1501
|
Pneumococcal Vaccines: Past Findings, Present Work, and Future Strategies. Vaccines (Basel) 2021; 9:vaccines9111338. [PMID: 34835269 PMCID: PMC8620834 DOI: 10.3390/vaccines9111338] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/12/2021] [Accepted: 11/13/2021] [Indexed: 01/24/2023] Open
Abstract
The importance of Streptococcus pneumoniae has been well established. These bacteria can colonize infants and adults without symptoms, but in some cases can spread, invade other tissues and cause disease with high morbidity and mortality. The development of pneumococcal conjugate vaccines (PCV) caused an enormous impact in invasive pneumococcal disease and protected unvaccinated people by herd effect. However, serotype replacement is a well-known phenomenon that has occurred after the introduction of the 7-valent pneumococcal conjugate vaccine (PCV7) and has also been reported for other PCVs. Therefore, it is possible that serotype replacement will continue to occur even with higher valence formulations, but the development of serotype-independent vaccines might overcome this problem. Alternative vaccines are under development in order to improve cost effectiveness, either using proteins or the pneumococcal whole cell. These approaches can be used as a stand-alone strategy or together with polysaccharide vaccines. Looking ahead, the next generation of pneumococcal vaccines can be impacted by the new technologies recently approved for human use, such as mRNA vaccines and viral vectors. In this paper, we will review the advantages and disadvantages of the addition of new polysaccharides in the current PCVs, mainly for low- and middle-income countries, and we will also address future perspectives.
Collapse
|
1502
|
Hagan K, Forman R, Mossialos E, Ndebele P, Hyder AA, Nasir K. COVID-19 vaccine mandate for healthcare workers in the United States: a social justice policy. Expert Rev Vaccines 2021; 21:37-45. [PMID: 34709969 DOI: 10.1080/14760584.2022.1999811] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Vaccination is the most effective strategy to mitigating COVID-19 and restoring societal function. As the pandemic evolves with no certainty of a herd immunity threshold, universal vaccination of at-risk populations is desirable. However, vaccine hesitancy threatens the return to normalcy, and healthcare workers (HCWs) must embrace their ambassadorial role of shoring up vaccine confidence. Unfortunately, voluntary vaccination has been suboptimal among HCWs in the United States, a priority group for whom immunization is essential for maintaining health system capacity and the safety of high-risk patients in their care. Consequently, some health systems have implemented mandates to improve compliance. AREAS COVERED This article discusses the ethical and practical considerations of mandatory COVID-19 vaccination policies for HCWs utilizing some components of the World Health Organization's framework and the unique context of a pandemic with evolving infection dynamics. EXPERT OPINION COVID-19 vaccine mandates for universal immunization of HCWs raise ethical and practical debates about their appropriateness, especially when the vaccines are pending full approval in most jurisdictions. Given the superiority of the vaccines to safety and testing protocols and their favorable safety profile, we encourage health systems to adopt vaccination mandates through participatory processes that address the concerns of stakeholders.
Collapse
Affiliation(s)
- K Hagan
- Division of Health Equity & Disparities Research, Center for Outcomes Research, Houston, TX, USA
| | - R Forman
- Department of Health Policy, London School of Economics and Political Sciences, UK
| | - Elias Mossialos
- Department of Health Policy, London School of Economics and Political Sciences, UK.,Center for Health Policy, Imperial College London, UK
| | - Paul Ndebele
- Department of Global Health, The George Washington University, Washington, DC, USA
| | - Adnan A Hyder
- Center on Commercial Determinants of Health, Milken Institute School of Public Health, The George Washington University, Washington, DC, USA
| | - Khurram Nasir
- Division of Health Equity & Disparities Research, Center for Outcomes Research, Houston, TX, USA.,Division of Cardiovascular Prevention and Wellness, Department of Cardiology, Houston Methodist DeBakey Heart & Vascular Center, Houston, TX, USA.,Center for Cardiovascular Computational & Precision Health (C3-PH), Houston Methodist DeBakey Heart & Vascular Center, Houston, TX, USA
| |
Collapse
|
1503
|
Feghali EJ, Zafar M, Abid S, Santoriello D, Mehta S. De-Novo Antineutrophil Cytoplasmic Antibody-Associated Vasculitis Following the mRNA-1273 (Moderna) Vaccine for COVID-19. Cureus 2021; 13:e19616. [PMID: 34956751 PMCID: PMC8675571 DOI: 10.7759/cureus.19616] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2021] [Indexed: 11/05/2022] Open
Abstract
Antineutrophil cytoplasmic antibody (ANCA) is a systemic autoimmune disorder characterized by antibodies directed against small- and moderate-sized vessels. While there are few reported cases of autoimmune illnesses associated with influenza vaccination, two cases of de-novo anti-proteinase (PR3) ANCA-associated pauci immune glomerulonephritis are reported after the mRNA-1273 coronavirus disease 2019 (COVID-19) vaccine. Here, we report the third case of ANCA-associated glomerulonephritis after the mRNA-1273 COVID-19 vaccine. Our patient presented with acute kidney injury and sub-nephrotic proteinuria four days after receiving the second dose of the COVID vaccine. He was found to have elevated c-ANCA and anti-PR3 antibodies. Renal biopsy confirmed focal necrotizing and diffuse crescentic glomerulonephritis. He was diagnosed with pauci immune glomerulonephritis. The patient achieved remission 10 weeks after the diagnosis with successful treatment.
Collapse
|
1504
|
Chen J, Vitetta L, Henson JD, Hall S. The intestinal microbiota and improving the efficacy of COVID-19 vaccinations. J Funct Foods 2021; 87:104850. [PMID: 34777578 PMCID: PMC8578005 DOI: 10.1016/j.jff.2021.104850] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 10/31/2021] [Accepted: 11/06/2021] [Indexed: 02/07/2023] Open
Abstract
Most COVID-19 cases are mild or asymptomatic and recover well, suggesting that effective immune responses ensue, which successfully eliminate SARS-CoV-2 viruses. However, a small proportion of patients develop severe COVID-19 with pathological immune responses. This indicates that a strong immune system balanced with anti-inflammatory mechanisms is critical for the recovery from SARS-CoV-2 infections. Many vaccines against SARS-CoV-2 have now been developed for eliciting effective immune responses to protect from SARS-CoV-2 infections or reduce the severity of the disease if infected. Although uncommon, serious morbidity and mortality have resulted from both COVID-19 vaccine adverse reactions and lack of efficacy, and further improvement of efficacy and prevention of adverse effects are urgently warranted. Many factors could affect efficacy of these vaccines to achieve optimal immune responses. Dysregulation of the gut microbiota (gut dysbiosis) could be an important risk factor as the gut microbiota is associated with the development and maintenance of an effective immune system response. In this narrative review, we discuss the immune responses to SARS-CoV-2, how COVID-19 vaccines elicit protective immune responses, gut dysbiosis involvement in inefficacy and adverse effects of COVID-19 vaccines and the modulation of the gut microbiota by functional foods to improve COVID-19 vaccine immunisations.
Collapse
Affiliation(s)
- Jiezhong Chen
- Medlab Clinical, Department of Research, Sydney 2015, Australia
| | - Luis Vitetta
- Medlab Clinical, Department of Research, Sydney 2015, Australia.,The University of Sydney, Faculty of Medicine and Health, Sydney 2006, Australia
| | - Jeremy D Henson
- Medlab Clinical, Department of Research, Sydney 2015, Australia.,The University of New South Wales, Faculty of Medicine, Prince of Wales Clinical School, Sydney, Australia
| | - Sean Hall
- Medlab Clinical, Department of Research, Sydney 2015, Australia
| |
Collapse
|
1505
|
Hendren NS, Carter S, Grodin JL. Severe COVID-19 vaccine associated myocarditis: Zebra or unicorn? Int J Cardiol 2021; 343:197-198. [PMID: 34560165 PMCID: PMC8453875 DOI: 10.1016/j.ijcard.2021.09.036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 09/17/2021] [Indexed: 12/29/2022]
Affiliation(s)
- Nicholas S Hendren
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA; Parkland Health and Hospital System, Dallas, TX, USA
| | - Spencer Carter
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA; Parkland Health and Hospital System, Dallas, TX, USA
| | - Justin L Grodin
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA; Parkland Health and Hospital System, Dallas, TX, USA.
| |
Collapse
|
1506
|
Heidari S, Palmer-Ross A, Goodman T. A Systematic Review of the Sex and Gender Reporting in COVID-19 Clinical Trials. Vaccines (Basel) 2021; 9:1322. [PMID: 34835253 PMCID: PMC8622702 DOI: 10.3390/vaccines9111322] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/02/2021] [Accepted: 11/09/2021] [Indexed: 01/12/2023] Open
Abstract
Sex and gender have implications for COVID-19 vaccine efficacy and adverse effects from the vaccine. As vaccination is one of the key responses to the COVID-19 pandemic, it is vital that sex and gender differences be acknowledged, measured, and analysed in clinical research. Here, we systematically review published COVID-19 vaccine trials, both interventional and observational, to assess the quality of reporting of sex and gender. Of the 75 clinical trials on COVID-19 vaccines included in this review, only 24% presented their main outcome data disaggregated by sex, and only 13% included any discussion of the implications of their study for women and men. Considering the sex differences in adverse events after vaccination, and the gendered aspects of vaccine hesitancy, these oversights in clinical research on vaccines have implications for recovery from the COVID-19 pandemic and for wider public health.
Collapse
Affiliation(s)
- Shirin Heidari
- Gender Equity and Human Rights, World Health Organisation, 1211 Geneva, Switzerland
| | | | | |
Collapse
|
1507
|
Uaprasert N, Panrong K, Rojnuckarin P, Chiasakul T. Thromboembolic and hemorrhagic risks after vaccination against SARS-CoV-2: a systematic review and meta-analysis of randomized controlled trials. Thromb J 2021; 19:86. [PMID: 34774069 PMCID: PMC8590131 DOI: 10.1186/s12959-021-00340-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 10/27/2021] [Indexed: 01/12/2023] Open
Abstract
Background Thromboembolic and bleeding events after vaccination against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are major public concerns leading to vaccine hesitancy. Due to low incidence, an individual randomized controlled trial (RCT) is underpowered to determine whether SARS-CoV-2 vaccines increase the risks of thromboembolism and hemorrhage. Methods We performed a literature search using PubMed, EMBASE, Cochrane, medRxiv databases, and reference lists of relevant articles to identify RCTs that reported thromboembolic, hemorrhagic events, and thromboembolism/hemorrhage-related death after SARS-CoV-2 vaccination. The primary aim of this systematic review and meta-analysis was to estimate the pooled thromboembolic risk related to SARS-CoV-2 vaccines compared to placebo. The secondary outcomes included estimating the risks of arterial thromboembolism (ATE), venous thromboembolisms (VTE), hemorrhage, thrombocytopenia, and thromboembolism/hemorrhage-related death. Results Eight RCTs of 4 vaccine platforms comprised of 195,196 participants were retrieved. SARS-CoV-2 vaccines were not associated with an increased risk of overall thromboembolism (risk ratio [RR], 1.14; 95% CI [confidence interval], 0.61–2.14; I2 = 35%), ATE (RR, 0.97; 95% CI, 0.46–2.06; I2 = 21%), VTE (RR, 1.47; 95% CI, 0.72–2.99; I2 = 0%), hemorrhage (RR, 0.97; 95% CI, 0.35–2.68; I2 = 0), and thromboembolism/hemorrhage-related death (RR, 0.53; 95% CI, 0.16–1.79; I2 = 0). Compared to the baseline estimated risk of these outcomes in participants administered placebos, the risk differences with vaccines were very small and not statistically significant. These findings were consistent in the subgroup analysis across 4 vaccine platforms. Conclusion Vaccines against SARS-CoV-2 are not associated with an increased risk of thromboembolism, hemorrhage, and thromboembolism/hemorrhage-related death. Supplementary Information The online version contains supplementary material available at 10.1186/s12959-021-00340-4.
Collapse
Affiliation(s)
- Noppacharn Uaprasert
- Division of Hematology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand. .,Research Unit in Translational Hematology, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand.
| | - Krissana Panrong
- Division of Hematology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand.,Research Unit in Translational Hematology, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Ponlapat Rojnuckarin
- Division of Hematology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand.,Research Unit in Translational Hematology, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Thita Chiasakul
- Division of Hematology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand.,Research Unit in Translational Hematology, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| |
Collapse
|
1508
|
Gross hematuria after SARS-CoV-2 vaccination: questionnaire survey in Japan. Clin Exp Nephrol 2021; 26:316-322. [PMID: 34773533 PMCID: PMC8590432 DOI: 10.1007/s10157-021-02157-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/04/2021] [Indexed: 11/25/2022]
Abstract
Background Recent clinical reports indicate a correlation between gross hematuria after the coronavirus 2019 (COVID-19) vaccination in patients with glomerulonephritis, especially immunoglobulin A nephropathy (IgAN). Furthermore, healthcare workers in Japan were initially vaccinated with an mRNA vaccine from February 17, 2021, and some of them experienced gross hematuria after receiving the vaccination. Methods We conducted a web-based survey of the councilor members of the Japanese Society of Nephrology (581 members, 382 facilities) to elucidate the relationship between gross hematuria and COVID-19 vaccination. Results In the first survey, 27 cases (female: 22, 81.5%) of gross hematuria were reported after receiving a COVID-19 vaccination. Of them, 19 (70.4%) patients were already diagnosed with IgAN at the occurrence of gross hematuria. Proteinuria appeared in eight of the 14 (57.1%) cases with no proteinuria before vaccination and hematuria in five of the seven (71.4%) cases with no hematuria before vaccination. The second survey revealed that a renal biopsy was performed after vaccination in four cases, all of whom were diagnosed with IgAN. Only one case showed a slightly increased serum creatinine level, and no patients progressed to severe renal dysfunction. Conclusion This study clarified the clinical features of gross hematuria after a COVID-19 vaccination. Because there was no obvious progression to severe renal dysfunction, safety of the COVID-19 vaccination is warranted at least in the protocol of inoculation twice. Supplementary Information The online version contains supplementary material available at 10.1007/s10157-021-02157-x.
Collapse
|
1509
|
Reddy KP, Fitzmaurice KP, Scott JA, Harling G, Lessells RJ, Panella C, Shebl FM, Freedberg KA, Siedner MJ. Clinical outcomes and cost-effectiveness of COVID-19 vaccination in South Africa. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.05.07.21256852. [PMID: 34013291 PMCID: PMC8132265 DOI: 10.1101/2021.05.07.21256852] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Low- and middle-income countries are implementing COVID-19 vaccination strategies in light of varying vaccine efficacies and costs, supply shortages, and resource constraints. Here, we use a microsimulation model to evaluate clinical outcomes and cost-effectiveness of a COVID-19 vaccination program in South Africa. We varied vaccination coverage, pace, acceptance, effectiveness, and cost as well as epidemic dynamics. Providing vaccines to at least 40% of the population and prioritizing vaccine rollout prevented >9 million infections and >73,000 deaths and reduced costs due to fewer hospitalizations. Model results were most sensitive to assumptions about epidemic growth and prevalence of prior immunity to SARS-CoV-2, though the vaccination program still provided high value and decreased both deaths and health care costs across a wide range of assumptions. Vaccination program implementation factors, including prompt procurement, distribution, and rollout, are likely more influential than characteristics of the vaccine itself in maximizing public health benefits and economic efficiency.
Collapse
Affiliation(s)
- Krishna P. Reddy
- Medical Practice Evaluation Center, Massachusetts General Hospital, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | - Justine A. Scott
- Medical Practice Evaluation Center, Massachusetts General Hospital, Boston, MA, USA
| | - Guy Harling
- Africa Health Research Institute, KwaZulu-Natal, South Africa
- MRC/Wits Rural Public Health & Health Transitions Research Unit (Agincourt), University of the Witwatersrand, South Africa
- School of Nursing & Public Health, College of Health Sciences, University of KwaZulu-Natal, KwaZulu-Natal, South Africa
- Institute for Global Health, University College London, London, UK
- Department of Epidemiology and Harvard Center for Population & Development Studies, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Richard J. Lessells
- KwaZulu-Natal Research Innovation and Sequencing (KRISP), College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Christopher Panella
- Medical Practice Evaluation Center, Massachusetts General Hospital, Boston, MA, USA
| | - Fatma M. Shebl
- Medical Practice Evaluation Center, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Kenneth A. Freedberg
- Medical Practice Evaluation Center, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA, USA
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA
- Department of Health Policy and Management, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Mark J. Siedner
- Medical Practice Evaluation Center, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Africa Health Research Institute, KwaZulu-Natal, South Africa
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
1510
|
Nishizawa Y, Hoshina Y, Baker V. Bell's palsy following the Ad26.COV2.S COVID-19 vaccination. QJM 2021; 114:657-658. [PMID: 34014316 PMCID: PMC8244554 DOI: 10.1093/qjmed/hcab143] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 05/13/2021] [Indexed: 02/06/2023] Open
Affiliation(s)
- Yoshiki Nishizawa
- Department of Neurology, U.S. Naval Hospital
Yokosuka, 82, Inaoka-Cho, Yokosuka-City, Kanagawa, 238-0001,
Japan
| | - Yoji Hoshina
- Department of Neurology, U.S. Naval Hospital
Yokosuka, 82, Inaoka-Cho, Yokosuka-City, Kanagawa, 238-0001,
Japan
- Address correspondence to author: Yoji Hoshina, MD,
Department of Neurology, U.S. Naval Hospital Yokosuka, 82, Inaoka-cho,
Yokosuka-city, Kanagawa, Japan, Tel: +81-46-816-7144,
| | - Virginia Baker
- Department of Neurology, U.S. Naval Hospital
Yokosuka, 82, Inaoka-Cho, Yokosuka-City, Kanagawa, 238-0001,
Japan
| |
Collapse
|
1511
|
Tang W, Gartshteyn Y, Ricker E, Inzerillo S, Murray S, Khalili L, Askanase A. The Use of COVID-19 Vaccines in Patients with SLE. Curr Rheumatol Rep 2021; 23:79. [PMID: 34767100 PMCID: PMC8586600 DOI: 10.1007/s11926-021-01046-2] [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] [Accepted: 09/27/2021] [Indexed: 02/06/2023]
Abstract
Purpose of Review Three COVID-19 vaccines obtained emergency authorization from the Food and Drug Administration (FDA) and are widely used in the USA. Unfortunately, there is a paucity of evidence on the safety and efficacy of these vaccines in patients with autoimmune inflammatory rheumatic diseases (AIIRD), as these patients were excluded from all phases of vaccine development. Here we reviewed current data on COVID-19 vaccination in patients with AIIRD, with emphasis on systemic lupus erythematosus (SLE), and provided a comprehensive update on the benefits and risks of vaccination. Recent Findings Patients with SLE have worse immune responses following SARS-CoV-2 vaccination than healthy controls. The efficacy of the COVID-19 vaccines seems to be further reduced by immunosuppressive medications, such as glucocorticoids (GC), methotrexate (MTX), mycophenolate/mycophenolic acid (MMF), and rituximab (RTX). However, these data do not substantiate that AIIRD patients are at greater risk of disease flares or have a higher incidence of side effects following vaccination. There is no significant safety concern for the use of COVID-19 vaccines in patients with AIIRD. Summary The benefits of vaccination far outweigh the risks in patients with AIIRD, including SLE. More data are needed to determine the necessity of a booster vaccine dose and appropriate adjustment of immunosuppressants around the administration of vaccine.
