1651
|
Caniels TG, Bontjer I, van der Straten K, Poniman M, Burger JA, Appelman B, Lavell HAA, Oomen M, Godeke GJ, Valle C, Mögling R, van Willigen HDG, Wynberg E, Schinkel M, van Vught LA, Guerra D, Snitselaar JL, Chaturbhuj DN, Cuella Martin I, Amsterdam UMC COVID-19 S3/HCW study group, Moore JP, de Jong MD, Reusken C, Sikkens JJ, Bomers MK, de Bree GJ, van Gils MJ, Eggink D, Sanders RW. Emerging SARS-CoV-2 variants of concern evade humoral immune responses from infection and vaccination. SCIENCE ADVANCES 2021; 7:eabj5365. [PMID: 34516917 PMCID: PMC8442901 DOI: 10.1126/sciadv.abj5365] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 07/13/2021] [Indexed: 05/21/2023]
Abstract
Emerging SARS-CoV-2 variants of concern (VOCs) pose a threat to human immunity induced by natural infection and vaccination. We assessed the recognition of three VOCs (B.1.1.7, B.1.351, and P.1) in cohorts of COVID-19 convalescent patients (n = 69) and Pfizer-BioNTech vaccine recipients (n = 50). Spike binding and neutralization against all three VOCs were substantially reduced in most individuals, with the largest four- to sevenfold reduction in neutralization being observed against B.1.351. While hospitalized patients with COVID-19 and vaccinees maintained sufficient neutralizing titers against all three VOCs, 39% of nonhospitalized patients exhibited no detectable neutralization against B.1.351. Moreover, monoclonal neutralizing antibodies show sharp reductions in their binding kinetics and neutralizing potential to B.1.351 and P.1 but not to B.1.1.7. These data have implications for the degree to which pre-existing immunity can protect against subsequent infection with VOCs and informs policy makers of susceptibility to globally circulating SARS-CoV-2 VOCs.
Collapse
Affiliation(s)
- Tom G. Caniels
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Ilja Bontjer
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Karlijn van der Straten
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
- Department of Internal Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Meliawati Poniman
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Judith A. Burger
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Brent Appelman
- Center for Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - H. A. Ayesha Lavell
- Department of Internal Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Melissa Oomen
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Gert-Jan Godeke
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Coralie Valle
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Ramona Mögling
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Hugo D. G. van Willigen
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Elke Wynberg
- Department of Infectious Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
- Public Health Service of Amsterdam, Amsterdam, Netherlands
| | - Michiel Schinkel
- Center for Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Lonneke A. van Vught
- Center for Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Denise Guerra
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Jonne L. Snitselaar
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Devidas N. Chaturbhuj
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, NY, USA
| | - Isabel Cuella Martin
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Amsterdam UMC COVID-19 S3/HCW study group
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
- Department of Internal Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
- Center for Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
- Department of Internal Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
- Department of Infectious Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
- Public Health Service of Amsterdam, Amsterdam, Netherlands
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, NY, USA
| | - John P. Moore
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, NY, USA
| | - Menno D. de Jong
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Chantal Reusken
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Jonne J. Sikkens
- Department of Internal Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Marije K. Bomers
- Department of Internal Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Godelieve J. de Bree
- Department of Internal Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Marit J. van Gils
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Dirk Eggink
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Rogier W. Sanders
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, NY, USA
| |
Collapse
|
1652
|
Huang Y, Borisov O, Kee JJ, Carpp LN, Wrin T, Cai S, Sarzotti-Kelsoe M, McDanal C, Eaton A, Pajon R, Hural J, Posavad CM, Gill K, Karuna S, Corey L, McElrath MJ, Gilbert PB, Petropoulos CJ, Montefiori DC. Calibration of Two Validated SARS-CoV-2 Pseudovirus Neutralization Assays for COVID-19 Vaccine Evaluation. RESEARCH SQUARE 2021:rs.3.rs-862572. [PMID: 34494017 PMCID: PMC8423224 DOI: 10.21203/rs.3.rs-862572/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Vaccine-induced neutralizing antibodies (nAbs) are key biomarkers considered to be associated with vaccine efficacy. In United States Government-sponsored phase 3 efficacy trials of COVID-19 vaccines, nAbs are measured by two different validated pseudovirus-based SARS-CoV-2 neutralization assays, with each trial using one of the two assays. Here we describe and compare the nAb titers obtained in the two assays. We observe that one assay consistently yielded higher nAb titers than the other when both assays were performed on the World Health Organization’s anti-SARS-CoV-2 immunoglobulin International Standard, COVID-19 convalescent sera, and mRNA-1273 vaccinee sera. To overcome the challenge this difference in readout poses in comparing/combining data from the two assays, we evaluate three calibration approaches and show that readouts from the two assays can be calibrated to a common scale. These results may aid decision-making based on data from these assays for the evaluation and licensure of new or adapted COVID-19 vaccines.
Collapse
Affiliation(s)
- Yunda Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Oleg Borisov
- Biomedical Advanced Research and Development Authority, Washington DC, USA
| | - Jia Jin Kee
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Lindsay N. Carpp
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Terri Wrin
- LabCorp-Monogram Biosciences, South San Francisco, CA, USA
| | - Suqin Cai
- LabCorp-Monogram Biosciences, South San Francisco, CA, USA
| | - Marcella Sarzotti-Kelsoe
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
- Department of Immunology, Duke University Medical Center, Durham, NC, USA
| | - Charlene McDanal
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Amanda Eaton
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | | | - John Hural
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Christine M. Posavad
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Katherine Gill
- Desmond Tutu HIV Centre, University of Cape Town, Cape Town, South Africa
| | - Shelly Karuna
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - M. Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Departments of Medicine and Laboratory Medicine, University of Washington, Seattle, WA, USA
| | - Peter B. Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | | | | |
Collapse
|
1653
|
Barouch DH, Stephenson KE, Sadoff J, Yu J, Chang A, Gebre M, McMahan K, Liu J, Chandrashekar A, Patel S, Le Gars M, de Groot AM, Heerwegh D, Struyf F, Douoguih M, van Hoof J, Schuitemaker H. Durable Humoral and Cellular Immune Responses 8 Months after Ad26.COV2.S Vaccination. N Engl J Med 2021; 385:951-953. [PMID: 34260834 PMCID: PMC8314733 DOI: 10.1056/nejmc2108829] [Citation(s) in RCA: 168] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Affiliation(s)
| | | | - Jerald Sadoff
- Janssen Vaccines and Prevention, Leiden, the Netherlands
| | - Jingyou Yu
- Beth Israel Deaconess Medical Center, Boston, MA
| | - Aiquan Chang
- Beth Israel Deaconess Medical Center, Boston, MA
| | - Makda Gebre
- Beth Israel Deaconess Medical Center, Boston, MA
| | | | - Jinyan Liu
- Beth Israel Deaconess Medical Center, Boston, MA
| | | | | | | | | | | | - Frank Struyf
- Janssen Research and Development, Beerse, Belgium
| | | | - Johan van Hoof
- Janssen Vaccines and Prevention, Leiden, the Netherlands
| | | |
Collapse
|
1654
|
|
1655
|
Cai C, Peng Y, Shen E, Huang Q, Chen Y, Liu P, Guo C, Feng Z, Gao L, Zhang X, Gao Y, Liu Y, Han Y, Zeng S, Shen H. A comprehensive analysis of the efficacy and safety of COVID-19 vaccines. Mol Ther 2021; 29:2794-2805. [PMID: 34365034 PMCID: PMC8342868 DOI: 10.1016/j.ymthe.2021.08.001] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/04/2021] [Accepted: 07/31/2021] [Indexed: 12/17/2022] Open
Abstract
The numbers of cases and deaths from coronavirus disease 2019 (COVID-19) are continuously increasing. Many people are concerned about the efficacy and safety of the COVID-19 vaccines. We performed a comprehensive analysis of the published trials of COVID-19 vaccines and the real-world data from the Vaccine Adverse Event Reporting System. Globally, our research found that the efficacy of all vaccines exceeded 70%, and RNA-based vaccines had the highest efficacy of 94.29%; moreover, Black or African American people, young people, and males may experience greater vaccine efficacy. The spectrum of vaccine-related adverse drug reactions (ADRs) is extremely broad, and the most frequent ADRs are pain, fatigue, and headache. Most ADRs are tolerable and are mainly grade 1 or 2 in severity. Some severe ADRs have been identified (thromboembolic events, 21-75 cases per million doses; myocarditis/pericarditis, 2-3 cases per million doses). In summary, vaccines are a powerful tool that can be used to control the COVID-19 pandemic, with high efficacy and tolerable ADRs. In addition, the spectrum of ADRs associated with the vaccines is broad, and most of the reactions appear within a week, although some may be delayed. Therefore, ADRs after vaccination need to be identified and addressed in a timely manner.
Collapse
Affiliation(s)
- Changjing Cai
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yinghui Peng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Edward Shen
- Department of Life Science, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Qiaoqiao Huang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yihong Chen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Ping Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Cao Guo
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Ziyang Feng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Le Gao
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xiangyang Zhang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yan Gao
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yihan Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Ying Han
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
| | - Shan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China.
| | - Hong Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China.
| |
Collapse
|
1656
|
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: 5.8] [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
|
1657
|
Russo V, Politano L. Unmet needs on the management of COVID-19 vaccination in patients with neuromuscular disorders. ACTA MYOLOGICA : MYOPATHIES AND CARDIOMYOPATHIES : OFFICIAL JOURNAL OF THE MEDITERRANEAN SOCIETY OF MYOLOGY 2021; 40:113-115. [PMID: 34632292 PMCID: PMC8489168 DOI: 10.36185/2532-1900-051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 09/13/2021] [Indexed: 11/12/2022]
Abstract
COVID-19 outbreak has quickly reached alarming morbidity and mortality with vaccines being the only weapon to fight. Although the critical situation, no international guidelines on the vaccination management of patients with neuromuscular disorders (NMDs) has still been issued. We aimed to address some unmet needs about the management of COVID-19 vaccination in patients with NMDs.
Collapse
Affiliation(s)
- Vincenzo Russo
- Department of Medical Translational Sciences, University of Campania “Luigi Vanvitelli”, Monaldi Hospital, Naples, Italy
| | - Luisa Politano
- Cardiomyology and Medical Genetics, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Gaetano Torre Association for Muscular Dystrophies, Research Unit, Naples, Italy
| |
Collapse
|
1658
|
Caldwell JM, Le X, McIntosh L, Meehan MT, Ogunlade S, Ragonnet R, O'Neill GK, Trauer JM, McBryde ES. Vaccines and variants: Modelling insights into emerging issues in COVID-19 epidemiology. Paediatr Respir Rev 2021; 39:32-39. [PMID: 34417121 PMCID: PMC8294600 DOI: 10.1016/j.prrv.2021.07.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 07/13/2021] [Indexed: 01/21/2023]
Abstract
Mathematical modelling has played a pivotal role in understanding the epidemiology of and guiding public health responses to the ongoing coronavirus disease of 2019 (COVID-19) pandemic. Here, we review the role of epidemiological models in understanding evolving epidemic characteristics, including the effects of vaccination and Variants of Concern (VoC). We highlight ways in which models continue to provide important insights, including (1) calculating the herd immunity threshold and evaluating its limitations; (2) verifying that nascent vaccines can prevent severe disease, infection, and transmission but may be less efficacious against VoC; (3) determining optimal vaccine allocation strategies under efficacy and supply constraints; and (4) determining that VoC are more transmissible and lethal than previously circulating strains, and that immune escape may jeopardize vaccine-induced herd immunity. Finally, we explore how models can help us anticipate and prepare for future stages of COVID-19 epidemiology (and that of other diseases) through forecasts and scenario projections, given current uncertainties and data limitations.
Collapse
Affiliation(s)
| | - Xuan Le
- Epidemiological Modelling Unit, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Lorin McIntosh
- Epidemiological Modelling Unit, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Michael T Meehan
- Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Australia
| | - Samson Ogunlade
- Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Australia
| | - Romain Ragonnet
- Epidemiological Modelling Unit, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Genevieve K O'Neill
- Epidemiological Modelling Unit, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - James M Trauer
- Epidemiological Modelling Unit, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Emma S McBryde
- Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Australia.
| |
Collapse
|
1659
|
Al Bahrani S, Albarrak A, Alghamdi OA, Alghamdi MA, Hakami FH, Al Abaadi AK, Alkhrashi SA, Alghamdi MY, Almershad MM, Alenazi MM, El Gezery MH, Jebakumar AZ, Al-Tawfiq JA. Safety and Reactogenicity of the ChAdOx1 (AZD1222) COVID-19 Vaccine in Saudi Arabia. Int J Infect Dis 2021; 110:359-362. [PMID: 34320413 PMCID: PMC8310569 DOI: 10.1016/j.ijid.2021.07.052] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/09/2021] [Accepted: 07/22/2021] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION The Kingdom of Saudi Arabia was one of the first countries to implement a COVID-19 vaccination program. This study estimated the safety and reactogenicity of the ChAdOx1-S vaccine after the first dose administered to adults. METHODS This cross-sectional study included 1592 randomly selected vaccinees from April to May 2021. A questionnaire was delivered to the vaccinees via phone calls 7 and 21 days after the first vaccine dose. RESULTS Of the 1592 vaccinees who had the first dose, the mean age was 37.4 (± 9.6) years and 81% were males. Of all the vaccinees, 553 (34.7%) reported an adverse reaction on the first telephone call. The most common symptoms were: pain at the site of injection (485, 30.5%), musculoskeletal symptoms (438, 27.5%), skin rash (307, 19.2%), gastrointestinal symptoms (379, 23.8%) and fever (498, 31.3%). Men were more likely to report fever (76.9% vs. 23.1%; P = 0.005), skin rash (81.1% vs. 18.9%, P = 0.005) and pain at the injection site (77.3% vs. 22.7%, P < 0.0001). Post-vaccine COVID-19 infection was 0.5% and there were no hospitalizations. CONCLUSION This study observed no major side effects of the ChAdOx1-S vaccine and no reported breakthrough infection during the observation period.
