1351
|
A novel STING agonist-adjuvanted pan-sarbecovirus vaccine elicits potent and durable neutralizing antibody and T cell responses in mice, rabbits and NHPs. Cell Res 2022; 32:269-287. [PMID: 35046518 PMCID: PMC8767042 DOI: 10.1038/s41422-022-00612-2] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/03/2022] [Indexed: 12/23/2022] Open
Abstract
The emergence of SARS-CoV-2 variants and potentially other highly pathogenic sarbecoviruses in the future highlights the need for pan-sarbecovirus vaccines. Here, we discovered a new STING agonist, CF501, and found that CF501-adjuvanted RBD-Fc vaccine (CF501/RBD-Fc) elicited significantly stronger neutralizing antibody (nAb) and T cell responses than Alum- and cGAMP-adjuvanted RBD-Fc in mice. Vaccination of rabbits and rhesus macaques (nonhuman primates, NHPs) with CF501/RBD-Fc elicited exceptionally potent nAb responses against SARS-CoV-2 and its nine variants and 41 S-mutants, SARS-CoV and bat SARSr-CoVs. CF501/RBD-Fc-immunized hACE2-transgenic mice were almost completely protected against SARS-CoV-2 challenge, even 6 months after the initial immunization. NHPs immunized with a single dose of CF501/RBD-Fc produced high titers of nAbs. The immunized macaques also exhibited durable humoral and cellular immune responses and showed remarkably reduced viral load in the upper and lower airways upon SARS-CoV-2 challenge even at 108 days post the final immunization. Thus, CF501/RBD-Fc can be further developed as a novel pan-sarbecovirus vaccine to combat current and future outbreaks of sarbecovirus diseases.
Collapse
|
1352
|
Jiang Y, Wu Q, Song P, You C. The Variation of SARS-CoV-2 and Advanced Research on Current Vaccines. Front Med (Lausanne) 2022; 8:806641. [PMID: 35118097 PMCID: PMC8804231 DOI: 10.3389/fmed.2021.806641] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 12/16/2021] [Indexed: 12/15/2022] Open
Abstract
Over the past 2 years, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) caused the coronavirus disease 2019 (COVID-19) and rapidly spread worldwide. In the process of evolution, new mutations of SARS-CoV-2 began to appear to be more adaptable to the diverse changes of various cellular environments and hosts. Generally, the emerging SARS-CoV-2 variants are characterized by high infectivity, augmented virulence, and fast transmissibility, posing a serious threat to the prevention and control of the global epidemic. At present, there is a paucity of effective measurements to cure COVID-19. It is extremely crucial to develop vaccines against SARS-CoV-2 and emerging variants to enhance individual immunity, but it is not yet known whether they are approved by the authority. Therefore, we systematically reviewed the main characteristics of the emerging various variants of SARS-CoV-2, including their distribution, mutations, transmissibility, severity, and susceptibility to immune responses, especially the Delta variant and the new emerging Omicron variant. Furthermore, we overviewed the suitable crowd, the efficacy, and adverse events (AEs) of current vaccines.
Collapse
Affiliation(s)
| | | | | | - Chongge You
- Laboratory Medicine Center, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
1353
|
The Population-Wide Risk-Benefit Profile of Extending the Primary COVID-19 Vaccine Course Compared with an mRNA Booster Dose Program. Vaccines (Basel) 2022; 10:vaccines10020140. [PMID: 35214599 PMCID: PMC8880242 DOI: 10.3390/vaccines10020140] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/05/2022] [Accepted: 01/14/2022] [Indexed: 12/10/2022] Open
Abstract
The vaccination program is reducing the burden of COVID-19. However, recently, COVID-19 infections have been increasing across Europe, providing evidence that vaccine efficacy is waning. Consequently, booster doses are required to restore immunity levels. However, the relative risk–benefit ratio of boosters, compared to pursuing a primary course in the unvaccinated population, remains uncertain. In this study, a susceptible-exposed-infectious-recovered (SEIR) transmission model of SARS-CoV-2 was used to investigate the impact of COVID-19 vaccine waning on disease burden, the benefit of a booster vaccine program compared to targeting the unvaccinated population, and the population-wide risk–benefit profile of vaccination. Our data demonstrates that the rate of vaccine efficacy waning has a significant impact on COVID-19 hospitalisations with the greatest effect in populations with lower vaccination coverage. There was greater benefit associated with a booster vaccination strategy compared to targeting the unvaccinated population, once >50% of the population had received their primary vaccination course. The population benefits of vaccination (reduced hospitalisations, long-COVID and deaths) outweighed the risks of myocarditis/pericarditis by an order of magnitude. Vaccination is important in ending the COVID-19 pandemic sooner, and the reduction in hospitalisations, death and long-COVID associated with vaccination significantly outweigh any risks. Despite these obvious benefits some people are vaccine reluctant, and as such remain unvaccinated. However, when most of a population have been vaccinated, a focus on a booster vaccine strategy for this group is likely to offer greater value, than targeting the proportion of the population who choose to remain unvaccinated.
Collapse
|
1354
|
Venous Thrombosis within 30 Days after Vaccination against SARS-CoV-2 in a Multinational Venous Thromboembolism Registry. Viruses 2022; 14:v14020178. [PMID: 35215771 PMCID: PMC8878689 DOI: 10.3390/v14020178] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/11/2022] [Accepted: 01/11/2022] [Indexed: 12/13/2022] Open
Abstract
Background: Venous thromboembolism (VTE)—including deep vein thrombosis, pulmonary embolism, and cerebral venous sinus thrombosis (CVST)—may occur early after vaccination against the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We sought to describe the site, clinical characteristics, and outcomes of VTE after vaccination against SARS-CoV-2. Methods: In a prospective study using the Registro Informatizado de Enfermedad TromboEmbólica (RIETE) platform, patients with VTE 4–30 days after vaccination against SARS-CoV-2 (1 February 2021 through 30 April 2021) were included. VTE patients recruited from the same centers into RIETE in the same months in 2018–2019 were selected as the reference group. All-cause mortality and major bleeding were the main study outcomes. Results: As of 30 April 2020, 102 patients with post-vaccination VTEs had been identified (28 after adenovirus-based vaccination [ChAdOx1 nCov-19; AstraZeneca] and 74 after mRNA-based vaccination [mRNA-1273; Moderna, and BNT162b2; Pfizer]). Compared with 911 historical controls, patients with VTE after adenovirus-based vaccination more frequently had CVST (10.7% vs. 0.4%, p < 0.001) or thrombosis at multiple sites (17.9% vs. 1.3%, p < 0.001), more frequently had thrombocytopenia (40.7% vs. 14.7%, p < 0.001), and had higher 14-day mortality (14.3% vs. 0.7%; odds ratio [OR]: 25.1; 95% confidence interval [CI]: 6.7–94.9) and major bleeding rates (10.3% vs. 1.0%, OR: 12.03, 95% CI: 3.07–47.13). The site of thrombosis, accompanying thrombocytopenia, and 14-day mortality rates were not significantly different for patients with VTE after mRNA-based vaccination, compared with historical controls. Conclusions: Compared with historical controls, VTE after adenovirus-based vaccination against SARS-CoV-2 is accompanied by thrombocytopenia, occurs in unusual sites, and is associated with worse clinical outcomes.
Collapse
|
1355
|
Taborda A, Murillo DA, Moreno C, Taborda PA, Fuquen M, Díaz PA, Londoño D. Análisis de impacto presupuestal de la vacunación contra COVID-19 en América Latina. Rev Panam Salud Publica 2022; 46:e5. [PMID: 35350457 PMCID: PMC8956969 DOI: 10.26633/rpsp.2022.5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 10/13/2021] [Indexed: 11/24/2022] Open
Abstract
Objetivo.
Estimar el impacto presupuestal de la vacunación contra COVID-19 en seis países de América Latina: Argentina, Brasil, Chile, Colombia, México y Perú, durante el periodo 2021-2022.
Métodos.
Se evaluaron las vacunas de Sinopharm (BBIBP-CorV), Janssen (JNJ-78436735), Instituto de Gamaleya (Gam-COVID-Vac), Sinovac (CoronaVac), CanSino (Convidecia), AstraZeneca (Vaxzevria), Moderna (mRNA-1273) y Pfizer (BNT162b2), según disponibilidad para cada país. Se adoptó la perspectiva del sistema de salud, de manera que solo se incluyeron costos médicos directos. El horizonte temporal se adoptó teniendo en cuenta los tiempos de implementación de cada plan de vacunación, excluyendo menores de 16 años y gestantes. Se incluyeron los siguientes costos: costo de la vacunación y aplicación, costos de la hospitalización general aislamiento, cuidado intermedio e intensivo. Se compararon dos escenarios de vacunación: 1) Población que desea vacunarse (según las encuestas nacionales) y 2) Población que debería vacunarse (total susceptible de vacunación). Los costos agregados para cada escenario de vacunación se compararon con el escenario de no vacunación. Adicionalmente, se realizaron análisis de sensibilidad determinísticos y probabilísticos.
Resultados.
Los diferentes esquemas de vacunación contra COVID-19 disponibles en América Latina genera ahorros potenciales que oscilan entre USD 100 y USD 1 500 millones de dólares por país para el período 2021-2022, asumiendo que se logra implementar en su totalidad el plan de vacunación previsto en cada país.
Conclusiones.
La vacunación contra COVID-19 es una estrategia que además de reducir la morbilidad y mortalidad para Latinoamérica, genera ahorros potenciales para los sistemas de salud en la región.
Collapse
Affiliation(s)
| | | | | | | | | | - Paula Andrea Díaz
- Facultad Nacional de Salud Pública. Universidad de Antioquia, Medellín, Colombia
| | - Darío Londoño
- Salud Poblacional. Fundación Santa Fe de Bogotá, Bogotá, Colombia
| |
Collapse
|
1356
|
Sobczak M, Pawliczak R. The risk of anaphylaxis behind authorized COVID-19 vaccines: a meta-analysis. Clin Mol Allergy 2022; 20:1. [PMID: 35039051 PMCID: PMC8762434 DOI: 10.1186/s12948-022-00167-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/06/2022] [Indexed: 12/31/2022] Open
Abstract
Background A serious allergic reaction that may occur in response to medical products is anaphylaxis, which potentially can lead to anaphylactic shock. In the light of recent COVID-19 pandemic, much public attention had been paid to the severe allergic reactions occurring after COVID-19 vaccination. Therefore, in our study we would like to investigate the risk of authorized COVID-19 vaccines to induce anaphylactic reaction, anaphylactoid reaction, anaphylactic shock and anaphylactoid shock. Methods We searched databases, such as PubMed, Web of Science and Embase and found eight articles about the incidence of anaphylactic and anaphylactoid reactions. Also, we used data from four databases from Canada, the U.S., the European Union and the United Kingdom. To calculate effect sizes, we used random effects model with inverse variance method. The risk ratio with 95% confidence interval were used for dichotomous outcomes. Statistical analysis was prepared in R. Results were considered statistically significant at p < 0.05. Results The most cases of anaphylactic reaction, anaphylactoid reaction, anaphylactic shock and anaphylactoid shock were reported in female aged 18–85 years after BNT162b2 vaccine according to data from the EU. Analyzed COVID-19 vaccines can cause the anaphylaxis/anaphylactic reaction with risk of 106.99 (95% CI [39.95; 286.57], p < 0.0001, I2 = 59%), whereas the anaphylactoid reaction, anaphylactic and anaphylactoid shocks with risk of 113.3 (95% CI [28.11; 456.53], p < 0.0001), 344.2 (95% CI [85.77; 1381.39], p < 0.0001), 14.9, 95% CI [1.96; 112.79], p = 0.009), respectively. Conclusions Our meta-analysis shows that the risk of anaphylactic reaction, anaphylactoid reaction, anaphylactic shock and anaphylactoid shock do not occur only after mRNA COVID-19 vaccines. Therefore, vaccination centers should be prepared to render assistance in the event of a reaction in all cases.
Collapse
Affiliation(s)
- Marharyta Sobczak
- Department of Immunopathology, Faculty of Medicine, Division of Biomedical Science, Medical University of Lodz, St. Zeligowskiego 7/9, 90-752, Lodz, Poland
| | - Rafał Pawliczak
- Department of Immunopathology, Faculty of Medicine, Division of Biomedical Science, Medical University of Lodz, St. Zeligowskiego 7/9, 90-752, Lodz, Poland.
| |
Collapse
|
1357
|
Halperin SA, Ye L, MacKinnon-Cameron D, Smith B, Cahn PE, Ruiz-Palacios GM, Ikram A, Lanas F, Lourdes Guerrero M, Muñoz Navarro SR, Sued O, Lioznov DA, Dzutseva V, Parveen G, Zhu F, Leppan L, Langley JM, Barreto L, Gou J, Zhu T. Final efficacy analysis, interim safety analysis, and immunogenicity of a single dose of recombinant novel coronavirus vaccine (adenovirus type 5 vector) in adults 18 years and older: an international, multicentre, randomised, double-blinded, placebo-controlled phase 3 trial. Lancet 2022; 399:237-248. [PMID: 34953526 PMCID: PMC8700283 DOI: 10.1016/s0140-6736(21)02753-7] [Citation(s) in RCA: 145] [Impact Index Per Article: 48.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/29/2021] [Accepted: 11/29/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND The Ad5-nCoV vaccine is a single-dose adenovirus type 5 (Ad5) vectored vaccine expressing the SARS-CoV-2 spike protein that was well-tolerated and immunogenic in phase 1 and 2 studies. In this study, we report results on the final efficacy and interim safety analyses of the phase 3 trial. METHODS This double-blind, randomised, international, placebo-controlled, endpoint-case driven, phase 3, clinical trial enrolled adults aged 18 years older at study centres in Argentina, Chile, Mexico, Pakistan, and Russia. Participants were eligible for the study if they had no unstable or severe underlying medical or psychiatric conditions; had no history of a laboratory-confirmed SARS-CoV-2 infection; were not pregnant or breastfeeding; and had no previous receipt of an adenovirus-vectored, coronavirus, or SARS-CoV-2 vaccine. After informed consent was obtained, 25 mL of whole blood was withdrawn from all eligible participants who were randomised in a 1:1 ratio to receive a single intramuscular dose of 0·5 mL placebo or a 0·5 mL dose of 5 × 1010 viral particle (vp)/mL Ad5-nCoV vaccine; study staff and participants were blinded to treatment allocation. All participants were contacted weekly by email, telephone, or text message to self-report any symptoms of COVID-19 illness, and laboratory testing for SARS-CoV-2 was done for all participants with any symptoms. The primary efficacy objective evaluated Ad5-nCoV in preventing symptomatic, PCR-confirmed COVID-19 infection occurring at least 28 days after vaccination in all participants who were at least 28 days postvaccination on Jan 15, 2021. The primary safety objective evaluated the incidence of any serious adverse events or medically attended adverse events postvaccination in all participants who received a study injection. This trial is closed for enrolment and is registered with ClinicalTrials.gov (NCT04526990). FINDINGS Study enrolment began on Sept 22, 2020, in Pakistan, Nov 6, 2020, in Mexico, Dec 2, 2020, in Russia and Chile, and Dec 17, 2020, in Argentina; 150 endpoint cases were reached on Jan 15, 2021, triggering the final primary efficacy analysis. One dose of Ad5-nCoV showed a 57·5% (95% CI 39·7-70·0, p=0·0026) efficacy against symptomatic, PCR-confirmed, COVID-19 infection at 28 days or more postvaccination (21 250 participants; 45 days median duration of follow-up [IQR 36-58]). In the primary safety analysis undertaken at the time of the efficacy analysis (36 717 participants), there was no significant difference in the incidence of serious adverse events (14 [0·1%] of 18 363 Ad5-nCoV recipients and 10 [0·1%] of 18 354 placebo recipients, p=0·54) or medically attended adverse events (442 [2·4%] of 18 363 Ad5-nCoV recipients and 411 [2·2%] of 18 354 placebo recipients, p=0·30) between the Ad5-nCoV or placebo groups, or any serious adverse events considered related to the study product (none in both Ad5-nCoV and placebo recipients). In the extended safety cohort, 1004 (63·5%) of 1582 of Ad5-nCoV recipients and 729 (46·4%) of 1572 placebo recipients reported a solicited systemic adverse event (p<0·0001), of which headache was the most common (699 [44%] of Ad5-nCoV recipients and 481 [30·6%] of placebo recipients; p<0·0001). 971 (61·3%) of 1584 Ad5-nCoV recipients and 314 (20·0%) of 1573 placebo recipients reported an injection-site adverse event (p<0·0001), of which pain at the injection site was the most frequent; reported by 939 (59%) Ad5-nCoV recipients and 303 (19%) placebo recipients. INTERPRETATION One dose of Ad5-nCoV is efficacious and safe in healthy adults aged 18 years and older. FUNDING CanSino Biologics and the Beijing Institute of Biotechnology.