Collapse
Affiliation(s)
- Wei Tang
- Division of Rheumatology, Department of Medicine, Columbia University Irving Medical Center, 630 West 168th Street, P&S 10-508, New York, NY, 10032, USA
| | - Yevgeniya Gartshteyn
- Division of Rheumatology, Department of Medicine, Columbia University Irving Medical Center, 630 West 168th Street, P&S 10-508, New York, NY, 10032, USA
| | - Edd Ricker
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Sean Inzerillo
- Division of Rheumatology, Department of Medicine, Columbia University Irving Medical Center, 630 West 168th Street, P&S 10-508, New York, NY, 10032, USA
| | - Shane Murray
- Division of Rheumatology, Department of Medicine, Columbia University Irving Medical Center, 630 West 168th Street, P&S 10-508, New York, NY, 10032, USA
| | - Leila Khalili
- Division of Rheumatology, Department of Medicine, Columbia University Irving Medical Center, 630 West 168th Street, P&S 10-508, New York, NY, 10032, USA
| | - Anca Askanase
- Division of Rheumatology, Department of Medicine, Columbia University Irving Medical Center, 630 West 168th Street, P&S 10-508, New York, NY, 10032, USA.
| |
Collapse
|
1512
|
Ben-Mayor Bashi T, Amikam U, Ashwal E, Hershkovitz G, Attali E, Berkovitz-Shperling R, Dominsky O, Halperin T, Goldshmidt H, Gamzu R, Yogev Y, Kuperminc M, Hiersch L. The association of maternal SARS-CoV-2 vaccination-to-delivery interval and the levels of maternal and cord blood antibodies. Int J Gynaecol Obstet 2021; 156:436-443. [PMID: 34762739 PMCID: PMC9087624 DOI: 10.1002/ijgo.14014] [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: 08/13/2021] [Revised: 11/16/2021] [Accepted: 11/09/2021] [Indexed: 12/25/2022]
Abstract
OBJECTIVE To evaluate the correlation of maternal and cord blood levels of SARS-CoV-2 antibodies in pregnant women immunized against COVID-19. METHODS A prospective cohort study was performed of pregnant women who delivered at a single university affiliated tertiary medical center. Women who received the COVID-19 vaccine (BNT162b2 Pfizer©) were approached. The correlation between levels of maternal sera and umbilical cord SARS-CoV-2 specific IgG was assessed. RESULTS Overall, 58 women were included; of them, 19 had received a single dose and 39 received two doses of the COVID-19 vaccine. Positive levels of umbilical cord IgG were found in 13/19 (68.4%) and 38/39 (97.4%) women after the administration of a single dose and two doses of the vaccine, respectively. The levels of SARS-CoV-2 IgG antibodies in the maternal sera of vaccinated women were positively correlated to their respective concentrations in cord blood sera (ρ = 0.857; R2 linear = 0.719; P < 0.001). Thirteen days after vaccination, the ratio of maternal-to-umbilical cord anti Spike IgG antibodies was approximately 1, indicating relatively similar levels in maternal and cord sera. CONCLUSION After the SARS-CoV-2 vaccine, levels of maternal and cord blood antibodies were positively correlated, especially when tested after 13 days following administration of the first dose of the vaccine.
Collapse
Affiliation(s)
- Tali Ben-Mayor Bashi
- Department of Obstetrics and Gynecology, Lis Maternity and Women's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Uri Amikam
- Department of Obstetrics and Gynecology, Lis Maternity and Women's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eran Ashwal
- Department of Obstetrics and Gynecology, Lis Maternity and Women's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Gal Hershkovitz
- Department of Obstetrics and Gynecology, Lis Maternity and Women's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Emmanuel Attali
- Department of Obstetrics and Gynecology, Lis Maternity and Women's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Roza Berkovitz-Shperling
- Department of Obstetrics and Gynecology, Lis Maternity and Women's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Omri Dominsky
- Department of Obstetrics and Gynecology, Lis Maternity and Women's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tami Halperin
- Laboratory Medicine Division, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Hanoch Goldshmidt
- Laboratory Medicine Division, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Ronni Gamzu
- Department of Obstetrics and Gynecology, Lis Maternity and Women's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yariv Yogev
- Department of Obstetrics and Gynecology, Lis Maternity and Women's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Michael Kuperminc
- Department of Obstetrics and Gynecology, Lis Maternity and Women's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Liran Hiersch
- Department of Obstetrics and Gynecology, Lis Maternity and Women's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
1513
|
Briggs FBS, Mateen FJ, Schmidt H, Currie KM, Siefers HM, Crouthamel S, Bebo BF, Fiol J, Racke MK, O'Connor KC, Kolaczkowski LG, Klein P, Loud S, McBurney RN. COVID-19 Vaccination Reactogenicity in Persons With Multiple Sclerosis. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 9:9/1/e1104. [PMID: 34753828 PMCID: PMC8579248 DOI: 10.1212/nxi.0000000000001104] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 09/21/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND OBJECTIVES There are limited data on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine reactogenicity in persons with multiple sclerosis (PwMS) and how reactogenicity is affected by disease-modifying therapies (DMTs). The objective of this retrospective cross-sectional study was to generate real-world multiple sclerosis-specific vaccine safety information, particularly in the context of specific DMTs, and provide information to mitigate specific concerns in vaccine hesitant PwMS. METHODS Between 3/2021 and 6/2021, participants in iConquerMS, an online people-powered research network, reported SARS-CoV-2 vaccines, experiences of local (itch, pain, redness, swelling, or warmth at injection site) and systemic (fever, chills, fatigue, headache, joint pain, malaise, muscle ache, nausea, allergic, and other) reactions within 24 hours (none, mild, moderate, and severe), DMT use, and other attributes. Multivariable models characterized associations between clinical factors and reactogenicity. RESULTS In 719 PwMS, 64% reported experiencing a reaction after their first vaccination shot, and 17% reported a severe reaction. The most common reactions were pain at injection site (54%), fatigue (34%), headache (28%), and malaise (21%). Younger age, being female, prior SARS-CoV-2 infection, and receiving the ChAdOx1 nCoV-19 (Oxford-AstraZeneca) vs BNT162b2 (Pfizer-BioNTech) vaccine were associated with experiencing a reaction after the first vaccine dose. Similar relationships were observed for a severe reaction, including higher odds of reactions among PwMS with more physical impairment and lower odds of reactions for PwMS on an alpha4-integrin blocker or sphingosine-1-phosphate receptor modulator. In 442 PwMS who received their second vaccination shot, 74% reported experiencing a reaction, whereas 22% reported a severe reaction. Reaction profiles after the second shot were similar to those reported after the first shot. Younger PwMS and those who received the mRNA-1273 (Moderna) vs BNT162b2 vaccine reported higher reactogenicity after the second shot, whereas those on a sphingosine-1-phosphate receptor modulator or fumarate were significantly less likely to report a reaction. DISCUSSION SARS-CoV-2 vaccine reactogenicity profiles and the associated factors in this convenience sample of PwMS appear similar to those reported in the general population. PwMS on specific DMTs were less likely to report vaccine reactions. Overall, the short-term vaccine reactions experienced in the study population were mostly self-limiting, including pain at the injection site, fatigue, headache, and fever.
Collapse
Affiliation(s)
- Farren Basil Shaw Briggs
- From the Department of Population and Quantitative Health Science (F.B.S.B.), School of Medicine, Cleveland, OH; Department of Neurology (F.J.M.), Massachusetts General Hospital, Boston; Accelerated Cure Project for MS (H.S., S.L., R.N.M.), Waltham, MA; Currie Consultancy (K.M.C.), LLC Eastover, SC; International AIDS Vaccine Initiative (H.M.S.), Frederick, MD; Mammoth Hospital (S.C.), Mammoth Lakes, CA; National Multiple Sclerosis Society (B.F.B., J.F.); Medical Affairs (M.K.R.), Quest Diagnostics, Secaucus, NJ; Departments of Neurology and Immunobiology (K.C.O.), Yale University School of Medicine, New Haven, CT; and iConquerMS (L.G.K., P.K.), Waltham, MA.
| | - Farrah J Mateen
- From the Department of Population and Quantitative Health Science (F.B.S.B.), School of Medicine, Cleveland, OH; Department of Neurology (F.J.M.), Massachusetts General Hospital, Boston; Accelerated Cure Project for MS (H.S., S.L., R.N.M.), Waltham, MA; Currie Consultancy (K.M.C.), LLC Eastover, SC; International AIDS Vaccine Initiative (H.M.S.), Frederick, MD; Mammoth Hospital (S.C.), Mammoth Lakes, CA; National Multiple Sclerosis Society (B.F.B., J.F.); Medical Affairs (M.K.R.), Quest Diagnostics, Secaucus, NJ; Departments of Neurology and Immunobiology (K.C.O.), Yale University School of Medicine, New Haven, CT; and iConquerMS (L.G.K., P.K.), Waltham, MA
| | - Hollie Schmidt
- From the Department of Population and Quantitative Health Science (F.B.S.B.), School of Medicine, Cleveland, OH; Department of Neurology (F.J.M.), Massachusetts General Hospital, Boston; Accelerated Cure Project for MS (H.S., S.L., R.N.M.), Waltham, MA; Currie Consultancy (K.M.C.), LLC Eastover, SC; International AIDS Vaccine Initiative (H.M.S.), Frederick, MD; Mammoth Hospital (S.C.), Mammoth Lakes, CA; National Multiple Sclerosis Society (B.F.B., J.F.); Medical Affairs (M.K.R.), Quest Diagnostics, Secaucus, NJ; Departments of Neurology and Immunobiology (K.C.O.), Yale University School of Medicine, New Haven, CT; and iConquerMS (L.G.K., P.K.), Waltham, MA
| | - Keisha M Currie
- From the Department of Population and Quantitative Health Science (F.B.S.B.), School of Medicine, Cleveland, OH; Department of Neurology (F.J.M.), Massachusetts General Hospital, Boston; Accelerated Cure Project for MS (H.S., S.L., R.N.M.), Waltham, MA; Currie Consultancy (K.M.C.), LLC Eastover, SC; International AIDS Vaccine Initiative (H.M.S.), Frederick, MD; Mammoth Hospital (S.C.), Mammoth Lakes, CA; National Multiple Sclerosis Society (B.F.B., J.F.); Medical Affairs (M.K.R.), Quest Diagnostics, Secaucus, NJ; Departments of Neurology and Immunobiology (K.C.O.), Yale University School of Medicine, New Haven, CT; and iConquerMS (L.G.K., P.K.), Waltham, MA
| | - Heather M Siefers
- From the Department of Population and Quantitative Health Science (F.B.S.B.), School of Medicine, Cleveland, OH; Department of Neurology (F.J.M.), Massachusetts General Hospital, Boston; Accelerated Cure Project for MS (H.S., S.L., R.N.M.), Waltham, MA; Currie Consultancy (K.M.C.), LLC Eastover, SC; International AIDS Vaccine Initiative (H.M.S.), Frederick, MD; Mammoth Hospital (S.C.), Mammoth Lakes, CA; National Multiple Sclerosis Society (B.F.B., J.F.); Medical Affairs (M.K.R.), Quest Diagnostics, Secaucus, NJ; Departments of Neurology and Immunobiology (K.C.O.), Yale University School of Medicine, New Haven, CT; and iConquerMS (L.G.K., P.K.), Waltham, MA
| | - Slavka Crouthamel
- From the Department of Population and Quantitative Health Science (F.B.S.B.), School of Medicine, Cleveland, OH; Department of Neurology (F.J.M.), Massachusetts General Hospital, Boston; Accelerated Cure Project for MS (H.S., S.L., R.N.M.), Waltham, MA; Currie Consultancy (K.M.C.), LLC Eastover, SC; International AIDS Vaccine Initiative (H.M.S.), Frederick, MD; Mammoth Hospital (S.C.), Mammoth Lakes, CA; National Multiple Sclerosis Society (B.F.B., J.F.); Medical Affairs (M.K.R.), Quest Diagnostics, Secaucus, NJ; Departments of Neurology and Immunobiology (K.C.O.), Yale University School of Medicine, New Haven, CT; and iConquerMS (L.G.K., P.K.), Waltham, MA
| | - Bruce F Bebo
- From the Department of Population and Quantitative Health Science (F.B.S.B.), School of Medicine, Cleveland, OH; Department of Neurology (F.J.M.), Massachusetts General Hospital, Boston; Accelerated Cure Project for MS (H.S., S.L., R.N.M.), Waltham, MA; Currie Consultancy (K.M.C.), LLC Eastover, SC; International AIDS Vaccine Initiative (H.M.S.), Frederick, MD; Mammoth Hospital (S.C.), Mammoth Lakes, CA; National Multiple Sclerosis Society (B.F.B., J.F.); Medical Affairs (M.K.R.), Quest Diagnostics, Secaucus, NJ; Departments of Neurology and Immunobiology (K.C.O.), Yale University School of Medicine, New Haven, CT; and iConquerMS (L.G.K., P.K.), Waltham, MA
| | - Julie Fiol
- From the Department of Population and Quantitative Health Science (F.B.S.B.), School of Medicine, Cleveland, OH; Department of Neurology (F.J.M.), Massachusetts General Hospital, Boston; Accelerated Cure Project for MS (H.S., S.L., R.N.M.), Waltham, MA; Currie Consultancy (K.M.C.), LLC Eastover, SC; International AIDS Vaccine Initiative (H.M.S.), Frederick, MD; Mammoth Hospital (S.C.), Mammoth Lakes, CA; National Multiple Sclerosis Society (B.F.B., J.F.); Medical Affairs (M.K.R.), Quest Diagnostics, Secaucus, NJ; Departments of Neurology and Immunobiology (K.C.O.), Yale University School of Medicine, New Haven, CT; and iConquerMS (L.G.K., P.K.), Waltham, MA
| | - Michael K Racke
- From the Department of Population and Quantitative Health Science (F.B.S.B.), School of Medicine, Cleveland, OH; Department of Neurology (F.J.M.), Massachusetts General Hospital, Boston; Accelerated Cure Project for MS (H.S., S.L., R.N.M.), Waltham, MA; Currie Consultancy (K.M.C.), LLC Eastover, SC; International AIDS Vaccine Initiative (H.M.S.), Frederick, MD; Mammoth Hospital (S.C.), Mammoth Lakes, CA; National Multiple Sclerosis Society (B.F.B., J.F.); Medical Affairs (M.K.R.), Quest Diagnostics, Secaucus, NJ; Departments of Neurology and Immunobiology (K.C.O.), Yale University School of Medicine, New Haven, CT; and iConquerMS (L.G.K., P.K.), Waltham, MA
| | - Kevin C O'Connor
- From the Department of Population and Quantitative Health Science (F.B.S.B.), School of Medicine, Cleveland, OH; Department of Neurology (F.J.M.), Massachusetts General Hospital, Boston; Accelerated Cure Project for MS (H.S., S.L., R.N.M.), Waltham, MA; Currie Consultancy (K.M.C.), LLC Eastover, SC; International AIDS Vaccine Initiative (H.M.S.), Frederick, MD; Mammoth Hospital (S.C.), Mammoth Lakes, CA; National Multiple Sclerosis Society (B.F.B., J.F.); Medical Affairs (M.K.R.), Quest Diagnostics, Secaucus, NJ; Departments of Neurology and Immunobiology (K.C.O.), Yale University School of Medicine, New Haven, CT; and iConquerMS (L.G.K., P.K.), Waltham, MA
| | - Laura G Kolaczkowski
- From the Department of Population and Quantitative Health Science (F.B.S.B.), School of Medicine, Cleveland, OH; Department of Neurology (F.J.M.), Massachusetts General Hospital, Boston; Accelerated Cure Project for MS (H.S., S.L., R.N.M.), Waltham, MA; Currie Consultancy (K.M.C.), LLC Eastover, SC; International AIDS Vaccine Initiative (H.M.S.), Frederick, MD; Mammoth Hospital (S.C.), Mammoth Lakes, CA; National Multiple Sclerosis Society (B.F.B., J.F.); Medical Affairs (M.K.R.), Quest Diagnostics, Secaucus, NJ; Departments of Neurology and Immunobiology (K.C.O.), Yale University School of Medicine, New Haven, CT; and iConquerMS (L.G.K., P.K.), Waltham, MA
| | - Phyllis Klein
- From the Department of Population and Quantitative Health Science (F.B.S.B.), School of Medicine, Cleveland, OH; Department of Neurology (F.J.M.), Massachusetts General Hospital, Boston; Accelerated Cure Project for MS (H.S., S.L., R.N.M.), Waltham, MA; Currie Consultancy (K.M.C.), LLC Eastover, SC; International AIDS Vaccine Initiative (H.M.S.), Frederick, MD; Mammoth Hospital (S.C.), Mammoth Lakes, CA; National Multiple Sclerosis Society (B.F.B., J.F.); Medical Affairs (M.K.R.), Quest Diagnostics, Secaucus, NJ; Departments of Neurology and Immunobiology (K.C.O.), Yale University School of Medicine, New Haven, CT; and iConquerMS (L.G.K., P.K.), Waltham, MA
| | - Sara Loud
- From the Department of Population and Quantitative Health Science (F.B.S.B.), School of Medicine, Cleveland, OH; Department of Neurology (F.J.M.), Massachusetts General Hospital, Boston; Accelerated Cure Project for MS (H.S., S.L., R.N.M.), Waltham, MA; Currie Consultancy (K.M.C.), LLC Eastover, SC; International AIDS Vaccine Initiative (H.M.S.), Frederick, MD; Mammoth Hospital (S.C.), Mammoth Lakes, CA; National Multiple Sclerosis Society (B.F.B., J.F.); Medical Affairs (M.K.R.), Quest Diagnostics, Secaucus, NJ; Departments of Neurology and Immunobiology (K.C.O.), Yale University School of Medicine, New Haven, CT; and iConquerMS (L.G.K., P.K.), Waltham, MA
| | - Robert Nicholas McBurney
- From the Department of Population and Quantitative Health Science (F.B.S.B.), School of Medicine, Cleveland, OH; Department of Neurology (F.J.M.), Massachusetts General Hospital, Boston; Accelerated Cure Project for MS (H.S., S.L., R.N.M.), Waltham, MA; Currie Consultancy (K.M.C.), LLC Eastover, SC; International AIDS Vaccine Initiative (H.M.S.), Frederick, MD; Mammoth Hospital (S.C.), Mammoth Lakes, CA; National Multiple Sclerosis Society (B.F.B., J.F.); Medical Affairs (M.K.R.), Quest Diagnostics, Secaucus, NJ; Departments of Neurology and Immunobiology (K.C.O.), Yale University School of Medicine, New Haven, CT; and iConquerMS (L.G.K., P.K.), Waltham, MA
| |
Collapse
|
1514
|
Tepasse PR, Vollenberg R, Nowacki TM. Vaccination against SARS-CoV-2 in Patients with Inflammatory Bowel Diseases: Where Do We Stand? Life (Basel) 2021; 11:life11111220. [PMID: 34833096 PMCID: PMC8620225 DOI: 10.3390/life11111220] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/04/2021] [Accepted: 11/09/2021] [Indexed: 12/15/2022] Open
Abstract
Crohn’s disease and ulcerative colitis are chronic inflammatory bowel diseases (IBDs). Immunosuppressive medication is the main therapeutic approach to reducing inflammation of the gastrointestinal tract. Immunocompromised patients are more vulnerable to severe courses of illness after infection with common pathogens. The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is the pathogen of the coronavirus disease 2019 (COVID-19) pandemic. COVID-19 leads to acute respiratory distress syndrome (ARDS) following severe pulmonal damage in a significant number of cases. The worldwide circulation of SARS-CoV-2 has led to major concerns about the management of IBD patients during the pandemic, as these patients are expected to be at greater risk of complications because of their underlying altered immunological condition and immunosuppressive therapies. Vaccination against SARS-CoV-2 is considered the main approach in containing the pandemic. Today, several vaccines have been shown to be highly effective in the prevention of SARS-CoV-2 infection and severe disease course in subjects without underlying conditions in respective registration studies. Patients with underlying conditions such as IBD and/or immunosuppressive therapies were not included in the registration studies, so little is known about effectiveness and safety of SARS-CoV-2 vaccination in immunocompromised IBD patients. This review provides an overview of the recent knowledge about vaccine response in IBD patients after vaccination against SARS-CoV-2.
