Units, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Sheima Farag
- Skin and Renal Units, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Isla Leslie
- Skin and Renal Units, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Aljosja Rogiers
- Skin and Renal Units, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Sunil Iyengar
- Haemato-oncology Unit, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Mark Ethell
- Haemato-oncology Unit, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Christina Messiou
- Department of Radiology, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - David Cunningham
- Gastrointestinal Unit, The Royal Marsden NHS Foundation Trust, London and Surrey SM2 5PT
| | - Ian Chau
- Gastrointestinal Unit, The Royal Marsden NHS Foundation Trust, London and Surrey SM2 5PT
| | - Naureen Starling
- Gastrointestinal Unit, The Royal Marsden NHS Foundation Trust, London and Surrey SM2 5PT
| | - Nicholas Turner
- Breast Unit, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Liam Welsh
- Neuro-oncology Unit, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Nicholas van As
- Clinical Oncology Unit, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Robin L. Jones
- Sarcoma Unit, The Royal Marsden NHS Foundation Trust and Institute of Cancer Research, London, SW3 6JJ, UK
| | - Joanne Droney
- Palliative Medicine, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Susana Banerjee
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Kate C. Tatham
- Anaesthetics, Perioperative Medicine and Pain Department, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Mary O’Brien
- Lung Unit, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Kevin Harrington
- Head and Neck, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
- Targeted Therapy Team, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Shreerang Bhide
- Head and Neck, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
- Targeted Therapy Team, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Alicia Okines
- Breast Unit, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
- Acute Oncology Service, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Alison Reid
- Uro-oncology unit, The Royal Marsden NHS Foundation Trust, Surrey, SM2 5PT
| | - Kate Young
- Skin and Renal Units, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Andrew J.S. Furness
- Skin and Renal Units, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Lisa Pickering
- Skin and Renal Units, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
- University College London Cancer Institute, London WC1E 6DD, UK
| | | | - Sonia Gandhi
- Neurodegeneration Biology Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
- UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG
| | - Steve Gamblin
- Structural Biology of Disease Processes Laboratory, The Francis Crick Institute, London, NW1 1AT, UK; Experimental Histopathology Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - David LV Bauer
- RNA Virus Replication Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - George Kassiotis
- Retroviral Immunology Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Sacheen Kumar
- Gastrointestinal Unit, The Royal Marsden NHS Foundation Trust, London and Surrey SM2 5PT
| | - Nadia Yousaf
- Lung Unit, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
- Acute Oncology Service, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Shaman Jhanji
- Anaesthetics, Perioperative Medicine and Pain Department, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Emma Nicholson
- Haemato-oncology Unit, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Michael Howell
- High Throughput Screening Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Susanna Walker
- Anaesthetics, Perioperative Medicine and Pain Department, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Robert J. Wilkinson
- Tuberculosis Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
- Wellcome Center for Infectious Disease Research in Africa, University of Cape Town, Observatory, Cape Town, Republic of South Africa
- Department of Infectious Disease, Imperial College London, London, UK
| | - James Larkin
- Skin and Renal Units, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Samra Turajlic
- Cancer Dynamics Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
- Skin and Renal Units, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| |
Collapse
|
162
|
Andersson O, Campos-Mercade P, Meier AN, Wengström E. Anticipation of COVID-19 vaccines reduces willingness to socially distance. JOURNAL OF HEALTH ECONOMICS 2021; 80:102530. [PMID: 34563830 PMCID: PMC8442531 DOI: 10.1016/j.jhealeco.2021.102530] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/31/2021] [Accepted: 09/01/2021] [Indexed: 05/05/2023]
Abstract
We investigate how the anticipation of COVID-19 vaccines affects voluntary social distancing. In a large-scale preregistered survey experiment with a representative sample, we study whether providing information about the safety, effectiveness, and availability of COVID-19 vaccines affects the willingness to comply with public health guidelines. We find that vaccine information reduces peoples' voluntary social distancing, adherence to hygiene guidelines, and their willingness to stay at home. Getting positive information on COVID-19 vaccines induces people to believe in a swifter return to normal life. The results indicate an important behavioral drawback of successful vaccine development: An increased focus on vaccines can lower compliance with public health guidelines and accelerate the spread of infectious disease. The results imply that, as vaccinations roll out and the end of a pandemic feels closer, policies aimed at increasing social distancing will be less effective, and stricter policies might be required.
Collapse
Affiliation(s)
- Ola Andersson
- Department of Economics, Uppsala University, Sweden; Uppsala Center for Fiscal Studies, Uppsala University, Sweden; Research Institute of Industrial Economics, Sweden.
| | | | - Armando N Meier
- Unisanté, University of Lausanne, Switzerland; Faculty of Business and Economics, University of Basel, Switzerland.
| | - Erik Wengström
- Department of Economics, Lund University, Sweden; Department of Finance and Economics, Hanken School of Economics, Finland.
| |
Collapse
|
163
|
Milota T, Strizova Z, Smetanova J, Sediva A. An immunologist's perspective on anti-COVID-19 vaccines. Curr Opin Allergy Clin Immunol 2021; 21:545-552. [PMID: 34545040 DOI: 10.1097/aci.0000000000000788] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
PURPOSE OF REVIEW Antisevere acute respiratory syndrome-corona virus 2 (SARS-CoV-2) vaccines may provide prompt, effective, and safe solution for the COVID-19 pandemic. Several vaccine candidates have been evaluated in randomized clinical trials (RCTs). Furthermore, data from observational studies mimicking real-life practice and studies on specific groups, such as pregnant women or immunocompromised patients who were excluded from RCTs, are currently available. The main aim of the review is to summarize and provide an immunologist's view on mechanism of action, efficacy and safety, and future challenges in vaccination against SARS-CoV-2. RECENT FINDINGS mRNA and recombinant viral vector-based vaccines have been approved for conditional use in Europe and the USA. They show robust humoral and cellular responses, high with efficacy in prevention of COVID-19 infection (66.9 95%) and favorable safety profile in RCTs. High efficacy of 80-92% was observed in real-life practice. A pilot study also confirmed good safety profile of the mRNA vaccines in pregnant women. Unlike in those with secondary immunodeficiencies where postvaccination responses did not occur, encouraging results were obtained in patients with inborn errors of immunity. SUMMARY Although both RCTs and observational studies suggest good efficacy and safety profiles of the vaccines, their long-term efficacy and safety are still being discussed. Despite the promising results, clinical evidence for specific groups such as children, pregnant and breastfeeding women, and immunocompromised patients, and for novel virus variants are lacking. VIDEO ABSTRACT http://links.lww.com/COAI/A21.
Collapse
Affiliation(s)
- Tomas Milota
- Department of Immunology, Second Faculty of Medicine, Charles University
- Department of Pediatric and Adult Rheumatology, Motol University Hospital, Prague, Czech Republic
| | - Zuzana Strizova
- Department of Immunology, Second Faculty of Medicine, Charles University
| | - Jitka Smetanova
- Department of Immunology, Second Faculty of Medicine, Charles University
| | - Anna Sediva
- Department of Immunology, Second Faculty of Medicine, Charles University
| |
Collapse
|
164
|
Pouwels KB, Pritchard E, Matthews PC, Stoesser N, Eyre DW, Vihta KD, House T, Hay J, Bell JI, Newton JN, Farrar J, Crook D, Cook D, Rourke E, Studley R, Peto TEA, Diamond I, Walker AS. Effect of Delta variant on viral burden and vaccine effectiveness against new SARS-CoV-2 infections in the UK. Nat Med 2021; 27:2127-2135. [PMID: 34650248 PMCID: PMC8674129 DOI: 10.1038/s41591-021-01548-7] [Citation(s) in RCA: 335] [Impact Index Per Article: 111.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 09/21/2021] [Indexed: 12/13/2022]
Abstract
The effectiveness of the BNT162b2 and ChAdOx1 vaccines against new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections requires continuous re-evaluation, given the increasingly dominant B.1.617.2 (Delta) variant. In this study, we investigated the effectiveness of these vaccines in a large, community-based survey of randomly selected households across the United Kingdom. We found that the effectiveness of BNT162b2 and ChAdOx1 against infections (new polymerase chain reaction (PCR)-positive cases) with symptoms or high viral burden is reduced with the B.1.617.2 variant (absolute difference of 10-13% for BNT162b2 and 16% for ChAdOx1) compared to the B.1.1.7 (Alpha) variant. The effectiveness of two doses remains at least as great as protection afforded by prior natural infection. The dynamics of immunity after second doses differed significantly between BNT162b2 and ChAdOx1, with greater initial effectiveness against new PCR-positive cases but faster declines in protection against high viral burden and symptomatic infection with BNT162b2. There was no evidence that effectiveness varied by dosing interval, but protection was higher in vaccinated individuals after a prior infection and in younger adults. With B.1.617.2, infections occurring after two vaccinations had similar peak viral burden as those in unvaccinated individuals. SARS-CoV-2 vaccination still reduces new infections, but effectiveness and attenuation of peak viral burden are reduced with B.1.617.2.
Collapse
Affiliation(s)
- Koen B Pouwels
- National Institute for Health Research Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance, University of Oxford, Oxford, UK.
- Health Economics Research Centre, Nuffield Department of Population Health, University of Oxford, Oxford, UK.
| | - Emma Pritchard
- National Institute for Health Research Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance, University of Oxford, Oxford, UK
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Philippa C Matthews
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Department of Infectious Diseases and Microbiology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK
- National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Nicole Stoesser
- National Institute for Health Research Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance, University of Oxford, Oxford, UK
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Department of Infectious Diseases and Microbiology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK
- National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - David W Eyre
- National Institute for Health Research Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance, University of Oxford, Oxford, UK
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Department of Infectious Diseases and Microbiology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK
- Big Data Institute, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Karina-Doris Vihta
- National Institute for Health Research Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance, University of Oxford, Oxford, UK
- Department of Engineering, University of Oxford, Oxford, UK
| | - Thomas House
- Department of Mathematics, University of Manchester, Manchester, UK
- IBM Research, Hartree Centre, Sci-Tech Daresbury, UK
| | - Jodie Hay
- Glasgow Lighthouse Laboratory, Glasgow, UK
- University of Glasgow, Glasgow, UK
| | - John I Bell
- Office of the Regius Professor of Medicine, University of Oxford, Oxford, UK
| | - John N Newton
- Health Improvement Directorate, Public Health England, London, UK
| | | | - Derrick Crook
- National Institute for Health Research Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance, University of Oxford, Oxford, UK
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Department of Infectious Diseases and Microbiology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK
- National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | | | | | | | - Tim E A Peto
- National Institute for Health Research Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance, University of Oxford, Oxford, UK
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Department of Infectious Diseases and Microbiology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK
- National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | | | - A Sarah Walker
- National Institute for Health Research Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance, University of Oxford, Oxford, UK
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Department of Infectious Diseases and Microbiology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK
- MRC Clinical Trials Unit at UCL, University College London, London, UK
| |
Collapse
|
165
|
A single dose, BCG-adjuvanted COVID-19 vaccine provides sterilising immunity against SARS-CoV-2 infection. NPJ Vaccines 2021; 6:143. [PMID: 34848711 PMCID: PMC8633321 DOI: 10.1038/s41541-021-00406-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 11/03/2021] [Indexed: 12/23/2022] Open
Abstract
Global control of COVID-19 requires broadly accessible vaccines that are effective against SARS-CoV-2 variants. In this report, we exploit the immunostimulatory properties of bacille Calmette-Guérin (BCG), the existing tuberculosis vaccine, to deliver a vaccination regimen with potent SARS-CoV-2-specific protective immunity. Combination of BCG with a stabilised, trimeric form of SARS-CoV-2 spike antigen promoted rapid development of virus-specific IgG antibodies in the blood of vaccinated mice, that was further augmented by the addition of alum. This vaccine formulation, BCG:CoVac, induced high-titre SARS-CoV-2 neutralising antibodies (NAbs) and Th1-biased cytokine release by vaccine-specific T cells, which correlated with the early emergence of T follicular helper cells in local lymph nodes and heightened levels of antigen-specific plasma B cells after vaccination. Vaccination of K18-hACE2 mice with a single dose of BCG:CoVac almost completely abrogated disease after SARS-CoV-2 challenge, with minimal inflammation and no detectable virus in the lungs of infected animals. Boosting BCG:CoVac-primed mice with a heterologous vaccine further increased SARS-CoV-2-specific antibody responses, which effectively neutralised B.1.1.7 and B.1.351 SARS-CoV-2 variants of concern. These findings demonstrate the potential for BCG-based vaccination to protect against major SARS-CoV-2 variants circulating globally.