Collapse
Affiliation(s)
- Scott A Halperin
- Canadian Center for Vaccinology, Dalhousie University, IWK Health, Nova Scotia Health, Halifax, Canada.
| | - Lingyun Ye
- Canadian Center for Vaccinology, Dalhousie University, IWK Health, Nova Scotia Health, Halifax, Canada
| | - Donna MacKinnon-Cameron
- Canadian Center for Vaccinology, Dalhousie University, IWK Health, Nova Scotia Health, Halifax, Canada
| | - Bruce Smith
- Canadian Center for Vaccinology, Dalhousie University, IWK Health, Nova Scotia Health, Halifax, Canada
| | | | | | - Aamer Ikram
- National Institute of Health, Islamabad, Pakistan
| | | | - M Lourdes Guerrero
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | | | - Omar Sued
- Fundación Huésped, Buenos Aires, Argentina
| | - Dmitry A Lioznov
- Federal State Budgetary Institution, Smorodintsev Research Institute of Influenza, St Petersburg, Russia
| | | | | | - Fengcai Zhu
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Laura Leppan
- Canadian Center for Vaccinology, Dalhousie University, IWK Health, Nova Scotia Health, Halifax, Canada
| | - Joanne M Langley
- Canadian Center for Vaccinology, Dalhousie University, IWK Health, Nova Scotia Health, Halifax, Canada
| | | | | | - Tao Zhu
- CanSino Biologics, Tianjin, China
| |
Collapse
|
1358
|
Garrett N, Tapley A, Andriesen J, Seocharan I, Fisher LH, Bunts L, Espy N, Wallis CL, Randhawa AK, Ketter N, Yacovone M, Goga A, Bekker LG, Gray GE, Corey L. High Rate of Asymptomatic Carriage Associated with Variant Strain Omicron. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2021.12.20.21268130. [PMID: 35043118 PMCID: PMC8764727 DOI: 10.1101/2021.12.20.21268130] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The early widespread dissemination of Omicron indicates the urgent need to better understand the transmission dynamics of this variant, including asymptomatic spread among immunocompetent and immunosuppressed populations. In early December 2021, the Ubuntu clinical trial, designed to evaluate efficacy of the mRNA-1273 vaccine (Moderna) among persons living with HIV (PLWH), began enrolling participants. Nasal swabs are routinely obtained at the initial vaccination visit, which requires participants to be clinically well to receive their initial jab. Of the initial 230 participants enrolled between December 2 and December 17, 2021, 71 (31%) were PCR positive for SARS-CoV-2: all of whom were subsequently confirmed by S gene dropout to be Omicron; 48% of the tested samples had cycle threshold (CT) values <25 and 18% less than 20, indicative of high titers of asymptomatic shedding. Asymptomatic carriage rates were similar in SARS-CoV-2 seropositive and seronegative persons (27% respectively). These data are in stark contrast to COVID-19 vaccine studies conducted pre-Omicron, where the SARS-CoV-2 PCR positivity rate at the first vaccination visit ranged from <1%-2.4%, including a cohort of over 1,200 PLWH largely enrolled in South Africa during the Beta outbreak. We also evaluated asymptomatic carriage in a sub study of the Sisonke vaccine trial conducted in South African health care workers, which indicated 2.6% asymptomatic carriage during the Beta and Delta outbreaks and subsequently rose to 16% in both PLWH and PHLWH during the Omicron period.
Collapse
Affiliation(s)
- Nigel Garrett
- Centre for the AIDS Programme of Research in South Africa, University of KwaZulu–Natal, Durban, South Africa
| | - Asa Tapley
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- University of Washington School of Medicine, Seattle, WA, USA
| | - Jessica Andriesen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Ishen Seocharan
- South African Medical Research Council, Pretoria, South Africa
| | - Leigh H. Fisher
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Lisa Bunts
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Nicole Espy
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Carole L. Wallis
- Bio Analytical Research Corporation South Africa and Lancet Laboratories, Johannesburg, South Africa
| | - April Kaur Randhawa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Nzeera Ketter
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | - Ameena Goga
- South African Medical Research Council, Pretoria, South Africa
| | - Linda-Gail Bekker
- Desmond Tutu HIV Centre, University of Cape Town, Cape Town, South Africa
| | - Glenda E. Gray
- South African Medical Research Council, Pretoria, South Africa
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
1359
|
Tomowiak C, Leblond V, Laribi K, Baron M, Puppinck C, Gérard P, Courret E, Gorochov G, Sterlin D, Tournilhac O, Morel P, Cymbalista F, Roos-Weil D. Response to vaccination against SARS-CoV-2 in 168 patients with Waldenström macroglobulinemia: a French Innovative Leukemia Organization (FILO) study. Br J Haematol 2022; 197:424-427. [PMID: 35029297 DOI: 10.1111/bjh.18055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/07/2022] [Accepted: 01/10/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Cécile Tomowiak
- Service d'Hématologie et thérapie cellulaire, CHU de Poitiers et CIC 1402, France
| | - Véronique Leblond
- Sorbonne Université, Service d'Hématologie Clinique, Hôpital Pitié-Salpêtrière, APHP, Paris, France
| | - Kamel Laribi
- Department of Hematology, Centre Hospitalier Le Mans, 194 Avenue Rubillard, 72000, Le Mans, France
| | - Marine Baron
- Sorbonne Université, Service d'Hématologie Clinique, Hôpital Pitié-Salpêtrière, APHP, Paris, France
| | - Christian Puppinck
- Association de Soutien et d'Information à la Leucémie Lymphoïde Chronique et la maladie de Waldenström (SILLC)
| | | | - Elodie Courret
- Sorbonne Université, Service d'Hématologie Clinique, Hôpital Pitié-Salpêtrière, APHP, Paris, France
| | - Guy Gorochov
- Sorbonne Université, Département d'Immunologie, Hôpital Pitié-Salpêtrière, APHP, Paris, France
| | - Delphine Sterlin
- Sorbonne Université, Département d'Immunologie, Hôpital Pitié-Salpêtrière, APHP, Paris, France
| | - Olivier Tournilhac
- Hematology Department, University Hospital Estaing, University Clermont Auvergne EA 7453, CIC1405, Clermont-Ferrand, France
| | - Pierre Morel
- Hematology Department, University Hospital Amiens-Picardie, Amiens, France
| | | | - Damien Roos-Weil
- Sorbonne Université, Service d'Hématologie Clinique, Hôpital Pitié-Salpêtrière, APHP, Paris, France
| | | |
Collapse
|
1360
|
Meng H, Mao J, Ye Q. Booster vaccination strategy: Necessity, Immunization Objectives, Immunization Strategy and Safety. J Med Virol 2022; 94:2369-2375. [PMID: 35028946 DOI: 10.1002/jmv.27590] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/09/2022] [Accepted: 01/10/2022] [Indexed: 12/14/2022]
Abstract
At present, the global COVID-19 epidemic has not been completely controlled, and epidemic prevention and control still face severe challenges. As there is no specific treatment for COVID-19, promoting roll-out vaccinations and building herd immunity are still the most effective and economic measures to control the COVID-19 pandemic. However, the neutralizing antibody level in the recipients decreases with time, and the vaccine's protective efficacy gradually weakens. It is still inconclusive whether it is necessary to carry out booster vaccination to strengthen the immune barrier to infection. In this paper, we combined the existing data on the effectiveness and persistence of COVID-19 vaccines. We found that it is necessary to carry out a booster vaccination strategy. However, not all subjects need to receive one more dose of vaccine six months after the initial immunization. Priority should be given to the high-risk groups, such as the elderly and people with immunodeficiency. A heterologous booster can induce higher immune responses and enhance immune protection than homologous vaccinations. However, more scientific data and clinical studies are needed to verify the safety of heterologous vaccination strategies. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Hanyan Meng
- The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, 310052, China
| | - Jianhua Mao
- The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, 310052, China
| | - Qing Ye
- The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, 310052, China
| |
Collapse
|
1361
|
Zaki HA, Zahran A, Abdelrahim M, Elnabawy WA, Kaber Y. A Case of Acute Viral Pericarditis Complicated With Pericardial Effusion Induced by Third Dose of COVID Vaccination. Cureus 2022; 14:e21207. [PMID: 35165640 PMCID: PMC8840804 DOI: 10.7759/cureus.21207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2022] [Indexed: 11/18/2022] Open
Abstract
COVID-19 vaccines were safe and efficacious in clinical trials. A two-dose regimen of the Pfizer-BioNTech COVID-19 vaccine confers no less than 95% protection against COVID-19 with an adequate safety profile. To date, no reports have been made in the literature regarding the onset of acute viral pericarditis after vaccination with the Pfizer BNT162b2 vaccine. But on the other hand, pericarditis is reported to occur in rare instances of COVID-19 infection, and this may be attributed to the pro-inflammatory effects of the spike protein. In this article, we describe the case of an elderly male patient with a known case of hypothyroidism who presented to our emergency department with fever, chills, and dry cough for ten days after the third dose of the Pfizer-BioNTech COVID-19 vaccine. Although we cannot mention a direct effect, it is essential to note a potential adverse reaction to vaccine administration following the expression of SARS-CoV-2 spike protein-induced from the vaccine’s mRNA.
Collapse
|
1362
|
Vollenberg R, Tepasse PR, Kühn JE, Hennies M, Strauss M, Rennebaum F, Schomacher T, Boeckel G, Lorentzen E, Bokemeyer A, Nowacki TM. Humoral Immune Response in IBD Patients Three and Six Months after Vaccination with the SARS-CoV-2 mRNA Vaccines mRNA-1273 and BNT162b2. Biomedicines 2022; 10:171. [PMID: 35052849 PMCID: PMC8774019 DOI: 10.3390/biomedicines10010171] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/09/2022] [Accepted: 01/11/2022] [Indexed: 12/13/2022] Open
Abstract
Severe acute respiratory syndrome coronovirus-2 (SARS-CoV-2) is the cause of the coronavirus disease 2019 (COVID-19) pandemic. Vaccination is considered the core approach to containing the pandemic. There is currently insufficient evidence on the efficacy of these vaccines in immunosuppressed inflammatory bowel disease (IBD) patients. The aim of this study was to investigate the humoral response in immunosuppressed IBD patients after COVID-19 mRNA vaccination. In this prospective study, IgG antibody levels (AB) against the SARS-CoV-2 receptor-binding domain (spike-protein) were quantitatively determined. For assessing the potential neutralizing capacity, a SARS-CoV-2 surrogate neutralization test (sVNT) was employed in IBD patients (n = 95) and healthy controls (n = 38). Sera were examined prior to the first/second vaccination and 3/6 months after second vaccination. Patients showed lower sVNT (%) and IgG-S (AU/mL) AB both before the second vaccination (sVNT p < 0.001; AB p < 0.001) and 3 (sVNT p = 0.002; AB p = 0.001) and 6 months (sVNT p = 0.062; AB p = 0.061) after the second vaccination. Although seroconversion rates (sVNT, IgG-S) did not differ between the two groups 3 months after second vaccination, a significant difference was seen 6 months after second vaccination (sVNT p = 0.045). Before and three months after the second vaccination, patients treated with anti-tumor necrosis factor (TNF) agents showed significantly lower AB than healthy subjects. In conclusion, an early booster shot vaccination should be discussed for IBD patients on anti-TNF therapy.
Collapse
Affiliation(s)
- Richard Vollenberg
- Department of Medicine B for Gastroenterology, Hepatology, Endocrinology and Clincial Infectiology University Hospital Muenster, 48149 Muenster, Germany; (F.R.); (T.S.); (T.M.N.)
| | - Phil-Robin Tepasse
- Department of Medicine B for Gastroenterology, Hepatology, Endocrinology and Clincial Infectiology University Hospital Muenster, 48149 Muenster, Germany; (F.R.); (T.S.); (T.M.N.)
| | - Joachim Ewald Kühn
- Institute of Virology, University Hospital Muenster, 48149 Muenster, Germany; (J.E.K.); (M.H.); (E.L.)
| | - Marc Hennies
- Institute of Virology, University Hospital Muenster, 48149 Muenster, Germany; (J.E.K.); (M.H.); (E.L.)
| | - Markus Strauss
- Department of Medicine C, Cardiology, University Hospital Muenster, 48149 Muenster, Germany;
| | - Florian Rennebaum
- Department of Medicine B for Gastroenterology, Hepatology, Endocrinology and Clincial Infectiology University Hospital Muenster, 48149 Muenster, Germany; (F.R.); (T.S.); (T.M.N.)
| | - Tina Schomacher
- Department of Medicine B for Gastroenterology, Hepatology, Endocrinology and Clincial Infectiology University Hospital Muenster, 48149 Muenster, Germany; (F.R.); (T.S.); (T.M.N.)
| | - Göran Boeckel
- Department of Medicine D, Division of General Internal and Emergency Medicine, Nephrology and Rheumatology, University Hospital Muenster, 48149 Muenster, Germany;
| | - Eva Lorentzen
- Institute of Virology, University Hospital Muenster, 48149 Muenster, Germany; (J.E.K.); (M.H.); (E.L.)
| | - Arne Bokemeyer
- Department of Gastroenterology, Hepatology and Transplant Medicine, University Hospital Essen, 45147 Essen, Germany;
| | - Tobias Max Nowacki
- Department of Medicine B for Gastroenterology, Hepatology, Endocrinology and Clincial Infectiology University Hospital Muenster, 48149 Muenster, Germany; (F.R.); (T.S.); (T.M.N.)