Collapse
Affiliation(s)
- Salma Al Bahrani
- King Fahad Military Medical Complex, Dhahran, Saudi Arabia; Infectious Disease Unit, Specialty Internal Medicine, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Ali Albarrak
- Infectious Disease Division, Internal Medicine Department, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | | | | | | | | | | | | | | | | | | | | | - Jaffar A Al-Tawfiq
- Specialty Internal Medicine and Quality Department, Johns Hopkins Aramco Healthcare, Dhahran, Saudi Arabia; Infectious Disease Division, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA; Infectious Disease Division, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
1660
|
Charmet T, Schaeffer L, Grant R, Galmiche S, Chény O, Von Platen C, Maurizot A, Rogoff A, Omar F, David C, Septfons A, Cauchemez S, Gaymard A, Lina B, Lefrancois LH, Enouf V, van der Werf S, Mailles A, Levy-Bruhl D, Carrat F, Fontanet A. Impact of original, B.1.1.7, and B.1.351/P.1 SARS-CoV-2 lineages on vaccine effectiveness of two doses of COVID-19 mRNA vaccines: Results from a nationwide case-control study in France. THE LANCET REGIONAL HEALTH. EUROPE 2021; 8:100171. [PMID: 34278372 PMCID: PMC8277121 DOI: 10.1016/j.lanepe.2021.100171] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND We aimed to assess the effectiveness of two doses of mRNA COVID-19 vaccines against COVID-19 with the original virus and other lineages circulating in France. METHODS In this nationwide case-control study, cases were SARS-CoV-2 infected adults with onset of symptoms between 14 February and 3 May 2021. Controls were non-infected adults from a national representative panel matched to cases by age, sex, region, population density and calendar week. Participants completed an online questionnaire on recent activity-related exposures and vaccination history. Information about the infecting virus was based on a screening RT-PCR for either B.1.1.7 or B.1.351/P.1 variants. FINDINGS Included in our analysis were 7 288 adults infected with the original SARS-CoV-2 virus, 31 313 with the B.1.1.7 lineage, 2 550 with B.1.351/P1 lineages, and 3 644 controls. In multivariable analysis, the vaccine effectiveness (95% confidence interval) seven days after the second dose of mRNA vaccine was estimated at 88% (81-92), 86% (81-90) and 77% (63-86) against COVID-19 with the original virus, the B.1.1.7 lineage, and the B.1.351/P.1 lineages, respectively. Recent (2 to 6 months) history of virologically confirmed SARS-CoV-2 infection was found to be 83% (76-88), 88% (85-91) and 83% (71-90) protective against COVID-19 with the original virus, the B.1.1.7 lineage, and the B.1.351/P.1 lineages, respectively; and more distant (> 6 months) infections were 76% (54-87), 84% (75-90), and 74% (41-89) protective against COVID-19 with the original virus, the B.1.1.7 lineage, and the B.1.351/P.1 lineages, respectively. INTERPRETATION In real-life settings, two doses of mRNA vaccines proved to be effective against COVID-19 with the original virus, B.1.1.7 lineage and B.1.351/P.1 lineages. FUNDING Institut Pasteur, Research & Action Emerging Infectious Diseases (REACTing), Fondation de France (Alliance "Tous unis contre le virus").
Collapse
Affiliation(s)
- Tiffany Charmet
- Institut Pasteur, Emerging Diseases Epidemiology Unit, Paris, France
| | - Laura Schaeffer
- Institut Pasteur, Emerging Diseases Epidemiology Unit, Paris, France
| | - Rebecca Grant
- Institut Pasteur, Emerging Diseases Epidemiology Unit, Paris, France
- Sorbonne University, Paris, France
| | - Simon Galmiche
- Institut Pasteur, Emerging Diseases Epidemiology Unit, Paris, France
| | - Olivia Chény
- Institut Pasteur, Centre for Translational Research, Paris, France
| | | | | | | | | | | | | | - Simon Cauchemez
- Institut Pasteur, Mathematical Modelling of Infectious Diseases Unit; UMR2000; CNRS, Paris, France
| | - Alexandre Gaymard
- CNR des Virus des Infections Respiratoires, Institut des Agents Infectieux, Hospices Civils de Lyon
- Virpath, Centre International de Recherche En Infectiologie, Université de Lyon, Inserm U1111, CNRS UMR5308, École Normale Supérieure de Lyon, UCBL, Lyon, France
| | - Bruno Lina
- CNR des Virus des Infections Respiratoires, Institut des Agents Infectieux, Hospices Civils de Lyon
- Virpath, Centre International de Recherche En Infectiologie, Université de Lyon, Inserm U1111, CNRS UMR5308, École Normale Supérieure de Lyon, UCBL, Lyon, France
| | - Louise H Lefrancois
- Molecular Genetics of RNA Viruses, Department of Virology, Institut Pasteur CNRS UMR 3569; Université de Paris, Paris, France
- National Reference Center for Respiratory Viruses, Institut Pasteur, Paris, France
| | - Vincent Enouf
- Molecular Genetics of RNA Viruses, Department of Virology, Institut Pasteur CNRS UMR 3569; Université de Paris, Paris, France
- National Reference Center for Respiratory Viruses, Institut Pasteur, Paris, France
- Mutualized Platform of Microbiology, Pasteur International Bioresources Network, Institut Pasteur, Paris, France
| | - Sylvie van der Werf
- Molecular Genetics of RNA Viruses, Department of Virology, Institut Pasteur CNRS UMR 3569; Université de Paris, Paris, France
- National Reference Center for Respiratory Viruses, Institut Pasteur, Paris, France
| | | | | | - Fabrice Carrat
- Sorbonne Université, Inserm, IPLESP, hôpital Saint-Antoine, APHP, 27 rue Chaligny, Paris F75571, France
| | - Arnaud Fontanet
- Institut Pasteur, Emerging Diseases Epidemiology Unit, Paris, France
- Conservatoire national des arts et métiers, Unité PACRI, Paris, France
| |
Collapse
|
1661
|
Chakraborty C, Sharma AR, Bhattacharya M, Agoramoorthy G, Lee SS. Evolution, Mode of Transmission, and Mutational Landscape of Newly Emerging SARS-CoV-2 Variants. mBio 2021; 12:e0114021. [PMID: 34465019 PMCID: PMC8406297 DOI: 10.1128/mbio.01140-21] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 07/19/2021] [Indexed: 12/13/2022] Open
Abstract
The recent emergence of multiple variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become a significant concern for public health worldwide. New variants have been classified either as variants of concern (VOCs) or variants of interest (VOIs) by the CDC (USA) and WHO. The VOCs include lineages such as B.1.1.7 (20I/501Y.V1 variant), P.1 (20J/501Y.V3 variant), B.1.351 (20H/501Y.V2 variant), and B.1.617.2. In contrast, the VOI category includes B.1.525, B.1.526, P.2, and B.1.427/B.1.429. The WHO provided the alert for last two variants (P.2 and B.1.427/B.1.429) and labeled them for further monitoring. As per the WHO, these variants can be reclassified due to their status at a particular time. At the same time, the CDC (USA) has marked these two variants as VOIs up through today. This article analyzes the evolutionary patterns of all these emerging variants, as well as their geographical distributions and transmission patterns, including the circulating frequency, entropy diversity, and mutational event diversity throughout the genomes of all SARS-CoV-2 lineages. The transmission pattern was observed highest in the B.1.1.7 lineage. Our frequency evaluation found that this lineage achieved 100% frequency in early October 2020. We also critically evaluated the above emerging variants mutational landscape and significant spike protein mutations (E484K, K417T/N, N501Y, and D614G) impacting public health. Finally, the effectiveness of vaccines against newly SARS-CoV-2 variants was also analyzed. IMPORTANCE Irrespective of the aggressive vaccination drive, the newly emerging multiple SARS-CoV-2 variants are causing havoc in several countries. As per the CDC (USA) and WHO, the VOCs include the B.1.1.7, P.1, B.1.351, and B.1.617.2 lineages, while the VOIs include the B.1.525, B.1.526, P.2, and B.1.427/B.1.429 lineages. This study analyzed the evolutionary patterns, geographical distributions and transmission patterns, circulating frequency, entropy diversity, and mutational event diversity throughout the genome of significant SARS-CoV-2 lineages. A higher transmission pattern was observed for the B.1.1.7 variant. The study also evaluated the mutational landscape and important spike protein mutations (E484K, K417T/N, N501Y, and D614G) of all of the above variants. Finally, a survey was performed on the efficacy of vaccines against these variants from the previously published literature. The results presented in this article will help design future countrywide pandemic planning strategies for the emerging variants, next-generation vaccine development using alternative wild-type antigens and significant viral antigens, and immediate planning for ongoing vaccination programs worldwide.
Collapse
Affiliation(s)
- Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, West Bengal, India
| | - Ashish Ranjan Sharma
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, Gangwon-do, Republic of Korea
| | | | | | - Sang-Soo Lee
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, Gangwon-do, Republic of Korea
| |
Collapse
|
1662
|
Beatty AL, Peyser ND, Butcher XE, Carton TW, Olgin JE, Pletcher MJ, Marcus GM. The COVID-19 Citizen Science Study: Protocol for a Longitudinal Digital Health Cohort Study. JMIR Res Protoc 2021; 10:e28169. [PMID: 34310336 PMCID: PMC8407439 DOI: 10.2196/28169] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/14/2021] [Accepted: 06/04/2021] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND The COVID-19 pandemic has catalyzed a global public response and innovation in clinical study methods. OBJECTIVE The COVID-19 Citizen Science study was designed to generate knowledge about participant-reported COVID-19 symptoms, behaviors, and disease occurrence. METHODS COVID-19 Citizen Science is a longitudinal cohort study launched on March 26, 2020, on the Eureka Research Platform. This study illustrates important advances in digital clinical studies, including entirely digital study participation, targeted recruitment strategies, electronic consent, recurrent and time-updated assessments, integration with smartphone-based measurements, analytics for recruitment and engagement, connection with partner studies, novel engagement strategies such as participant-proposed questions, and feedback in the form of real-time results to participants. RESULTS As of February 2021, the study has enrolled over 50,000 participants. Study enrollment and participation are ongoing. Over the lifetime of the study, an average of 59% of participants have completed at least one survey in the past 4 weeks. CONCLUSIONS Insights about COVID-19 symptoms, behaviors, and disease occurrence can be drawn through digital clinical studies. Continued innovation in digital clinical study methods represents the future of clinical research. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) DERR1-10.2196/28169.
Collapse
Affiliation(s)
- Alexis L Beatty
- Division of Cardiology, University of California, San Francisco, San Francisco, CA, United States
| | - Noah D Peyser
- Division of Cardiology, University of California, San Francisco, San Francisco, CA, United States
| | - Xochitl E Butcher
- Division of Cardiology, University of California, San Francisco, San Francisco, CA, United States
| | - Thomas W Carton
- Louisiana Public Health Institute, New Orleans, LA, United States
| | - Jeffrey E Olgin
- Division of Cardiology, University of California, San Francisco, San Francisco, CA, United States
| | - Mark J Pletcher
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, United States
| | - Gregory M Marcus
- Division of Cardiology, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
1663
|
Beginning to understand clinical events and immune responses of hematopoietic cell transplant recipients receiving SARS-CoV-2 vaccination. Transplant Cell Ther 2021; 27:700-701. [PMID: 34452720 PMCID: PMC8384341 DOI: 10.1016/j.jtct.2021.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
1664
|
Thakkar A, Mishra S, Warner JL. COVID-19 Vaccine among Actively-Treated People with Cancer: A Glimpse into the Known Unknowns? J Natl Cancer Inst 2021; 114:169-171. [PMID: 34453849 DOI: 10.1093/jnci/djab175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 08/24/2021] [Indexed: 11/14/2022] Open
Affiliation(s)
- Astha Thakkar
- Department of Medicine, Division of Oncology, Montefiore Medical Center, Bronx, NY, USA
| | - Sanjay Mishra
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jeremy L Warner
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Medicine, Division of Hematology/Oncology, Vanderbilt University, Nashville, TN, USA.,Department of Biomedical Informatics, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
1665
|
Buehrle DJ, Sutton RR, McCann EL, Lucas AE. A Review of Treatment and Prevention of Coronavirus Disease 2019 among Solid Organ Transplant Recipients. Viruses 2021; 13:1706. [PMID: 34578287 PMCID: PMC8471770 DOI: 10.3390/v13091706] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/12/2021] [Accepted: 08/19/2021] [Indexed: 12/15/2022] Open
Abstract
Therapeutic management of solid organ transplant (SOT) recipients infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the cause of coronavirus disease 2019 (COVID-19), may challenge healthcare providers given a paucity of clinical data specific to this cohort. Herein, we summarize and review the studies that have formed the framework for current COVID-19 consensus management guidelines. Our review focuses on COVID-19 treatment options including monoclonal antibody products, antiviral agents such as remdesivir, and immunomodulatory agents such as corticosteroids, interleukin inhibitors, and kinase inhibitors. We highlight the presence or absence of clinical data of these therapeutics related to the SOT recipient with COVID-19. We also describe data surrounding COVID-19 vaccination of the SOT recipient. Understanding the extent and limitations of observational and clinical trial data for the prevention and treatment of COVID-19 specific to the SOT population is crucial for optimal management. Although minimal data exist on clinical outcomes among SOT recipients treated with varying COVID-19 therapeutics, reviewing these agents and the studies that have led to their inclusion or exclusion in clinical management of COVID-19 highlights the need for further studies of these therapeutics in SOT patients with COVID-19.