Collapse
|
166
|
Prates-Syed WA, Chaves LCS, Crema KP, Vuitika L, Lira A, Côrtes N, Kersten V, Guimarães FEG, Sadraeian M, Barroso da Silva FL, Cabral-Marques O, Barbuto JAM, Russo M, Câmara NOS, Cabral-Miranda G. VLP-Based COVID-19 Vaccines: An Adaptable Technology against the Threat of New Variants. Vaccines (Basel) 2021; 9:1409. [PMID: 34960155 PMCID: PMC8708688 DOI: 10.3390/vaccines9121409] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/18/2021] [Accepted: 11/20/2021] [Indexed: 12/23/2022] Open
Abstract
Virus-like particles (VLPs) are a versatile, safe, and highly immunogenic vaccine platform. Recently, there are developmental vaccines targeting SARS-CoV-2, the causative agent of COVID-19. The COVID-19 pandemic affected humanity worldwide, bringing out incomputable human and financial losses. The race for better, more efficacious vaccines is happening almost simultaneously as the virus increasingly produces variants of concern (VOCs). The VOCs Alpha, Beta, Gamma, and Delta share common mutations mainly in the spike receptor-binding domain (RBD), demonstrating convergent evolution, associated with increased transmissibility and immune evasion. Thus, the identification and understanding of these mutations is crucial for the production of new, optimized vaccines. The use of a very flexible vaccine platform in COVID-19 vaccine development is an important feature that cannot be ignored. Incorporating the spike protein and its variations into VLP vaccines is a desirable strategy as the morphology and size of VLPs allows for better presentation of several different antigens. Furthermore, VLPs elicit robust humoral and cellular immune responses, which are safe, and have been studied not only against SARS-CoV-2 but against other coronaviruses as well. Here, we describe the recent advances and improvements in vaccine development using VLP technology.
Collapse
Affiliation(s)
- Wasim A. Prates-Syed
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo 05508000, SP, Brazil; (W.A.P.-S.); (K.P.C.); (L.V.); (A.L.); (N.C.); (V.K.); (O.C.-M.); (J.A.M.B.); (M.R.); (N.O.S.C.)
- Institute of Research and Education in Child Health (PENSI), São Paulo 01228200, SP, Brazil
| | - Lorena C. S. Chaves
- Department of Microbiology and Immunology, School of Medicine, Emory University, Claudia Nance Rollins Building, Atlanta, GA 30329, USA;
| | - Karin P. Crema
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo 05508000, SP, Brazil; (W.A.P.-S.); (K.P.C.); (L.V.); (A.L.); (N.C.); (V.K.); (O.C.-M.); (J.A.M.B.); (M.R.); (N.O.S.C.)
- Institute of Research and Education in Child Health (PENSI), São Paulo 01228200, SP, Brazil
| | - Larissa Vuitika
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo 05508000, SP, Brazil; (W.A.P.-S.); (K.P.C.); (L.V.); (A.L.); (N.C.); (V.K.); (O.C.-M.); (J.A.M.B.); (M.R.); (N.O.S.C.)
| | - Aline Lira
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo 05508000, SP, Brazil; (W.A.P.-S.); (K.P.C.); (L.V.); (A.L.); (N.C.); (V.K.); (O.C.-M.); (J.A.M.B.); (M.R.); (N.O.S.C.)
- Institute of Research and Education in Child Health (PENSI), São Paulo 01228200, SP, Brazil
| | - Nelson Côrtes
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo 05508000, SP, Brazil; (W.A.P.-S.); (K.P.C.); (L.V.); (A.L.); (N.C.); (V.K.); (O.C.-M.); (J.A.M.B.); (M.R.); (N.O.S.C.)
- Institute of Research and Education in Child Health (PENSI), São Paulo 01228200, SP, Brazil
| | - Victor Kersten
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo 05508000, SP, Brazil; (W.A.P.-S.); (K.P.C.); (L.V.); (A.L.); (N.C.); (V.K.); (O.C.-M.); (J.A.M.B.); (M.R.); (N.O.S.C.)
| | | | - Mohammad Sadraeian
- São Carlos Institute of Physics, IFSC-USP, São Carlos 13566590, SP, Brazil; (F.E.G.G.); (M.S.)
- Institute for Biomedical Materials & Devices (IBMD), Faculty of Science, University of Technology, Sydney, NSW 2007, Australia
| | - Fernando L. Barroso da Silva
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040903, SP, Brazil;
- Department of Chemical and Biomolecular Engeneering, North Carolina State University, Raleigh, NC 27695, USA
| | - Otávio Cabral-Marques
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo 05508000, SP, Brazil; (W.A.P.-S.); (K.P.C.); (L.V.); (A.L.); (N.C.); (V.K.); (O.C.-M.); (J.A.M.B.); (M.R.); (N.O.S.C.)
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508000, SP, Brazil
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Children’s Medical Center, Tehran 1419733151, Iran
| | - José A. M. Barbuto
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo 05508000, SP, Brazil; (W.A.P.-S.); (K.P.C.); (L.V.); (A.L.); (N.C.); (V.K.); (O.C.-M.); (J.A.M.B.); (M.R.); (N.O.S.C.)
- Laboratory of Medical Investigation in Pathogenesis and Targeted Therapy in Onco-Immuno-Hematology (LIM-31), Department of Hematology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo 0124690, SP, Brazil
| | - Momtchilo Russo
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo 05508000, SP, Brazil; (W.A.P.-S.); (K.P.C.); (L.V.); (A.L.); (N.C.); (V.K.); (O.C.-M.); (J.A.M.B.); (M.R.); (N.O.S.C.)
| | - Niels O. S. Câmara
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo 05508000, SP, Brazil; (W.A.P.-S.); (K.P.C.); (L.V.); (A.L.); (N.C.); (V.K.); (O.C.-M.); (J.A.M.B.); (M.R.); (N.O.S.C.)
| | - Gustavo Cabral-Miranda
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo 05508000, SP, Brazil; (W.A.P.-S.); (K.P.C.); (L.V.); (A.L.); (N.C.); (V.K.); (O.C.-M.); (J.A.M.B.); (M.R.); (N.O.S.C.)
- Institute of Research and Education in Child Health (PENSI), São Paulo 01228200, SP, Brazil
| |
Collapse
|
167
|
dos Santos Ferreira CE, Gómez-Dantés H, Junqueira Bellei NC, López E, Nogales Crespo KA, O’Ryan M, Villegas J. The Role of Serology Testing in the Context of Immunization Policies for COVID-19 in Latin American Countries. Viruses 2021; 13:2391. [PMID: 34960660 PMCID: PMC8706237 DOI: 10.3390/v13122391] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/18/2021] [Accepted: 11/18/2021] [Indexed: 12/16/2022] Open
Abstract
This review aims to explore the role and value of serology testing in the context of COVID-19 immunization policies in Latin American countries and the barriers and challenges to the adequate use and uptake of this tool. It builds on a review of the academic literature, evidence, and existing policies, and includes a multistage process of discussion and feedback by a group of five experts. Regional and country-level evidence and resources from five focus countries-Argentina, Brazil, Chile, Colombia, and Mexico-were collected and analyzed. This review contains an overview of (1) the impact of the SARS-CoV-2 pandemic, the variants of concern and current testing strategies, (2) the introduction of COVID-19 vaccination, (3) the potential use of serology testing to support immunization initiatives, (4) the current frameworks for the use of serology testing in the region, and (5) the barriers and challenges to implementing serology testing in the context of COVID-19 immunization policies, including a discussion on the potential actions required to address these barriers and facilitate the uptake of this strategy in the region. Stakeholders can use elements of this document to guide timely decision-making, raise awareness, and inspire further studies.
Collapse
Affiliation(s)
- Carlos E. dos Santos Ferreira
- Clinical Pathology, Hospital Israelita Albert Einstein, São Paulo 05652-900, Brazil;
- Microbiology Sector, Federal University of São Paulo’s Central Laboratory Activities, São Paulo 04088-002, Brazil
- Brazilian Society of Clinical Pathology and Laboratory Medicine, Rio de Janeiro 22220-040, Brazil
| | | | | | - Eduardo López
- Department of Medicine, Hospital de Niños Gutiérrez, Buenos Aires C1425-EFD, Argentina;
- Pediatric Infectious Diseases Program, Faculty of Medicine, University of Buenos Aires, Buenos Aires C1121-ABG, Argentina
- Pediatrics and Vaccinology, Faculty of Medicine, University of Salvador, Buenos Aires C1055-AAG, Argentina
| | | | - Miguel O’Ryan
- Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago de Chile 8380000, Chile;
- Millennium Institute of Immunology and Immunotherapy, University of Chile, Santiago de Chile 8331150, Chile
- Chilean Academy of Medicine, Santiago de Chile 6500445, Chile
| | | |
Collapse
|
168
|
Ghosh AK, Kaiser M, Molla MMA, Nafisa T, Yeasmin M, Ratul RH, Sharif MM, Akram A, Hosen N, Mamunur R, Amin MR, Islam A, Hoque ME, Landt O, Lytton SD. Molecular and Serological Characterization of the SARS-CoV-2 Delta Variant in Bangladesh in 2021. Viruses 2021; 13:v13112310. [PMID: 34835116 PMCID: PMC8623815 DOI: 10.3390/v13112310] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/09/2021] [Accepted: 11/12/2021] [Indexed: 12/13/2022] Open
Abstract
Novel SARS-CoV-2 variants are emerging at an alarming rate. The delta variant and other variants of concern (VoC) carry spike (S)-protein mutations, which have the potential to evade protective immunity, to trigger break-through infections after COVID-19 vaccination, and to propagate future waves of COVID-19 pandemic. To identify SARS CoV-2 variants in Bangladesh, patients who are RT-PCR-positive for COVID-19 infections in Dhaka were screened by a RT-PCR melting curve analysis for spike protein mutations. To assess the anti-SARS CoV-2 antibody responses, the levels of the anti-S -proteins IgA and IgG and the anti-N-protein IgG were measured by ELISA. Of a total of 36 RT-PCR positive samples (75%), 27 were identified as delta variants, with one carrying an additional Q677H mutation and two with single nucleotide substitutions at position 23029 (compared to Wuhan-Hu-1 reference NC 045512) in the genome sequence. Three (8.3%) were identified as beta variants, two (5.5%) were identified as alpha variants, three (8.3%) were identified as having a B.1.1.318 lineage, and one sample was identified as an eta variant (B.1.525) carrying an additional V687L mutation. The trend of higher viral load (lower Cp values) among delta variants than in the alpha and beta variants was of borderline statistical significance (p = 0.045). Prospective studies with larger Bangladeshi cohorts are warranted to confirm the emergence of S-protein mutations and their association with antibody response in natural infection and potential breakthrough in vaccinated subjects.
Collapse
Affiliation(s)
- Asish Kumar Ghosh
- Department of Virology, Dhaka Medical College Hospital, Dhaka 1000, Bangladesh; (A.K.G.); (R.H.R.); (M.M.S.); (M.R.A.)
| | - Marco Kaiser
- GenExpress Gesellschaft für Proteindesign GmbH, Eresburgstraße 22-23 D, 12103 Berlin, Germany;
| | - Md. Maruf Ahmed Molla
- National Institute of Laboratory Medicine and Referral Center, Sher E-Bangla Nagar, Dhaka 1207, Bangladesh; (M.M.A.M.); (T.N.); (M.Y.); (A.A.); (N.H.); (M.E.H.)
| | - Tasnim Nafisa
- National Institute of Laboratory Medicine and Referral Center, Sher E-Bangla Nagar, Dhaka 1207, Bangladesh; (M.M.A.M.); (T.N.); (M.Y.); (A.A.); (N.H.); (M.E.H.)
| | - Mahmuda Yeasmin
- National Institute of Laboratory Medicine and Referral Center, Sher E-Bangla Nagar, Dhaka 1207, Bangladesh; (M.M.A.M.); (T.N.); (M.Y.); (A.A.); (N.H.); (M.E.H.)
| | - Rifat Hossain Ratul
- Department of Virology, Dhaka Medical College Hospital, Dhaka 1000, Bangladesh; (A.K.G.); (R.H.R.); (M.M.S.); (M.R.A.)
| | - Md. Mohiuddin Sharif
- Department of Virology, Dhaka Medical College Hospital, Dhaka 1000, Bangladesh; (A.K.G.); (R.H.R.); (M.M.S.); (M.R.A.)
| | - Arifa Akram
- National Institute of Laboratory Medicine and Referral Center, Sher E-Bangla Nagar, Dhaka 1207, Bangladesh; (M.M.A.M.); (T.N.); (M.Y.); (A.A.); (N.H.); (M.E.H.)
| | - Nur Hosen
- National Institute of Laboratory Medicine and Referral Center, Sher E-Bangla Nagar, Dhaka 1207, Bangladesh; (M.M.A.M.); (T.N.); (M.Y.); (A.A.); (N.H.); (M.E.H.)
| | - Rashid Mamunur
- Bangladesh Institute Tropical Infectious Disease (BITID), Fouzderhat, Chittagong 4317, Bangladesh;
| | - Md. Robed Amin
- Department of Virology, Dhaka Medical College Hospital, Dhaka 1000, Bangladesh; (A.K.G.); (R.H.R.); (M.M.S.); (M.R.A.)
| | - Alimul Islam
- Department of Microbiology and Hygiene, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh;
| | - Md. Ehsanul Hoque
- National Institute of Laboratory Medicine and Referral Center, Sher E-Bangla Nagar, Dhaka 1207, Bangladesh; (M.M.A.M.); (T.N.); (M.Y.); (A.A.); (N.H.); (M.E.H.)