- Department of Medicine, Gastroenterology, Marienhospital Steinfurt, 48565 Steinfurt, Germany
| |
Collapse
|
1363
|
Presby DM, Capodilupo ER. Biometrics from a Wearable Device Reveals Temporary Effects of COVID-19 Vaccines on Cardiovascular, Respiratory, and Sleep Physiology. J Appl Physiol (1985) 2022; 132:448-458. [PMID: 35019761 PMCID: PMC8816631 DOI: 10.1152/japplphysiol.00420.2021] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Although vaccines against SARS-CoV-2 have been proven safe and effective, transient side-effects lasting 24-48 h postvaccination have been reported. To better understand the subjective and objective response to COVID-19 vaccination, we conducted a retrospective analysis on 69,619 subscribers to a wrist-worn biometric device (WHOOP Inc., Boston, MA) who received either the AstraZeneca, Janssen/Johnson & Johnson, Moderna, or Pfizer/BioNTech vaccine. The WHOOP device measures resting heart rate (RHR), heart rate variability (HRV), respiratory rate (RR), and sleep architecture, and these physiological measures were normalized to the same day of the week, 1 wk before vaccination. Averaging across vaccines, RHR, RR, and percent sleep derived from light sleep were elevated on the first night following vaccination and returned to baseline within 4 nights postvaccination. When statistical differences were observed between doses on the first night postvaccination, larger deviations in physiological measures were observed following the first dose of AstraZeneca and the second dose of Moderna and Pfizer/BioNTech. When statistical differences were observed between age groups or gender on the first night postvaccination, larger deviations in physiological measures were observed in younger populations and in females (compared with males). When combining self-reported symptoms (fatigue, muscle aches, headache, chills, or fever) with the objectively measured physiological parameters, we found that self-reporting fever or chills had the strongest association with deviations in physiological measures following vaccination. In summary, these results suggest that COVID-19 vaccines temporarily affect cardiovascular, respiratory, and sleep physiology and that dose, gender, and age affect the physiological response to vaccination. NEW & NOTEWORTHY Here we report the first large-scale study investigating the effect of COVID-19 vaccines on cardiovascular, respiratory, and sleep physiology. We find that vaccines temporarily impact measures of cardiovascular, respiratory, and sleep physiology and that the degree of change in physiology is influenced by the manufacturer and dose of the vaccine and the gender and age of the vaccine recipient. These results provide insights into physiological changes that occur with COVID-19 vaccination and indicate that the unique responses that occur postvaccination may depend on manufacturer, dose, gender, and age.
Collapse
Affiliation(s)
- David M Presby
- Department of Data Science and Research, Whoop, Inc., Boston, Boston, MA, United States
| | - Emily R Capodilupo
- Department of Data Science and Research, Whoop, Inc., Boston, Boston, MA, United States
| |
Collapse
|
1364
|
Berlansky S, Sallinger M, Grabmayr H, Humer C, Bernhard A, Fahrner M, Frischauf I. Calcium Signals during SARS-CoV-2 Infection: Assessing the Potential of Emerging Therapies. Cells 2022; 11:253. [PMID: 35053369 PMCID: PMC8773957 DOI: 10.3390/cells11020253] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/05/2022] [Accepted: 01/11/2022] [Indexed: 01/09/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a positive-sense single-stranded RNA virus that causes coronavirus disease 2019 (COVID-19). This respiratory illness was declared a pandemic by the world health organization (WHO) in March 2020, just a few weeks after being described for the first time. Since then, global research effort has considerably increased humanity's knowledge about both viruses and disease. It has also spawned several vaccines that have proven to be key tools in attenuating the spread of the pandemic and severity of COVID-19. However, with vaccine-related skepticism being on the rise, as well as breakthrough infections in the vaccinated population and the threat of a complete immune escape variant, alternative strategies in the fight against SARS-CoV-2 are urgently required. Calcium signals have long been known to play an essential role in infection with diverse viruses and thus constitute a promising avenue for further research on therapeutic strategies. In this review, we introduce the pivotal role of calcium signaling in viral infection cascades. Based on this, we discuss prospective calcium-related treatment targets and strategies for the cure of COVID-19 that exploit viral dependence on calcium signals.
Collapse
Affiliation(s)
| | | | | | | | | | - Marc Fahrner
- Institute of Biophysics, Johannes Kepler University Linz, Gruberstrasse 40, 4020 Linz, Austria; (S.B.); (M.S.); (H.G.); (C.H.); (A.B.)
| | - Irene Frischauf
- Institute of Biophysics, Johannes Kepler University Linz, Gruberstrasse 40, 4020 Linz, Austria; (S.B.); (M.S.); (H.G.); (C.H.); (A.B.)
| |
Collapse
|
1365
|
Clinical validation of engineered CRISPR/Cas12a for rapid SARS-CoV-2 detection. COMMUNICATIONS MEDICINE 2022; 2:7. [PMID: 35603267 PMCID: PMC9053293 DOI: 10.1038/s43856-021-00066-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 11/30/2021] [Indexed: 12/26/2022] Open
Abstract
Background The coronavirus disease (COVID-19) caused by SARS-CoV-2 has swept through the globe at an unprecedented rate. CRISPR-based detection technologies have emerged as a rapid and affordable platform that can shape the future of diagnostics. Methods We developed ENHANCEv2 that is composed of a chimeric guide RNA, a modified LbCas12a enzyme, and a dual reporter construct to improve the previously reported ENHANCE system. We validated both ENHANCE and ENHANCEv2 using 62 nasopharyngeal swabs and compared the results to RT-qPCR. We created a lyophilized version of ENHANCEv2 and characterized its detection capability and stability. Results Here we demonstrate that when coupled with an RT-LAMP step, ENHANCE detects COVID-19 samples down to a few copies with 95% accuracy while maintaining a high specificity towards various isolates of SARS-CoV-2 against 31 highly similar and common respiratory pathogens. ENHANCE works robustly in a wide range of magnesium concentrations (3 mM-13 mM), allowing for further assay optimization. Our clinical validation results for both ENHANCE and ENHANCEv2 show 60/62 (96.7%) sample agreement with RT-qPCR results while only using 5 µL of sample and 20 minutes of CRISPR reaction. We show that the lateral flow assay using paper-based strips displays 100% agreement with the fluorescence-based reporter assay during clinical validation. Finally, we demonstrate that a lyophilized version of ENHANCEv2 shows high sensitivity and specificity for SARS-CoV-2 detection while reducing the CRISPR reaction time to as low as 3 minutes while maintaining its detection capability for several weeks upon storage at room temperature. Conclusions CRISPR-based diagnostic platforms offer many advantages as compared to conventional qPCR-based detection methods. Our work here provides clinical validation of ENHANCE and its improved form ENHANCEv2 for the detection of COVID-19. Nguyen et al. describe the clinical validation of the ENHANCE system, a method to detect SARS-CoV-2 based on engineered crRNAs for Cas12a and preceded by an RT-LAMP amplification step. Authors also describe the development and clinical validation of a version of this system, ENHANCEv2, that can be lyophilized and that uses another mutated Cas12a for further signal amplification. The COVID-19 pandemic has underscored the need for rapid and accurate tests to detect SARS-CoV-2 infection. The tests commonly used have limitations, and a detection system based on CRISPR technology could offer a useful alternative. CRISPR is a technology derived from bacteria that can specifically detect pieces of DNA. We have previously developed ENHANCE, a detection system that converts the SARS-CoV-2 genetic material into DNA that is then detected by an engineered CRISPR technology. Here, we develop an improved version of this method, ENHANCEv2, that has an extended shelf life and less need for refrigeration, facilitating transportation of the components required for the test and its use. We show that both ENHANCE and ENHACEv2 can quickly and accurately detect SARS-CoV-2 in swabs from infected people. This is a step towards having more versatile tools to detect SARS-CoV-2 infection quickly and accurately.
Collapse
|
1366
|
Lin N, Fu H, Pu D, Quan Y, Li Y, Yin X, Wei Y, Wang H, Ma X, Wei X. Criteria for judging the immune markers of COVID-19 disease vaccines. MedComm (Beijing) 2022; 3:1-12. [PMID: 35005708 PMCID: PMC8719528 DOI: 10.1002/mco2.109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 11/27/2021] [Accepted: 11/28/2021] [Indexed: 02/05/2023] Open
Abstract
As severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) sweeping the world, effective and affordable vaccines are in urgent need. A reliable system for the assessment of SARS‐CoV‐2 vaccines would boost the development of vaccines and reduce the research cost. We constructed a logistic regression model and analyzed the relationship between antibody (Ab) level and efficacy of different vaccine types. The relationship between assessment dates and Ab levels was depicted by plotting the mean of Ab levels evolved over time and a fitted cubic polynomial model. Anti‐spike immunoglobulin G (IgG) could best estimate the vaccine efficacy (VE) (adjusted R2 = 0.731) and neutralizing Ab to live SARS‐CoV‐2 also explained a fine relationship (adjusted R2 = 0.577). Neutralizing Abs to live SARS‐CoV‐2 in inactivated virus vaccines reached a peak during days 40–60, and their receptor‐binding domain (RBD)‐IgG peaked during days 40–50. For messenger RNA (mRNA) and viral vector vaccines, their neutralizing Ab to live SARS‐CoV‐2 peaked later than day 40, and for RBD‐IgG during days 30–50. For mRNA and viral vector vaccines, their peak time of Abs was later than that in inactivated virus vaccines. RBD‐IgG peaked earlier than Ab to live SARS‐CoV‐2. Anti‐spike IgG and Ab to live SARS‐CoV‐2 may be good immune markers for VE assessment.
Collapse
Affiliation(s)
- Nan Lin
- West China School of Medicine West China Hospital Sichuan University Chengdu China
| | - Haoxuan Fu
- Department of Statistics University of Illinois at Urbana Champaign Urbana Illinois USA
| | - Dan Pu
- Department of Radiation Oncology Cancer Center, West China Hospital, Sichuan University Chengdu China
| | - Yuxin Quan
- West China School of Medicine West China Hospital Sichuan University Chengdu China
| | - Yueyi Li
- Department of Biotherapy Cancer Center, West China Hospital, Sichuan University Chengdu China
| | - Xiaomeng Yin
- Department of Biotherapy Cancer Center, West China Hospital, Sichuan University Chengdu China
| | - Yuhao Wei
- West China School of Medicine West China Hospital Sichuan University Chengdu China
| | - Hang Wang
- West China School of Medicine West China Hospital Sichuan University Chengdu China
| | - Xuelei Ma
- Department of Biotherapy Cancer Center, West China Hospital, Sichuan University Chengdu China
| | - Xiawei Wei
- Laboratory of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University Chengdu China
| |
Collapse
|
1367
|
Cocchio S, Zabeo F, Facchin G, Piva N, Furlan P, Nicoletti M, Saia M, Tonon M, Mongillo M, Russo F, Baldo V. The Effectiveness of a Diverse COVID-19 Vaccine Portfolio and Its Impact on the Persistence of Positivity and Length of Hospital Stays: The Veneto Region's Experience. Vaccines (Basel) 2022; 10:vaccines10010107. [PMID: 35062767 PMCID: PMC8777781 DOI: 10.3390/vaccines10010107] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/09/2022] [Accepted: 01/10/2022] [Indexed: 02/04/2023] Open
Abstract
The vaccination campaign for the Veneto region (northeastern Italy) started on 27 December 2020. As of early December 2021, 75.1% of the whole Veneto population has been fully vaccinated. Vaccine efficacy has been demonstrated in many clinical trials, but reports on real-world contexts are still necessary. We conducted a retrospective cohort study on 2,233,399 residents in the Veneto region to assess the reduction in the COVID-19 burden, taking different outcomes into consideration. First, we adopted a non-brand-specific approach borrowed from survival analysis to estimate the effectiveness of vaccination against SARS-CoV-2 in preventing infections, hospitalizations, and deaths. We used t-tests and multivariate regressions to examine vaccine impact on breakthrough infections, in terms of the persistence of positivity and the length of hospital stays. Evidence emerging from this study suggests that unvaccinated individuals are significantly more likely to become infected, need hospitalization, and are at a higher risk of death from COVID-19 than those given at least one dose of vaccine. Cox models indicate that the effectiveness of full vaccination is 88% against infection, 94% against hospitalization, and 95% against death. Multivariate regressions suggest that vaccination is significantly correlated with a shorter period of positivity and shorter hospital stays, with each step toward completion of the vaccination cycle coinciding with a reduction of 3.3 days in the persistence of positivity and 2.3 days in the length of hospital stay.
Collapse
Affiliation(s)
- Silvia Cocchio
- Department of Cardiac Thoracic and Vascular Sciences and Public Health, University of Padua, 35131 Padua, Italy; (S.C.); (F.Z.); (G.F.); (N.P.); (P.F.); (M.N.)
| | - Federico Zabeo
- Department of Cardiac Thoracic and Vascular Sciences and Public Health, University of Padua, 35131 Padua, Italy; (S.C.); (F.Z.); (G.F.); (N.P.); (P.F.); (M.N.)
| | - Giacomo Facchin
- Department of Cardiac Thoracic and Vascular Sciences and Public Health, University of Padua, 35131 Padua, Italy; (S.C.); (F.Z.); (G.F.); (N.P.); (P.F.); (M.N.)
| | - Nicolò Piva
- Department of Cardiac Thoracic and Vascular Sciences and Public Health, University of Padua, 35131 Padua, Italy; (S.C.); (F.Z.); (G.F.); (N.P.); (P.F.); (M.N.)
| | - Patrizia Furlan
- Department of Cardiac Thoracic and Vascular Sciences and Public Health, University of Padua, 35131 Padua, Italy; (S.C.); (F.Z.); (G.F.); (N.P.); (P.F.); (M.N.)
| | - Michele Nicoletti
- Department of Cardiac Thoracic and Vascular Sciences and Public Health, University of Padua, 35131 Padua, Italy; (S.C.); (F.Z.); (G.F.); (N.P.); (P.F.); (M.N.)
| | - Mario Saia
- Azienda Zero of Veneto Region, 35100 Padua, Italy;
| | - Michele Tonon
- Regional Directorate of Prevention, Food Safety, Veterinary, Public Health—Regione del Veneto, 30123 Venezia, Italy; (M.T.); (M.M.); (F.R.)
| | - Michele Mongillo
- Regional Directorate of Prevention, Food Safety, Veterinary, Public Health—Regione del Veneto, 30123 Venezia, Italy; (M.T.); (M.M.); (F.R.)
| | - Francesca Russo
- Regional Directorate of Prevention, Food Safety, Veterinary, Public Health—Regione del Veneto, 30123 Venezia, Italy; (M.T.); (M.M.); (F.R.)
| | - Vincenzo Baldo
- Department of Cardiac Thoracic and Vascular Sciences and Public Health, University of Padua, 35131 Padua, Italy; (S.C.); (F.Z.); (G.F.); (N.P.); (P.F.); (M.N.)