Collapse
Affiliation(s)
- Deanna J. Buehrle
- Department of Medicine, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA;
| | - Robert R. Sutton
- Department of Pharmacy, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA; (R.R.S.); (E.L.M.)
| | - Erin L. McCann
- Department of Pharmacy, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA; (R.R.S.); (E.L.M.)
| | - Aaron E. Lucas
- Department of Medicine, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA;
| |
Collapse
|
1666
|
Rhabdomyolysis Following Ad26.COV2.S COVID-19 Vaccination. Vaccines (Basel) 2021; 9:vaccines9090956. [PMID: 34579193 PMCID: PMC8472996 DOI: 10.3390/vaccines9090956] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/21/2021] [Accepted: 08/23/2021] [Indexed: 12/23/2022] Open
Abstract
We report the case of a 19-year-old male who complained of myalgia, muscle weakness, and darkened urine two days after receiving his Ad26.COV2.S (Johnson & Johnson, New Brunswick, New Jersey, United States) COVID-19 vaccination. Blood examination revealed an increased creatine kinase (CK) level, and his urinary dipstick tested positive for blood, indicative of acute rhabdomyolysis. Serum creatinine levels were normal. Rhabdomyolysis due to strenuous physical activity was ruled out and further diagnostics excluded an autoimmune cause. Under repeated treatment with intravenous fluid resuscitation (outpatient treatment), his symptoms resolved and peak CK levels of 44,180 U/L returned to almost normal levels within two weeks. Rhabdomyolysis is a rare, potentially fatal vaccine-induced reaction. Further research is needed to better understand the underlying pathomechanism and to investigate whether subcutaneous injection of vaccines may be able to prevent rhabdomyolysis.
Collapse
|
1667
|
Evaluation of Antibody Responses to COVID-19 Vaccines among Solid Tumor and Hematologic Patients. Cancers (Basel) 2021; 13:cancers13174312. [PMID: 34503127 PMCID: PMC8430869 DOI: 10.3390/cancers13174312] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/17/2021] [Accepted: 08/19/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Vaccination is the primary public health strategy to cope with the COVID-19 pandemic. Although solid tumor and hematologic patients are at higher risk of serious COVID-19-related complications and mortality, data on immune responses to COVID-19 vaccines in this patient cohort are particularly scarce. Our results show that antibody titers against the SARS-CoV-2 spike protein are significantly higher in solid tumor vs. hematologic patients. While SARS-CoV-2 antibody titers were equal among sexes, an age-dependent decrease could be observed. Of note, our studies additionally show that complete vaccination represents a valuable predictor for high anti-SARS-CoV-2 antibody responses in solid tumor and hematologic patients. Our findings aim to support future vaccination strategies in these highly vulnerable patients, including vaccination booster programs and alternative protective approaches. Abstract Vaccination is the primary public health strategy to cope with the COVID-19 pandemic. Although solid tumor and hematologic patients are at higher risk of serious COVID-19-related complications, data on immune responses to COVID-19 vaccines in this patient cohort are particularly scarce. The present study, therefore, aimed at the standardized determination of anti-SARS-CoV-2 spike protein antibody titers among non-vaccinated versus vaccinated solid tumor and hematologic patients who are under clinical observation or under treatment at the University Hospital Krems. Standardized anti-SARS-CoV-2 S antibody titers of a total of 441 patients were retrospectively analyzed. Our results show that antibody titers against the SARS-CoV-2 spike protein are significantly higher in solid tumor versus hematologic patients. While SARS-CoV-2 antibody titers were equal among sexes, an age-dependent decrease was observed. Of note, our studies additionally show that complete vaccination represents a valuable predictor for high anti-SARS-CoV-2 antibody responses in solid tumor and hematologic patients. In summary, to date, this is one of the largest studies to comprehensively evaluate the impact of various COVID-19 vaccines on anti-SARS-CoV-2 S antibody production in solid tumor and hematologic patients. Our findings aim to support future vaccination strategies in these highly vulnerable patients, including vaccination booster programs and alternative protective approaches.
Collapse
|
1668
|
Ying B, Whitener B, VanBlargan LA, Hassan AO, Shrihari S, Liang CY, Karl CE, Mackin S, Chen RE, Kafai NM, Wilks SH, Smith DJ, Carreño JM, Singh G, Krammer F, Carfi A, Elbashir S, Edwards DK, Thackray LB, Diamond MS. Protective activity of mRNA vaccines against ancestral and variant SARS-CoV-2 strains. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.08.25.457693. [PMID: 34462745 PMCID: PMC8404887 DOI: 10.1101/2021.08.25.457693] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Although mRNA vaccines prevent COVID-19, variants jeopardize their efficacy as immunity wanes. Here, we assessed the immunogenicity and protective activity of historical (mRNA-1273, designed for Wuhan-1 spike) or modified (mRNA-1273.351, designed for B.1.351 spike) preclinical Moderna mRNA vaccines in 129S2 and K18-hACE2 mice. Immunization with high or low dose formulations of mRNA vaccines induced neutralizing antibodies in serum against ancestral SARS-CoV-2 and several variants, although levels were lower particularly against the B.1.617.2 (Delta) virus. Protection against weight loss and lung pathology was observed with all high-dose vaccines against all viruses. Nonetheless, low-dose formulations of the vaccines, which produced lower magnitude antibody and T cell responses, and serve as a possible model for waning immunity, showed breakthrough lung infection and pneumonia with B.1.617.2. Thus, as levels of immunity induced by mRNA vaccines decline, breakthrough infection and disease likely will occur with some SARS-CoV-2 variants, suggesting a need for additional booster regimens.
Collapse
Affiliation(s)
- Baoling Ying
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Bradley Whitener
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Laura A. VanBlargan
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ahmed O. Hassan
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Swathi Shrihari
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Chieh-Yu Liang
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Courtney E. Karl
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine. St. Louis, MO, USA
| | - Samantha Mackin
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Rita E. Chen
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Natasha M. Kafai
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Samuel H. Wilks
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge UK
| | - Derek J. Smith
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge UK
| | - Juan Manuel Carreño
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gagandeep Singh
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | | | - Larissa B. Thackray
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michael S. Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine. St. Louis, MO, USA
| |
Collapse
|
1669
|
Zhou YW, Xie Y, Tang LS, Pu D, Zhu YJ, Liu JY, Ma XL. Therapeutic targets and interventional strategies in COVID-19: mechanisms and clinical studies. Signal Transduct Target Ther 2021; 6:317. [PMID: 34446699 PMCID: PMC8390046 DOI: 10.1038/s41392-021-00733-x] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/27/2021] [Accepted: 07/14/2021] [Indexed: 02/06/2023] Open
Abstract
Owing to the limitations of the present efforts on drug discovery against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the lack of the understanding of the biological regulation mechanisms underlying COVID-19, alternative or novel therapeutic targets for COVID-19 treatment are still urgently required. SARS-CoV-2 infection and immunity dysfunction are the two main courses driving the pathogenesis of COVID-19. Both the virus and host factors are potential targets for antiviral therapy. Hence, in this study, the current therapeutic strategies of COVID-19 have been classified into "target virus" and "target host" categories. Repurposing drugs, emerging approaches, and promising potential targets are the implementations of the above two strategies. First, a comprehensive review of the highly acclaimed old drugs was performed according to evidence-based medicine to provide recommendations for clinicians. Additionally, their unavailability in the fight against COVID-19 was analyzed. Next, a profound analysis of the emerging approaches was conducted, particularly all licensed vaccines and monoclonal antibodies (mAbs) enrolled in clinical trials against primary SARS-CoV-2 and mutant strains. Furthermore, the pros and cons of the present licensed vaccines were compared from different perspectives. Finally, the most promising potential targets were reviewed, and the update of the progress of treatments has been summarized based on these reviews.
Collapse
Affiliation(s)
- Yu-Wen Zhou
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yao Xie
- Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Department of Dermatovenerology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Lian-Sha Tang
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Dan Pu
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ya-Juan Zhu
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ji-Yan Liu
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
- Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| | - Xue-Lei Ma
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
- Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
1670
|
A Retrospective Survey among SARS-CoV-1 Infected Healthcare Workers after Three Years Post-Infection. Pathogens 2021; 10:pathogens10091078. [PMID: 34578111 PMCID: PMC8465910 DOI: 10.3390/pathogens10091078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/12/2021] [Accepted: 08/23/2021] [Indexed: 11/26/2022] Open
Abstract
Healthcare workers (HCWs) are on the frontline fighting several infectious diseases including SARS-CoV-1 and COVID-19. Coronavirus neutralizing antibodies (nAbs) were recently reported to last for a certain period. The factors affecting nAbs’ existence remain unclear. Here, we retrospectively analyzed the factors correlating with nAbs’ from SARS-CoV-1 long-term convalescence HCWs in Taiwan. One hundred and thirty SARS-CoV-1 convalescent patients were recruited between August 2006 and March 2007. Blood samples were collected to determine the anti-nucleocapsid (N) and anti-spike (S) antibodies’ existence status and neutralization ability. Neutralization ability was measured using SARS-CoV-1 pseudotyped viruses. Statistical analysis of factors associated with anti-SARS-CoV-1 antibodies’ existence status was determined using SAS software. 46.2% SARS-CoV-1 convalescent patients presented anti-N antibody after three years post-infection. Among sixty participants, ten participants co-presented anti-S antibodies. Eight participants with anti-S antibody displayed neutralization ability to SARS-CoV-1. The gender, age, and disease severity of participants did not affect the anti-N antibody existence status, whereas the anti-S antibody is significantly reduced in participants with old age (>50 years, p = 0.0434) after three years post SARS-CoV-1 infection. This study suggests that age is an important factor correlated with the duration of SARS-CoV-1 protective antibody existence status.
Collapse
|
1671
|
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: 1.8] [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
|
1672
|
Shazley O, Alshazley M. A COVID-Positive 52-Year-Old Man Presented With Venous Thromboembolism and Disseminated Intravascular Coagulation Following Johnson & Johnson Vaccination: A Case-Study. Cureus 2021; 13:e16383. [PMID: 34408937 PMCID: PMC8362796 DOI: 10.7759/cureus.16383] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/14/2021] [Indexed: 01/22/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) is caused by the severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2). Infection by the SARS-CoV-2 increases the risk for systematic multi-organ complications and venous, arterial thromboembolism. The need for an effective vaccine to combat the pandemic prompted the Centers for Disease Control and Prevention (CDC) and Food and Drug Administration (FDA) to approve a nationwide distribution of the Ad26.COV2.S vaccine manufactured by Johnson & Johnson (J&J). The use of the vaccine was halted after reported cases of cerebral venous sinus thrombosis (CVST) and thrombocytopenia among recipients. Researchers have postulated these rare occurrences as potentially immune-triggered responses associated with complement-mediated thrombotic microangiopathy (TMA). Thrombotic complications and thrombocytopenia increase the risk for blood clot growth due to the inflammation of immune complexes by pro-thrombotic activation of anti-platelet antibodies. A 52-year-old man presented to the intensive care unit (ICU) with severe dyspnea. He required bilevel positive airway pressure (BiPAP) for supplemental oxygen therapy. Endotracheal intubation was performed due to his worsened respiratory deterioration. Lab results suggested respiratory failure due to decreased partial pressure of oxygen (pO2) and increased partial pressure of carbon dioxide (pCO2). Findings of elevated D‐dimer levels with decreased fibrinogen and thrombocytopenia with prolonged prothrombin clotting time were consistent for disseminated intravascular coagulation (DIC). Chest radiography displayed moderate to heavy bilateral airspace consolidations, consistent with multifocal pneumonia suspicious for COVID-19. A computed tomography angiogram (CTA) revealed a mildly enlarged right ventricle and interventricular septum consistent for right heart strain due to a saddle pulmonary embolism (PE) that extended into the main pulmonary lobar segmental arteries bilaterally. The patient was transferred to a higher-level (tertiary) care for radiology intervention to remove the pulmonary embolism found on his lungs. This patient presented with severe dyspnea secondary to massive PE and deep venous thrombosis (DVT) due to SARS-CoV2 infection following the administration of the J&J vaccine. Bilateral thrombus opacities and pulmonary emboli are consistent among COVID-19 patients by intravascular coagulation with increased prothrombin time and D-dimer concentration with a low platelet count. Adverse emboli growths with increased D-dimer and thrombocytopenia strikes a similarity in recipients of the AstraZeneca vaccine due to vaccine-induced immune thrombotic thrombocytopenia (VITT). Administrative use of the J&J vaccine resumed in May 2021. The FDA's reassurance stemmed from their conclusive findings that the vaccine's benefits far outweigh these rare developments, which account for less than 0.01% of the total recipient population. Nevertheless, a further detailed analysis must be conducted on the adverse thrombotic manifestations following adenoviral-based COVID-19 vaccines (J&J, AstraZeneca) compared to mRNA-based vaccines (Moderna, Pfizer) to assess causality with higher specificity.
Collapse
Affiliation(s)
- Omar Shazley
- Basic Sciences, Saint James School of Medicine, Kingstown, VCT
| | - Moudar Alshazley
- Emergency Medicine, Internal Medicine, Santa Rosa Medical Center, Pensacola, USA
| |
Collapse
|
1673
|
Nathan A, Rossin EJ, Kaseke C, Park RJ, Khatri A, Koundakjian D, Urbach JM, Singh NK, Bashirova A, Tano-Menka R, Senjobe F, Waring MT, Piechocka-Trocha A, Garcia-Beltran WF, Iafrate AJ, Naranbhai V, Carrington M, Walker BD, Gaiha GD. Structure-guided T cell vaccine design for SARS-CoV-2 variants and sarbecoviruses. Cell 2021; 184:4401-4413.e10. [PMID: 34265281 PMCID: PMC8241654 DOI: 10.1016/j.cell.2021.06.029] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 04/02/2021] [Accepted: 06/24/2021] [Indexed: 12/05/2022]
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants that escape convalescent and vaccine-induced antibody responses has renewed focus on the development of broadly protective T-cell-based vaccines. Here, we apply structure-based network analysis and assessments of HLA class I peptide stability to define mutationally constrained CD8+ T cell epitopes across the SARS-CoV-2 proteome. Highly networked residues are conserved temporally among circulating variants and sarbecoviruses and disproportionately impair spike pseudotyped lentivirus infectivity when mutated. Evaluation of HLA class I stabilizing activity for 18 globally prevalent alleles identifies CD8+ T cell epitopes within highly networked regions with limited mutational frequencies in circulating SARS-CoV-2 variants and deep-sequenced primary isolates. Moreover, these epitopes elicit demonstrable CD8+ T cell reactivity in convalescent individuals but reduced recognition in recipients of mRNA-based vaccines. These data thereby elucidate key mutationally constrained regions and immunogenic epitopes in the SARS-CoV-2 proteome for a global T-cell-based vaccine against emerging variants and SARS-like coronaviruses.