| | - Olfert Landt
- TIB Molbiol GmbH, Eresburgstraße 22-23, 12103 Berlin, Germany;
| | - Simon D. Lytton
- SeraDiaLogistics, 81545 Munich, Germany
- Correspondence: ; Tel.: +49-172-49-11169
| |
Collapse
|
169
|
Halperin DT, Hearst N, Hodgins S, Bailey RC, Klausner JD, Jackson H, Wamai RG, Ladapo JA, Over M, Baral S, Escandón K, Gandhi M. Revisiting COVID-19 policies: 10 evidence-based recommendations for where to go from here. BMC Public Health 2021; 21:2084. [PMID: 34774012 PMCID: PMC8590121 DOI: 10.1186/s12889-021-12082-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 10/22/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Strategies to control coronavirus 2019 disease (COVID-19) have often been based on preliminary and limited data and have tended to be slow to evolve as new evidence emerges. Yet knowledge about COVID-19 has grown exponentially, and the expanding rollout of vaccines presents further opportunity to reassess the response to the pandemic more broadly. MAIN TEXT We review the latest evidence concerning 10 key COVID-19 policy and strategic areas, specifically addressing: 1) the expansion of equitable vaccine distribution, 2) the need to ease restrictions as hospitalization and mortality rates eventually fall, 3) the advantages of emphasizing educational and harm reduction approaches over coercive and punitive measures, 4) the need to encourage outdoor activities, 5) the imperative to reopen schools, 6) the far-reaching and long-term economic and psychosocial consequences of sustained lockdowns, 7) the excessive focus on surface disinfection and other ineffective measures, 8) the importance of reassessing testing policies and practices, 9) the need for increasing access to outpatient therapies and prophylactics, and 10) the necessity to better prepare for future pandemics. CONCLUSIONS While remarkably effective vaccines have engendered great hope, some widely held assumptions underlying current policy approaches call for an evidence-based reassessment. COVID-19 will require ongoing mitigation for the foreseeable future as it transforms from a pandemic into an endemic infection, but maintaining a constant state of emergency is not viable. A more realistic public health approach is to adjust current mitigation goals to be more data-driven and to minimize unintended harms associated with unfocused or ineffective control efforts. Based on the latest evidence, we therefore present recommendations for refining 10 key policy areas, and for applying lessons learned from COVID-19 to prevent and prepare for future pandemics.
Collapse
Affiliation(s)
- Daniel T Halperin
- Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Norman Hearst
- Department of Family and Community Medicine, School of Medicine, University of California, San Francisco, CA, USA
| | - Stephen Hodgins
- School of Public Health, University of Alberta, Edmonton, AB, Canada
| | - Robert C Bailey
- School of Public Health, University of Illinois, Chicago, IL, USA
| | - Jeffrey D Klausner
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | - Richard G Wamai
- Integrated Initiative for Global Health, Northeastern University, Boston, MA, USA
- School of Public Health, University of Nairobi, Nairobi, Kenya
| | - Joseph A Ladapo
- Division of General Internal Medicine and Health Services Research, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Mead Over
- Center for Global Development, Washington, D.C, USA
| | - Stefan Baral
- Department of Epidemiology, Johns Hopkins School of Public Health, Baltimore, MD, USA
| | - Kevin Escandón
- School of Medicine, Universidad del Valle, Cali, Colombia.
- Department of Microbiology, Universidad del Valle, Grupo de Investigación en Virus Emergentes VIREM, Cali, Colombia.
| | - Monica Gandhi
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| |
Collapse
|
170
|
Kennedy-Shaffer L, Kahn R, Lipsitch M. Estimating Vaccine Efficacy Against Transmission via Effect on Viral Load. Epidemiology 2021; 32:820-828. [PMID: 34469363 PMCID: PMC8478108 DOI: 10.1097/ede.0000000000001415] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 08/23/2021] [Indexed: 12/23/2022]
Abstract
Determining policies to end the SARS-CoV-2 pandemic will require an understanding of the efficacy and effectiveness (hereafter, efficacy) of vaccines. Beyond the efficacy against severe disease and symptomatic and asymptomatic infection, understanding vaccine efficacy against virus transmission, including efficacy against transmission of different viral variants, will help model epidemic trajectory and determine appropriate control measures. Recent studies have proposed using random virologic testing in individual randomized controlled trials to improve estimation of vaccine efficacy against infection. We propose to further use the viral load measures from these tests to estimate efficacy against transmission. This estimation requires a model of the relationship between viral load and transmissibility and assumptions about the vaccine effect on transmission and the progress of the epidemic. We describe these key assumptions, potential violations of them, and solutions that can be implemented to mitigate these violations. Assessing these assumptions and implementing this random sampling, with viral load measures, will enable better estimation of the crucial measure of vaccine efficacy against transmission.
Collapse
Affiliation(s)
- Lee Kennedy-Shaffer
- From the Department of Mathematics and Statistics, Vassar College, Poughkeepsie, NY
| | - Rebecca Kahn
- Center for Communicable Disease Dynamics, Harvard T H Chan School of Public Health, Boston, MA
- Department of Epidemiology, Harvard T H Chan School of Public Health, Boston, MA
| | - Marc Lipsitch
- Center for Communicable Disease Dynamics, Harvard T H Chan School of Public Health, Boston, MA
- Department of Epidemiology, Harvard T H Chan School of Public Health, Boston, MA
| |
Collapse
|
171
|
Feng S, Phillips DJ, White T, Sayal H, Aley PK, Bibi S, Dold C, Fuskova M, Gilbert SC, Hirsch I, Humphries HE, Jepson B, Kelly EJ, Plested E, Shoemaker K, Thomas KM, Vekemans J, Villafana TL, Lambe T, Pollard AJ, Voysey M. Correlates of protection against symptomatic and asymptomatic SARS-CoV-2 infection. Nat Med 2021; 27:2032-2040. [PMID: 34588689 PMCID: PMC8604724 DOI: 10.1038/s41591-021-01540-1] [Citation(s) in RCA: 746] [Impact Index Per Article: 248.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 09/14/2021] [Indexed: 12/20/2022]
Abstract
The global supply of COVID-19 vaccines remains limited. An understanding of the immune response that is predictive of protection could facilitate rapid licensure of new vaccines. Data from a randomized efficacy trial of the ChAdOx1 nCoV-19 (AZD1222) vaccine in the United Kingdom was analyzed to determine the antibody levels associated with protection against SARS-CoV-2. Binding and neutralizing antibodies at 28 days after the second dose were measured in infected and noninfected vaccine recipients. Higher levels of all immune markers were correlated with a reduced risk of symptomatic infection. A vaccine efficacy of 80% against symptomatic infection with majority Alpha (B.1.1.7) variant of SARS-CoV-2 was achieved with 264 (95% CI: 108, 806) binding antibody units (BAU)/ml: and 506 (95% CI: 135, not computed (beyond data range) (NC)) BAU/ml for anti-spike and anti-RBD antibodies, and 26 (95% CI: NC, NC) international unit (IU)/ml and 247 (95% CI: 101, NC) normalized neutralization titers (NF50) for pseudovirus and live-virus neutralization, respectively. Immune markers were not correlated with asymptomatic infections at the 5% significance level. These data can be used to bridge to new populations using validated assays, and allow extrapolation of efficacy estimates to new COVID-19 vaccines.
Collapse
Affiliation(s)
- Shuo Feng
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Daniel J Phillips
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Thomas White
- Late-stage development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Homesh Sayal
- Late-stage development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Parvinder K Aley
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Sagida Bibi
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Christina Dold
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Michelle Fuskova
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Sarah C Gilbert
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Ian Hirsch
- Late-stage development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | | | - Brett Jepson
- Late-stage development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
- Cytel Inc., Cambridge, MA, USA
| | - Elizabeth J Kelly
- Microbial Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Emma Plested
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Kathryn Shoemaker
- Late-stage development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Kelly M Thomas
- National Infection Service, Public Health England, Salisbury, UK
| | - Johan Vekemans
- Late-stage development Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Tonya L Villafana
- Late-stage development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Teresa Lambe
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Centre, Oxford, UK
| | - Merryn Voysey
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK.
- NIHR Oxford Biomedical Centre, Oxford, UK.
| |
Collapse
|
172
|
Wei J, Matthews PC, Stoesser N, Maddox T, Lorenzi L, Studley R, Bell JI, Newton JN, Farrar J, Diamond I, Rourke E, Howarth A, Marsden BD, Hoosdally S, Jones EY, Stuart DI, Crook DW, Peto TEA, Pouwels KB, Walker AS, Eyre DW. Anti-spike antibody response to natural SARS-CoV-2 infection in the general population. Nat Commun 2021; 12:6250. [PMID: 34716320 PMCID: PMC8556331 DOI: 10.1038/s41467-021-26479-2] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 10/06/2021] [Indexed: 01/08/2023] Open
Abstract
Understanding the trajectory, duration, and determinants of antibody responses after SARS-CoV-2 infection can inform subsequent protection and risk of reinfection, however large-scale representative studies are limited. Here we estimated antibody response after SARS-CoV-2 infection in the general population using representative data from 7,256 United Kingdom COVID-19 infection survey participants who had positive swab SARS-CoV-2 PCR tests from 26-April-2020 to 14-June-2021. A latent class model classified 24% of participants as 'non-responders' not developing anti-spike antibodies, who were older, had higher SARS-CoV-2 cycle threshold values during infection (i.e. lower viral burden), and less frequently reported any symptoms. Among those who seroconverted, using Bayesian linear mixed models, the estimated anti-spike IgG peak level was 7.3-fold higher than the level previously associated with 50% protection against reinfection, with higher peak levels in older participants and those of non-white ethnicity. The estimated anti-spike IgG half-life was 184 days, being longer in females and those of white ethnicity. We estimated antibody levels associated with protection against reinfection likely last 1.5-2 years on average, with levels associated with protection from severe infection present for several years. These estimates could inform planning for vaccination booster strategies.
Collapse
Affiliation(s)
- Jia Wei
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Big Data Institute, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Philippa C Matthews
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Department of Infectious Diseases and Microbiology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK
| | - Nicole Stoesser
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Department of Infectious Diseases and Microbiology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK
- The National Institute for Health Research Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at the University of Oxford, Oxford, UK
- The National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | | | | | | | - John I Bell
- Office of the Regius Professor of Medicine, University of Oxford, Oxford, UK
| | - John N Newton
- Health Improvement Directorate, Public Health England, London, UK
| | | | | | | | - Alison Howarth
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- The National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Brian D Marsden
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Sarah Hoosdally
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - E Yvonne Jones
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - David I Stuart
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Derrick W Crook
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Department of Infectious Diseases and Microbiology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK
- The National Institute for Health Research Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at the University of Oxford, Oxford, UK
- The National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Tim E A Peto
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Department of Infectious Diseases and Microbiology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK
- The National Institute for Health Research Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at the University of Oxford, Oxford, UK
- The National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Koen B Pouwels
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Big Data Institute, Nuffield Department of Population Health, University of Oxford, Oxford, UK
- The National Institute for Health Research Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at the University of Oxford, Oxford, UK
- Health Economics Research Centre, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - A Sarah Walker
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Big Data Institute, Nuffield Department of Population Health, University of Oxford, Oxford, UK
- The National Institute for Health Research Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at the University of Oxford, Oxford, UK
- MRC Clinical Trials Unit at UCL, UCL, London, UK
| | - David W Eyre
- Big Data Institute, Nuffield Department of Population Health, University of Oxford, Oxford, UK.
- Department of Infectious Diseases and Microbiology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK.
- The National Institute for Health Research Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at the University of Oxford, Oxford, UK.
- The National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, UK.
| |
Collapse
|
173
|
Estella Á, Cantón ML, Muñoz L, Higueras IR, Recuerda Núñez M, Tejero Aranguren J, Zaya B, Gómez C, Amaya R, Hurtado Martinez Á, del Valle Odero Bernal M, De la Fuente C, Alados JC, Garnacho-Montero J. Vaccinated Patients Admitted in ICU with Severe Pneumonia Due to SARS-CoV-2: A Multicenter Pilot Study. J Pers Med 2021; 11:1086. [PMID: 34834437 PMCID: PMC8625038 DOI: 10.3390/jpm11111086] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/15/2021] [Accepted: 10/22/2021] [Indexed: 11/16/2022] Open
Abstract
Background: The aim of this study was to analyze the percentage of patients admitted to the ICU having received the vaccine against COVID-19, to describe the clinical profile of vaccinated patients admitted to the ICU, and to assess the humoral immune response to vaccination. Methods: In this multicenter prospective descriptive cohort study, consecutive critically ill patients with confirmed SARS-CoV-2 pneumonia who received at least one dose of the SARS-CoV-2 vaccine were included. The time of study was from 1 July to 10 August of 2021. Results: Of the 94 consecutive patients from seven Andalusian ICUs admitted during the time of study, 50 (53.2%) received at least one dose of anti SARS-CoV-2 vaccine. No patient was admitted having previously had SARS-CoV-2 infection. The B.1.617.2 (Delta) variant was the most frequently identified, in 80.76% of cases. Patients with a complete vaccination with non-optimal antibody levels were immunocompromised. Fifteen patients were admitted to the ICU with Acute Respiratory Distress Syndrome (ARDS) without having completed their vaccination; the clinical profile was younger and with less comorbidities compared to patients with full vaccination. There were no differences in severity of ARDS. Conclusions: Most of the patients who were admitted to the ICU having received a dose of the vaccine were not optimally vaccinated; fully vaccinated patients who did not obtain optimal serum antibody levels were patients considered immunocompromised.