Collapse
|
1515
|
Keeton R, Richardson SI, Moyo-Gwete T, Hermanus T, Tincho MB, Benede N, Manamela NP, Baguma R, Makhado Z, Ngomti A, Motlou T, Mennen M, Chinhoyi L, Skelem S, Maboreke H, Doolabh D, Iranzadeh A, Otter AD, Brooks T, Noursadeghi M, Moon JC, Grifoni A, Weiskopf D, Sette A, Blackburn J, Hsiao NY, Williamson C, Riou C, Goga A, Garrett N, Bekker LG, Gray G, Ntusi NAB, Moore PL, Burgers WA. Prior infection with SARS-CoV-2 boosts and broadens Ad26.COV2.S immunogenicity in a variant-dependent manner. Cell Host Microbe 2021; 29:1611-1619.e5. [PMID: 34688376 PMCID: PMC8511649 DOI: 10.1016/j.chom.2021.10.003] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/10/2021] [Accepted: 10/07/2021] [Indexed: 01/02/2023]
Abstract
The Johnson and Johnson Ad26.COV2.S single-dose vaccine represents an attractive option for coronavirus disease 2019 (COVID-19) vaccination in countries with limited resources. We examined the effect of prior infection with different SARS-CoV-2 variants on Ad26.COV2.S immunogenicity. We compared participants who were SARS-CoV-2 naive with those either infected with the ancestral D614G virus or infected in the second wave when Beta predominated. Prior infection significantly boosts spike-binding antibodies, antibody-dependent cellular cytotoxicity, and neutralizing antibodies against D614G, Beta, and Delta; however, neutralization cross-reactivity varied by wave. Robust CD4 and CD8 T cell responses are induced after vaccination, regardless of prior infection. T cell recognition of variants is largely preserved, apart from some reduction in CD8 recognition of Delta. Thus, Ad26.COV2.S vaccination after infection could result in enhanced protection against COVID-19. The impact of the infecting variant on neutralization breadth after vaccination has implications for the design of second-generation vaccines based on variants of concern.
Collapse
Affiliation(s)
- Roanne Keeton
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Simone I Richardson
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa; MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Thandeka Moyo-Gwete
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa; MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Tandile Hermanus
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa; MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Marius B Tincho
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Ntombi Benede
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Nelia P Manamela
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa; MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Richard Baguma
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Zanele Makhado
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa; MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Amkele Ngomti
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Thopisang Motlou
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa; MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Mathilda Mennen
- Department of Medicine, University of Cape Town and Groote Schuur Hospital, South Africa
| | - Lionel Chinhoyi
- Department of Medicine, University of Cape Town and Groote Schuur Hospital, South Africa
| | - Sango Skelem
- Department of Medicine, University of Cape Town and Groote Schuur Hospital, South Africa
| | - Hazel Maboreke
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Chemical and Systems Biology, Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, South Africa
| | - Deelan Doolabh
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Arash Iranzadeh
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Ashley D Otter
- National Infection Service, Public Health England, Porton Down, UK
| | - Tim Brooks
- National Infection Service, Public Health England, Porton Down, UK
| | - Mahdad Noursadeghi
- Division of Infection and Immunity, University College London, London, UK
| | - James C Moon
- Institute of Cardiovascular Sciences, University College London, London, UK; Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego, La Jolla, CA, USA
| | - Jonathan Blackburn
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Chemical and Systems Biology, Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, South Africa
| | - Nei-Yuan Hsiao
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa; NHLS Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Carolyn Williamson
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa; Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Cape Town, South Africa
| | - Catherine Riou
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa; Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Cape Town, South Africa
| | - Ameena Goga
- South African Medical Research Council, Cape Town, South Africa
| | - Nigel Garrett
- Centre for the AIDS Programme of Research in South Africa, Durban, South Africa; Discipline of Public Health Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Linda-Gail Bekker
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Desmond Tutu HIV Centre, Cape Town, South Africa
| | - Glenda Gray
- South African Medical Research Council, Cape Town, South Africa
| | - Ntobeko A B Ntusi
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Department of Medicine, University of Cape Town and Groote Schuur Hospital, South Africa; Hatter Institute for Cardiovascular Research in Africa, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
| | - Penny L Moore
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa; MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa.
| | - Wendy A Burgers
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa; Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa; Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
1516
|
Hayawi K, Shahriar S, Serhani MA, Alashwal H, Masud MM. Vaccine versus Variants (3Vs): Are the COVID-19 Vaccines Effective against the Variants? A Systematic Review. Vaccines (Basel) 2021; 9:1305. [PMID: 34835238 PMCID: PMC8622454 DOI: 10.3390/vaccines9111305] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/07/2021] [Accepted: 11/08/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND With the emergence and spread of new SARS-CoV-2 variants, concerns are raised about the effectiveness of the existing vaccines to protect against these new variants. Although many vaccines were found to be highly effective against the reference COVID-19 strain, the same level of protection may not be found against mutation strains. The objective of this study is to systematically review relevant studies in the literature and compare the efficacy of COVID-19 vaccines against new variants. METHODS We conducted a systematic review of research published in Scopus, PubMed, and Google Scholar until 30 August 2021. Studies including clinical trials, prospective cohorts, retrospective cohorts, and test negative case-controls that reported vaccine effectiveness against any COVID-19 variants were considered. PRISMA recommendations were adopted for screening, eligibility, and inclusion. RESULTS 129 unique studies were reviewed by the search criteria, of which 35 met the inclusion criteria. These comprised of 13 test negative case-control studies, 6 Phase 1-3 clinical trials, and 16 observational studies. The study location, type, vaccines used, variants considered, and reported efficacies were highlighted. CONCLUSION Full vaccination (two doses) offers strong protection against Alpha (B.1.1.7) with 13 out of 15 studies reporting more than 84% efficacy. The results are not conclusive against the Beta (B.1.351) variant for fully vaccinated individuals with 4 out of 7 studies reporting efficacies between 22 and 60% and 3 out of 7 studies reporting efficacies between 75 and 100%. Protection against Gamma (P.1) variant was lower than 50% according to two studies in fully vaccinated individuals. The data on Delta (B.1.617.2) variant is limited but indicates lower protection compared to other variants.
Collapse
Affiliation(s)
- Kadhim Hayawi
- College of Technological Innovation, Zayed University, Abu Dhabi 51133, United Arab Emirates; (K.H.); (S.S.)
| | - Sakib Shahriar
- College of Technological Innovation, Zayed University, Abu Dhabi 51133, United Arab Emirates; (K.H.); (S.S.)
| | - Mohamed Adel Serhani
- College of Information Technology, UAE University, Abu Dhabi 15551, United Arab Emirates; (H.A.); (M.M.M.)
| | - Hany Alashwal
- College of Information Technology, UAE University, Abu Dhabi 15551, United Arab Emirates; (H.A.); (M.M.M.)
| | - Mohammad M. Masud
- College of Information Technology, UAE University, Abu Dhabi 15551, United Arab Emirates; (H.A.); (M.M.M.)
| |
Collapse
|
1517
|
Rzymski P, Camargo CA, Fal A, Flisiak R, Gwenzi W, Kelishadi R, Leemans A, Nieto JJ, Ozen A, Perc M, Poniedziałek B, Sedikides C, Sellke F, Skirmuntt EC, Stashchak A, Rezaei N. COVID-19 Vaccine Boosters: The Good, the Bad, and the Ugly. Vaccines (Basel) 2021; 9:1299. [PMID: 34835230 PMCID: PMC8623745 DOI: 10.3390/vaccines9111299] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/04/2021] [Accepted: 11/05/2021] [Indexed: 02/06/2023] Open
Abstract
Pursuing vaccinations against COVID-19 brings hope to limit the spread of SARS-CoV-2 and remains the most rational decision under pandemic conditions. However, it does not come without challenges, including temporary shortages in vaccine doses, significant vaccine inequity, and questions regarding the durability of vaccine-induced immunity that remain unanswered. Moreover, SARS-CoV-2 has undergone evolution with the emergence of its novel variants, characterized by enhanced transmissibility and ability to at least partially evade neutralizing antibodies. At the same time, serum antibody levels start to wane within a few months after vaccination, ultimately increasing the risk of breakthrough infections. This article discusses whether the administration of booster doses of COVID-19 vaccines is urgently needed to control the pandemic. We conclude that, at present, optimizing the immunity level of wealthy populations cannot come at the expense of low-income regions that suffer from vaccine unavailability. Although the efficiency of vaccination in protecting from infection may decrease over time, current data show that efficacy against severe disease, hospitalization, and death remains at a high level. If vaccine coverage continues at extremely low levels in various regions, including African countries, SARS-CoV-2 may sooner or later evolve into variants better adapted to evade natural and vaccine-induced immunity, ultimately bringing a global threat that, of course, includes wealthy populations. We offer key recommendations to increase vaccination rates in low-income countries. The pandemic is, by definition, a major epidemiological event and requires looking beyond one's immediate self-interest; otherwise, efforts to contain it will be futile.
Collapse
Affiliation(s)
- Piotr Rzymski
- Department of Environmental Medicine, Poznan University of Medical Sciences, 60-806 Poznań, Poland;
- Universal Scientific Education and Research Network (USERN), https://usern.tums.ac.ir, Tehran 1417614411, Iran; (C.A.C.J.); (A.F.); (R.K.); (A.L.); (J.J.N.); (A.O.); (M.P.); (C.S.); (F.S.); (A.S.)
| | - Carlos A. Camargo
- Universal Scientific Education and Research Network (USERN), https://usern.tums.ac.ir, Tehran 1417614411, Iran; (C.A.C.J.); (A.F.); (R.K.); (A.L.); (J.J.N.); (A.O.); (M.P.); (C.S.); (F.S.); (A.S.)
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Andrzej Fal
- Universal Scientific Education and Research Network (USERN), https://usern.tums.ac.ir, Tehran 1417614411, Iran; (C.A.C.J.); (A.F.); (R.K.); (A.L.); (J.J.N.); (A.O.); (M.P.); (C.S.); (F.S.); (A.S.)
- Collegium Medicum, Warsaw Faculty of Medicine, Cardinal Stefan Wyszyński University, 01-938 Warsaw, Poland
| | - Robert Flisiak
- Department of Infectious Diseases and Hepatology, Medical University of Bialystok, 15-540 Białystok, Poland;
| | - Willis Gwenzi
- Biosystems and Environmental Engineering Research Group, Department of Agricultural and Biosystems Engineering, University of Zimbabwe, Mount Pleasant, Harare P.O. Box MP167, Zimbabwe;
| | - Roya Kelishadi
- Universal Scientific Education and Research Network (USERN), https://usern.tums.ac.ir, Tehran 1417614411, Iran; (C.A.C.J.); (A.F.); (R.K.); (A.L.); (J.J.N.); (A.O.); (M.P.); (C.S.); (F.S.); (A.S.)
- Department of Pediatrics, Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan 8174673441, Iran
| | - Alexander Leemans
- Universal Scientific Education and Research Network (USERN), https://usern.tums.ac.ir, Tehran 1417614411, Iran; (C.A.C.J.); (A.F.); (R.K.); (A.L.); (J.J.N.); (A.O.); (M.P.); (C.S.); (F.S.); (A.S.)
- PROVIDI Lab, Image Sciences Institute, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Juan J. Nieto
- Universal Scientific Education and Research Network (USERN), https://usern.tums.ac.ir, Tehran 1417614411, Iran; (C.A.C.J.); (A.F.); (R.K.); (A.L.); (J.J.N.); (A.O.); (M.P.); (C.S.); (F.S.); (A.S.)
- Instituto de Matemáticas, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Ahmet Ozen
- Universal Scientific Education and Research Network (USERN), https://usern.tums.ac.ir, Tehran 1417614411, Iran; (C.A.C.J.); (A.F.); (R.K.); (A.L.); (J.J.N.); (A.O.); (M.P.); (C.S.); (F.S.); (A.S.)
- Department of Pediatric Allergy and Immunology, Marmara University School of Medicine, 34854 Istanbul, Turkey
| | - Matjaž Perc
- Universal Scientific Education and Research Network (USERN), https://usern.tums.ac.ir, Tehran 1417614411, Iran; (C.A.C.J.); (A.F.); (R.K.); (A.L.); (J.J.N.); (A.O.); (M.P.); (C.S.); (F.S.); (A.S.)
- Faculty of Natural Sciences and Mathematics, University of Maribor, 2000 Maribor, Slovenia
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404332, Taiwan
| | - Barbara Poniedziałek
- Department of Environmental Medicine, Poznan University of Medical Sciences, 60-806 Poznań, Poland;
- Universal Scientific Education and Research Network (USERN), https://usern.tums.ac.ir, Tehran 1417614411, Iran; (C.A.C.J.); (A.F.); (R.K.); (A.L.); (J.J.N.); (A.O.); (M.P.); (C.S.); (F.S.); (A.S.)
| | - Constantine Sedikides
- Universal Scientific Education and Research Network (USERN), https://usern.tums.ac.ir, Tehran 1417614411, Iran; (C.A.C.J.); (A.F.); (R.K.); (A.L.); (J.J.N.); (A.O.); (M.P.); (C.S.); (F.S.); (A.S.)
- Center for Research on Self and Identity, School of Psychology, University of Southampton, Southampton SO17 1BJ, UK
| | - Frank Sellke
- Universal Scientific Education and Research Network (USERN), https://usern.tums.ac.ir, Tehran 1417614411, Iran; (C.A.C.J.); (A.F.); (R.K.); (A.L.); (J.J.N.); (A.O.); (M.P.); (C.S.); (F.S.); (A.S.)
- Alpert Medical School of Brown University, Division of Cardiothoracic Surgery, Rhode Island Hospital, Providence, RI 02905, USA
| | - Emilia C. Skirmuntt
- Peter Medawar Building for Pathogen Research, Department of Zoology, University of Oxford, Oxford OX1 3SY, UK;
| | - Anzhela Stashchak
- Universal Scientific Education and Research Network (USERN), https://usern.tums.ac.ir, Tehran 1417614411, Iran; (C.A.C.J.); (A.F.); (R.K.); (A.L.); (J.J.N.); (A.O.); (M.P.); (C.S.); (F.S.); (A.S.)
- International Relations Office, Kharkiv National Medical University, 61000 Kharkiv, Ukraine
| | - Nima Rezaei
- Universal Scientific Education and Research Network (USERN), https://usern.tums.ac.ir, Tehran 1417614411, Iran; (C.A.C.J.); (A.F.); (R.K.); (A.L.); (J.J.N.); (A.O.); (M.P.); (C.S.); (F.S.); (A.S.)