- Correspondence: ; Tel.: +39-049-8275381
| |
Collapse
|
1368
|
Li W, Chen Y, Prévost J, Ullah I, Lu M, Gong SY, Tauzin A, Gasser R, Vézina D, Anand SP, Goyette G, Chaterjee D, Ding S, Tolbert WD, Grunst MW, Bo Y, Zhang S, Richard J, Zhou F, Huang RK, Esser L, Zeher A, Côté M, Kumar P, Sodroski J, Xia D, Uchil PD, Pazgier M, Finzi A, Mothes W. Structural basis and mode of action for two broadly neutralizing antibodies against SARS-CoV-2 emerging variants of concern. Cell Rep 2022; 38:110210. [PMID: 34971573 PMCID: PMC8673750 DOI: 10.1016/j.celrep.2021.110210] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/26/2021] [Accepted: 12/13/2021] [Indexed: 01/15/2023] Open
Abstract
Emerging variants of concern for the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can transmit more efficiently and partially evade protective immune responses, thus necessitating continued refinement of antibody therapies and immunogen design. Here, we elucidate the structural basis and mode of action for two potent SARS-CoV-2 spike (S)-neutralizing monoclonal antibodies, CV3-1 and CV3-25, which remain effective against emerging variants of concern in vitro and in vivo. CV3-1 binds to the (485-GFN-487) loop within the receptor-binding domain (RBD) in the "RBD-up" position and triggers potent shedding of the S1 subunit. In contrast, CV3-25 inhibits membrane fusion by binding to an epitope in the stem helix region of the S2 subunit that is highly conserved among β-coronaviruses. Thus, vaccine immunogen designs that incorporate the conserved regions in the RBD and stem helix region are candidates to elicit pan-coronavirus protective immune responses.
Collapse
Affiliation(s)
- Wenwei Li
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Yaozong Chen
- Infectious Disease Division, Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814-4712, USA
| | - Jérémie Prévost
- Centre de Recherche du CHUM (CRCHUM), Montreal, QC H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC H2X 0A9, Canada
| | - Irfan Ullah
- Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Maolin Lu
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Shang Yu Gong
- Centre de Recherche du CHUM (CRCHUM), Montreal, QC H2X 0A9, Canada; Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 2B4, Canada
| | - Alexandra Tauzin
- Centre de Recherche du CHUM (CRCHUM), Montreal, QC H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC H2X 0A9, Canada
| | - Romain Gasser
- Centre de Recherche du CHUM (CRCHUM), Montreal, QC H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC H2X 0A9, Canada
| | - Dani Vézina
- Centre de Recherche du CHUM (CRCHUM), Montreal, QC H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC H2X 0A9, Canada
| | - Sai Priya Anand
- Centre de Recherche du CHUM (CRCHUM), Montreal, QC H2X 0A9, Canada; Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 2B4, Canada
| | | | | | - Shilei Ding
- Centre de Recherche du CHUM (CRCHUM), Montreal, QC H2X 0A9, Canada
| | - William D Tolbert
- Infectious Disease Division, Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814-4712, USA
| | - Michael W Grunst
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Yuxia Bo
- Department of Biochemistry, Microbiology and Immunology, and Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Shijian Zhang
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA
| | - Jonathan Richard
- Centre de Recherche du CHUM (CRCHUM), Montreal, QC H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC H2X 0A9, Canada
| | - Fei Zhou
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rick K Huang
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lothar Esser
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Allison Zeher
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Marceline Côté
- Department of Biochemistry, Microbiology and Immunology, and Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Priti Kumar
- Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Joseph Sodroski
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA
| | - Di Xia
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Pradeep D Uchil
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Marzena Pazgier
- Infectious Disease Division, Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814-4712, USA.
| | - Andrés Finzi
- Centre de Recherche du CHUM (CRCHUM), Montreal, QC H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC H2X 0A9, Canada; Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 2B4, Canada.
| | - Walther Mothes
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
1369
|
Tostanoski LH, Chandrashekar A, Patel S, Yu J, Jacob-Dolan C, Chang A, Powers OC, Sellers D, Gardner S, Barrett J, Sanborn O, Stephenson KE, Ansel JL, Jaegle K, Seaman MS, Porto M, Lok M, Spence B, Cayer K, Nase D, Holman S, Bradette H, Kar S, Andersen H, Lewis MG, Cox F, Tolboom JTBM, de Groot AM, Heerwegh D, Le Gars M, Sadoff J, Wegmann F, Zahn RC, Schuitemaker H, Barouch DH. Passive transfer of Ad26.COV2.S-elicited IgG from humans attenuates SARS-CoV-2 disease in hamsters. NPJ Vaccines 2022; 7:2. [PMID: 35013325 PMCID: PMC8748674 DOI: 10.1038/s41541-021-00427-z] [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: 10/04/2021] [Accepted: 12/14/2021] [Indexed: 12/27/2022] Open
Abstract
SARS-CoV-2 Spike-specific binding and neutralizing antibodies, elicited either by natural infection or vaccination, have emerged as potential correlates of protection. An important question, however, is whether vaccine-elicited antibodies in humans provide direct, functional protection from SARS-CoV-2 infection and disease. In this study, we explored directly the protective efficacy of human antibodies elicited by Ad26.COV2.S vaccination by adoptive transfer studies. IgG from plasma of Ad26.COV2.S vaccinated individuals was purified and transferred into naïve golden Syrian hamster recipients, followed by intra-nasal challenge of the hamsters with SARS-CoV-2. IgG purified from Ad26.COV2.S-vaccinated individuals provided dose-dependent protection in the recipient hamsters from weight loss following challenge. In contrast, IgG purified from placebo recipients provided no protection in this adoptive transfer model. Attenuation of weight loss correlated with binding and neutralizing antibody titers of the passively transferred IgG. This study suggests that Ad26.COV2.S-elicited antibodies in humans are mechanistically involved in protection against SARS-CoV-2.
Collapse
Affiliation(s)
- Lisa H Tostanoski
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Abishek Chandrashekar
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Shivani Patel
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jingyou Yu
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Catherine Jacob-Dolan
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Aiquan Chang
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Olivia C Powers
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Daniel Sellers
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Sarah Gardner
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Julia Barrett
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Owen Sanborn
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Kathryn E Stephenson
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jessica L Ansel
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Kate Jaegle
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Michael S Seaman
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | | | | - Freek Cox
- Janssen Vaccines & Prevention, Leiden, The Netherlands
| | | | | | | | | | - Jerald Sadoff
- Janssen Vaccines & Prevention, Leiden, The Netherlands
| | - Frank Wegmann
- Janssen Vaccines & Prevention, Leiden, The Netherlands
| | - Roland C Zahn
- Janssen Vaccines & Prevention, Leiden, The Netherlands
| | | | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.
- Massachusetts Consortium on Pathogen Readiness, Boston, MA, USA.
| |
Collapse
|
1370
|
Paz-Bailey G, Sternberg M, Kugeler K, Hoots B, Amin AB, Johnson AG, Barbeau B, Bayoumi NS, Bertolino D, Boulton R, Brown CM, Busen K, Cima M, Drenzek C, Gent A, Haney G, Hicks L, Hook S, Jara A, Jones A, Kamal-Ahmed I, Kangas S, Kanishka FNU, Khan SI, Kirkendall SK, Kocharian A, Lyons BC, Lauro P, McCormick D, McMullen C, Milroy L, Reese HE, Sell J, Sierocki A, Smith E, Sosin D, Stanislawski E, Strand K, Troelstrup T, Turner KA, Vest H, Warner S, Wiedeman C, Silk B, Scobie HM. Covid-19 Rates by Time since Vaccination during Delta Variant Predominance. NEJM EVIDENCE 2022; 1:10.1056/evidoa2100057. [PMID: 37207114 PMCID: PMC10193243 DOI: 10.1056/evidoa2100057] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
BACKGROUND With the emergence of the delta variant, the United States experienced a rapid increase in Covid-19 cases in 2021. We estimated the risk of breakthrough infection and death by month of vaccination as a proxy for waning immunity during a period of delta variant predominance. METHODS Covid-19 case and death data from 15 U.S. jurisdictions during January 3 to September 4, 2021 were used to estimate weekly hazard rates among fully vaccinated persons, stratified by age group and vaccine product. Case and death rates during August 1 to September 4, 2021 were presented across four cohorts defined by month of vaccination. Poisson models were used to estimate adjusted rate ratios comparing the earlier cohorts to July rates. RESULTS During August 1 to September 4, 2021, case rates per 100,000 person-weeks among all vaccine recipients for the January to February, March to April, May to June, and July cohorts were 168.8 (95% confidence interval [CI], 167.5 to 170.1), 123.5 (95% CI, 122.8 to 124.1), 83.6 (95% CI, 82.9 to 84.3), and 63.1 (95% CI, 61.6 to 64.6), respectively. Similar trends were observed by age group for BNT162b2 (Pfizer-BioNTech) and mRNA-1273 (Moderna) vaccine recipients. Rates for the Ad26.COV2.S (Janssen-Johnson & Johnson) vaccine were higher; however, trends were inconsistent. BNT162b2 vaccine recipients 65 years of age or older had higher death rates among those vaccinated earlier in the year. Protection against death was sustained for the mRNA-1273 vaccine recipients. Across age groups and vaccine types, people who were vaccinated 6 months ago or longer (January-February) were 3.44 (3.36 to 3.53) times more likely to be infected and 1.70 (1.29 to 2.23) times more likely to die from COVID-19 than people vaccinated recently in July 2021. CONCLUSIONS Our study suggests that protection from SARS-CoV-2 infection among all ages or death among older adults waned with increasing time since vaccination during a period of delta predominance. These results add to the evidence base that supports U.S. booster recommendations, especially for older adults vaccinated with BNT162b2 and recipients of the Ad26.COV2.S vaccine. (Funded by the Centers for Disease Control and Prevention.).
Collapse
Affiliation(s)
- Gabriela Paz-Bailey
- Epidemiology Task Force, COVID-19 Response Team, Centers for Disease Control and Prevention, Atlanta
| | - Maya Sternberg
- Epidemiology Task Force, COVID-19 Response Team, Centers for Disease Control and Prevention, Atlanta
| | - Kiersten Kugeler
- Epidemiology Task Force, COVID-19 Response Team, Centers for Disease Control and Prevention, Atlanta
| | - Brooke Hoots
- Epidemiology Task Force, COVID-19 Response Team, Centers for Disease Control and Prevention, Atlanta
| | - Avnika B Amin
- Epidemiology Task Force, COVID-19 Response Team, Centers for Disease Control and Prevention, Atlanta
| | - Amelia G Johnson
- Epidemiology Task Force, COVID-19 Response Team, Centers for Disease Control and Prevention, Atlanta
| | | | | | - Daniel Bertolino
- New York City Department of Health and Mental Hygiene, Long Island City
| | | | | | | | | | | | | | | | - Liam Hicks
- Arizona Department of Health Services, Phoenix
| | - Sarah Hook
- Epidemiology Task Force, COVID-19 Response Team, Centers for Disease Control and Prevention, Atlanta
| | | | - Amanda Jones
- Center for Surveillance, Epidemiology, and Laboratory Services, Centers for Disease Control and Prevention, Atlanta
| | | | - Sarah Kangas
- Wisconsin Department of Health Services, Madison
| | - F N U Kanishka
- Nebraska Department of Health and Human Services, Lincoln
| | | | | | | | - B Casey Lyons
- Data Analytics and Visualization Task Force, COVID-19 Response Team, Centers for Disease Control and Prevention, Atlanta
| | | | | | | | | | - Heather E Reese
- Epidemiology Task Force, COVID-19 Response Team, Centers for Disease Control and Prevention, Atlanta
| | - Jessica Sell
- New York City Department of Health and Mental Hygiene, Long Island City
| | | | | | | | | | - Kyle Strand
- Nebraska Department of Health and Human Services, Lincoln
| | | | | | | | - Sydni Warner
- Wisconsin Department of Health Services, Madison
| | | | - Benjamin Silk
- Epidemiology Task Force, COVID-19 Response Team, Centers for Disease Control and Prevention, Atlanta
| | - Heather M Scobie
- Epidemiology Task Force, COVID-19 Response Team, Centers for Disease Control and Prevention, Atlanta
| |
Collapse
|
1371
|
Ozturk D, Gareayaghi N, Tahtasakal CA, Calik M, Altinbilek E. Antibody responses after two doses of CoronaVac of the participants with or without the diagnosis of COVID-19. Ir J Med Sci 2022; 191:2833-2838. [PMID: 35001336 PMCID: PMC8743087 DOI: 10.1007/s11845-021-02883-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 12/01/2021] [Indexed: 01/06/2023]
Abstract
Background CoronaVac, an inactivated whole-virion vaccine against COVID-19, has been shown to be safe with acceptable antibody responses by various clinical trials. Aims The objective was to investigate the post-vaccination antibody levels of both symptomatic and asymptomatic healthcare workers with or without the diagnosis of COVID-19 in an emergency department (ED) of a hospital serving as a pandemic hospital. Methods This single-centred, prospective study was conducted on 86 participants who were working as nurse or doctor in the ED. The volunteers were older than 18 years and either positive or negative for either computed tomography (CT), real-time reverse transcription polymerase chain reaction (qRT-PCR), or both. Thirty days after the second dose of CoronaVac (3 µg), the antibody levels were chemiluminescent microparticle immunoassay. Results Mean age of all participants were 33.1 ± 9.1 years. The antibody levels in the qRT-PCR( +) and CT( +) groups were significantly higher than the qRT-PCR( −) and CT( −) groups, respectively (p < 0.05). In the CT( +)/qRT-PCR( +) group, the antibody level was significantly higher than the CT( −)/qRT-PCR( −) and CT( −)/qRT-PCR( +) or CT( +)/qRT-PCR( −) group (p < 0.05). On the other hand, antibody levels in the hospitalized group were significantly higher than in the non-hospitalized group (p < 0.05). A significant positive correlation was observed between the time elapsed after vaccination and antibody levels of the participants (r = 0.343; p = 0.000). Conclusion In conclusion, antibody responses of recovered patients COVID-19 diagnosed by both CT and qRT-PCR were much robust than the patients diagnosed by either one of the techniques or undiagnosed/disease-free participants suggesting that severity of the disease likely contributes to the antibody responses after vaccination with CoronaVac.