Collapse
Affiliation(s)
- Ángel Estella
- Intensive Care Unit, Medicine Department University of Cadiz, University Hospital of Jerez, INiBICA, 11001 Jerez, Spain
| | - Mª Luisa Cantón
- Intensive Care Unit, University Hospital Virgen Macarena, 41013 Sevilla, Spain; (M.L.C.); (J.G.-M.)
| | - Laura Muñoz
- Intensive Care Unit, Hospital Costa del Sol de Marbella, 29001 Málaga, Spain; (L.M.); (B.Z.); (C.G.)
| | | | - María Recuerda Núñez
- Intensive Care Unit, University Hospital of Jerez, INiBICA, 11001 Jerez, Spain; (M.R.N.); (M.d.V.O.B.); (J.C.A.)
| | | | - Benito Zaya
- Intensive Care Unit, Hospital Costa del Sol de Marbella, 29001 Málaga, Spain; (L.M.); (B.Z.); (C.G.)
| | - Carmen Gómez
- Intensive Care Unit, Hospital Costa del Sol de Marbella, 29001 Málaga, Spain; (L.M.); (B.Z.); (C.G.)
| | - Rosario Amaya
- Intensive Care Unit, University Hospital Virgen del Rocío, 41013 Sevilla, Spain;
| | | | - María del Valle Odero Bernal
- Intensive Care Unit, University Hospital of Jerez, INiBICA, 11001 Jerez, Spain; (M.R.N.); (M.d.V.O.B.); (J.C.A.)
| | | | - Juan Carlos Alados
- Intensive Care Unit, University Hospital of Jerez, INiBICA, 11001 Jerez, Spain; (M.R.N.); (M.d.V.O.B.); (J.C.A.)
| | - Jose Garnacho-Montero
- Intensive Care Unit, University Hospital Virgen Macarena, 41013 Sevilla, Spain; (M.L.C.); (J.G.-M.)
| | | |
Collapse
|
174
|
Ramesh S, Govindarajulu M, Parise RS, Neel L, Shankar T, Patel S, Lowery P, Smith F, Dhanasekaran M, Moore T. Emerging SARS-CoV-2 Variants: A Review of Its Mutations, Its Implications and Vaccine Efficacy. Vaccines (Basel) 2021; 9:1195. [PMID: 34696303 PMCID: PMC8537675 DOI: 10.3390/vaccines9101195] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/26/2021] [Accepted: 10/08/2021] [Indexed: 12/21/2022] Open
Abstract
The widespread increase in multiple severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) variants is causing a significant health concern in the United States and worldwide. These variants exhibit increased transmissibility, cause more severe disease, exhibit evasive immune properties, impair neutralization by antibodies from vaccinated individuals or convalescence sera, and reinfection. The Centers for Disease Control and Prevention (CDC) has classified SARS-CoV-2 variants into variants of interest, variants of concern, and variants of high consequence. Currently, four variants of concern (B.1.1.7, B.1.351, P.1, and B.1.617.2) and several variants of interests (B.1.526, B.1.525, and P.2) are characterized and are essential for close monitoring. In this review, we discuss the different SARS-CoV-2 variants, emphasizing variants of concern circulating the world and highlight the various mutations and how these mutations affect the characteristics of the virus. In addition, we discuss the most common vaccines and the various studies concerning the efficacy of these vaccines against different variants of concern.
Collapse
Affiliation(s)
- Sindhu Ramesh
- Department of Drug Discovery and Development, Auburn University Harrison School of Pharmacy, Auburn, AL 36849, USA; (S.R.); (M.G.); (R.S.P.); (L.N.); (P.L.); (F.S.); (M.D.)
| | - Manoj Govindarajulu
- Department of Drug Discovery and Development, Auburn University Harrison School of Pharmacy, Auburn, AL 36849, USA; (S.R.); (M.G.); (R.S.P.); (L.N.); (P.L.); (F.S.); (M.D.)
| | - Rachel S. Parise
- Department of Drug Discovery and Development, Auburn University Harrison School of Pharmacy, Auburn, AL 36849, USA; (S.R.); (M.G.); (R.S.P.); (L.N.); (P.L.); (F.S.); (M.D.)
| | - Logan Neel
- Department of Drug Discovery and Development, Auburn University Harrison School of Pharmacy, Auburn, AL 36849, USA; (S.R.); (M.G.); (R.S.P.); (L.N.); (P.L.); (F.S.); (M.D.)
| | - Tharanath Shankar
- Department of Internal Medicine, Ramaiah Medical College and Hospital, Bengaluru 560054, Karnataka, India;
| | - Shriya Patel
- Department of Neuroscience, Middlebury College, Middlebury, VT 05753, USA;
| | - Payton Lowery
- Department of Drug Discovery and Development, Auburn University Harrison School of Pharmacy, Auburn, AL 36849, USA; (S.R.); (M.G.); (R.S.P.); (L.N.); (P.L.); (F.S.); (M.D.)
| | - Forrest Smith
- Department of Drug Discovery and Development, Auburn University Harrison School of Pharmacy, Auburn, AL 36849, USA; (S.R.); (M.G.); (R.S.P.); (L.N.); (P.L.); (F.S.); (M.D.)
| | - Muralikrishnan Dhanasekaran
- Department of Drug Discovery and Development, Auburn University Harrison School of Pharmacy, Auburn, AL 36849, USA; (S.R.); (M.G.); (R.S.P.); (L.N.); (P.L.); (F.S.); (M.D.)
| | - Timothy Moore
- Department of Drug Discovery and Development, Auburn University Harrison School of Pharmacy, Auburn, AL 36849, USA; (S.R.); (M.G.); (R.S.P.); (L.N.); (P.L.); (F.S.); (M.D.)
| |
Collapse
|
175
|
Rovida F, Cassaniti I, Paolucci S, Percivalle E, Sarasini A, Piralla A, Giardina F, Sammartino JC, Ferrari A, Bergami F, Muzzi A, Novelli V, Meloni A, Cutti S, Grugnetti AM, Grugnetti G, Rona C, Daglio M, Marena C, Triarico A, Lilleri D, Baldanti F. SARS-CoV-2 vaccine breakthrough infections with the alpha variant are asymptomatic or mildly symptomatic among health care workers. Nat Commun 2021; 12:6032. [PMID: 34654808 PMCID: PMC8521593 DOI: 10.1038/s41467-021-26154-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 09/17/2021] [Indexed: 11/09/2022] Open
Abstract
Vaccine breakthrough SARS-CoV-2 infection has been monitored in 3720 healthcare workers receiving 2 doses of BNT162b2. SARS-CoV-2 infection is detected in 33 subjects, with a 100-day cumulative incidence of 0.93%. Vaccine protection against acquisition of SARS-CoV-2 infection is 83% (95%CI: 58-93%) in the overall population and 93% (95%CI: 69-99%) in SARS-CoV-2-experienced subjects, when compared with a non-vaccinated control group from the same Institution, in which SARS-CoV-2 infection occurs in 20/346 subjects (100-day cumulative incidence: 5.78%). The infection is symptomatic in 16 (48%) vaccinated subjects vs 17 (85%) controls (p = 0.01). All analyzed patients, in whom the amount of viral RNA was sufficient for genome sequencing, results infected by the alpha variant. Antibody and T-cell responses are not reduced in subjects with breakthrough infection. Evidence of virus transmission, determined by contact tracing, is observed in two (6.1%) cases. This real-world data support the protective effect of BNT162b2 vaccine. A triple antigenic exposure, such as two-dose vaccine schedule in experienced subjects, may confer a higher protection.
Collapse
Affiliation(s)
- Francesca Rovida
- Molecular Virology Unit, Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Irene Cassaniti
- Molecular Virology Unit, Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Stefania Paolucci
- Molecular Virology Unit, Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Elena Percivalle
- Molecular Virology Unit, Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Antonella Sarasini
- Molecular Virology Unit, Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Antonio Piralla
- Molecular Virology Unit, Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Federica Giardina
- Molecular Virology Unit, Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Josè Camilla Sammartino
- Molecular Virology Unit, Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Alessandro Ferrari
- Molecular Virology Unit, Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Federica Bergami
- Molecular Virology Unit, Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Alba Muzzi
- Medical Direction, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Viola Novelli
- Medical Direction, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Alessandro Meloni
- Medical Direction, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Public Health, Experimental and Forensic Medicine, Section of Hygiene, University of Pavia, Pavia, Italy
| | - Sara Cutti
- Medical Direction, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Anna Maria Grugnetti
- Health Professions Direction, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Giuseppina Grugnetti
- Health Professions Direction, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Claudia Rona
- Medical Direction, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Marinella Daglio
- Medical Direction, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Carlo Marena
- Medical Direction, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Antonio Triarico
- Direzione Sanitaria, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Daniele Lilleri
- Molecular Virology Unit, Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.
| | - Fausto Baldanti
- Molecular Virology Unit, Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
176
|
Sachak-Patwa R, Byrne HM, Dyson L, Thompson RN. The risk of SARS-CoV-2 outbreaks in low prevalence settings following the removal of travel restrictions. COMMUNICATIONS MEDICINE 2021; 1:39. [PMID: 35602220 PMCID: PMC9053223 DOI: 10.1038/s43856-021-00038-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 09/03/2021] [Indexed: 12/23/2022] Open
Abstract
Background Countries around the world have introduced travel restrictions to reduce SARS-CoV-2 transmission. As vaccines are gradually rolled out, attention has turned to when travel restrictions and other non-pharmaceutical interventions (NPIs) can be relaxed. Methods Using SARS-CoV-2 as a case study, we develop a mathematical branching process model to assess the risk that, following the removal of NPIs, cases arriving in low prevalence settings initiate a local outbreak. Our model accounts for changes in background population immunity due to vaccination. We consider two locations with low prevalence in which the vaccine rollout has progressed quickly – specifically, the Isle of Man (a British crown dependency in the Irish Sea) and the country of Israel. Results We show that the outbreak risk is unlikely to be eliminated completely when travel restrictions and other NPIs are removed. This general result is the most important finding of this study, rather than exact quantitative outbreak risk estimates in different locations. It holds even once vaccine programmes are completed. Key factors underlying this result are the potential for transmission even following vaccination, incomplete vaccine uptake, and the recent emergence of SARS-CoV-2 variants with increased transmissibility. Conclusions Combined, the factors described above suggest that, when travel restrictions are relaxed, it may still be necessary to implement surveillance of incoming passengers to identify infected individuals quickly. This measure, as well as tracing and testing (and/or isolating) contacts of detected infected passengers, remains useful to suppress potential outbreaks while global case numbers are high. The effectiveness of public health measures against COVID-19 has varied between countries, with some experiencing many infections and others containing transmission successfully. As vaccines are deployed, an important challenge is deciding when to relax measures. Here, we consider locations with few cases, and investigate whether vaccination can ever eliminate the risk of COVID-19 outbreaks completely, allowing measures to be removed risk-free. Using a mathematical model, we demonstrate that there is still a risk that imported cases initiate outbreaks when measures are removed, even if most of the population is fully vaccinated. This highlights the need for continued vigilance in low prevalence settings to prevent imported cases leading to local transmission. Until case numbers are reduced globally, so that SARS-CoV-2 spread between countries is less likely, the risk of outbreaks in low prevalence settings will remain. Sachak-Patwa et al. estimate the risk of SARS-CoV-2 outbreaks in low prevalence settings following the removal of travel restrictions and other non-pharmaceutical interventions, with the Isle of Man and Israel as case studies. Using a branching process mathematical model, the authors show that even after a large proportion of the population is vaccinated, there remains a risk of local outbreaks from imported cases.
Collapse
|
177
|
González S, Olszevicki S, Salazar M, Calabria A, Regairaz L, Marín L, Campos P, Varela T, Martínez VVG, Ceriani L, Garcia E, Kreplak N, Pifano M, Estenssoro E, Marsico F. Effectiveness of the first component of Gam-COVID-Vac (Sputnik V) on reduction of SARS-CoV-2 confirmed infections, hospitalisations and mortality in patients aged 60-79: a retrospective cohort study in Argentina. EClinicalMedicine 2021; 40:101126. [PMID: 34541480 PMCID: PMC8435263 DOI: 10.1016/j.eclinm.2021.101126] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND A first-dose of various vaccines provides acceptable protection against infections by SARS-CoV-2 and evolution to the most severe forms of COVID-19. The recombinant adenovirus (rAd)-based vaccine, Gam-COVID-Vac (Sputnik V), was proven efficacious but information about effectiveness in the real-world setting is lacking. The aim of our study was to investigate the association between the rollout of the first component (rAd26) of Gam-COVID-Vac and PCR-positive tests, hospitalisations and deaths. METHODS We conducted a retrospective cohort study which analyzed individuals aged 60-79 who self-registered in the online vaccination system of the Province of Buenos Aires, Argentina, from December 29, 2020 to March 21, 2021. Exclusion criteria were having a previous positive RT-PCR or antigen tests for SARS-CoV-2, having received other vaccines, or two doses of any vaccine.Proportions of new laboratory-confirmed SARS-CoV-2 infections, hospitalisations and deaths until 83 days of vaccination were compared between vaccinated and unvaccinated subjects. Vaccine effectiveness for the three outcomes was calculated as (1-OR) × 100. Kaplan-Meier cumulative incidence curves were constructed. FINDINGS During the study period 415995 registered subjects received the first component of Gam-COVID-Vac; 40387 belonged to the 60-79 age group, and were compared to 38978 unvaccinated. Vaccine effectiveness for preventing laboratory-confirmed infections was 78•6% [CI95% 74·8 - 81·7]; and for reducing hospitalizations and deaths was, respectively, 87·6% [CI95% 80·3 - 92·2] and 84·8% [CI95% 75·0 - 90·7]. Effectiveness was high across all subgroups. INTERPRETATION Similarly to other vaccines, the administration of one dose of Gam-COVID-Vac was effective for a wide range of COVID-19-related outcomes. FUNDING This study did not receive any funding.