- Research Center for Immunodeficiencies, Children’s Medical Center, Tehran University of Medical Sciences, Tehran 1417614411, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| |
Collapse
|
1518
|
Dong Y, Dai T, Wang B, Zhang L, Zeng LH, Huang J, Yan H, Zhang L, Zhou F. The way of SARS-CoV-2 vaccine development: success and challenges. Signal Transduct Target Ther 2021; 6:387. [PMID: 34753918 PMCID: PMC8575680 DOI: 10.1038/s41392-021-00796-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 10/10/2021] [Indexed: 01/08/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of coronavirus disease 2019 (COVID-19). To halt the pandemic, multiple SARS-CoV-2 vaccines have been developed and several have been allowed for emergency use and rollout worldwide. With novel SARS-CoV-2 variants emerging and circulating widely, whether the original vaccines that were designed based on the wild-type SARS-CoV-2 were effective against these variants has been a contentious discussion. Moreover, some studies revealed the long-term changes of immune responses post SARS-CoV-2 infection or vaccination and the factors that might impact the vaccine-induced immunity. Thus, in this review, we have summarized the influence of mutational hotspots on the vaccine efficacy and characteristics of variants of interest and concern. We have also discussed the reasons that might result in discrepancies in the efficacy of different vaccines estimated in different trials. Furthermore, we provided an overview of the duration of immune responses after natural infection or vaccination and shed light on the factors that may affect the immunity induced by the vaccines, such as special disease conditions, sex, and pre-existing immunity, with the aim of aiding in combating COVID-19 and distributing SARS-CoV-2 vaccines under the prevalence of diverse SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Yetian Dong
- School of Medicine, Zhejiang University City College, Hangzhou, 310015, Zhejiang, China
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Department of Orthopaedic Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Rui'an, China
| | - Tong Dai
- Institutes of Biology and Medical Science, Soochow University, Suzhou 215123, China
| | - Bin Wang
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Lei Zhang
- Department of Orthopaedic Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Rui'an, China
| | - Ling-Hui Zeng
- School of Medicine, Zhejiang University City College, Hangzhou, 310015, Zhejiang, China
| | - Jun Huang
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Haiyan Yan
- School of Medicine, Zhejiang University City College, Hangzhou, 310015, Zhejiang, China
| | - Long Zhang
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| | - Fangfang Zhou
- Institutes of Biology and Medical Science, Soochow University, Suzhou 215123, China.
| |
Collapse
|
1519
|
Amjadi MF, Adyniec RR, Gupta S, Bashar SJ, Mergaert AM, Braun KM, Moreno GK, O'Connor DH, Friedrich TC, Safdar N, McCoy SS, Shelef MA. Anti-membrane and anti-spike antibodies are long-lasting and together discriminate between past COVID-19 infection and vaccination. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.11.02.21265750. [PMID: 34790984 PMCID: PMC8597887 DOI: 10.1101/2021.11.02.21265750] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The consequences of past COVID-19 infection for personal health and long-term population immunity are only starting to be revealed. Unfortunately, detecting past infection is currently a challenge, limiting clinical and research endeavors. Widely available anti-SARS-CoV-2 antibody tests cannot differentiate between past infection and vaccination given vaccine-induced anti-spike antibodies and the rapid loss of infection-induced anti-nucleocapsid antibodies. Anti-membrane antibodies develop after COVID-19, but their long-term persistence is unknown. Here, we demonstrate that anti-membrane IgG is a sensitive and specific marker of past COVID-19 infection and persists at least one year. We also confirm that anti-receptor binding domain (RBD) Ig is a long-lasting, sensitive, and specific marker of past infection and vaccination, while anti-nucleocapsid IgG lacks specificity and quickly declines after COVID-19. Thus, a combination of anti-membrane and anti-RBD antibodies can accurately differentiate between distant COVID-19 infection, vaccination, and naïve states to advance public health, individual healthcare, and research goals.
Collapse
|
1520
|
Alhassan RK, Owusu-Agyei S, Ansah EK, Gyapong M. COVID-19 vaccine uptake among health care workers in Ghana: a case for targeted vaccine deployment campaigns in the global south. HUMAN RESOURCES FOR HEALTH 2021; 19:136. [PMID: 34742301 PMCID: PMC8571849 DOI: 10.1186/s12960-021-00657-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/10/2021] [Indexed: 05/05/2023]
Abstract
BACKGROUND Health care workers (HCWs) are among the high-risk groups in contracting and dying from COVID-19. World Health Organization estimates that over 10,000 HCWs in Africa have been infected with COVID-19 making it a significant occupational health hazard to HCWs. In Ghana, over 100 HCWs have already been infected and dozen others died from the virus. Acceptability and uptake of the COVID-19 vaccine is therefore critical to promote health and safety of HCWs as the country battles out of a third wave of the pandemic. OBJECTIVE The study sought to ascertain the correlates of HCWs likelihood of participating in a COVID-19 vaccine trial and accepting the vaccine when given the opportunity. METHODS The study was a web-based cross-sectional survey among HCWs (n = 1605) in all sixteen (16) administrative regions in Ghana. Data were analyzed with STATA statistical analysis software (version 14). Chi-square (X2) and Fisher's exact tests were used to test for differences in categorical variables; bivariate probit regression analysis with Average Marginal Effect (AME) was employed to ascertain the determinants of HCWs' likelihood of participating in a COVID-19 vaccine trial and taking the vaccine. RESULTS It was found that 48% of HCWs will participate in a COVID-19 vaccine trial when given the opportunity; 70% will accept the COVID-19 vaccine; younger HCWs (AME = 0.28, SE = 0.16, p < 0.1), non-Christians (AME = 21, SE = 0.09, p < 0.05) and those who worked in faith-based health facilities (AME = 18, SE = 0.07, p < 0.05) were more likely to participate in a COVID-19 vaccine trial. Female HCWs (AME = - 11, SE = 0.04, p < 0.05) and those with lower educational qualification were less likely to accept a COVID-19 vaccine (AME = - 0.16, SE = 0.08, p < 0.1). Reasons cited for unwillingness to participate in a COVID-19 vaccine trial or uptake the vaccine were mainly fear, safety concerns, mistrust, uncertainty, spiritual and religious beliefs. CONCLUSIONS Acceptance of the COVID-19 vaccine appear to be high among HCWs; conversely, willingness to volunteer for the vaccine trial was low. Continuous targeted and integrated public health education for HCWs will enhance vaccine acceptability to promote safety and population health in the global south as Ghana intensifies efforts to produce COVID-19 vaccines locally.
Collapse
Affiliation(s)
- Robert Kaba Alhassan
- Institute of Health Research (IHR), University of Health and Allied Sciences, PMB 31, Volta Region, Ho, Ghana
| | - Seth Owusu-Agyei
- Institute of Health Research (IHR), University of Health and Allied Sciences, PMB 31, Volta Region, Ho, Ghana
| | - Evelyn Korkor Ansah
- Institute of Health Research (IHR), University of Health and Allied Sciences, PMB 31, Volta Region, Ho, Ghana
| | - Margaret Gyapong
- Institute of Health Research (IHR), University of Health and Allied Sciences, PMB 31, Volta Region, Ho, Ghana
| |
Collapse
|
1521
|
Lai CY, To A, Wong TAS, Lieberman MM, Clements DE, Senda JT, Ball AH, Pessaint L, Andersen H, Furuyama W, Marzi A, Donini O, Lehrer AT. Recombinant protein subunit SARS-CoV-2 vaccines formulated with CoVaccine HT adjuvant induce broad, Th1 biased, humoral and cellular immune responses in mice. Vaccine X 2021; 9:100126. [PMID: 34778744 PMCID: PMC8570651 DOI: 10.1016/j.jvacx.2021.100126] [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: 03/15/2021] [Revised: 10/20/2021] [Accepted: 10/29/2021] [Indexed: 12/23/2022] Open
Abstract
The speed at which several COVID-19 vaccines went from conception to receiving FDA and EMA approval for emergency use is an achievement unrivaled in the history of vaccine development. Mass vaccination efforts using the highly effective vaccines are currently underway to generate sufficient herd immunity and reduce transmission of the SARS-CoV-2 virus. Despite the most advanced vaccine technology, global recipient coverage, especially in resource-poor areas remains a challenge as genetic drift in naïve population pockets threatens overall vaccine efficacy. In this study, we described the production of insect-cell expressed SARS-CoV-2 spike protein ectodomain constructs and examined their immunogenicity in mice. We demonstrated that, when formulated with CoVaccine HTTM adjuvant, an oil-in-water nanoemulsion compatible with lyophilization, our vaccine candidates elicit a broad-spectrum IgG response, high neutralizing antibody (NtAb) titers against SARS-CoV-2 prototype and variants of concern, specifically B.1.351 (Beta) and P.1. (Gamma), and an antigen-specific IFN-γ secreting response in outbred mice. Of note, different ectodomain constructs yielded variations in NtAb titers against the prototype strain and some VOC. Dose response experiments indicated that NtAb titers increased with antigen dose, but not adjuvant dose, and may be higher with a lower adjuvant dose. Our findings lay the immunological foundation for the development of a dry-thermostabilized vaccine that is deployable without refrigeration.
Collapse
Affiliation(s)
- Chih-Yun Lai
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology,
John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI,
USA
- Pacific Center for Emerging Infectious Disease Research, John A. Burns
School of Medicine, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Albert To
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology,
John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI,
USA
| | - Teri Ann S. Wong
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology,
John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI,
USA
| | - Michael M. Lieberman
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology,
John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI,
USA
| | | | | | - Aquena H. Ball
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology,
John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI,
USA
| | | | | | - Wakako Furuyama
- Laboratory of Virology, Division of Intramural Research, NIAID, NIH,
Hamilton, Montana, MT, USA
| | - Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, NIAID, NIH,
Hamilton, Montana, MT, USA
| | | | - Axel T. Lehrer
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology,
John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI,
USA
- Pacific Center for Emerging Infectious Disease Research, John A. Burns
School of Medicine, University of Hawaii at Manoa, Honolulu, HI, USA
| |
Collapse
|
1522
|
Rzymski P, Poniedziałek B, Fal A. Willingness to Receive the Booster COVID-19 Vaccine Dose in Poland. Vaccines (Basel) 2021; 9:1286. [PMID: 34835217 PMCID: PMC8624071 DOI: 10.3390/vaccines9111286] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/18/2021] [Accepted: 11/03/2021] [Indexed: 12/29/2022] Open
Abstract
COVID-19 vaccinations are essential to mitigate the pandemic and prevent severe SARS-CoV-2 infections. However, the serum antibody levels in vaccinated individuals gradually decrease over time, while SARS-CoV-2 is undergoing an evolution toward more transmissible variants, such as B.1.617.2, ultimately increasing the risk of breakthrough infections and further virus spread. This cross-sectional online study of adult Poles (n = 2427) was conducted in September 2021 (before a general recommendation to administer a booster COVID-19 vaccine dose in Poland was issued) to assess the attitude of individuals who completed the current vaccination regime toward a potential booster dose of the COVID-19 vaccine and identify potential factors that may influence it. Overall, 71% of participants declared willingness to receive a booster COVID-19 dose, with a low median level of fear of receiving it of 1.0 (measured by the 10-point Likert-type scale), which was increased particularly in those having a worse experience (in terms of severity of side effects and associated fear) with past COVID-19 vaccination. The lowest frequency of willingness to receive a booster dose (26.7%) was seen in the group previously vaccinated with Ad26.COV2.S. The majority of individuals vaccinated previously with mRNA vaccines wished to receive the same vaccine, while in the case of AZD1222, such accordance was observed only in 9.1%. The main reasons against accepting a booster COVID-19 dose included the side effects experienced after previous doses, the opinion that further vaccination is unnecessary, and safety uncertainties. Women, older individuals (≥50 years), subjects with obesity, chronic diseases, and pre-vaccination and post-vaccination SARS-CoV-2 infections, and those with a history of vaccination against influenza were significantly more frequently willing to receive a booster COVID-19 dose. Moreover, the majority of immunosuppressed individuals (88%) were willing to receive an additional dose. The results emphasize some hesitancy toward potential further COVID-19 vaccination in the studied group of Poles and indicate the main groups to be targeted with effective science communication regarding the booster doses.
Collapse
Affiliation(s)
- Piotr Rzymski
- Department of Environmental Medicine, Poznan University of Medical Sciences, 60-806 Poznań, Poland;
- Integrated Science Association (ISA), Universal Scientific Education and Research Network (USERN), 60-806 Poznań, Poland
| | - Barbara Poniedziałek
- Department of Environmental Medicine, Poznan University of Medical Sciences, 60-806 Poznań, Poland;
| | - Andrzej Fal
- Collegium Medicum, Warsaw Faculty of Medicine, Cardinal Stefan Wyszyński University, 01-938 Warsaw, Poland;
| |
Collapse
|
1523
|
Tatsi EB, Filippatos F, Michos A. SARS-CoV-2 variants and effectiveness of vaccines: a review of current evidence. Epidemiol Infect 2021; 149:e237. [PMID: 34732275 PMCID: PMC8632374 DOI: 10.1017/s0950268821002430] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 10/29/2021] [Accepted: 10/31/2021] [Indexed: 12/23/2022] Open
Abstract
The SARS-CoV-2 virus is rapidly evolving via mutagenesis, lengthening the pandemic, and threatening the public health. Until August 2021, 12 variants of SARS-CoV-2 named as variants of concern (VOC; Alpha to Delta) or variants of interest (VOI; Epsilon to Mu), with significant impact on transmissibility, morbidity, possible reinfection and mortality, have been identified. The VOC Delta (B.1.617.2) of Indian origin is now the dominant and the most contagious variant worldwide as it provokes a strong binding to the human ACE2 receptor, increases transmissibility and manifests considerable immune escape strategies after natural infection or vaccination. Although the development and administration of SARS-CoV-2 vaccines, based on different technologies (mRNA, adenovirus carrier, recombinant protein, etc.), are very promising for the control of the pandemic, their effectiveness and neutralizing activity against VOCs varies significantly. In this review, we describe the most significant circulating variants of SARS-CoV-2, and the known effectiveness of currently available vaccines against them.
Collapse
Affiliation(s)
- Elizabeth-Barbara Tatsi
- First Department of Pediatrics, Infectious Diseases and Chemotherapy Research Laboratory, Medical School, National and Kapodistrian University of Athens, ‘Aghia Sophia’ Children's Hospital, 11527Athens, Greece
| | - Filippos Filippatos
- First Department of Pediatrics, Infectious Diseases and Chemotherapy Research Laboratory, Medical School, National and Kapodistrian University of Athens, ‘Aghia Sophia’ Children's Hospital, 11527Athens, Greece
| | - Athanasios Michos
- First Department of Pediatrics, Infectious Diseases and Chemotherapy Research Laboratory, Medical School, National and Kapodistrian University of Athens, ‘Aghia Sophia’ Children's Hospital, 11527Athens, Greece
| |
Collapse
|
1524
|
Acute disseminated encephalomyelitis (ADEM) following recent Oxford/AstraZeneca COVID-19 vaccination. Forensic Sci Med Pathol 2021; 18:74-79. [PMID: 34735684 PMCID: PMC8567127 DOI: 10.1007/s12024-021-00440-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2021] [Indexed: 02/02/2023]
Abstract
This report describes the clinical context and autopsy findings in the first reported fatal case of acute disseminated encephalomyelitis (ADEM), developed after being vaccinated using the Oxford/AstraZeneca COVID-19 vaccine. ADEM is a rare autoimmune disease, causing demyelination in the brain and spinal cord. A wide variety of precipitating factors can trigger ADEM, and it has long been known to be a rare adverse event following some types of vaccinations. Recently, ADEM has also been associated with COVID-19 infection and (very rarely) with COVID-19 vaccination. The reports of the latter however all pertain to living patients. Our case demonstrates that ADEM should be considered in patients developing neurological symptoms post COVID-19 vaccination, although that this adverse reaction is likely to remain extremely rare. Our report further emphasizes the added value of comprehensive post mortem investigation to confirm ante mortem diagnosis and to determine vaccination safety.
Collapse
|
1525
|
Guven DC, Sahin TK, Kilickap S, Uckun FM. Antibody Responses to COVID-19 Vaccination in Cancer: A Systematic Review. Front Oncol 2021; 11:759108. [PMID: 34804957 PMCID: PMC8599356 DOI: 10.3389/fonc.2021.759108] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 10/18/2021] [Indexed: 02/05/2023] Open
Abstract
INTRODUCTION After the results of phase III vaccine studies became available, the leading oncology societies recommended two doses of COVID-19 vaccination to all patients with cancer with no specific recommendation for tumor type and active treatments. However, the data on the COVID-19 vaccine efficacy in cancer patients is limited due to exclusion of cancer patients from most vaccine clinical trials. Therefore, we systemically reviewed the available evidence evaluating the antibody responses in cancer patients. METHODS We conducted a systematic search from the Pubmed database and calculated risk differences (RD) and 95% confidence intervals (CI) to compare seroconversion rates between cancer patients and controls using the Review Manager software, version 5.3. RESULTS Our systematic search retrieved a total 27 studies and we included 17 studies with control arms in the analyses. Cancer patients had significantly lower seroconversion rates (37.3%) than controls (74.1%) (RD: -0.44, 95% CI: -0.52, -0.35, p<0.001) with first vaccine dose. After two doses, the seroconversion rates were 99.6% in control arm and 78.3% in cancer patients (RD: -0.19, 95% CI: -0.28, -0.10, p<0.001). The difference in seroconversion rates was more pronounced patients with hematologic malignancies (72.6%) (RD: -0.25, 95% CI: -0.27, -0.22, p<0.001) than patients with solid tumors (91.6%) (RD: -0.09, 95% CI: -0.13, -0.04, p<0.003) and patients in remission (RD: -0.10, 95% CI: -0.14, -0.06, p<0.001). CONCLUSION In conclusion, COVID-19 vaccine seroconversion rates were significantly lower in patients with hematological malignancies and patients under active treatment. Further research focusing on the approaches to improve vaccine efficacy and exploration of novel treatment options is urgently needed for these patients.