Collapse
Affiliation(s)
- Anusha Nathan
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Program in Health Sciences & Technology, Harvard Medical School & Massachusetts Institute of Technology, Boston, MA 02115, USA
| | - Elizabeth J Rossin
- The Broad Institute, Cambridge, MA 02142, USA; Harvard Medical School Department of Ophthalmology, Massachusetts Eye and Ear, Boston, MA 02114, USA
| | - Clarety Kaseke
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Ryan J Park
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Harvard Radiation Oncology Program, Boston, MA 02114, USA
| | - Ashok Khatri
- Massachusetts General Hospital Endocrine Division and Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | | | | | - Nishant K Singh
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA 02142, USA
| | - Arman Bashirova
- Basic Science Program, Frederick National Laboratory for Cancer Research in the Laboratory of Integrative Cancer Immunology, National Cancer Institute, Bethesda, MD 20892, USA
| | - Rhoda Tano-Menka
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Fernando Senjobe
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Program in Virology, Harvard Medical School, Boston, MA 02115, USA
| | - Michael T Waring
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Alicja Piechocka-Trocha
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Wilfredo F Garcia-Beltran
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Department of Pathology, Massachusetts General Hospital, MA 02115, USA
| | - A John Iafrate
- Department of Pathology, Massachusetts General Hospital, MA 02115, USA
| | - Vivek Naranbhai
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02115, USA; Dana-Farber Cancer Institute, Boston, MA 02215, USA; Center for the AIDS Programme of Research in South Africa, Durban 4001, South Africa
| | - Mary Carrington
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Basic Science Program, Frederick National Laboratory for Cancer Research in the Laboratory of Integrative Cancer Immunology, National Cancer Institute, Bethesda, MD 20892, USA
| | - Bruce D Walker
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; The Broad Institute, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; Center for the AIDS Programme of Research in South Africa, Durban 4001, South Africa; Institute for Medical Engineering and Science and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Gaurav D Gaiha
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Division of Gastroenterology, Massachusetts General Hospital, Boston, MA 02114, USA.
| |
Collapse
|
1674
|
Salzberger B, Buder F, Lampl B, Ehrenstein B, Hitzenbichler F, Bauernfeind S, Holzmann T, Schmidt B, Hanses F. [Update on: SARS-CoV-2/COVID-19-epidemiology and prevention]. DER NEPHROLOGE 2021; 16:307-310. [PMID: 34426750 PMCID: PMC8374405 DOI: 10.1007/s11560-021-00529-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 07/23/2021] [Indexed: 11/23/2022]
Affiliation(s)
- Bernd Salzberger
- Abt. Krankenhaushygiene und Infektiologie, Universitätsklinikum Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Deutschland
| | - Felix Buder
- Abt. Krankenhaushygiene und Infektiologie, Universitätsklinikum Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Deutschland
| | - Benedikt Lampl
- Sachgebiet Infektionsschutz und Hygiene, Gesundheitsamt Regensburg, Regensburg, Deutschland
| | - Boris Ehrenstein
- Klinik für Rheumatologie und Klinische Immunologie, Fachklinikum Bad Abbach, Bad Abbach, Deutschland
| | - Florian Hitzenbichler
- Abt. Krankenhaushygiene und Infektiologie, Universitätsklinikum Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Deutschland
| | - Stilla Bauernfeind
- Abt. Krankenhaushygiene und Infektiologie, Universitätsklinikum Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Deutschland
| | - Thomas Holzmann
- Abt. Krankenhaushygiene und Infektiologie, Universitätsklinikum Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Deutschland
| | - Barbara Schmidt
- Institut für Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Regensburg, Regensburg, Deutschland
| | - Frank Hanses
- Abt. Krankenhaushygiene und Infektiologie, Universitätsklinikum Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Deutschland
| |
Collapse
|
1675
|
Ng XL, Betzler BK, Testi I, Ho SL, Tien M, Ngo WK, Zierhut M, Chee SP, Gupta V, Pavesio CE, de Smet MD, Agrawal R. Ocular Adverse Events After COVID-19 Vaccination. Ocul Immunol Inflamm 2021; 29:1216-1224. [PMID: 34559576 PMCID: PMC8477588 DOI: 10.1080/09273948.2021.1976221] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 08/24/2021] [Accepted: 08/29/2021] [Indexed: 12/23/2022]
Abstract
PURPOSE The COVID-19 pandemic has galvanized the development of new vaccines at an unprecedented pace. Since the widespread implementation of vaccination campaigns, reports of ocular adverse effects after COVID-19 vaccinations have emerged. This review summarizes ocular adverse effects possibly associated with COVID-19 vaccination, and discusses their clinical characteristics and management. METHODS Narrative Literature Review. RESULTS Ocular adverse effects of COVID-19 vaccinations include facial nerve palsy, abducens nerve palsy, acute macular neuroretinopathy, central serous retinopathy, thrombosis, uveitis, multiple evanescent white dot syndrome, Vogt-Koyanagi-Harada disease reactivation, and new-onset Graves' Disease. Studies in current literature are primarily retrospective case series or isolated case reports - these are inherently weak in establishing association or causality. Nevertheless, the described presentations resemble the reported ocular manifestations of the COVID-19 disease itself. Hence, we hypothesize that the human body's immune response to COVID-19 vaccinations may be involved in the pathogenesis of the ocular adverse effects post-COVID-19 vaccination. CONCLUSION Ophthalmologists and generalists should be aware of the possible, albeit rare, ocular adverse effects after COVID-19 vaccination.
Collapse
Affiliation(s)
- Xin Le Ng
- National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore
| | | | - Ilaria Testi
- Moorfields Eye Hospital, NHS Foundation Trust, London, UK
| | - Su Ling Ho
- National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore
| | - Melissa Tien
- National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore
| | - Wei Kiong Ngo
- National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore
| | - Manfred Zierhut
- Centre of Ophthalmology, University of Tuebingen, Tuebingen, Germany
| | - Soon Phaik Chee
- Department of Ocular Inflammation and Immunology, Singapore National Eye Centre, Singapore
- Singapore Eye Research Institute, The Academia, Singapore
- Department of Ophthalmology & Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore
| | - Vishali Gupta
- Department of Ophthalmology, Advanced Eye Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Carlos E Pavesio
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Marc D. de Smet
- MicroInvasive Ocular Surgery Clinic, Lausanne, Switzerland
- Department of Ophthalmology, University of Leiden, Leiden, The Netherlands
| | - Rupesh Agrawal
- National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Ocular Inflammation and Immunology, Singapore National Eye Centre, Singapore
- Singapore Eye Research Institute, The Academia, Singapore
- Department of Ophthalmology & Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| |
Collapse
|
1676
|
Abstract
The COVID-19 pandemic has motivated the rapid development of numerous vaccines that have proven effective against SARS-CoV-2. Several of these successful vaccines are based on the adenoviral vector platform. The mass manufacturing of these vaccines poses great challenges, especially in the context of a pandemic where extremely large quantities must be produced quickly at an affordable cost. In this work, two baseline processes for the production of a COVID-19 adenoviral vector vaccine, B1 and P1, were designed, simulated and economically evaluated with the aid of the software SuperPro Designer. B1 used a batch cell culture viral production step, with a viral titer of 5 × 1010 viral particles (VP)/mL in both stainless-steel and disposable equipment. P1 used a perfusion cell culture viral production step, with a viral titer of 1 × 1012 VP/mL in exclusively disposable equipment. Both processes were sized to produce 400 M/yr vaccine doses. P1 led to a smaller cost per dose than B1 ($0.15 vs. $0.23) and required a much smaller capital investment ($126 M vs. $299 M). The media and facility-dependent expenses were found to be the main contributors to the operating cost. The results indicate that adenoviral vector vaccines can be practically manufactured at large scale and low cost.
Collapse
|
1677
|
A Modeling Study on Vaccination and Spread of SARS-CoV-2 Variants in Italy. Vaccines (Basel) 2021; 9:vaccines9080915. [PMID: 34452040 PMCID: PMC8402493 DOI: 10.3390/vaccines9080915] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/29/2021] [Accepted: 08/12/2021] [Indexed: 12/16/2022] Open
Abstract
From the end of 2020, different vaccines against COVID-19 have been approved, offering a glimmer of hope and relief worldwide. However, in late 2020, new cases of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) started to re-surge, worsened by the emergence of highly infectious variants. To study this scenario, we extend the Susceptible-Exposed-Infectious-Removed model with lockdown measures used in our previous work with the inclusion of new lineages and mass vaccination campaign. We estimate model parameters using the Bayesian method Conditional Robust Calibration in two case studies: Italy and the Umbria region, the Italian region being worse affected by the emergence of variants. We then use the model to explore the dynamics of COVID-19, given different vaccination paces and a policy of gradual reopening. Our findings confirm the higher reproduction number of Umbria and the increase of transmission parameters due to the presence of new variants. The results illustrate the importance of preserving population-wide interventions, especially during the beginning of vaccination. Finally, under the hypothesis of waning immunity, the predictions show that a seasonal vaccination with a constant rate would probably be necessary to control the epidemic.
Collapse
|
1678
|
Paul E, Brown GW, Kalk A, Ridde V. Playing vaccine roulette: Why the current strategy of staking everything on Covid-19 vaccines is a high-stakes wager. Vaccine 2021; 39:4921-4924. [PMID: 34315610 PMCID: PMC8289694 DOI: 10.1016/j.vaccine.2021.07.045] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/14/2021] [Accepted: 07/16/2021] [Indexed: 12/15/2022]
Affiliation(s)
- Elisabeth Paul
- Université libre de Bruxelles, School of Public Health, Campus Erasme, Route de Lennik 808, CP 591, 1070 Brussels, Belgium.
| | - Garrett W Brown
- University of Leeds, Global Health Theme, POLIS, Leeds, United Kingdom.
| | - Andreas Kalk
- Deutsche Gesellschaft für Internationale Zusammenarbeit, Kinshasa Country Office, Kinshasa, The Democratic Republic of the Congo.
| | - Valéry Ridde
- Institut de Recherche pour le Développement (IRD), CEPED (IRD-Universités de Paris), INSERM, Paris, France.
| |
Collapse
|
1679
|
Altawalah H. Antibody Responses to Natural SARS-CoV-2 Infection or after COVID-19 Vaccination. Vaccines (Basel) 2021; 9:910. [PMID: 34452035 PMCID: PMC8402626 DOI: 10.3390/vaccines9080910] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/03/2021] [Accepted: 08/11/2021] [Indexed: 12/14/2022] Open
Abstract
The novel coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is the causative agent of the ongoing pandemic of coronavirus disease 2019 (COVID-19). The clinical severity of COVID-19 ranges from asymptomatic to critical disease and, eventually, death in smaller subsets of patients. The first case of COVID-19 was declared at the end of 2019 and it has since spread worldwide and remained a challenge in 2021, with the emergence of variants of concern. In fact, new concerns were the still unclear situation of SARS-CoV-2 immunity during the ongoing pandemic and progress with vaccination. If maintained at sufficiently high levels, the immune response could effectively block reinfection, which might confer long-lived protection. Understanding the protective capacity and the duration of humoral immunity during SARS-CoV-2 infection or after vaccination is critical for managing the pandemic and would also provide more evidence about the efficacy of SARS-CoV-2 vaccines. However, the exact features of antibody responses that govern SARS-CoV-2 infection or after vaccination remain unclear. This review summarizes the main knowledge that we have about the humoral immune response during COVID-19 disease or after vaccination. Such knowledge should help to optimize vaccination strategies and public health decisions.
Collapse
Affiliation(s)
- Haya Altawalah
- Department of Microbiology, Faculty of Medicine, Kuwait University, Safat 24923, Kuwait; or
- Virology Unit, Yacoub Behbehani Center, Sabah Hospital, Ministry of Health, Safat 24923, Kuwait
| |
Collapse
|
1680
|
Heinz FX, Stiasny K. Distinguishing features of current COVID-19 vaccines: knowns and unknowns of antigen presentation and modes of action. NPJ Vaccines 2021; 6:104. [PMID: 34400651 PMCID: PMC8368295 DOI: 10.1038/s41541-021-00369-6] [Citation(s) in RCA: 243] [Impact Index Per Article: 60.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/30/2021] [Indexed: 12/23/2022] Open
Abstract
COVID-19 vaccines were developed with an unprecedented pace since the beginning of the pandemic. Several of them have reached market authorization and mass production, leading to their global application on a large scale. This enormous progress was achieved with fundamentally different vaccine technologies used in parallel. mRNA, adenoviral vector as well as inactivated whole-virus vaccines are now in widespread use, and a subunit vaccine is in a final stage of authorization. They all rely on the native viral spike protein (S) of SARS-CoV-2 for inducing potently neutralizing antibodies, but the presentation of this key antigen to the immune system differs substantially between the different categories of vaccines. In this article, we review the relevance of structural modifications of S in different vaccines and the different modes of antigen expression after vaccination with genetic adenovirus-vector and mRNA vaccines. Distinguishing characteristics and unknown features are highlighted in the context of protective antibody responses and reactogenicity of vaccines.