Collapse
Affiliation(s)
- Soledad González
- Ministry of Health of the Province of Buenos Aires, La Plata, Buenos Aires, Argentina
| | - Santiago Olszevicki
- Ministry of Health of the Province of Buenos Aires, La Plata, Buenos Aires, Argentina
| | - Martín Salazar
- Faculty of Medical Sciences - National University of La Plata
| | - Ana Calabria
- Ministry of Health of the Province of Buenos Aires, La Plata, Buenos Aires, Argentina
| | - Lorena Regairaz
- Immunology Unit, Children's Hospital Sor Maria Ludovica, La Plata, Buenos Aires, Argentina
| | - Lupe Marín
- Ministry of Health of the Province of Buenos Aires, La Plata, Buenos Aires, Argentina
| | - Patricia Campos
- Ministry of Health of the Province of Buenos Aires, La Plata, Buenos Aires, Argentina
| | - Teresa Varela
- Ministry of Health of the Province of Buenos Aires, La Plata, Buenos Aires, Argentina
| | | | - Leticia Ceriani
- Ministry of Health of the Province of Buenos Aires, La Plata, Buenos Aires, Argentina
| | - Enio Garcia
- Ministry of Health of the Province of Buenos Aires, La Plata, Buenos Aires, Argentina
| | - Nicolás Kreplak
- Ministry of Health of the Province of Buenos Aires, La Plata, Buenos Aires, Argentina
| | - Marina Pifano
- Ministry of Health of the Province of Buenos Aires, La Plata, Buenos Aires, Argentina
| | - Elisa Estenssoro
- Ministry of Health of the Province of Buenos Aires, La Plata, Buenos Aires, Argentina
| | - Franco Marsico
- Calculus Institute, University of Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
178
|
Petráš M. Highly Effective Naturally Acquired Protection Against COVID-19 Persists for at Least 1 Year: A Meta-Analysis. J Am Med Dir Assoc 2021; 22:2263-2265. [PMID: 34582779 PMCID: PMC8443339 DOI: 10.1016/j.jamda.2021.08.042] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/24/2021] [Accepted: 08/24/2021] [Indexed: 10/27/2022]
Affiliation(s)
- Marek Petráš
- Department of Epidemiology and Biostatistics, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
179
|
Swan DA, Goyal A, Bracis C, Moore M, Krantz E, Brown E, Cardozo-Ojeda F, Reeves DB, Gao F, Gilbert PB, Corey L, Cohen MS, Janes H, Dimitrov D, Schiffer JT. Mathematical Modeling of Vaccines That Prevent SARS-CoV-2 Transmission. Viruses 2021; 13:1921. [PMID: 34696352 PMCID: PMC8539635 DOI: 10.3390/v13101921] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 09/01/2021] [Accepted: 09/16/2021] [Indexed: 12/22/2022] Open
Abstract
SARS-CoV-2 vaccine clinical trials assess efficacy against disease (VEDIS), the ability to block symptomatic COVID-19. They only partially discriminate whether VEDIS is mediated by preventing infection completely, which is defined as detection of virus in the airways (VESUSC), or by preventing symptoms despite infection (VESYMP). Vaccine efficacy against transmissibility given infection (VEINF), the decrease in secondary transmissions from infected vaccine recipients, is also not measured. Using mathematical modeling of data from King County Washington, we demonstrate that if the Moderna (mRNA-1273QS) and Pfizer-BioNTech (BNT162b2) vaccines, which demonstrated VEDIS > 90% in clinical trials, mediate VEDIS by VESUSC, then a limited fourth epidemic wave of infections with the highly infectious B.1.1.7 variant would have been predicted in spring 2021 assuming rapid vaccine roll out. If high VEDIS is explained by VESYMP, then high VEINF would have also been necessary to limit the extent of this fourth wave. Vaccines which completely protect against infection or secondary transmission also substantially lower the number of people who must be vaccinated before the herd immunity threshold is reached. The limited extent of the fourth wave suggests that the vaccines have either high VESUSC or both high VESYMP and high VEINF against B.1.1.7. Finally, using a separate intra-host mathematical model of viral kinetics, we demonstrate that a 0.6 log vaccine-mediated reduction in average peak viral load might be sufficient to achieve 50% VEINF, which suggests that human challenge studies with a relatively low number of infected participants could be employed to estimate all three vaccine efficacy metrics.
Collapse
Affiliation(s)
- David A. Swan
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (D.A.S.); (A.G.); (M.M.); (E.K.); (E.B.); (F.C.-O.); (D.B.R.); (F.G.); (P.B.G.); (L.C.); (H.J.); (D.D.)
| | - Ashish Goyal
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (D.A.S.); (A.G.); (M.M.); (E.K.); (E.B.); (F.C.-O.); (D.B.R.); (F.G.); (P.B.G.); (L.C.); (H.J.); (D.D.)
| | - Chloe Bracis
- TIMC-IMAG/BCM, Université Grenoble Alpes, 38000 Grenoble, France;
| | - Mia Moore
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (D.A.S.); (A.G.); (M.M.); (E.K.); (E.B.); (F.C.-O.); (D.B.R.); (F.G.); (P.B.G.); (L.C.); (H.J.); (D.D.)
| | - Elizabeth Krantz
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (D.A.S.); (A.G.); (M.M.); (E.K.); (E.B.); (F.C.-O.); (D.B.R.); (F.G.); (P.B.G.); (L.C.); (H.J.); (D.D.)
| | - Elizabeth Brown
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (D.A.S.); (A.G.); (M.M.); (E.K.); (E.B.); (F.C.-O.); (D.B.R.); (F.G.); (P.B.G.); (L.C.); (H.J.); (D.D.)
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Fabian Cardozo-Ojeda
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (D.A.S.); (A.G.); (M.M.); (E.K.); (E.B.); (F.C.-O.); (D.B.R.); (F.G.); (P.B.G.); (L.C.); (H.J.); (D.D.)
| | - Daniel B. Reeves
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (D.A.S.); (A.G.); (M.M.); (E.K.); (E.B.); (F.C.-O.); (D.B.R.); (F.G.); (P.B.G.); (L.C.); (H.J.); (D.D.)
| | - Fei Gao
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (D.A.S.); (A.G.); (M.M.); (E.K.); (E.B.); (F.C.-O.); (D.B.R.); (F.G.); (P.B.G.); (L.C.); (H.J.); (D.D.)
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Peter B. Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (D.A.S.); (A.G.); (M.M.); (E.K.); (E.B.); (F.C.-O.); (D.B.R.); (F.G.); (P.B.G.); (L.C.); (H.J.); (D.D.)
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (D.A.S.); (A.G.); (M.M.); (E.K.); (E.B.); (F.C.-O.); (D.B.R.); (F.G.); (P.B.G.); (L.C.); (H.J.); (D.D.)
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Department of Medicine, University of Washington, Seattle, WA 98195, USA
- Department of Laboratory Medicine, University of Washington, Seattle, WA 98195, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Myron S. Cohen
- Institute of Global Health and Infectious Diseases, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Holly Janes
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (D.A.S.); (A.G.); (M.M.); (E.K.); (E.B.); (F.C.-O.); (D.B.R.); (F.G.); (P.B.G.); (L.C.); (H.J.); (D.D.)
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Dobromir Dimitrov
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (D.A.S.); (A.G.); (M.M.); (E.K.); (E.B.); (F.C.-O.); (D.B.R.); (F.G.); (P.B.G.); (L.C.); (H.J.); (D.D.)
- Department of Applied Mathematics, University of Washington, Seattle, WA 98195, USA
| | - Joshua T. Schiffer
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (D.A.S.); (A.G.); (M.M.); (E.K.); (E.B.); (F.C.-O.); (D.B.R.); (F.G.); (P.B.G.); (L.C.); (H.J.); (D.D.)
- Department of Medicine, University of Washington, Seattle, WA 98195, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| |
Collapse
|
180
|
Weng NP, Pawelec G. Validation of the effectiveness of SARS-CoV-2 vaccines in older adults in "real-world" settings. Immun Ageing 2021; 18:36. [PMID: 34551812 PMCID: PMC8455805 DOI: 10.1186/s12979-021-00248-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 09/10/2021] [Indexed: 01/06/2023]
Abstract
The rapidity of SARS-CoV-2 vaccination around the world has substantially reduced the number of new cases of COVID-19 and their severity in highly vaccinated countries. The unanticipated efficacy of SARS-CoV-2 vaccines in older adults has been very encouraging but the longevity of vaccine immunity is currently unknown and protection against emerging variants may be lower. Adoptive immunotherapy with neutralizing mAb may offer an alternative for poor vaccine responders, while the mechanisms underlying failure to respond are still unclear. Further studies of B and T cell responses and their regulation particularly in older populations will provide a more solid foundation to develop suitable approaches to optimize vaccine responses of older adults who fail to mount a durable response.
Collapse
Affiliation(s)
- Nan-Ping Weng
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, MD, USA.
| | - Graham Pawelec
- Department of Immunology, University of Tübingen, Tübingen, Germany.
- Health Sciences North Research Institute, Sudbury, ON, Canada.
| |
Collapse
|
181
|
Mostaghimi D, Valdez CN, Larson HT, Kalinich CC, Iwasaki A. Prevention of host-to-host transmission by SARS-CoV-2 vaccines. THE LANCET. INFECTIOUS DISEASES 2021; 22:e52-e58. [PMID: 34534512 PMCID: PMC8439617 DOI: 10.1016/s1473-3099(21)00472-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/09/2021] [Accepted: 07/28/2021] [Indexed: 01/19/2023]
Abstract
As the number of individuals vaccinated against SARS-CoV-2 rises worldwide, population-level data regarding the vaccines' ability to reduce infection are being generated. Randomised trials have shown that these vaccines dramatically reduce symptomatic COVID-19; however, less is known about their effects on transmission between individuals. The natural course of infection with SARS-CoV-2 involves infection of the respiratory epithelia and replication within the mucosa to sufficient viral titres for transmission via aerosol particles and droplets. Here we discuss the available data on the existing, approved SARS-CoV-2 vaccines' capacity to reduce transmissibility by reducing primary infection, viral replication, capacity for transmission, and symptomaticity. The potential for mucosal-targeted SARS-CoV-2 vaccine strategies to more effectively limit transmission than intramuscular vaccines is considered with regard to known immunological mechanisms. Finally, we enumerate the population-level effects of approved vaccines on transmission through observational studies following clinical trials and vaccine distribution in real-world settings.
Collapse
Affiliation(s)
- Darius Mostaghimi
- Department of Immunobiology, New Haven, CT, USA; Yale University School of Medicine, New Haven, CT, USA
| | | | - Haleigh T Larson
- Yale University School of Medicine, New Haven, CT, USA; Department of Cardiac Surgery, Yale New Haven Hospital, New Haven, CT, USA
| | - Chaney C Kalinich
- Yale University School of Medicine, New Haven, CT, USA; Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Akiko Iwasaki
- Department of Immunobiology, New Haven, CT, USA; Yale University School of Medicine, New Haven, CT, USA; Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| |
Collapse
|
182
|
Sharma K, Koirala A, Nicolopoulos K, Chiu C, Wood N, Britton PN. Vaccines for COVID-19: Where do we stand in 2021? Paediatr Respir Rev 2021; 39:22-31. [PMID: 34362666 PMCID: PMC8274273 DOI: 10.1016/j.prrv.2021.07.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 07/06/2021] [Indexed: 12/20/2022]
Abstract
As of July 2021, over 3 billion doses of a COVID-19 vaccines have been administered globally, and there are now 19 COVID-19 vaccines approved for use in at least one country. Several of these have been shown to be highly effective both in clinical trials and real-world observational studies, some of which have included special populations of interest. A small number of countries have approved a COVID-19 vaccine for use in adolescents or children. These are laudable achievements, but the global vaccination effort has been challenged by inequitable distribution of vaccines predominantly to high income countries, with only 0.9% of people in low-income countries having received at least one dose of a COVID-19 vaccine. Addressing this inequity is of critical importance and will result in better control of SARS-CoV-2 globally. Other challenges include: the reduced protection from COVID-19 vaccines against some strains of SARS-CoV-2, necessitating the development of variant specific vaccines; and uncertainties around the duration of protection from vaccine-induced immunity.