Collapse
Affiliation(s)
- Deniz C. Guven
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Taha K. Sahin
- Department of Internal Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Saadettin Kilickap
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
- Department of Medical Oncology, Istinye University, Istanbul, Turkey
| | - Fatih M. Uckun
- Department of Immunology and Inflammatory Disorders, Reven Pharmaceuticals, Westminster, CO, United States
- Immuno-Oncology Program and COVID-19 Task Force, Ares Pharmaceuticals, St. Paul, MN, United States
| |
Collapse
|
1526
|
Tostanoski LH, Yu J, Mercado NB, McMahan K, Jacob-Dolan C, Martinot AJ, Piedra-Mora C, Anioke T, Chang A, Giffin VM, Hope DL, Wan H, Bondzie EA, Mahrokhian SH, Wrijil LM, Bauer K, Pessaint L, Porto M, Piegols J, Faudree A, Spence B, Kar S, Amanat F, Krammer F, Andersen H, Lewis MG, Wegmann F, Zahn R, Schuitemaker H, Barouch DH. Immunity elicited by natural infection or Ad26.COV2.S vaccination protects hamsters against SARS-CoV-2 variants of concern. Sci Transl Med 2021; 13:eabj3789. [PMID: 34705477 PMCID: PMC8818312 DOI: 10.1126/scitranslmed.abj3789] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 08/25/2021] [Indexed: 01/07/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern have emerged and may pose a threat to both the efficacy of vaccines based on the original WA1/2020 strain and the natural immunity induced by infection with earlier SARS-CoV-2 variants. We investigated how mutations in the spike protein of circulating SARS-CoV-2 variants, which have been shown to partially evade neutralizing antibodies, affect natural and vaccine-induced immunity. We adapted a Syrian hamster model of moderate to severe clinical disease for two variant strains of SARS-CoV-2: B.1.1.7 (alpha variant) and B.1.351 (beta variant). We then assessed the protective efficacy conferred by either natural immunity from WA1/2020 infection or by vaccination with a single dose of the adenovirus serotype 26 vaccine, Ad26.COV2.S. Primary infection with the WA1/2020 strain provided potent protection against weight loss and viral replication in lungs after rechallenge with WA1/2020, B.1.1.7, or B.1.351. Ad26.COV2.S induced cross-reactive binding and neutralizing antibodies that were reduced against the B.1.351 strain compared with WA1/2020 but nevertheless still provided robust protection against B.1.351 challenge, as measured by weight loss and pathology scoring in the lungs. Together, these data support hamsters as a preclinical model to study protection against emerging variants of SARS-CoV-2 conferred by prior infection or vaccination.
Collapse
Affiliation(s)
- Lisa H. Tostanoski
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Jingyou Yu
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Noe B. Mercado
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Katherine McMahan
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Catherine Jacob-Dolan
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Amanda J. Martinot
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
- Department of Biomedical Sciences, Section of Pathology, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA 01536, USA
| | - Cesar Piedra-Mora
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
- Department of Biomedical Sciences, Section of Pathology, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA 01536, USA
| | - Tochi Anioke
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Aiquan Chang
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Victoria M. Giffin
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - David L. Hope
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Huahua Wan
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Esther A. Bondzie
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Shant H. Mahrokhian
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Linda M. Wrijil
- Department of Biomedical Sciences, Section of Pathology, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA 01536, USA
| | - Katherine Bauer
- Department of Biomedical Sciences, Section of Pathology, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA 01536, USA
| | | | | | | | | | | | | | - Fatima Amanat
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Florian Krammer
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | | | - Frank Wegmann
- Janssen Vaccines & Prevention BV, Leiden, 2333 CN, Netherlands
| | - Roland Zahn
- Janssen Vaccines & Prevention BV, Leiden, 2333 CN, Netherlands
| | | | - Dan H. Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
- Massachusetts Consortium on Pathogen Readiness, Boston, MA 02115, USA
| |
Collapse
|
1527
|
Gregory ME, Powell JR, MacEwan SR, Kurth JD, Kenah E, Panchal AR, McAlearney AS. COVID-19 Vaccinations in EMS Professionals: Prevalence and Predictors. PREHOSP EMERG CARE 2021; 26:632-640. [PMID: 34644239 PMCID: PMC9190028 DOI: 10.1080/10903127.2021.1993391] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Background: Immunizations for emergency medical services (EMS) professionals during pandemics are an important tool to increase the safety of the workforce as well as their patients. The purpose of this study was to better understand EMS professionals’ decisions to receive or decline a COVID-19 vaccine. Methods: We conducted a cross-sectional analysis of nationally certified EMS professionals (18–85 years) in April 2021. Participants received an electronic survey asking whether they received a vaccine, why or why not, and their associated beliefs using three validated scales: perceived risk of COVID-19, medical mistrust, and confidence in the COVID-19 vaccine. Data were merged with National Registry dataset demographics. Analyses included descriptive analysis and multivariable logistic regression (OR, 95% CI). Multivariate imputation by chained equations was used for missingness. Results: A total of 2,584 respondents satisfied inclusion criteria (response rate = 14%). Overall, 70% of EMS professionals were vaccinated. Common reasons for vaccination among vaccinated respondents were to protect oneself (76%) and others (73%). Common reasons for non-vaccination among non-vaccinated respondents included concerns about vaccine safety (53%) and beliefs that vaccination was not necessary (39%). Most who had not received the vaccine did not plan to get it in the future (84%). Hesitation was most frequently related to wanting to see how the vaccine was working for others (55%). Odds of COVID-19 vaccination were associated with demographics including age (referent <28 years; 39–50 years: 1.56, 1.17–2.08; >51 years: 2.22, 1.64–3.01), male sex (1.26, 1.01–1.58), residing in an urban/suburban area (referent rural; 1.36, 1.08–1.70), advanced education (referent GED/high school and below; bachelor’s and above: 1.72, 1.19–2.47), and working at a hospital (referent fire-based agency; 1.53, 1.04–2.24). Additionally, vaccination odds were significantly higher with greater perceived risk of COVID-19 (2.05, 1.68–2.50), and higher vaccine confidence (2.84, 2.40–3.36). Odds of vaccination were significantly lower with higher medical mistrust (0.54, 0.46–0.63). Conclusion: Despite vaccine availability, not all EMS professionals had been vaccinated. The decision to receive a COVID-19 vaccine was associated with demographics, beliefs regarding COVID-19 and the vaccine, and medical mistrust. Efforts to increase COVID-19 vaccination rates should emphasize the safety and efficacy of vaccines.
Collapse
Affiliation(s)
- Megan E Gregory
- Received August 24, 2021; Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, Ohio (MEG, ASM); The Center for the Advancement of Team Science, Analytics, and Systems Thinking in Health Services and Implementation Science Research (CATALYST), College of Medicine, The Ohio State University, Columbus, Ohio (MEG, SRM, ARP, ASM); National Registry of Emergency Medical Technicians, Columbus, Ohio (JRP, JDK, ARP); Division of Epidemiology, The Ohio State University College of Public Health, Columbus, Ohio (JRP, ARP); Division of Biostatistics, The Ohio State University College of Public Health, Columbus, Ohio (EK); Department of Emergency Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio (ARP); Department of Family and Community Medicine, College of Medicine, The Ohio State University, Columbus, Ohio (ASM). Revision received October 8, 2021; accepted for publication October 11, 2021
| | - Jonathan R Powell
- Received August 24, 2021; Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, Ohio (MEG, ASM); The Center for the Advancement of Team Science, Analytics, and Systems Thinking in Health Services and Implementation Science Research (CATALYST), College of Medicine, The Ohio State University, Columbus, Ohio (MEG, SRM, ARP, ASM); National Registry of Emergency Medical Technicians, Columbus, Ohio (JRP, JDK, ARP); Division of Epidemiology, The Ohio State University College of Public Health, Columbus, Ohio (JRP, ARP); Division of Biostatistics, The Ohio State University College of Public Health, Columbus, Ohio (EK); Department of Emergency Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio (ARP); Department of Family and Community Medicine, College of Medicine, The Ohio State University, Columbus, Ohio (ASM). Revision received October 8, 2021; accepted for publication October 11, 2021
| | - Sarah R MacEwan
- Received August 24, 2021; Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, Ohio (MEG, ASM); The Center for the Advancement of Team Science, Analytics, and Systems Thinking in Health Services and Implementation Science Research (CATALYST), College of Medicine, The Ohio State University, Columbus, Ohio (MEG, SRM, ARP, ASM); National Registry of Emergency Medical Technicians, Columbus, Ohio (JRP, JDK, ARP); Division of Epidemiology, The Ohio State University College of Public Health, Columbus, Ohio (JRP, ARP); Division of Biostatistics, The Ohio State University College of Public Health, Columbus, Ohio (EK); Department of Emergency Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio (ARP); Department of Family and Community Medicine, College of Medicine, The Ohio State University, Columbus, Ohio (ASM). Revision received October 8, 2021; accepted for publication October 11, 2021
| | - Jordan D Kurth
- Received August 24, 2021; Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, Ohio (MEG, ASM); The Center for the Advancement of Team Science, Analytics, and Systems Thinking in Health Services and Implementation Science Research (CATALYST), College of Medicine, The Ohio State University, Columbus, Ohio (MEG, SRM, ARP, ASM); National Registry of Emergency Medical Technicians, Columbus, Ohio (JRP, JDK, ARP); Division of Epidemiology, The Ohio State University College of Public Health, Columbus, Ohio (JRP, ARP); Division of Biostatistics, The Ohio State University College of Public Health, Columbus, Ohio (EK); Department of Emergency Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio (ARP); Department of Family and Community Medicine, College of Medicine, The Ohio State University, Columbus, Ohio (ASM). Revision received October 8, 2021; accepted for publication October 11, 2021
| | - Eben Kenah
- Received August 24, 2021; Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, Ohio (MEG, ASM); The Center for the Advancement of Team Science, Analytics, and Systems Thinking in Health Services and Implementation Science Research (CATALYST), College of Medicine, The Ohio State University, Columbus, Ohio (MEG, SRM, ARP, ASM); National Registry of Emergency Medical Technicians, Columbus, Ohio (JRP, JDK, ARP); Division of Epidemiology, The Ohio State University College of Public Health, Columbus, Ohio (JRP, ARP); Division of Biostatistics, The Ohio State University College of Public Health, Columbus, Ohio (EK); Department of Emergency Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio (ARP); Department of Family and Community Medicine, College of Medicine, The Ohio State University, Columbus, Ohio (ASM). Revision received October 8, 2021; accepted for publication October 11, 2021
| | - Ashish R Panchal
- Received August 24, 2021; Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, Ohio (MEG, ASM); The Center for the Advancement of Team Science, Analytics, and Systems Thinking in Health Services and Implementation Science Research (CATALYST), College of Medicine, The Ohio State University, Columbus, Ohio (MEG, SRM, ARP, ASM); National Registry of Emergency Medical Technicians, Columbus, Ohio (JRP, JDK, ARP); Division of Epidemiology, The Ohio State University College of Public Health, Columbus, Ohio (JRP, ARP); Division of Biostatistics, The Ohio State University College of Public Health, Columbus, Ohio (EK); Department of Emergency Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio (ARP); Department of Family and Community Medicine, College of Medicine, The Ohio State University, Columbus, Ohio (ASM). Revision received October 8, 2021; accepted for publication October 11, 2021
| | - Ann Scheck McAlearney
- Received August 24, 2021; Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, Ohio (MEG, ASM); The Center for the Advancement of Team Science, Analytics, and Systems Thinking in Health Services and Implementation Science Research (CATALYST), College of Medicine, The Ohio State University, Columbus, Ohio (MEG, SRM, ARP, ASM); National Registry of Emergency Medical Technicians, Columbus, Ohio (JRP, JDK, ARP); Division of Epidemiology, The Ohio State University College of Public Health, Columbus, Ohio (JRP, ARP); Division of Biostatistics, The Ohio State University College of Public Health, Columbus, Ohio (EK); Department of Emergency Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio (ARP); Department of Family and Community Medicine, College of Medicine, The Ohio State University, Columbus, Ohio (ASM). Revision received October 8, 2021; accepted for publication October 11, 2021
| |
Collapse
|
1528
|
Martinez DR, Schäfer A, Gobeil S, Li D, De la Cruz G, Parks R, Lu X, Barr M, Stalls V, Janowska K, Beaudoin E, Manne K, Mansouri K, Edwards RJ, Cronin K, Yount B, Anasti K, Montgomery SA, Tang J, Golding H, Shen S, Zhou T, Kwong PD, Graham BS, Mascola JR, Montefiori DC, Alam SM, Sempowski GD, Khurana S, Wiehe K, Saunders KO, Acharya P, Haynes BF, Baric RS. A broadly cross-reactive antibody neutralizes and protects against sarbecovirus challenge in mice. Sci Transl Med 2021; 14:eabj7125. [PMID: 34726473 DOI: 10.1126/scitranslmed.abj7125] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- David R Martinez
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alexandra Schäfer
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sophie Gobeil
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Dapeng Li
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Gabriela De la Cruz
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Robert Parks
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Xiaozhi Lu
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Maggie Barr
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Victoria Stalls
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Katarzyna Janowska
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Esther Beaudoin
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Kartik Manne
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Katayoun Mansouri
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Robert J Edwards
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Kenneth Cronin
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Boyd Yount
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kara Anasti
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Stephanie A Montgomery
- Department of Laboratory Medicine and Pathology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Juanjie Tang
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, Maryland, USA, 20871
| | - Hana Golding
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, Maryland, USA, 20871
| | - Shaunna Shen
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Tongqing Zhou
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - Peter D Kwong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - John R Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - David C Montefiori
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - S Munir Alam
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Gregory D Sempowski
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Surender Khurana
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, Maryland, USA, 20871
| | - Kevin Wiehe
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Kevin O Saunders
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA.,Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Priyamvada Acharya
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA.,Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Barton F Haynes
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
1529
|
Barandalla I, Alvarez C, Barreiro P, de Mendoza C, González-Crespo R, Soriano V. Impact of scaling up SARS-CoV-2 vaccination on COVID-19 hospitalizations in Spain. Int J Infect Dis 2021; 112:81-88. [PMID: 34536609 PMCID: PMC8442297 DOI: 10.1016/j.ijid.2021.09.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND The advent of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines has been associated with a significant decline in coronavirus disease 2019 (COVID-19) hospitalizations and deaths. However, little is known about the benefits experienced by different population groups and/or using distinct vaccines. METHODS The Spanish public registry was analyzed to examine associations between weekly vaccination scale-up and the incidence of COVID-19 hospitalizations by age, sex, and vaccine modality. The study period extended from January 2020 to June 2021. RESULTS A total of 363 960 COVID-19 hospitalizations were recorded in Spain during the study period, with three peaks in March 2020, November 2020, and January 2021. The incidence of COVID-19 hospitalizations per 100 000 population increased exponentially with age, on average 71.5% for each decade older. Overall, individuals older than 60 years of age accounted for 65% of all COVID-19 hospitalizations. The speedy vaccination rollout since the end of 2020, with prioritization of the elderly groups, resulted in a rapid fall in COVID-19 hospitalizations starting in February 2021. The benefit was already noticed 3-4 weeks after the first dose, regardless of the vaccine modality. CONCLUSIONS COVID-19 hospitalizations increased exponentially with age in all three peaks of SARS-CoV-2 infection in Spain. Early mass vaccination of people over 60 years of age prevented a fourth wave of COVID-19 hospitalizations during the spring of 2021.
Collapse
Affiliation(s)
| | - Carmen Alvarez
- UNIR Health Sciences School and Medical Center, Madrid, Spain
| | - Pablo Barreiro
- Consejería de Sanidad, Comunidad de Madrid, Madrid, Spain
| | - Carmen de Mendoza
- Puerta de Hierro Research Institute and University Hospital, Madrid, Spain
| | | | - Vicente Soriano
- UNIR Health Sciences School and Medical Center, Madrid, Spain.
| |
Collapse
|
1530
|
Affiliation(s)
- Zahra Rikhtegaran Tehrani
- Division of Clinical Care and Research, Institute of Human Virology, University of Maryland School of Medicine, Baltimore
| | - Mohammad M Sajadi
- Division of Clinical Care and Research, Institute of Human Virology, University of Maryland School of Medicine, Baltimore
- Department of Medicine, Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| |
Collapse
|
1531
|
Mlcochova P, Kemp SA, Dhar MS, Papa G, Meng B, Ferreira IATM, Datir R, Collier DA, Albecka A, Singh S, Pandey R, Brown J, Zhou J, Goonawardane N, Mishra S, Whittaker C, Mellan T, Marwal R, Datta M, Sengupta S, Ponnusamy K, Radhakrishnan VS, Abdullahi A, Charles O, Chattopadhyay P, Devi P, Caputo D, Peacock T, Wattal C, Goel N, Satwik A, Vaishya R, Agarwal M, Mavousian A, Lee JH, Bassi J, Silacci-Fegni C, Saliba C, Pinto D, Irie T, Yoshida I, Hamilton WL, Sato K, Bhatt S, Flaxman S, James LC, Corti D, Piccoli L, Barclay WS, Rakshit P, Agrawal A, Gupta RK. SARS-CoV-2 B.1.617.2 Delta variant replication and immune evasion. Nature 2021; 599:114-119. [PMID: 34488225 DOI: 10.1101/2021.05.08.443253] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 08/23/2021] [Indexed: 05/23/2023]
Abstract
The B.1.617.2 (Delta) variant of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was first identified in the state of Maharashtra in late 2020 and spread throughout India, outcompeting pre-existing lineages including B.1.617.1 (Kappa) and B.1.1.7 (Alpha)1. In vitro, B.1.617.2 is sixfold less sensitive to serum neutralizing antibodies from recovered individuals, and eightfold less sensitive to vaccine-elicited antibodies, compared with wild-type Wuhan-1 bearing D614G. Serum neutralizing titres against B.1.617.2 were lower in ChAdOx1 vaccinees than in BNT162b2 vaccinees. B.1.617.2 spike pseudotyped viruses exhibited compromised sensitivity to monoclonal antibodies to the receptor-binding domain and the amino-terminal domain. B.1.617.2 demonstrated higher replication efficiency than B.1.1.7 in both airway organoid and human airway epithelial systems, associated with B.1.617.2 spike being in a predominantly cleaved state compared with B.1.1.7 spike. The B.1.617.2 spike protein was able to mediate highly efficient syncytium formation that was less sensitive to inhibition by neutralizing antibody, compared with that of wild-type spike. We also observed that B.1.617.2 had higher replication and spike-mediated entry than B.1.617.1, potentially explaining the B.1.617.2 dominance. In an analysis of more than 130 SARS-CoV-2-infected health care workers across three centres in India during a period of mixed lineage circulation, we observed reduced ChAdOx1 vaccine effectiveness against B.1.617.2 relative to non-B.1.617.2, with the caveat of possible residual confounding. Compromised vaccine efficacy against the highly fit and immune-evasive B.1.617.2 Delta variant warrants continued infection control measures in the post-vaccination era.