Collapse
Affiliation(s)
- Franz X Heinz
- Center for Virology, Medical University of Vienna, Vienna, Austria.
| | - Karin Stiasny
- Center for Virology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
1681
|
Douxfils J, Gillot C, De Gottal É, Vandervinne S, Bayart JL, Dogné JM, Favresse J. Efficient Maternal to Neonate Transfer of Neutralizing Antibodies after SARS-CoV-2 Vaccination with BNT162b2: A Case-Report and Discussion of the Literature. Vaccines (Basel) 2021; 9:907. [PMID: 34452032 PMCID: PMC8402444 DOI: 10.3390/vaccines9080907] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 12/20/2022] Open
Abstract
This case reports on the successful maternal to fetal transfer of neutralizing antibodies after vaccination with BNT162b2 in a pregnant woman at 25 weeks of gestation. The levels of neutralizing antibodies were approximately 5-fold higher in the umbilical cord than in the maternal blood while the level of total antibodies showed only a 2-fold increase. This suggest that the antibodies that crossed the syncytiotrophoblast cell barrier have specific characteristics that correlate to functional neutralizing capacity. Although pregnant and lactating women have been excluded from clinical trials for several reasons including ethical concerns about fetal exposure, accumulating evidence has now revealed that these vaccines are safe and efficient for both the fetus and the woman. Vaccination against COVID-19 in pregnancy is vital to control disease burden and to decrease morbidity in the ante-, peri- and post-natal periods. Inclusion of pregnant women in research programs for the development of SARS-CoV-2 vaccines should be mandatory to provide this population with the equitable benefits of vaccine research.
Collapse
Affiliation(s)
- Jonathan Douxfils
- Qualiblood s.a., 5000 Namur, Belgium
- Namur Thrombosis and Hemostasis Center, Namur Research Institute for Life Sciences, Department of Pharmacy, Faculty of Medicine, University of Namur, 5000 Namur, Belgium; (C.G.); (J.-M.D.); (J.F.)
| | - Constant Gillot
- Namur Thrombosis and Hemostasis Center, Namur Research Institute for Life Sciences, Department of Pharmacy, Faculty of Medicine, University of Namur, 5000 Namur, Belgium; (C.G.); (J.-M.D.); (J.F.)
| | - Émilie De Gottal
- Département de Gynécologie, Centre Hospitalier Régional de Huy, 4500 Liège, Belgium;
| | - Stéphanie Vandervinne
- Laboratoire de Biologie Clinique, Centre Hospitalier Régional Huy, 4500 Liège, Belgium;
| | - Jean-Louis Bayart
- Department of Laboratory Medicine, Clinique Saint-Pierre Ottignies, 1340 Ottignies, Belgium;
| | - Jean-Michel Dogné
- Namur Thrombosis and Hemostasis Center, Namur Research Institute for Life Sciences, Department of Pharmacy, Faculty of Medicine, University of Namur, 5000 Namur, Belgium; (C.G.); (J.-M.D.); (J.F.)
| | - Julien Favresse
- Namur Thrombosis and Hemostasis Center, Namur Research Institute for Life Sciences, Department of Pharmacy, Faculty of Medicine, University of Namur, 5000 Namur, Belgium; (C.G.); (J.-M.D.); (J.F.)
- Department of Laboratory Medicine, Clinique Saint-Luc Bouge, 5004 Namur, Belgium
| |
Collapse
|
1682
|
Pascual-Iglesias A, Canton J, Ortega-Prieto AM, Jimenez-Guardeño JM, Regla-Nava JA. An Overview of Vaccines against SARS-CoV-2 in the COVID-19 Pandemic Era. Pathogens 2021; 10:1030. [PMID: 34451494 PMCID: PMC8402174 DOI: 10.3390/pathogens10081030] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/01/2021] [Accepted: 08/11/2021] [Indexed: 12/14/2022] Open
Abstract
The emergence of SARS-CoV-2 in late 2019 led to the COVID-19 pandemic all over the world. When the virus was first isolated and its genome was sequenced in the early months of 2020, the efforts to develop a vaccine began. Based on prior well-known knowledge about coronavirus, the SARS-CoV-2 spike (S) protein was selected as the main target. Currently, more than one hundred vaccines are being investigated and several of them are already authorized by medical agencies. This review summarizes and compares the current knowledge about main approaches for vaccine development, focusing on those authorized and specifically their immunogenicity, efficacy preventing severe disease, adverse side effects, protection, and ability to cope with emergent SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Alejandro Pascual-Iglesias
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, 28046 Madrid, Spain;
- Tumor Immunology Laboratory, IdiPAZ, La Paz University Hospital, 28046 Madrid, Spain
| | - Javier Canton
- International Institute for Defense and Security (CISDE), 41007 Sevilla, Spain;
| | - Ana Maria Ortega-Prieto
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King’s College London, London SE1 9RT, UK;
| | - Jose M. Jimenez-Guardeño
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King’s College London, London SE1 9RT, UK;
| | - Jose Angel Regla-Nava
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
- Department of Microbiology and Pathology, University Center for Health Science (CUCS), University of Guadalajara, Guadalajara 44340, Mexico
| |
Collapse
|
1683
|
Moline HL, Whitaker M, Deng L, Rhodes JC, Milucky J, Pham H, Patel K, Anglin O, Reingold A, Chai SJ, Alden NB, Kawasaki B, Meek J, Yousey-Hindes K, Anderson EJ, Farley MM, Ryan PA, Kim S, Nunez VT, Como-Sabetti K, Lynfield R, Sosin DM, McMullen C, Muse A, Barney G, Bennett NM, Bushey S, Shiltz J, Sutton M, Abdullah N, Talbot HK, Schaffner W, Chatelain R, Ortega J, Murthy BP, Zell E, Schrag SJ, Taylor C, Shang N, Verani JR, Havers FP. Effectiveness of COVID-19 Vaccines in Preventing Hospitalization Among Adults Aged ≥65 Years - COVID-NET, 13 States, February-April 2021. MMWR. MORBIDITY AND MORTALITY WEEKLY REPORT 2021; 70:1088-1093. [PMID: 34383730 PMCID: PMC8360274 DOI: 10.15585/mmwr.mm7032e3] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Clinical trials of COVID-19 vaccines currently authorized for emergency use in the United States (Pfizer-BioNTech, Moderna, and Janssen [Johnson & Johnson]) indicate that these vaccines have high efficacy against symptomatic disease, including moderate to severe illness (1-3). In addition to clinical trials, real-world assessments of COVID-19 vaccine effectiveness are critical in guiding vaccine policy and building vaccine confidence, particularly among populations at higher risk for more severe illness from COVID-19, including older adults. To determine the real-world effectiveness of the three currently authorized COVID-19 vaccines among persons aged ≥65 years during February 1-April 30, 2021, data on 7,280 patients from the COVID-19-Associated Hospitalization Surveillance Network (COVID-NET) were analyzed with vaccination coverage data from state immunization information systems (IISs) for the COVID-NET catchment area (approximately 4.8 million persons). Among adults aged 65-74 years, effectiveness of full vaccination in preventing COVID-19-associated hospitalization was 96% (95% confidence interval [CI] = 94%-98%) for Pfizer-BioNTech, 96% (95% CI = 95%-98%) for Moderna, and 84% (95% CI = 64%-93%) for Janssen vaccine products. Effectiveness of full vaccination in preventing COVID-19-associated hospitalization among adults aged ≥75 years was 91% (95% CI = 87%-94%) for Pfizer-BioNTech, 96% (95% CI = 93%-98%) for Moderna, and 85% (95% CI = 72%-92%) for Janssen vaccine products. COVID-19 vaccines currently authorized in the United States are highly effective in preventing COVID-19-associated hospitalizations in older adults. In light of real-world data demonstrating high effectiveness of COVID-19 vaccines among older adults, efforts to increase vaccination coverage in this age group are critical to reducing the risk for COVID-19-related hospitalization.
Collapse
|
1684
|
Corbett KS, Gagne M, Wagner DA, Connell SO, Narpala SR, Flebbe DR, Andrew SF, Davis RL, Flynn B, Johnston TS, Stringham C, Lai L, Valentin D, Van Ry A, Flinchbaugh Z, Werner AP, Moliva JI, Sriparna M, O'Dell S, Schmidt SD, Tucker C, Choi A, Koch M, Bock KW, Minai M, Nagata BM, Alvarado GS, Henry AR, Laboune F, Schramm CA, Zhang Y, Wang L, Choe M, Boyoglu-Barnum S, Shi W, Lamb E, Nurmukhambetova ST, Provost SJ, Donaldson MM, Marquez J, Todd JPM, Cook A, Dodson A, Pekosz A, Boritz E, Ploquin A, Doria-Rose N, Pessaint L, Andersen H, Foulds KE, Misasi J, Wu K, Carfi A, Nason MC, Mascola J, Moore IN, Edwards DK, Lewis MG, Suthar MS, Roederer M, McDermott A, Douek DC, Sullivan NJ, Graham BS, Seder RA. Protection against SARS-CoV-2 Beta Variant in mRNA-1273 Boosted Nonhuman Primates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.08.11.456015. [PMID: 34426813 PMCID: PMC8382125 DOI: 10.1101/2021.08.11.456015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2023]
Abstract
UNLABELLED Neutralizing antibody responses gradually wane after vaccination with mRNA-1273 against several variants of concern (VOC), and additional boost vaccinations may be required to sustain immunity and protection. Here, we evaluated the immune responses in nonhuman primates that received 100 µg of mRNA-1273 vaccine at 0 and 4 weeks and were boosted at week 29 with mRNA-1273 (homologous) or mRNA-1273.β (heterologous), which encompasses the spike sequence of the B.1.351 (beta or β) variant. Reciprocal ID 50 pseudovirus neutralizing antibody geometric mean titers (GMT) against live SARS-CoV-2 D614G and the β variant, were 4700 and 765, respectively, at week 6, the peak of primary response, and 644 and 553, respectively, at a 5-month post-vaccination memory time point. Two weeks following homologous or heterologous boost β-specific reciprocal ID 50 GMT were 5000 and 3000, respectively. At week 38, animals were challenged in the upper and lower airway with the β variant. Two days post-challenge, viral replication was low to undetectable in both BAL and nasal swabs in most of the boosted animals. These data show that boosting with the homologous mRNA-1273 vaccine six months after primary immunization provides up to a 20-fold increase in neutralizing antibody responses across all VOC, which may be required to sustain high-level protection against severe disease, especially for at-risk populations. ONE-SENTENCE SUMMARY mRNA-1273 boosted nonhuman primates have increased immune responses and are protected against SARS-CoV-2 beta infection.
Collapse
|
1685
|
Bernardeau-Serra L, Nguyen-Huynh A, Sponagel L, Sernizon Guimarães N, Teixeira de Aguiar RA, Soriano Marcolino M. The COVID-19 Vaccination Strategy in Brazil-A Case Study. EPIDEMIOLOGIA 2021; 2:338-359. [PMID: 36417230 PMCID: PMC9620893 DOI: 10.3390/epidemiologia2030026] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 07/23/2021] [Accepted: 08/03/2021] [Indexed: 12/14/2022] Open
Abstract
Brazil is among the countries which have faced two devastating infection waves of COVID-19 in the past year. Despite the fact the country has one of the world's leading immunization programs, Brazil only slowly established a national COVID-19 vaccination strategy and campaign. This case study is based on an integrative review of primary and secondary literature sources. Different search strategies on Medline and Google Scholar were performed for the case presentation, for the management and outcome of the COVID-19 outbreak and for the state of the COVID-19 vaccination program. Official documents from the Brazilian Ministry of Health, the website of the World Health Organization and pharmaceutical companies were also reviewed. Searches were limited to English, French, German, Portuguese and Spanish. This article describes the Brazilian COVID-19 vaccination campaign and the drivers and barriers to its implementation; and evaluates further investigations needed to have a conclusive overview over the constantly evolving situation. Healthcare inequalities, which were widened during the pandemic, a lack of coordination at the federal level, the absence of federal government support for scientific research and the lack of endorsement and commitment to the mitigation of the COVID-19 pandemic set the country's COVID-19 vaccination campaign off to a challenging start. However, Brazil had a well-developed primary care system and national vaccination program prior to the pandemic, which are both important facilitators. At the time of writing, six vaccines are currently available in the country, and the program is advancing. The scientific community needs to continue to investigate the country's vaccination strategy and its implementation to make sure that maximum effort is undertaken for the health of the Brazilian population.
Collapse
Affiliation(s)
| | | | - Lara Sponagel
- Global Studies Institute, Université de Genève, 1211 Geneva, Switzerland
| | - Nathalia Sernizon Guimarães
- Infectious Disease and Tropical Medicine Postgraduation Program, Medical School, Universidade Federal de Minas Gerais, Belo Horizonte 30130-100, Brazil;
| | | | - Milena Soriano Marcolino
- Department of Internal Medicine, Medical School, Universidade Federal de Minas Gerais, Belo Horizonte 30130-100, Brazil;
| |
Collapse
|
1686
|
Yen CC, Lin SY, Chen SC, Chiu YW, Chang JM, Hwang SJ. COVID-19 Vaccines in Patients with Maintenance Hemodialysis. J Pers Med 2021; 11:789. [PMID: 34442432 PMCID: PMC8399537 DOI: 10.3390/jpm11080789] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/08/2021] [Accepted: 08/11/2021] [Indexed: 12/15/2022] Open
Abstract
The COVID-19 pandemic has infected more than 180 million people and caused more than 3.95 million deaths worldwide. In addition to personal hygiene, augmented cleaning, social distancing, and isolation, vaccine development and immunization are essential for this highly contagious disease. Patients with maintenance hemodialysis (MHD) have a greater risk of COVID-19 infection owing to their comorbidities, defective immunity, and repeated crowded in-center dialysis settings. However, many patients hesitate to get vaccinated because of their misunderstandings. The Efficacy of COVID-19 vaccination has been intensively discussed in the general population, whereas the data concerning the effectiveness of vaccination in MHD patients are relatively scanty. Nevertheless, those limited publications can provide some valuable information. Overall, lower and more delayed antibody responses following COVID-19 vaccination were observed in patients with MHD than in healthy controls in the settings of different populations, vaccines and dosage, definitions of the immune response, and antibody detection timepoints. Younger age, previous COVID-19 infection, and higher serum albumin level were positively associated with antibody formation, whereas older age and receiving immunosuppressive therapy were unfavorable factors. However, it remains uncertain between the elicited antibodies following vaccination and the genuine protection against COVID-19 infection. Patients with MHD should make their COVID-19 vaccination a priority in addition to other protective measures. More studies focusing on different vaccines, non-humoral immune responses, and risk-benefit analyses are warranted.