Collapse
Affiliation(s)
- Ketaki Sharma
- National Centre for Immunisation Research and Surveillance, Westmead, NSW, Australia; Sydney Medical School, The University of Sydney, NSW, Australia
| | - Archana Koirala
- National Centre for Immunisation Research and Surveillance, Westmead, NSW, Australia; Sydney Medical School, The University of Sydney, NSW, Australia; Department of Infectious Diseases, Nepean Hospital, Penrith, NSW, Australia
| | - Katrina Nicolopoulos
- National Centre for Immunisation Research and Surveillance, Westmead, NSW, Australia
| | - Clayton Chiu
- National Centre for Immunisation Research and Surveillance, Westmead, NSW, Australia; Sydney Medical School, The University of Sydney, NSW, Australia
| | - Nicholas Wood
- National Centre for Immunisation Research and Surveillance, Westmead, NSW, Australia; Sydney Medical School, The University of Sydney, NSW, Australia
| | - Philip N Britton
- Sydney Medical School, The University of Sydney, NSW, Australia; Department of Infectious Diseases and Microbiology, The Children's Hospital at Westmead, NSW, Australia.
| |
Collapse
|
183
|
Wei J, Stoesser N, Matthews PC, Ayoubkhani D, Studley R, Bell I, Bell JI, Newton JN, Farrar J, Diamond I, Rourke E, Howarth A, Marsden BD, Hoosdally S, Jones EY, Stuart DI, Crook DW, Peto TEA, Pouwels KB, Eyre DW, Walker AS. Antibody responses to SARS-CoV-2 vaccines in 45,965 adults from the general population of the United Kingdom. Nat Microbiol 2021; 6:1140-1149. [PMID: 34290390 PMCID: PMC8294260 DOI: 10.1038/s41564-021-00947-3] [Citation(s) in RCA: 208] [Impact Index Per Article: 69.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 07/01/2021] [Indexed: 02/08/2023]
Abstract
We report that in a cohort of 45,965 adults, who were receiving either the ChAdOx1 or the BNT162b2 SARS-CoV-2 vaccines, in those who had no prior infection with SARS-CoV-2, seroconversion rates and quantitative antibody levels after a single dose were lower in older individuals, especially in those aged >60 years. Two vaccine doses achieved high responses across all ages. Antibody levels increased more slowly and to lower levels with a single dose of ChAdOx1 compared with a single dose of BNT162b2, but waned following a single dose of BNT162b2 in older individuals. In descriptive latent class models, we identified four responder subgroups, including a 'low responder' group that more commonly consisted of people aged >75 years, males and individuals with long-term health conditions. Given our findings, we propose that available vaccines should be prioritized for those not previously infected and that second doses should be prioritized for individuals aged >60 years. Further data are needed to better understand the extent to which quantitative antibody responses are associated with vaccine-mediated protection.
Collapse
Affiliation(s)
- Jia Wei
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Big Data Institute, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Nicole Stoesser
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- The National Institute for Health Research Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at the University of Oxford, Oxford, UK
- The National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
- Department of Infectious Diseases and Microbiology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK
| | - Philippa C Matthews
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Department of Infectious Diseases and Microbiology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK
| | | | | | - Iain Bell
- Office for National Statistics, Newport, UK
| | - John I Bell
- Office of the Regius Professor of Medicine, University of Oxford, Oxford, UK
| | - John N Newton
- Health Improvement Directorate, Public Health England, London, UK
| | | | | | | | - Alison Howarth
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Department of Infectious Diseases and Microbiology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK
| | - Brian D Marsden
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Sarah Hoosdally
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - E Yvonne Jones
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - David I Stuart
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Derrick W Crook
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- The National Institute for Health Research Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at the University of Oxford, Oxford, UK
- The National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
- Department of Infectious Diseases and Microbiology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK
| | - Tim E A Peto
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- The National Institute for Health Research Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at the University of Oxford, Oxford, UK
- The National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
- Department of Infectious Diseases and Microbiology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK
| | - Koen B Pouwels
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Big Data Institute, Nuffield Department of Population Health, University of Oxford, Oxford, UK
- The National Institute for Health Research Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at the University of Oxford, Oxford, UK
- Health Economics Research Centre, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - David W Eyre
- Big Data Institute, Nuffield Department of Population Health, University of Oxford, Oxford, UK.
- The National Institute for Health Research Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at the University of Oxford, Oxford, UK.
- The National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, UK.
- Department of Infectious Diseases and Microbiology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK.
| | - A Sarah Walker
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Big Data Institute, Nuffield Department of Population Health, University of Oxford, Oxford, UK
- The National Institute for Health Research Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at the University of Oxford, Oxford, UK
- MRC Clinical Trials Unit at UCL, UCL, London, UK
| |
Collapse
|
184
|
Bauer S, Contreras S, Dehning J, Linden M, Iftekhar E, Mohr SB, Olivera-Nappa A, Priesemann V. Relaxing restrictions at the pace of vaccination increases freedom and guards against further COVID-19 waves. PLoS Comput Biol 2021; 17:e1009288. [PMID: 34473693 PMCID: PMC8412259 DOI: 10.1371/journal.pcbi.1009288] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/19/2021] [Indexed: 12/12/2022] Open
Abstract
Mass vaccination offers a promising exit strategy for the COVID-19 pandemic. However, as vaccination progresses, demands to lift restrictions increase, despite most of the population remaining susceptible. Using our age-stratified SEIRD-ICU compartmental model and curated epidemiological and vaccination data, we quantified the rate (relative to vaccination progress) at which countries can lift non-pharmaceutical interventions without overwhelming their healthcare systems. We analyzed scenarios ranging from immediately lifting restrictions (accepting high mortality and morbidity) to reducing case numbers to a level where test-trace-and-isolate (TTI) programs efficiently compensate for local spreading events. In general, the age-dependent vaccination roll-out implies a transient decrease of more than ten years in the average age of ICU patients and deceased. The pace of vaccination determines the speed of lifting restrictions; Taking the European Union (EU) as an example case, all considered scenarios allow for steadily increasing contacts starting in May 2021 and relaxing most restrictions by autumn 2021. Throughout summer 2021, only mild contact restrictions will remain necessary. However, only high vaccine uptake can prevent further severe waves. Across EU countries, seroprevalence impacts the long-term success of vaccination campaigns more strongly than age demographics. In addition, we highlight the need for preventive measures to reduce contagion in school settings throughout the year 2021, where children might be drivers of contagion because of them remaining susceptible. Strategies that maintain low case numbers, instead of high ones, reduce infections and deaths by factors of eleven and five, respectively. In general, policies with low case numbers significantly benefit from vaccination, as the overall reduction in susceptibility will further diminish viral spread. Keeping case numbers low is the safest long-term strategy because it considerably reduces mortality and morbidity and offers better preparedness against emerging escape or more contagious virus variants while still allowing for higher contact numbers (freedom) with progressing vaccinations.
Collapse
Affiliation(s)
- Simon Bauer
- Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
| | - Sebastian Contreras
- Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
- Centre for Biotechnology and Bioengineering, Universidad de Chile, Santiago, Chile
| | - Jonas Dehning
- Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
| | - Matthias Linden
- Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
- Institute for Theoretical Physics, Leibniz University, Hannover, Germany
| | - Emil Iftekhar
- Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
| | - Sebastian B. Mohr
- Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
| | - Alvaro Olivera-Nappa
- Centre for Biotechnology and Bioengineering, Universidad de Chile, Santiago, Chile
| | - Viola Priesemann
- Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
- Institute for the Dynamics of Complex Systems, University of Göttingen, Göttingen, Germany
| |
Collapse
|
185
|
Hakim A, Hasan MM, Hasan M, Lokman SM, Azim KF, Raihan T, Chowdhury PA, Azad AK. Major Insights in Dynamics of Host Response to SARS-CoV-2: Impacts and Challenges. Front Microbiol 2021; 12:637554. [PMID: 34512561 PMCID: PMC8424194 DOI: 10.3389/fmicb.2021.637554] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 07/28/2021] [Indexed: 01/08/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19), a pandemic declared by the World Health Organization on March 11, 2020, is caused by the infection of highly transmissible species of a novel coronavirus called severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). As of July 25, 2021, there are 194,372,584 cases and 4,167,937 deaths with high variability in clinical manifestations, disease burden, and post-disease complications among different people around the globe. Overall, COVID-19 is manifested as mild to moderate in almost 90% of the cases and only the rest 10% of the cases need hospitalization. However, patients with older age and those having different comorbidities have made worst the pandemic scenario. The variability of pathological consequences and clinical manifestations of COVID-19 is associated with differential host-SARS-CoV-2 interactions, which are influenced by the factors that originated from the SARS-CoV-2 and the host. These factors usually include the genomic attributes and virulent factors of the SARS-CoV-2, the burden of coinfection with other viruses and bacteria, age and gender of the individuals, different comorbidities, immune suppressions/deficiency, genotypes of major histocompatibility complex, and blood group antigens and antibodies. We herein retrieved and reviewed literatures from PubMed, Scopus, and Google relevant to clinical complications and pathogenesis of COVID-19 among people of different age, sex, and geographical locations; genomic characteristics of SARS-CoV-2 including its variants, host response under different variables, and comorbidities to summarize the dynamics of the host response to SARS-CoV-2 infection; and host response toward approved vaccines and treatment strategies against COVID-19. After reviewing a large number of published articles covering different aspects of host response to SARS-CoV-2, it is clear that one aspect from one region is not working with the scenario same to others, as studies have been done separately with a very small number of cases from a particular area/region of a country. Importantly, to combat such a pandemic as COVID-19, a conclusive understanding of the disease dynamics is required. This review emphasizes on the identification of the factors influencing the dynamics of host responses to SARS-CoV-2 and offers a future perspective to explore the molecular insights of COVID-19.
Collapse
Affiliation(s)
- Al Hakim
- Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Md. Mahbub Hasan
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King’s College London, Franklin-Wilkins Building, London, United Kingdom
| | - Mahmudul Hasan
- Department of Pharmaceutical and Industrial Biotechnology, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Syed Mohammad Lokman
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Kazi Faizul Azim
- Department of Microbial Biotechnology, Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Topu Raihan
- Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | | | - Abul Kalam Azad
- Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| |
Collapse
|
186
|
Iheanacho CO, Eze UIH, Adida EA. A systematic review of effectiveness of BNT162b2 mRNA and ChAdOx1 adenoviral vector COVID-19 vaccines in the general population. BULLETIN OF THE NATIONAL RESEARCH CENTRE 2021; 45:150. [PMID: 34456555 PMCID: PMC8383253 DOI: 10.1186/s42269-021-00607-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 08/15/2021] [Indexed: 05/14/2023]
Abstract
BACKGROUND High effectiveness of COVID-19 vaccines is essential for the pandemic control. This study systematically reviewed available evidence on effectiveness of ChAdOx1 and BNT162b2 vaccines in the general population, for improved vaccine policies and strategies. MAIN BODY OF THE ABSTRACT Using several keywords, a search of Scopus, PubMed, Google scholar and Hinari databases was conducted from December 1, 2020 to June 9, 2021. Eligible studies comprising original studies reporting effectiveness of the vaccines, were included following PRISMA guidelines. Individual studies were assessed for quality using National Heart, Lung and Blood Institute quality assessment tool. A total of 1766 titles were retrieved and 11 were included, out of which only 5 were peer-reviewed. Although data availability was limited, studies suggest equivalent effectiveness of BNT162b2 and ChAdOx1 COVID-19 vaccine against SARS-CoV-2 infection and COVID-19 related morbidity and mortality. Vaccine effectiveness increased steadily to about 35 days, with an enhanced effectiveness following the second dose. SHORT CONCLUSION BNT162 and ChAdOx1 vaccines were associated with equivalent and high effectiveness which increased with time and a second dose in the general population. This encourages continued practice of other preventive measures, particularly during the first week of vaccination, and reinforces the need for a second dose.
Collapse
Affiliation(s)
- Chinonyerem O. Iheanacho
- Department of Clinical Pharmacy and Public Health, Faculty of Pharmacy, University of Calabar, Calabar, PMB 1115 Nigeria
| | - Uchenna I. H. Eze
- Department of Clinical Pharmacy and Biopharmarcy, Faculty of Pharmacy, Olabisi Onabanjo University, Sagamu, Nigeria
| | - Emmanuel A. Adida
- Department of Clinical Pharmacy and Public Health, Faculty of Pharmacy, University of Calabar, Calabar, PMB 1115 Nigeria
| |
Collapse
|
187
|
Mor O, Mandelboim M, Fleishon S, Bucris E, Bar-Ilan D, Linial M, Nemet I, Kliker L, Lustig Y, Mendelson ES, Zuckerman NS. The Rise and Fall of a Local SARS-CoV-2 Variant with the Spike Protein Mutation L452R. Vaccines (Basel) 2021; 9:937. [PMID: 34452062 PMCID: PMC8402656 DOI: 10.3390/vaccines9080937] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/16/2021] [Accepted: 08/19/2021] [Indexed: 12/21/2022] Open
Abstract
Emerging SARS-CoV-2 variants may threaten global vaccination efforts and the awaited reduction in outbreak burden. In this study, we report a novel variant carrying the L452R mutation that emerged from a local B.1.362 lineage, B.1.362+L452R. The L452R mutation is associated with the Delta and Epsilon variants and was shown to cause increased infection and reduction in neutralization in pseudoviruses. Indeed, the B.1.362+L452R variant demonstrated a X4-fold reduction in neutralization capacity of sera from BNT162b2-vaccinated individuals compared to a wild-type strain. The variant infected 270 individuals in Israel between December 2020 and March 2021, until diminishing due to the gain in dominance of the Alpha variant in February 2021. This study demonstrates an independent, local emergence of a variant carrying a critical mutation, L452R, which may have the potential of becoming a variant of concern and emphasizes the importance of routine surveillance and detection of novel variants among efforts undertaken to prevent further disease spread.