Collapse
Affiliation(s)
- Petra Mlcochova
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Steven A Kemp
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
- University College London, London, UK
| | | | - Guido Papa
- MRC - Laboratory of Molecular Biology, Cambridge, UK
| | - Bo Meng
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Isabella A T M Ferreira
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Rawlings Datir
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Dami A Collier
- Department of Medicine, University of Cambridge, Cambridge, UK
- University College London, London, UK
| | - Anna Albecka
- MRC - Laboratory of Molecular Biology, Cambridge, UK
| | - Sujeet Singh
- National Centre for Disease Control, Delhi, India
| | - Rajesh Pandey
- CSIR Institute of Genomics and Integrative Biology, Delhi, India
| | - Jonathan Brown
- Department of Infectious Diseases, Imperial College London, London, UK
| | - Jie Zhou
- Department of Infectious Diseases, Imperial College London, London, UK
| | | | - Swapnil Mishra
- Medical Research Council (MRC) Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK
| | - Charles Whittaker
- Medical Research Council (MRC) Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK
| | - Thomas Mellan
- Medical Research Council (MRC) Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK
| | - Robin Marwal
- National Centre for Disease Control, Delhi, India
| | - Meena Datta
- National Centre for Disease Control, Delhi, India
| | | | | | | | - Adam Abdullahi
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | | | | | - Priti Devi
- CSIR Institute of Genomics and Integrative Biology, Delhi, India
| | | | - Tom Peacock
- Medical Research Council (MRC) Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK
| | | | | | | | | | | | | | - Joo Hyeon Lee
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Jessica Bassi
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | | | - Christian Saliba
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Dora Pinto
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Takashi Irie
- Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Isao Yoshida
- Tokyo Metropolitan Institute of Public Health, Tokyo, Japan
| | | | - Kei Sato
- Division of Systems Virology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- CREST, Japan Science and Technology Agency, Saitama, Japan
| | - Samir Bhatt
- National Centre for Disease Control, Delhi, India
- Section of Epidemiology, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Seth Flaxman
- Department of Computer Science, University of Oxford, Oxford, UK
| | - Leo C James
- MRC - Laboratory of Molecular Biology, Cambridge, UK
| | - Davide Corti
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Luca Piccoli
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Wendy S Barclay
- Medical Research Council (MRC) Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK
| | | | - Anurag Agrawal
- CSIR Institute of Genomics and Integrative Biology, Delhi, India.
| | - Ravindra K Gupta
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK.
- Department of Medicine, University of Cambridge, Cambridge, UK.
- Africa Health Research Institute, Durban, South Africa.
| |
Collapse
|
1532
|
Aguiar M, Van-Dierdonck JB, Mar J, Stollenwerk N. The role of mild and asymptomatic infections on COVID-19 vaccines performance: A modeling study. J Adv Res 2021; 39:157-166. [PMID: 35777906 PMCID: PMC8592646 DOI: 10.1016/j.jare.2021.10.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 10/25/2021] [Accepted: 10/27/2021] [Indexed: 01/01/2023] Open
Affiliation(s)
- Maíra Aguiar
- Basque Center for Applied Mathematics (BCAM), Bilbao, Spain; Dipartimento di Matematica, Università degli Studi di Trento, Italy; Ikerbasque, Basque Foundation for Science, Bilbao, Spain.
| | | | - Javier Mar
- Osakidetza Basque Health Service, Debagoiena Integrated Healthcare Organisation, Research Unit, Arrasate-Mondragón, Guipúzcoa, Spain; Biodonostia Health Research Institute, Donostia-San Sebastián, Guipúzcoa, Spain; Kronikgune Institute for Health Services Research, Economic Evaluation Unit, Barakaldo, Spain
| | - Nico Stollenwerk
- Basque Center for Applied Mathematics (BCAM), Bilbao, Spain; Dipartimento di Matematica, Università degli Studi di Trento, Italy
| |
Collapse
|
1533
|
Gong SY, Chatterjee D, Richard J, Prévost J, Tauzin A, Gasser R, Bo Y, Vézina D, Goyette G, Gendron-Lepage G, Medjahed H, Roger M, Côté M, Finzi A. Contribution of single mutations to selected SARS-CoV-2 emerging variants spike antigenicity. Virology 2021; 563:134-145. [PMID: 34536797 PMCID: PMC8433594 DOI: 10.1016/j.virol.2021.09.001] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/01/2021] [Accepted: 09/01/2021] [Indexed: 12/17/2022]
Abstract
Towards the end of 2020, multiple variants of concern (VOCs) and variants of interest (VOIs) have arisen from the original SARS-CoV-2 Wuhan-Hu-1 strain. Mutations in the Spike protein are highly scrutinized for their impact on transmissibility, pathogenesis and vaccine efficacy. Here, we contribute to the growing body of literature on emerging variants by evaluating the impact of single mutations on the overall antigenicity of selected variants and their binding to the ACE2 receptor. We observe a differential contribution of single mutants to the global variants phenotype related to ACE2 interaction and antigenicity. Using biolayer interferometry, we observe that enhanced ACE2 interaction is mostly modulated by a decrease in off-rate. Finally, we made the interesting observation that the Spikes from tested emerging variants bind better to ACE2 at 37°C compared to the D614G variant. Whether improved ACE2 binding at higher temperature facilitates emerging variants transmission remain to be demonstrated.
Collapse
Affiliation(s)
- Shang Yu Gong
- Centre de Recherche du CHUM, QC, H2X 0A9, Canada; Department of Microbiology and Immunology, McGill University, Montreal, QC, H3A 0G4, Canada
| | | | - Jonathan Richard
- Centre de Recherche du CHUM, QC, H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, H2X 0A9, Canada
| | - Jérémie Prévost
- Centre de Recherche du CHUM, QC, H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, H2X 0A9, Canada
| | - Alexandra Tauzin
- Centre de Recherche du CHUM, QC, H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, H2X 0A9, Canada
| | - Romain Gasser
- Centre de Recherche du CHUM, QC, H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, H2X 0A9, Canada
| | - Yuxia Bo
- Department of Biochemistry, Microbiology and Immunology, and Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Dani Vézina
- Centre de Recherche du CHUM, QC, H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, H2X 0A9, Canada
| | | | | | | | - Michel Roger
- Centre de Recherche du CHUM, QC, H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, H2X 0A9, Canada; Laboratoire de Santé Publique du Québec, Institut Nationale de Santé Publique du Québec, Sainte-Anne-de-Bellevue, QC, H9X 3R5, Canada
| | - Marceline Côté
- Department of Biochemistry, Microbiology and Immunology, and Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Andrés Finzi
- Centre de Recherche du CHUM, QC, H2X 0A9, Canada; Department of Microbiology and Immunology, McGill University, Montreal, QC, H3A 0G4, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, H2X 0A9, Canada.
| |
Collapse
|
1534
|
Buchy P, Buisson Y, Cintra O, Dwyer DE, Nissen M, Ortiz de Lejarazu R, Petersen E. COVID-19 pandemic: lessons learned from more than a century of pandemics and current vaccine development for pandemic control. Int J Infect Dis 2021; 112:300-317. [PMID: 34563707 PMCID: PMC8459551 DOI: 10.1016/j.ijid.2021.09.045] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 09/17/2021] [Accepted: 09/19/2021] [Indexed: 01/04/2023] Open
Abstract
Pandemic dynamics and health care responses are markedly different during the COVID-19 pandemic than in earlier outbreaks. Compared with established infectious disease such as influenza, we currently know relatively little about the origin, reservoir, cross-species transmission and evolution of SARS-CoV-2. Health care services, drug availability, laboratory testing, research capacity and global governance are more advanced than during 20th century pandemics, although COVID-19 has highlighted significant gaps. The risk of zoonotic transmission and an associated new pandemic is rising substantially. COVID-19 vaccine development has been done at unprecedented speed, with the usual sequential steps done in parallel. The pandemic has illustrated the feasibility of this approach and the benefits of a globally coordinated response and infrastructure. Some of the COVID-19 vaccines recently developed or currently in development might offer flexibility or sufficiently broad protection to swiftly respond to antigenic drift or emergence of new coronaviruses. Yet many challenges remain, including the large-scale production of sufficient quantity of vaccines, delivery of vaccines to all countries and ensuring vaccination of relevant age groups. This wide vaccine technology approach will be best employed in tandem with active surveillance for emerging variants or new pathogens using antigen mapping, metagenomics and next generation sequencing.
Collapse
Affiliation(s)
| | | | | | - Dominic E Dwyer
- New South Wales Health Pathology - Institute of Clinical Pathology and Medical Research, Westmead Hospital, New South Wales, Australia.
| | - Michael Nissen
- Consultant in Infectious Diseases, University of Queensland, Brisbane, Australia.
| | - Raul Ortiz de Lejarazu
- Scientific Advisor & Emeritus director at Valladolid NIC (National Influenza Centre) Spain, School of Medicine, Avd Ramón y Cajal s/n 47005 Valladolid, Spain.
| | - Eskild Petersen
- European Society for Clinical Microbiology and Infectious Diseases, Basel, Switzerland; Department of Molecular Medicine, The University of Pavia, Pavia, Italy; Department of Clinical, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
1535
|
Shrotri M, Krutikov M, Palmer T, Giddings R, Azmi B, Subbarao S, Fuller C, Irwin-Singer A, Davies D, Tut G, Lopez Bernal J, Moss P, Hayward A, Copas A, Shallcross L. Vaccine effectiveness of the first dose of ChAdOx1 nCoV-19 and BNT162b2 against SARS-CoV-2 infection in residents of long-term care facilities in England (VIVALDI): a prospective cohort study. THE LANCET. INFECTIOUS DISEASES 2021; 21:1529-1538. [PMID: 34174193 PMCID: PMC8221738 DOI: 10.1016/s1473-3099(21)00289-9] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/04/2021] [Accepted: 05/11/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND The effectiveness of SARS-CoV-2 vaccines in older adults living in long-term care facilities is uncertain. We investigated the protective effect of the first dose of the Oxford-AstraZeneca non-replicating viral-vectored vaccine (ChAdOx1 nCoV-19; AZD1222) and the Pfizer-BioNTech mRNA-based vaccine (BNT162b2) in residents of long-term care facilities in terms of PCR-confirmed SARS-CoV-2 infection over time since vaccination. METHODS The VIVALDI study is a prospective cohort study that commenced recruitment on June 11, 2020, to investigate SARS-CoV-2 transmission, infection outcomes, and immunity in residents and staff in long-term care facilities in England that provide residential or nursing care for adults aged 65 years and older. In this cohort study, we included long-term care facility residents undergoing routine asymptomatic SARS-CoV-2 testing between Dec 8, 2020 (the date the vaccine was first deployed in a long-term care facility), and March 15, 2021, using national testing data linked within the COVID-19 Datastore. Using Cox proportional hazards regression, we estimated the relative hazard of PCR-positive infection at 0-6 days, 7-13 days, 14-20 days, 21-27 days, 28-34 days, 35-48 days, and 49 days and beyond after vaccination, comparing unvaccinated and vaccinated person-time from the same cohort of residents, adjusting for age, sex, previous infection, local SARS-CoV-2 incidence, long-term care facility bed capacity, and clustering by long-term care facility. We also compared mean PCR cycle threshold (Ct) values for positive swabs obtained before and after vaccination. The study is registered with ISRCTN, number 14447421. FINDINGS 10 412 care home residents aged 65 years and older from 310 LTCFs were included in this analysis. The median participant age was 86 years (IQR 80-91), 7247 (69·6%) of 10 412 residents were female, and 1155 residents (11·1%) had evidence of previous SARS-CoV-2 infection. 9160 (88·0%) residents received at least one vaccine dose, of whom 6138 (67·0%) received ChAdOx1 and 3022 (33·0%) received BNT162b2. Between Dec 8, 2020, and March 15, 2021, there were 36 352 PCR results in 670 628 person-days, and 1335 PCR-positive infections (713 in unvaccinated residents and 612 in vaccinated residents) were included. Adjusted hazard ratios (HRs) for PCR-positive infection relative to unvaccinated residents declined from 28 days after the first vaccine dose to 0·44 (95% CI 0·24-0·81) at 28-34 days and 0·38 (0·19-0·77) at 35-48 days. Similar effect sizes were seen for ChAdOx1 (adjusted HR 0·32, 95% CI 0·15-0·66) and BNT162b2 (0·35, 0·17-0·71) vaccines at 35-48 days. Mean PCR Ct values were higher for infections that occurred at least 28 days after vaccination than for those occurring before vaccination (31·3 [SD 8·7] in 107 PCR-positive tests vs 26·6 [6·6] in 552 PCR-positive tests; p<0·0001). INTERPRETATION Single-dose vaccination with BNT162b2 and ChAdOx1 vaccines provides substantial protection against infection in older adults from 4-7 weeks after vaccination and might reduce SARS-CoV-2 transmission. However, the risk of infection is not eliminated, highlighting the ongoing need for non-pharmaceutical interventions to prevent transmission in long-term care facilities. FUNDING UK Government Department of Health and Social Care.
Collapse
Affiliation(s)
- Madhumita Shrotri
- UCL Institute of Health Informatics, UCL, London, UK; Public Health England, London, UK
| | | | - Tom Palmer
- UCL Institute for Global Health, UCL, London, UK
| | | | - Borscha Azmi
- UCL Institute of Health Informatics, UCL, London, UK
| | | | | | | | | | - Gokhan Tut
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | | | - Paul Moss
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Andrew Hayward
- UCL Institute of Epidemiology & Healthcare, UCL, London, UK; Health Data Research UK, London, UK
| | - Andrew Copas
- UCL Institute for Global Health, UCL, London, UK
| | | |
Collapse
|
1536
|
Adamson PC, Pfeffer MA, Arboleda VA, Garner OB, de St Maurice A, von Bredow B, Flint J, Kruglyak L, Currier JS. Lower Severe Acute Respiratory Syndrome Coronavirus 2 Viral Shedding Following Coronavirus Disease 2019 Vaccination Among Healthcare Workers in Los Angeles, California. Open Forum Infect Dis 2021; 8:ofab526. [PMID: 35005055 PMCID: PMC8689969 DOI: 10.1093/ofid/ofab526] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 10/13/2021] [Indexed: 11/13/2022] Open
Abstract
Among 880 healthcare workers with a positive severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) test, 264 (30.0%) infections were identified following receipt of at least 1 vaccine dose. Median SARS-CoV-2 cycle threshold values were highest among individuals receiving 2 vaccine doses, corresponding to lower viral shedding. Vaccination might lead to lower transmissibility of SARS-CoV-2.
Collapse
Affiliation(s)
- Paul C Adamson
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Michael A Pfeffer
- Department of Medicine/Department of Information Services and Solutions, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Valerie A Arboleda
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Omai B Garner
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Annabelle de St Maurice
- Division of Pediatric Infectious Diseases, Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Benjamin von Bredow
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Jonathan Flint
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Leonid Kruglyak
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Howard Hughes Medical Institute, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Department of Biological Chemistry, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Judith S Currier
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
1537
|
Duarte G, Coutinho CM, Rolnik DL, Quintana SM, Rabelo e Silva AC, Poon LC, Costa FDS. Perspectives on administration of COVID-19 vaccine to pregnant and lactating women: a challenge for low- and middle-income countries. AJOG GLOBAL REPORTS 2021; 1:100020. [PMID: 34494014 PMCID: PMC8413092 DOI: 10.1016/j.xagr.2021.100020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 12/13/2022] Open
Abstract
Women who are in the pregnancy-puerperal cycle or are lactating have been deliberately excluded from participating in COVID-19 vaccine clinical trials that aimed to evaluate either the efficacy of the vaccines in inducing the formation of neutralizing antibodies or the investigational products' safety profile. The exclusion of pregnant and lactating women from such studies certainly and inequitably denies these women access to COVID-19 vaccines, since these products have become increasingly available to nonpregnant people and even to those who are pregnant and are in high-income settings. In this clinical opinion article, we discuss some aspects of the prolonged pandemic, the emergence of viral variants, the risks of severe complications of COVID-19 in pregnant women, and the disproportionate impact of the above on low- and middle-income countries. We argue that the decision to receive the COVID-19 vaccine should be a joint decision between the pregnant or lactating women and the healthcare providers, while considering the available data on vaccine efficacy, safety, the risks of SARS-CoV-2 infection in pregnant women, and the women's individual risks for infection and serious illness. The various types of vaccines that are already in use and their safety, effectiveness, and the potential risks and benefits of their administration to pregnant or lactating women are also reviewed.