Collapse
Affiliation(s)
- Derya Ozturk
- Department of Emergency Medicine, University of Health Sciences, Sisli Hamidiye Etfal Training and Research Hospital, Halaskargazi St, 34371, Sisli/Istanbul, Turkey
| | - Nesrin Gareayaghi
- Department of Microbiology, University of Health Sciences, Sisli Hamidiye Etfal Training and Research Hospital, Halaskargazi St, 34371, Sisli/Istanbul, Turkey
| | - Ceren Atasoy Tahtasakal
- Department of Infectious Diseases, University of Health Sciences, Sisli Hamidiye Etfal Training and Research Hospital, Halaskargazi St, 34371, Sisli/Istanbul, Turkey
| | - Mustafa Calik
- Department of Emergency Medicine, University of Health Sciences, Gaziosmanpaşa Training and Research Hospital, Osmanbey St, 34255, Gaziosmanpasa/Istanbul, Turkey.
| | - Ertugrul Altinbilek
- Department of Emergency Medicine, University of Health Sciences, Sisli Hamidiye Etfal Training and Research Hospital, Halaskargazi St, 34371, Sisli/Istanbul, Turkey
| |
Collapse
|
1372
|
Noureen S, Rehman K, Hamid Akash MS. Natural immunity boosters as therapeutic interventions in the era of COVID-19 pandemic. Endocr Metab Immune Disord Drug Targets 2022; 22:842-851. [PMID: 35016600 DOI: 10.2174/1871530322666220110113028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/04/2021] [Accepted: 11/26/2021] [Indexed: 12/15/2022]
Abstract
COVID-19, a pandemic caused by SARS-CoV2 has been spread all over the world and is responsible for serious fatalities. SARS-CoV2 is included in the family of β-coronavirus that affects pulmonary gas exchange and triggers cytokines storm. Vigorous inflammation, hyper-coagulation, a decrease in lymphocytic count and an increase in neutrophilic count are observed in the second week after the onset of disease. Fever, dry cough, sneezing, shortness of breath and respiratory distress are the symptoms of COVID-19. Different preventive measures are taken to prevent the attack of coronavirus amongst social distancing, vaccination, wearing gloves and face masks and the use of sanitizers plays pivotal role. People with weak immunity are more susceptible to the attack of coronavirus. Various natural immunity boosters are known for their immune boosting properties among them are vitamin C, D, and B complex, medicinal mushrooms, plant-based stuff and minerals play important role by increasing the beneficial flora of human body. All these natural immunity boosters improve the innate and adaptive immune response against coronavirus. Hence, we conclude that the use of natural immunity boosters prevents the attack of coronavirus and make a person stronger against the suspected attack of COVID-19 and/or other viral diseases.
Collapse
Affiliation(s)
- Sibgha Noureen
- Department of Pharmaceutical Chemistry, Government College University, Faisalabad, Pakistan
| | - Kanwal Rehman
- Department of Pharmacy, University of Agriculture, Faisalabad, Pakistan
| | | |
Collapse
|
1373
|
Dynamics of spike-and nucleocapsid specific immunity during long-term follow-up and vaccination of SARS-CoV-2 convalescents. Nat Commun 2022; 13:153. [PMID: 35013191 PMCID: PMC8748966 DOI: 10.1038/s41467-021-27649-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 12/02/2021] [Indexed: 12/23/2022] Open
Abstract
Anti-viral immunity continuously declines over time after SARS-CoV-2 infection. Here, we characterize the dynamics of anti-viral immunity during long-term follow-up and after BNT162b2 mRNA-vaccination in convalescents after asymptomatic or mild SARS-CoV-2 infection. Virus-specific and virus-neutralizing antibody titers rapidly declined in convalescents over 9 months after infection, whereas virus-specific cytokine-producing polyfunctional T cells persisted, among which IL-2-producing T cells correlated with virus-neutralizing antibody titers. Among convalescents, 5% of individuals failed to mount long-lasting immunity after infection and showed a delayed response to vaccination compared to 1% of naïve vaccinees, but successfully responded to prime/boost vaccination. During the follow-up period, 8% of convalescents showed a selective increase in virus-neutralizing antibody titers without accompanying increased frequencies of circulating SARS-CoV-2-specific T cells. The same convalescents, however, responded to vaccination with simultaneous increase in antibody and T cell immunity revealing the strength of mRNA-vaccination to increase virus-specific immunity in convalescents. Waning immunity to SARS-CoV-2 is of concern. Here the authors follow spike- and nucleocapsid specific immunity in convalescent individuals for 9 months observing a decline in antibody levels but persisting T cell response. Vaccination approximately 11 months after infection boosts antibody and T cell immunity.
Collapse
|
1374
|
Li R, Li Y, Zou Z, Liu Y, Li X, Zhuang G, Shen M, Zhang L. Evaluating the Impact of SARS-CoV-2 Variants on the COVID-19 Epidemic and Social Restoration in the United States: A Mathematical Modelling Study. Front Public Health 2022; 9:801763. [PMID: 35083192 PMCID: PMC8786080 DOI: 10.3389/fpubh.2021.801763] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/22/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Multiple SARS-CoV-2 variants are still rampant across the United States (US). We aimed to evaluate the impact of vaccination scale-up and potential reduction in the vaccination effectiveness on the COVID-19 epidemic and social restoration in the US. Methods: We extended a published compartmental model and calibrated the model to the latest US COVID-19 data. We estimated the vaccine effectiveness against the variant and evaluated the impact of a potential reduction in vaccine effectiveness on the epidemics. We explored the epidemic trends under different levels of social restoration. Results: We estimated the overall existing vaccine effectiveness against the variant as 88.5% (95% CI: 87.4-89.5%) with the vaccination coverage of 70% by the end of August, 2021. With this vaccine effectiveness and coverage, there would be 498,972 (109,998-885,947) cumulative infections and 15,443 (3,828-27,057) deaths nationwide over the next 12 months, of which 95.0% infections and 93.3% deaths were caused by the variant. Complete social restoration at 60, 65, 70% vaccination coverage would increase cumulative infections to 1.6 (0.2-2.9) million 0.7 (0.1-1.2) million, and 511,159 (110,578-911,740), respectively. At same time it would increase cumulative deaths to 39,040 (5,509-72,570), 19,562 (3,873-35,250), 15,739 (3,841-27,638), respectively. However, if the vaccine effectiveness were reduced to 75%, 50% or 25% due to new SARS-CoV-2 variants, there would be 667,075 (130,682-1,203,468), 1.7 (0.2-3.2) million, 19.0 (5.3-32.7) million new infections and 19,249 (4,281-34,217), 42,265 (5,081-79,448), 426,860 (117,229-736,490) cumulative deaths to occur over the next 12 months. Further, social restoration at a lower vaccination coverage would lead to even greater secondary outbreaks. Conclusion: Current COVID-19 vaccines remain effective against the SARS-CoV-2 variant, and 70% vaccination coverage would be sufficient to restore social activities to a pre-pandemic level. Further reduction in vaccine effectiveness against SARS-CoV-2 variants would result in a potential surge of the epidemic. Multiple measures, including public health interventions, vaccination scale-up and development of a new vaccine booster, should be integrated to counter the new challenges of new SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Rui Li
- China-Australia Joint Research Center for Infectious Diseases, Xi'an Jiaotong University Health Science Center, School of Public Health, Xi'an, China
| | - Yan Li
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Obstetrics, Gynecology, and Reproductive Science, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Zhuoru Zou
- China-Australia Joint Research Center for Infectious Diseases, Xi'an Jiaotong University Health Science Center, School of Public Health, Xi'an, China
| | - Yiming Liu
- China-Australia Joint Research Center for Infectious Diseases, Xi'an Jiaotong University Health Science Center, School of Public Health, Xi'an, China
| | - Xinghui Li
- School of Public Health, Shaanxi University of Chinese Medicine, Xi'an, China
| | - Guihua Zhuang
- China-Australia Joint Research Center for Infectious Diseases, Xi'an Jiaotong University Health Science Center, School of Public Health, Xi'an, China
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, China
| | - Mingwang Shen
- China-Australia Joint Research Center for Infectious Diseases, Xi'an Jiaotong University Health Science Center, School of Public Health, Xi'an, China
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, China
| | - Lei Zhang
- China-Australia Joint Research Center for Infectious Diseases, Xi'an Jiaotong University Health Science Center, School of Public Health, Xi'an, China
- Melbourne Sexual Health Centre, Alfred Health, Melbourne, VIC, Australia
- Faculty of Medicine, Nursing and Health Sciences, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
1375
|
Szczepanek J, Skorupa M, Goroncy A, Jarkiewicz-Tretyn J, Wypych A, Sandomierz D, Jarkiewicz-Tretyn A, Dejewska J, Ciechanowska K, Pałgan K, Rajewski P, Tretyn A. Anti-SARS-CoV-2 IgG against the S Protein: A Comparison of BNT162b2, mRNA-1273, ChAdOx1 nCoV-2019 and Ad26.COV2.S Vaccines. Vaccines (Basel) 2022; 10:99. [PMID: 35062760 PMCID: PMC8778136 DOI: 10.3390/vaccines10010099] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/28/2021] [Accepted: 01/07/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND COVID-19 vaccines induce a differentiated humoral and cellular response, and one of the comparable parameters of the vaccine response is the determination of IgG antibodies. MATERIALS AND METHODS Concentrations of IgG anti-SARS-CoV-2 antibodies were analyzed at three time points (at the beginning of May, at the end of June and at the end of September). Serum samples were obtained from 954 employees of the Nicolaus Copernicus University in Toruń (a total of three samples each were obtained from 511 vaccinated participants). IgG antibody concentrations were determined by enzyme immunoassay. The statistical analysis included comparisons between vaccines, between convalescents and COVID-19 non-patients, between individual measurements and included the gender, age and blood groups of participants. RESULTS There were significant differences in antibody levels between mRNA and vector vaccines. People vaccinated with mRNA-1273 achieved the highest levels of antibodies, regardless of the time since full vaccination. People vaccinated with ChAdOx1 nCoV-2019 produced several times lower antibody levels compared to the mRNA vaccines, while the antibody levels were more stable. In the case of each of the vaccines, the factor having the strongest impact on the level and stability of the IgG antibody titers was previous SARS-CoV-2 infection. There were no significant correlations with age, gender and blood type. SUMMARY mRNA vaccines induce a stronger humoral response of the immune system with the fastest loss of antibodies over time.
Collapse
Affiliation(s)
- Joanna Szczepanek
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University, 87-100 Torun, Poland; (M.S.); (A.W.)
| | - Monika Skorupa
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University, 87-100 Torun, Poland; (M.S.); (A.W.)
- Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 87-100 Torun, Poland;
| | - Agnieszka Goroncy
- Faculty of Mathematics and Computer Science, Nicolaus Copernicus University, 87-100 Torun, Poland;
| | - Joanna Jarkiewicz-Tretyn
- Cancer Genetics Laboratory Ltd., 87-100 Torun, Poland; (J.J.-T.); (D.S.); (A.J.-T.); (J.D.); (K.C.)
| | - Aleksandra Wypych
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University, 87-100 Torun, Poland; (M.S.); (A.W.)
- Academic Research Center AKAMED Ltd., 87-100 Torun, Poland
| | - Dorota Sandomierz
- Cancer Genetics Laboratory Ltd., 87-100 Torun, Poland; (J.J.-T.); (D.S.); (A.J.-T.); (J.D.); (K.C.)
| | - Aleksander Jarkiewicz-Tretyn
- Cancer Genetics Laboratory Ltd., 87-100 Torun, Poland; (J.J.-T.); (D.S.); (A.J.-T.); (J.D.); (K.C.)
- Polish-Japanese Academy of Information Technology, 02-008 Warszawa, Poland
| | - Joanna Dejewska
- Cancer Genetics Laboratory Ltd., 87-100 Torun, Poland; (J.J.-T.); (D.S.); (A.J.-T.); (J.D.); (K.C.)
| | - Karolina Ciechanowska
- Cancer Genetics Laboratory Ltd., 87-100 Torun, Poland; (J.J.-T.); (D.S.); (A.J.-T.); (J.D.); (K.C.)
| | - Krzysztof Pałgan
- Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum, Nicolaus Copernicus University, 85-067 Bydgoszcz, Poland;
| | - Paweł Rajewski
- Department of Internal and Infectious Diseases, Provincial Infectious Disease Hospital, 85-067 Bydgoszcz, Poland;
| | - Andrzej Tretyn
- Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 87-100 Torun, Poland;
| |
Collapse
|
1376
|
Efficacy and Safety of Third Dose of the COVID-19 Vaccine among Solid Organ Transplant Recipients: A Systemic Review and Meta-Analysis. Vaccines (Basel) 2022; 10:vaccines10010095. [PMID: 35062756 PMCID: PMC8778934 DOI: 10.3390/vaccines10010095] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/02/2022] [Accepted: 01/05/2022] [Indexed: 12/22/2022] Open
Abstract
Solid organ transplant recipients were demonstrated to have reduced antibody response to the first and second doses of the COVID-19 mRNA vaccine. This review evaluated published data on the efficacy and safety of the third dose among solid organ transplant recipients. We performed a systematic search of PubMed, EMBASE, and Web of Science to retrieve studies evaluating the efficacy of the third dose of anti-SARS-CoV-2 vaccines in adult solid organ transplant recipients. Serologic response after the third vaccine was pooled using inverse variance and generalized linear mixed and random-effects models. Seven studies met our inclusion criteria. A total of 853 patients received the third dose. Except for one randomized controlled trial, all studies were retrospective in design. Following the third COVID-19 vaccine dose, antibody response occurred in 6.4–69.2% of patients. The pooled proportion of antibody response rate after the third vaccine was 50.3% (95% confidence interval (CI): 37.1–63.5, I2 = 90%). Five papers reported the safety profile. No severe adverse events were observed after the third vaccine dose. In conclusion, a third dose of the SARS-CoV-2 mRNA vaccine in solid organ transplant recipients is associated with improved immunogenicity and appears to be safe. Nevertheless, a significant portion of patients remain seronegative.
Collapse
|
1377
|
Capoferri AA, Shao W, Spindler J, Coffin JM, Rausch JW, Kearney MF. A Pre-Vaccination Baseline of SARS-CoV-2 Genetic Surveillance and Diversity in the United States. Viruses 2022; 14:104. [PMID: 35062308 PMCID: PMC8778900 DOI: 10.3390/v14010104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 01/04/2022] [Indexed: 12/14/2022] Open
Abstract
COVID-19 vaccines were first administered on 15 December 2020, marking an important transition point for the spread of SARS-CoV-2 in the United States (U.S.). Prior to this point in time, the virus spread to an almost completely immunologically naïve population, whereas subsequently, vaccine-induced immune pressure and prior infections might be expected to influence viral evolution. Accordingly, we conducted a study to characterize the spread of SARS-CoV-2 in the U.S. pre-vaccination, investigate the depth and uniformity of genetic surveillance during this period, and measure and otherwise characterize changing viral genetic diversity, including by comparison with more recently emergent variants of concern (VOCs). In 2020, SARS-CoV-2 spread across the U.S. in three phases distinguishable by peaks in the numbers of infections and shifting geographical distributions. Virus was genetically sampled during this period at an overall rate of ~1.2%, though there was a substantial mismatch between case rates and genetic sampling nationwide. Viral genetic diversity tripled over this period but remained low in comparison to other widespread RNA virus pathogens, and although 54 amino acid changes were detected at frequencies exceeding 5%, linkage among them was not observed. Based on our collective observations, our analysis supports a targeted strategy for worldwide genetic surveillance as perhaps the most sensitive and efficient means of detecting new VOCs.