Collapse
Affiliation(s)
- Orna Mor
- Central Virology Laboratory, Public Health Services, Ministry of Health, Sheba Medical Center, Tel-Hashomer 52621, Israel; (O.M.); (M.M.); (S.F.); (E.B.); (D.B.-I.); (I.N.); (L.K.); (Y.L.); (E.S.M.)
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Michal Mandelboim
- Central Virology Laboratory, Public Health Services, Ministry of Health, Sheba Medical Center, Tel-Hashomer 52621, Israel; (O.M.); (M.M.); (S.F.); (E.B.); (D.B.-I.); (I.N.); (L.K.); (Y.L.); (E.S.M.)
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Shay Fleishon
- Central Virology Laboratory, Public Health Services, Ministry of Health, Sheba Medical Center, Tel-Hashomer 52621, Israel; (O.M.); (M.M.); (S.F.); (E.B.); (D.B.-I.); (I.N.); (L.K.); (Y.L.); (E.S.M.)
| | - Efrat Bucris
- Central Virology Laboratory, Public Health Services, Ministry of Health, Sheba Medical Center, Tel-Hashomer 52621, Israel; (O.M.); (M.M.); (S.F.); (E.B.); (D.B.-I.); (I.N.); (L.K.); (Y.L.); (E.S.M.)
| | - Dana Bar-Ilan
- Central Virology Laboratory, Public Health Services, Ministry of Health, Sheba Medical Center, Tel-Hashomer 52621, Israel; (O.M.); (M.M.); (S.F.); (E.B.); (D.B.-I.); (I.N.); (L.K.); (Y.L.); (E.S.M.)
| | - Michal Linial
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel;
| | - Ital Nemet
- Central Virology Laboratory, Public Health Services, Ministry of Health, Sheba Medical Center, Tel-Hashomer 52621, Israel; (O.M.); (M.M.); (S.F.); (E.B.); (D.B.-I.); (I.N.); (L.K.); (Y.L.); (E.S.M.)
| | - Limor Kliker
- Central Virology Laboratory, Public Health Services, Ministry of Health, Sheba Medical Center, Tel-Hashomer 52621, Israel; (O.M.); (M.M.); (S.F.); (E.B.); (D.B.-I.); (I.N.); (L.K.); (Y.L.); (E.S.M.)
| | - Yaniv Lustig
- Central Virology Laboratory, Public Health Services, Ministry of Health, Sheba Medical Center, Tel-Hashomer 52621, Israel; (O.M.); (M.M.); (S.F.); (E.B.); (D.B.-I.); (I.N.); (L.K.); (Y.L.); (E.S.M.)
| | | | - Ella S. Mendelson
- Central Virology Laboratory, Public Health Services, Ministry of Health, Sheba Medical Center, Tel-Hashomer 52621, Israel; (O.M.); (M.M.); (S.F.); (E.B.); (D.B.-I.); (I.N.); (L.K.); (Y.L.); (E.S.M.)
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Neta S. Zuckerman
- Central Virology Laboratory, Public Health Services, Ministry of Health, Sheba Medical Center, Tel-Hashomer 52621, Israel; (O.M.); (M.M.); (S.F.); (E.B.); (D.B.-I.); (I.N.); (L.K.); (Y.L.); (E.S.M.)
| |
Collapse
|
188
|
Mor O, Mandelboim M, Fleishon S, Bucris E, Bar-Ilan D, Linial M, Nemet I, Kliker L, Lustig Y, Mendelson ES, Zuckerman NS. The Rise and Fall of a Local SARS-CoV-2 Variant with the Spike Protein Mutation L452R. Vaccines (Basel) 2021; 9. [PMID: 34452062 DOI: 10.1101/2021.07.03.21259957] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/16/2021] [Accepted: 08/19/2021] [Indexed: 05/20/2023] Open
Abstract
Emerging SARS-CoV-2 variants may threaten global vaccination efforts and the awaited reduction in outbreak burden. In this study, we report a novel variant carrying the L452R mutation that emerged from a local B.1.362 lineage, B.1.362+L452R. The L452R mutation is associated with the Delta and Epsilon variants and was shown to cause increased infection and reduction in neutralization in pseudoviruses. Indeed, the B.1.362+L452R variant demonstrated a X4-fold reduction in neutralization capacity of sera from BNT162b2-vaccinated individuals compared to a wild-type strain. The variant infected 270 individuals in Israel between December 2020 and March 2021, until diminishing due to the gain in dominance of the Alpha variant in February 2021. This study demonstrates an independent, local emergence of a variant carrying a critical mutation, L452R, which may have the potential of becoming a variant of concern and emphasizes the importance of routine surveillance and detection of novel variants among efforts undertaken to prevent further disease spread.
Collapse
Affiliation(s)
- Orna Mor
- Central Virology Laboratory, Public Health Services, Ministry of Health, Sheba Medical Center, Tel-Hashomer 52621, Israel
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Michal Mandelboim
- Central Virology Laboratory, Public Health Services, Ministry of Health, Sheba Medical Center, Tel-Hashomer 52621, Israel
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Shay Fleishon
- Central Virology Laboratory, Public Health Services, Ministry of Health, Sheba Medical Center, Tel-Hashomer 52621, Israel
| | - Efrat Bucris
- Central Virology Laboratory, Public Health Services, Ministry of Health, Sheba Medical Center, Tel-Hashomer 52621, Israel
| | - Dana Bar-Ilan
- Central Virology Laboratory, Public Health Services, Ministry of Health, Sheba Medical Center, Tel-Hashomer 52621, Israel
| | - Michal Linial
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Ital Nemet
- Central Virology Laboratory, Public Health Services, Ministry of Health, Sheba Medical Center, Tel-Hashomer 52621, Israel
| | - Limor Kliker
- Central Virology Laboratory, Public Health Services, Ministry of Health, Sheba Medical Center, Tel-Hashomer 52621, Israel
| | - Yaniv Lustig
- Central Virology Laboratory, Public Health Services, Ministry of Health, Sheba Medical Center, Tel-Hashomer 52621, Israel
| | - Ella S Mendelson
- Central Virology Laboratory, Public Health Services, Ministry of Health, Sheba Medical Center, Tel-Hashomer 52621, Israel
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Neta S Zuckerman
- Central Virology Laboratory, Public Health Services, Ministry of Health, Sheba Medical Center, Tel-Hashomer 52621, Israel
| |
Collapse
|
189
|
Gallo V, Chiodini P, Bruzzese D, Kondilis E, Howdon D, Mierau J, Bhopal R. Comparing the COVID-19 pandemic in space and over time in Europe, using numbers of deaths, crude rates and adjusted mortality trend ratios. Sci Rep 2021; 11:16443. [PMID: 34385482 PMCID: PMC8361083 DOI: 10.1038/s41598-021-95658-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 07/23/2021] [Indexed: 11/09/2022] Open
Abstract
Comparison of COVID-19 trends in space and over time is essential to monitor the pandemic and to indirectly evaluate non-pharmacological policies aimed at reducing the burden of disease. Given the specific age- and sex- distribution of COVID-19 mortality, the underlying sex- and age-distribution of populations need to be accounted for. The aim of this paper is to present a method for monitoring trends of COVID-19 using adjusted mortality trend ratios (AMTRs). Age- and sex-mortality distribution of a reference European population (N = 14,086) was used to calculate age- and sex-specific mortality rates. These were applied to each country to calculate the expected deaths. Adjusted Mortality Trend Ratios (AMTRs) with 95% confidence intervals (C.I.) were calculated for selected European countries on a daily basis from 17th March 2020 to 29th April 2021 by dividing observed cumulative mortality, by expected mortality, times the crude mortality of the reference population. These estimated the sex- and age-adjusted mortality for COVID-19 per million population in each country. United Kingdom experienced the highest number of COVID-19 related death in Europe. Crude mortality rates were highest Hungary, Czech Republic, and Luxembourg. Accounting for the age-and sex-distribution of the underlying populations with AMTRs for each European country, four different patterns were identified: countries which experienced a two-wave pandemic, countries with almost undetectable first wave, but with either a fast or a slow increase of mortality during the second wave; countries with consistently low rates throughout the period. AMTRs were highest in Eastern European countries (Hungary, Czech Republic, Slovakia, and Poland). Our methods allow a fair comparison of mortality in space and over time. These might be of use to indirectly estimating the efficacy of non-pharmacological health policies. The authors urge the World Health Organisation, given the absence of age and sex-specific mortality data for direct standardisation, to adopt this method to estimate the comparative mortality from COVID-19 pandemic worldwide.
Collapse
Affiliation(s)
- Valentina Gallo
- University of Groningen, Campus Fryslân, Wirdumerdijk 34, 8911 CE, Leeuwarden, The Netherlands. .,Queen Mary University of London, London, UK. .,London School of Hygiene and Tropical Medicine, London, UK.
| | - Paolo Chiodini
- Medical Statistics Unit, University of Campania "L. Vanvitelli", Naples, Italy
| | - Dario Bruzzese
- Medical Statistics, University of Naples "Federico II", Naples, Italy
| | | | | | - Jochen Mierau
- Aletta Jacobs School of Public Health, University of Groningen, Groningen, The Netherlands
| | - Raj Bhopal
- Usher Institute, University of Edinburgh, Edinburgh, Scotland, UK
| |
Collapse
|
190
|
Hou YC, Lu KC, Kuo KL. The Efficacy of COVID-19 Vaccines in Chronic Kidney Disease and Kidney Transplantation Patients: A Narrative Review. Vaccines (Basel) 2021; 9:885. [PMID: 34452010 PMCID: PMC8402591 DOI: 10.3390/vaccines9080885] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/06/2021] [Accepted: 08/08/2021] [Indexed: 12/13/2022] Open
Abstract
The SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) pandemic has posed a huge threat to global health because of its rapid spread and various mutant variants. Critical illness occurs in the elderly and vulnerable individuals, such as those with chronic kidney disease. The severity of SARS-CoV-2 infection is associated with the severity of chronic kidney disease (CKD)and even kidney transplantation (KT) because of the chronic use of immunosuppressive agents. To develop adaptive immunity against SARS-CoV-2, vaccination against the spike protein is important. Current phase III trials of vaccines against SARS-CoV-2 have not focused on a specific group of individuals, such as patients with CKD or those undergoing dialysis or kidney transplantation. Chronic use of immunosuppressive agents might disturb the immune response to the SARS-CoV-2 spike protein. On the basis of limited evidence, the immune compromised status of CKD patients might decrease neutralizing antibody development after a single dose of a specific vaccine. Boosting dosage more than the protocol might increase the titer of the neutralizing antibody in CKD patients. Further evidence is needed to understand the factors disturbing the immunogenicity of the SARS-CoV-2 vaccine, and CKD patients should receive the recommended dose of the SARS-CoV-2 vaccine due to their relatively immune compromised status.
Collapse
Affiliation(s)
- Yi-Chou Hou
- Division of Nephrology, Department of Medicine, Cardinal-Tien Hospital, New Taipei City 231, Taiwan;
- School of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
| | - Kuo-Cheng Lu
- Division of Nephrology, Department of Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan;
- School of Medicine, Buddhist Tzu Chi University, Hualien 970, Taiwan
| | - Ko-Lin Kuo
- Division of Nephrology, Department of Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan;
- School of Medicine, Buddhist Tzu Chi University, Hualien 970, Taiwan
| |
Collapse
|
191
|
Bok K, Sitar S, Graham BS, Mascola JR. Accelerated COVID-19 vaccine development: milestones, lessons, and prospects. Immunity 2021; 54:1636-1651. [PMID: 34348117 PMCID: PMC8328682 DOI: 10.1016/j.immuni.2021.07.017] [Citation(s) in RCA: 132] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 07/19/2021] [Accepted: 07/21/2021] [Indexed: 02/09/2023]
Abstract
The development of effective vaccines to combat infectious diseases is a complex multi-year and multi-stakeholder process. To accelerate the development of vaccines for coronavirus disease 2019 (COVID-19), a novel pathogen emerging in late 2019 and spreading globally by early 2020, the United States government (USG) mounted an operation bridging public and private sector expertise and infrastructure. The success of the endeavor can be seen in the rapid advanced development of multiple vaccine candidates, with several demonstrating efficacy and now being administered around the globe. Here, we review the milestones enabling the USG-led effort, the methods utilized, and ensuing outcomes. We discuss the current status of COVID-19 vaccine development and provide a perspective for how partnership and preparedness can be better utilized in response to future public-health pandemic emergencies.