Collapse
Affiliation(s)
- Geraldo Duarte
- Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil (Drs Duarte, Coutinho, and Quintana and Ms Silva)
| | - Conrado Milani Coutinho
- Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil (Drs Duarte, Coutinho, and Quintana and Ms Silva)
| | - Daniel Lorber Rolnik
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia (Dr Rolnik)
| | - Silvana Maria Quintana
- Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil (Drs Duarte, Coutinho, and Quintana and Ms Silva)
| | - Ana Cláudia Rabelo e Silva
- Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil (Drs Duarte, Coutinho, and Quintana and Ms Silva)
| | - Liona C. Poon
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong SAR, China (Dr Poon)
| | - Fabrício da Silva Costa
- Maternal-Fetal Medicine Unit, Gold Coast University Hospital and School of Medicine, Griffith University, Gold Coast, Queensland, Australia (Dr Costa)
| |
Collapse
|
1538
|
Corchado-Garcia J, Zemmour D, Hughes T, Bandi H, Cristea-Platon T, Lenehan P, Pawlowski C, Bade S, O’Horo JC, Gores GJ, Williams AW, Badley AD, Halamka J, Virk A, Swift MD, Wagner T, Soundararajan V. Analysis of the Effectiveness of the Ad26.COV2.S Adenoviral Vector Vaccine for Preventing COVID-19. JAMA Netw Open 2021; 4:e2132540. [PMID: 34726743 PMCID: PMC8564583 DOI: 10.1001/jamanetworkopen.2021.32540] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
IMPORTANCE Continuous assessment of the effectiveness and safety of the US Food and Drug Administration-authorized SARS-CoV-2 vaccines is critical to amplify transparency, build public trust, and ultimately improve overall health outcomes. OBJECTIVE To evaluate the effectiveness of the Johnson & Johnson Ad26.COV2.S vaccine for preventing SARS-CoV-2 infection. DESIGN, SETTING, AND PARTICIPANTS This comparative effectiveness research study used large-scale longitudinal curation of electronic health records from the multistate Mayo Clinic Health System (Minnesota, Arizona, Florida, Wisconsin, and Iowa) to identify vaccinated and unvaccinated adults between February 27 and July 22, 2021. The unvaccinated cohort was matched on a propensity score derived from age, sex, zip code, race, ethnicity, and previous number of SARS-CoV-2 polymerase chain reaction tests. The final study cohort consisted of 8889 patients in the vaccinated group and 88 898 unvaccinated matched patients. EXPOSURE Single dose of the Ad26.COV2.S vaccine. MAIN OUTCOMES AND MEASURES The incidence rate ratio of SARS-CoV-2 infection in the vaccinated vs unvaccinated control cohorts, measured by SARS-CoV-2 polymerase chain reaction testing. RESULTS The study was composed of 8889 vaccinated patients (4491 men [50.5%]; mean [SD] age, 52.4 [16.9] years) and 88 898 unvaccinated patients (44 748 men [50.3%]; mean [SD] age, 51.7 [16.7] years). The incidence rate ratio of SARS-CoV-2 infection in the vaccinated vs unvaccinated control cohorts was 0.26 (95% CI, 0.20-0.34) (60 of 8889 vaccinated patients vs 2236 of 88 898 unvaccinated individuals), which corresponds to an effectiveness of 73.6% (95% CI, 65.9%-79.9%) and a 3.73-fold reduction in SARS-CoV-2 infections. CONCLUSIONS AND RELEVANCE This study's findings are consistent with the clinical trial-reported efficacy of Ad26.COV2.S and the first retrospective analysis, suggesting that the vaccine is effective at reducing SARS-CoV-2 infection, even with the spread of variants such as Alpha or Delta that were not present in the original studies, and reaffirm the urgent need to continue mass vaccination efforts globally.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Sairam Bade
- nference Labs, Murgesh Pallya, Bengaluru, Karnataka, India
| | - John C. O’Horo
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Gregory J. Gores
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Amy W. Williams
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Andrew D. Badley
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - John Halamka
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Abinash Virk
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Melanie D. Swift
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | | | - Venky Soundararajan
- nference, Cambridge, Massachusetts
- nference Labs, Murgesh Pallya, Bengaluru, Karnataka, India
| |
Collapse
|
1539
|
Moghadas SM, Sah P, Shoukat A, Meyers LA, Galvani AP. Population Immunity Against COVID-19 in the United States. Ann Intern Med 2021; 174:1586-1591. [PMID: 34516275 PMCID: PMC8442744 DOI: 10.7326/m21-2721] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND As of 28 July 2021, 60% of adults in the United States had been fully vaccinated against COVID-19, and more than 34 million cases had been reported. Given the uncertainty regarding undocumented infections, the population level of immunity against COVID-19 in the United States remains undetermined. OBJECTIVE To estimate the population immunity, defined as the proportion of the population that is protected against SARS-CoV-2 infection due to prior infection or vaccination. DESIGN Statistical and simulation modeling to estimate overall and age-specific population immunity. SETTING United States. PARTICIPANTS Simulated age-stratified population representing U.S. demographic characteristics. MEASUREMENTS The true number of SARS-CoV-2 infections in the United States was inferred from data on reported deaths using age-specific infection-fatality rates (IFRs). Taking into account the estimates for vaccine effectiveness and protection against reinfection, the overall population immunity was determined as the sum of protection levels in vaccinated persons and those who were previously infected but not vaccinated. RESULTS Using age-specific IFR estimates from the Centers for Disease Control and Prevention, it was estimated that as of 15 July 2021, 114.9 (95% credible interval [CrI], 103.2 to 127.4) million persons had been infected with SARS-CoV-2 in the United States. The mean overall population immunity was 62.0% (CrI, 58.4% to 66.4%). Adults aged 65 years or older were estimated to have the highest immunity level (77.2% [CrI, 76.2% to 78.6%]), and children younger than 12 years had the lowest immunity level (17.9% [CrI, 14.4% to 21.9%]). LIMITATION Publicly reported deaths may underrepresent actual deaths. CONCLUSION As of 15 July 2021, the U.S. population immunity against COVID-19 may still have been insufficient to contain the outbreaks and safely revert to prepandemic social behavior. PRIMARY FUNDING SOURCE National Science Foundation, National Institutes of Health, Notsew Orm Sands Foundation, Canadian Institutes of Health Research, and Natural Sciences and Engineering Research Council of Canada.
Collapse
Affiliation(s)
| | - Pratha Sah
- Yale School of Public Health, New Haven, Connecticut (P.S., A.S., A.P.G.)
| | - Affan Shoukat
- Yale School of Public Health, New Haven, Connecticut (P.S., A.S., A.P.G.)
| | - Lauren Ancel Meyers
- The University of Texas at Austin, Austin, Texas, and Santa Fe Institute, Santa Fe, New Mexico (L.A.M.)
| | - Alison P Galvani
- Yale School of Public Health, New Haven, Connecticut (P.S., A.S., A.P.G.)
| |
Collapse
|
1540
|
Shrotri M, Krutikov M, Palmer T, Giddings R, Azmi B, Subbarao S, Fuller C, Irwin-Singer A, Davies D, Tut G, Lopez Bernal J, Moss P, Hayward A, Copas A, Shallcross L. Vaccine effectiveness of the first dose of ChAdOx1 nCoV-19 and BNT162b2 against SARS-CoV-2 infection in residents of long-term care facilities in England (VIVALDI): a prospective cohort study. THE LANCET. INFECTIOUS DISEASES 2021. [PMID: 34174193 DOI: 10.1101/2021.03.26.21254391] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
BACKGROUND The effectiveness of SARS-CoV-2 vaccines in older adults living in long-term care facilities is uncertain. We investigated the protective effect of the first dose of the Oxford-AstraZeneca non-replicating viral-vectored vaccine (ChAdOx1 nCoV-19; AZD1222) and the Pfizer-BioNTech mRNA-based vaccine (BNT162b2) in residents of long-term care facilities in terms of PCR-confirmed SARS-CoV-2 infection over time since vaccination. METHODS The VIVALDI study is a prospective cohort study that commenced recruitment on June 11, 2020, to investigate SARS-CoV-2 transmission, infection outcomes, and immunity in residents and staff in long-term care facilities in England that provide residential or nursing care for adults aged 65 years and older. In this cohort study, we included long-term care facility residents undergoing routine asymptomatic SARS-CoV-2 testing between Dec 8, 2020 (the date the vaccine was first deployed in a long-term care facility), and March 15, 2021, using national testing data linked within the COVID-19 Datastore. Using Cox proportional hazards regression, we estimated the relative hazard of PCR-positive infection at 0-6 days, 7-13 days, 14-20 days, 21-27 days, 28-34 days, 35-48 days, and 49 days and beyond after vaccination, comparing unvaccinated and vaccinated person-time from the same cohort of residents, adjusting for age, sex, previous infection, local SARS-CoV-2 incidence, long-term care facility bed capacity, and clustering by long-term care facility. We also compared mean PCR cycle threshold (Ct) values for positive swabs obtained before and after vaccination. The study is registered with ISRCTN, number 14447421. FINDINGS 10 412 care home residents aged 65 years and older from 310 LTCFs were included in this analysis. The median participant age was 86 years (IQR 80-91), 7247 (69·6%) of 10 412 residents were female, and 1155 residents (11·1%) had evidence of previous SARS-CoV-2 infection. 9160 (88·0%) residents received at least one vaccine dose, of whom 6138 (67·0%) received ChAdOx1 and 3022 (33·0%) received BNT162b2. Between Dec 8, 2020, and March 15, 2021, there were 36 352 PCR results in 670 628 person-days, and 1335 PCR-positive infections (713 in unvaccinated residents and 612 in vaccinated residents) were included. Adjusted hazard ratios (HRs) for PCR-positive infection relative to unvaccinated residents declined from 28 days after the first vaccine dose to 0·44 (95% CI 0·24-0·81) at 28-34 days and 0·38 (0·19-0·77) at 35-48 days. Similar effect sizes were seen for ChAdOx1 (adjusted HR 0·32, 95% CI 0·15-0·66) and BNT162b2 (0·35, 0·17-0·71) vaccines at 35-48 days. Mean PCR Ct values were higher for infections that occurred at least 28 days after vaccination than for those occurring before vaccination (31·3 [SD 8·7] in 107 PCR-positive tests vs 26·6 [6·6] in 552 PCR-positive tests; p<0·0001). INTERPRETATION Single-dose vaccination with BNT162b2 and ChAdOx1 vaccines provides substantial protection against infection in older adults from 4-7 weeks after vaccination and might reduce SARS-CoV-2 transmission. However, the risk of infection is not eliminated, highlighting the ongoing need for non-pharmaceutical interventions to prevent transmission in long-term care facilities. FUNDING UK Government Department of Health and Social Care.
Collapse
Affiliation(s)
- Madhumita Shrotri
- UCL Institute of Health Informatics, UCL, London, UK; Public Health England, London, UK
| | | | - Tom Palmer
- UCL Institute for Global Health, UCL, London, UK
| | | | - Borscha Azmi
- UCL Institute of Health Informatics, UCL, London, UK
| | | | | | | | | | - Gokhan Tut
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | | | - Paul Moss
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Andrew Hayward
- UCL Institute of Epidemiology & Healthcare, UCL, London, UK; Health Data Research UK, London, UK
| | - Andrew Copas
- UCL Institute for Global Health, UCL, London, UK
| | | |
Collapse
|
1541
|
Gong SY, Chatterjee D, Richard J, Prévost J, Tauzin A, Gasser R, Bo Y, Vézina D, Goyette G, Gendron-Lepage G, Medjahed H, Roger M, Côté M, Finzi A. Contribution of single mutations to selected SARS-CoV-2 emerging variants spike antigenicity. Virology 2021. [PMID: 34536797 DOI: 10.1101/2021.08.04.455140v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Towards the end of 2020, multiple variants of concern (VOCs) and variants of interest (VOIs) have arisen from the original SARS-CoV-2 Wuhan-Hu-1 strain. Mutations in the Spike protein are highly scrutinized for their impact on transmissibility, pathogenesis and vaccine efficacy. Here, we contribute to the growing body of literature on emerging variants by evaluating the impact of single mutations on the overall antigenicity of selected variants and their binding to the ACE2 receptor. We observe a differential contribution of single mutants to the global variants phenotype related to ACE2 interaction and antigenicity. Using biolayer interferometry, we observe that enhanced ACE2 interaction is mostly modulated by a decrease in off-rate. Finally, we made the interesting observation that the Spikes from tested emerging variants bind better to ACE2 at 37°C compared to the D614G variant. Whether improved ACE2 binding at higher temperature facilitates emerging variants transmission remain to be demonstrated.
Collapse
Affiliation(s)
- Shang Yu Gong
- Centre de Recherche du CHUM, QC, H2X 0A9, Canada; Department of Microbiology and Immunology, McGill University, Montreal, QC, H3A 0G4, Canada
| | | | - Jonathan Richard
- Centre de Recherche du CHUM, QC, H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, H2X 0A9, Canada
| | - Jérémie Prévost
- Centre de Recherche du CHUM, QC, H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, H2X 0A9, Canada
| | - Alexandra Tauzin
- Centre de Recherche du CHUM, QC, H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, H2X 0A9, Canada
| | - Romain Gasser
- Centre de Recherche du CHUM, QC, H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, H2X 0A9, Canada
| | - Yuxia Bo
- Department of Biochemistry, Microbiology and Immunology, and Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Dani Vézina
- Centre de Recherche du CHUM, QC, H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, H2X 0A9, Canada
| | | | | | | | - Michel Roger
- Centre de Recherche du CHUM, QC, H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, H2X 0A9, Canada; Laboratoire de Santé Publique du Québec, Institut Nationale de Santé Publique du Québec, Sainte-Anne-de-Bellevue, QC, H9X 3R5, Canada
| | - Marceline Côté
- Department of Biochemistry, Microbiology and Immunology, and Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Andrés Finzi
- Centre de Recherche du CHUM, QC, H2X 0A9, Canada; Department of Microbiology and Immunology, McGill University, Montreal, QC, H3A 0G4, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, H2X 0A9, Canada.
| |
Collapse
|
1542
|
Butler M, Tamborska A, Wood GK, Ellul M, Thomas RH, Galea I, Pett S, Singh B, Solomon T, Pollak TA, Michael BD, Nicholson TR. Considerations for causality assessment of neurological and neuropsychiatric complications of SARS-CoV-2 vaccines: from cerebral venous sinus thrombosis to functional neurological disorder. J Neurol Neurosurg Psychiatry 2021; 92:1144-1151. [PMID: 34362855 DOI: 10.1136/jnnp-2021-326924] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 06/21/2021] [Indexed: 12/26/2022]
Affiliation(s)
- Matt Butler
- Institute of Psychiatry Psychology and Neuroscience, London, UK
| | - Arina Tamborska
- Department of Neurology, The Walton Centre NHS Foundation Trust, Liverpool, UK.,University of Liverpool, Liverpool, UK
| | - Greta K Wood
- Department of Neurology, The Walton Centre NHS Foundation Trust, Liverpool, UK.,University of Liverpool, Liverpool, UK
| | - Mark Ellul
- Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
| | - Rhys H Thomas
- Department of Neuroscience, Newcastle University, Newcastle upon Tyne, Tyne and Wear, UK.,Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | - Ian Galea
- Faculty of Medicine, University of Southampton, Southampton, UK
| | - Sarah Pett
- MRC CTU at UCL, Institute for Global Health and Institute for Clinical Trials Methodology, University College London, London, UK
| | | | - Tom Solomon
- Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
| | - Thomas Arthur Pollak
- Department of Psychological Medicine, Institute of Psychiatry, King's College London, London, UK
| | - Benedict D Michael
- Department of Neurology, The Walton Centre NHS Foundation Trust, Liverpool, UK.,University of Liverpool, Liverpool, UK
| | | |
Collapse
|
1543
|
Tsai SJ, Atai NA, Cacciottolo M, Nice J, Salehi A, Guo C, Sedgwick A, Kanagavelu S, Gould SJ. Exosome-mediated mRNA delivery in vivo is safe and can be used to induce SARS-CoV-2 immunity. J Biol Chem 2021; 297:101266. [PMID: 34600888 PMCID: PMC8483990 DOI: 10.1016/j.jbc.2021.101266] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 09/17/2021] [Accepted: 09/28/2021] [Indexed: 12/15/2022] Open
Abstract
Functional delivery of mRNA has high clinical potential. Previous studies established that mRNAs can be delivered to cells in vitro and in vivo via RNA-loaded lipid nanoparticles (LNPs). Here we describe an alternative approach using exosomes, the only biologically normal nanovesicle. In contrast to LNPs, which elicited pronounced cellular toxicity, exosomes had no adverse effects in vitro or in vivo at any dose tested. Moreover, mRNA-loaded exosomes were characterized by efficient mRNA encapsulation (∼90%), high mRNA content, consistent size, and a polydispersity index under 0.2. Using an mRNA encoding the red light-emitting luciferase Antares2, we observed that mRNA-loaded exosomes were superior to mRNA-loaded LNPs at delivering functional mRNA into human cells in vitro. Injection of Antares2 mRNA-loaded exosomes also led to strong light emission following injection into the vitreous fluid of the eye or into the tissue of skeletal muscle in mice. Furthermore, we show that repeated injection of Antares2 mRNA-loaded exosomes drove sustained luciferase expression across six injections spanning at least 10 weeks, without evidence of signal attenuation or adverse injection site responses. Consistent with these findings, we observed that exosomes loaded with mRNAs encoding immunogenic forms of the SARS-CoV-2 Spike and Nucleocapsid proteins induced long-lasting cellular and humoral responses to both. Taken together, these results demonstrate that exosomes can be used to deliver functional mRNA to and into cells in vivo.