Collapse
Affiliation(s)
- Adam A Capoferri
- HIV Dynamics and Replication Program, Center for Cancer Research, NCI-Frederick, Frederick, MD 21702, USA
- Department of Microbiology and Immunology, Georgetown University, Washington, DC 20007, USA
| | - Wei Shao
- Advanced Biomedical Computing Science, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Jon Spindler
- HIV Dynamics and Replication Program, Center for Cancer Research, NCI-Frederick, Frederick, MD 21702, USA
| | - John M Coffin
- Department of Molecular Biology and Microbiology, Tufts University, Boston, MA 02129, USA
| | - Jason W Rausch
- HIV Dynamics and Replication Program, Center for Cancer Research, NCI-Frederick, Frederick, MD 21702, USA
| | - Mary F Kearney
- HIV Dynamics and Replication Program, Center for Cancer Research, NCI-Frederick, Frederick, MD 21702, USA
| |
Collapse
|
1378
|
Hafiz I, Illian DN, Meila O, Utomo ARH, Susilowati A, Susetya IE, Desrita D, Siregar GA, Basyuni M. Effectiveness and Efficacy of Vaccine on Mutated SARS-CoV-2 Virus and Post Vaccination Surveillance: A Narrative Review. Vaccines (Basel) 2022; 10:82. [PMID: 35062743 PMCID: PMC8777947 DOI: 10.3390/vaccines10010082] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/19/2021] [Accepted: 01/04/2022] [Indexed: 02/04/2023] Open
Abstract
The ongoing COVID-19 pandemic, as a result of the SARS-CoV-2 virus, since December 2019, is a major health problem and concern worldwide. The pandemic has impacted various fields, from the social to the development of health science and technology. The virus has been mutating and thus producing several new variants, rushing research in the field of molecular biology to develop rapidly to overcome the problems that occur. Vaccine clinical studies are developing promptly with the aim of obtaining vaccines that are effective in suppressing the spread of the virus; however, the development of viral mutations raises concerns about the decreasing effectiveness of the resulting vaccine, which also results in the need for more in-depth studies. There have been 330 vaccines developed, including 136 clinical developments and 194 pre-clinical developments. The SARS-CoV-2 variant continues to evolve today, and it poses a challenge in testing the effectiveness of existing vaccines. This is a narrative review describing the emergence of the COVID-19 pandemic, development of vaccine platforms, identification of concerning mutations and virus variants in various countries of the world, and real-world monitoring of post-vaccination effectiveness and surveillance.
Collapse
Affiliation(s)
- Ihsanul Hafiz
- Department of Pharmacology, Faculty of Pharmacy and Health, Institut Kesehatan Helvetia, Medan 20124, Indonesia;
- Doctoral Program in Pharmacy, Department of Pharmaceutical Biology, School of Pharmacy, Institut Teknologi Bandung, Bandung 40132, Indonesia
| | - Didi Nurhadi Illian
- Department of Pharmacy, Faculty of Mathematics and Natural Sciences, Universitas Syiah Kuala, Banda Aceh 23111, Indonesia; (D.N.I.); (O.M.)
| | - Okpri Meila
- Department of Pharmacy, Faculty of Mathematics and Natural Sciences, Universitas Syiah Kuala, Banda Aceh 23111, Indonesia; (D.N.I.); (O.M.)
- Doctoral Program of Clinical Pharmacy and Pharmacology, Faculty of Pharmacy, Universitas Indonesia, Depok 16424, Indonesia
| | | | - Arida Susilowati
- Center of Excellence for Mangrove, Universitas Sumatera Utara, Medan 20155, Indonesia; (A.S.); (I.E.S.); (D.D.); (G.A.S.)
- Department of Forestry, Faculty of Forestry, Universitas Sumatera Utara, Medan 20155, Indonesia
| | - Ipanna Enggar Susetya
- Center of Excellence for Mangrove, Universitas Sumatera Utara, Medan 20155, Indonesia; (A.S.); (I.E.S.); (D.D.); (G.A.S.)
- Department of Aquatic Resource Management, Faculty of Agriculture, Universitas Sumatera Utara, Medan 20155, Indonesia
| | - Desrita Desrita
- Center of Excellence for Mangrove, Universitas Sumatera Utara, Medan 20155, Indonesia; (A.S.); (I.E.S.); (D.D.); (G.A.S.)
- Department of Aquatic Resource Management, Faculty of Agriculture, Universitas Sumatera Utara, Medan 20155, Indonesia
| | - Gontar Alamsyah Siregar
- Center of Excellence for Mangrove, Universitas Sumatera Utara, Medan 20155, Indonesia; (A.S.); (I.E.S.); (D.D.); (G.A.S.)
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, Faculty of Medicine, Universitas Sumatera Utara, Medan 20155, Indonesia
| | - Mohammad Basyuni
- Center of Excellence for Mangrove, Universitas Sumatera Utara, Medan 20155, Indonesia; (A.S.); (I.E.S.); (D.D.); (G.A.S.)
- Department of Forestry, Faculty of Forestry, Universitas Sumatera Utara, Medan 20155, Indonesia
| |
Collapse
|
1379
|
Gagne M, Corbett KS, Flynn BJ, Foulds KE, Wagner DA, Andrew SF, Todd JPM, Honeycutt CC, McCormick L, Nurmukhambetova ST, Davis-Gardner ME, Pessaint L, Bock KW, Nagata BM, Minai M, Werner AP, Moliva JI, Tucker C, Lorang CG, Zhao B, McCarthy E, Cook A, Dodson A, Teng IT, Mudvari P, Roberts-Torres J, Laboune F, Wang L, Goode A, Kar S, Boyoglu-Barnum S, Yang ES, Shi W, Ploquin A, Doria-Rose N, Carfi A, Mascola JR, Boritz EA, Edwards DK, Andersen H, Lewis MG, Suthar MS, Graham BS, Roederer M, Moore IN, Nason MC, Sullivan NJ, Douek DC, Seder RA. Protection from SARS-CoV-2 Delta one year after mRNA-1273 vaccination in rhesus macaques coincides with anamnestic antibody response in the lung. Cell 2022; 185:113-130.e15. [PMID: 34921774 PMCID: PMC8639396 DOI: 10.1016/j.cell.2021.12.002] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/23/2021] [Accepted: 11/30/2021] [Indexed: 01/07/2023]
Abstract
mRNA-1273 vaccine efficacy against SARS-CoV-2 Delta wanes over time; however, there are limited data on the impact of durability of immune responses on protection. Here, we immunized rhesus macaques and assessed immune responses over 1 year in blood and upper and lower airways. Serum neutralizing titers to Delta were 280 and 34 reciprocal ID50 at weeks 6 (peak) and 48 (challenge), respectively. Antibody-binding titers also decreased in bronchoalveolar lavage (BAL). Four days after Delta challenge, the virus was unculturable in BAL, and subgenomic RNA declined by ∼3-log10 compared with control animals. In nasal swabs, sgRNA was reduced by 1-log10, and the virus remained culturable. Anamnestic antibodies (590-fold increased titer) but not T cell responses were detected in BAL by day 4 post-challenge. mRNA-1273-mediated protection in the lungs is durable but delayed and potentially dependent on anamnestic antibody responses. Rapid and sustained protection in upper and lower airways may eventually require a boost.
Collapse
Affiliation(s)
- Matthew Gagne
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kizzmekia S Corbett
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Barbara J Flynn
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kathryn E Foulds
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Danielle A Wagner
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Shayne F Andrew
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - John-Paul M Todd
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Christopher Cole Honeycutt
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lauren McCormick
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Saule T Nurmukhambetova
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Meredith E Davis-Gardner
- Department of Pediatrics, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | - Kevin W Bock
- Infectious Disease Pathogenesis Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20892, USA
| | - Bianca M Nagata
- Infectious Disease Pathogenesis Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20892, USA
| | - Mahnaz Minai
- Infectious Disease Pathogenesis Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20892, USA
| | - Anne P Werner
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Juan I Moliva
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Courtney Tucker
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Cynthia G Lorang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Bingchun Zhao
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Elizabeth McCarthy
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | - I-Ting Teng
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Prakriti Mudvari
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jesmine Roberts-Torres
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Farida Laboune
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lingshu Wang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | - Seyhan Boyoglu-Barnum
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Eun Sung Yang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wei Shi
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Aurélie Ploquin
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nicole Doria-Rose
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - John R Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Eli A Boritz
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | - Mehul S Suthar
- Department of Pediatrics, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ian N Moore
- Infectious Disease Pathogenesis Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20892, USA
| | - Martha C Nason
- Biostatistics Research Branch, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nancy J Sullivan
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Daniel C Douek
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
1380
|
Avelino-Silva VI, de Barros MTL, Class 8 FICSAE Working Group. Assessment of novel technologies in healthcare - off-label use of drugs and the ethics of implementation and distribution of COVID-19 vaccines. EINSTEIN-SAO PAULO 2022; 19:eED6840. [PMID: 35019043 PMCID: PMC8693880 DOI: 10.31744/einstein_journal/2021ed6840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Vivian Iida Avelino-Silva
- Faculdade Israelita de Ciências da Saúde Albert EinsteinHospital Israelita Albert EinsteinSão PauloSPBrazilFaculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | - Mario Thadeu Leme de Barros
- Faculdade Israelita de Ciências da Saúde Albert EinsteinHospital Israelita Albert EinsteinSão PauloSPBrazilFaculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | - Class 8 FICSAE Working Group
- Faculdade Israelita de Ciências da Saúde Albert EinsteinHospital Israelita Albert EinsteinSão PauloSPBrazilFaculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| |
Collapse
|
1381
|
Vilches TN, Moghadas SM, Sah P, Fitzpatrick MC, Shoukat A, Pandey A, Galvani AP. Estimating COVID-19 Infections, Hospitalizations, and Deaths Following the US Vaccination Campaigns During the Pandemic. JAMA Netw Open 2022; 5:e2142725. [PMID: 35015067 PMCID: PMC8753510 DOI: 10.1001/jamanetworkopen.2021.42725] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
This decision analytic modeling study uses a simulation model to evaluate the association of US COVID-19 vaccination campaigns with infections, hospitalizations, and deaths.
Collapse
Affiliation(s)
- Thomas N. Vilches
- Agent-Based Modelling Laboratory, York University, Toronto, Ontario, Canada
- Center for Infectious Disease Modeling and Analysis, Yale School of Public Health, New Haven, Connecticut
| | - Seyed M. Moghadas
- Agent-Based Modelling Laboratory, York University, Toronto, Ontario, Canada
| | - Pratha Sah
- Center for Infectious Disease Modeling and Analysis, Yale School of Public Health, New Haven, Connecticut
| | - Meagan C. Fitzpatrick
- Center for Infectious Disease Modeling and Analysis, Yale School of Public Health, New Haven, Connecticut
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore
| | - Affan Shoukat
- Center for Infectious Disease Modeling and Analysis, Yale School of Public Health, New Haven, Connecticut
| | - Abhishek Pandey
- Center for Infectious Disease Modeling and Analysis, Yale School of Public Health, New Haven, Connecticut
| | - Alison P. Galvani
- Center for Infectious Disease Modeling and Analysis, Yale School of Public Health, New Haven, Connecticut
| |
Collapse
|
1382
|
Liu J, Chandrashekar A, Sellers D, Barrett J, Lifton M, McMahan K, Sciacca M, VanWyk H, Wu C, Yu J, Collier ARY, Barouch DH. Vaccines Elicit Highly Cross-Reactive Cellular Immunity to the SARS-CoV-2 Omicron Variant. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.01.02.22268634. [PMID: 35018387 PMCID: PMC8750713 DOI: 10.1101/2022.01.02.22268634] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The highly mutated SARS-CoV-2 Omicron (B.1.1.529) variant has been shown to evade a substantial fraction of neutralizing antibody responses elicited by current vaccines that encode the WA1/2020 Spike immunogen 1 , resulting in increased breakthrough infections and reduced vaccine efficacy. Cellular immune responses, particularly CD8+ T cell responses, are likely critical for protection against severe SARS-CoV-2 disease 2-6 . Here we show that cellular immunity induced by current SARS-CoV-2 vaccines is highly cross-reactive against the SARS-CoV-2 Omicron variant. Individuals who received Ad26.COV2.S or BNT162b2 vaccines demonstrated durable CD8+ and CD4+ T cell responses that showed extensive cross-reactivity against both the Delta and Omicron variants, including in central and effector memory cellular subpopulations. Median Omicron-specific CD8+ T cell responses were 82-84% of WA1/2020-specific CD8+ T cell responses. These data suggest that current vaccines may provide considerable protection against severe disease with the SARS-CoV-2 Omicron variant despite the substantial reduction of neutralizing antibody responses.
Collapse
Affiliation(s)
- Jinyan Liu
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | | | - Julia Barrett
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | | | | | - Haley VanWyk
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Cindy Wu
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Jingyou Yu
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | - Dan H. Barouch
- Beth Israel Deaconess Medical Center, Boston, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| |
Collapse
|
1383
|
Marco JJG, Pasquín MJÁ, Martín SM, Miranda APJ. Papel protector de las actuales vacunas para las variantes del virus SARS-CoV-2 y la COVID persistente. FMC : FORMACION MEDICA CONTINUADA EN ATENCION PRIMARIA 2022; 29:16-25. [PMID: 35095266 PMCID: PMC8788134 DOI: 10.1016/j.fmc.2021.10.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 06/14/2023]
Abstract
Puntos para una lectura rápida•La COVID-19 persistente es un complejo plurisintomático de difícil manejo clínico. •La vacunación por sí sola no es suficiente para controlar la pandemia. •La aparición de variantes del SARS-CoV-2 puede condicionar la respuesta vacunal. •La variante de mayor interés epidemiológico y la predominante actualmente es la delta. •Las vacunas con tecnología ARNm son efectivas contra las variantes. •La mejor estrategia contra las variantes es estar correctamente vacunado.