Collapse
Affiliation(s)
- Cheng-Chieh Yen
- Division of Nephrology, Department of Internal Medicine, Ditmanson Medical Foundation, Chia-Yi Christian Hospital, Chia-Yi City 60002, Taiwan;
- Division of Nephrology, Chiayi Hospital, Ministry of Health and Welfare, Chia-Yi City 60096, Taiwan
| | - Shang-Yi Lin
- Division of Infectious Diseases, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan;
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80756, Taiwan; (S.-C.C.); (Y.-W.C.); (J.-M.C.)
| | - Szu-Chia Chen
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80756, Taiwan; (S.-C.C.); (Y.-W.C.); (J.-M.C.)
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
- Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung 81267, Taiwan
| | - Yi-Wen Chiu
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80756, Taiwan; (S.-C.C.); (Y.-W.C.); (J.-M.C.)
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
| | - Jer-Ming Chang
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80756, Taiwan; (S.-C.C.); (Y.-W.C.); (J.-M.C.)
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
| | - Shang-Jyh Hwang
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80756, Taiwan; (S.-C.C.); (Y.-W.C.); (J.-M.C.)
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
- Institute of Population Health Sciences, National Health Research Institutes, Miaoli 35053, Taiwan
| |
Collapse
|
1687
|
Schulze C, Welker A, Kühn A, Schwertz R, Otto B, Moraldo L, Dentz U, Arends A, Welk E, Wendorff JJ, Koller H, Kuss D, Ries M. Public Health Leadership in a VUCA World Environment: Lessons Learned during the Regional Emergency Rollout of SARS-CoV-2 Vaccinations in Heidelberg, Germany, during the COVID-19 Pandemic. Vaccines (Basel) 2021; 9:vaccines9080887. [PMID: 34452012 PMCID: PMC8402600 DOI: 10.3390/vaccines9080887] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/30/2021] [Accepted: 08/08/2021] [Indexed: 11/16/2022] Open
Abstract
The purpose of this work is to share methods used and lessons learned during a comprehensive inter-institutional pandemic disaster response in Heidelberg, Germany, conveying experiences of the regional SARS-CoV-2 vaccination rollout campaign for up to 1,000,000 vaccines in the year 2020. In this volatile, uncertain, complex, and ambiguous (VUCA) environment, the following five strategic elements were pertinent for institutional arrangements so that specific contributions of the various project partners would be available fast without the necessity of extensive negotiations or information exchange: (1) robust mandate, (2) use of established networks, (3) fast onboarding and securing of commitment of project partners, (4) informed planning of supply capacity, and (5) securing the availability of critical items. Planning tools included analyses through a VUCA lens, analyses of stakeholders and their management, possible failures, and management of main risks including mitigation strategies. The method of the present analysis (VUCA factors combined with analyses of possible failures, and management of stakeholders and risks) can theoretically be adjusted to any public health care emergency anywhere across the globe. Lessons learned include ten tactical leadership priorities and ten major pitfalls.
Collapse
Affiliation(s)
- Christoph Schulze
- Public Health Service Rhein-Neckar District and Heidelberg, 69115 Heidelberg, Germany; (C.S.); (A.W.); (A.K.); (R.S.); (B.O.); (L.M.); (D.K.)
| | - Andreas Welker
- Public Health Service Rhein-Neckar District and Heidelberg, 69115 Heidelberg, Germany; (C.S.); (A.W.); (A.K.); (R.S.); (B.O.); (L.M.); (D.K.)
| | - Anne Kühn
- Public Health Service Rhein-Neckar District and Heidelberg, 69115 Heidelberg, Germany; (C.S.); (A.W.); (A.K.); (R.S.); (B.O.); (L.M.); (D.K.)
| | - Rainer Schwertz
- Public Health Service Rhein-Neckar District and Heidelberg, 69115 Heidelberg, Germany; (C.S.); (A.W.); (A.K.); (R.S.); (B.O.); (L.M.); (D.K.)
| | - Benjamin Otto
- Public Health Service Rhein-Neckar District and Heidelberg, 69115 Heidelberg, Germany; (C.S.); (A.W.); (A.K.); (R.S.); (B.O.); (L.M.); (D.K.)
- Medical Faculty, University of Heidelberg, 69120 Heidelberg, Germany;
| | - Laura Moraldo
- Public Health Service Rhein-Neckar District and Heidelberg, 69115 Heidelberg, Germany; (C.S.); (A.W.); (A.K.); (R.S.); (B.O.); (L.M.); (D.K.)
| | - Udo Dentz
- Fire and Disaster Management Agency Rhein-Neckar District, 68526 Ladenburg, Germany;
| | - Albertus Arends
- Medical Faculty, University of Heidelberg, 69120 Heidelberg, Germany;
| | - Eckhard Welk
- CIMIC District Liaison Commands Heidelberg and Rhein-Neckar, 3rd Medical Regiment, German Federal Armed Forces, 89160 Dornstadt, Germany;
| | - Jean-Jacques Wendorff
- CIMIC District Liaison Command Heidelberg, German Federal Armed Forces, 70374 Stuttgart, Germany;
| | - Hans Koller
- Institute of Technology and Innovation Management, Helmut-Schmidt-University, University of the German Federal Armed Forces Hamburg, 22043 Hamburg, Germany;
| | - Doreen Kuss
- Public Health Service Rhein-Neckar District and Heidelberg, 69115 Heidelberg, Germany; (C.S.); (A.W.); (A.K.); (R.S.); (B.O.); (L.M.); (D.K.)
| | - Markus Ries
- CIMIC District Liaison Commands Heidelberg and Rhein-Neckar, 3rd Medical Regiment, German Federal Armed Forces, 89160 Dornstadt, Germany;
- Pediatric Neurology and Metabolic Medicine, Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, 69120 Heidelberg, Germany
- Center for Virtual Patients, Medical Faculty, University of Heidelberg, 69120 Heidelberg, Germany
- Correspondence:
| |
Collapse
|
1688
|
Duerr R, Dimartino D, Marier C, Zappile P, Wang G, Lighter J, Elbel B, Troxel AB, Heguy A. Dominance of alpha and Iota variants in SARS-CoV-2 vaccine breakthrough infections in New York City. J Clin Invest 2021; 131:e152702. [PMID: 34375308 DOI: 10.1172/jci152702] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 08/05/2021] [Indexed: 11/17/2022] Open
Abstract
The efficacy of COVID-19 mRNA vaccines is high, but breakthrough infections still occur. We compared the SARS-CoV-2 genomes of 76 breakthrough cases after full vaccination with BNT162b2 (Pfizer/BioNTech), mRNA-1273 (Moderna), or JNJ-78436735 (Janssen) to unvaccinated controls (February-April 2021) in metropolitan New York, including their phylogenetic relationship, distribution of variants, and full spike mutation profiles. Their median age was 48 years; seven required hospitalization and one died. Most breakthrough infections (57/76) occurred with B.1.1.7 (Alpha) or B.1.526 (Iota). Among the 7 hospitalized cases, 4 were infected with B.1.1.7, including 1 death. Both unmatched and matched statistical analyses considering age, sex, vaccine type, and study month as covariates supported the null hypothesis of equal variant distributions between vaccinated and unvaccinated in chi-squared and McNemar tests (p>0.1) highlighting a high vaccine efficacy against B.1.1.7 and B.1.526. There was no clear association among breakthroughs between type of vaccine received and variant. In the vaccinated group, spike mutations in the N-terminal domain and receptor-binding domain that have been associated with immune evasion were overrepresented. The evolving dynamic of SARS-CoV-2 variants requires broad genomic analyses of breakthrough infections to provide real-life information on immune escape mediated by circulating variants and their spike mutations.
Collapse
Affiliation(s)
- Ralf Duerr
- Department of Microbiology, NYU Langone Health, New York, United States of America
| | - Dacia Dimartino
- Office for Science and Research, NYU Langone Health, New York, United States of America
| | - Christian Marier
- Office for Science and Research, NYU Langone Health, New York, United States of America
| | - Paul Zappile
- Office for Science and Research, NYU Langone Health, New York, United States of America
| | - Guiqing Wang
- Department of Pathology, NYU Langone Health, New York, United States of America
| | - Jennifer Lighter
- Department of Pediatric Infectious Diseases, NYU Langone Health, New York, United States of America
| | - Brian Elbel
- Department of Population Health, NYU Langone Health, New York, United States of America
| | - Andrea B Troxel
- Department of Population Health, NYU Langone Health, New York, United States of America
| | - Adriana Heguy
- NYU Langone Health, New York, United States of America
| |
Collapse
|
1689
|
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: 52] [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] [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
|
1690
|
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: 151] [Impact Index Per Article: 37.8] [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
|
1691
|
Cimica V, Galarza JM, Rashid S, Stedman TT. Current development of Zika virus vaccines with special emphasis on virus-like particle technology. Expert Rev Vaccines 2021; 20:1483-1498. [PMID: 34148481 DOI: 10.1080/14760584.2021.1945447] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction: Zika virus disease received little attention until its recent explosive emergence around the globe. The devastating consequences of this pandemic include congenital Zika syndrome (CZS) and the neurological autoimmune disorder Guillain-Barré syndrome. These potential outcomes prompted massive efforts to understand the course of Zika infection and to develop therapeutic and prophylactic strategies for treatment and prevention of disease.Area covered: Preclinical and clinical data demonstrate that a safe and efficacious vaccine for protection against Zika virus infection is possible in the near future. Nevertheless, significant knowledge gaps regarding the outcome of a mass vaccination strategy exist and must be addressed. Zika virus circulates in flavivirus-endemic regions, an ideal Zika vaccine should avoid the potential of antibody-dependent enhancement from exposure to dengue virus. Prevention of CZS is the primary goal for immunization, and the vaccine must provide protection against intrauterine transmission for use during pregnancy and in women of childbearing age. Ideally, a vaccine should also prevent sexual transmission of the virus through mucosal protection.Expert opinion: This review describes current vaccine approaches against Zika virus with particular attention to the application of virus-like particle (VLP) technology as a strategy for solving the challenges of Zika virus immunization.
Collapse
Affiliation(s)
- Velasco Cimica
- American Type Culture Collection (ATCC), Manassas, VA, USA
| | | | - Sujatha Rashid
- American Type Culture Collection (ATCC), Manassas, VA, USA
| | | |
Collapse
|
1692
|
Thakkar A, Gonzalez-Lugo JD, Goradia N, Gali R, Shapiro LC, Pradhan K, Rahman S, Kim SY, Ko B, Sica RA, Kornblum N, Bachier-Rodriguez L, McCort M, Goel S, Perez-Soler R, Packer S, Sparano J, Gartrell B, Makower D, Goldstein YD, Wolgast L, Verma A, Halmos B. Seroconversion rates following COVID-19 vaccination among patients with cancer. Cancer Cell 2021; 39:1081-1090.e2. [PMID: 34133951 PMCID: PMC8179248 DOI: 10.1016/j.ccell.2021.06.002] [Citation(s) in RCA: 259] [Impact Index Per Article: 64.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 05/26/2021] [Accepted: 06/02/2021] [Indexed: 12/12/2022]
Abstract
As COVID-19 adversely affects patients with cancer, prophylactic strategies are critically needed. Using a validated antibody assay against SARS-CoV-2 spike protein, we determined a high seroconversion rate (94%) in 200 patients with cancer in New York City that had received full dosing with one of the FDA-approved COVID-19 vaccines. On comparison with solid tumors (98%), a significantly lower rate of seroconversion was observed in patients with hematologic malignancies (85%), particularly recipients following highly immunosuppressive therapies such as anti-CD20 therapies (70%) and stem cell transplantation (73%). Patients receiving immune checkpoint inhibitor therapy (97%) or hormonal therapies (100%) demonstrated high seroconversion post vaccination. Patients with prior COVID-19 infection demonstrated higher anti-spike IgG titers post vaccination. Relatively lower IgG titers were observed following vaccination with the adenoviral than with mRNA-based vaccines. These data demonstrate generally high immunogenicity of COVID-19 vaccination in oncology patients and identify immunosuppressed cohorts that need novel vaccination or passive immunization strategies.