Collapse
Affiliation(s)
- Karin Bok
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sandra Sitar
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - John R Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
192
|
Benning L, Töllner M, Hidmark A, Schaier M, Nusshag C, Kälble F, Reichel P, Buylaert M, Grenz J, Ponath G, Klein K, Zeier M, Süsal C, Schnitzler P, Morath C, Speer C. Heterologous ChAdOx1 nCoV-19/BNT162b2 Prime-Boost Vaccination Induces Strong Humoral Responses among Health Care Workers. Vaccines (Basel) 2021; 9:857. [PMID: 34451982 PMCID: PMC8402499 DOI: 10.3390/vaccines9080857] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/27/2021] [Accepted: 08/02/2021] [Indexed: 11/21/2022] Open
Abstract
Despite limited data on safety and immunogenicity, heterologous prime-boost vaccination is currently recommended for individuals with ChAdOx1 nCoV-19 prime immunization in certain age groups. In this prospective, single-center study we included 166 health care workers from Heidelberg University Hospital who received either heterologous ChAdOx1 nCoV-19/BNT162b2, homologous BNT162b2 or homologous ChAdOx1 nCoV-19 vaccination between December 2020 and May 2021. We measured anti-S1 IgG, SARS-CoV-2 specific neutralizing antibodies, and antibodies against different SARS-CoV-2 fragments 0-3 days before and 19-21 days after boost vaccination. Before boost, 55/70 (79%) ChAdOx1 nCoV-19-primed compared with 44/45 (98%) BNT162b2-primed individuals showed positive anti-S1 IgG with a median (IQR) anti-S1 IgG index of 1.95 (1.05-2.99) compared to 9.38 (6.26-17.12). SARS-CoV-2 neutralizing antibodies exceeded the threshold in 24/70 (34%) of ChAdOx1 nCoV-19-primed and 43/45 (96%) of BNT162b2-primed individuals. After boosting dose, median (IQR) anti-S1 IgG index in heterologous ChAdOx1 nCoV-19/BNT162b2 vaccinees was 116.2 (61.84-170), compared to 13.09 (7.03-29.02) in homologous ChAdOx1 nCoV-19 and 145.5 (100-291.1) in homologous BNT162b2 vaccinees. All boosted vaccinees exceeded the threshold for neutralization, irrespective of their vaccination scheme. Vaccination was well-tolerated overall. We show that heterologous ChAdOx1 nCoV-19/BNT162b2 vaccination is safe and induces a strong and broad humoral response in healthy individuals.
Collapse
Affiliation(s)
- Louise Benning
- Department of Nephrology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.T.); (A.H.); (M.S.); (C.N.); (F.K.); (P.R.); (M.B.); (J.G.); (G.P.); (K.K.); (M.Z.); (C.M.)
| | - Maximilian Töllner
- Department of Nephrology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.T.); (A.H.); (M.S.); (C.N.); (F.K.); (P.R.); (M.B.); (J.G.); (G.P.); (K.K.); (M.Z.); (C.M.)
| | - Asa Hidmark
- Department of Nephrology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.T.); (A.H.); (M.S.); (C.N.); (F.K.); (P.R.); (M.B.); (J.G.); (G.P.); (K.K.); (M.Z.); (C.M.)
| | - Matthias Schaier
- Department of Nephrology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.T.); (A.H.); (M.S.); (C.N.); (F.K.); (P.R.); (M.B.); (J.G.); (G.P.); (K.K.); (M.Z.); (C.M.)
| | - Christian Nusshag
- Department of Nephrology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.T.); (A.H.); (M.S.); (C.N.); (F.K.); (P.R.); (M.B.); (J.G.); (G.P.); (K.K.); (M.Z.); (C.M.)
| | - Florian Kälble
- Department of Nephrology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.T.); (A.H.); (M.S.); (C.N.); (F.K.); (P.R.); (M.B.); (J.G.); (G.P.); (K.K.); (M.Z.); (C.M.)
| | - Paula Reichel
- Department of Nephrology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.T.); (A.H.); (M.S.); (C.N.); (F.K.); (P.R.); (M.B.); (J.G.); (G.P.); (K.K.); (M.Z.); (C.M.)
| | - Mirabel Buylaert
- Department of Nephrology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.T.); (A.H.); (M.S.); (C.N.); (F.K.); (P.R.); (M.B.); (J.G.); (G.P.); (K.K.); (M.Z.); (C.M.)
| | - Julia Grenz
- Department of Nephrology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.T.); (A.H.); (M.S.); (C.N.); (F.K.); (P.R.); (M.B.); (J.G.); (G.P.); (K.K.); (M.Z.); (C.M.)
| | - Gerald Ponath
- Department of Nephrology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.T.); (A.H.); (M.S.); (C.N.); (F.K.); (P.R.); (M.B.); (J.G.); (G.P.); (K.K.); (M.Z.); (C.M.)
| | - Katrin Klein
- Department of Nephrology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.T.); (A.H.); (M.S.); (C.N.); (F.K.); (P.R.); (M.B.); (J.G.); (G.P.); (K.K.); (M.Z.); (C.M.)
| | - Martin Zeier
- Department of Nephrology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.T.); (A.H.); (M.S.); (C.N.); (F.K.); (P.R.); (M.B.); (J.G.); (G.P.); (K.K.); (M.Z.); (C.M.)
| | - Caner Süsal
- Institute of Immunology, University Hospital Heidelberg, 69120 Heidelberg, Germany;
| | - Paul Schnitzler
- Department of Infectious Diseases, Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany;
| | - Christian Morath
- Department of Nephrology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.T.); (A.H.); (M.S.); (C.N.); (F.K.); (P.R.); (M.B.); (J.G.); (G.P.); (K.K.); (M.Z.); (C.M.)
| | - Claudius Speer
- Department of Nephrology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.T.); (A.H.); (M.S.); (C.N.); (F.K.); (P.R.); (M.B.); (J.G.); (G.P.); (K.K.); (M.Z.); (C.M.)
- Molecular Medicine Partnership Unit Heidelberg, EMBL, 69117 Heidelberg, Germany
| |
Collapse
|
193
|
Kow CS, Hasan SS. Real-world effectiveness of BNT162b2 mRNA vaccine: a meta-analysis of large observational studies. Inflammopharmacology 2021; 29:1075-1090. [PMID: 34241782 PMCID: PMC8266992 DOI: 10.1007/s10787-021-00839-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 06/16/2021] [Indexed: 11/18/2022]
Abstract
This paper aims to summarize through meta-analyses the overall vaccine effectiveness of the BNT162b2 mRNA vaccine from observational studies. A systematic literature search with no language restriction was performed in electronic databases to identify eligible observational studies which reported the adjusted effectiveness of the BNT162b2 mRNA vaccine to prevent RT-PCR confirmed COVID-19. Meta-analyses with the random-effects model were used to calculate the pooled hazard ratio (HR) and pooled incidence rate ratio (IRR) at 95% confidence intervals, and the vaccine effectiveness was indicated as (pooled HR - 1)/HR or (pooled IRR - 1)/IRR. Nineteen studies were included for this meta-analysis. The meta-analysis revealed significant protective effect against RT-PCR confirmed COVID-19 ≥ 14 days after the first dose, with vaccine effectiveness of 53% (95% confidence interval 32-68%), and ≥ 7 days after the second dose, with vaccine effectiveness of 95% (95% confidence interval: 96-97%). Despite its effectiveness, reporting vaccine safety data by relevant stakeholders should be encouraged as BNT162b2 mRNA is a new vaccine that has not gained full approval. There have been limited data about vaccine effectiveness among immunocompromised patients; thus, the vaccine should be used cautiously in this patient population.
Collapse
Affiliation(s)
- Chia Siang Kow
- School of Postgraduate Studies, International Medical University, Kuala Lumpur, Malaysia.
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Petaling Jaya, Selangor, Malaysia.
| | - Syed Shahzad Hasan
- School of Applied Sciences, University of Huddersfield, Huddersfield, United Kingdom
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, Australia
| |
Collapse
|
194
|
Xiaoni C, Pengxiang W, Zhun W. Emergency use of COVID-19 vaccines recommended by the World Health Organization (WHO) as of June 2021. Drug Discov Ther 2021; 15:222-224. [PMID: 34275974 DOI: 10.5582/ddt.2021.01064] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
In December 2019, the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) caused the outbreak of coronavirus disease 2019 (COVID-19), and the resulting pandemic has caused widespread health problems and social and economic disruption. Thus far in 2021, more than 4 million people worldwide have died from COVID-19, so safe and efficacious vaccines are urgently needed to restore normal economic and social activities. According to the official guidance documents of the World Health Organization (WHO), vaccines based on four major strategies including mRNA, adenoviral vectors, inactivated viruses, and recombinant proteins have entered the stage of emergency use authorization and pre-certification evaluation. The current review summarizes these vaccines and it looks ahead to the development of additional COVID-19 vaccines in the future.
Collapse
Affiliation(s)
- Cui Xiaoni
- Sino-Cell Biomed Co., Ltd., Qingdao, Shandong, China
| | | | - Wei Zhun
- Institute of Innovative Drugs and Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
195
|
Rzymski P, Pazgan-Simon M, Simon K, Łapiński T, Zarębska-Michaluk D, Szczepańska B, Chojnicki M, Mozer-Lisewska I, Flisiak R. Clinical Characteristics of Hospitalized COVID-19 Patients Who Received at Least One Dose of COVID-19 Vaccine. Vaccines (Basel) 2021; 9:781. [PMID: 34358197 PMCID: PMC8310296 DOI: 10.3390/vaccines9070781] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/07/2021] [Accepted: 07/09/2021] [Indexed: 12/20/2022] Open
Abstract
The clinical trials of the COVID-19 vaccines that are authorized in the European Union have revealed high efficacy in preventing symptomatic infections. However, during vaccination campaigns, some vaccine recipients, including those partially and fully vaccinated, will experience severe COVID-19, requiring hospitalization. This may particularly concern patients with a diminished immune response to the vaccine, as well as non-responders. This work has retrospectively analyzed the 92 cases of patients who were hospitalized between 27 December 2020 and 31 May 2021 in four Polish healthcare units due to COVID-19, and who have previously received the COVID-19 vaccine (54.3% ≤ 14 days after the first dose, 26.1% > 14 days after the first dose, 7.6% ≤ 14 days after the second dose, and 12% > 14 days after the second dose). These patients represented a minute fraction (1.2%) of all the COVID-19 patients who were hospitalized during the same period in the same healthcare institutions. No significant differences in white blood count, absolute lymphocyte count nadir, C-reactive protein, interleukin-6, procalcitonin, oxygen saturation, lung involvement, and fever frequency were found between the recipients of the first and second vaccine dose. A total of 15 deaths were noted (1.1% of all fatal COVID-19 cases in the considered period and healthcare units), including six in patients who received the second dose (five > 14 days after the second dose)-three of these subjects were using immunosuppressive medicines, and two were confirmed to be vaccine non-responders. The study reassures that severe COVID-19 and deaths are not common in vaccinated individuals, highlights that the clinical course in such patients may not reveal any distinctive features, and advocates for close monitoring of those at a higher risk of vaccine failure.
Collapse
Affiliation(s)
- Piotr Rzymski
- Department of Environmental Medicine, Poznan University of Medical Sciences, 60-806 Poznań, Poland
- Integrated Science Association (ISA), Universal Scientific Education and Research Network (USERN), 60-806 Poznań, Poland
| | - Monika Pazgan-Simon
- 1st Infectious Diseases Ward, Gromkowski Regional Specialist Hospital, 50-149 Wroclaw, Poland;
- Department of Infectious Diseases and Hepatology, Wrocław Medical University, 51-149 Wrocław, Poland;
| | - Krzysztof Simon
- Department of Infectious Diseases and Hepatology, Wrocław Medical University, 51-149 Wrocław, Poland;
| | - Tadeusz Łapiński
- Department of Infectious Diseases and Hepatology, Medical University of Bialystok, 15-540 Białystok, Poland;
| | | | - Barbara Szczepańska
- Department of the Children’s Diseases, The Infectious-Neurological Subdivision, Jan Kochanowski University, 25-369 Kielce, Poland;
| | - Michał Chojnicki
- Department of Immunobiology, Poznan University of Medical Sciences, 60-806 Poznań, Poland;
- Department of Infectious Diseases, Jozef Strus Hospital, 61-285 Poznań, Poland;
| | | | - Robert Flisiak
- Department of Infectious Diseases and Hepatology, Medical University of Bialystok, 15-540 Białystok, Poland;
| |
Collapse
|
196
|
van den Borst B. Recovery after Covid-19. LANCET REGIONAL HEALTH-WESTERN PACIFIC 2021; 12:100208. [PMID: 34250517 PMCID: PMC8262450 DOI: 10.1016/j.lanwpc.2021.100208] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 06/22/2021] [Indexed: 01/10/2023]
Affiliation(s)
- Bram van den Borst
- Radboud University Medical Center, Department of Pulmonary Diseases, Nijmegen, The Netherlands
| |
Collapse
|