Collapse
Affiliation(s)
- José Javier Gómez Marco
- Médico de familia. Miembro del Grupo de Infecciosas PAPPS-semFYC. CSU Las Calesas, SERMAS. Madrid, España
| | | | - Susana Martín Martín
- Médica de familia. Coordinadora del Grupo de Infecciosas PAPPS-semFYC. Centro de Salud Balmaseda, Balmaseda, Vizcaya. España
| | | |
Collapse
|
1384
|
Akbarzadehlaleh P, Farjami A, Montazersaheb S, Soofiyani S, Salatin S. Biopharmaceuticals for prevention of COVID-19: A scoping review. ASIAN PAC J TROP MED 2022. [DOI: 10.4103/1995-7645.348158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
1385
|
Gupta S, Sudhindran S, Saraf N, Vijai A, Swaminathan S, Panackel C, Mehta NN, Varghese J, Singh S, Reddy MS, M. Sivaramakrishnan V, Bhangui P, Mohanka R, Asthana S, Rohatgi S. Liver Transplant Society of India Guidelines for Liver Transplant During COVID-19 times. J Clin Exp Hepatol 2022; 12:180-185. [PMID: 34429571 PMCID: PMC8378015 DOI: 10.1016/j.jceh.2021.08.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/15/2021] [Indexed: 12/12/2022] Open
Abstract
Coronavirus disease-2019 (COVID-19) pandemic has affected liver transplantation in many ways. There is risk of infection to the transplant recipients; and COVID-19 is associated with significant risk of mortality in patients on wait list. The Liver Transplant Society of India (LTSI) has prepared guidelines regarding selection of adult and pediatric patients for liver transplantation, transplant for acute liver failure, use of deceased donor organs, transplant techniques and minimally invasive donor hepatectomy, pre- and postsurgery testing for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-related coronavirus disease 2019 in donors and recipients, role of COVID-19 antibody testing, shifting of recipients from COVID-19 to non-COVID-19 areas after recovery, isolation policy of team members exposed to COVID-19 patients, drug therapy of proven or suspected COVID-19 infection early posttransplant, care of SARS-CoV-2 positive donors and recipients and a separate COVID-19 consent for surgery.
Collapse
Affiliation(s)
| | | | - Neeraj Saraf
- Medanta The Medicity, Gurgaon, India,Address for correspondence: Neeraj Saraf. Medanta Institute of Liver Transplantation and Regenerative Medicine, Medanta The Medicity Hospital, Sector 38, Gurgaon, Delhi (NCR), India.
| | - Anand Vijai
- GEM Hospital and Research Center, Coimbatore, India
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1386
|
Akram M, Inder D, Manak S, Kumar P. Recent update on treatment and preventive modalities for COVID-19 Omicron variant (B.1.1.529) in India: A comprehensive review. SAUDI JOURNAL FOR HEALTH SCIENCES 2022. [DOI: 10.4103/sjhs.sjhs_42_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
1387
|
Simnani FZ, Singh D, Kaur R. COVID-19 phase 4 vaccine candidates, effectiveness on SARS-CoV-2 variants, neutralizing antibody, rare side effects, traditional and nano-based vaccine platforms: a review. 3 Biotech 2022; 12:15. [PMID: 34926119 PMCID: PMC8665991 DOI: 10.1007/s13205-021-03076-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 11/26/2021] [Indexed: 12/12/2022] Open
Abstract
The COVID-19 pandemic has endangered world health and the economy. As the number of cases is increasing, different companies have started developing potential vaccines using both traditional and nano-based platforms to overcome the pandemic. Several countries have approved a few vaccine candidates for emergency use authorization (EUA), showing significant effectiveness and inducing a robust immune response. Oxford-AstraZeneca, Pfizer-BioNTech's BNT162, Moderna's mRNA-1273, Sinovac's CoronaVac, Johnson & Johnson, Sputnik-V, and Sinopharm's vaccine candidates are leading the race. However, the SARS-CoV-2 is constantly mutating, making the vaccines less effective, possibly by escaping immune response for some variants. Besides, some EUA vaccines have been reported to induce rare side effects such as blood clots, cardiac injury, anaphylaxis, and some neurological effects. Although the COVID-19 vaccine candidates promise to overcome the pandemic, a more significant and clear understanding is needed. In this review, we brief about the clinical trial of some leading candidates, their effectiveness, and their neutralizing effect on SARS-CoV-2 variants. Further, we have discussed the rare side effects, different traditional and nano-based platforms to understand the scope of future development.
Collapse
Affiliation(s)
| | - Dibyangshee Singh
- KIIT School of Biotechnology, KIIT University, Bhubaneswar, 751024 India
| | - Ramneet Kaur
- Department of Life Sciences, RIMT University, Ludhiana, Punjab India
| |
Collapse
|
1388
|
Baena-García L, Aparicio VA, Molina-López A, Aranda P, Cámara-Roca L, Ocón-Hernández O. Premenstrual and menstrual changes reported after COVID-19 vaccination: The EVA project. WOMEN'S HEALTH (LONDON, ENGLAND) 2022; 18:17455057221112237. [PMID: 35833668 PMCID: PMC9289916 DOI: 10.1177/17455057221112237] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/13/2022] [Accepted: 06/16/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND Menstrual disorders were not reported as a possible secondary effect in any of the clinical trials for the SARS-CoV-2 vaccines. AIM To describe the prevalence of perceived premenstrual and menstrual changes after COVID-19 vaccine administration. DESIGN Cross-sectional study. METHODS A total of 14,153 women (mean age 31.5 ± 9.3 years old) who had received the full course of vaccination at least three months earlier were included in this cross-sectional study. Data including the type of vaccine administered, perceived changes in the amount and duration of menstrual bleeding, presence of clots, cycle length, and premenstrual symptoms were collected through a retrospective online survey from June to September 2021. RESULTS Of the women who participated in this study, 3136 reported no menstrual changes and 11,017 (78% of the study sample) reported experiencing menstrual cycle changes after vaccination. In summary, women who reported menstrual changes after vaccination were older (overall p < 0.001) and slightly more smokers (p = 0.05) than women who did not report any changes. The most prevalent changes in relation to premenstrual symptoms were increased fatigue (43%), abdominal bloating (37%), irritability (29%), sadness (28%), and headaches (28%). The most predominant menstrual changes were more menstrual bleeding (43%), more menstrual pain (41%), delayed menstruation (38%), fewer days of menstrual bleeding (34.5%), and shorter cycle length (32%). CONCLUSION Women vaccinated against COVID-19 usually perceive mild menstrual and premenstrual changes. Future studies are warranted to clarify the physiological mechanisms behind these widely reported changes.
Collapse
Affiliation(s)
- Laura Baena-García
- Department of Nursing, Faculty of
Health Sciences, University of Granada, Ceuta, Spain
- Sport and Health University Research
Institute (iMUDS), Granada, Spain
- iInstituto de Investigación
Biosanitaria, ibs. Granada, Spain
| | - Virginia A Aparicio
- Sport and Health University Research
Institute (iMUDS), Granada, Spain
- Institute of Nutrition and Food
Technology (INYTA), Biomedical Research Centre (CIBM), University of Granada,
Granada, Spain
- Department of Physiology, Faculty of
Pharmacy, University of Granada, Granada, Spain
| | - Ana Molina-López
- Gynaecology and Obstetrics Unit, ‘San
Cecilio’ University Hospital, Granada, Spain
| | - Pilar Aranda
- Department of Physiology, Faculty of
Pharmacy, University of Granada, Granada, Spain
| | - Laura Cámara-Roca
- Gynaecology and Obstetrics Unit,
‘Virgen de las Nieves’ University Hospital, Granada, Spain
| | - Olga Ocón-Hernández
- iInstituto de Investigación
Biosanitaria, ibs. Granada, Spain
- Gynaecology and Obstetrics Unit, ‘San
Cecilio’ University Hospital, Granada, Spain
| |
Collapse
|
1389
|
Gyang TV, Evans JP, Miller JS, Alcorn K, Peng J, Bell EH, Zeng C, Gumina R, Liu SL, Segal BM. Neutralizing antibody responses against SARS-CoV-2 in vaccinated people with multiple sclerosis. Mult Scler J Exp Transl Clin 2022; 8:20552173221087357. [PMID: 35342640 PMCID: PMC8941285 DOI: 10.1177/20552173221087357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 02/25/2022] [Indexed: 11/27/2022] Open
Abstract
Background Patients with multiple sclerosis (pwMS) are often treated with disease
modifying therapies (DMT) with immunomodulatory effects. This is of
particular concern following the development of several vaccines to combat
coronavirus disease 19 (COVD-19), a potentially fatal illness caused by
SARS-CoV-2. Objectives To determine the efficacy of SARS-CoV-2 vaccination in pwMS and the impact of
disease modifying therapies (DMT) on vaccine response. Methods This is a prospective longitudinal study in pwMS. Longitudinal serum samples
were obtained prior to, and after SARS-CoV-2 mRNA vaccination. A novel
neutralizing antibody (nAb) assay was used to determine nAbs titres against
SARS-CoV-2 spike. Results We observed that (1) pwMS on B-cell depleting therapies exhibited reduced
response to vaccination compared to other pwMS, correlating with time from
last anti-CD20 infusion, (2) prior COVID-19 illness, DMT category, and
pyramidal function were significant predictors of vaccine responsiveness,
and (3) circulating absolute lymphocyte count (ALC) and IgG levels
correlated with nAb levels. Conclusions We demonstrate that pwMS exhibit reduced nAb response to mRNA vaccination
dependent on DMT status and identify predictive biomarkers for vaccine
efficacy. We conclude that additional vaccination strategies may be
necessary to achieve protective immunity in pwMS.
Collapse
Affiliation(s)
- Tirisham V Gyang
- Department of Neurology, The Ohio State University, Columbus, OH 43210, USA
| | - John P Evans
- Center for Retrovirus Research, The Ohio State University, Columbus, OH 43210, USA
| | - Joseph S Miller
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Kariss Alcorn
- College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Juan Peng
- Department of Biomedical Informatics, Center for Biostatistics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Erica H Bell
- Department of Neurology, The Ohio State University, Columbus, OH 43210, USA
| | - Cong Zeng
- Center for Retrovirus Research, The Ohio State University, Columbus, OH 43210, USA
| | - Richard Gumina
- Department of Internal Medicine, Division of Cardiovascular Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Shan-Lu Liu
- Center for Retrovirus Research, The Ohio State University, Columbus, OH 43210, USA
| | - Benjamin M Segal
- Department of Neurology, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
1390
|
Yilmaz IC, Ipekoglu EM, Bulbul A, Turay N, Yildirim M, Evcili I, Yilmaz NS, Guvencli N, Aydin Y, Gungor B, Saraydar B, Bartan AG, Ibibik B, Bildik T, Baydemir İ, Sanli HA, Kayaoglu B, Ceylan Y, Yildirim T, Abras I, Ayanoglu IC, Cam SB, Ciftci Dede E, Gizer M, Erganis O, Sarac F, Uzar S, Enul H, Adiay C, Aykut G, Polat H, Yildirim IS, Tekin S, Korukluoglu G, Zeytin HE, Korkusuz P, Gursel I, Gursel M. Development and preclinical evaluation of virus-like particle vaccine against COVID-19 infection. Allergy 2022; 77:258-270. [PMID: 34519053 PMCID: PMC8653174 DOI: 10.1111/all.15091] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 08/02/2021] [Accepted: 08/15/2021] [Indexed: 12/16/2022]
Abstract
Background Vaccines that incorporate multiple SARS‐CoV‐2 antigens can further broaden the breadth of virus‐specific cellular and humoral immunity. This study describes the development and immunogenicity of SARS‐CoV‐2 VLP vaccine that incorporates the four structural proteins of SARS‐CoV‐2. Methods VLPs were generated in transiently transfected HEK293 cells, purified by multimodal chromatography, and characterized by tunable‐resistive pulse sensing, AFM, SEM, and TEM. Immunoblotting studies verified the protein identities of VLPs. Cellular and humoral immune responses of immunized animals demonstrated the immune potency of the formulated VLP vaccine. Results Transiently transfected HEK293 cells reproducibly generated vesicular VLPs that were similar in size to and expressing all four structural proteins of SARS‐CoV‐2. Alum adsorbed, K3‐CpG ODN‐adjuvanted VLPs elicited high titer anti‐S, anti‐RBD, anti‐N IgG, triggered multifunctional Th1‐biased T‐cell responses, reduced virus load, and prevented lung pathology upon live virus challenge in vaccinated animals. Conclusion These data suggest that VLPs expressing all four structural protein antigens of SARS‐CoV‐2 are immunogenic and can protect animals from developing COVID‐19 infection following vaccination.
Collapse
|
1391
|
Valente M, Dalmonte G, Riccò M, Prioriello C, Ballabeni L, Peruzzi S, Marchesi F. Knowledge, Attitudes and Practices towards SARS-CoV-2 vaccination among morbid obese individuals: a pilot study. ACTA BIO-MEDICA : ATENEI PARMENSIS 2022; 93:e2022234. [PMID: 35775776 PMCID: PMC9335429 DOI: 10.23750/abm.v93i3.12386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 10/10/2021] [Indexed: 11/23/2022]
Abstract
BACKGROUND AND AIM Vaccinations have dramatically impacted on the ongoing pandemic of COVID-19, the disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). As morbid obese (MO) individuals are at high risk for severe complications, their acceptance of SARS-CoV-2 vaccines is of certain public health interest. METHODS We investigated the knowledge, attitudes and eventual acceptance of SARS-CoV-2/COVID-19 vaccination among MO individuals either in waiting list, or recipients of bariatric surgery from a reference center (Parma University Hospital) shortly before the inception of the Italian mass vaccination campaign (March 2021). Data were collected through a web-based questionnaire. Association of individual factors with acceptance of SARS-CoV-2 vaccine was assessed by means of a logistic regression analysis with eventual calculation of adjusted Odds Ratios (aOR) and corresponding 95% Confidence Intervals (95%CI). RESULTS Adequate, general knowledge of SARS-CoV-2/COVID-19 was found in the majority of MO patients. High perception of SARS-CoV-2 risk was found in around 80% of participants (79.2% regarding its occurrence, 73.6% regarding its potential severity). Acceptance of SARS-CoV-2/COVID-19 vaccination was reported by 65.3% of participants, and was more likely endorsed by MO patients who were likely to accept some sort of payment/copayment (aOR 5.783; 1.426; 23.456), or who were more likely towards a vaccination mandate (aOR 7.920; 1.995; 31.444). CONCLUSIONS Around one third of the MO individuals among potential recipient of bariatric surgery exhibited some significant hesitancy towards SARS-CoV-2 vaccine, and a rational approach may fail to capture and address specific barriers/motivators in this subset of individuals, stressing the importance for alternative interventions. (www.actabiomedica.it).