Collapse
Affiliation(s)
- Astha Thakkar
- Department of Oncology, Montefiore Medical Center, Albert Einstein Cancer Center, Bronx, NY 10461, USA
| | - Jesus D Gonzalez-Lugo
- Department of Oncology, Montefiore Medical Center, Albert Einstein Cancer Center, Bronx, NY 10461, USA
| | - Niyati Goradia
- Department of Oncology, Montefiore Medical Center, Albert Einstein Cancer Center, Bronx, NY 10461, USA
| | - Radhika Gali
- Department of Oncology, Montefiore Medical Center, Albert Einstein Cancer Center, Bronx, NY 10461, USA
| | - Lauren C Shapiro
- Department of Oncology, Montefiore Medical Center, Albert Einstein Cancer Center, Bronx, NY 10461, USA
| | - Kith Pradhan
- Department of Oncology, Montefiore Medical Center, Albert Einstein Cancer Center, Bronx, NY 10461, USA
| | - Shafia Rahman
- Department of Oncology, Montefiore Medical Center, Albert Einstein Cancer Center, Bronx, NY 10461, USA
| | - So Yeon Kim
- Department of Oncology, Montefiore Medical Center, Albert Einstein Cancer Center, Bronx, NY 10461, USA
| | - Brian Ko
- Department of Oncology, Montefiore Medical Center, Albert Einstein Cancer Center, Bronx, NY 10461, USA
| | - R Alejandro Sica
- Department of Oncology, Montefiore Medical Center, Albert Einstein Cancer Center, Bronx, NY 10461, USA
| | - Noah Kornblum
- Department of Oncology, Montefiore Medical Center, Albert Einstein Cancer Center, Bronx, NY 10461, USA
| | | | - Margaret McCort
- Department of Medicine, Division of Infectious Diseases, Montefiore Medical Center, Bronx, NY 10461, USA
| | - Sanjay Goel
- Department of Oncology, Montefiore Medical Center, Albert Einstein Cancer Center, Bronx, NY 10461, USA
| | - Roman Perez-Soler
- Department of Oncology, Montefiore Medical Center, Albert Einstein Cancer Center, Bronx, NY 10461, USA
| | - Stuart Packer
- Department of Oncology, Montefiore Medical Center, Albert Einstein Cancer Center, Bronx, NY 10461, USA
| | - Joseph Sparano
- Department of Oncology, Montefiore Medical Center, Albert Einstein Cancer Center, Bronx, NY 10461, USA
| | - Benjamin Gartrell
- Department of Oncology, Montefiore Medical Center, Albert Einstein Cancer Center, Bronx, NY 10461, USA
| | - Della Makower
- Department of Oncology, Montefiore Medical Center, Albert Einstein Cancer Center, Bronx, NY 10461, USA
| | - Yitz D Goldstein
- Department of Pathology, Montefiore Medical Center, Bronx, NY 10461, USA
| | - Lucia Wolgast
- Department of Pathology, Montefiore Medical Center, Bronx, NY 10461, USA
| | - Amit Verma
- Department of Oncology, Montefiore Medical Center, Albert Einstein Cancer Center, Bronx, NY 10461, USA.
| | - Balazs Halmos
- Department of Oncology, Montefiore Medical Center, Albert Einstein Cancer Center, Bronx, NY 10461, USA.
| |
Collapse
|
1693
|
King RG, Silva-Sanchez A, Peel JN, Botta D, Dickson AM, Pinto AK, Meza-Perez S, Allie SR, Schultz MD, Liu M, Bradley JE, Qiu S, Yang G, Zhou F, Zumaquero E, Simpler TS, Mousseau B, Killian JT, Dean B, Shang Q, Tipper JL, Risley CA, Harrod KS, Feng T, Lee Y, Shiberu B, Krishnan V, Peguillet I, Zhang J, Green TJ, Randall TD, Suschak JJ, Georges B, Brien JD, Lund FE, Roberts MS. Single-Dose Intranasal Administration of AdCOVID Elicits Systemic and Mucosal Immunity against SARS-CoV-2 and Fully Protects Mice from Lethal Challenge. Vaccines (Basel) 2021; 9:881. [PMID: 34452006 PMCID: PMC8402488 DOI: 10.3390/vaccines9080881] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/29/2021] [Accepted: 08/03/2021] [Indexed: 02/08/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has highlighted the urgent need for effective prophylactic vaccination to prevent the spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Intranasal vaccination is an attractive strategy to prevent COVID-19 as the nasal mucosa represents the first-line barrier to SARS-CoV-2 entry. The current intramuscular vaccines elicit systemic immunity but not necessarily high-level mucosal immunity. Here, we tested a single intranasal dose of our candidate adenovirus type 5-vectored vaccine encoding the receptor-binding domain (RBD) of the SARS-CoV-2 spike protein (AdCOVID) in inbred, outbred, and transgenic mice. A single intranasal vaccination with AdCOVID elicited a strong and focused immune response against RBD through the induction of mucosal IgA in the respiratory tract, serum neutralizing antibodies, and CD4+ and CD8+ T cells with a Th1-like cytokine expression profile. A single AdCOVID dose resulted in immunity that was sustained for over six months. Moreover, a single intranasal dose completely protected K18-hACE2 mice from lethal SARS-CoV-2 challenge, preventing weight loss and mortality. These data show that AdCOVID promotes concomitant systemic and mucosal immunity and represents a promising vaccine candidate.
Collapse
Affiliation(s)
- R. Glenn King
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (R.G.K.); (J.N.P.); (D.B.); (M.D.S.); (S.Q.); (G.Y.); (F.Z.); (E.Z.); (T.S.S.); (B.M.); (J.T.K.J.); (B.D.); (Q.S.); (C.A.R.); (T.J.G.)
| | - Aaron Silva-Sanchez
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (A.S.-S.); (S.M.-P.); (S.R.A.); (M.L.); (J.E.B.); (T.D.R.)
| | - Jessica N. Peel
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (R.G.K.); (J.N.P.); (D.B.); (M.D.S.); (S.Q.); (G.Y.); (F.Z.); (E.Z.); (T.S.S.); (B.M.); (J.T.K.J.); (B.D.); (Q.S.); (C.A.R.); (T.J.G.)
| | - Davide Botta
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (R.G.K.); (J.N.P.); (D.B.); (M.D.S.); (S.Q.); (G.Y.); (F.Z.); (E.Z.); (T.S.S.); (B.M.); (J.T.K.J.); (B.D.); (Q.S.); (C.A.R.); (T.J.G.)
| | - Alexandria M. Dickson
- Department of Molecular Microbiology & Immunology, Saint Louis University, St. Louis, MO 63104, USA; (A.M.D.); (A.K.P.); (J.D.B.)
| | - Amelia K. Pinto
- Department of Molecular Microbiology & Immunology, Saint Louis University, St. Louis, MO 63104, USA; (A.M.D.); (A.K.P.); (J.D.B.)
| | - Selene Meza-Perez
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (A.S.-S.); (S.M.-P.); (S.R.A.); (M.L.); (J.E.B.); (T.D.R.)
| | - S. Rameeza Allie
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (A.S.-S.); (S.M.-P.); (S.R.A.); (M.L.); (J.E.B.); (T.D.R.)
| | - Michael D. Schultz
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (R.G.K.); (J.N.P.); (D.B.); (M.D.S.); (S.Q.); (G.Y.); (F.Z.); (E.Z.); (T.S.S.); (B.M.); (J.T.K.J.); (B.D.); (Q.S.); (C.A.R.); (T.J.G.)
| | - Mingyong Liu
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (A.S.-S.); (S.M.-P.); (S.R.A.); (M.L.); (J.E.B.); (T.D.R.)
| | - John E. Bradley
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (A.S.-S.); (S.M.-P.); (S.R.A.); (M.L.); (J.E.B.); (T.D.R.)
| | - Shihong Qiu
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (R.G.K.); (J.N.P.); (D.B.); (M.D.S.); (S.Q.); (G.Y.); (F.Z.); (E.Z.); (T.S.S.); (B.M.); (J.T.K.J.); (B.D.); (Q.S.); (C.A.R.); (T.J.G.)
| | - Guang Yang
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (R.G.K.); (J.N.P.); (D.B.); (M.D.S.); (S.Q.); (G.Y.); (F.Z.); (E.Z.); (T.S.S.); (B.M.); (J.T.K.J.); (B.D.); (Q.S.); (C.A.R.); (T.J.G.)
| | - Fen Zhou
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (R.G.K.); (J.N.P.); (D.B.); (M.D.S.); (S.Q.); (G.Y.); (F.Z.); (E.Z.); (T.S.S.); (B.M.); (J.T.K.J.); (B.D.); (Q.S.); (C.A.R.); (T.J.G.)
| | - Esther Zumaquero
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (R.G.K.); (J.N.P.); (D.B.); (M.D.S.); (S.Q.); (G.Y.); (F.Z.); (E.Z.); (T.S.S.); (B.M.); (J.T.K.J.); (B.D.); (Q.S.); (C.A.R.); (T.J.G.)
| | - Thomas S. Simpler
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (R.G.K.); (J.N.P.); (D.B.); (M.D.S.); (S.Q.); (G.Y.); (F.Z.); (E.Z.); (T.S.S.); (B.M.); (J.T.K.J.); (B.D.); (Q.S.); (C.A.R.); (T.J.G.)
| | - Betty Mousseau
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (R.G.K.); (J.N.P.); (D.B.); (M.D.S.); (S.Q.); (G.Y.); (F.Z.); (E.Z.); (T.S.S.); (B.M.); (J.T.K.J.); (B.D.); (Q.S.); (C.A.R.); (T.J.G.)
| | - John T. Killian
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (R.G.K.); (J.N.P.); (D.B.); (M.D.S.); (S.Q.); (G.Y.); (F.Z.); (E.Z.); (T.S.S.); (B.M.); (J.T.K.J.); (B.D.); (Q.S.); (C.A.R.); (T.J.G.)
| | - Brittany Dean
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (R.G.K.); (J.N.P.); (D.B.); (M.D.S.); (S.Q.); (G.Y.); (F.Z.); (E.Z.); (T.S.S.); (B.M.); (J.T.K.J.); (B.D.); (Q.S.); (C.A.R.); (T.J.G.)
| | - Qiao Shang
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (R.G.K.); (J.N.P.); (D.B.); (M.D.S.); (S.Q.); (G.Y.); (F.Z.); (E.Z.); (T.S.S.); (B.M.); (J.T.K.J.); (B.D.); (Q.S.); (C.A.R.); (T.J.G.)
| | - Jennifer L. Tipper
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (J.L.T.); (K.S.H.)
| | - Christopher A. Risley
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (R.G.K.); (J.N.P.); (D.B.); (M.D.S.); (S.Q.); (G.Y.); (F.Z.); (E.Z.); (T.S.S.); (B.M.); (J.T.K.J.); (B.D.); (Q.S.); (C.A.R.); (T.J.G.)
| | - Kevin S. Harrod
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (J.L.T.); (K.S.H.)
| | - Tsungwei Feng
- Altimmune Inc., Gaithersburg, MD 20878, USA; (T.F.); (Y.L.); (B.S.); (V.K.); (I.P.); (J.Z.); (J.J.S.); (B.G.)
| | - Young Lee
- Altimmune Inc., Gaithersburg, MD 20878, USA; (T.F.); (Y.L.); (B.S.); (V.K.); (I.P.); (J.Z.); (J.J.S.); (B.G.)
| | - Bethlehem Shiberu
- Altimmune Inc., Gaithersburg, MD 20878, USA; (T.F.); (Y.L.); (B.S.); (V.K.); (I.P.); (J.Z.); (J.J.S.); (B.G.)
| | - Vyjayanthi Krishnan
- Altimmune Inc., Gaithersburg, MD 20878, USA; (T.F.); (Y.L.); (B.S.); (V.K.); (I.P.); (J.Z.); (J.J.S.); (B.G.)
| | - Isabelle Peguillet
- Altimmune Inc., Gaithersburg, MD 20878, USA; (T.F.); (Y.L.); (B.S.); (V.K.); (I.P.); (J.Z.); (J.J.S.); (B.G.)
| | - Jianfeng Zhang
- Altimmune Inc., Gaithersburg, MD 20878, USA; (T.F.); (Y.L.); (B.S.); (V.K.); (I.P.); (J.Z.); (J.J.S.); (B.G.)
| | - Todd J. Green
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (R.G.K.); (J.N.P.); (D.B.); (M.D.S.); (S.Q.); (G.Y.); (F.Z.); (E.Z.); (T.S.S.); (B.M.); (J.T.K.J.); (B.D.); (Q.S.); (C.A.R.); (T.J.G.)
| | - Troy D. Randall
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (A.S.-S.); (S.M.-P.); (S.R.A.); (M.L.); (J.E.B.); (T.D.R.)
| | - John J. Suschak
- Altimmune Inc., Gaithersburg, MD 20878, USA; (T.F.); (Y.L.); (B.S.); (V.K.); (I.P.); (J.Z.); (J.J.S.); (B.G.)
| | - Bertrand Georges
- Altimmune Inc., Gaithersburg, MD 20878, USA; (T.F.); (Y.L.); (B.S.); (V.K.); (I.P.); (J.Z.); (J.J.S.); (B.G.)
| | - James D. Brien
- Department of Molecular Microbiology & Immunology, Saint Louis University, St. Louis, MO 63104, USA; (A.M.D.); (A.K.P.); (J.D.B.)
| | - Frances E. Lund
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (R.G.K.); (J.N.P.); (D.B.); (M.D.S.); (S.Q.); (G.Y.); (F.Z.); (E.Z.); (T.S.S.); (B.M.); (J.T.K.J.); (B.D.); (Q.S.); (C.A.R.); (T.J.G.)
| | - M. Scot Roberts
- Altimmune Inc., Gaithersburg, MD 20878, USA; (T.F.); (Y.L.); (B.S.); (V.K.); (I.P.); (J.Z.); (J.J.S.); (B.G.)
| |
Collapse
|
1694
|
Addeo A, Shah PK, Bordry N, Hudson RD, Albracht B, Di Marco M, Kaklamani V, Dietrich PY, Taylor BS, Simand PF, Patel D, Wang J, Labidi-Galy I, Fertani S, Leach RJ, Sandoval J, Mesa R, Lathrop K, Mach N, Shah DP. Immunogenicity of SARS-CoV-2 messenger RNA vaccines in patients with cancer. Cancer Cell 2021; 39:1091-1098.e2. [PMID: 34214473 PMCID: PMC8218532 DOI: 10.1016/j.ccell.2021.06.009] [Citation(s) in RCA: 168] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/14/2021] [Accepted: 06/15/2021] [Indexed: 12/12/2022]
Abstract
Patients with cancer experience a higher burden of SARS-CoV-2 infection, disease severity, complications, and mortality, than the general population. SARS-CoV-2 mRNA vaccines are highly effective in the general population; however, few data are available on their efficacy in patients with cancer. Using a prospective cohort, we assessed the seroconversion rates and anti-SARS-CoV-2 spike protein antibody titers following the first and second dose of BNT162b2 and mRNA-1273 SARS-CoV-2 vaccines in patients with cancer in US and Europe from January to April 2021. Among 131 patients, most (94%) achieved seroconversion after receipt of two vaccine doses. Seroconversion rates and antibody titers in patients with hematological malignancy were significantly lower than those with solid tumors. None of the patients with history of anti-CD-20 antibody in the 6 months before vaccination developed antibody response. Antibody titers were highest for clinical surveillance or endocrine therapy groups and lowest for cytotoxic chemotherapy or monoclonal antibody groups.