Collapse
Affiliation(s)
- Shang Jui Tsai
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nadia A Atai
- Capricor Therapeutics, Inc, Beverly Hills, California, USA
| | | | - Justin Nice
- Capricor Therapeutics, Inc, Beverly Hills, California, USA
| | - Arjang Salehi
- Capricor Therapeutics, Inc, Beverly Hills, California, USA
| | - Chenxu Guo
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | - Stephen J Gould
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
1544
|
Delmonte OM, Bergerson JRE, Burbelo PD, Durkee-Shock JR, Dobbs K, Bosticardo M, Keller MD, McDermott DH, Rao VK, Dimitrova D, Quiros-Roldan E, Imberti L, Ferrè EMN, Schmitt M, Lafeer C, Pfister J, Shaw D, Draper D, Truong M, Ulrick J, DiMaggio T, Urban A, Holland SM, Lionakis MS, Cohen JI, Ricotta EE, Notarangelo LD, Freeman AF. Antibody responses to the SARS-CoV-2 vaccine in individuals with various inborn errors of immunity. J Allergy Clin Immunol 2021; 148:1192-1197. [PMID: 34492260 PMCID: PMC8418380 DOI: 10.1016/j.jaci.2021.08.016] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/19/2021] [Accepted: 08/24/2021] [Indexed: 01/05/2023]
Abstract
BACKGROUND SARS-CoV-2 vaccination is recommended in patients with inborn errors of immunity (IEIs); however, little is known about immunogenicity and safety in these patients. OBJECTIVE We sought to evaluate the impact of genetic diagnosis, age, and treatment on antibody response to COVID-19 vaccine and related adverse events in a cohort of patients with IEIs. METHODS Plasma was collected from 22 health care worker controls, 81 patients with IEIs, and 2 patients with thymoma; the plasma was collected before immunization, 1 to 6 days before the second dose of mRNA vaccine, and at a median of 30 days after completion of the immunization schedule with either mRNA vaccine or a single dose of Johnson & Johnson's Janssen vaccine. Anti-spike (anti-S) and anti-nucleocapsid antibody titers were measured by using a luciferase immunoprecipitation systems method. Information on T- and B-cell counts and use of immunosuppressive drugs was extracted from medical records, and information on vaccine-associated adverse events was collected after each dose. RESULTS Anti-S antibodies were detected in 27 of 46 patients (58.7%) after 1 dose of mRNA vaccine and in 63 of 74 fully immunized patients (85.1%). A lower rate of seroconversion (7 of 11 [63.6%]) was observed in patients with autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy. Previous use of rituximab and baseline counts of less than 1000 CD3+ T cells/mL and less than 100 CD19+ B cells/mL were associated with lower anti-S IgG levels. No significant adverse events were reported. CONCLUSION Vaccinating patients with IEIs is safe, but immunogenicity is affected by certain therapies and gene defects. These data may guide the counseling of patients with IEIs regarding prevention of SARS-CoV-2 infection and the need for subsequent boosts.
Collapse
Affiliation(s)
- Ottavia M Delmonte
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Jenna R E Bergerson
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Peter D Burbelo
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Md
| | - Jessica R Durkee-Shock
- Center for Cancer and Immunology Research and Division of Allergy and Immunology, Children's National Hospital, Washington, DC
| | - Kerry Dobbs
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Marita Bosticardo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Michael D Keller
- Center for Cancer and Immunology Research and Division of Allergy and Immunology, Children's National Hospital, Washington, DC
| | - David H McDermott
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - V Koneti Rao
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Dimana Dimitrova
- Experimental Transplantation and Immunotherapy Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md
| | - Eugenia Quiros-Roldan
- Department of Infectious and Tropical Diseases, University of Brescia and ASST Spedali Civili di Brescia, Brescia, Italy; CREA Laboratory, Diagnostic Laboratory, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Luisa Imberti
- CREA Laboratory, Diagnostic Laboratory, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Elise M N Ferrè
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Monica Schmitt
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Christine Lafeer
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Justina Pfister
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Dawn Shaw
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Deborah Draper
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Meng Truong
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Jean Ulrick
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Tom DiMaggio
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Amanda Urban
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Steven M Holland
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Michail S Lionakis
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Jeffrey I Cohen
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Emily E Ricotta
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md.
| | - Alexandra F Freeman
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md.
| |
Collapse
|
1545
|
Rapid and Quantitative Detection of Human Antibodies against the 2019 Novel Coronavirus SARS CoV2 and Its Variants as a Result of Vaccination and Infection. Microbiol Spectr 2021; 9:e0089021. [PMID: 34585942 PMCID: PMC8557906 DOI: 10.1128/spectrum.00890-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Measuring the antibody response to 2019 SARS CoV2 is critical for diagnostic purposes, for monitoring the prevalence of infection, and for gauging the efficacy of the worldwide vaccination effort for COVID-19. In this study, a microchip-based grating-coupled fluorescent plasmonic (GC-FP) assay was used to measure antibody levels that resulted from COVID-19 infection and vaccination. In addition, we measured the relative antibody binding toward antigens from the CoV2 virus variants strains B.1.1.7 (Alpha) and B.1.351 (Beta). Antibody levels against multiple antigens within the SARS CoV2 spike protein were significantly elevated for both vaccinated and infected individuals, while those against the nucleocapsid (N) protein were only elevated for infected individuals. GC-FP was effective for monitoring the IgG-based serological response to vaccination throughout the vaccination sequence and also resolved acute (within hours) increases in antibody levels. A significant decrease in antibody binding to antigens from the B.1.351 variant, but not B.1.1.7, was observed for all vaccinated subjects when measured by GC-FP compared to the 2019 SARS CoV2 antigens. These results were corroborated by competitive enzyme-linked immunosorbent assay (ELISA). Collectively, the findings suggest that GC-FP is a viable, rapid, and accurate method for measuring both overall antibody levels to SARS CoV2 and relative antibody binding to viral variants during infection or vaccination. IMPORTANCE In this work, a novel biosensor technology was used to measure antibody levels that resulted from vaccination against COVID-19 and/or from infection with the virus. Importantly, this approach enables quantification of antibody levels, which can provide information about the timing and level of immune response. Due the multiplexed nature of this approach, antibody binding to both the original 2019 SARS CoV-2 strain and variant strains can be performed simultaneously and in a short (30-min) time frame.
Collapse
|
1546
|
Itell HL, Weight H, Fish CS, Logue JK, Franko N, Wolf CR, McCulloch DJ, Galloway J, Matsen FA, Chu HY, Overbaugh J. SARS-CoV-2 Antibody Binding and Neutralization in Dried Blood Spot Eluates and Paired Plasma. Microbiol Spectr 2021; 9:e0129821. [PMID: 34668728 PMCID: PMC8528110 DOI: 10.1128/spectrum.01298-21] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 09/17/2021] [Indexed: 11/20/2022] Open
Abstract
Wide-scale assessment of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific antibodies is critical to understanding population seroprevalence, correlates of protection, and the longevity of vaccine-elicited responses. Most SARS-CoV-2 studies characterize antibody responses in plasma/sera. While reliable and broadly used, these samples pose several logistical restrictions, such as requiring venipuncture for collection and a cold chain for transportation and storage. Dried blood spots (DBS) overcome these barriers as they can be self-collected by fingerstick and mailed and stored at ambient temperature. Here, we evaluate the suitability of DBS for SARS-CoV-2 antibody assays by comparing several antibody responses between paired plasma and DBS from SARS-CoV-2 convalescent and vaccinated individuals. We found that DBS not only reflected plasma antibody binding by enzyme-linked immunosorbent assay (ELISA) and epitope profiles using phage display, but also yielded SARS-CoV-2 neutralization titers that highly correlated with paired plasma. Neutralization measurement was further streamlined by adapting assays to a high-throughput 384-well format. This study supports the adoption of DBS for numerous SARS-CoV-2 binding and neutralization assays. IMPORTANCE Plasma and sera isolated from venous blood represent conventional sample types used for the evaluation of SARS-CoV-2 antibody responses after infection or vaccination. However, collection of these samples is invasive and requires trained personnel and equipment for immediate processing. Once collected, plasma and sera must be stored and shipped at cold temperatures. To define the risk of emerging SARS-CoV-2 variants and the longevity of immune responses to natural infection and vaccination, it will be necessary to measure various antibody features in populations around the world, including in resource-limited areas. A sampling method that is compatible with these settings and is suitable for a variety of SARS-CoV-2 antibody assays is therefore needed to continue to understand and curb the COVID-19 pandemic.
Collapse
Affiliation(s)
- Hannah L. Itell
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Molecular and Cellular Biology Graduate Program, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Haidyn Weight
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Carolyn S. Fish
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Jennifer K. Logue
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Nicholas Franko
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Caitlin R. Wolf
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | | | - Jared Galloway
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Frederick A. Matsen
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Helen Y. Chu
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Julie Overbaugh
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| |
Collapse
|
1547
|
Morawska M. Reasons and consequences of COVID-19 vaccine failure in patients with chronic lymphocytic leukemia. Eur J Haematol 2021; 108:91-98. [PMID: 34717004 PMCID: PMC8652891 DOI: 10.1111/ejh.13722] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/19/2021] [Accepted: 10/25/2021] [Indexed: 12/15/2022]
Abstract
People with hematologic malignancies are at a high risk of morbidity and mortality from COVID-19. The response to vaccination is highly limited in patients with chronic lymphocytic leukemia. Less than half of the patients develop antibody response, suggesting that they remain at risk of SARS-CoV-2 infection even after the vaccination. Reasons for inadequate response to COVID-19 vaccination in chronic lymphocytic leukemia are multifactorial and attributed to disease-related immune dysregulation and patient- and therapy-related factors. The negative predictors of response to vaccination include hypogammaglobulinemia, advanced age, current active treatment, and past treatment anti-CD20 monoclonal antibodies. Despite using booster doses and heterologous immunization to improve humoral and cellular immunity, some patients with chronic lymphocytic leukemia will fail to respond. Active treatment at the time of vaccination and a recent history of anti-CD20 monoclonal antibodies use are the strongest predictors of the non-response. Current data support informing patients with chronic lymphocytic leukemia and other hematologic malignancies about the risk of infection regardless of vaccination. These individuals and members of their households should continue extreme preventive actions despite relaxed local regulations. Other emerging non-vaccine preventive strategies include passive and post-exposure prevention with monoclonal antibodies.
Collapse
Affiliation(s)
- Marta Morawska
- Experimental Hematooncology Department, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
1548
|
Shoukat A, Vilches TN, Moghadas SM, Sah P, Schneider EC, Shaff J, Ternier A, Chokshi DA, Galvani AP. Lives saved and hospitalizations averted by COVID-19 vaccination in New York City: a modeling study. LANCET REGIONAL HEALTH. AMERICAS 2021; 5:100085. [PMID: 34746912 PMCID: PMC8556548 DOI: 10.1016/j.lana.2021.100085] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/29/2021] [Accepted: 09/08/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Following the start of COVID-19 vaccination in New York City (NYC), cases have declined over 10-fold from the outbreak peak in January 2020, despite the emergence of highly transmissible variants. We evaluated the impact of NYC's vaccination campaign on saving lives as well as averting hospitalizations and cases. METHODS We used an age-stratified agent-based model of COVID-19 to include transmission dynamics of Alpha, Gamma, Delta and Iota variants as identified in NYC. The model was calibrated and fitted to reported incidence in NYC, accounting for the relative transmissibility of each variant and vaccination rollout data. We simulated COVID-19 outbreak in NYC under the counterfactual scenario of no vaccination and compared the resulting disease burden with the number of cases, hospitalizations and deaths reported under the actual pace of vaccination. FINDINGS We found that without vaccination, there would have been a spring-wave of COVID-19 in NYC due to the spread of Alpha and Delta variants. The COVID-19 vaccination campaign in NYC prevented such a wave, and averted 290,467 (95% CrI: 232,551 - 342,664) cases, 48,076 (95% CrI: 42,264 - 53,301) hospitalizations, and 8,508 (95% CrI: 7,374 - 9,543) deaths from December 14, 2020 to July 15, 2021. INTERPRETATION Our study demonstrates that the vaccination program in NYC was instrumental to substantially reducing the COVID-19 burden and suppressing a surge of cases attributable to more transmissible variants. As the Delta variant sweeps predominantly among unvaccinated individuals, our findings underscore the urgent need to accelerate vaccine uptake and close the vaccination coverage gaps. FUNDING This study was supported by The Commonwealth Fund.
Collapse
Affiliation(s)
- Affan Shoukat
- Center for Infectious Disease Modeling and Analysis (CIDMA), Yale School of Public Health, New Haven, Connecticut, USA
| | - Thomas N. Vilches
- Center for Infectious Disease Modeling and Analysis (CIDMA), Yale School of Public Health, New Haven, Connecticut, USA,Agent-Based Modelling Laboratory, York University, Toronto, Ontario, Canada
| | - Seyed M. Moghadas
- Agent-Based Modelling Laboratory, York University, Toronto, Ontario, Canada
| | - Pratha Sah
- Center for Infectious Disease Modeling and Analysis (CIDMA), Yale School of Public Health, New Haven, Connecticut, USA
| | | | - Jaimie Shaff
- New York City Department of Health and Mental Hygiene, New York, NY, USA
| | - Alexandra Ternier
- New York City Department of Health and Mental Hygiene, New York, NY, USA
| | - Dave A. Chokshi
- New York City Department of Health and Mental Hygiene, New York, NY, USA
| | - Alison P. Galvani
- Center for Infectious Disease Modeling and Analysis (CIDMA), Yale School of Public Health, New Haven, Connecticut, USA,Corresponding author: Alison P. Galvani. Yale University School of Public Health, Center for Infectious Disease Modeling and Analysis, 135 College, New Haven, Connecticut 06510, USA.
| |
Collapse
|
1549
|
Lazaros G, Anastassopoulou C, Hatziantoniou S, Kalos T, Soulaidopoulos S, Lazarou E, Vlachopoulos C, Vassilopoulos D, Tsakris A, Tsioufis C. A case series of acute pericarditis following COVID-19 vaccination in the context of recent reports from Europe and the United States. Vaccine 2021; 39:6585-6590. [PMID: 34635376 PMCID: PMC8491922 DOI: 10.1016/j.vaccine.2021.09.078] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 12/14/2022]
Abstract
Background COVID-19 vaccines were efficacious and safe in clinical trials. We report nine events of acute pericarditis (AP) in eight patients following COVID-19 vaccination with BNT162b2 (6/9), AZD1222 (2/9) and mRNA-1273 (1/9). Methods All patients were referred for AP temporally linked with COVID-19 vaccination. Chest pain was the most common clinical manifestation. Alternative etiologies were excluded upon thorough diagnostic work up. AP diagnosis was established according to ESC guidelines. Findings Five events occurred after the first vaccine dose and four after the second. The mean age in this cohort was 65.8 ± 10.2 years and the men/women ratio 3/5. All events resolved without sequelae; two events were complicated by cardiac tamponade requiring emergent pericardial decompression. Hospitalization was required in four cases. Interpretation Although causality cannot be firmly established, AP has emerged as a possible complication following COVID-19 vaccination. Further investigation is indispensable to fully characterize this new entity.
Collapse
Affiliation(s)
- George Lazaros
- First Cardiology Department, School of Medicine, Hippokration General Hospital, National and Kapodistrian University of Athens, Athens, Greece.
| | - Cleo Anastassopoulou
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Sophia Hatziantoniou
- Laboratory of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, University of Patras, Patras, Greece
| | - Theodoros Kalos
- First Cardiology Department, School of Medicine, Hippokration General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Stergios Soulaidopoulos
- First Cardiology Department, School of Medicine, Hippokration General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Emilia Lazarou
- First Cardiology Department, School of Medicine, Hippokration General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Charalambos Vlachopoulos
- First Cardiology Department, School of Medicine, Hippokration General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios Vassilopoulos
- Second Department of Medicine and Laboratory, Clinical Immunology-Rheumatology Unit, School of Medicine, Hippokration General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Athanasios Tsakris
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Costas Tsioufis
- First Cardiology Department, School of Medicine, Hippokration General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
1550
|
Insights into COVID-19 Vaccine Development Based on Immunogenic Structural Proteins of SARS-CoV-2, Host Immune Responses, and Herd Immunity. Cells 2021; 10:cells10112949. [PMID: 34831172 PMCID: PMC8616290 DOI: 10.3390/cells10112949] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 10/05/2021] [Accepted: 10/18/2021] [Indexed: 01/18/2023] Open
Abstract
The first quarter of the 21st century has remarkably been characterized by a multitude of challenges confronting human society as a whole in terms of several outbreaks of infectious viral diseases, such as the 2003 severe acute respiratory syndrome (SARS), China; the 2009 influenza H1N1, Mexico; the 2012 Middle East respiratory syndrome (MERS), Saudi Arabia; and the ongoing coronavirus disease 19 (COVID-19), China. COVID-19, caused by SARS-CoV-2, reportedly broke out in December 2019, Wuhan, the capital of China’s Hubei province, and continues unabated, leading to considerable devastation and death worldwide. The most common target organ of SARS-CoV-2 is the lungs, especially the bronchial and alveolar epithelial cells, culminating in acute respiratory distress syndrome (ARDS) in severe patients. Nevertheless, other tissues and organs are also known to be critically affected following infection, thereby complicating the overall aetiology and prognosis. Excluding H1N1, the SARS-CoV (also referred as SARS-CoV-1), MERS, and SARS-CoV-2 are collectively referred to as coronaviruses, and taxonomically placed under the realm Riboviria, order Nidovirales, suborder Cornidovirineae, family Coronaviridae, subfamily Orthocoronavirinae, genus Betacoronavirus, and subgenus Sarbecovirus. As of 23 September 2021, the ongoing SARS-CoV-2 pandemic has globally resulted in around 229 million and 4.7 million reported infections and deaths, respectively, apart from causing huge psychosomatic debilitation, academic loss, and deep economic recession. Such an unprecedented pandemic has compelled researchers, especially epidemiologists and immunologists, to search for SARS-CoV-2-associated potential immunogenic molecules to develop a vaccine as an immediate prophylactic measure. Amongst multiple structural and non-structural proteins, the homotrimeric spike (S) glycoprotein has been empirically found as the most suitable candidate for vaccine development owing to its immense immunogenic potential, which makes it capable of eliciting both humoral and cell-mediated immune responses. As a consequence, it has become possible to design appropriate, safe, and effective vaccines, apart from related therapeutic agents, to reduce both morbidity and mortality. As of 23 September 2021, four vaccines, namely, Comirnaty, COVID-19 vaccine Janssen, Spikevax, and Vaxzevria, have received the European Medicines Agency’s (EMA) approval, and around thirty are under the phase three clinical trial with emergency authorization by the vaccine-developing country-specific National Regulatory Authority (NRA). In addition, 100–150 vaccines are under various phases of pre-clinical and clinical trials. The mainstay of global vaccination is to introduce herd immunity, which would protect the majority of the population, including immunocompromised individuals, from infection and disease. Here, we primarily discuss category-wise vaccine development, their respective advantages and disadvantages, associated efficiency and potential safety aspects, antigenicity of SARS-CoV-2 structural proteins and immune responses to them along with the emergence of SARS-CoV-2 VOC, and the urgent need of achieving herd immunity to contain the pandemic.
Collapse
|