Collapse
Affiliation(s)
- Marina Valente
- University of Parma, Department of Medicine and Surgery, School of General Surgery, Parma (PR), Italy
| | - Giorgio Dalmonte
- University of Parma, Department of Medicine and Surgery, School of General Surgery, Parma (PR), Italy
| | - Matteo Riccò
- AUSL – IRCCS di Reggio Emilia, Servizio di Prevenzione e Sicurezza negli ambienti di Lavoro (SPSAL), Reggio Emilia (RE), Italy
| | - Concetta Prioriello
- University of Parma, Department of Medicine and Surgery, School of General Surgery, Parma (PR), Italy
| | - Lucia Ballabeni
- University of Parma, Department of Medicine and Surgery, School of General Surgery, Parma (PR), Italy
| | - Simona Peruzzi
- AUSL – IRCCS di Reggio Emilia, Laboratorio Analisi Chimico Cliniche e Microbiologiche, Ospedale Civile di Guastalla, Guastalla (RE), Italy
| | - Federico Marchesi
- University of Parma, Department of Medicine and Surgery, School of General Surgery, Parma (PR), Italy
| |
Collapse
|
1392
|
Zhang M, Liang Y, Yu D, Du B, Cheng W, Li L, Yu Z, Luo S, Zhang Y, Wang H, Zhang X, Zhang W. A systematic review of Vaccine Breakthrough Infections by SARS-CoV-2 Delta Variant. Int J Biol Sci 2022; 18:889-900. [PMID: 35002532 PMCID: PMC8741840 DOI: 10.7150/ijbs.68973] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/02/2021] [Indexed: 12/11/2022] Open
Abstract
Vaccines are proving to be highly effective in controlling hospitalization and deaths associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, as shown by clinical trials and real-world evidence. However, a deadly second wave of coronavirus disease 2019 (COVID-19), infected by SARS-CoV-2 variants, especially the Delta (B.1.617.2) variant, with an increased number of post-vaccination breakthrough infections were reported in the world recently. Actually, Delta variant not only resulted in a severe surge of vaccine breakthrough infections which was accompanied with high viral load and transmissibility, but also challenged the development of effective vaccines. Therefore, the biological characteristics and epidemiological profile of Delta variant, the current status of Delta variant vaccine breakthrough infections and the mechanism of vaccine breakthrough infections were discussed in this article. In addition, the significant role of the Delta variant spike (S) protein in the mechanism of immune escape of SARS-CoV-2 was highlighted in this article. In particular, we further discussed key points on the future SARS-CoV-2 vaccine research and development, hoping to make a contribution to the early, accurate and rapid control of the COVID-19 epidemic.
Collapse
Affiliation(s)
- Mengxin Zhang
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| | - Ying Liang
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| | - Dongsheng Yu
- Department of Chinese Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Bang Du
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| | - Weyland Cheng
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| | - Lifeng Li
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| | - Zhidan Yu
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| | - Shuying Luo
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| | - Yaodong Zhang
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| | - Huanmin Wang
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| | - Xianwei Zhang
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| | - Wancun Zhang
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| |
Collapse
|
1393
|
Sanyaolu A, Marinkovic A, Prakash S, Desai P, Haider N, Abbasi AF, Mehraban N, Jain I, Ekeh A, Shazley O, Okorie C, Orish VN. Reactogenicity to COVID-19 vaccination in the United States of America. Clin Exp Vaccine Res 2022; 11:104-115. [PMID: 35223671 PMCID: PMC8844673 DOI: 10.7774/cevr.2022.11.1.104] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 11/15/2021] [Indexed: 12/21/2022] Open
Abstract
PURPOSE In the United States, Pfizer-BioNTech, Moderna, and Janssen's coronavirus disease 2019 (COVID-19) vaccines have been granted Emergency Use Authorization (EUA) with the Pfizer-BioNTech vaccine presently approved by the US Food and Drug Administration. The purpose of this study is to analyze passive surveillance data on COVID-19 vaccine adverse reaction in the United States. MATERIALS AND METHODS We analyzed passive surveillance data on COVID-19 vaccine adverse reactions which were retrieved from the Vaccine Adverse Event Reporting System database. Retrieved records on demographic information as well as the top 10 common vaccine adverse events were extracted and assessed from 200 of the most recently reported cases for the study analysis. RESULTS Local and systemic adverse reactions were reported in the study. A significant difference (p<0.05) was recorded for the top 10 systemic reactions by age category (0.041) and by gender (0.002). Analysis of the top five systemic reactions, stratified by vaccine type yielded a significant difference (p<0.05) for chills (p=0.044), and when stratified by age group and type of vaccination received, it yielded a significant difference (p<0.05) for fatigue (p=0.023). Overall, Pfizer had 182 persons (91.0%) reporting adverse events, Moderna with 13 (6.5%), and Janssen with 5 (2.5%). CONCLUSION Mild side effects were reported following vaccination with the EUA COVID-19 vaccines in the United States. Thus, continuous monitoring and reporting of all adverse events are recommended to ensure the safety of vaccination.
Collapse
Affiliation(s)
| | | | | | - Priyank Desai
- American University of Saint Vincent School of Medicine, Kingstown, Saint Vincent and the Grenadines
| | - Nafees Haider
- All Saints University School of Medicine, Roseau, Dominica
| | | | - Nasima Mehraban
- Saint James School of Medicine, Anguilla, British West Indies
| | - Isha Jain
- Windsor University School of Medicine, Cayon, Saint Kitts and Nevis
| | - Amarachi Ekeh
- Avalon University School of Medicine, Willemstad, Curacao
| | - Omar Shazley
- Saint James School of Medicine, Arnos Vale, St. Vincent and the Grenadines
| | - Chuku Okorie
- Union County College (Plainfield Campus), Plainfield, NJ, USA
| | | |
Collapse
|
1394
|
Pecetta S, Kratochvil S, Kato Y, Vadivelu K, Rappuoli R. Immunology and Technology of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Vaccines. Pharmacol Rev 2022; 74:313-339. [PMID: 35101964 DOI: 10.1124/pharmrev.120.000285] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
We have experienced an enormous cohesive effort of the scientific community to understand how the immune system reacts to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and how to elicit protective immunity via vaccination. This effort resulted in the development of vaccines in record time with high levels of safety, efficacy, and real-life effectiveness. However, the rapid diffusion of viral variants that escape protective antibodies prompted new studies to understand SARS-CoV-2 vulnerabilities and strategies to guide follow-up actions to increase, and maintain, the protection offered by vaccines. In this review, we report the main findings on human immunity to SARS-CoV-2 after natural infection and vaccination; we dissect the immunogenicity and efficacy of the different vaccination strategies that resulted in products widely used in the population; and we describe the impact of viral variants on vaccine-elicited immunity, summarizing the main discoveries and challenges to stay ahead of SARS-CoV-2 evolution. SIGNIFICANCE STATEMENT: This study reviewed findings on human immunity to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), analyzed the immunogenicity and efficacy of the various vaccines currently used in large vaccination campaigns or candidates in advanced clinical development, and discussed the challenging task to ensure high protective efficacy against the rapidly evolving SARS-CoV-2 virus. This manuscript was completed prior to the emergence of the Omicron variant and to global vaccine boosting efforts.
Collapse
Affiliation(s)
- Simone Pecetta
- Research and Development Centre, GSK, Siena, Italy (S.P., K.V., R.R.); Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, Massachusetts (S.K.); IconOVir Bio, San Diego, California (Y.K.); and La Jolla Institute for Immunology, La Jolla, California (Y.K.)
| | - Sven Kratochvil
- Research and Development Centre, GSK, Siena, Italy (S.P., K.V., R.R.); Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, Massachusetts (S.K.); IconOVir Bio, San Diego, California (Y.K.); and La Jolla Institute for Immunology, La Jolla, California (Y.K.)
| | - Yu Kato
- Research and Development Centre, GSK, Siena, Italy (S.P., K.V., R.R.); Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, Massachusetts (S.K.); IconOVir Bio, San Diego, California (Y.K.); and La Jolla Institute for Immunology, La Jolla, California (Y.K.)
| | - Kumaran Vadivelu
- Research and Development Centre, GSK, Siena, Italy (S.P., K.V., R.R.); Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, Massachusetts (S.K.); IconOVir Bio, San Diego, California (Y.K.); and La Jolla Institute for Immunology, La Jolla, California (Y.K.)
| | - Rino Rappuoli
- Research and Development Centre, GSK, Siena, Italy (S.P., K.V., R.R.); Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, Massachusetts (S.K.); IconOVir Bio, San Diego, California (Y.K.); and La Jolla Institute for Immunology, La Jolla, California (Y.K.)
| |
Collapse
|
1395
|
Chen Z, Gao X, Yu D. Longevity of vaccine protection: Immunological mechanism, assessment methods, and improving strategy. VIEW 2022. [DOI: 10.1002/viw.20200103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Zhian Chen
- The University of Queensland Diamantina Institute, Faculty of Medicine The University of Queensland Brisbane Queensland Australia
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research Australian National University Canberra Australia
| | - Xin Gao
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research Australian National University Canberra Australia
| | - Di Yu
- The University of Queensland Diamantina Institute, Faculty of Medicine The University of Queensland Brisbane Queensland Australia
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research Australian National University Canberra Australia
| |
Collapse
|
1396
|
Đorđević Betetto L, Luzar B, Pipan Tkalec Ž, Ponorac S. Cutaneous leukocytoclastic vasculitis following COVID-19 vaccination with Ad26.COV2.S vaccine: a case report and literature review. ACTA DERMATOVENEROLOGICA ALPINA PANNONICA ET ADRIATICA 2022. [DOI: 10.15570/actaapa.2022.12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
1397
|
Ashrani AA, Crusan DJ, Petterson T, Bailey K, Heit JA. Age- and Sex-Specific Incidence of Cerebral Venous Sinus Thrombosis Associated With Ad26.COV2.S COVID-19 Vaccination. JAMA Intern Med 2022; 182:80-83. [PMID: 34724036 PMCID: PMC8561428 DOI: 10.1001/jamainternmed.2021.6352] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
This cohort study compares the sex- and age-adjusted incidence of cerebral venous sinus thrombosis before the COVID-19 pandemic with that during the first 92 days after Ad26.COV2.S vaccination.
Collapse
Affiliation(s)
- Aneel A Ashrani
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Daniel J Crusan
- Divisions of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Tanya Petterson
- Divisions of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Kent Bailey
- Divisions of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - John A Heit
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota.,Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota.,Division of Epidemiology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
1398
|
Finsterer J, Scorza CA, Scorza FA. Guillain-Barre syndrome related to SARS-CoV-2 vaccinations. Clinics (Sao Paulo) 2022; 77:100113. [PMID: 36137343 PMCID: PMC9464593 DOI: 10.1016/j.clinsp.2022.100113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 08/16/2022] [Accepted: 08/31/2022] [Indexed: 12/15/2022] Open
Key Words
- aidp, acute, inflammatory demyelinating polyneuropathy
- aman, acute, motor, axonal neuropathy
- amsan, acute, motor, and sensory axonal neuropathy
- azv, astra zeneca vaccine
- cns, central nervous system
- fda, food and drug administration
- gbs, guillain barres syndrome
- ivig, ntravenous immunoglobulins
- mhra, medicine and healthcare products regulatory agency
- pcb, pharyngo-cervico-brachial
- pe, plasma exchange
- pns, pcripheral nervous system
- sc2vag, sars-cov-2 vaccination associated gbs
Collapse
Affiliation(s)
| | - Carla A Scorza
- Disciplina de Neurociência, Universidade Federal de São Paulo, Escola Paulista de Medicina (UNIFESP/EPM), São Paulo, SP, Brasil
| | - Fulvio A Scorza
- Disciplina de Neurociência, Universidade Federal de São Paulo, Escola Paulista de Medicina (UNIFESP/EPM), São Paulo, SP, Brasil
| |
Collapse
|
1399
|
Breeher LE, Wolf ME, Geyer H, Brinker T, Tommaso C, Kohlnhofer S, Hainy C, Swift M. Work Absence Following COVID-19 Vaccination in a Cohort of Healthcare Personnel. J Occup Environ Med 2022; 64:6-9. [PMID: 34982070 PMCID: PMC8715930 DOI: 10.1097/jom.0000000000002376] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To identify rates of work absence following receipt of COVID-19 vaccine in a cohort of healthcare personnel (HCP). METHODS Short-term disability (STD) usage by HCP attributed to side effects of the COVID-19 vaccine was calculated for each vaccine manufacturer, job category, age group, and work region. Analysis was performed for the cohort of HCP during the initial vaccination campaign. RESULTS 4.1% of COVID-19 vaccinations generated a STD claim for lost work due to side effects, with increased STD rates after dose 2 than dose 1 (7.4% and 0.9%, respectively). Rates were higher for younger HCP and allied health staff. CONCLUSIONS While side effects from mRNA vaccine dose 2 resulted in more work absence, statistically significant geographic differences in STD suggest cultural and staffing factors may impact HCP to utilize STD following vaccination.
Collapse
Affiliation(s)
- Laura E Breeher
- Occupational Health Services, Practice Administration, Mayo Clinic, Rochester, Minnesota (Dr Breeher, Dr Tommaso, Hainy, Dr Swift), Division of Preventive, Occupational, and Aerospace Medicine, Mayo Clinic, Rochester, Minnesota (Dr Breeher, Dr Wolf, Dr Swift), Occupational Health Services, Mayo Clinic, Scottsdale, Arizona (Dr Geyer), Department of Hospital Internal Medicine, Mayo Clinic, Scottsdale, Arizona (Dr Geyer), Employee Health Services, Mayo Clinic, Jacksonville, Florida (Dr Brinker), Department of Family Medicine, Mayo Clinic, Jacksonville, Florida (Dr Brinker), Recovery and Claims Services, Mayo Clinic, Rochester, Minnesota (Kohlnhofer)
| | | | | | | | | | | | | | | |
Collapse
|
1400
|
Cauchi M, Ball H, Ben-Shlomo Y, Robertson N. Interpretation of vaccine associated neurological adverse events: a methodological and historical review. J Neurol 2022; 269:493-503. [PMID: 34398270 PMCID: PMC8366487 DOI: 10.1007/s00415-021-10747-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 08/06/2021] [Indexed: 12/18/2022]
Abstract
As a result of significant recent scientific investment, the range of vaccines available for COVID-19 prevention continues to expand and uptake is increasing globally. Although initial trial safety data have been generally reassuring, a number of adverse events, including vaccine induced thrombosis and thrombocytopenia (VITT), have come to light which have the potential to undermine the success of the vaccination program. However, it can be difficult to interpret available data and put these into context and to communicate this effectively. In this review, we discuss contemporary methodologies employed to investigate possible associations between vaccination and adverse neurological outcomes and why determining causality can be challenging. We demonstrate these issues by discussing relevant historical exemplars and explore the relevance for the current pandemic and vaccination program. We also discuss challenges in understanding and communicating such risks to clinicians and the general population within the context of the 'infodemic' facilitated by the Internet and other media.
Collapse
Affiliation(s)
- Marija Cauchi
- Division of Psychological Medicine and Clinical Neuroscience, Department of Neurology, University Hospital of Wales, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK.
| | - Harriet Ball
- Population Health Sciences, Bristol Medical School, Bristol, BS8 2PS UK
| | - Yoav Ben-Shlomo
- Population Health Sciences, Bristol Medical School, Bristol, BS8 2PS UK
| | - Neil Robertson
- Division of Psychological Medicine and Clinical Neuroscience, Department of Neurology, University Hospital of Wales, Cardiff University, Heath Park, Cardiff, CF14 4XN UK
| |
Collapse
|