Collapse
Affiliation(s)
- Alfredo Addeo
- Department of Oncology, Geneva University Hospitals, University of Geneva, Swiss Cancer Center Leman, Switzerland.
| | - Pankil K Shah
- Mays Cancer Center at UT Health San Antonio MD Anderson, San Antonio, TX, USA
| | - Natacha Bordry
- Department of Oncology, Geneva University Hospitals, University of Geneva, Swiss Cancer Center Leman, Switzerland
| | - Robert D Hudson
- Mays Cancer Center at UT Health San Antonio MD Anderson, San Antonio, TX, USA
| | - Brenna Albracht
- Mays Cancer Center at UT Health San Antonio MD Anderson, San Antonio, TX, USA
| | - Mariagrazia Di Marco
- Department of Oncology, Geneva University Hospitals, University of Geneva, Swiss Cancer Center Leman, Switzerland
| | - Virginia Kaklamani
- Mays Cancer Center at UT Health San Antonio MD Anderson, San Antonio, TX, USA
| | - Pierre-Yves Dietrich
- Department of Oncology, Geneva University Hospitals, University of Geneva, Swiss Cancer Center Leman, Switzerland
| | - Barbara S Taylor
- Division of Infectious Diseases, Department of Medicine, Joe R. and Teresa Lozano Long School of Medicine, UT Health San Antonio, San Antonio, TX, USA
| | - Pierre-Francois Simand
- Department of Oncology, Geneva University Hospitals, University of Geneva, Swiss Cancer Center Leman, Switzerland
| | - Darpan Patel
- Mays Cancer Center at UT Health San Antonio MD Anderson, San Antonio, TX, USA
| | - Jing Wang
- Mays Cancer Center at UT Health San Antonio MD Anderson, San Antonio, TX, USA
| | - Intidhar Labidi-Galy
- Department of Oncology, Geneva University Hospitals, University of Geneva, Swiss Cancer Center Leman, Switzerland; Center of Translational Research in Onco-Hematology, Faculty of Medicine, University of Geneva, Swiss Cancer Center Leman, Geneva, Switzerland
| | - Sara Fertani
- Department of Oncology, Geneva University Hospitals, University of Geneva, Swiss Cancer Center Leman, Switzerland
| | - Robin J Leach
- Mays Cancer Center at UT Health San Antonio MD Anderson, San Antonio, TX, USA
| | - Jose Sandoval
- Department of Oncology, Geneva University Hospitals, University of Geneva, Swiss Cancer Center Leman, Switzerland
| | - Ruben Mesa
- Mays Cancer Center at UT Health San Antonio MD Anderson, San Antonio, TX, USA
| | - Kate Lathrop
- Mays Cancer Center at UT Health San Antonio MD Anderson, San Antonio, TX, USA
| | - Nicolas Mach
- Department of Oncology, Geneva University Hospitals, University of Geneva, Swiss Cancer Center Leman, Switzerland
| | - Dimpy P Shah
- Mays Cancer Center at UT Health San Antonio MD Anderson, San Antonio, TX, USA.
| |
Collapse
|
1695
|
North CM, Barczak A, Goldstein RH, Healy BC, Finkelstein DM, Ding DD, Kim A, Boucau J, Shaw B, Gilbert RF, Vyas T, Reynolds Z, Siddle KJ, MacInnis BL, Regan J, Flynn JP, Choudhary MC, Vyas JM, Laskowski K, Dighe AS, Lemieux JE, Li JZ, Baden LR, Siedner MJ, Woolley AE, Sacks CA. Determining the Incidence of Asymptomatic SARS-CoV-2 among Early Recipients of COVID-19 Vaccines: A Prospective Cohort Study of Healthcare Workers before, during and after Vaccination [DISCOVER-COVID-19]. Clin Infect Dis 2021; 74:1275-1278. [PMID: 34363462 PMCID: PMC8436402 DOI: 10.1093/cid/ciab643] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Indexed: 01/19/2023] Open
Abstract
The impact of coronavirus disease 2019 vaccination on viral characteristics of breakthrough infections is unknown. In this prospective cohort study, incidence of severe acute respiratory syndrome coronavirus 2 infection decreased following vaccination. Although asymptomatic positive tests were observed following vaccination, the higher cycle thresholds, repeat negative tests, and inability to culture virus raise questions about their clinical significance.
Collapse
Affiliation(s)
- Crystal M North
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Amy Barczak
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,The Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Robert H Goldstein
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Brian C Healy
- Harvard Medical School, Boston, MA, USA.,Department of Biostatistics, Massachusetts General Hospital, Boston, MA, USA
| | | | - Delaney D Ding
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Andy Kim
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Julie Boucau
- The Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Bennett Shaw
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Rebecca F Gilbert
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Tammy Vyas
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Zahra Reynolds
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | | | | | - James Regan
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - James P Flynn
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Manish C Choudhary
- Harvard Medical School, Boston, MA, USA.,Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Jatin M Vyas
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Karl Laskowski
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Anand S Dighe
- Harvard Medical School, Boston, MA, USA.,Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Jacob E Lemieux
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Jonathan Z Li
- Harvard Medical School, Boston, MA, USA.,Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Lindsey R Baden
- Harvard Medical School, Boston, MA, USA.,Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Mark J Siedner
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Ann E Woolley
- Harvard Medical School, Boston, MA, USA.,Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Chana A Sacks
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| |
Collapse
|
1696
|
Merritt-Genore H, Moosdorf R, Gillaspie E, Lother S, Engelman D, Ahmed S, Baciewicz FA, Grant MC, Milewski R, Cawcutt K, Hayanga JA, Chatterjee S, Arora RC. Perioperative Coronavirus Vaccination - Timing and Implications: A Guidance Document. Ann Thorac Surg 2021; 112:1707-1715. [PMID: 34370980 PMCID: PMC8349423 DOI: 10.1016/j.athoracsur.2021.07.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 11/13/2022]
Abstract
Cardiothoracic surgical patients are at risk of increased coronavirus disease severity. Several important factors influence the administration of the coronavirus disease vaccine in the perioperative period. This guidance statement outlines current information regarding vaccine types, summarizes recommendations regarding appropriate timing of administration, and provides information regarding side effects in the perioperative period for cardiac and thoracic surgical patients.
Collapse
Affiliation(s)
| | - Rainer Moosdorf
- Department for Cardiovascular Surgery, Phillips University, Marburg, Germany
| | - Erin Gillaspie
- Assistant Professor of Thoracic Surgery, Department of Thoracic Surgery, Vanderbilt University Medical Center, Nashville, TN
| | - Sylvain Lother
- Division of Critical Care and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba
| | - Daniel Engelman
- Heart and Vascular Program, Baystate Health and University of Massachusetts Medical School-Baystate, Springfield, Massachusetts
| | - Shahnur Ahmed
- School of Medicine, Wayne State University School of Medicine, Detroit, MI
| | - Frank A Baciewicz
- Professor of Cardiothoracic Surgery, Department of Surgery, Wayne State University, Detroit, MI
| | - Michael C Grant
- Associate Professor, Division of Cardiac Anesthesia, Surgical Critical Care and Acute Care Surgery, The Johns Hopkins University School of Medicine, Baltimore MD
| | - Rita Milewski
- Associate Professor of Surgery, Division of Cardiac Surgery; Yale University, New Haven, CT
| | - Kelly Cawcutt
- Assistant Professor, Division of Infectious Diseases & Pulmonary and Critical Care Medicine, University of Nebraska Medical Center, Omaha, NE
| | - J Awori Hayanga
- Professor, Department of Cardiovascular and Thoracic Surgery, West Virginia University School of Medicine, Morgantown, WV
| | - Subhasis Chatterjee
- Assistant Professor, Division of General and Cardiothoracic Surgery, Michael E DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX
| | - Rakesh C Arora
- Department of Surgery, Section of Cardiac Surgery, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | | |
Collapse
|
1697
|
Tada T, Zhou H, Samanovic MI, Dcosta BM, Cornelius A, Mulligan MJ, Landau NR. Comparison of Neutralizing Antibody Titers Elicited by mRNA and Adenoviral Vector Vaccine against SARS-CoV-2 Variants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021. [PMID: 34312623 DOI: 10.1101/2021.07.19.452771] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The increasing prevalence of SARS-CoV-2 variants has raised concerns regarding possible decreases in vaccine efficacy. Here, neutralizing antibody titers elicited by mRNA-based and an adenoviral vector-based vaccine against variant pseudotyped viruses were compared. BNT162b2 and mRNA-1273-elicited antibodies showed modest neutralization resistance against Beta, Delta, Delta plus and Lambda variants whereas Ad26.COV2.S-elicited antibodies from a significant fraction of vaccinated individuals were of low neutralizing titer (IC 50 <50). The data underscore the importance of surveillance for breakthrough infections that result in severe COVID-19 and suggest the benefit of a second immunization following Ad26.COV2.S to increase protection against the variants.
Collapse
|
1698
|
Francis AI, Ghany S, Gilkes T, Umakanthan S. Review of COVID-19 vaccine subtypes, efficacy and geographical distributions. Postgrad Med J 2021; 98:389-394. [PMID: 37066438 DOI: 10.1136/postgradmedj-2021-140654] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 07/04/2021] [Indexed: 12/11/2022]
Abstract
As of 1 May 2021, there have been 152 661 445 Covid-19 cases with 3 202 256 deaths globally. This pandemic led to the race to discover a vaccine to achieve herd immunity and curtail the damaging effects of Covid-19. This study aims to discuss the most recent WHO-approved Covid-19 vaccine subtypes, their status and geographical scheduled updates as of 4 May 2021. The keywords "Covid-19, Vaccines, Pfizer, BNT162b2, AstraZeneca, AZD1222, Moderna, mRNA-1273, Janssen, Ad26.COV2.S" were typed into PubMed. Thirty Two relevant PubMed articles were included in the study. The vaccines discussed are Pfizer/BNT162b2, Moderna Vaccine/mRNA1273, AstraZeneca/AZD122/ChAdOx1 n-CoV-19 and the Janssen vaccines/Ad26.COV2.S, as well as their platforms, trials, limitations and geographical distributions. As of 16 May 2021, the number of countries that have approved the use of the following vaccines is Pfizer in 85, Moderna in 46, Oxford/AstraZeneca in 101, and Janssen in 41.
Collapse
Affiliation(s)
- Andre Ian Francis
- Department of Clinical Medical Sciences, The Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago
| | - Saudah Ghany
- Department of Clinical Medical Sciences, The Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago
| | - Tia Gilkes
- Department of Clinical Medical Sciences, The Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago
| | - Srikanth Umakanthan
- Pathology unit, Department of Paraclinical Sciences, The University Of The West Indies, St. Augustine, Trinidad and Tobago
| |
Collapse
|
1699
|
Boschiero MN, Palamim CVC, Marson FAL. The hindrances to perform the COVID-19 vaccination in Brazil. Hum Vaccin Immunother 2021; 17:3989-4004. [PMID: 34353218 DOI: 10.1080/21645515.2021.1955607] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Brazil is one of the epicenters of COVID-19 pandemic and faces several hindrances to make his COVID-19 vaccination plan efficient. METHODS The Brazilian COVID-19 vaccination plan was evaluated and the hindrances to make the COVID-19 vaccination plan efficient were described and discussed. RESULTS High territorial extension might contribute to a delay on the COVID-19 vaccination, due to difficulty in delivering vaccines to furthest Brazilian states and to all the interior cities. The choice among the vaccines should be done based on the type of storage and must consider the transport conditions necessary to maintain its effectiveness. The indigenous individuals were included with health-care workers as the first group to be vaccinated, inflaming the number of vaccines doses distributed in states where the indigenous population have higher prevalence. The antivaccine movement and the politicization of the vaccine are also hindrances to be overcome in Brazil. The COVID-19 incidence or mortality rate and the distribution of intensive care units (ICUs) are not a criterion to distribute the vaccines, as we did not identify a correlation between these markers and the number of vaccines. However, a strong or very strong correlation occurred between the number of COVID-19 vaccines and the number of COVID-19 cases, deaths by COVID-19, gross domestic product, as well as populational density. A total of 83,280,475 doses of COVID-19 vaccines were distributed in Brazil. In the first dose, the Coronavac (Sinovac™), AZD1222 (AstraZeneca/Oxford™), and BNT162b (Pfizer/BioNTech™) vaccines were responsible to vaccinate, respectively, 9.61%, 6.69%, and 0.35% of the Brazilian population. In the second dose, the Coronavac, AZD1222, and BNT162b vaccines were responsible to vaccinate, respectively, 7.52%, 0.53%, and <0.01% of the Brazilian population. CONCLUSIONS The Federal Government must evaluate the hindrances and propose solutions to maximize the immunization against COVID-19 on Brazil.
Collapse
Affiliation(s)
- Matheus Negri Boschiero
- Laboratory of Cell and Molecular Tumor Biology and Bioactive Compounds, São Francisco University, Bragança Paulista, SP, Brazil.,Laboratory of Human and Medical Genetics, São Francisco University, Bragança Paulista, SP, Brazil
| | - Camila Vantini Capasso Palamim
- Laboratory of Cell and Molecular Tumor Biology and Bioactive Compounds, São Francisco University, Bragança Paulista, SP, Brazil.,Laboratory of Human and Medical Genetics, São Francisco University, Bragança Paulista, SP, Brazil
| | - Fernando Augusto Lima Marson
- Laboratory of Cell and Molecular Tumor Biology and Bioactive Compounds, São Francisco University, Bragança Paulista, SP, Brazil.,Laboratory of Human and Medical Genetics, São Francisco University, Bragança Paulista, SP, Brazil
| |
Collapse
|
1700
|
Expert Perspectives on COVID-19 Vaccination for People Living with Multiple Sclerosis. Neurol Ther 2021; 10:415-425. [PMID: 34347280 PMCID: PMC8336531 DOI: 10.1007/s40120-021-00266-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 07/08/2021] [Indexed: 11/25/2022] Open
Abstract
COVID-19 vaccines are safe for people living with MS on or off disease-modifying therapies and are important for the prevention of COVID-19. Antibody responses for individuals on certain DMTs may be diminished, however, T-cell responses may be preserved in those individuals. Data are lacking regarding optimal timing of vaccinations, and delaying disease-modifying therapies may increase the risk of disease activity and progression. In this perspective podcast, the authors recommend COVID-19 vaccination as soon as possible, regardless of timing considerations, in most cases. ![]()
Collapse
|